1
|
Thom RP, Warren TL, Khan S, Muhle RA, Wang PP, Brennand K, Zürcher NR, Veenstra-VanderWeele J, Hoffman EJ. A Blueprint for Translational Precision Medicine in Autism Spectrum Disorder and Related Neurogenetic Syndromes. J Child Adolesc Psychopharmacol 2025; 35:178-193. [PMID: 40138183 DOI: 10.1089/cap.2025.0023] [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] [Indexed: 03/29/2025]
Abstract
Objectives: Despite growing knowledge of the underlying neurobiology of autism spectrum disorder (ASD) and related neurogenetic syndromes, treatment discovery has remained elusive. In this review, we provide a blueprint for translational precision medicine in ASD and related neurogenetic syndromes. Methods: The discovery of trofinetide for Rett syndrome (RTT) is described, and the role of nonmammalian, mammalian, and stem cell model systems in the identification of molecular targets and drug screening is discussed. We then provide a framework for translating preclinical findings to human clinical trials, including the role of biomarkers in selecting molecular targets and evaluating target engagement, and discuss how to leverage these findings for future ASD drug development. Results: Multiple preclinical model systems for ASD have been developed, each with tradeoffs with regard to suitability for high-throughput small molecule screening, conservation across species, and behavioral face validity. Future clinical trials should incorporate biomarkers and intermediate phenotypes to demonstrate target engagement. Factors that contributed to the approval of trofinetide for RTT included replicated findings in mouse models, a well-studied natural history of the syndrome, development of RTT-specific outcome measures, and strong engagement of the RTT family community. Conclusions: The translation of our growing understanding of the neurobiology of ASD to human drug discovery will require a precision medicine approach, including the use of multiple model systems for molecular target selection, evaluation of target engagement, and clinical trial design strategies that address heterogeneity, power, and the placebo response.
Collapse
Affiliation(s)
- Robyn P Thom
- Massachusetts General Hospital Lurie Center for Autism, Harvard Medical School, Lexington, Massachusetts, USA
| | | | - Suha Khan
- Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Rebecca A Muhle
- Columbia University and New York State Psychiatric Institute, New York, New York, USA
| | - Paul P Wang
- Yale School of Medicine, New Haven, Connecticut, USA
- Simons Foundation Autism Research Initiative, New Haven, Connecticut, USA
| | | | - Nicole R Zürcher
- Massachusetts General Hospital Lurie Center for Autism, Harvard Medical School, Lexington, Massachusetts, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Boston, Massachusetts, USA
| | | | - Ellen J Hoffman
- Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
2
|
Neziri S, Köseoğlu AE, Deniz Köseoğlu G, Özgültekin B, Özgentürk NÖ. Animal models in neuroscience with alternative approaches: Evolutionary, biomedical, and ethical perspectives. Animal Model Exp Med 2024; 7:868-880. [PMID: 39375824 DOI: 10.1002/ame2.12487] [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: 05/27/2024] [Accepted: 08/07/2024] [Indexed: 10/09/2024] Open
Abstract
Animal models have been a crucial tool in neuroscience research for decades, providing insights into the biomedical and evolutionary mechanisms of the nervous system, disease, and behavior. However, their use has raised concerns on several ethical, clinical, and scientific considerations. The welfare of animals and the 3R principles (replacement, reduction, refinement) are the focus of the ethical concerns, targeting the importance of reducing the stress and suffering of these models. Several laws and guidelines are applied and developed to protect animal rights during experimenting. Concurrently, in the clinic and biomedical fields, discussions on the relevance of animal model findings on human organisms have increased. Latest data suggest that in a considerable amount of time the animal model results are not translatable in humans, costing time and money. Alternative methods, such as in vitro (cell culture, microscopy, organoids, and micro physiological systems) techniques and in silico (computational) modeling, have emerged as potential replacements for animal models, providing more accurate data in a minimized cost. By adopting alternative methods and promoting ethical considerations in research practices, we can achieve the 3R goals while upholding our responsibility to both humans and other animals. Our goal is to present a thorough review of animal models used in neuroscience from the biomedical, evolutionary, and ethical perspectives. The novelty of this research lies in integrating diverse points of views to provide an understanding of the advantages and disadvantages of animal models in neuroscience and in discussing potential alternative methods.
Collapse
Affiliation(s)
- Sabina Neziri
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Yıldız Technical University, Istanbul, Turkey
| | | | | | - Buminhan Özgültekin
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acıbadem University, Istanbul, Turkey
| | - Nehir Özdemir Özgentürk
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Yıldız Technical University, Istanbul, Turkey
| |
Collapse
|
3
|
Hong T, Park J, An G, Song J, Song G, Lim W. Evaluation of organ developmental toxicity of environmental toxicants using zebrafish embryos. Mol Cells 2024; 47:100144. [PMID: 39489379 PMCID: PMC11635654 DOI: 10.1016/j.mocell.2024.100144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/04/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024] Open
Abstract
There is increasing global concern about environmental pollutants, such as heavy metals, plastics, pharmaceuticals, personal care products, and pesticides, which have been detected in a variety of environments and are likely to be exposed to nontarget organisms, including humans. Various animal models have been utilized for toxicity assessment, and zebrafish are particularly valuable for studying the toxicity of various compounds owing to their similarity to other aquatic organisms and 70% genetic similarity to humans. Their development is easy to observe, and transgenic models for organs such as the heart, liver, blood vessels, and nervous system enable efficient studies of organ-specific toxicity. This suggests that zebrafish are a valuable tool for evaluating toxicity in specific organs and forecasting the potential impacts on other nontarget species. This review describes organ toxicity caused by various toxic substances and their mechanisms in zebrafish.
Collapse
Affiliation(s)
- Taeyeon Hong
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Junho Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Garam An
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jisoo Song
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| | - Whasun Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
4
|
He QK, Wang XY, Hu W, Cai J, Chen P, Liu MW, Wu YH. Therapeutic potential of Canna edulis RS3-resistant starch in alleviating neuroinflammation and apoptosis in a Parkinson's disease rat model. Heliyon 2024; 10:e38072. [PMID: 39347419 PMCID: PMC11438014 DOI: 10.1016/j.heliyon.2024.e38072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 09/02/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
This study aimed to investigate the effects of Miao medicinal Canna edulis RS3-resistant starch on behavioral performance and substantia nigra neuron apoptosis-related indicators in a rat model of Parkinson's disease (PD). Among the experimental groups, except for the control group, we induced PD rat models by subcutaneous injection of rotenone in the neck and back. After model induction, a 28-day drug intervention was conducted. Various techniques have been employed, including behavioral analysis, Real-time Polymerase Chain Reaction (RT-PCR), western blotting, enzyme-linked immunosorbent assay (ELISA), immunofluorescence, and terminal deoxynucleotidyltransferase-mediated UTP nick-ends. labeling (TUNEL) and Nissl staining to investigate the effect of Canna edulis RS3-resistant starch on the substantia nigra and neuronal apoptosis-related markers in the brains of PD model rats. Our study revealed that Canna edulis RS3, a resistant starch, significantly reduced the climbing time of PD model rats, prolonged their hanging time, lowered the expression levels of the inflammatory factors IL-1β, IL-6, and TNF-α, increased the number of TH-positive neurons in the substantia nigra, and decreased the levels of IL-1β, IL-6, and TNF-α. Furthermore, Canna edulis RS3 elevated the protein expression levels of tyrosine hydroxylase (TH) and Bcl-2 while reducing those of Bax, TLR4, NLRP3,and p-P65, and mitigated apoptosis and morphological changes in dopaminergic neurons in the substantia nigra region. Our results suggest that Canna edulis RS3-resistant starch may offer therapeutic benefits for PD patients with PD by potentially influencing inflammation and apoptosis in the dopaminergic system.
Collapse
Affiliation(s)
- Qian-Kun He
- Department of Neurology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China
- Department of Neurology, Traditional Chinese Medicine Hospital of Yuxi City, Yuxi, Yunnan, 6527000, China
| | - Xue-Yong Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Wei Hu
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Jing Cai
- Department of Neurology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China
| | - Peng Chen
- Department of Basic Clinical Teaching and Research of Traditional Chinese Medicine, School of Basic Medicine, Guizhou University of Chinese Medicine, Guiyang, Guizhou, 550001, China
| | - Ming-Wei Liu
- Department of Emergency Medicine, Dali Bai Autonomous Prefecture People's Hospital, Dali, 671000, China
| | - Yuan-Hua Wu
- Department of Neurology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China
| |
Collapse
|
5
|
De Meulemeester AS, Reid C, Auvin S, Carlen PL, Andrew CJ, Szlendak R, Di Sapia R, Moshé SL, Sankar R, O’Brien TJ, Baulac S, Henshall DC, Akman Ö, Galanopoulou AS. WONOEP appraisal: Modeling early onset epilepsies. Epilepsia 2024; 65:2553-2566. [PMID: 39042520 PMCID: PMC11534511 DOI: 10.1111/epi.18063] [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: 03/20/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/25/2024]
Abstract
Epilepsy has a peak incidence during the neonatal to early childhood period. These early onset epilepsies may be severe conditions frequently associated with comorbidities such as developmental deficits and intellectual disability and, in a significant percentage of patients, may be medication-resistant. The use of adult rodent models in the exploration of mechanisms and treatments for early life epilepsies is challenging, as it ignores significant age-specific developmental differences. More recently, models developed in immature animals, such as rodent pups, or in three-dimensional organoids may more closely model aspects of the immature brain and could result in more translatable findings. Although models are not perfect, they may offer a more controlled screening platform in studies of mechanisms and treatments, which cannot be done in pediatric patient cohorts. On the other hand, more simplified models with higher throughput capacities are required to deal with the large number of epilepsy candidate genes and the need for new treatment options. Therefore, a combination of different modeling approaches will be beneficial in addressing the unmet needs of pediatric epilepsy patients. In this review, we summarize the discussions on this topic that occurred during the XVI Workshop on Neurobiology of Epilepsy, organized in 2022 by the Neurobiology Commission of the International League Against Epilepsy. We provide an overview of selected models of early onset epilepsies, discussing their advantages and disadvantages. Heterologous expression models provide initial functional insights, and zebrafish, rodent models, and brain organoids present increasingly complex platforms for modeling and validating epilepsy-related phenomena. Together, these models offer valuable insights into early onset epilepsies and accelerate hypothesis generation and therapy discovery.
Collapse
Affiliation(s)
- Ann-Sofie De Meulemeester
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, Inserm, CNRS, Paris, France
- Laboratory for Molecular Biodiscovery, KU Leuven, Leuven, Belgium
| | - Christopher Reid
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
- Department of Medicine, Epilepsy Research Centre, University of Melbourne, Austin Health, Heidelberg, VIC 3084, Australia
| | - Stéphane Auvin
- AP-HP, Robert-Debré University Hospital, Pediatric Neurology Department, CRMR épilepsies rares, EpiCARE member, Paris, France
- Université Paris Cité, INSERM NeuroDiderot, Paris, France
- Institut Universitaire de France (IUF), Paris, France
| | - Peter L. Carlen
- Krembil Research Institute, 60 Leonard Ave, 7KDT430, Toronto, ON, Canada M5T 0S8
- Institute of Biomedical Engineering, University of Toronto, Rosebrugh Building, 164 College St Room 407, Toronto, ON, Canada, M5S 3G9
- Departments of Medicine and Physiology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Toronto, ON, Canada, M5S 1A8
| | - Cole J. Andrew
- MGH Epilepsy Service, Division of Clinical Neurophysiology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Roza Szlendak
- Department of Medical Genetics, Institute of Mother and Child, Kasprzaka 17A, 01-211 Warsaw, Poland
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Rossella Di Sapia
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Solomon L. Moshé
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York
- Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
- Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Raman Sankar
- Department of Neurology and Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Terence J. O’Brien
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Royal Parade, Parkville, Victoria 3050, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, 3004 Victoria, Australia
- Department of Neurology, The Alfred Hospital, Commercial Road, Melbourne, Victoria, 3004 Victoria, Australia
| | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, Inserm, CNRS, Paris, France
| | - David C. Henshall
- FutureNeuro SFI Research Centre, RCSI University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, D02 YN7
| | - Özlem Akman
- Demiroglu Bilim University, Faculty of Medicine Department of Physiology, Istanbul, Turkey
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York
- Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
- Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
6
|
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
|
7
|
Alberti G, Amico MD, Caruso Bavisotto C, Rappa F, Marino Gammazza A, Bucchieri F, Cappello F, Scalia F, Szychlinska MA. Speeding up Glioblastoma Cancer Research: Highlighting the Zebrafish Xenograft Model. Int J Mol Sci 2024; 25:5394. [PMID: 38791432 PMCID: PMC11121320 DOI: 10.3390/ijms25105394] [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: 04/03/2024] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Glioblastoma multiforme (GBM) is a very aggressive and lethal primary brain cancer in adults. The multifaceted nature of GBM pathogenesis, rising from complex interactions between cells and the tumor microenvironment (TME), has posed great treatment challenges. Despite significant scientific efforts, the prognosis for GBM remains very poor, even after intensive treatment with surgery, radiation, and chemotherapy. Efficient GBM management still requires the invention of innovative treatment strategies. There is a strong necessity to complete cancer in vitro studies and in vivo studies to properly evaluate the mechanisms of tumor progression within the complex TME. In recent years, the animal models used to study GBM tumors have evolved, achieving highly invasive GBM models able to provide key information on the molecular mechanisms of GBM onset. At present, the most commonly used animal models in GBM research are represented by mammalian models, such as mouse and canine ones. However, the latter present several limitations, such as high cost and time-consuming management, making them inappropriate for large-scale anticancer drug evaluation. In recent years, the zebrafish (Danio rerio) model has emerged as a valuable tool for studying GBM. It has shown great promise in preclinical studies due to numerous advantages, such as its small size, its ability to generate a large cohort of genetically identical offspring, and its rapid development, permitting more time- and cost-effective management and high-throughput drug screening when compared to mammalian models. Moreover, due to its transparent nature in early developmental stages and genetic and anatomical similarities with humans, it allows for translatable brain cancer research and related genetic screening and drug discovery. For this reason, the aim of the present review is to highlight the potential of relevant transgenic and xenograft zebrafish models and to compare them to the traditionally used animal models in GBM research.
