1
|
McAtee D, Abdelmoneim A. A peripheral irritant motor response (PIMR) assay to identify chemical-induced locomotor deficits in larval zebrafish (Danio rerio). Neurotoxicology 2025; 108:344-353. [PMID: 40348092 DOI: 10.1016/j.neuro.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Zebrafish (Danio rerio) behavioral assays provide valuable insights into the effects of environmental chemicals on the developing nervous system, primarily through motor responses triggered by stimuli-induced CNS activation. However, as these responses rely on the locomotor integrity of larval zebrafish, chemical-induced impairments to their locomotor capacity could obscure behavioral observations and confound findings concerning the developmental neurotoxicity of the tested chemicals. This limitation emphasizes the need for supporting assays designed to specifically evaluate the locomotor capacity of larval zebrafish. In the present study, we evaluated the use of peripheral irritant-elicited motor responses as a method to identify chemical-induced locomotor deficits. The motor activity of 120 hours post-fertilization (hpf) zebrafish larvae exposed to different concentrations of two peripheral stimulants-mustard oil and cinnamon oil-was evaluated. Subsequently, we assessed changes to central (visual and acoustic) and peripheral (irritant) motor responses after tricaine-s (MS-222; neurodepressant) and tubocurarine (neuromuscular blocker) exposures. Additional investigations were also carried out to assess the central and peripheral motor activity of larvae after developmental exposures (114 h) to lead (Pb) and cadmium (Cd)-two suggested developmental neurotoxic environmental contaminants. Our observations revealed that exposure to mustard oil (12.5 µM) elicits the strongest motor response. Larvae exposed to MS-222 showed decreases in motor responses to visual and acoustic stimuli, but the same exposure induced limited effects on motor responses elicited by the peripheral irritant. Exposure to tubocurarine depressed all motor responses examined. Finally, both Pb and Cd exposures induced hypoactivation of central motor responses, but only Cd showed a significant depression in the peripheral irritant motor response (PIMR) in both intact and developmentally deformed larvae. This finding suggests that loss of locomotor capacity might be confounding the behavioral observations associated with Cd exposures. This research underscores the utility of this zebrafish-based PIMR assay in elucidating locomotor impairments induced by chemicals, which may obscure the behavioral findings of motor response assays designed to evaluate developmental neurotoxicity.
Collapse
Affiliation(s)
- Demetrius McAtee
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Ahmed Abdelmoneim
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
2
|
Hirao H, Honda M, Tomita M, Li L, Adawy A, Xue W, Hibi T. Intravital Imaging of Immune Responses in the Cancer Microenvironment. Cancer Med 2025; 14:e70899. [PMID: 40257446 PMCID: PMC12010765 DOI: 10.1002/cam4.70899] [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: 02/08/2025] [Revised: 03/18/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND To date, many types of immune cells have been identified, but their precise role in cancer immunity remains unclear. Understanding the immune responses involved in cancer and the cancer microenvironment is becoming increasingly important for elucidating disease mechanisms. In recent years, the application of intravital imaging in cancer research has provided new insights into the mechanisms of cancer-specific immune events, including innate and adaptive immunity. RESULTS In this review, we focus on the emerging role of intravital imaging in cancer research and describe how cancer and immune cells can be observed using intravital imaging in vivo. We also discuss new insights gained by this state-of-the-art technique. CONCLUSIONS Intravital imaging is a relatively new field of research that offers significant advantages, including the ability to directly capture cell-cell interactions, pathophysiology, and immune cell dynamics in the cancer microenvironment in vivo.
Collapse
Affiliation(s)
- Hiroki Hirao
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Masaki Honda
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Masahiro Tomita
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Lianbo Li
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Ahmad Adawy
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Weijie Xue
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Taizo Hibi
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| |
Collapse
|
3
|
Sudhakaran G, Ramamurthy K, Dhaareshwar VN, Rajakrishnan R, Alfarhan A, Arockiaraj J. Liquid mosquito repellent induces toxic effects in zebrafish. Drug Chem Toxicol 2025; 48:404-415. [PMID: 39686665 DOI: 10.1080/01480545.2024.2436511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024]
Abstract
Mosquitoes serve as vectors for life-threatening parasitic diseases, presenting a continuous threat throughout human history. This has resulted in the extensive utilization of various mosquito repellents, including liquid mosquito repellents (LMRs), roll-ons, and topical creams. While these products demonstrate significant efficacy, the toxicological implications associated with their use are not yet fully understood and continue to be a subject of debate. The analysis conducted using gas chromatography-mass spectrometry (GC-MS) on LMR revealed the presence of 158 distinct compounds, among which were Piperazine 2,5-dimethyl propyl and a range of hydrocarbons. The analysis of network toxicology indicated that 78 of the examined compounds contravened Lipinski's rule of five and exhibited considerable overlap with target genes associated with lung cancer pathways, thereby highlighting potential concerns regarding their carcinogenic properties. The exposure of zebrafish embryos to LMR concentrations between 0.1 and 14 µg/mL resulted in developmental toxicity assays that demonstrated a dose-dependent escalation in mortality rates and the occurrence of morphological abnormalities, such as pericardial edema and skeletal deformities. Behavioral assays demonstrated a marked decrease in locomotor activity at elevated LMR concentrations, indicating potential neurotoxic effects. Biochemical analyses revealed elevated levels of reactive oxygen species (ROS), enhanced lipid peroxidation, and diminished glutathione, which are indicative of oxidative stress. Enzyme activity assays indicated a reduction in superoxide dismutase (SOD) and catalase (CAT) activities, alongside an increase in lactate dehydrogenase (LDH) activity, which suggests the occurrence of cellular damage. Analysis of gene expression demonstrated significant dysregulation in genes associated with oxidative stress (SOD1, CAT), inflammatory markers (TNF-α, IL-1β), apoptotic regulators (p53, bcl2), and neurobiological genes (brain-derived neurotrophic factor, bdnf). The results highlight the possible health hazards linked to LMR exposure, which manifest as developmental, biochemical, and genetic alterations in zebrafish embryos.
Collapse
Affiliation(s)
- Gokul Sudhakaran
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Karthikeyan Ramamurthy
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, India
| | - V N Dhaareshwar
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, India
| | - R Rajakrishnan
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Alfarhan
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, India
| |
Collapse
|
4
|
Lachowicz-Radulska J, Widelski J, Nowaczyński F, Serefko A, Sobczyński J, Ludwiczuk A, Kasica N, Szopa A. Zebrafish as a Suitable Model for Utilizing the Bioactivity of Coumarins and Coumarin-Based Compounds. Int J Mol Sci 2025; 26:1444. [PMID: 40003910 PMCID: PMC11855297 DOI: 10.3390/ijms26041444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/03/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
The aim of this review is to summarize the current knowledge on the use of coumarin-derived compounds in the zebrafish (Danio rerio) model. Coumarins, a class of naturally occurring compounds with diverse biological activities, including compounds such as coumarin, angelicin, and warfarin, have attracted considerable attention in the study of potential therapeutic agents for cancer, central nervous system disorders, and infectious diseases. The capabilities of coumarins as active compounds have led to synthesizing various derivatives with their own properties. While such variety is certainly promising, it is also cumbersome due to the large amount of research needed to find the most optimal compounds. The zebrafish model offers unique advantages for such studies, including high genetic and physiological homology to mammals, optical transparency of the embryos, and rapid developmental processes, facilitating the assessment of compound toxicity and underlying mechanisms of action. This review provides an in-depth analysis of the chemical properties of coumarins, their mechanisms of biological activity, and the results of previous studies evaluating the toxicity and efficacy of these compounds in zebrafish assays. The zebrafish model allows for a holistic assessment of the therapeutic potential of coumarin derivatives, offering valuable insights for advancing drug discovery and development.
Collapse
Affiliation(s)
- Joanna Lachowicz-Radulska
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Lublin, 7 Chodźki Street, 20-093 Lublin, Poland; (J.L.-R.); (F.N.); (A.S.); (J.S.)
| | - Jarosław Widelski
- Department of Pharmacognosy with Medicinal Plants Garden, Medical University of Lublin, 1 Chodźki Street, 20-093 Lublin, Poland; (J.W.); (A.L.)
| | - Filip Nowaczyński
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Lublin, 7 Chodźki Street, 20-093 Lublin, Poland; (J.L.-R.); (F.N.); (A.S.); (J.S.)
- Department of Pharmacognosy with Medicinal Plants Garden, Medical University of Lublin, 1 Chodźki Street, 20-093 Lublin, Poland; (J.W.); (A.L.)
| | - Anna Serefko
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Lublin, 7 Chodźki Street, 20-093 Lublin, Poland; (J.L.-R.); (F.N.); (A.S.); (J.S.)
| | - Jan Sobczyński
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Lublin, 7 Chodźki Street, 20-093 Lublin, Poland; (J.L.-R.); (F.N.); (A.S.); (J.S.)
| | - Agnieszka Ludwiczuk
- Department of Pharmacognosy with Medicinal Plants Garden, Medical University of Lublin, 1 Chodźki Street, 20-093 Lublin, Poland; (J.W.); (A.L.)
| | - Natalia Kasica
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Aleksandra Szopa
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Lublin, 7 Chodźki Street, 20-093 Lublin, Poland; (J.L.-R.); (F.N.); (A.S.); (J.S.)
| |
Collapse
|
5
|
Sudhakaran G, Karthikeyan R, Madesh S, Dhaareshwar VN, Pachaiappan R, Rajagopal R, Soundharrajan I, Karthick Raja Namasivayam S, Arockiaraj J. Neurotoxic effects and cognitive decline induced by Aniline Blue dye in an in-vivo zebrafish model. Toxicology 2025; 511:154065. [PMID: 39880357 DOI: 10.1016/j.tox.2025.154065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 01/31/2025]
Abstract
Aniline Blue is a synthetic dye extensively used in various industries, including textiles, plastics, and biological research due to its effective staining properties. However, its environmental and health impacts, particularly its neurotoxic effects, are poorly understood. While the dye has been associated with carcinogenicity and organ toxicity, the neurobehavioral consequences of Aniline Blue exposure remain underexplored. This study aims to address this research gap by evaluating the neurotoxic effects of Aniline Blue using adult zebrafish as a model organism. We conducted a series of experiments to assess the dose-dependent toxicity of Aniline Blue, examining survival rates, cognitive function, anxiety-related behaviors, locomotor activity, oxidative stress markers, histopathological changes, and gene expression profiles. Zebrafish were exposed to varying concentrations of Aniline Blue over a specified period, followed by behavioral assays, biochemical analyses, and gene expression studies. The results revealed an apparent dose-dependent decline in survival rates, with higher concentrations of Aniline Blue significantly impairing cognitive function and increasing anxiety-like behaviors. Biochemical assays showed a reduction in antioxidant enzyme activities (SOD and CAT) and an increase in LDH activity, indicating oxidative stress. Histopathological analysis confirmed severe neuronal damage, including neuroinflammation and cell death. Gene expression analysis further highlighted the modulation of critical genes associated with neuroinflammation, neurotrophic support, and synaptic function, with potential implications for neurodegenerative diseases such as Alzheimer's. In conclusion, the study provides critical insights into the neurotoxic effects of Aniline Blue, emphasizing the need for caution in its use across industries.
Collapse
Affiliation(s)
- Gokul Sudhakaran
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - R Karthikeyan
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu 603203, India
| | - S Madesh
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu 603203, India
| | - V N Dhaareshwar
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu 603203, India
| | - Raman Pachaiappan
- Department of Biotechnology, School of Bioengineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, District, Tamil Nadu 603203, India
| | - Rajakrishnan Rajagopal
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Ilavenil Soundharrajan
- Grassland Forages Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea
| | - S Karthick Raja Namasivayam
- Centre for Applied Research, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu 602105, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu 603203, India.
| |
Collapse
|
6
|
Gao N, Yang L, Zhu L, Zhu L, Feng J. New Insights into the Visual Toxicity of Organophosphate Esters: An Integrated Quantitative Adverse Outcome Pathway and Cross-chemical Extrapolation. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:22039-22052. [PMID: 39631370 DOI: 10.1021/acs.est.4c08176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Organophosphate esters (OPEs) have been documented to adversely affect visual function, potentially impacting wildlife survival and human health, thereby necessitating a comprehensive risk assessment. Despite the quantitative adverse outcome pathway (qAOP) holding promise for addressing this concern mechanistically, unclear mode of action and inadequate quantitative understanding across biological levels currently impede its development. Herein, we employed an integrated strategy, combining multiomics analyses, targeted bioassays, and modular model-fitting, to develop and validate a qAOP framework for visual toxicity of OPEs, exemplified by tris(2-butoxyethyl) phosphate, triphenyl phosphate, and tris(1,3-dichloro-2-propyl) phosphate. Our results revealed that these OPEs induce visual dysfunction in zebrafish larvae primarily via oxidative stress, then cascade to damaging photoreceptors and retinal structures, ultimately resulting in the disruption of visual behaviors (i.e., decreased optokinetic response, phototaxis, and visual motor response). The qAOP, validated through cross-chemical extrapolation, enabled the prediction for vision-related effects of OPEs within a certain domain. Integrating toxicokinetic modeling could compensate for the uncertainty in qAOP predictions, since adjusting for internal concentrations as inputs significantly enhanced the accuracy and applicability of the predictions. This work contributes to a better understanding of visual toxicity by OPEs and presents a promising paradigm for quantitative risk assessment based on the qAOP framework.