Collapse
Affiliation(s)
- Giusi Alberti
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (M.D.A.); (C.C.B.); (F.R.); (A.M.G.); (F.B.); (F.C.); (F.S.)
| | - Maria Denise Amico
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (M.D.A.); (C.C.B.); (F.R.); (A.M.G.); (F.B.); (F.C.); (F.S.)
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (M.D.A.); (C.C.B.); (F.R.); (A.M.G.); (F.B.); (F.C.); (F.S.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Francesca Rappa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (M.D.A.); (C.C.B.); (F.R.); (A.M.G.); (F.B.); (F.C.); (F.S.)
- The Institute of Translational Pharmacology, National Research Council of Italy (CNR), 90146 Palermo, Italy
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (M.D.A.); (C.C.B.); (F.R.); (A.M.G.); (F.B.); (F.C.); (F.S.)
| | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (M.D.A.); (C.C.B.); (F.R.); (A.M.G.); (F.B.); (F.C.); (F.S.)
| | - Francesco Cappello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (M.D.A.); (C.C.B.); (F.R.); (A.M.G.); (F.B.); (F.C.); (F.S.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Federica Scalia
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (M.D.A.); (C.C.B.); (F.R.); (A.M.G.); (F.B.); (F.C.); (F.S.)
| | - Marta Anna Szychlinska
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
8
|
Locubiche S, Ordóñez V, Abad E, Scotto di Mase M, Di Donato V, De Santis F. A Zebrafish-Based Platform for High-Throughput Epilepsy Modeling and Drug Screening in F0. Int J Mol Sci 2024; 25:2991. [PMID: 38474238 DOI: 10.3390/ijms25052991] [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: 11/20/2023] [Revised: 02/22/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
The zebrafish model has emerged as a reference tool for phenotypic drug screening. An increasing number of molecules have been brought from bench to bedside thanks to zebrafish-based assays over the last decade. The high homology between the zebrafish and the human genomes facilitates the generation of zebrafish lines carrying loss-of-function mutations in disease-relevant genes; nonetheless, even using this alternative model, the establishment of isogenic mutant lines requires a long generation time and an elevated number of animals. In this study, we developed a zebrafish-based high-throughput platform for the generation of F0 knock-out (KO) models and the screening of neuroactive compounds. We show that the simultaneous inactivation of a reporter gene (tyrosinase) and a second gene of interest allows the phenotypic selection of F0 somatic mutants (crispants) carrying the highest rates of mutations in both loci. As a proof of principle, we targeted genes associated with neurodevelopmental disorders and we efficiently generated de facto F0 mutants in seven genes involved in childhood epilepsy. We employed a high-throughput multiparametric behavioral analysis to characterize the response of these KO models to an epileptogenic stimulus, making it possible to employ kinematic parameters to identify seizure-like events. The combination of these co-injection, screening and phenotyping methods allowed us to generate crispants recapitulating epilepsy features and to test the efficacy of compounds already during the first days post fertilization. Since the strategy can be applied to a wide range of indications, this study paves the ground for high-throughput drug discovery and promotes the use of zebrafish in personalized medicine and neurotoxicity assessment.
Collapse
Affiliation(s)
- Sílvia Locubiche
- ZeClinics S.L., Carrer de Laureà Miró, 408-410, 08980 Sant Feliu de Llobregat, Spain
- Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
| | - Víctor Ordóñez
- ZeClinics S.L., Carrer de Laureà Miró, 408-410, 08980 Sant Feliu de Llobregat, Spain
| | - Elena Abad
- ZeClinics S.L., Carrer de Laureà Miró, 408-410, 08980 Sant Feliu de Llobregat, Spain
| | | | - Vincenzo Di Donato
- ZeClinics S.L., Carrer de Laureà Miró, 408-410, 08980 Sant Feliu de Llobregat, Spain
| | - Flavia De Santis
- ZeClinics S.L., Carrer de Laureà Miró, 408-410, 08980 Sant Feliu de Llobregat, Spain
| |
Collapse
|
9
|
Moussavi N, van der Ent W, Diallo D, Sanogo R, Malterud KE, Esguerra CV, Wangensteen H. Inhibition of Seizure-Like Paroxysms and Toxicity Effects of Securidaca longepedunculata Extracts and Constituents in Zebrafish Danio rerio. ACS Chem Neurosci 2024; 15:617-628. [PMID: 38270158 PMCID: PMC10853935 DOI: 10.1021/acschemneuro.3c00642] [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: 10/06/2023] [Revised: 01/08/2024] [Accepted: 01/15/2024] [Indexed: 01/26/2024] Open
Abstract
Plants used in traditional medicine in the management of epilepsy could potentially yield novel drug compounds with antiepileptic properties. The medicinal plant Securidaca longepedunculata is widely used in traditional medicine in the African continent, and epilepsy is among several indications. Limited knowledge is available on its toxicity and medicinal effects, such as anticonvulsant activities. This study explores the potential in vivo inhibition of seizure-like paroxysms and toxicity effects of dichloromethane (DCM) and ethanol (EtOH) extracts, as well as isolated xanthones and benzoates of S. longepedunculata. Ten phenolic compounds were isolated from the DCM extract. All of the substances were identified by nuclear magnetic resonance spectroscopy. Assays for toxicity and inhibition of pentylenetetrazole (PTZ)-induced seizure-like paroxysms were performed in zebrafish larvae. Among the compounds assessed in the assay for maximum tolerated concentration (MTC), benzyl-2-hydroxy-6-methoxy-benzoate (MTC 12.5 μM), 4,8-dihydroxy-1,2,3,5,6-pentamethoxyxanthone (MTC 25 μM), and 1,7-dihydroxy-4-methoxyxanthone (MTC 6.25 μM) were the most toxic. The DCM extract, 1,7-dihydroxy-4-methoxyxanthone and 2-hydroxy-1,7-dimethoxyxanthone displayed the most significant inhibition of paroxysms by altering the locomotor behavior in GABAA receptor antagonist, PTZ, which induced seizures in larval zebrafish. The EtOH extract, benzyl benzoate, and benzyl-2-hydroxy-6-methoxy-benzoate unexpectedly increased locomotor activity in treated larval zebrafish and decreased locomotor activity in nontreated larval zebrafish, seemingly due to paradoxical excitation. The results reveal promising medicinal activities of this plant, contributing to our understanding of its use as an antiepileptic drug. It also shows us the presence of potentially new lead compounds for future drug development.
Collapse
Affiliation(s)
- Nastaran Moussavi
- Section
for Pharmaceutical Chemistry, Department of Pharmacy, University of Oslo, P.O. Box 1068, Oslo 0316, Norway
| | - Wietske van der Ent
- NCMM,
Chemical Neuroscience Group, Centre for Molecular Medicine Norway,
Faculty of Medicine, University of Oslo, Oslo 0349, Norway
| | - Drissa Diallo
- Department
of Traditional Medicine, National Institute
of Public Health, PB, Bamako 1746, Mali
- Faculty
of Pharmacy, University of Sciences, Techniques
and Technologies of Bamako (USTTB), Bamako 1746, Mali
| | - Rokia Sanogo
- Department
of Traditional Medicine, National Institute
of Public Health, PB, Bamako 1746, Mali
- Faculty
of Pharmacy, University of Sciences, Techniques
and Technologies of Bamako (USTTB), Bamako 1746, Mali
| | - Karl E. Malterud
- Section
for Pharmaceutical Chemistry, Department of Pharmacy, University of Oslo, P.O. Box 1068, Oslo 0316, Norway
| | - Camila V. Esguerra
- Section
for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O.
Box 1068, Oslo 0316, Norway
| | - Helle Wangensteen
- Section
for Pharmaceutical Chemistry, Department of Pharmacy, University of Oslo, P.O. Box 1068, Oslo 0316, Norway
| |
Collapse
|
10
|
Jin DS, Neelakantan U, Lacadie CM, Chen T, Rooney B, Liu Y, Wu W, Wang Z, Papademetris X, Hoffman EJ. Brain Registration and Evaluation for Zebrafish (BREEZE)-mapping: A pipeline for whole-brain structural and activity analyses. STAR Protoc 2023; 4:102647. [PMID: 37897734 PMCID: PMC10641303 DOI: 10.1016/j.xpro.2023.102647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/05/2023] [Accepted: 09/26/2023] [Indexed: 10/30/2023] Open
Abstract
Here, we present Brain Registration and Evaluation for Zebrafish (BREEZE)-mapping, a user-friendly pipeline for the registration and analysis of whole-brain images in larval zebrafish. We describe steps for pre-processing, registration, quantification, and visualization of whole-brain phenotypes in zebrafish mutants of genes associated with neurodevelopmental and neuropsychiatric disorders. By utilizing BioImage Suite Web, an open-source software package originally developed for processing human brain imaging data, we provide a highly accessible whole-brain mapping protocol developed for users with general computational proficiency. For complete details on the use and execution of this protocol, please refer to Weinschutz Mendes et al. (2023).1.
Collapse
Affiliation(s)
- David S Jin
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale University, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Uma Neelakantan
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Cheryl M Lacadie
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06510, USA
| | - Tianying Chen
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Brendan Rooney
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yunqing Liu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Weimiao Wu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Zuoheng Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Xenophon Papademetris
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06510, USA; Section of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, CT 06510, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Ellen J Hoffman
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale University, New Haven, CT 06510, USA.
| |
Collapse
|
11
|
Rajesh V, Divya PK. Embryonic exposure to decitabine induces multiple neural tube defects in developing zebrafish. FISH PHYSIOLOGY AND BIOCHEMISTRY 2023; 49:1357-1379. [PMID: 37982970 DOI: 10.1007/s10695-023-01261-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 11/01/2023] [Indexed: 11/21/2023]
Abstract
Neural tube defects are severe congenital disorders of the central nervous system that originate during embryonic development when the neural tube fails to close completely. It affects one to two infants per 1000 births. The aetiology is multifactorial with contributions from both genetic and environmental factors. Dysregulated epigenetic mechanisms, in particular the abnormal genome-wide methylation during embryogenesis, have been linked to developmental abnormalities including neural tube defects. The current study investigated the influence of decitabine (DCT), a DNA methylation inhibitor, on embryonic development in zebrafish, with a focus on neural tube formation. The developing zebrafish embryos were exposed to graded concentrations of decitabine (from 13.69 μM to 1 mM) before the onset of neurulation. The developmental process was monitored at regular time intervals post fertilization. At 120 h post fertilization, the developing embryos were inspected individually to determine the incidence and severity of neural tube defects. Using alizarin red staining, the cranial and caudal neural tube morphology was examined in formaldehyde fixed larvae. Anomalies in neural tube and somite development, as well as a delay in hatching, were discovered at an early stage of development. As development continued, neural tube defects became increasingly evident, and there was a concentration-dependent rise in the prevalence and severity of various neural tube defects. 90% of growing embryos in the group exposed to decitabine 1 mM had multiple neural tube malformations, and 10% had isolated neural tube defects. With several abnormalities, the caudal region of the neural tube was seriously compromised. The histopathological studies supported the malformations in neural tube. Our study revealed the harmful impact of decitabine on the development of the neural tube in growing zebrafish. Moreover, these findings support the hypothesis that the hypomethylation during embryonic development causes neural tube defects.