Collapse
Affiliation(s)
- Ning Gao
- Key Laboratory of Pollution Process and Environmental Criteria of Ministry of Education and Tianjin Key Laboratory of Environmental Technology for Complex Trans-Media Pollution, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Lanpeng Yang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon 999077, Hong Kong, China
| | - Lingyan Zhu
- Key Laboratory of Pollution Process and Environmental Criteria of Ministry of Education and Tianjin Key Laboratory of Environmental Technology for Complex Trans-Media Pollution, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Lin Zhu
- Key Laboratory of Pollution Process and Environmental Criteria of Ministry of Education and Tianjin Key Laboratory of Environmental Technology for Complex Trans-Media Pollution, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Jianfeng Feng
- Key Laboratory of Pollution Process and Environmental Criteria of Ministry of Education and Tianjin Key Laboratory of Environmental Technology for Complex Trans-Media Pollution, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| |
Collapse
|
7
|
de Araujo MH, Muñoz Sánchez S, Simão TLBV, Nowik N, Antunes SS, Pinto SC, Sorze D, Boldrin F, Manganelli R, Correia Romeiro N, Lasunskaia EB, Verbeek FJ, Spaink HP, Muzitano MF. Exploring the Antimycobacterial Potential of Podocarpusflavone A from Kielmeyera membranacea: In Vitro and In Vivo Insights. Pharmaceuticals (Basel) 2024; 17:1560. [PMID: 39770402 PMCID: PMC11676425 DOI: 10.3390/ph17121560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Tuberculosis (TB) is one of the leading infectious causes of death worldwide, highlighting the importance of identifying new anti-TB agents. In previous research, our team identified antimycobacterial activity in Kielmeyera membranacea leaf extract; therefore, this study aims to conduct further exploration of its potential. Methods: Classical chromatography was applied for fractionation and spectrometric techniques were utilized for chemical characterization. For in vitro tests, samples were assessed against Mycobacterium tuberculosis and Mycobacterium marinum. The toxicity and efficacy of active samples were evaluated in vivo using different zebrafish models. Chemogenomics studies were applied to predict the isolated active compound's potential mode of action. Results: We performed fractionation of K. membranacea ethanolic extract (EE) and then its dichloromethane fraction (DCM), and the biflavonoid podocarpusflavone A (PCFA) was isolated and identified as a promising active compound. The EE and PCFA were found to be non-toxic to zebrafish larvae and were able to inhibit M. tuberculosis growth extracellularly. Additionally, PCFA demonstrated antimycobacterial activity within infected macrophages, especially when combined with isoniazid. In addition, the EE, DCM, and PCFA have shown the ability to inhibit M. marinum's growth during in vivo zebrafish larvae yolk infection. Notably, PCFA also effectively countered systemic infection established through the caudal vein, showing a similar inhibitory activity profile to rifampicin, both at 32 µM. A reduction in the transcriptional levels of pro-inflammatory cytokines confirmed the infection resolution. The protein tyrosine phosphatase B (PtpB) of M. tuberculosis, which inhibits the macrophage immune response, was predicted as a theoretical target of PCFA. This finding is in agreement with the higher activity observed for PCFA intracellularly and in vivo on zebrafish, compared with the direct action in M. tuberculosis. Conclusions: Here, we describe the discovery of PCFA as an intracellular inhibitor of M. tuberculosis and provide evidence of its in vivo efficacy and safety, encouraging its further development as a combination drug in novel therapeutic regimens for TB.
Collapse
Affiliation(s)
- Marlon Heggdorne de Araujo
- Laboratório de Produtos Bioativos (LPBio), Instituto de Ciências Farmacêuticas, Universidade Federal do Rio de Janeiro, Campus Macaé, Macaé 27930-560, RJ, Brazil; (M.H.d.A.); (S.S.A.); (S.C.P.)
- Department of Animal Sciences and Health, Institute of Biology (IBL), Leiden University, 2333 BE Leiden, The Netherlands; (S.M.S.); (H.P.S.)
| | - Salomé Muñoz Sánchez
- Department of Animal Sciences and Health, Institute of Biology (IBL), Leiden University, 2333 BE Leiden, The Netherlands; (S.M.S.); (H.P.S.)
| | - Thatiana Lopes Biá Ventura Simão
- Laboratório de Biologia do Reconhecer (LBR), Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes 28013-602, RJ, Brazil; (T.L.B.V.S.); (E.B.L.)
| | - Natalia Nowik
- Department of Animal Sciences and Health, Institute of Biology (IBL), Leiden University, 2333 BE Leiden, The Netherlands; (S.M.S.); (H.P.S.)
| | - Stella Schuenck Antunes
- Laboratório de Produtos Bioativos (LPBio), Instituto de Ciências Farmacêuticas, Universidade Federal do Rio de Janeiro, Campus Macaé, Macaé 27930-560, RJ, Brazil; (M.H.d.A.); (S.S.A.); (S.C.P.)
- Laboratório Integrado de Computação Científica (LICC), Universidade Federal do Rio de Janeiro, Campus Macaé, Macaé 27930-560, RJ, Brazil;
| | - Shaft Corrêa Pinto
- Laboratório de Produtos Bioativos (LPBio), Instituto de Ciências Farmacêuticas, Universidade Federal do Rio de Janeiro, Campus Macaé, Macaé 27930-560, RJ, Brazil; (M.H.d.A.); (S.S.A.); (S.C.P.)
| | - Davide Sorze
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (D.S.); (F.B.); (R.M.)
| | - Francesca Boldrin
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (D.S.); (F.B.); (R.M.)
| | - Riccardo Manganelli
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (D.S.); (F.B.); (R.M.)
| | - Nelilma Correia Romeiro
- Laboratório Integrado de Computação Científica (LICC), Universidade Federal do Rio de Janeiro, Campus Macaé, Macaé 27930-560, RJ, Brazil;
| | - Elena B. Lasunskaia
- Laboratório de Biologia do Reconhecer (LBR), Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes 28013-602, RJ, Brazil; (T.L.B.V.S.); (E.B.L.)
| | - Fons J. Verbeek
- Leiden Institute of Advanced Computer Science, Leiden University, 2333 CA Leiden, The Netherlands;
| | - Herman P. Spaink
- Department of Animal Sciences and Health, Institute of Biology (IBL), Leiden University, 2333 BE Leiden, The Netherlands; (S.M.S.); (H.P.S.)
| | - Michelle Frazão Muzitano
- Laboratório de Produtos Bioativos (LPBio), Instituto de Ciências Farmacêuticas, Universidade Federal do Rio de Janeiro, Campus Macaé, Macaé 27930-560, RJ, Brazil; (M.H.d.A.); (S.S.A.); (S.C.P.)
| |
Collapse
|
8
|
Verkerk L, Verkerk AO, Wilders R. Zebrafish as a Model System for Brugada Syndrome. Rev Cardiovasc Med 2024; 25:313. [PMID: 39355588 PMCID: PMC11440409 DOI: 10.31083/j.rcm2509313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 10/03/2024] Open
Abstract
Brugada syndrome (BrS) is an inheritable cardiac arrhythmogenic disease, associated with an increased risk of sudden cardiac death. It is most common in males around the age of 40 and the prevalence is higher in Asia than in Europe and the United States. The pathophysiology underlying BrS is not completely understood, but several hypotheses have been proposed. So far, the best effective treatment is the implantation of an implantable cardioverter-defibrillator (ICD), but device-related complications are not uncommon. Therefore, there is an urgent need to improve diagnosis and risk stratification and to find new treatment options. To this end, research should further elucidate the genetic basis and pathophysiological mechanisms of BrS. Several experimental models are being used to gain insight into these aspects. The zebrafish (Danio rerio) is a widely used animal model for the study of cardiac arrhythmias, as its cardiac electrophysiology shows interesting similarities to humans. However, zebrafish have only been used in a limited number of studies on BrS, and the potential role of zebrafish in studying the mechanisms of BrS has not been reviewed. Therefore, the present review aims to evaluate zebrafish as an animal model for BrS. We conclude that zebrafish can be considered as a valuable experimental model for BrS research, not only for gene editing technologies, but also for screening potential BrS drugs.
Collapse
Affiliation(s)
- Leonie Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
9
|
López-Cuevas P, Oates TCL, Tong Q, McGowan LM, Cross SJ, Xu C, Zhao Y, Yin Z, Toye AM, Boussahel A, Hammond CL, Mann S, Martin P. Reprogramming macrophages with R848-loaded artificial protocells to modulate skin and skeletal wound healing. J Cell Sci 2024; 137:jcs262202. [PMID: 39078119 PMCID: PMC11385641 DOI: 10.1242/jcs.262202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024] Open
Abstract
After tissue injury, inflammatory cells are rapidly recruited to the wound where they clear microbes and other debris, and coordinate the behaviour of other cell lineages at the repair site in both positive and negative ways. In this study, we take advantage of the translucency and genetic tractability of zebrafish to evaluate the feasibility of reprogramming innate immune cells in vivo with cargo-loaded protocells and investigate how this alters the inflammatory response in the context of skin and skeletal repair. Using live imaging, we show that protocells loaded with R848 cargo (which targets TLR7 and TLR8 signalling), are engulfed by macrophages resulting in their switching to a pro-inflammatory phenotype and altering their regulation of angiogenesis, collagen deposition and re-epithelialization during skin wound healing, as well as dampening osteoblast and osteoclast recruitment and bone mineralization during fracture repair. For infected skin wounds, R848-reprogrammed macrophages exhibited enhanced bactericidal activities leading to improved healing. We replicated our zebrafish studies in cultured human macrophages, and showed that R848-loaded protocells similarly reprogramme human cells, indicating how this strategy might be used to modulate wound inflammation in the clinic.
Collapse
Affiliation(s)
- Paco López-Cuevas
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Tiah C L Oates
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Qiao Tong
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Lucy M McGowan
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Stephen J Cross
- Wolfson Bioimaging Facility, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Can Xu
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Yu Zhao
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Zhuping Yin
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Ashley M Toye
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell Products, University of Bristol, Bristol BS34 7QH, UK
| | - Asme Boussahel
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Chrissy L Hammond
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Stephen Mann
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
- Max Planck Bristol Centre for Minimal Biology, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Paul Martin
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
10
|
Dey S, Thamaraikani T, Vellapandian C. Advancing Alzheimer's Research With Zebrafish Models: Current Insights, Addressing Challenges, and Charting Future Courses. Cureus 2024; 16:e66935. [PMID: 39280389 PMCID: PMC11401598 DOI: 10.7759/cureus.66935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024] Open
Abstract
Alzheimer's disease (AD) is a neurological condition that progressively impairs cognitive function and results in memory loss. Despite substantial research efforts, little is known about the specific processes driving AD, and there are few proven therapies. Because of their physiological and genetic resemblance to humans, zebrafish (Danio rerio) have become an important model organism for furthering research on AD. This abstract discusses the difficulties faced, looks at the insights currently garnered from zebrafish models, and suggests future research options. AD knowledge has greatly benefited from the use of zebrafish models. Transgenic zebrafish that express human AD-associated genes, such as tau and amyloid precursor protein (APP), display tau neurofibrillary tangles (NFTs) and amyloid-beta (Aβ) plaques, two of the disease's main clinical characteristics. These models have clarified the roles of oxidative stress, inflammation, and calcium homeostasis in the course of AD and allowed for the purpose of high-throughput screening of potential therapeutic agents. Understanding the growth and deterioration of neurons has been greatly aided by real-time zebrafish imaging. Fully using zebrafish models in AD research requires addressing a number of issues. The dissimilarities in zebrafish anatomy and physiology from humans, the difficulty of developing models that replicate progressive and late-onset AD (LOAD), and the requirement for standardized procedures to evaluate alterations in zebrafish cognition and behavior are a few issues. Furthermore, variations in the genetic makeup of zebrafish strains might affect the results of experiments. Future directions include developing standardized behavioral assays and cognitive tests, working together to create extensive databases of zebrafish genetic and phenotypic data, and using genetic engineering techniques like CRISPR/Cas9 to create more complex zebrafish models. Combining zebrafish models with other model species helps expedite the conversion of research results into therapeutic applications and offers a more thorough knowledge of AD. To sum up, zebrafish models have made a substantial contribution to Alzheimer's research by offering insightful information on the causes of the illness and possible therapies. By tackling present issues and formulating a planned future path, we can improve the use of zebrafish to decipher the mysteries of Alzheimer's and help create successful treatments.