Collapse
Affiliation(s)
- Venugopalan Rajesh
- Department of Pharmacology, The Erode College of Pharmacy and Research Institute affiliated to The Tamil Nadu Dr. M.G.R. Medical University, Veppampalayam, Vallipurathampalayam (Po), Erode, Chennai, Tamil Nadu, 638112, India.
| | - Pachangattupalayam Karuppusamy Divya
- Department of Pharmacology, The Erode College of Pharmacy and Research Institute affiliated to The Tamil Nadu Dr. M.G.R. Medical University, Veppampalayam, Vallipurathampalayam (Po), Erode, Chennai, Tamil Nadu, 638112, India
| |
Collapse
|
12
|
Serrano RJ, Oorschot V, Palipana D, Calcinotto V, Sonntag C, Ramm G, Bryson-Richardson RJ. Genetic model of UBA5 deficiency highlights the involvement of both peripheral and central nervous systems and identifies widespread mitochondrial abnormalities. Brain Commun 2023; 5:fcad317. [PMID: 38046095 PMCID: PMC10691876 DOI: 10.1093/braincomms/fcad317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 10/10/2023] [Accepted: 11/19/2023] [Indexed: 12/05/2023] Open
Abstract
Variants in UBA5 have been reported to cause neurological disease with impaired motor function, developmental delay, intellectual disability and brain pathology as recurrent clinical manifestations. UBA5 encodes a ubiquitin-activating-like enzyme that activates ufmylation, a post-translational ubiquitin-like modification pathway, which has been implicated in neurodevelopment and neuronal survival. The reason behind the variation in severity and clinical manifestations in affected individuals and the signal transduction pathways regulated by ufmylation that compromise the nervous system remains unknown. Zebrafish have emerged as a powerful model to study neurodegenerative disease due to its amenability for in vivo analysis of muscle and neuronal tissues, high-throughput examination of motor function and rapid embryonic development allowing an examination of disease progression. Using clustered regularly interspaced short palindromic repeats-associated protein 9 genome editing, we developed and characterized zebrafish mutant models to investigate disease pathophysiology. uba5 mutant zebrafish showed a significantly impaired motor function accompanied by delayed growth and reduced lifespan, reproducing key phenotypes observed in affected individuals. Our study demonstrates the suitability of zebrafish to study the pathophysiology of UBA5-related disease and as a powerful tool to identify pathways that could reduce disease progression. Furthermore, uba5 mutants exhibited widespread mitochondrial damage in both the nervous system and the skeletal muscle, suggesting that a perturbation of mitochondrial function may contribute to disease pathology.
Collapse
Affiliation(s)
- Rita J Serrano
- School of Biological Sciences, Monash University, Melbourne 3800, Australia
| | - Viola Oorschot
- Monash Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Melbourne 3800, Australia
| | - Dashika Palipana
- School of Biological Sciences, Monash University, Melbourne 3800, Australia
| | - Vanessa Calcinotto
- School of Biological Sciences, Monash University, Melbourne 3800, Australia
| | - Carmen Sonntag
- School of Biological Sciences, Monash University, Melbourne 3800, Australia
| | - Georg Ramm
- Monash Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Melbourne 3800, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | | |
Collapse
|
13
|
Zhong Y, Zhang N, Zhao F, Chang S, Chen W, Cao Q, Sun L, Wang Y, Gong Z, Lu L, Liu D, Yang L. RBFOX1 and Working Memory: From Genome to Transcriptome Revealed Posttranscriptional Mechanism Separate From Attention-Deficit/Hyperactivity Disorder. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:1042-1052. [PMID: 37881587 PMCID: PMC10593897 DOI: 10.1016/j.bpsgos.2022.08.006] [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: 05/27/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/22/2022] Open
Abstract
Background Many psychiatric disorders share a working memory (WM) impairment phenotype, yet the genetic causes remain unclear. Here, we generated genetic profiles of WM deficits using attention-deficit/hyperactivity disorder samples and validated the results in zebrafish models. Methods We used 2 relatively large attention-deficit/hyperactivity disorder cohorts, 799 and 776 cases, respectively. WM impairment was assessed using the Rey Complex Figure Test. First, association analyses were conducted at single-variant, gene-based, and gene-set levels. Deeper insights into the biological mechanism were gained from further functional exploration by bioinformatic analyses and zebrafish models. Results Genomic analyses identified and replicated a locus with rs75885813 as the index single nucleotide polymorphism showing significant association with WM defects but not with attention-deficit/hyperactivity disorder. Functional feature exploration found that these single nucleotide polymorphisms may regulate the expression level of RBFOX1 through chromatin interaction. Further pathway enrichment analysis of potential associated single nucleotide polymorphisms revealed the involvement of posttranscription regulation that affects messenger RNA stability and/or alternative splicing. Zebrafish with functionally knocked down or genome-edited rbfox1 exhibited WM impairment but no hyperactivity. Transcriptome profiling of rbfox1-defective zebrafish indicated that alternative exon usages of snap25a might partially lead to reduced WM learning of larval zebrafish. Conclusions The locus with rs75885813 in RBFOX1 was identified as associated with WM. Rbfox1 regulates synaptic and long-term potentiation-related gene snap25a to adjust WM at the posttranscriptional level.
Collapse
Affiliation(s)
- Yuanxin Zhong
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Na Zhang
- School of Life Science, Southern University of Science and Technology, Shenzhen, China
- Department of Biological Science, National University of Singapore, Singapore
| | - Feng Zhao
- School of Life Science, Southern University of Science and Technology, Shenzhen, China
| | - Suhua Chang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Wei Chen
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Qingjiu Cao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Li Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Yufeng Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Zhiyuan Gong
- Department of Biological Science, National University of Singapore, Singapore
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
- Peking-Tsinghua Center for Life Sciences, International Data Group, McGovern Institute for Brain Research at Peking University, Peking University, Beijing, China
| | - Dong Liu
- School of Life Science, Southern University of Science and Technology, Shenzhen, China
| | - Li Yang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| |
Collapse
|
14
|
Da Silveira Cavalcante L, Higuita ML, González-Rosa JM, Marques B, To S, Pendexter CA, Cronin SE, Gopinathan K, de Vries RJ, Ellett F, Uygun K, Langenau DM, Toner M, Tessier SN. Zebrafish as a high throughput model for organ preservation and transplantation research. FASEB J 2023; 37:e23187. [PMID: 37718489 PMCID: PMC10754348 DOI: 10.1096/fj.202300076r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 08/15/2023] [Accepted: 08/24/2023] [Indexed: 09/19/2023]
Abstract
Despite decades of effort, the preservation of complex organs for transplantation remains a significant barrier that exacerbates the organ shortage crisis. Progress in organ preservation research is significantly hindered by suboptimal research tools that force investigators to sacrifice translatability over throughput. For instance, simple model systems, such as single cell monolayers or co-cultures, lack native tissue structure and functional assessment, while mammalian whole organs are complex systems with confounding variables not compatible with high-throughput experimentation. In response, diverse fields and industries have bridged this experimental gap through the development of rich and robust resources for the use of zebrafish as a model organism. Through this study, we aim to demonstrate the value zebrafish pose for the fields of solid organ preservation and transplantation, especially with respect to experimental transplantation efforts. A wide array of methods were customized and validated for preservation-specific experimentation utilizing zebrafish, including the development of assays at multiple developmental stages (larvae and adult), methods for loading and unloading preservation agents, and the development of viability scores to quantify functional outcomes. Using this platform, the largest and most comprehensive screen of cryoprotectant agents (CPAs) was performed to determine their toxicity and efficiency at preserving complex organ systems using a high subzero approach called partial freezing (i.e., storage in the frozen state at -10°C). As a result, adult zebrafish cardiac function was successfully preserved after 5 days of partial freezing storage. In combination, the methods and techniques developed have the potential to drive and accelerate research in the fields of solid organ preservation and transplantation.
Collapse
Affiliation(s)
- Luciana Da Silveira Cavalcante
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Manuela Lopera Higuita
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Juan Manuel González-Rosa
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute, Harvard Medical School, Charlestown MA, USA
| | - Beatriz Marques
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
| | - Samantha To
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute, Harvard Medical School, Charlestown MA, USA
| | - Casie A. Pendexter
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Stephanie E.J. Cronin
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Kaustav Gopinathan
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
| | - Reinier J. de Vries
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Felix Ellett
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Korkut Uygun
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - David M. Langenau
- Molecular Pathology Unit and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Mehmet Toner
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Shannon N. Tessier
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| |
Collapse
|
15
|
Xu J, Casanave R, Chitre AS, Wang Q, Nguyen KM, Blake C, Wagle M, Cheng R, Polesskaya O, Palmer AA, Guo S. Causal Genetic Loci for a Motivated Behavior Spectrum Harbor Psychiatric Risk Genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.06.556529. [PMID: 37732200 PMCID: PMC10508786 DOI: 10.1101/2023.09.06.556529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Behavioral diversity is critical for population fitness. Individual differences in risk-taking are observed across species, but underlying genetic mechanisms and conservation are largely unknown. We examined dark avoidance in larval zebrafish, a motivated behavior reflecting an approach-avoidance conflict. Brain-wide calcium imaging revealed significant neural activity differences between approach-inclined versus avoidance-inclined individuals. We used a population of ∼6,000 to perform the first genome-wide association study (GWAS) in zebrafish, which identified 34 genomic regions harboring many genes that are involved in synaptic transmission and human psychiatric diseases. We used CRISPR to study several causal genes: serotonin receptor-1b ( htr1b ), nitric oxide synthase-1 ( nos1 ), and stress-induced phosphoprotein-1 ( stip1 ). We further identified 52 conserved elements containing 66 GWAS significant variants. One encoded an exonic regulatory element that influenced tissue-specific nos1 expression. Together, these findings reveal new genetic loci and establish a powerful, scalable animal system to probe mechanisms underlying motivation, a critical dimension of psychiatric diseases.
Collapse
|
16
|
Hedge JM, Hunter DL, Sanders E, Jarema KA, Olin JK, Britton KN, Lowery M, Knapp BR, Padilla S, Hill BN. Influence of Methylene Blue or Dimethyl Sulfoxide on Larval Zebrafish Development and Behavior. Zebrafish 2023; 20:132-145. [PMID: 37406269 PMCID: PMC10627343 DOI: 10.1089/zeb.2023.0017] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023] Open
Abstract
The use of larval zebrafish developmental testing and assessment, specifically larval zebrafish locomotor activity, has been recognized as a higher throughput testing strategy to identify developmentally toxic and neurotoxic chemicals. There are, however, no standardized protocols for this type of assay, which could result in confounding variables being overlooked. Two chemicals commonly employed during early-life stage zebrafish assays, methylene blue (antifungal agent) and dimethyl sulfoxide (DMSO, a commonly used vehicle) have been reported to affect the morphology and behavior of freshwater fish. In this study, we conducted developmental toxicity (morphology) and neurotoxicity (behavior) assessments of commonly employed concentrations for both chemicals (0.6-10.0 μM methylene blue; 0.3%-1.0% v/v DMSO). A light-dark transition behavioral testing paradigm was applied to morphologically normal, 6 days postfertilization (dpf) zebrafish larvae kept at 26°C. Additionally, an acute DMSO challenge was administered based on early-life stage zebrafish assays typically used in this research area. Results from developmental toxicity screens were similar between both chemicals with no morphological abnormalities detected at any of the concentrations tested. However, neurodevelopmental results were mixed between the two chemicals of interest. Methylene blue resulted in no behavioral changes up to the highest concentration tested, 10.0 μM. By contrast, DMSO altered larval behavior following developmental exposure at concentrations as low as 0.5% (v/v) and exhibited differential concentration-response patterns in the light and dark photoperiods. These results indicate that developmental DMSO exposure can affect larval zebrafish locomotor activity at routinely used concentrations in developmental neurotoxicity assessments, whereas methylene blue does not appear to be developmentally or neurodevelopmentally toxic to larval zebrafish at routinely used concentrations. These results also highlight the importance of understanding the influence of experimental conditions on larval zebrafish locomotor activity that may ultimately confound the interpretation of results.
Collapse
Affiliation(s)
- Joan M. Hedge
- Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Advanced Experimental Toxicology Models Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Deborah L. Hunter
- Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Erik Sanders
- Aquatics Lab Services LLC 1112 Nashville Street St. Peters, MO 63376, USA
| | - Kimberly A. Jarema
- Office of Research and Development, Center for Public Health and Environmental Assessment, Immediate Office, Program Operations Staff, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Jeanene K. Olin
- Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Katy N. Britton
- ORAU Research Participation Program hosted by EPA, Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Morgan Lowery
- Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Bridget R. Knapp
- ORISE Research Participation Program hosted by EPA, Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Stephanie Padilla
- Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Bridgett N. Hill
- ORISE Research Participation Program hosted by EPA, Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| |
Collapse
|
17
|
Huo WB, Jia PP, Li WG, Xie XY, Yang G, Pei DS. Sulfonamides (SAs) exposure causes neurobehavioral toxicity at environmentally relevant concentrations (ERCs) in early development of zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 261:106614. [PMID: 37390778 DOI: 10.1016/j.aquatox.2023.106614] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 07/02/2023]
Abstract
Antibiotics, due to their stability and persistence in the environment, can have chronic impacts on various ecosystems and organisms. However, the molecular mechanisms underlying antibiotic toxicity at environmental concentrations, particularly the neurotoxic effects of sulfonamides (SAs), remain poorly understood. In this study, we assessed the neurotoxicity of six SAs including the sulfadiazine (SD), sulfathiazole (ST), sulfamethoxazole (SMX), sulfisoxazole (SIZ), sulfapyridine (SPD), and sulfadimethoxine (SDM) by exposing zebrafish to environmentally relevant concentrations (ERCs). The SAs exhibited concentration-dependent effects on zebrafish behavior, including spontaneous movement, heartbeat, survival rate, and body metrics, ultimately leading to depressive-like symptoms and sublethal toxicity during early life stages. Notably, even the lowest SA concentration (0.05 μg/L) induced neurotoxicity and behavioral impairment in zebrafish. We observed a dose-dependent increase in melancholy behavior as indicated by increased resting time and decreased motor activity in zebrafish larvae. Following exposure to SAs from 4 to 120 h post-fertilization (hpf), key genes involved in folate synthesis [sepiapterin reductase a (spra), phenylalanine hydroxylase (pah), tyrosine hydroxylase (th), and tryptophan hydroxylase 1 (tryptophan 5-monooxygenase) a tryptophan hydroxylase (tph1a)] and carbonic anhydrase (CA) metabolism [carbonic anhydrase II (ca2), carbonic anhydrase IV a (ca4a), carbonic anhydrase VII (ca7), and carbonic anhydrase XIV (ca14)] were significantly downregulated or inhibited at different concentrations. Our findings demonstrate that acute exposure to six SAs at environmentally relevant concentrations induces developmental and neurotoxic effects in zebrafish, impacting folate synthesis pathways and CA metabolism. These results provide valuable insights into the potential role of antibiotics in depressive disorders and neuroregulatory pathways.