Collapse
Affiliation(s)
- Shreya Dey
- Pharmacy/Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu, IND
| | - Tamilanban Thamaraikani
- Pharmacy/Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu, IND
| | - Chitra Vellapandian
- Pharmacy/Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu, IND
| |
Collapse
|
11
|
McAtee D, Abdelmoneim A. A zebrafish-based acoustic motor response (AMR) assay to evaluate chemical-induced developmental neurotoxicity. Neurotoxicology 2024; 103:60-70. [PMID: 38851595 DOI: 10.1016/j.neuro.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 05/20/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Behavioral assays using early-developing zebrafish (Danio rerio) offer a valuable supplement to the in vitro battery adopted as new approach methodologies (NAMs) for assessing risk of chemical-induced developmental neurotoxicity. However, the behavioral assays primarily adopted rely on visual stimulation to elicit behavioral responses, known as visual motor response (VMR) assays. Ocular deficits resulting from chemical exposures can, therefore, confound the behavioral responses, independent of effects on the nervous system. This highlights the need for complementary assays employing alternative forms of sensory stimulation. In this study, we investigated the efficacy of acoustic stimuli as triggers of behavioral responses in larval zebrafish, determined the most appropriate data acquisition mode, and evaluated the suitability of an acoustic motor response (AMR) assay as means to assess alterations in brain activity and risk of chemical-induced developmental neurotoxicity. We quantified the motor responses of 120 h post-fertilization (hpf) larvae to acoustic stimuli with varying patterns and frequencies, and determined the optimal time intervals for data acquisition. Following this, we examined changes in acoustic and visual motor responses resulting from exposures to pharmacological agents known to impact brain activity (pentylenetetrazole (PTZ) and tricaine-s (MS-222)). Additionally, we examined the AMR and VMR of larvae following exposure to two environmental contaminants associated with developmental neurotoxicity: arsenic (As) and cadmium (Cd). Our findings indicate that exposure to a 100 Hz sound frequency in 100 ms pulses elicits the strongest behavioral response among the acoustic stimuli tested and data acquisition in 2 s time intervals is suitable for response assessment. Exposure to PTZ exaggerated and depressed both AMR and VMR in a concentration-dependent manner, while exposure to MS-222 only depressed them. Similarly, exposure to As and Cd induced respective hyper- and hypo-activation of both motor responses. This study highlights the efficiency of the proposed zebrafish-based AMR assay in demonstrating risk of chemical-induced developmental neurotoxicity and its suitability as a complement to the widely adopted VMR assay.
Collapse
Affiliation(s)
- Demetrius McAtee
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Ahmed Abdelmoneim
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
12
|
Lee H, Park W, An G, Park J, Lim W, Song G. Hexaconazole induces developmental toxicities via apoptosis, inflammation, and alterations of Akt and MAPK signaling cascades. Comp Biochem Physiol C Toxicol Pharmacol 2024; 279:109872. [PMID: 38423198 DOI: 10.1016/j.cbpc.2024.109872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/16/2024] [Accepted: 02/25/2024] [Indexed: 03/02/2024]
Abstract
Hexaconazole is a highly effective triazole fungicide that is frequently applied in various countries to elevate crop productivity. Given its long half-life and high water solubility, this fungicide is frequently detected in the environment, including water sources. Moreover, hexaconazole exerts hazardous effects on nontarget organisms. However, little is known about the toxic effects of hexaconazole on animal development. Thus, this study aimed to investigate the developmental toxicity of hexaconazole to zebrafish, a valuable animal model for toxicological studies, and elucidate the underlying mechanisms. Results showed that hexaconazole affected the viability and hatching rate of zebrafish at 96 h postfertilization. Hexaconazole-treated zebrafish showed phenotypic defects, such as reduced size of head and eyes and enlarged pericardiac edema. Moreover, hexaconazole induced apoptosis, DNA fragmentation, and inflammation in developing zebrafish. Various organ defects, including neurotoxicity, cardiovascular toxicity, and hepatotoxicity, were observed in transgenic zebrafish models olig2:dsRed, fli1:eGFP, and l-fabp:dsRed. Furthermore, hexaconazole treatment altered the Akt and MAPK signaling pathways, which possibly triggered the organ defects and other toxic mechanisms. This study demonstrated the developmental toxicity of hexaconazole to zebrafish and elucidated the underlying mechanisms.
Collapse
Affiliation(s)
- Hojun Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Wonhyoung Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Garam An
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Junho Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Whasun Lim
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
13
|
Camilli MP, Simko OM, Bevelander B, Thebeau JM, Masood F, da Silva MCB, Raza MF, Markova S, Obshta O, Jose MS, Biganski S, Kozii IV, Zabrodski MW, Moshynskyy I, Simko E, Wood SC. Fetal Alcohol Spectrum Disorder: The Honey Bee as a Social Animal Model. Life (Basel) 2024; 14:434. [PMID: 38672706 PMCID: PMC11051024 DOI: 10.3390/life14040434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Animal models have been essential for advancing research of fetal alcohol spectrum disorder (FASD) in humans, but few animal species effectively replicate the behavioural and clinical signs of FASD. The honey bee (Apis mellifera) is a previously unexplored research model for FASD that offers the distinct benefit of highly social behaviour. In this study, we chronically exposed honey bee larvae to incremental concentrations of 0, 3, 6, and 10% ethanol in the larval diet using an in vitro rearing protocol and measured developmental time and survival to adult eclosion, as well as body weight and motor activity of newly emerged adult bees. Larvae reared on 6 and 10% dietary ethanol demonstrated significant, dose-responsive delays to pupation and decreased survival and adult body weight. All ethanol-reared adults showed significantly decreased motor activity. These results suggest that honey bees may be a suitable social animal model for future FASD research.
Collapse
Affiliation(s)
- Marcelo P. Camilli
- Department of Veterinary Pathology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada; (M.P.C.)
| | - Olena M. Simko
- Department of Veterinary Pathology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada; (M.P.C.)
| | - Breanne Bevelander
- Department of Veterinary Pathology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada; (M.P.C.)
| | - Jenna M. Thebeau
- Department of Veterinary Pathology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada; (M.P.C.)
| | - Fatima Masood
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada
| | - Marina C. Bezerra da Silva
- Department of Veterinary Pathology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada; (M.P.C.)
| | - Muhammad Fahim Raza
- Department of Veterinary Pathology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada; (M.P.C.)
| | - Sofiia Markova
- Department of Veterinary Pathology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada; (M.P.C.)
| | - Oleksii Obshta
- Department of Veterinary Pathology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada; (M.P.C.)
| | - Midhun S. Jose
- Department of Veterinary Pathology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada; (M.P.C.)
| | - Sarah Biganski
- Department of Veterinary Pathology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada; (M.P.C.)
| | - Ivanna V. Kozii
- Prairie Diagnostic Services Inc., Saskatoon, SK S7N 5B4, Canada
| | | | - Igor Moshynskyy
- Department of Veterinary Pathology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada; (M.P.C.)
| | - Elemir Simko
- Department of Veterinary Pathology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada; (M.P.C.)
| | - Sarah C. Wood
- Department of Veterinary Pathology, Western College of Veterinary Medicine, Saskatoon, SK S7N 5B4, Canada; (M.P.C.)
| |
Collapse
|
14
|
Verma SK, Nandi A, Sinha A, Patel P, Mohanty S, Jha E, Jena S, Kumari P, Ghosh A, Jerman I, Chouhan RS, Dutt A, Samal SK, Mishra YK, Varma RS, Panda PK, Kaushik NK, Singh D, Suar M. The posterity of Zebrafish in paradigm of in vivo molecular toxicological profiling. Biomed Pharmacother 2024; 171:116160. [PMID: 38237351 DOI: 10.1016/j.biopha.2024.116160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
The aggrandised advancement in utility of advanced day-to-day materials and nanomaterials has raised serious concern on their biocompatibility with human and other biotic members. In last few decades, understanding of toxicity of these materials has been given the centre stage of research using many in vitro and in vivo models. Zebrafish (Danio rerio), a freshwater fish and a member of the minnow family has garnered much attention due to its distinct features, which make it an important and frequently used animal model in various fields of embryology and toxicological studies. Given that fertilization and development of zebrafish eggs take place externally, they serve as an excellent model organism for studying early developmental stages. Moreover, zebrafish possess a comparable genetic composition to humans and share almost 70% of their genes with mammals. This particular model organism has become increasingly popular, especially for developmental research. Moreover, it serves as a link between in vitro studies and in vivo analysis in mammals. It is an appealing choice for vertebrate research, when employing high-throughput methods, due to their small size, swift development, and relatively affordable laboratory setup. This small vertebrate has enhanced comprehension of pathobiology and drug toxicity. This review emphasizes on the recent developments in toxicity screening and assays, and the new insights gained about the toxicity of drugs through these assays. Specifically, the cardio, neural, and, hepatic toxicology studies inferred by applications of nanoparticles have been highlighted.
Collapse
Affiliation(s)
- Suresh K Verma
- School of Biotechnology, KIIT University, Bhubaneswar, India.
| | - Aditya Nandi
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Adrija Sinha
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Paritosh Patel
- School of Biotechnology, KIIT University, Bhubaneswar, India; Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897, Seoul, South Korea
| | | | - Ealisha Jha
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Snehasmita Jena
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Puja Kumari
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno 61137, Czech Republic
| | - Aishee Ghosh
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Ivan Jerman
- National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Raghuraj Singh Chouhan
- Department of Environmental Sciences, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Ateet Dutt
- Instituto de Investigaciones en Materiales, UNAM, CDMX, Mexico
| | - Shailesh Kumar Samal
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Yogendra Kumar Mishra
- Mads Clausen Institute, NanoSYD, University of Southern Denmark, Alsion 2, Sønderborg DK-6400, Denmark
| | - Rajender S Varma
- Institute for Nanomaterials, Advanced Technologies and Innovation (CxI), Technical University of Liberec (TUL), Studentská 1402/2, Liberec 1 461 17, Czech Republic
| | - Pritam Kumar Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897, Seoul, South Korea.
| | - Deobrat Singh
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden.
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar, India.
| |
Collapse
|
15
|
Ding Z, Jia H, Yang Z, Yao N, Wang Y. The cardiovascular toxicity of clozapine in embryonic zebrafish and RNA sequencing-based transcriptome analysis. J Appl Toxicol 2024; 44:175-183. [PMID: 37605992 DOI: 10.1002/jat.4530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 07/29/2023] [Accepted: 08/07/2023] [Indexed: 08/23/2023]
Abstract
Clozapine (CLZ) is the most prescribed medication for treating refractory schizophrenia but is associated with significant cardiovascular toxicity. This study aimed to investigate the cardiovascular toxicity induced by CLZ using zebrafish as a model animal. For this purpose, zebrafish developed to 80-h post-fertilization were exposed to different CLZ concentration solutions for 24 h followed by cardiac morphological observations in yolk sac edema, pericardial edema, and blood coagulation, in addition to increased SV-BA distance, functionally manifested as bradycardia, and decreased cardiac ejection fraction using the untreated embryos as control. At the same time, RNA sequencing was used to study the possible molecular mechanism of CLZ-induced cardiovascular toxicity. The results indicated that compared to the control group, the experimental groups possessed a total of 5888 differentially expressed genes (DEGs), where gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment of analysis indicated that DEGs were mainly enriched in the pathways related to ion channels. These findings may provide new insights and directions for the subsequent in-depth study of the molecular mechanism of CLZ-induced cardiovascular toxicity.
Collapse
Affiliation(s)
- Zijiao Ding
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huiting Jia
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ziqian Yang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Nan Yao
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Yunyun Wang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
16
|
Karaman GE, Ünal İ, Beler M, Üstündağ FD, Cansız D, Üstündağ ÜV, Emekli-Alturfan E, Akyüz S. Toothpastes for children and their detergent contents affect molecular mechanisms of odontogenesis in zebrafish embryos. Drug Chem Toxicol 2024; 47:15-25. [PMID: 36444776 DOI: 10.1080/01480545.2022.2150208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 09/29/2022] [Accepted: 11/12/2022] [Indexed: 12/03/2022]
Abstract
We aimed to evaluate how different types of toothpaste (TP) for children affected molecular mechanisms of odontogenesis in zebrafish embryos. Commercially available TPs were selected according to their detergent contents as the cocamidopropyl betaine (CAPB) containing TP (TP1) and sodium lauryl sulfate (SLS) containing TP (TP2). TP3 contained no detergent. Effects of SLS, and CAPB alone were also examined. TP and detergent concentrations affecting development were determined as 750 mg/L and 4 mg/L, respectively. Embryos were exposed to TP1, TP2, TP3, SLS, CAPB, and embryo medium (control) for 72 h post fertilization. Acetylcholinesterase (AChE) activity and oxidant-antioxidant parameters were analyzed spectrophotometrically. Expressions of tooth development genes were evaluated by reverse transcription PCR (RT-PCR). Intraocular distance, lower jaw, and ceratohyal cartilage length were displayed using Alcian Blue staining. axin2 and wnt10a expressions increased in SLS and TP2 groups. igf2a and eve1 expressions decreased in all groups except TP3. nrOb1 expression decreased in TP1, SLS, and CAPB groups. Oxidant-antioxidant balance was disturbed in all groups except TP3, evidenced by increased lipid peroxidation, nitric oxide. SLS, and CAPB groups were more affected in terms of AChE, glutathione-S-transferase, and superoxide dismutase; perturbations were observed in cartilage structures. Altered expression of tooth development gene axin2 correlated with wnt10a, and with changes in cartilage structures in SLS and TP2 groups. TP3 group presented no disruptions in oxidant-antioxidant balance. Our study shows the availability of externally developing zebrafish embryos in examining the effects of TP' contents on embryogenesis.
Collapse
Affiliation(s)
- Gözde Ece Karaman
- Department of Paediatric Dentistry, Institute of Health Sciences, Marmara University, Istanbul, Turkey
| | - İsmail Ünal
- Department of Biochemistry, Institute of Health Sciences, Marmara University, Istanbul, Turkey
| | - Merih Beler
- Department of Biochemistry, Institute of Health Sciences, Marmara University, Istanbul, Turkey
| | - Fümet Duygu Üstündağ
- Department of Biophysics, Institute of Health Sciences, Marmara University, Istanbul, Turkey
| | - Derya Cansız
- Department of Biochemistry, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ünsal Veli Üstündağ
- Department of Biochemistry, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ebru Emekli-Alturfan
- Department of Basic Medical Sciences, Faculty of Dentistry, Marmara University, Istanbul, Turkey
| | - Serap Akyüz
- Department of Paediatric Dentistry, Faculty of Dentistry, Marmara University, Istanbul, Turkey
| |
Collapse
|
17
|
Chahardehi AM, Hosseini Y, Mahdavi SM, Naseh I. The Zebrafish Model as a New Discovery Path for Medicinal Plants in the Treatment of Parkinson's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:306-314. [PMID: 36999188 DOI: 10.2174/1871527322666230330111712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 12/29/2022] [Accepted: 01/11/2023] [Indexed: 04/01/2023]
Abstract
Parkinson's disease (PD) is one of the most frequent degenerative central nervous system disorders affecting older adults. Dopaminergic neuron failure in the substantia nigra is a pathological sign connected with the motor shortfall of PD. Due to their low teratogenic and adverse effect potential, medicinal herbs have emerged as a promising therapy option for preventing and curing PD and other neurodegenerative disorders. However, the mechanism through which natural compounds provide neuroprotection against PD remains unknown. While testing compounds in vertebrates such as mice is prohibitively expensive and time-consuming, zebrafish (Danio rerio) may offer an appealing alternative because they are vertebrates and share many of the same characteristics as humans. Zebrafish are commonly used as animal models for studying many human diseases, and their molecular history and bioimaging properties are appropriate for the study of PD. However, a literature review indicated that only six plants, including Alpinia oxyhylla, Bacopa monnieri, Canavalia gladiate, Centella asiatica, Paeonia suffruticosa, and Stachytarpheta indica had been investigated as potential PD treatments using the zebrafish model. Only C. asiatica and B. monnieri were found to have potential anti-PD activity. In addition to reviewing the current state of research in this field, these plants' putative mechanisms of action against PD are explored, and accessible assays for investigation are made.
Collapse
Affiliation(s)
| | - Yasaman Hosseini
- Cognitive Neuroscience Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Mahdavi
- Department of Bioscience and Biotechnology, Malek Ashtar University of Technology (MUT), Tehran, Iran
| | - Iman Naseh
- Cognitive Neuroscience Research Center, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Ja'apar SAN, Ichwan SJA, Mustaffa M. In vivo cytotoxicity analysis of bioceramic root canal sealers on zebrafish embryo. J Dent Res Dent Clin Dent Prospects 2023; 17:242-249. [PMID: 38584992 PMCID: PMC10998162 DOI: 10.34172/joddd.2023.39163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 11/13/2023] [Indexed: 04/09/2024] Open
Abstract
Background This study evaluated the cytotoxicity of four bioceramic root canal sealers (RCSs) in vivo. The embryonic zebrafish characteristics, such as mortality, survival, hatching, and general morphology, served as the parameters for assessing cytotoxicity. Methods The RCSs, namely GuttaFlow Bioseal, MTA Fillapex, CeraSeal Bioceramic, and iRoot SP, were mixed according to the manufacturer's guidelines. The extract solution was prepared by immersing the set RCS into 1X dilution of E3 solution. Then, the extract solution was delivered into a Petri dish where zebrafish embryos were allowed to develop. Cytotoxicity was evaluated 24, 48, 72, and 96 hours after fertilization. Results The Kruskal-Wallis test showed that except for GuttaFlow Bioseal, the mortality, survival, and hatching of zebrafish embryos for the remaining three bioceramic RCSs were significantly different from the negative controls (P<0.05). Significant differences were also evident in the mortality, survival, and hatching of zebrafish embryos between GuttaFlow Bioseal and three other RCSs (P<0.05). Conclusion GuttaFlow Bioseal was less cytotoxic than other bioceramics RCSs; MTA Fillapex, CeraSeal Bioceramic root canal sealer, and iRoot SP root canal sealer exhibited comparable cytotoxicity.
Collapse
Affiliation(s)
- Siti Aisyah Nadirah Ja'apar
- Department of Biotechnology, Kulliyyah of Sciences, International Islamic University Malaysia (IIUM), Jalan Sultan Ahmad Shah, Bandar Indera Mahkota, 25200, Kuantan, Pahang, Malaysia
| | - Solachuddin Jauhari Arief Ichwan
- Department of Fundamental Dental and Medical Sciences, Kulliyyah of Dentistry (KOD), IIUM, Jalan Sultan Ahmad Shah, Bandar Indera Mahkota, 25200, Kuantan, Pahang, Malaysia
- Dentistry Programme, PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, BE1410, Brunei
| | - Musliana Mustaffa
- Department of Restorative Dentistry, KOD, IIUM, Jalan Sultan Ahmad Shah, Bandar Indera Mahkota, 25200, Kuantan, Pahang, Malaysia
| |
Collapse
|
19
|
Kumar N, Marée R, Geurts P, Muller M. Recent Advances in Bioimage Analysis Methods for Detecting Skeletal Deformities in Biomedical and Aquaculture Fish Species. Biomolecules 2023; 13:1797. [PMID: 38136667 PMCID: PMC10742266 DOI: 10.3390/biom13121797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Detecting skeletal or bone-related deformities in model and aquaculture fish is vital for numerous biomedical studies. In biomedical research, model fish with bone-related disorders are potential indicators of various chemically induced toxins in their environment or poor dietary conditions. In aquaculture, skeletal deformities are affecting fish health, and economic losses are incurred by fish farmers. This survey paper focuses on showcasing the cutting-edge image analysis tools and techniques based on artificial intelligence that are currently applied in the analysis of bone-related deformities in aquaculture and model fish. These methods and tools play a significant role in improving research by automating various aspects of the analysis. This paper also sheds light on some of the hurdles faced when dealing with high-content bioimages and explores potential solutions to overcome these challenges.
Collapse
Affiliation(s)
- Navdeep Kumar
- Department of Computer Science and Electrical Engineering, Montefiore Institute, University of Liège, 4000 Liège, Belgium; (R.M.); (P.G.)
| | - Raphaël Marée
- Department of Computer Science and Electrical Engineering, Montefiore Institute, University of Liège, 4000 Liège, Belgium; (R.M.); (P.G.)
| | - Pierre Geurts
- Department of Computer Science and Electrical Engineering, Montefiore Institute, University of Liège, 4000 Liège, Belgium; (R.M.); (P.G.)
| | - Marc Muller
- Laboratory for Organogenesis and Regeneration (LOR), GIGA Institute, University of Liège, 4000 Liège, Belgium;
| |
Collapse
|
20
|
Santos N, Oliveira M, Domingues I. Influence of exposure scenario on the sensitivity to caffeine. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:122808-122821. [PMID: 37978123 PMCID: PMC10724325 DOI: 10.1007/s11356-023-30945-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
The chorion acts as a protective barrier, restricting some chemical absorption into the embryo and the surrounding fluids. In this sense, larvae may only have direct contact with some chemicals after dechorionation. This study aimed to evaluate the effects of caffeine (CAF) (0, 13, 20, 44, 67, and 100 mg.L-1) under different exposure scenarios (embryos with chorion or embryos/larvae already hatched) and rank the stage sensitivity. Thus, three scenarios were investigated: from 2 to 120 hours post fertilization (hpf) (5 days of exposure- 5dE), from 72 to 120 hpf (2dE), and from 96 to 120 hpf (1dE). Heart rate (48 hpf) and energy reserves (120 hpf) were measured in the 5dE scenario, and behavior and acetylcholinesterase (AChE) activity were evaluated at 120 hpf in all scenarios (5dE, 2dE, and 1dE). At 120 hpf, some of the fish was transferred to clean medium for a 10 days depuration period (10dPE). Behavior and AChE activity were assessed after this period. In the 5dE scenario, CAF increased heartbeat (13, 20, and 30 mg.L-1) and reduced carbohydrates (67, and 100 mg.L-1), while inhibiting AChE activity (100 mg.L-1) in the 5dE, 2dE, and 1dE scenarios. CAF reduced the total distance moved in the 5dE (67, and 100 mg.L-1), 2dE (20, 30, 44, 67, and 100 mg.L-1), and 1dE fish (67, and 100 mg.L-1) and increased erratic movements. Based on the lowest observed effect concentration (LOEC) for total distance moved (20 mg.L-1) and higher inhibition of AChE activity (100 mg.L-1) (65%), 2dE fish appear to be more sensitive to CAF. After 10dPE, a recovery in behavior was detected in all scenarios (5dE, 2dE, and 1dE). AChE activity remained inhibited in the 2dE scenario while increasing in the 1dE scenario. This study demonstrated that the presence of the chorion is an important factor for the analysis of CAF toxicity. After the loss of the chorion, organisms show greater sensitivity to CAF and can be used to evaluate the toxicity of various substances, including nanomaterials or chemicals with low capacity to cross the chorion. Therefore, the use of hatched embryos in toxicity tests is suggested, as they allow a shorter and less expensive exposure scenario that provides similar outcome as the conventional scenario.
Collapse
Affiliation(s)
- Niedja Santos
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Miguel Oliveira
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Inês Domingues
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
21
|
Zarrabi A, Perrin D, Kavoosi M, Sommer M, Sezen S, Mehrbod P, Bhushan B, Machaj F, Rosik J, Kawalec P, Afifi S, Bolandi SM, Koleini P, Taheri M, Madrakian T, Łos MJ, Lindsey B, Cakir N, Zarepour A, Hushmandi K, Fallah A, Koc B, Khosravi A, Ahmadi M, Logue S, Orive G, Pecic S, Gordon JW, Ghavami S. Rhabdomyosarcoma: Current Therapy, Challenges, and Future Approaches to Treatment Strategies. Cancers (Basel) 2023; 15:5269. [PMID: 37958442 PMCID: PMC10650215 DOI: 10.3390/cancers15215269] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/18/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Rhabdomyosarcoma is a rare cancer arising in skeletal muscle that typically impacts children and young adults. It is a worldwide challenge in child health as treatment outcomes for metastatic and recurrent disease still pose a major concern for both basic and clinical scientists. The treatment strategies for rhabdomyosarcoma include multi-agent chemotherapies after surgical resection with or without ionization radiotherapy. In this comprehensive review, we first provide a detailed clinical understanding of rhabdomyosarcoma including its classification and subtypes, diagnosis, and treatment strategies. Later, we focus on chemotherapy strategies for this childhood sarcoma and discuss the impact of three mechanisms that are involved in the chemotherapy response including apoptosis, macro-autophagy, and the unfolded protein response. Finally, we discuss in vivo mouse and zebrafish models and in vitro three-dimensional bioengineering models of rhabdomyosarcoma to screen future therapeutic approaches and promote muscle regeneration.
Collapse
Affiliation(s)
- Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Türkiye; (A.Z.); (A.Z.)
| | - David Perrin
- Section of Orthopaedic Surgery, Department of Surgery, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; (D.P.); (M.S.)
| | - Mahboubeh Kavoosi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Biotechnology Center, Silesian University of Technology, 8 Krzywousty St., 44-100 Gliwice, Poland;
| | - Micah Sommer
- Section of Orthopaedic Surgery, Department of Surgery, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; (D.P.); (M.S.)
- Section of Physical Medicine and Rehabilitation, Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Serap Sezen
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
| | - Parvaneh Mehrbod
- Department of Influenza and Respiratory Viruses, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Bhavya Bhushan
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Science, McGill University, Montreal, QC H3A 0C7, Canada
| | - Filip Machaj
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Jakub Rosik
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Philip Kawalec
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Section of Neurosurgery, Department of Surgery, University of Manitoba, Health Sciences Centre, Winnipeg, MB R3A 1R9, Canada
| | - Saba Afifi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Seyed Mohammadreza Bolandi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Peiman Koleini
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Mohsen Taheri
- Genetics of Non-Communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran;
| | - Tayyebeh Madrakian
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (T.M.); (M.A.)
| | - Marek J. Łos
- Biotechnology Center, Silesian University of Technology, 8 Krzywousty St., 44-100 Gliwice, Poland;
| | - Benjamin Lindsey
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Nilufer Cakir
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
| | - Atefeh Zarepour
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Türkiye; (A.Z.); (A.Z.)
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran;
| | - Ali Fallah
- Integrated Manufacturing Technologies Research and Application Center, Sabanci University, Tuzla, Istanbul 34956, Türkiye;
| | - Bahattin Koc
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
- Integrated Manufacturing Technologies Research and Application Center, Sabanci University, Tuzla, Istanbul 34956, Türkiye;
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Türkiye
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Türkiye;
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (T.M.); (M.A.)
| | - Susan Logue
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), 01007 Vitoria-Gasteiz, Spain;
- University Institute for Regenerative Medicine and Oral Implantology–UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01007 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, USA;
| | - Joseph W. Gordon
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- College of Nursing, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555 Katowice, Poland
- Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| |
Collapse
|
22
|
Huang M, Zhang Y, Gong Y, Liang Z, Chen X, Ni Y, Pan X, Wu W, Chen J, Huang Z, Sun J. 8-Hydroxyquinoline ruthenium(II) complexes induce ferroptosis in HeLa cells by down-regulating GPX4 and ferritin. J Inorg Biochem 2023; 248:112365. [PMID: 37690267 DOI: 10.1016/j.jinorgbio.2023.112365] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/12/2023]
Abstract
Ruthenium complexes are one of the most promising anticancer drugs triggered extensive research. Here, the synthesis and characterization of two ruthenium(II) polypyridine complexes containing 8-hydroxylquinoline as ligand, [Ru(dip)2(8HQ)]PF6 (Ru1), [Ru(dpq)2(8HQ)]PF6 (Ru2) (8HQ = 8-hydroxylquinoline; dip = 4,7-diphenyl-1,10-phenanthroline; dpq = pyrazino[2,3-f][1,10]phenanthroline) were reported. On the basis of cytotoxicity tests, Ru1 (IC50 = 1.98 ± 0.02 μM) and Ru2 (IC50 = 10.02 ± 0.19 μM) both showed good anticancer activity in a panel of cell lines, especially in HeLa cells. Researches on mechanism indicated that Ru1 and Ru2 acted on mitochondria and nuclei and induced reactive oxygen species (ROS) accumulation, while the morphology of nuclei and cell cycle had no significant change. Western blot assay further proved that GPX4 and Ferritin were down-regulated, which eventually triggered ferroptosis in HeLa cells. In addition, the toxicity test of zebrafish embryos showed that the concentrations of Ru1 and Ru2 below 120 μM and 60 μM were safe and did not have obvious effect on the normal development of zebrafish embryos.