Collapse
Affiliation(s)
- Wen-Bo Huo
- College of Life Science, Henan Normal University, Xinxiang 453007, China; School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Pan-Pan Jia
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Wei-Guo Li
- College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Xiao-Yu Xie
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Guan Yang
- Environmental Science and Engineering, Chongqing Jiaotong University, Chongqing 400074, China
| | - De-Sheng Pei
- School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
18
|
Davidović P, Blagojević D, Meriluoto J, Simeunović J, Svirčev Z. Biotests in Cyanobacterial Toxicity Assessment-Efficient Enough or Not? BIOLOGY 2023; 12:biology12050711. [PMID: 37237524 DOI: 10.3390/biology12050711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/27/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023]
Abstract
Cyanobacteria are a diverse group of organisms known for producing highly potent cyanotoxins that pose a threat to human, animal, and environmental health. These toxins have varying chemical structures and toxicity mechanisms and several toxin classes can be present simultaneously, making it difficult to assess their toxic effects using physico-chemical methods, even when the producing organism and its abundance are identified. To address these challenges, alternative organisms among aquatic vertebrates and invertebrates are being explored as more assays evolve and diverge from the initially established and routinely used mouse bioassay. However, detecting cyanotoxins in complex environmental samples and characterizing their toxic modes of action remain major challenges. This review provides a systematic overview of the use of some of these alternative models and their responses to harmful cyanobacterial metabolites. It also assesses the general usefulness, sensitivity, and efficiency of these models in investigating the mechanisms of cyanotoxicity expressed at different levels of biological organization. From the reported findings, it is clear that cyanotoxin testing requires a multi-level approach. While studying changes at the whole-organism level is essential, as the complexities of whole organisms are still beyond the reach of in vitro methodologies, understanding cyanotoxicity at the molecular and biochemical levels is necessary for meaningful toxicity evaluations. Further research is needed to refine and optimize bioassays for cyanotoxicity testing, which includes developing standardized protocols and identifying novel model organisms for improved understanding of the mechanisms with fewer ethical concerns. In vitro models and computational modeling can complement vertebrate bioassays and reduce animal use, leading to better risk assessment and characterization of cyanotoxins.
Collapse
Affiliation(s)
- Petar Davidović
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia
| | - Dajana Blagojević
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia
| | - Jussi Meriluoto
- Faculty of Science and Engineering, Biochemistry, Åbo Akademi, Tykistökatu 6 A, 20520 Turku, Finland
| | - Jelica Simeunović
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia
| | - Zorica Svirčev
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia
- Faculty of Science and Engineering, Biochemistry, Åbo Akademi, Tykistökatu 6 A, 20520 Turku, Finland
| |
Collapse
|
19
|
Weinschutz Mendes H, Neelakantan U, Liu Y, Fitzpatrick SE, Chen T, Wu W, Pruitt A, Jin DS, Jamadagni P, Carlson M, Lacadie CM, Enriquez KD, Li N, Zhao D, Ijaz S, Sakai C, Szi C, Rooney B, Ghosh M, Nwabudike I, Gorodezky A, Chowdhury S, Zaheer M, McLaughlin S, Fernandez JM, Wu J, Eilbott JA, Vander Wyk B, Rihel J, Papademetris X, Wang Z, Hoffman EJ. High-throughput functional analysis of autism genes in zebrafish identifies convergence in dopaminergic and neuroimmune pathways. Cell Rep 2023; 42:112243. [PMID: 36933215 PMCID: PMC10277173 DOI: 10.1016/j.celrep.2023.112243] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/15/2022] [Accepted: 02/23/2023] [Indexed: 03/19/2023] Open
Abstract
Advancing from gene discovery in autism spectrum disorders (ASDs) to the identification of biologically relevant mechanisms remains a central challenge. Here, we perform parallel in vivo functional analysis of 10 ASD genes at the behavioral, structural, and circuit levels in zebrafish mutants, revealing both unique and overlapping effects of gene loss of function. Whole-brain mapping identifies the forebrain and cerebellum as the most significant contributors to brain size differences, while regions involved in sensory-motor control, particularly dopaminergic regions, are associated with altered baseline brain activity. Finally, we show a global increase in microglia resulting from ASD gene loss of function in select mutants, implicating neuroimmune dysfunction as a key pathway relevant to ASD biology.
Collapse
Affiliation(s)
| | - Uma Neelakantan
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yunqing Liu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Sarah E Fitzpatrick
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; MD-PhD Program, Yale School of Medicine, New Haven, CT 06510, USA
| | - Tianying Chen
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Weimiao Wu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - April Pruitt
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA
| | - David S Jin
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Marina Carlson
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA
| | - Cheryl M Lacadie
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Ningshan Li
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA; SJTU-Yale Joint Center for Biostatistics and Data Science, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dejian Zhao
- Department of Genetics, Yale Center for Genome Analysis, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sundas Ijaz
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Catalina Sakai
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Christina Szi
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Brendan Rooney
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Marcus Ghosh
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Ijeoma Nwabudike
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; MD-PhD Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA
| | - Andrea Gorodezky
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sumedha Chowdhury
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Meeraal Zaheer
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sarah McLaughlin
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Jia Wu
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jeffrey A Eilbott
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Brent Vander Wyk
- Department of Internal Medicine, Section of Geriatrics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Xenophon Papademetris
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06510, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Zuoheng Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Ellen J Hoffman
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
20
|
Hill BN, Britton KN, Hunter DL, Olin JK, Lowery M, Hedge JM, Knapp BR, Jarema KA, Rowson Z, Padilla S. Inconsistencies in variable reporting and methods in larval zebrafish behavioral assays. Neurotoxicol Teratol 2023; 96:107163. [PMID: 36758822 PMCID: PMC10337341 DOI: 10.1016/j.ntt.2023.107163] [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: 09/23/2022] [Revised: 01/06/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
New approaches in developmental neurotoxicity (DNT) screening are needed due to the tens of thousands of chemicals requiring hazard assessments. Zebrafish (Danio rerio) are an alternative vertebrate model for DNT testing, but without a standardized protocol for larval behavioral assays, comparison of results among laboratories is challenging. To evaluate the congruence of protocols across laboratories, we conducted a literature review of DNT studies focusing on larval zebrafish behavior assays and cataloged experimental design consistencies. Our review focused on 51 unique method variables in publications where chemical exposure occurred in early development and subsequent larval locomotor evaluation focused on assays that included a light/dark photoperiod transition. We initially identified 94 publications, but only 31 exclusively met our inclusion criteria which focused on parameters that are important to an assay employed by our laboratory. No publication reported 100% of the targeted variables; only 51 to 86% of those variables were reported in the reviewed publications, with some aspects of the experimental design consistent among laboratories. However, no protocol was exactly the same for any two publications. Many of these variables had more than one parameter/design reported, highlighting the inconsistencies among methods. Overall, there is not only a strong need for the development of a standardized testing protocol for larval zebrafish locomotor assays, but there is also a need for a standardized protocol for reporting experimental variables in the literature. Here we include an extensive guideline checklist for conducting larval zebrafish developmental behavior assays.
Collapse
Affiliation(s)
- Bridgett N Hill
- ORISE Research Participation Program hosted by EPA, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Katy N Britton
- ORAU Research Participation Program hosted by EPA, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Deborah L Hunter
- Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Jeanene K Olin
- Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Morgan Lowery
- Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Joan M Hedge
- Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Advanced Experimental Toxicology Models Branch, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Bridget R Knapp
- ORISE Research Participation Program hosted by EPA, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Kimberly A Jarema
- Center for Public Health and Environmental Assessment, Immediate Office, Program Operations Staff, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Zachary Rowson
- ORISE Research Participation Program hosted by EPA, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Stephanie Padilla
- Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| |
Collapse
|
21
|
Integrative Roles of Dopamine Pathway and Calcium Channels Reveal a Link between Schizophrenia and Opioid Use Disorder. Int J Mol Sci 2023; 24:ijms24044088. [PMID: 36835497 PMCID: PMC9966501 DOI: 10.3390/ijms24044088] [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: 12/30/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
Several theories have been proposed to explain the mechanisms of substance use in schizophrenia. Brain neurons pose a potential to provide novel insights into the association between opioid addiction, withdrawal, and schizophrenia. Thus, we exposed zebrafish larvae at 2 days post-fertilization (dpf) to domperidone (DPM) and morphine, followed by morphine withdrawal. Drug-induced locomotion and social preference were assessed, while the level of dopamine and the number of dopaminergic neurons were quantified. In the brain tissue, the expression levels of genes associated with schizophrenia were measured. The effects of DMP and morphine were compared to vehicle control and MK-801, a positive control to mimic schizophrenia. Gene expression analysis revealed that α1C, α1Sa, α1Aa, drd2a, and th1 were up-regulated after 10 days of exposure to DMP and morphine, while th2 was down-regulated. These two drugs also increased the number of positive dopaminergic neurons and the total dopamine level but reduced the locomotion and social preference. The termination of morphine exposure led to the up-regulation of th2, drd2a, and c-fos during the withdrawal phase. Our integrated data implicate that the dopamine system plays a key role in the deficits in social behavior and locomotion that are common in the schizophrenia-like symptoms and opioid dependence.
Collapse
|
22
|
Calderon-Garcia AA, Perez-Fernandez M, Curto-Aguilera D, Rodriguez-Martin I, Sánchez-Barba M, Gonzalez-Nunez V. Exposure to Morphine and Cocaine Modify the Transcriptomic Landscape in Zebrafish Embryos. Neuroscience 2022; 507:14-27. [PMID: 36404518 DOI: 10.1016/j.neuroscience.2022.10.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/19/2022]
Abstract
Morphine and other opioid analgesics are the drugs of election to treat moderate-to-severe pain, and they elicit their actions by binding to the opioid receptors. Cocaine is a potent inhibitor of dopamine, serotonin, and noradrenaline reuptake, as it blocks DAT, the dopamine transporter, causing an increase in the local concentration of these neurotransmitters in the synaptic cleft. The molecular effects of these drugs have been studied in specific brain areas or nuclei, but the systemic effects in the whole organism have not been comprehensively analyzed. This study aims to analyze the transcriptomic changes elicited by morphine (10 uM) and cocaine (15 uM) in zebrafish embryos. An RNAseq assay was performed with tissues extracts from zebrafish embryos treated from 5 hpf (hours post fertilization) to 72 hpf, and the most representative deregulated genes were experimentally validated by qPCR. We have found changes in the expression of genes related to lipid metabolism, chemokine receptor ligands, visual system, hemoglobins, and metabolic detoxification pathways. Besides, morphine and cocaine modified the global DNA methylation pattern in zebrafish embryos, which would explain the changes in gene expression elicited by these two drugs of abuse.
Collapse
Affiliation(s)
- Andrés Angel Calderon-Garcia
- Dept. Biochemistry and Molecular Biology, Faculty of Medicine, University of Salamanca, Spain; Instituto de Neurociencias de Castilla y León (INCYL), Faculty of Medicine, University of Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Spain
| | - Maria Perez-Fernandez
- Instituto de Neurociencias de Castilla y León (INCYL), Faculty of Medicine, University of Salamanca, Spain
| | - Daniel Curto-Aguilera
- Dept. Biochemistry and Molecular Biology, Faculty of Medicine, University of Salamanca, Spain
| | - Ivan Rodriguez-Martin
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Mercedes Sánchez-Barba
- Institute of Biomedical Research of Salamanca (IBSAL), Spain; Dept. Statistics. Faculty of Medicine, University of Salamanca, Spain
| | - Veronica Gonzalez-Nunez
- Dept. Biochemistry and Molecular Biology, Faculty of Medicine, University of Salamanca, Spain; Instituto de Neurociencias de Castilla y León (INCYL), Faculty of Medicine, University of Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Spain.
| |
Collapse
|
23
|
Tsai JF, Wu TS, Yu FY, Liu BH. Neurotoxicity of mycotoxin citrinin: Novel evidence in developing zebrafish and underlying mechanisms in human neuron cells. Food Chem Toxicol 2022; 171:113543. [PMID: 36460223 DOI: 10.1016/j.fct.2022.113543] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/03/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022]
Abstract
Citrinin (CTN) is a mycotoxin that is found as a contaminant in various types of food/feed grains and fermented food supplements. Previous studies have already established the nephrotoxicity and hepatotoxicity of CTN, but the neurotoxicity of CTN has not been clearly examined. In this study, CTN at 2-20 μM was first found to interfere with the neural ganglia formation and locomotive behavior of embryonic zebrafish, a vertebrate animal model, at 24 hpf and 6 dpf, respectively. Further exposure of human neuroblastoma SH-SY5Y cells to 10 and 20 μM CTN for 72 h indicated that pathways responsible for neuron differentiation and projection guidance were down-regulated while oxidative stress and electron transport chain pathways were up-regulated based on the enrichment results of GSEA in the transcriptomic profiling. PCR analysis verified that CTN significantly down-regulated the expression of marker genes involved in neuron differentiation and synaptic signaling. CTN at the doses impairing cellular neurite outgrowth did not trigger mitochondrial oxidative stress and dysfunction. The neurotoxic mechanisms of CTN provide new information that is valuable in the assessment of CTN-related health risk for the general public.
Collapse
Affiliation(s)
- Jui-Feng Tsai
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ting-Shuan Wu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Feng-Yih Yu
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Biing-Hui Liu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
24
|
Shenoy A, Banerjee M, Upadhya A, Bagwe-Parab S, Kaur G. The Brilliance of the Zebrafish Model: Perception on Behavior and Alzheimer's Disease. Front Behav Neurosci 2022; 16:861155. [PMID: 35769627 PMCID: PMC9234549 DOI: 10.3389/fnbeh.2022.861155] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/21/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease (AD) has become increasingly prevalent in the elderly population across the world. It's pathophysiological markers such as overproduction along with the accumulation of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFT) are posing a serious challenge to novel drug development processes. A model which simulates the human neurodegenerative mechanism will be beneficial for rapid screening of potential drug candidates. Due to the comparable neurological network with humans, zebrafish has emerged as a promising AD model. This model has been thoroughly validated through research in aspects of neuronal pathways analogous to the human brain. The cholinergic, glutamatergic, and GABAergic pathways, which play a role in the manifested behavior of the zebrafish, are well defined. There are several behavioral models in both adult zebrafish and larvae to establish various aspects of cognitive impairment including spatial memory, associative memory, anxiety, and other such features that are manifested in AD. The zebrafish model eliminates the shortcomings of previously recognized mammalian models, in terms of expense, extensive assessment durations, and the complexity of imaging the brain to test the efficacy of therapeutic interventions. This review highlights the various models that analyze the changes in the normal behavioral patterns of the zebrafish when exposed to AD inducing agents. The mechanistic pathway adopted by drugs and novel therapeutic strategies can be explored via these behavioral models and their efficacy to slow the progression of AD can be evaluated.
Collapse
Affiliation(s)
| | | | | | | | - Ginpreet Kaur
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s Narsee Monjee Institute of Management Studies, Mumbai, India
| |
Collapse
|
25
|
Beppi C, Penner M, Straumann D, Bögli SY. A non-invasive biomechanical model of mild TBI in larval zebrafish. PLoS One 2022; 17:e0268901. [PMID: 35622781 PMCID: PMC9140253 DOI: 10.1371/journal.pone.0268901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/05/2022] [Indexed: 11/18/2022] Open
Abstract
A mild traumatic brain injury is a neurological dysfunction caused by biomechanical forces transmitted to the brain in physical impacts. The current understanding of the neuropathological cascade resulting in the manifested clinical signs and symptoms is limited due to the absence of sensitive brain imaging methods. Zebrafish are established models for the reproduction and study of neurobiological pathologies. However, all available models mostly recreate moderate-to-severe focal injuries in adult zebrafish. The present work has induced a mild brain trauma in larval zebrafish through a non-invasive biomechanical approach. A custom-made apparatus with a commercially available motor was employed to expose larvae to rapidly decelerating linear movements. The neurophysiological changes following concussion were assessed through behavioural quantifications of startle reflex locomotor distance and habituation metrics. Here we show that the injury was followed, within five minutes, by a transient anxiety state and CNS dysfunction manifested by increased startle responsivity with impaired startle habituation, putatively mirroring the human clinical sign of hypersensitivity to noise. Within a day after the injury, chronic effects arose, as evidenced by an overall reduced responsivity to sensory stimulation (lower amplitude and distance travelled along successive stimuli), reflecting the human post-concussive symptomatology. This study represents a step forward towards the establishment of a parsimonious (simple, less ethically concerning, yet sensitive) animal model of mild TBI. Our behavioural findings mimic aspects of acute and chronic effects of human concussion, which warrant further study at molecular, cellular and circuit levels. While our model opens wide avenues for studying the underlying cellular and molecular pathomechanisms, it also enables high-throughput testing of therapeutic interventions to accelerate post-concussive recovery.
Collapse
Affiliation(s)
- Carolina Beppi
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Swiss Concussion Center, Schulthess Clinic, Zurich, Switzerland
- * E-mail:
| | - Marco Penner
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Dominik Straumann
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Swiss Concussion Center, Schulthess Clinic, Zurich, Switzerland
| | - Stefan Yu Bögli
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Swiss Concussion Center, Schulthess Clinic, Zurich, Switzerland
| |
Collapse
|
26
|
Tayanloo-Beik A, Hamidpour SK, Abedi M, Shojaei H, Tavirani MR, Namazi N, Larijani B, Arjmand B. Zebrafish Modeling of Autism Spectrum Disorders, Current Status and Future Prospective. Front Psychiatry 2022; 13:911770. [PMID: 35911241 PMCID: PMC9329562 DOI: 10.3389/fpsyt.2022.911770] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Autism spectrum disorder (ASD) refers to a complicated range of childhood neurodevelopmental disorders which can occur via genetic or non-genetic factors. Clinically, ASD is associated with problems in relationships, social interactions, and behaviors that pose many challenges for children with ASD and their families. Due to the complexity, heterogeneity, and association of symptoms with some neuropsychiatric disorders such as ADHD, anxiety, and sleep disorders, clinical trials have not yielded reliable results and there still remain challenges in drug discovery and development pipeline for ASD patients. One of the main steps in promoting lead compounds to the suitable drug for commercialization is preclinical animal testing, in which the efficacy and toxicity of candidate drugs are examined in vivo. In recent years, zebrafish have been able to attract the attention of many researchers in the field of neurological disorders such as ASD due to their outstanding features. The presence of orthologous genes for ASD modeling, the anatomical similarities of parts of the brain, and similar neurotransmitter systems between zebrafish and humans are some of the main reasons why scientists draw attention to zebrafish as a prominent animal model in preclinical studies to discover highly effective treatment approaches for the ASD through genetic and non-genetic modeling methods.
Collapse
Affiliation(s)
- Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shayesteh Kokabi Hamidpour
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Abedi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamide Shojaei
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Nazli Namazi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
An Overview of Zebrafish Modeling Methods in Drug Discovery and Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1387:145-169. [PMID: 34961915 DOI: 10.1007/5584_2021_684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Animal studies are recognized as a significant step forward in the bridging between drug discovery and clinical applications. Animal models, due to their relative genetic, molecular, physiological, and even anatomical similarities to humans, can provide a suitable platform for unraveling the mechanisms underlying human diseases and discovering new therapeutic approaches as well. Recently, zebrafish has attracted attention as a valuable experimental and pharmacological model in drug discovery and development studies due to its prominent characteristics such as the high degree of genetic similarity with humans, genetic manipulability, and prominent clinical features. Since advancing a theory to a valid and reliable observation requires the manipulation of animals, it is, therefore, essential to use efficient modeling methods appropriate to the different aspects of experimental conditions. In this context, applying several various approaches such as using chemicals, pathogens, and genetic manipulation approaches allows zebrafish development into a preferable model that mimics some human disease pathophysiology. Thus, such modeling approaches not only can provide a framework for a comprehensive understanding of the human disease mechanisms that have a counterpart in zebrafish but also can pave the way for discovering new drugs that are accompanied by higher amelioration effects on different human diseases.
Collapse
|
28
|
Closset M, Cailliau K, Slaby S, Marin M. Effects of Aluminium Contamination on the Nervous System of Freshwater Aquatic Vertebrates: A Review. Int J Mol Sci 2021; 23:ijms23010031. [PMID: 35008450 PMCID: PMC8744726 DOI: 10.3390/ijms23010031] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 12/18/2022] Open
Abstract
Aluminium (Al) is the most common natural metallic element in the Earth’s crust. It is released into the environment through natural processes and human activities and accumulates in aquatic environments. This review compiles scientific data on the neurotoxicity of aluminium contamination on the nervous system of aquatic organisms. More precisely, it helps identify biomarkers of aluminium exposure for aquatic environment biomonitoring in freshwater aquatic vertebrates. Al is neurotoxic and accumulates in the nervous system of aquatic vertebrates, which is why it could be responsible for oxidative stress. In addition, it activates and inhibits antioxidant enzymes and leads to changes in acetylcholinesterase activity, neurotransmitter levels, and in the expression of several neural genes and nerve cell components. It also causes histological changes in nerve tissue, modifications of organism behaviour, and cognitive deficit. However, impacts of aluminium exposure on the early stages of aquatic vertebrate development are poorly described. Lastly, this review also poses the question of how accurate aquatic vertebrates (fishes and amphibians) could be used as model organisms to complement biological data relating to the developmental aspect. This “challenge” is very relevant since freshwater pollution with heavy metals has increased in the last few decades.
Collapse
Affiliation(s)
- Marie Closset
- University Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (M.C.); (K.C.)
| | - Katia Cailliau
- University Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (M.C.); (K.C.)
| | - Sylvain Slaby
- Normandie University, UNILEHAVRE, CNRS, UMR 3730 SCALE, Environmental Stress and Aquatic Biomonitoring (SEBIO), F-76600 Le Havre, France;
| | - Matthieu Marin
- University Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (M.C.); (K.C.)
- Correspondence:
| |
Collapse
|
29
|
Bauer B, Mally A, Liedtke D. Zebrafish Embryos and Larvae as Alternative Animal Models for Toxicity Testing. Int J Mol Sci 2021; 22:13417. [PMID: 34948215 PMCID: PMC8707050 DOI: 10.3390/ijms222413417] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 02/07/2023] Open
Abstract
Prerequisite to any biological laboratory assay employing living animals is consideration about its necessity, feasibility, ethics and the potential harm caused during an experiment. The imperative of these thoughts has led to the formulation of the 3R-principle, which today is a pivotal scientific standard of animal experimentation worldwide. The rising amount of laboratory investigations utilizing living animals throughout the last decades, either for regulatory concerns or for basic science, demands the development of alternative methods in accordance with 3R to help reduce experiments in mammals. This demand has resulted in investigation of additional vertebrate species displaying favourable biological properties. One prominent species among these is the zebrafish (Danio rerio), as these small laboratory ray-finned fish are well established in science today and feature outstanding biological characteristics. In this review, we highlight the advantages and general prerequisites of zebrafish embryos and larvae before free-feeding stages for toxicological testing, with a particular focus on cardio-, neuro, hepato- and nephrotoxicity. Furthermore, we discuss toxicokinetics, current advances in utilizing zebrafish for organ toxicity testing and highlight how advanced laboratory methods (such as automation, advanced imaging and genetic techniques) can refine future toxicological studies in this species.
Collapse
Affiliation(s)
- Benedikt Bauer
- Institute of Pharmacology and Toxicology, Julius-Maximilians-University, 97078 Würzburg, Germany; (B.B.); (A.M.)
| | - Angela Mally
- Institute of Pharmacology and Toxicology, Julius-Maximilians-University, 97078 Würzburg, Germany; (B.B.); (A.M.)
| | - Daniel Liedtke
- Institute of Human Genetics, Julius-Maximilians-University, 97074 Würzburg, Germany
| |
Collapse
|
30
|
Kakebeen AD, Niswander L. Micronutrient imbalance and common phenotypes in neural tube defects. Genesis 2021; 59:e23455. [PMID: 34665506 PMCID: PMC8599664 DOI: 10.1002/dvg.23455] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 12/24/2022]
Abstract
Neural tube defects (NTDs) are among the most common birth defects, with a prevalence of close to 19 per 10,000 births worldwide. The etiology of NTDs is complex involving the interplay of genetic and environmental factors. Since nutrient deficiency is a risk factor and dietary changes are the major preventative measure to reduce the risk of NTDs, a more detailed understanding of how common micronutrient imbalances contribute to NTDs is crucial. While folic acid has been the most discussed environmental factor due to the success that population-wide fortification has had on prevention of NTDs, folic acid supplementation does not prevent all NTDs. The imbalance of several other micronutrients has been implicated as risks for NTDs by epidemiological studies and in vivo studies in animal models. In this review, we highlight recent literature deciphering the multifactorial mechanisms underlying NTDs with an emphasis on mouse and human data. Specifically, we focus on advances in our understanding of how too much or too little retinoic acid, zinc, and iron alter gene expression and cellular processes contributing to the pathobiology of NTDs. Synthesis of the discussed literature reveals common cellular phenotypes found in embryos with NTDs resulting from several micronutrient imbalances. The goal is to combine knowledge of these common cellular phenotypes with mechanisms underlying micronutrient imbalances to provide insights into possible new targets for preventative measures against NTDs.