Collapse
Affiliation(s)
- Minying Huang
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yuqing Zhang
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yao Gong
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zhijun Liang
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Xide Chen
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710, China.
| | - Yunxin Ni
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Xinjie Pan
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Wei Wu
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Jiaxi Chen
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Zunnan Huang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Medical University, Dongguan 523808, China
| | - Jing Sun
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China; Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Medical University, Dongguan 523808, China; The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710, China.
| |
Collapse
|
23
|
Wang D, Leng X, Tian Y, Liu J, Zou J, Xie S. Toxic Effects of Koumine on the Early-Life Development Stage of Zebrafish. TOXICS 2023; 11:853. [PMID: 37888703 PMCID: PMC10611223 DOI: 10.3390/toxics11100853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/16/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023]
Abstract
Koumine is one of the most abundant alkaloids found in Gelsemium elegans, and it has a wide range of pharmacological effects including antitumor, anti-inflammatory, analgesic treatment effects, and antianxiety. However, its high toxicity and unclear mechanism of action have greatly limited the medicinal development and use of koumine. We investigated the toxic effects of koumine on the developmental toxicity and behavioral neurotoxicity of zebrafish embryos and larvae. Embryos at 6 h postfertilization (hpf) were exposed to 12.5, 25, 50, 75, and 100 mg/L of koumine until 120 hpf. Koumine affected the hatching and heartbeats of the embryos. The morphological analysis also revealed many abnormalities, such as shortened bodies, yolk sac edemas, tail malformations, and pericardial edemas. To identify the neurotoxicity of koumine, the behavior of the larvae was measured. Koumine at 50 and 100 mg/L affect the escape response. The embryos exhibited uncoordinated muscle contractions along the body axis in response to touch at 36 hpf. More importantly, we found that the neurotoxicity of koumine is mainly caused by influencing the ACh content and the activity of AChE without impairing motor neuron development. A comprehensive analysis shows that a high concentration of koumine has obvious toxic effects on zebrafish, and the safe concentration of koumine for zebrafish should be less than 25 mg/L. These results will be valuable for better understanding the toxicity of koumine and provide new insights into the application of koumine.
Collapse
Affiliation(s)
- Dongjie Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China;
| | - Xinyi Leng
- College of Life Sciences, Wuhan University, Wuhan 430000, China; (X.L.); (J.L.)
| | - Yao Tian
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland;
| | - Jiangdong Liu
- College of Life Sciences, Wuhan University, Wuhan 430000, China; (X.L.); (J.L.)
| | - Jixing Zou
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China;
| | - Shaolin Xie
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China;
| |
Collapse
|
24
|
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: 7] [Impact Index Per Article: 3.5] [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
|
25
|
Samrani LMM, Dumont F, Hallmark N, Bars R, Tinwell H, Pallardy M, Piersma AH. Retinoic acid signaling pathway perturbation impacts mesodermal-tissue development in the zebrafish embryo: Biomarker candidate identification using transcriptomics. Reprod Toxicol 2023; 119:108404. [PMID: 37207909 DOI: 10.1016/j.reprotox.2023.108404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/11/2023] [Accepted: 05/14/2023] [Indexed: 05/21/2023]
Abstract
The zebrafish embryo (ZE) model provides a developmental model well conserved throughout vertebrate embryogenesis, with relevance for early human embryo development. It was employed to search for gene expression biomarkers of compound-induced disruption of mesodermal development. We were particularly interested in the expression of genes related to the retinoic acid signaling pathway (RA-SP), as a major morphogenetic regulating mechanism. We exposed ZE to teratogenic concentrations of valproic acid (VPA) and all-trans retinoic acid (ATRA), using folic acid (FA) as a non-teratogenic control compound shortly after fertilization for 4 h, and performed gene expression analysis by RNA sequencing. We identified 248 genes specifically regulated by both teratogens but not by FA. Further analysis of this gene set revealed 54 GO-terms related to the development of mesodermal tissues, distributed along the paraxial, intermediate, and lateral plate sections of the mesoderm. Gene expression regulation was specific to tissues and was observed for somites, striated muscle, bone, kidney, circulatory system, and blood. Stitch analysis revealed 47 regulated genes related to the RA-SP, which were differentially expressed in the various mesodermal tissues. These genes provide potential molecular biomarkers of mesodermal tissue and organ (mal)formation in the early vertebrate embryo.
Collapse
Affiliation(s)
- Laura M M Samrani
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Université Paris-Saclay, Inflammation, Microbiome and Immunosurveillance, INSERM, Faculté Pharmacie, 91104 Orsay, France; Institute for Risk Assessment Sciences (IRAS), Utrecht University, the Netherlands.
| | | | | | | | | | - Marc Pallardy
- Université Paris-Saclay, Inflammation, Microbiome and Immunosurveillance, INSERM, Faculté Pharmacie, 91104 Orsay, France
| | - Aldert H Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, the Netherlands
| |
Collapse
|
26
|
Samrani LMM, Dumont F, Hallmark N, Bars R, Tinwell H, Pallardy M, Piersma AH. Nervous system development related gene expression regulation in the zebrafish embryo after exposure to valproic acid and retinoic acid: A genome wide approach. Toxicol Lett 2023; 384:96-104. [PMID: 37451652 DOI: 10.1016/j.toxlet.2023.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/04/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
The evaluation of chemical and pharmaceutical safety for humans is moving from animal studies to New Approach Methodologies (NAM), reducing animal use and focusing on mechanism of action, whilst enhancing human relevance. In developmental toxicology, the mechanistic approach is facilitated by the assessment of predictive biomarkers, which allow mechanistic pathways perturbation monitoring at the basis of human hazard assessment. In our search for biomarkers of maldevelopment, we focused on chemically-induced perturbation of the retinoic acid signaling pathway (RA-SP), a major pathway implicated in a plethora of developmental processes. A genome-wide expression screening was performed on zebrafish embryos treated with two teratogens, all-trans retinoic acid (ATRA) and valproic acid (VPA), and a non-teratogen reference compound, folic acid (FA). Each compound was found to have a specific mRNA expression profile with 248 genes commonly dysregulated by both teratogenic compounds but not by FA. These genes were implicated in several developmental processes (e.g., the circulatory and nervous system). Given the prominent response of neurodevelopmental gene sets, and the crucial need to better understand developmental neurotoxicity, our study then focused on nervous system development. We found 62 genes that are potential early neurodevelopmental toxicity biomarker candidates. These results advance NAM-based safety assessment evaluation by highlighting the usefulness of the RA-SP in providing early toxicity biomarker candidates.
Collapse
Affiliation(s)
- Laura M M Samrani
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Université Paris-Saclay, Inflammation, Microbiome and Immunosurveillance, INSERM, Faculté Pharmacie, 91104 Orsay, France; Institute for Risk Assessment Sciences (IRAS), Utrecht University, the Netherlands.
| | | | | | | | | | - Marc Pallardy
- Université Paris-Saclay, Inflammation, Microbiome and Immunosurveillance, INSERM, Faculté Pharmacie, 91104 Orsay, France
| | - Aldert H Piersma
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, the Netherlands
| |
Collapse
|
27
|
Zhou H, Arechavala-Gomeza V, Garanto A. Experimental Model Systems Used in the Preclinical Development of Nucleic Acid Therapeutics. Nucleic Acid Ther 2023; 33:238-247. [PMID: 37145922 PMCID: PMC10457615 DOI: 10.1089/nat.2023.0001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/23/2023] [Indexed: 05/07/2023] Open
Abstract
Preclinical evaluation of nucleic acid therapeutics (NATs) in relevant experimental model systems is essential for NAT drug development. As part of COST Action "DARTER" (Delivery of Antisense RNA ThERapeutics), a network of researchers in the field of RNA therapeutics, we have conducted a survey on the experimental model systems routinely used by our members in preclinical NAT development. The questionnaire focused on both cellular and animal models. Our survey results suggest that skin fibroblast cultures derived from patients is the most commonly used cellular model, while induced pluripotent stem cell-derived models are also highly reported, highlighting the increasing potential of this technology. Splice-switching antisense oligonucleotide is the most frequently investigated RNA molecule, followed by small interfering RNA. Animal models are less prevalent but also widely used among groups in the network, with transgenic mouse models ranking the top. Concerning the research fields represented in our survey, the mostly studied disease area is neuromuscular disorders, followed by neurometabolic diseases and cancers. Brain, skeletal muscle, heart, and liver are the top four tissues of interest reported. We expect that this snapshot of the current preclinical models will facilitate decision making and the share of resources between academics and industry worldwide to facilitate the development of NATs.
Collapse
Affiliation(s)
- Haiyan Zhou
- Genetics and Genomic Medicine Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- NIHR Great Ormond Street Hospital Biomedical Research Center, London, United Kingdom
| | - Virginia Arechavala-Gomeza
- Nucleic Acid Therapeutics for Rare Disorders (NAT-RD), Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Alejandro Garanto
- Department of Pediatrics, Amalia Children's Hospital, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
28
|
Bondue T, Berlingerio SP, van den Heuvel L, Levtchenko E. The Zebrafish Embryo as a Model Organism for Testing mRNA-Based Therapeutics. Int J Mol Sci 2023; 24:11224. [PMID: 37446400 DOI: 10.3390/ijms241311224] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
mRNA-based therapeutics have revolutionized the world of molecular therapy and have proven their potential in the vaccination campaigns for SARS-CoV2 and clinical trials for hereditary disorders. Preclinical studies have mainly focused on in vitro and rodent studies. However, research in rodents is costly and labour intensive, and requires ethical approval for all interventions. Zebrafish embryonic disease models are not always classified as laboratory animals and have been shown to be extremely valuable for high-throughput drug testing. Zebrafish larvae are characterized by their small size, optical transparency and high number of embryos, and are therefore also suited for the study of mRNA-based therapeutics. First, the one-cell stage injection of naked mRNA can be used to assess the effectivity of gene addition in vivo. Second, the intravascular injection in older larvae can be used to assess tissue targeting efficiency of (packaged) mRNA. In this review, we describe how zebrafish can be used as a steppingstone prior to testing mRNA in rodent models. We define the procedures that can be employed for both the one-cell stage and later-stage injections, as well as the appropriate procedures for post-injection follow-up.
Collapse
Affiliation(s)
- Tjessa Bondue
- Department of Development and Regeneration, KU Leuven Campus Gasthuisberg, 3000 Leuven, Belgium
| | | | - Lambertus van den Heuvel
- Department of Development and Regeneration, KU Leuven Campus Gasthuisberg, 3000 Leuven, Belgium
- Department of Pediatric Nephrology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Elena Levtchenko
- Department of Development and Regeneration, KU Leuven Campus Gasthuisberg, 3000 Leuven, Belgium
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
29
|
Bilardo R, Traldi F, Brennan CH, Resmini M. The Role of Crosslinker Content of Positively Charged NIPAM Nanogels on the In Vivo Toxicity in Zebrafish. Pharmaceutics 2023; 15:1900. [PMID: 37514086 PMCID: PMC10383542 DOI: 10.3390/pharmaceutics15071900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/29/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Polymeric nanogels as drug delivery systems offer great advantages, such as high encapsulation capacity and easily tailored formulations; however, data on biocompatibility are still limited. We synthesized N-isopropylacrylamide nanogels, with crosslinker content between 5 and 20 mol%, functionalized with different positively charged co-monomers, and investigated the in vivo toxicity in zebrafish. Our results show that the chemical structure of the basic unit impacts the toxicity profile depending on the degree of ionization and hydrogen bonding capability. When the degree of crosslinking of the polymer was altered, from 5 mol% to 20 mol%, the distribution of the positively charged monomer 2-tert-butylaminoethyl methacrylate was significantly altered, leading to higher surface charges for the more rigid nanogels (20 mol% crosslinker), which resulted in >80% survival rate (48 h, up to 0.5 mg/mL), while the more flexible polymers (5 mol% crosslinker) led to 0% survival rate (48 h, up to 0.5 mg/mL). These data show the importance of tailoring both chemical composition and rigidity of the formulation to minimize toxicity and demonstrate that using surface charge data to guide the design of nanogels for drug delivery may be insufficient.
Collapse
Affiliation(s)
- Roberta Bilardo
- Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Federico Traldi
- Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Caroline H Brennan
- School of Biological and Behavioural Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Marina Resmini
- Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK
| |
Collapse
|
30
|
Haraoka Y, Miyake M, Ishitani T. Zebrafish imaging reveals hidden oncogenic-normal cell communication during primary tumorigenesis. Cell Struct Funct 2023; 48:113-121. [PMID: 37164759 PMCID: PMC10721949 DOI: 10.1247/csf.23026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/08/2023] [Indexed: 05/12/2023] Open
Abstract
Oncogenic mutations drive tumorigenesis, and single cells with oncogenic mutations act as the tumor seeds that gradually evolve into fully transformed tumors. However, oncogenic cell behavior and communication with neighboring cells during primary tumorigenesis remain poorly understood. We used the zebrafish, a small vertebrate model suitable for in vivo cell biology, to address these issues. We describe the cooperative and competitive communication between oncogenic cells and neighboring cells, as revealed by our recent zebrafish imaging studies. Newly generated oncogenic cells are actively eliminated by neighboring cells in healthy epithelia, whereas oncogenic cells cooperate with their neighbors to prime tumorigenesis in unhealthy epithelia via additional mutations or inflammation. In addition, we discuss the potential of zebrafish in vivo imaging to determine the initial steps of human tumorigenesis.Key words: zebrafish, imaging, cell-cell communication, cell competition, EDAC, senescence, primary tumorigenesis.
Collapse
Affiliation(s)
- Yukinari Haraoka
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mai Miyake
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Tohru Ishitani
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
31
|
Li K, Liu B, Wang Z, Li Y, Li H, Wu S, Li Z. Quantitative characterization of zebrafish development based on multiple classifications using Mueller matrix OCT. BIOMEDICAL OPTICS EXPRESS 2023; 14:2889-2904. [PMID: 37342688 PMCID: PMC10278635 DOI: 10.1364/boe.488614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023]
Abstract
Organ development analysis plays an important role in assessing an individual' s growth health. In this study, we present a non-invasive method for the quantitative characterization of zebrafish multiple organs during their growth, utilizing Mueller matrix optical coherence tomography (Mueller matrix OCT) in combination with deep learning. Firstly, Mueller matrix OCT was employed to acquire 3D images of zebrafish during development. Subsequently, a deep learning based U-Net network was applied to segment various anatomical structures, including the body, eyes, spine, yolk sac, and swim bladder of the zebrafish. Following segmentation, the volume of each organ was calculated. Finally, the development and proportional trends of zebrafish embryos and organs from day 1 to day 19 were quantitatively analyzed. The obtained quantitative results revealed that the volume development of the fish body and individual organs exhibited a steady growth trend. Additionally, smaller organs, such as the spine and swim bladder, were successfully quantified during the growth process. Our findings demonstrate that the combination of Mueller matrix OCT and deep learning effectively quantify the development of various organs throughout zebrafish embryonic development. This approach offers a more intuitive and efficient monitoring method for clinical medicine and developmental biology studies.
Collapse
Affiliation(s)
- Ke Li
- Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Provincial Engineering Technology Research Center of Photoelectric Sensing Application, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, Fujian, 350007, China
| | - Bin Liu
- Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Provincial Engineering Technology Research Center of Photoelectric Sensing Application, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, Fujian, 350007, China
| | - Zaifan Wang
- Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Provincial Engineering Technology Research Center of Photoelectric Sensing Application, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, Fujian, 350007, China
| | - Yao Li
- Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Provincial Engineering Technology Research Center of Photoelectric Sensing Application, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, Fujian, 350007, China
| | - Hui Li
- Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Provincial Engineering Technology Research Center of Photoelectric Sensing Application, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, Fujian, 350007, China
| | - Shulian Wu
- Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Provincial Engineering Technology Research Center of Photoelectric Sensing Application, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, Fujian, 350007, China
| | - Zhifang Li
- Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Provincial Engineering Technology Research Center of Photoelectric Sensing Application, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, Fujian, 350007, China
- Bionovel Lab, Guangzhou, Guangdong, 510407, China
| |
Collapse
|
32
|
Schippers P, Rasheed S, Park YM, Risch T, Wagmann L, Hemmer S, Manier SK, Müller R, Herrmann J, Meyer MR. Evaluation of extraction methods for untargeted metabolomic studies for future applications in zebrafish larvae infection models. Sci Rep 2023; 13:7489. [PMID: 37161044 PMCID: PMC10170104 DOI: 10.1038/s41598-023-34593-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/04/2023] [Indexed: 05/11/2023] Open
Abstract
Sample preparation in untargeted metabolomics should allow reproducible extractions of as many molecules as possible. Thus, optimizing sample preparation is crucial. This study compared six different extraction procedures to find the most suitable for extracting zebrafish larvae in the context of an infection model. Two one-phase extractions employing methanol (I) and a single miscible phase of methanol/acetonitrile/water (II) and two two-phase methods using phase separation between chloroform and methanol/water combinations (III and IV) were tested. Additional bead homogenization was used for methods III and IV (III_B and IV_B). Nine internal standards and 59 molecules of interest (MoInt) related to mycobacterial infection were used for method evaluation. Two-phase methods (III and IV) led to a lower feature count, higher peak areas of MoInt, especially amino acids, and higher coefficients of variation in comparison to one-phase extractions. Adding bead homogenization increased feature count, peak areas, and CVs. Extraction I showed higher peak areas and lower CVs than extraction II, thus being the most suited one-phase method. Extraction III and IV showed similar results, with III being easier to execute and less prone to imprecisions. Thus, for future applications in zebrafish larvae metabolomics and infection models, extractions I and III might be chosen.
Collapse
Affiliation(s)
- Philip Schippers
- Department of Experimental and Clinical Toxicology, Center for Molecular Signaling (PZMS), Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, 66421, Homburg, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken, Germany
| | - Sari Rasheed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover, Braunschweig, Germany
| | - Yu Mi Park
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken, Germany
| | - Timo Risch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover, Braunschweig, Germany
| | - Lea Wagmann
- Department of Experimental and Clinical Toxicology, Center for Molecular Signaling (PZMS), Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, 66421, Homburg, Germany
| | - Selina Hemmer
- Department of Experimental and Clinical Toxicology, Center for Molecular Signaling (PZMS), Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, 66421, Homburg, Germany
| | - Sascha K Manier
- Department of Experimental and Clinical Toxicology, Center for Molecular Signaling (PZMS), Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, 66421, Homburg, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover, Braunschweig, Germany
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover, Braunschweig, Germany
| | - Markus R Meyer
- Department of Experimental and Clinical Toxicology, Center for Molecular Signaling (PZMS), Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, 66421, Homburg, Germany.
| |
Collapse
|
33
|
Adhish M, Manjubala I. Effectiveness of zebrafish models in understanding human diseases-A review of models. Heliyon 2023; 9:e14557. [PMID: 36950605 PMCID: PMC10025926 DOI: 10.1016/j.heliyon.2023.e14557] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/01/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Understanding the detailed mechanism behind every human disease, disorder, defect, and deficiency is a daunting task concerning the clinical diagnostic tools for patients. Hence, a closely resembling living or simulated model is of paramount interest for the development and testing of a probable novel drug for rectifying the conditions pertaining to the various ailments. The animal model that can be easily genetically manipulated to suit the study of the therapeutic motive is an indispensable asset and within the last few decades, the zebrafish models have proven their effectiveness by becoming such potent human disease models with their use being extended to various avenues of research to understand the underlying mechanisms of the diseases. As zebrafish are explored as model animals in understanding the molecular basis and genetics of many diseases owing to the 70% genetic homology between the human and zebrafish genes; new and fascinating facts about the diseases are being surfaced, establishing it as a very powerful tool for upcoming research. These prospective research areas can be explored in the near future using zebrafish as a model. In this review, the effectiveness of the zebrafish as an animal model against several human diseases such as osteoporosis, atrial fibrillation, Noonan syndrome, leukemia, autism spectrum disorders, etc. has been discussed.
Collapse
Affiliation(s)
- Mazumder Adhish
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, India
| | - I. Manjubala
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, India
| |
Collapse
|
34
|
Balitaan JNI, Luo WJ, Su YW, Yu CY, Wu TY, Chang CA, Jia HW, Lin SR, Hsiao CD, Yeh JM. Healing Wounds Efficiently with Biomimetic Soft Matter: Injectable Self-Healing Neutral Glycol Chitosan/Dibenzaldehyde-Terminated Poly(ethylene glycol) Hydrogel with Inherent Antibacterial Properties. ACS APPLIED BIO MATERIALS 2023; 6:552-565. [PMID: 36759183 DOI: 10.1021/acsabm.2c00859] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The high prevalence of acquiring skin wounds, along with the emergence of antibiotic-resistant strains that lead to infections, impose a threat to the physical, mental, and socioeconomic health of society. Among the wide array of wound dressings developed, hydrogels are regarded as a biomimetic soft matter of choice owing to their ability to provide a moist environment ideal for healing. Herein, neutral glycol chitosan (GC) was cross-linked via imine bonds with varying concentrations of dibenzaldehyde-terminated polyethylene glycol (DP) to give glycol chitosan/dibenzaldehyde-terminated polyethylene glycol hydrogels (GC/DP). These dynamic Schiff base linkages (absorption peak at 1638 cm-1) within the hydrogel structure endowed their ability to recover from damage as characterized by high-low strain exposure in continuous step strain rheology. Along with their good injectability and biodegradability, the hydrogels exhibited remarkable inhibition against E. coli, P. aeruginosa, and S. aureus. GC/DP hydrogels demonstrated high LC50 values in vivo using zebrafish embryos as a model system due to their relative biocompatibility and a remarkable 93.4 ± 0.88% wound contraction at 30-dpw against 49.1 ± 3.40% of the control. To the best of our knowledge, this is the first study that developed injectable glycol chitosan/dibenzaldehyde-terminated polyethylene glycol self-healing hydrogels for application in wound healing with intrinsic bacteriostatic properties against the three bacteria.
Collapse
|
35
|
Zebrafish, a biological model for pharmaceutical research for the management of anxiety. Mol Biol Rep 2023; 50:3863-3872. [PMID: 36757551 DOI: 10.1007/s11033-023-08263-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/10/2023] [Indexed: 02/10/2023]
Abstract
The zebrafish (Danio rerio) is a valuable animal model rapidly becoming more commonly used in pharmaceutical studies. Due to its low-cost maintenance and high breeding potential, the zebrafish is a suitable substitute for most adult rodents (mice and rats) in neuroscience research. It is widely used in various anxiety models. This species has been used to develop a conceptual framework for anxiety behavior studies with broad applications in the laboratory, including the study of herbal and chemical drugs. This review discusses the latest studies of anxiety-related behavior in the zebrafish model.
Collapse
|
36
|
Dal NJK, Schäfer G, Thompson AM, Schmitt S, Redinger N, Alonso-Rodriguez N, Johann K, Ojong J, Wohlmann J, Best A, Koynov K, Zentel R, Schaible UE, Griffiths G, Barz M, Fenaroli F. Π-Π interactions stabilize PeptoMicelle-based formulations of Pretomanid derivatives leading to promising therapy against tuberculosis in zebrafish and mouse models. J Control Release 2023; 354:851-868. [PMID: 36681282 DOI: 10.1016/j.jconrel.2023.01.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/15/2022] [Accepted: 01/14/2023] [Indexed: 01/23/2023]
Abstract
Tuberculosis is the deadliest bacterial disease globally, threatening the lives of millions every year. New antibiotic therapies that can shorten the duration of treatment, improve cure rates, and impede the development of drug resistance are desperately needed. Here, we used polymeric micelles to encapsulate four second-generation derivatives of the antitubercular drug pretomanid that had previously displayed much better in vivo activity against Mycobacterium tuberculosis than pretomanid itself. Because these compounds were relatively hydrophobic and had limited bioavailability, we expected that their micellar formulations would overcome these limitations, reduce toxicities, and improve therapeutic outcomes. The polymeric micelles were based on polypept(o)ides (PeptoMicelles) and were stabilized in their hydrophobic core by π-π interactions, allowing the efficient encapsulation of aromatic pretomanid derivatives. The stability of these π-π-stabilized PeptoMicelles was demonstrated in water, blood plasma, and lung surfactant by fluorescence cross-correlation spectroscopy and was further supported by prolonged circulation times of several days in the vasculature of zebrafish larvae. The most efficacious PeptoMicelle formulation tested in the zebrafish larvae infection model almost completely eradicated the bacteria at non-toxic doses. This lead formulation was further assessed against Mycobacterium tuberculosis in the susceptible C3HeB/FeJ mouse model, which develops human-like necrotic granulomas. Following intravenous administration, the drug-loaded PeptoMicelles significantly reduced bacterial burden and inflammatory responses in the lungs and spleens of infected mice.