Collapse
Affiliation(s)
- Anneke Dixie Kakebeen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Lee Niswander
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
31
|
Melatonin Pretreatment Protects Against Status epilepticus, Glutamate Transport, and Oxidative Stress Induced by Kainic Acid in Zebrafish. Mol Neurobiol 2021; 59:266-275. [PMID: 34665406 DOI: 10.1007/s12035-021-02579-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/24/2021] [Indexed: 02/07/2023]
Abstract
Status epilepticus (SE) develops from abnormal electrical discharges, resulting in neuronal damage. Current treatments include antiepileptic drugs. However, the most common drugs used to treat seizures may sometimes be ineffective and have many side effects. Melatonin is an endogenous physiological hormone that is considered an alternative treatment for neurological disorders because of its free radical scavenging property. Thus, this study aimed to determine the effects of melatonin pretreatment on SE by inducing glutamatergic hyperstimulation in zebrafish. Seizures were induced in zebrafish using kainic acid (KA), a glutamate analog, and the seizure intensity was recorded for 60 min. Melatonin treatment for 7 days showed a decrease in seizure intensity (28%), latency to reach score 5 (14 min), and duration of SE (29%). In addition, melatonin treatment attenuated glutamate transporter levels, which significantly decreased in the zebrafish brain after 12 h of KA-induced seizures. Melatonin treatment reduced the increase in oxidative stress by reactive oxygen species formation through thiobarbituric acid reactive substances and 2',7'-dichiorofluorescin, induced by KA-seizure. An imbalance of antioxidant enzyme activities such as superoxide dismutase and catalase was influenced by melatonin and KA-induced seizures. Our study indicates that melatonin promotes a neuroprotective response against the epileptic profile in zebrafish. These effects could be related to the modulation of glutamatergic neurotransmission, recovery of glutamate uptake, and oxidative stress parameters in the zebrafish brain.
Collapse
|
32
|
Hubená P, Horký P, Slavík O. Fish self-awareness: limits of current knowledge and theoretical expectations. Anim Cogn 2021; 25:447-461. [PMID: 34655023 DOI: 10.1007/s10071-021-01566-5] [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: 02/25/2021] [Revised: 09/15/2021] [Accepted: 10/08/2021] [Indexed: 10/20/2022]
Abstract
Animal self-awareness is divided into three levels: bodily, social, and introspective self-awareness. Research has focused mainly on the introspection of so-called higher organisms such as mammals. Herein, we turn our attention to fish and provide opinions on their self-awareness based on a review of the scientific literature. Our specific aims are to discuss whether fish (A) could have a neural substrate supporting self-awareness and whether they display signs of (B) social and (C) introspective self-awareness. The present knowledge does not exclude the possibility that fish could have a simple neocortex or other structures that support certain higher cognitive processes, as the function of the primate cerebral cortex can be replaced by other neurological structures. Fish are known to display winner, loser, and audience effects, which could be interpreted as signs of social self-awareness. The audience effect may be explained not only by ethological cost and benefit theory but also by the concept of public self-awareness, which comes from human studies. The behavioural and neural manifestations of depression may be induced in fish under social subordination and may be viewed as certain awareness of a social status. The current findings on fish introspective self-awareness have been debated in the scientific community and, therefore, demand replication to provide more evidence. Further research is needed to verify the outlined ideas; however, the current knowledge indicates that fish are capable of certain higher cognitive processes, which raises questions and implications regarding ethics and welfare in fish-related research and husbandry.
Collapse
Affiliation(s)
- Pavla Hubená
- Department of Zoology and Fisheries, Czech University of Life Sciences Prague, Kamýcká 129, Prague 6, 165 00, Suchdol, Czech Republic.
| | - Pavel Horký
- Department of Zoology and Fisheries, Czech University of Life Sciences Prague, Kamýcká 129, Prague 6, 165 00, Suchdol, Czech Republic
| | - Ondřej Slavík
- Department of Zoology and Fisheries, Czech University of Life Sciences Prague, Kamýcká 129, Prague 6, 165 00, Suchdol, Czech Republic
| |
Collapse
|
33
|
Kim GHJ, Mo H, Liu H, Wu Z, Chen S, Zheng J, Zhao X, Nucum D, Shortland J, Peng L, Elepano M, Tang B, Olson S, Paras N, Li H, Renslo AR, Arkin MR, Huang B, Lu B, Sirota M, Guo S. A zebrafish screen reveals Renin-angiotensin system inhibitors as neuroprotective via mitochondrial restoration in dopamine neurons. eLife 2021; 10:69795. [PMID: 34550070 PMCID: PMC8457844 DOI: 10.7554/elife.69795] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/27/2021] [Indexed: 01/12/2023] Open
Abstract
Parkinson’s disease (PD) is a common neurodegenerative disorder without effective disease-modifying therapeutics. Here, we establish a chemogenetic dopamine (DA) neuron ablation model in larval zebrafish with mitochondrial dysfunction and robustness suitable for high-content screening. We use this system to conduct an in vivo DA neuron imaging-based chemical screen and identify the Renin-Angiotensin-Aldosterone System (RAAS) inhibitors as significantly neuroprotective. Knockdown of the angiotensin receptor 1 (agtr1) in DA neurons reveals a cell-autonomous mechanism of neuroprotection. DA neuron-specific RNA-seq identifies mitochondrial pathway gene expression that is significantly restored by RAAS inhibitor treatment. The neuroprotective effect of RAAS inhibitors is further observed in a zebrafish Gaucher disease model and Drosophila pink1-deficient PD model. Finally, examination of clinical data reveals a significant effect of RAAS inhibitors in delaying PD progression. Our findings reveal the therapeutic potential and mechanisms of targeting the RAAS pathway for neuroprotection and demonstrate a salient approach that bridges basic science to translational medicine. Parkinson’s disease is caused by the slow death and deterioration of brain cells, in particular of the neurons that produce a chemical messenger known as dopamine. Certain drugs can mitigate the resulting drop in dopamine levels and help to manage symptoms, but they cause dangerous side-effects. There is no treatment that can slow down or halt the progress of the condition, which affects 0.3% of the population globally. Many factors, both genetic and environmental, contribute to the emergence of Parkinson’s disease. For example, dysfunction of the mitochondria, the internal structures that power up cells, is a known mechanism associated with the death of dopamine-producing neurons. Zebrafish are tiny fish which can be used to study Parkinson’s disease, as they are easy to manipulate in the lab and share many characteristics with humans. In particular, they can be helpful to test the effects of various potential drugs on the condition. Here, Kim et al. established a new zebrafish model in which dopamine-producing brain cells die due to their mitochondria not working properly; they then used this assay to assess the impact of 1,403 different chemicals on the integrity of these cells. A group of molecules called renin-angiotensin-aldosterone (RAAS) inhibitors was shown to protect dopamine-producing neurons and stopped them from dying as often. These are already used to treat high blood pressure as they help to dilate blood vessels. In the brain, however, RAAS worked by restoring certain mitochondrial processes. Kim et al. then investigated whether these results are relevant in other, broader contexts. They were able to show that RAAS inhibitors have the same effect in other animals, and that Parkinson’s disease often progresses more slowly in patients that already take these drugs for high blood pressure. Taken together, these findings therefore suggest that RAAS inhibitors may be useful to treat Parkinson’s disease, as well as other brain illnesses that emerge because of mitochondria not working properly. Clinical studies and new ways to improve these drugs are needed to further investigate and capitalize on these potential benefits.
Collapse
Affiliation(s)
- Gha-Hyun J Kim
- Department of Bioengineering and Therapeutic Sciences and Programs in BiologicalSciences and Human Genetics, University of California, San Francisco, San Francisco, United States.,Graduate Program of Pharmaceutical Sciences and Pharmacogenomics, University of California, San Francisco, San Francisco, United States
| | - Han Mo
- Department of Bioengineering and Therapeutic Sciences and Programs in BiologicalSciences and Human Genetics, University of California, San Francisco, San Francisco, United States.,Tsinghua-Peking Center for Life Sciences, McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Harrison Liu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States.,Graduate Program of Bioengineering, University of California, San Francisco, San Francisco, United States
| | - Zhihao Wu
- Department of Pathology, Stanford University School of Medicine, Stanford, United States
| | - Steven Chen
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States.,Small Molecule Discovery Center, University of California, San Francisco, San Francisco, United States
| | - Jiashun Zheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - Xiang Zhao
- Department of Bioengineering and Therapeutic Sciences and Programs in BiologicalSciences and Human Genetics, University of California, San Francisco, San Francisco, United States
| | - Daryl Nucum
- Department of Bioengineering and Therapeutic Sciences and Programs in BiologicalSciences and Human Genetics, University of California, San Francisco, San Francisco, United States
| | - James Shortland
- Department of Bioengineering and Therapeutic Sciences and Programs in BiologicalSciences and Human Genetics, University of California, San Francisco, San Francisco, United States
| | - Longping Peng
- Department of Bioengineering and Therapeutic Sciences and Programs in BiologicalSciences and Human Genetics, University of California, San Francisco, San Francisco, United States.,Department of Cardiovascular Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mannuel Elepano
- Institute for Neurodegenerative Diseases (IND), UCSF Weill Institute forNeurosciences, University of California, San Francisco, San Francisco, United States
| | - Benjamin Tang
- Department of Pathology, Stanford University School of Medicine, Stanford, United States.,Institute for Neurodegenerative Diseases (IND), UCSF Weill Institute forNeurosciences, University of California, San Francisco, San Francisco, United States
| | - Steven Olson
- Small Molecule Discovery Center, University of California, San Francisco, San Francisco, United States.,Institute for Neurodegenerative Diseases (IND), UCSF Weill Institute forNeurosciences, University of California, San Francisco, San Francisco, United States
| | - Nick Paras
- Institute for Neurodegenerative Diseases (IND), UCSF Weill Institute forNeurosciences, University of California, San Francisco, San Francisco, United States
| | - Hao Li
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - Adam R Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States.,Small Molecule Discovery Center, University of California, San Francisco, San Francisco, United States
| | - Michelle R Arkin
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States.,Small Molecule Discovery Center, University of California, San Francisco, San Francisco, United States
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States.,Graduate Program of Bioengineering, University of California, San Francisco, San Francisco, United States.,Chan Zuckerberg Biohub, San Francisco, United States
| | - Bingwei Lu
- Department of Pathology, Stanford University School of Medicine, Stanford, United States
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, United States
| | - Su Guo
- Department of Bioengineering and Therapeutic Sciences and Programs in BiologicalSciences and Human Genetics, University of California, San Francisco, San Francisco, United States.,Graduate Program of Pharmaceutical Sciences and Pharmacogenomics, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
34
|
Di Mauro G, Rauti R, Casani R, Chimowa G, Galibert AM, Flahaut E, Cellot G, Ballerini L. Tuning the Reduction of Graphene Oxide Nanoflakes Differently Affects Neuronal Networks in the Zebrafish. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:2161. [PMID: 34578477 PMCID: PMC8468975 DOI: 10.3390/nano11092161] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 01/05/2023]
Abstract
The increasing engineering of biomedical devices and the design of drug-delivery platforms enriched by graphene-based components demand careful investigations of the impact of graphene-related materials (GRMs) on the nervous system. In addition, the enhanced diffusion of GRM-based products and technologies that might favor the dispersion in the environment of GRMs nanoparticles urgently requires the potential neurotoxicity of these compounds to be addressed. One of the challenges in providing definite evidence supporting the harmful or safe use of GRMs is addressing the variety of this family of materials, with GRMs differing for size and chemistry. Such a diversity impairs reaching a unique and predictive picture of the effects of GRMs on the nervous system. Here, by exploiting the thermal reduction of graphene oxide nanoflakes (GO) to generate materials with different oxygen/carbon ratios, we used a high-throughput analysis of early-stage zebrafish locomotor behavior to investigate if modifications of a specific GRM chemical property influenced how these nanomaterials affect vertebrate sensory-motor neurophysiology-exposing zebrafish to GO downregulated their swimming performance. Conversely, reduced GO (rGO) treatments boosted locomotor activity. We concluded that the tuning of single GRM chemical properties is sufficient to produce differential effects on nervous system physiology, likely interfering with different signaling pathways.
Collapse
Affiliation(s)
- Giuseppe Di Mauro
- Neuron Physiology and Technology Lab, International School for Advanced Studies (SISSA), Neuroscience, Via Bonomea 265, 34136 Trieste, Italy; (G.D.M.); (R.R.); (R.C.)
| | - Rossana Rauti
- Neuron Physiology and Technology Lab, International School for Advanced Studies (SISSA), Neuroscience, Via Bonomea 265, 34136 Trieste, Italy; (G.D.M.); (R.R.); (R.C.)
| | - Raffaele Casani
- Neuron Physiology and Technology Lab, International School for Advanced Studies (SISSA), Neuroscience, Via Bonomea 265, 34136 Trieste, Italy; (G.D.M.); (R.R.); (R.C.)
| | - George Chimowa
- CIRIMAT, UMR CNRS 5085, Université Toulouse Paul Sabatier, Bat. CIRIMAT, 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (G.C.); (A.M.G.); (E.F.)
| | - Anne Marie Galibert
- CIRIMAT, UMR CNRS 5085, Université Toulouse Paul Sabatier, Bat. CIRIMAT, 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (G.C.); (A.M.G.); (E.F.)
| | - Emmanuel Flahaut
- CIRIMAT, UMR CNRS 5085, Université Toulouse Paul Sabatier, Bat. CIRIMAT, 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (G.C.); (A.M.G.); (E.F.)
| | - Giada Cellot
- Neuron Physiology and Technology Lab, International School for Advanced Studies (SISSA), Neuroscience, Via Bonomea 265, 34136 Trieste, Italy; (G.D.M.); (R.R.); (R.C.)
| | - Laura Ballerini
- Neuron Physiology and Technology Lab, International School for Advanced Studies (SISSA), Neuroscience, Via Bonomea 265, 34136 Trieste, Italy; (G.D.M.); (R.R.); (R.C.)
| |
Collapse
|
35
|
Nasri A, Mezni A, Lafon PA, Wahbi A, Cubedo N, Clair P, Harrath AH, Beyrem H, Rossel M, Perrier V. Ethinylestradiol (EE2) residues from birth control pills impair nervous system development and swimming behavior of zebrafish larvae. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 770:145272. [PMID: 33497902 DOI: 10.1016/j.scitotenv.2021.145272] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 06/12/2023]
Abstract
The ubiquitous use of ethinylestradiol (EE2), an active constituent of birth control preparations, results in continuous release of this synthetic estrogen to surface waters. Many studies document the untoward effects of EE2 on the endocrine system of aquatic organisms. Effects of environmental EE2 on the nervous system are still poorly documented. We studied effects of pico- to nanomolar concentrations of EE2 on early nervous system development of zebrafish larvae. EE2 disrupted axonal nerve regeneration and hair cell regeneration up to 50%. Gene expression in larval brain tissues showed significantly upregulated expression of target genes, such as estrogen and progesterone receptors, and aromatase B. In contrast, downregulation of the tyrosine hydroxylase, involved in the synthesis of neurotransmitters, occurred concomitant with diminution of proliferating cells. Overall, the size of exposed fish larvae decreased by 25% and their swimming behavior was modified compared to non-treated larvae. EE2 interferes with nervous system development, both centrally and peripherally, with negative effects on regeneration and swimming behavior. Survival of fish and other aquatic species may be at risk in chronically EE2-contaminated environments.
Collapse
Affiliation(s)
- Ahmed Nasri
- Laboratory of Environment Biomonitoring, Unit of coastal Ecology and Ecotoxicology, Faculty of Sciences of Bizerte, University of Carthage, 7021, Zarzouna, Tunisia; U1198 MMDN (Molecular Mechanisms of Neurodegenerative Diseases), Inserm (National Institute for Health and Medical Research), MGX (Montpellier GenomiX), BioCampus, University of Montpellier, 34095, Montpellier, France.
| | - Ali Mezni
- Laboratory of Environment Biomonitoring, Unit of coastal Ecology and Ecotoxicology, Faculty of Sciences of Bizerte, University of Carthage, 7021, Zarzouna, Tunisia
| | - Pierre-André Lafon
- U1198 MMDN (Molecular Mechanisms of Neurodegenerative Diseases), Inserm (National Institute for Health and Medical Research), MGX (Montpellier GenomiX), BioCampus, University of Montpellier, 34095, Montpellier, France
| | - Aymen Wahbi
- Laboratory of Hetero-Organic Compounds and Nanostructured Materials (LR18ES11), Department of Chemistry, Faculty of Sciences of Bizerte, University of Carthage, 7021, Zarzouna, Tunisia
| | - Nicolas Cubedo
- U1198 MMDN (Molecular Mechanisms of Neurodegenerative Diseases), Inserm (National Institute for Health and Medical Research), MGX (Montpellier GenomiX), BioCampus, University of Montpellier, 34095, Montpellier, France
| | - Philippe Clair
- U1198 MMDN (Molecular Mechanisms of Neurodegenerative Diseases), Inserm (National Institute for Health and Medical Research), MGX (Montpellier GenomiX), BioCampus, University of Montpellier, 34095, Montpellier, France
| | - Abdel Halim Harrath
- King Saud University, Zoology Department, College of Science, Box 2455, Riyadh, 11451, Saudi Arabia
| | - Hamouda Beyrem
- Laboratory of Environment Biomonitoring, Unit of coastal Ecology and Ecotoxicology, Faculty of Sciences of Bizerte, University of Carthage, 7021, Zarzouna, Tunisia
| | - Mireille Rossel
- U1198 MMDN (Molecular Mechanisms of Neurodegenerative Diseases), Inserm (National Institute for Health and Medical Research), MGX (Montpellier GenomiX), BioCampus, University of Montpellier, 34095, Montpellier, France
| | - Véronique Perrier
- U1198 MMDN (Molecular Mechanisms of Neurodegenerative Diseases), Inserm (National Institute for Health and Medical Research), MGX (Montpellier GenomiX), BioCampus, University of Montpellier, 34095, Montpellier, France
| |
Collapse
|
36
|
Thompson WA, Vijayan MM. Zygotic exposure to venlafaxine disrupts cortisol stress axis activity in multiple generations of zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 274:116535. [PMID: 33524651 DOI: 10.1016/j.envpol.2021.116535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 06/12/2023]
Abstract
Ubiquitous use of antidepressants has resulted in increased concentrations of these pharmaceuticals in waterways receiving municipal wastewater effluent. Amongst these, venlafaxine, a selective serotonin and norepinephrine reuptake inhibitor, is commonly found at concentrations surpassing 1 ppb in surface waters. We recently showed that the deposition of venlafaxine in zebrafish (Danio rerio) embryos impacts neural development in the hypothalamus, suggesting the possibility of neuroendocrine disruptions due to this antidepressant. Here, we tested the hypothesis that early developmental exposure to venlafaxine disrupts the long-term functioning of the hypothalamus-pituitary-interrenal (HPI) axis in zebrafish. Embryos (1-4 cell stage) were injected with either 0, 1, or 10 ng venlafaxine, and the ontogeny of cortisol content, as well as changes in cortisol levels following a stressor in larvae and adults were assessed across 3 generations. Zygotic venlafaxine exposure did not affect the ontogeny of cortisol production, but there was a disruption in the cortisol response to stressor exposure, which was also evident in multiple generations. In the F0 generation, venlafaxine exposure did not affect cortisol levels in response to stressor exposure in larvae, but adult females, and not males, showed an attenuated cortisol response compared to control fish. This reduction in cortisol levels in the females was rescued by stimulation with adrenocorticotropic hormone, suggesting that the disruption was at the level of the hypothalamus-pituitary axis. Venlafaxine-mediated disruption in HPI axis functioning was also evident in the F1 and F2 generations, including impaired cortisol responses to a stressor in adult female and larval fish, respectively. Taken together, our results suggest that venlafaxine is an endocrine disruptor, and early developmental exposure to this antidepressant may have long-term and generational consequences on cortisol stress axis activity in zebrafish.
Collapse
Affiliation(s)
- William A Thompson
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| | - Mathilakath M Vijayan
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada.
| |
Collapse
|
37
|
Reimunde P, Pensado-López A, Carreira Crende M, Lombao Iglesias V, Sánchez L, Torrecilla-Parra M, Ramírez CM, Anfray C, Torres Andón F. Cellular and Molecular Mechanisms Underlying Glioblastoma and Zebrafish Models for the Discovery of New Treatments. Cancers (Basel) 2021; 13:1087. [PMID: 33802571 PMCID: PMC7961726 DOI: 10.3390/cancers13051087] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is the most common of all brain malignant tumors; it displays a median survival of 14.6 months with current complete standard treatment. High heterogeneity, aggressive and invasive behavior, the impossibility of completing tumor resection, limitations for drug administration and therapeutic resistance to current treatments are the main problems presented by this pathology. In recent years, our knowledge of GBM physiopathology has advanced significantly, generating relevant information on the cellular heterogeneity of GBM tumors, including cancer and immune cells such as macrophages/microglia, genetic, epigenetic and metabolic alterations, comprising changes in miRNA expression. In this scenario, the zebrafish has arisen as a promising animal model to progress further due to its unique characteristics, such as transparency, ease of genetic manipulation, ethical and economic advantages and also conservation of the major brain regions and blood-brain-barrier (BBB) which are similar to a human structure. A few papers described in this review, using genetic and xenotransplantation zebrafish models have been used to study GBM as well as to test the anti-tumoral efficacy of new drugs, their ability to interact with target cells, modulate the tumor microenvironment, cross the BBB and/or their toxicity. Prospective studies following these lines of research may lead to a better diagnosis, prognosis and treatment of patients with GBM.
Collapse
Affiliation(s)
- Pedro Reimunde
- Department of Medicine, Campus de Oza, Universidade da Coruña, 15006 A Coruña, Spain
- Department of Neurosurgery, Hospital Universitario Lucus Augusti, 27003 Lugo, Spain
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Martín Carreira Crende
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Vanesa Lombao Iglesias
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Marta Torrecilla-Parra
- IMDEA Research Institute of Food and Health Sciences, 28049 Madrid, Spain; (M.T.-P.); (C.M.R.)
| | - Cristina M. Ramírez
- IMDEA Research Institute of Food and Health Sciences, 28049 Madrid, Spain; (M.T.-P.); (C.M.R.)
| | - Clément Anfray
- IRCCS Istituto Clinico Humanitas, Via A. Manzoni 56, 20089 Rozzano, Milan, Italy;
| | - Fernando Torres Andón
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
- IRCCS Istituto Clinico Humanitas, Via A. Manzoni 56, 20089 Rozzano, Milan, Italy;
| |
Collapse
|
38
|
Chen X, Li Y, Yao T, Jia R. Benefits of Zebrafish Xenograft Models in Cancer Research. Front Cell Dev Biol 2021; 9:616551. [PMID: 33644052 PMCID: PMC7905065 DOI: 10.3389/fcell.2021.616551] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
As a promising in vivo tool for cancer research, zebrafish have been widely applied in various tumor studies. The zebrafish xenograft model is a low-cost, high-throughput tool for cancer research that can be established quickly and requires only a small sample size, which makes it favorite among researchers. Zebrafish patient-derived xenograft (zPDX) models provide promising evidence for short-term clinical treatment. In this review, we discuss the characteristics and advantages of zebrafish, such as their transparent and translucent features, the use of vascular fluorescence imaging, the establishment of metastatic and intracranial orthotopic models, individual pharmacokinetics measurements, and tumor microenvironment. Furthermore, we introduce how these characteristics and advantages are applied other in tumor studies. Finally, we discuss the future direction of the use of zebrafish in tumor studies and provide new ideas for the application of it.
Collapse
Affiliation(s)
- Xingyu Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Tengteng Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
39
|
Zhang K, Liang J, Brun NR, Zhao Y, Werdich AA. Rapid Zebrafish Behavioral Profiling Assay Accelerates the Identification of Environmental Neurodevelopmental Toxicants. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2021; 55:1919-1929. [PMID: 33470099 DOI: 10.1021/acs.est.0c06949] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Rapid and cost-effective in vivo assays to screen potential environmental neurodevelopmental toxicants are necessary to address the limitations of in vitro platforms, such as the inability to fully recapitulate the developmental and physiological processes of whole organisms. In the present study, a rapid zebrafish behavioral profiling assay was developed to characterize the neurodevelopmental effects of environmental substances by quantitatively evaluating multiple spontaneous movement features of zebrafish embryos. This video analysis-based assay automatically segmented every embryo and thus was able to accurately quantify spontaneous movement features, including frequency, duration, intensity, interval, and the number of continuous movements. When tested with eight environmental substances known to be neurodevelopmental toxicants, such as chlorpyrifos and bisphenol A, the assay successfully captured frequency alterations that were well-documented in previous studies while also providing additional information. Using an optimized procedure, we further assessed 132 potential neurotoxins that spanned a wide range of molecular targets, many of which were previously detected in environmental waterbodies. The distinct altered behavioral barcodes indicated that the spontaneous movement was impacted by diverse neuroactive substances, and the effects could be effectively evaluated with the developed assay. The web-based tool, named EMAnalysis, is further provided at http://www.envh.sjtu.edu.cn/zebrafish_contraction.jsp. Thus, this assay provides an efficient platform to accelerate the pace of neurotoxic environmental contaminant discoveries.
Collapse
Affiliation(s)
- Kun Zhang
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
- Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, Massachusetts 02115, United States
| | - Jiahui Liang
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Nadja R Brun
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, United States
| | - Yanbin Zhao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
- Shanghai Institute of Pollution Control and Ecological Security, Shanghai 200092, China
| | - Andreas A Werdich
- Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, Massachusetts 02115, United States
| |
Collapse
|
40
|
Li M, Yang T, Gao L, Xu H. An inadvertent issue of human retina exposure to endocrine disrupting chemicals: A safety assessment. CHEMOSPHERE 2021; 264:128484. [PMID: 33022499 DOI: 10.1016/j.chemosphere.2020.128484] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/07/2020] [Accepted: 09/27/2020] [Indexed: 06/11/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are a group of chemical compounds that present a considerable public health problem due to their pervasiveness and associations with chronic diseases. EDCs can interrupt the endocrine system and interfere with hormone homeostasis, leading to abnormalities in human physiology. Much attention has been focused on the adverse effects EDCs have on the reproductive system, neurogenesis, neuroendocrine system, and thyroid dysfunction. The eye is usually directly exposed to the surrounding environment; however, the influences of EDCs on the eye have received comparatively little attention. Ocular diseases, such as ocular surface diseases and retinal diseases, have been implicated in hormone deficiency or excess. Epidemiologic studies have shown that EDC exposure not only causes ocular surface disorders, such as dry eye, but also associates with visual deficits and retinopathy. EDCs can pass through the human blood-retinal barrier and enter the neural retina, and can then accumulate in the retina. The retina is an embryologic extension of the central nervous system, and is extremely sensitive and vulnerable to EDCs that could be passed across the placenta during critical periods of retinal development. Subtle alterations in the retinal development process usually result in profound immediate, long-term, and delayed effects late in life. This review, based on extensive literature survey, briefly summarizes the current knowledge about the impact of representative manufactured EDCs on retinal toxicity, including retinal structure alterations and dysfunction. We also highlight the potential mechanism of action of EDCs on the retina, and the predictive retinal models of EDC exposure.