Collapse
Affiliation(s)
- Nils-Jørgen K Dal
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Gabriela Schäfer
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany; Leiden Academic Center for Drug Research (LACDR), Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC, Leiden, the Netherlands
| | - Andrew M Thompson
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Sascha Schmitt
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Natalja Redinger
- Forschungszentrum Borstel, Leibniz Lungenzentrum, Program Area Infections, Div. Cellular Microbiology; University of Lübeck, Immunochemistry and Biochemical Microbiology, & German Center for Infection Research, partner site Hamburg-Lübeck - Borstel - Riems, 23845 Borstel, Germany
| | | | - Kerstin Johann
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Jessica Ojong
- Forschungszentrum Borstel, Leibniz Lungenzentrum, Program Area Infections, Div. Cellular Microbiology; University of Lübeck, Immunochemistry and Biochemical Microbiology, & German Center for Infection Research, partner site Hamburg-Lübeck - Borstel - Riems, 23845 Borstel, Germany
| | - Jens Wohlmann
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Andreas Best
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Kaloian Koynov
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Rudolf Zentel
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Ulrich E Schaible
- Forschungszentrum Borstel, Leibniz Lungenzentrum, Program Area Infections, Div. Cellular Microbiology; University of Lübeck, Immunochemistry and Biochemical Microbiology, & German Center for Infection Research, partner site Hamburg-Lübeck - Borstel - Riems, 23845 Borstel, Germany
| | - Gareth Griffiths
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Matthias Barz
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany; Leiden Academic Center for Drug Research (LACDR), Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC, Leiden, the Netherlands.
| | - Federico Fenaroli
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway; Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4021 Stavanger, Norway.
| |
Collapse
|
37
|
Sathiyanarayanan A, Yashwanth BS, Pinto N, Thakuria D, Chaudhari A, Gireesh Babu P, Goswami M. Establishment and characterization of a new fibroblast-like cell line from the skin of a vertebrate model, zebrafish (Danio rerio). Mol Biol Rep 2023; 50:19-29. [PMID: 36289143 DOI: 10.1007/s11033-022-08009-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/05/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND The available fully sequenced genome and genetic similarities compared to humans make zebrafish a prominent in vitro vertebrate model for drug discovery & screening, toxicology, and radiation biology. Zebrafish also possess well developed immune systems which is ideal for studying infectious diseases. Fish skin confers immunity by serving as a physical barrier against the invading pathogens in the aquatic habitat. Therefore in vitro models from the skin tissue of zebrafish help to study the physiology, functional genes in vitro, wound healing, and pathogenicity of microbes. Hence the study aimed to develop and characterize a skin cell line from the wild-type zebrafish Danio rerio. METHODS AND RESULTS A novel cell line designated as DRS (D. rerio skin) was established and characterized from the skin tissue of wild-type zebrafish, D. rerio, by the explant technique. The cells thrived well in the Leibovitz's -15 medium supplemented with 15% FBS and routinely passaged at regular intervals. The DRS cells mainly feature fibroblast-like morphology. The culture conditions of the cells were determined by incubating the cells at varying concentrations of FBS and temperature; the optimum was 15% FBS and 28 °C, respectively. Cells were cryopreserved and revived with 70-75% viability at different passage levels. Two extracellular products from bacterial species Aeromonas hydrophila and Edwardsiella tarda were tested and found toxic to the DRS cells. Mitochondrial genes, namely COI and 16S rRNA PCR amplification and partial sequencing authenticated the species of origin of cells. The modal diploid (2n) chromosome number of the cells was 50. The cell line DRS was found to be free from mycoplasma. The cells were transfected with pMaxGFP plasmid and tested positive for green fluorescence at 24-48 h post-transfection. CONCLUSION The findings from this study thus confirm the usefulness of the developed cell line in bacterial susceptibility and transgene expression studies.
Collapse
Affiliation(s)
- Arjunan Sathiyanarayanan
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Panch Marg, Off Yari Road, Versova, Andheri West, Mumbai, Maharashtra, 400061, India
| | - B S Yashwanth
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Panch Marg, Off Yari Road, Versova, Andheri West, Mumbai, Maharashtra, 400061, India
| | - Nevil Pinto
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Panch Marg, Off Yari Road, Versova, Andheri West, Mumbai, Maharashtra, 400061, India
| | - Dimpal Thakuria
- ICAR-Directorate of Coldwater Fisheries Research, Anusandhan Bhawan, Industrial Area, Bhimtal, 263136, India
| | - Aparna Chaudhari
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Panch Marg, Off Yari Road, Versova, Andheri West, Mumbai, Maharashtra, 400061, India
| | - P Gireesh Babu
- ICAR-National Research Centre on Meat, Chengicherla, Boduppal Post, Hyderabad, 500092, India
| | - Mukunda Goswami
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Panch Marg, Off Yari Road, Versova, Andheri West, Mumbai, Maharashtra, 400061, India.
| |
Collapse
|
38
|
Samrani LMM, Pennings JLA, Hallmark N, Bars R, Tinwell H, Pallardy M, Piersma AH. Dynamic regulation of gene expression and morphogenesis in the zebrafish embryo test after exposure to all-trans retinoic acid. Reprod Toxicol 2023; 115:8-16. [PMID: 36375755 DOI: 10.1016/j.reprotox.2022.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/13/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
Abstract
The zebrafish embryotoxicity test (ZET) is widely used in developmental toxicology. The analysis of gene expression regulation in ZET after chemical exposure provides mechanistic information about the effects of chemicals on morphogenesis in the test. The gene expression response magnitude has been shown to change with exposure duration. The objective of this work is to study the effect of the exposure duration on the magnitude of gene expression changes in the all-trans retinoic acid (ATRA) signaling pathway in the ZET. Retinoic acid regulation is a key driver of morphogenesis and is therefore employed here as an indicator for the regulation of developmental genes. A teratogenic concentration of 7.5 nM of ATRA was given at 3 hrs post fertilization (hpf) for a range of exposure durations until 120 hrs of development. The expression of a selection of genes related to ATRA signaling and downstream developmental genes was determined. The highest magnitudes of gene expression regulation were observed after 2-24 hrs exposure with an optimal response after 4 hrs. Longer exposures showed a decrease in the gene expression response, although continued exposure to 120 hpf caused malformations and lethality. This study shows that assessment of gene expression regulation at early time points after the onset of exposure in the ZET may be optimal for the prediction of developmental toxicity. We believe these results could help optimize sensitivity in future studies with ZET.
Collapse
Affiliation(s)
- Laura M M Samrani
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Université Paris-Saclay, Inflammation, Microbiome and Immunosurveillance, INSERM, Faculté Pharmacie, Châtenay-Malabry 92296, France; Institute for Risk Assessment Sciences (IRAS), Utrecht University, the Netherlands.
| | - Jeroen L A Pennings
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | | | | | | | - Marc Pallardy
- Université Paris-Saclay, Inflammation, Microbiome and Immunosurveillance, INSERM, Faculté Pharmacie, Châtenay-Malabry 92296, France
| | - Aldert H Piersma
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, the Netherlands
| |
Collapse
|
39
|
Alffenaar JWC, de Steenwinkel JEM, Diacon AH, Simonsson USH, Srivastava S, Wicha SG. Pharmacokinetics and pharmacodynamics of anti-tuberculosis drugs: An evaluation of in vitro, in vivo methodologies and human studies. Front Pharmacol 2022; 13:1063453. [PMID: 36569287 PMCID: PMC9780293 DOI: 10.3389/fphar.2022.1063453] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
There has been an increased interest in pharmacokinetics and pharmacodynamics (PKPD) of anti-tuberculosis drugs. A better understanding of the relationship between drug exposure, antimicrobial kill and acquired drug resistance is essential not only to optimize current treatment regimens but also to design appropriately dosed regimens with new anti-tuberculosis drugs. Although the interest in PKPD has resulted in an increased number of studies, the actual bench-to-bedside translation is somewhat limited. One of the reasons could be differences in methodologies and outcome assessments that makes it difficult to compare the studies. In this paper we summarize most relevant in vitro, in vivo, in silico and human PKPD studies performed to optimize the drug dose and regimens for treatment of tuberculosis. The in vitro assessment focuses on MIC determination, static time-kill kinetics, and dynamic hollow fibre infection models to investigate acquisition of resistance and killing of Mycobacterium tuberculosis populations in various metabolic states. The in vivo assessment focuses on the various animal models, routes of infection, PK at the site of infection, PD read-outs, biomarkers and differences in treatment outcome evaluation (relapse and death). For human PKPD we focus on early bactericidal activity studies and inclusion of PK and therapeutic drug monitoring in clinical trials. Modelling and simulation approaches that are used to evaluate and link the different data types will be discussed. We also describe the concept of different studies, study design, importance of uniform reporting including microbiological and clinical outcome assessments, and modelling approaches. We aim to encourage researchers to consider methods of assessing and reporting PKPD of anti-tuberculosis drugs when designing studies. This will improve appropriate comparison between studies and accelerate the progress in the field.
Collapse
Affiliation(s)
- Jan-Willem C. Alffenaar
- Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, Australia,School of Pharmacy, The University of Sydney Faculty of Medicine and Health, Sydney, NSW, Australia,Westmead Hospital, Sydney, NSW, Australia,*Correspondence: Jan-Willem C. Alffenaar,
| | | | | | | | - Shashikant Srivastava
- Department of Pulmonary Immunology, University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Sebastian G. Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| |
Collapse
|
40
|
Chen Y, Wisner AS, Schiefer IT, Williams FE, Hall FS. Methamphetamine-induced lethal toxicity in zebrafish larvae. Psychopharmacology (Berl) 2022; 239:3833-3846. [PMID: 36269378 PMCID: PMC10593407 DOI: 10.1007/s00213-022-06252-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 09/27/2022] [Indexed: 10/24/2022]
Abstract
RATIONALE The use of novel psychoactive substances has been steadily increasing in recent years. Given the rapid emergence of new substances and their constantly changing chemical structure, it is necessary to develop an efficient and expeditious approach to examine the mechanisms underlying their pharmacological and toxicological effects. Zebrafish (Danio rerio) have become a popular experimental subject for drug screening due to their amenability to high-throughput approaches. OBJECTIVES In this study, we used methamphetamine (METH) as an exemplary psychoactive substance to investigate its acute toxicity and possible underlying mechanisms in 5-day post-fertilization (5 dpf) zebrafish larvae. METHODS Lethality and toxicity of different concentrations of METH were examined in 5-dpf zebrafish larvae using a 96-well plate format. RESULTS METH induced lethality in zebrafish larvae in a dose-dependent manner, which was associated with initial sympathomimetic activation, followed by cardiotoxicity. This was evidenced by significant heart rate increases at low doses, followed by decreased cardiac function at high doses and later time points. Levels of ammonia in the excreted water were increased but decreased internally. There was also evidence of seizures. Co-administration of the glutamate AMPA receptor antagonist GYKI-52466 and the dopamine D2 receptor antagonist raclopride significantly attenuated METH-induced lethality, suggesting that this lethality may be mediated synergistically or independently by glutamatergic and dopaminergic systems. CONCLUSIONS These experiments provide a baseline for the study of the toxicity of related amphetamine compounds in 5-dpf zebrafish as well as a new high-throughput approach for investigating the toxicities of rapidly emerging new psychoactive substances.
Collapse
Affiliation(s)
- Yu Chen
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, 3000 Arlington Ave., MS 1015, Toledo, OH, 43614-2598, USA
- College of Pharmacy, The University of Tennessee Health Science Center, 881 Madison Ave Room 610, Memphis, TN, 38163, USA
| | - Alexander S Wisner
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, 3000 Arlington Ave., MS 1015, Toledo, OH, 43614-2598, USA
| | - Isaac T Schiefer
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
- Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Frederick E Williams
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, 3000 Arlington Ave., MS 1015, Toledo, OH, 43614-2598, USA
| | - F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, 3000 Arlington Ave., MS 1015, Toledo, OH, 43614-2598, USA.
| |
Collapse
|
41
|
Dubale NM, Kapron CM, West SL. Commentary: Zebrafish as a Model for Osteoporosis-An Approach to Accelerating Progress in Drug and Exercise-Based Treatment. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:15866. [PMID: 36497941 PMCID: PMC9739463 DOI: 10.3390/ijerph192315866] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 06/17/2023]
Abstract
Osteoporosis (OP) is a degenerative disease characterized by reduced bone strength and increased fracture risk. As the global population continues to age, the prevalence and economic burden of osteoporosis can be expected to rise substantially, but there remain various gaps in the field of OP care. For instance, there is a lack of anti-fracture drugs with proven long-term efficacy. Likewise, though exercise remains widely recommended in OP prevention and management, data regarding the safety and efficacy for patients after vertebral fracture remain limited. This lack of evidence may be due to the cost and inherent difficulties associated with exercise-based OP research. Thus, the current research landscape highlights the need for novel research strategies that accelerate OP drug discovery and allow for the low-cost study of exercise interventions. Here, we outline an example of one strategy, the use of zebrafish, which has emerged as a potential model for the discovery of anti-osteoporosis therapeutics and study of exercise interventions. The strengths, limitations, and potential applications of zebrafish in OP research will be outlined.
Collapse
Affiliation(s)
- Natnaiel M. Dubale
- Department of Biology, Trent University, Peterborough, ON K9L 0G2, Canada
| | - Carolyn M. Kapron
- Department of Biology, Trent University, Peterborough, ON K9L 0G2, Canada
| | - Sarah L. West
- Department of Biology, Trent University, Peterborough, ON K9L 0G2, Canada
- Department of Kinesiology, Trent University, Peterborough, ON K9L 0G2, Canada
- Trent/Fleming School of Nursing, Trent University, Peterborough, ON K9L 0G2, Canada
| |
Collapse
|
42
|
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: 2] [Impact Index Per Article: 0.7] [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
|
43
|
Machikhin A, Huang CC, Khokhlov D, Galanova V, Burlakov A. Single-shot Mueller-matrix imaging of zebrafish tissues: In vivo analysis of developmental and pathological features. JOURNAL OF BIOPHOTONICS 2022; 15:e202200088. [PMID: 35582886 DOI: 10.1002/jbio.202200088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/05/2022] [Accepted: 05/14/2022] [Indexed: 06/15/2023]
Abstract
Zebrafish is a well-established animal model for developmental and disease studies. Its optical transparency at early developmental stages allows in vivo tissues visualization. Interaction of polarized light with these tissues provides information on their structure and properties. This approach is effective for muscle tissue analysis due to its birefringence. To enable real-time Mueller-matrix characterization of unanesthetized fish, we assembled a microscope for single-shot Mueller-matrix imaging. First, we performed a continuous observation of 48 species within the period of 2 to 96 hpf and measured temporal dependencies of the polarization features in different tissues. These measurements show that hatching was accompanied by a sharp change in the angle and degree of linearly polarized light after interaction with muscles. Second, we analyzed nine species with skeletal disorders and demonstrated that the spatial distribution of light depolarization features clearly indicated them. Obtained results demonstrated that real-time Mueller-matrix imaging is a powerful tool for label-free monitoring zebrafish embryos.
Collapse
Affiliation(s)
- Alexander Machikhin
- Laboratory of Acousto-optical Spectroscopy, Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
| | - Chih-Chung Huang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Demid Khokhlov
- Laboratory of Acousto-optical Spectroscopy, Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
| | - Victoria Galanova
- Laboratory of Acousto-optical Spectroscopy, Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
- Department of Laser and Opto-Electronic Systems, Bauman Moscow State Technical University, Moscow, Russia
| | - Alexander Burlakov
- Laboratory of Acousto-optical Spectroscopy, Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
- Department of Ichthyology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
44
|
Demery-Poulos C, Chambers JM. Identification, conservation, and expression of tiered pharmacogenes in zebrafish. PLoS One 2022; 17:e0273582. [PMID: 36040978 PMCID: PMC9426904 DOI: 10.1371/journal.pone.0273582] [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: 05/10/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
The number of adverse drug events in the United States is critically high, with annual rates exceeding 1 million cases over the last nine years. One cause of adverse drug events is the underlying genetic variation that can alter drug responses. Pharmacogenomics is a growing field that seeks to better understand the relationship between a patient’s genetics and drug efficacy. Currently, pharmacogenomics relies largely on human trials, as there is not a well-developed animal model for studying preventative measures and alternative treatments. Here, we analyzed pharmacogene expression at two developmental time points in zebrafish to demonstrate the potential of using this model organism for high-throughput pharmacogenomics research. We found that 76% of tiered human pharmacogenes have a zebrafish ortholog, and of these, many have highly conserved amino acid sequences. Additional gene ontology analysis was used to classify pharmacogenes and identify candidate pathways for future modeling in zebrafish. As precision medicine burgeons, adopting a high-throughput in vivo model such as the zebrafish could greatly increase our understanding of the molecular pathology underlying adverse drug events.
Collapse
Affiliation(s)
- Catherine Demery-Poulos
- Department of Pharmaceutical Sciences, College of Pharmacy, Natural and Health Sciences, Manchester University, Fort Wayne, Indiana, United States
| | - Joseph M. Chambers
- Department of Pharmaceutical Sciences, College of Pharmacy, Natural and Health Sciences, Manchester University, Fort Wayne, Indiana, United States
- * E-mail:
| |
Collapse
|
45
|
Li Y, Ren B, Zhao T, Chen H, Zhao Y, Liang H, Liang H. Enantioselective toxic effects of mefentrifluconazole in the early life stage of zebrafish (Danio rerio). ENVIRONMENTAL TOXICOLOGY 2022; 37:1662-1674. [PMID: 35297557 DOI: 10.1002/tox.23515] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/22/2022] [Accepted: 03/06/2022] [Indexed: 06/14/2023]
Abstract
The research on the enantioselective toxic effects of chiral pesticides on non-target aquatic organisms has attracted more and more attention. This study investigated the enantioselective toxic effects of mefentrifluconazole (MFZ) on acute toxicity, developmental toxicity, locomotor behaviors, and the mRNA relative expression levels of genes related to neurodevelopment and cardiac development in zebrafish embryos or larvae. The 96-h lethal concentration 50 (LC50 ) values (exposed to racemate and enantiomers of MFZ, that is, rac-MFZ/(-)-MFZ/(+)-MFZ) were 1.010, 1.552, and 0.753 mg/L for embryo, and 0.753, 1.187, and 0.553 mg/L for larvae. The rac-MFZ/(-)-MFZ/(+)-MFZ can affect the heart development of zebrafish embryos, accompanied by heart rate inhibition, yolk sac deformities, pericardial deformities, and down-regulation of genes related to cardiotoxicity in larvae in an enantioselective manner. Moreover, the rac-MFZ/(-)-MFZ/(+)-MFZ also can affect the neural development of zebrafish embryos, accompanied by autonomic movement inhibition, swimming speed and swimming distance abnormalities, and down-regulation of genes related to neurotoxicity in larvae in an enantioselective manner. For all toxicity endpoints, the effect of the (+)-MFZ to early-staged zebrafish were significantly greater than that of (-)-MFZ. These results will help distinguishing the difference of MFZ enantiomers to zebrafish, and provide scientific reference for improving the risk assessment of chiral pesticides MFZ.
Collapse
Affiliation(s)
- Yanhong Li
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, China
| | - Bo Ren
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, China
| | - Tingting Zhao
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, China
| | - Haiyue Chen
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, China
| | - Yuexing Zhao
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, China
| | - Hanlin Liang
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, China
| | - Hongwu Liang
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, China
| |
Collapse
|
46
|
Varela T, Varela D, Martins G, Conceição N, Cancela ML. Cdkl5 mutant zebrafish shows skeletal and neuronal alterations mimicking human CDKL5 deficiency disorder. Sci Rep 2022; 12:9325. [PMID: 35665761 PMCID: PMC9167277 DOI: 10.1038/s41598-022-13364-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/12/2022] [Indexed: 12/17/2022] Open
Abstract
CDKL5 deficiency disorder (CDD) is a rare neurodevelopmental condition characterized primarily by seizures and impairment of cognitive and motor skills. Additional phenotypes include microcephaly, dysmorphic facial features, and scoliosis. Mutations in cyclin-dependent kinase-like 5 (CDKL5) gene, encoding a kinase essential for normal brain development and function, are responsible for CDD. Zebrafish is an accepted biomedical model for the study of several genetic diseases and has many advantages over other models. Therefore, this work aimed to characterize the phenotypic, behavioral, and molecular consequences of the Cdkl5 protein disruption in a cdkl5 mutant zebrafish line (sa21938). cdkl5sa21938 mutants displayed a reduced head size, suggesting microcephaly, a feature frequently observed in CDD individuals. Double staining revealed shorter craniofacial cartilage structures and decrease bone mineralization in cdkl5 homozygous zebrafish indicating an abnormal craniofacial cartilage development and impaired skeletal development. Motor behavior analysis showed that cdkl5sa21938 embryos had less frequency of double coiling suggesting impaired glutamatergic neurotransmission. Locomotor behavior analysis revealed that homozygous embryos swim shorter distances, indicative of impaired motor activity which is one of the main traits of CCD. Although no apparent spontaneous seizures were observed in these models, upon treatment with pentylenetetrazole, seizure behavior and an increase in the distance travelled were observed. Quantitative PCR showed that neuronal markers, including glutamatergic genes were dysregulated in cdkl5sa21938 mutant embryos. In conclusion, homozygous cdkl5sa21938 zebrafish mimic several characteristics of CDD, thus validating them as a suitable animal model to better understand the physiopathology of this disorder.
Collapse
Affiliation(s)
- Tatiana Varela
- Centre of Marine Sciences, University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal
| | - Débora Varela
- Centre of Marine Sciences, University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal
| | - Gil Martins
- Centre of Marine Sciences, University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal
| | - Natércia Conceição
- Centre of Marine Sciences, University of Algarve, Faro, Portugal.
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal.
- Algarve Biomedical Center, University of Algarve, Faro, Portugal.
| | - M Leonor Cancela
- Centre of Marine Sciences, University of Algarve, Faro, Portugal.
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal.
- Algarve Biomedical Center, University of Algarve, Faro, Portugal.
| |
Collapse
|
47
|
Caioni G, Cimini A, Benedetti E. Food Contamination: An Unexplored Possible Link between Dietary Habits and Parkinson’s Disease. Nutrients 2022; 14:nu14071467. [PMID: 35406080 PMCID: PMC9003245 DOI: 10.3390/nu14071467] [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: 02/23/2022] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 12/10/2022] Open
Abstract
Importance of a healthy lifestyle in maintaining the population’s well-being and health, especially in terms of balanced nutrition, is well known. Food choice of and dieting habits could impact disease management, which is especially true for Parkinson’s disease (PD). However, nowadays, it is not that simple to maintain a balance in nutrition, and the idea of a healthy diet tends to fade as the consequence of a western lifestyle. This should not only be dealt with in the context of food choice, but also from an environmental point of view. What we put into our bodies is strictly related to the quality of ecosystems we live in. For these reasons, attention should be directed to all the pollutants, which in many cases, we unknowingly ingest. It will be necessary to explore the interaction between food and environment, since human activity also influences the raw materials destined for consumption. This awareness can be achieved by means of an innovative scientific approach, which involves the use of new models, in order to overcome the traditional scientific investigations included in the study of Parkinson’s disease.
Collapse
Affiliation(s)
- Giulia Caioni
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.C.)
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.C.)
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.C.)
- Correspondence: ; Tel.: +39-086-243-3267
| |
Collapse
|
48
|
Li C, Wang M, Fu T, Li Z, Chen Y, He T, Feng D, Wang Z, Fan Q, Chen M, Zhang H, Lin R, Zhao C. Lipidomics Indicates the Hepatotoxicity Effects of EtOAc Extract of Rhizoma Paridis. Front Pharmacol 2022; 13:799512. [PMID: 35211012 PMCID: PMC8861452 DOI: 10.3389/fphar.2022.799512] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/17/2022] [Indexed: 12/18/2022] Open
Abstract
Rhizoma Paridis is a traditional Chinese medicine commonly used in the clinical treatment of gynecological diseases. Previous studies have shown that aqueous extracts of Rhizoma Paridis exhibit some hepatotoxicity to hepatocytes. Here, using lipidomics analysis, we investigated the potential hepatotoxicity of Rhizoma Paridis and its possible mechanism. The hepatic damaging of different solvent extracts of Rhizoma Paridis on zebrafish larvae were determined by a combination of mortality dose, biochemical, morphological, and functional tests. We found that ethyl acetate extracts (AcOEtE) were the most toxic fraction. Notably, lipidomic responsible for the pharmacological effects of AcOEtE were investigated by Q-Exactive HF-X mass spectrometer (Thermo Scientific high-resolution) coupled in tandem with a UHPLC system. Approximately 1958 unique spectral features were detected, of which 325 were identified as unique lipid species. Among these lipid species, phosphatidylethanolamine cardiolipin Ceramide (Cer), lysophosphatidylinositol sphingosine (Sph), etc., were significantly upregulated in the treated group. Pathway analysis indicates that Rhizoma Paridis may cause liver damage via interfering with the glycerophospholipid metabolism. Collectively, this study has revealed previously uncharacterized lipid metabolic disorder involving lipid synthesis, metabolism, and transport that functionally determines hepatic fibrosis procession.
Collapse
Affiliation(s)
- Chaofeng Li
- Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Mingshuang Wang
- Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Tingting Fu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiqi Li
- Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Chen
- Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Tao He
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Dan Feng
- Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhaoyi Wang
- Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Qiqi Fan
- Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Meilin Chen
- Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Honggui Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ruichao Lin
- Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chongjun Zhao
- Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
49
|
Sudhakaran G, Guru A, Hari Deva Muthu B, Murugan R, Arshad A, Arockiaraj J. Evidence-based hormonal, mutational, and endocrine-disrupting chemical-induced zebrafish as an alternative model to study PCOS condition similar to mammalian PCOS model. Life Sci 2022; 291:120276. [PMID: 34990650 DOI: 10.1016/j.lfs.2021.120276] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/15/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022]
Abstract
Polycystic ovarian syndrome (PCOS) causes swollen ovaries in women at reproductive age due to hormonal disorder with small cysts on the outer edges. The cause of the disorder is still yet to be found. Multiple factors have increased PCOS prevalence, hyperandrogenism, oxidative stress, inflammation, and insulin resistance. Various animal PCOS models have been developed to imitate the pathophysiology of PCOS in humans. Zebrafish is one of the most versatile animal experimental models because of the transparency of the embryos, small size, and rapid growth. The zebrafish similarity to higher vertebrates made it a useful non-mammalian model for PCOS drug testing and screening. This review provides an insight into the usage of zebrafish, a non-mammalian model for PCOS, as an opportunity for evaluating future initiatives in such a research domain.
Collapse
Affiliation(s)
- Gokul Sudhakaran
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - Ajay Guru
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - B Hari Deva Muthu
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - Raghul Murugan
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - Aziz Arshad
- International Institute of Aquaculture and Aquatic Sciences (I-AQUAS), Universiti Putra Malaysia, 71050 Port Dickson, Negeri Sembilan, Malaysia
| | - Jesu Arockiaraj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India; Foundation for Aquaculture Innovations and Technology Transfer (FAITT), Thoraipakkam, Chennai 600 097, Tamil Nadu, India.
| |
Collapse
|
50
|
Age-dependent effects of embryonic ethanol exposure on anxiety-like behaviours in young zebrafish: A genotype comparison study. Pharmacol Biochem Behav 2022; 214:173342. [DOI: 10.1016/j.pbb.2022.173342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/07/2022] [Accepted: 01/27/2022] [Indexed: 11/17/2022]
|