Collapse
Affiliation(s)
- Minghui Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Tian Yang
- Department of Cold Environmental Medicine, College of High Altitude Military Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lixiong Gao
- Department of Ophthalmology, Third Medical Center of PLA General Hospital, Beijing, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China.
| |
Collapse
|
41
|
A model-based quantification of startle reflex habituation in larval zebrafish. Sci Rep 2021; 11:846. [PMID: 33436805 PMCID: PMC7804396 DOI: 10.1038/s41598-020-79923-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/14/2020] [Indexed: 01/29/2023] Open
Abstract
Zebrafish is an established animal model for the reproduction and study of neurobiological pathogenesis of human neurological conditions. The 'startle reflex' in zebrafish larvae is an evolutionarily preserved defence response, manifesting as a quick body-bend in reaction to sudden sensory stimuli. Changes in startle reflex habituation characterise several neuropsychiatric disorders and hence represent an informative index of neurophysiological health. This study aimed at establishing a simple and reliable experimental protocol for the quantification of startle reflex response and habituation. The fish were stimulated with 20 repeated pulses of specific vibratory frequency, acoustic intensity/power, light-intensity and interstimulus-interval, in three separate studies. The cumulative distance travelled, namely the sum of the distance travelled (mm) during all 20 stimuli, was computed as a group-level description for all the experimental conditions in each study. Additionally, by the use of bootstrapping, the data was fitted to a model of habituation with a first-order exponential representing the decay of locomotor distance travelled over repeated stimulation. Our results suggest that startle habituation is a stereotypic first-order process with a decay constant ranging from 1 to 2 stimuli. Habituation memory lasts no more than 5 min, as manifested by the locomotor activity recovering to baseline levels. We further observed significant effects of vibratory frequency, acoustic intensity/power and interstimulus-interval on the amplitude, offset, decay constant and cumulative distance travelled. Instead, the intensity of the flashed light did not contribute to significant behavioural variations. The findings provide novel insights as to the influence of different stimuli parameters on the startle reflex habituation and constitute a helpful reference framework for further investigation.
Collapse
|
42
|
Campos-Rodriguez C, Fredrick E, Ramirez-San Juan E, Olsson R. Enantiomeric N-substituted phthalimides with excitatory amino acids protect zebrafish larvae against PTZ-induced seizures. Eur J Pharmacol 2020; 888:173489. [DOI: 10.1016/j.ejphar.2020.173489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 10/23/2022]
|
43
|
Maphanga VB, Skalicka-Woźniak K, Budzynska B, Enslin GM, Viljoen AM. Screening selected medicinal plants for potential anxiolytic activity using an in vivo zebrafish model. Psychopharmacology (Berl) 2020; 237:3641-3652. [PMID: 32840669 DOI: 10.1007/s00213-020-05642-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 08/13/2020] [Indexed: 02/03/2023]
Abstract
RATIONALE Medicinal plants are used extensively in many countries to treat conditions related to the central nervous system (CNS), and there is renewed interest to explore natural products, which may exhibit CNS activity. OBJECTIVE In this study, seven plants were selected based on literature reports of their ethnopharmacological use in treating anxiety-related conditions and assayed in a zebrafish model. METHODS Crude extracts were prepared with solvents of different polarities, and the maximum tolerated concentration (MTC) of these crude extracts was established. The anxiolytic activity of the crude extracts was determined using 5-day post-fertilization (dpf) zebrafish larvae. General locomotor activity and reverse-thigmotaxis behavior (indicative of anxiolytic activity) were analyzed under continuous illumination and alternating light-dark challenges, which induced anxiety in the zebrafish larvae. RESULTS Of the 28 extracts tested, 13 were toxic according to the MTC values obtained. Larvae were subsequently treated with the 15 non-toxic extracts, at a dose determined in the MTC assay or with 1% DMSO as control. The anxiolytic activity (reverse-thigmotaxis) was demonstrated by an increase in the percentage time spent by the larvae in the central arena of the well. Of the 15 non-toxic extracts tested, the Sceletium tortuosum water extract exhibited the most promising anxiolytic activity. CONCLUSIONS The zebrafish model was effective in studying anxiety-related behavior. Thus, the study confirmed that S. tortuosum mitigates anxiety in zebrafish larvae, a step towards the full in vivo validation of the traditional use of the plant.
Collapse
Affiliation(s)
- Veronica B Maphanga
- Department of Pharmaceutical Sciences, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
| | - Krystyna Skalicka-Woźniak
- Department of Pharmacognosy with Medicinal Plant Unit, Medical University of Lublin, 1 Chodzki Street, 20-093, Lublin, Poland
| | - Barbara Budzynska
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, Jaczewskiego 4, 20-090, Lublin, Poland
| | - Gill M Enslin
- Department of Pharmaceutical Sciences, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
| | - Alvaro M Viljoen
- Department of Pharmaceutical Sciences, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa. .,SAMRC Herbal Drugs Research Unit, Tshwane University of Technology, Pretoria, 0001, South Africa.
| |
Collapse
|
44
|
Pensado-López A, Veiga-Rúa S, Carracedo Á, Allegue C, Sánchez L. Experimental Models to Study Autism Spectrum Disorders: hiPSCs, Rodents and Zebrafish. Genes (Basel) 2020; 11:E1376. [PMID: 33233737 PMCID: PMC7699923 DOI: 10.3390/genes11111376] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/26/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023] Open
Abstract
Autism Spectrum Disorders (ASD) affect around 1.5% of the global population, which manifest alterations in communication and socialization, as well as repetitive behaviors or restricted interests. ASD is a complex disorder with known environmental and genetic contributors; however, ASD etiology is far from being clear. In the past decades, many efforts have been put into developing new models to study ASD, both in vitro and in vivo. These models have a lot of potential to help to validate some of the previously associated risk factors to the development of the disorder, and to test new potential therapies that help to alleviate ASD symptoms. The present review is focused on the recent advances towards the generation of models for the study of ASD, which would be a useful tool to decipher the bases of the disorder, as well as to conduct drug screenings that hopefully lead to the identification of useful compounds to help patients deal with the symptoms of ASD.
Collapse
Affiliation(s)
- Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (S.V.-R.)
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Sara Veiga-Rúa
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (S.V.-R.)
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Ángel Carracedo
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), CIMUS, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Catarina Allegue
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (S.V.-R.)
| |
Collapse
|
45
|
Guo SY, Zhang Y, Zhu XY, Zhou JL, Li J, Li CQ, Wu LR. Developmental neurotoxicity and toxic mechanisms induced by olaquindox in zebrafish. J Appl Toxicol 2020; 41:549-560. [PMID: 33111391 DOI: 10.1002/jat.4062] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 12/30/2022]
Abstract
Olaquindox (OLA) has been widely used as an animal feed additive in China for decades; however, its toxicity and toxic mechanisms have not been well investigated. In this study, the developmental neurotoxicity and toxic mechanisms of OLA were evaluated in zebrafish. Zebrafish embryos were exposed to different concentrations of OLA (25-1,000 mg/L) from 6 to 120 hours post fertilization (hpf). OLA exposure resulted in many abnormal phenotypes in zebrafish, including shortened body length, notochord degeneration, spinal curvature, brain apoptosis, damage of axon and peripheral motor neuron, and hepatotoxicity. Interestingly, OLA increased zebrafish spontaneous tail coiling, while reduced locomotor capacity. Quantitative polymerase chain reaction (Q-PCR) showed that the expression levels of nine marker genes for nervous system functions or development, namely, α1-tubulin, glial fibrillary acidic protein (gfap), myelin basic protein (mbp), synapsinII a (syn2a), sonic hedgehog a (shha), encoding HuC (elavl3), mesencephalic astrocyte-derived neurotrophic factor (manf) growth associated protein 43 (gap43), and acetylcholinesterase (ache) were all down-regulated significantly in zebrafish after treated with OLA. Besides, the anti-apoptotic and pro-apoptotic genes bcl-2/bax ratio was reduced. These results show that OLA exposure could cause severe developmental neurotoxicity in the early stages of zebrafish life and OLA might induce neurotoxicity by inhibiting the expression of neuro-developmental genes and promoting apoptosis.
Collapse
Affiliation(s)
- Sheng-Ya Guo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Binwen Road, Hangzhou, 310053, China.,Research and Development Department, Hunter Biotechnology, Inc., Jiangling Road, Hangzhou, 310051, China
| | - Yong Zhang
- Research and Development Department, Hunter Biotechnology, Inc., Jiangling Road, Hangzhou, 310051, China
| | - Xiao-Yu Zhu
- Research and Development Department, Hunter Biotechnology, Inc., Jiangling Road, Hangzhou, 310051, China
| | - Jia-Li Zhou
- Research and Development Department, Hunter Biotechnology, Inc., Jiangling Road, Hangzhou, 310051, China
| | - Jiao Li
- Research and Development Department, Hunter Biotechnology, Inc., Jiangling Road, Hangzhou, 310051, China
| | - Chun-Qi Li
- Research and Development Department, Hunter Biotechnology, Inc., Jiangling Road, Hangzhou, 310051, China.,Research and Development Department, New Hunter Testing and Technology Co., Ltd, Xinjinhu Road, Nanjing, 210046, China
| | - Li-Ren Wu
- Laboratory Animal Regulatory Center, Hangzhou Medical College, Tianmushan Road, Hangzhou, 310013, China
| |
Collapse
|
46
|
He Y, Tsang KF, Kong RYC, Chow YT. Indication of Electromagnetic Field Exposure via RBF-SVM Using Time-Series Features of Zebrafish Locomotion. SENSORS 2020; 20:s20174818. [PMID: 32858993 PMCID: PMC7506915 DOI: 10.3390/s20174818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 12/23/2022]
Abstract
This paper introduces a novel model based on support vector machine with radial basis function kernel (RBF-SVM) using time-series features of zebrafish (Danio rerio) locomotion exposed to different electromagnetic fields (EMFs) to indicate the corresponding EMF exposure. A group of 14 adult zebrafish was randomly divided into two groups, 7 in each group; the fish of each group have the novel tank test under a sham or real magnetic exposure of 6.78 MHz and about 1 A/m. Their locomotion in the tests was videotaped to convert into the x, y coordinate time-series of the trajectories for reforming time-series matrices according to different time-series lengths. The time-series features of zebrafish locomotion were calculated by the comparative time-series analyzing framework highly comparative time-series analysis (HCTSA), and a limited number of the time-series features that were most relevant to the EMF exposure conditions were selected using the minimum redundancy maximum relevance (mRMR) algorithm for RBF-SVM classification training. Before this, ambient environmental parameters (AEPs) had little effect on the locomotion performance of zebrafish processed by the empirical method, which had been quantitatively verified by regression using another group of 14 adult zebrafish. The results have demonstrated that the purposed model is capable of accurately indicating different EMF exposures. All classification accuracies can be 100%, and the classification precision of several classifiers based on specific parameters and feature sets with specific dimensions can reach higher than 95%. The speculative reason for this result is that the specified EMF has affected the zebrafish neural aspect, which is then reflected in their behaviors. The outcomes of this study have provided a new indication model for EMF exposures and provided a reference for the investigation of the impact of EMF exposure.
Collapse
Affiliation(s)
- Yaqing He
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong 999077, China; (K.F.T.); (Y.-T.C.)
- Correspondence:
| | - Kim Fung Tsang
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong 999077, China; (K.F.T.); (Y.-T.C.)
| | - Richard Yuen-Chong Kong
- Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong 999077, China;
- Futian-CityU Mangrove Research & Development Centre (FCMC), Futian National Nature Reserve, Mangrove Road, Shenzhen 518040, China
| | - Yuk-Tak Chow
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong 999077, China; (K.F.T.); (Y.-T.C.)
| |
Collapse
|
47
|
Liu Y, Zhang T, Li G, Li S, Li J, Zhao Q, Wu Q, Xu D, Hu X, Zhang L, Li Q, Zhang H, Liu B. Radiosensitivity enhancement by Co-NMS-mediated mitochondrial impairment in glioblastoma. J Cell Physiol 2020; 235:9623-9634. [PMID: 32394470 DOI: 10.1002/jcp.29774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 02/06/2023]
Abstract
We investigated the radiosensitizing effects of Co-NMS, a derivative of nimesulide based on a cobalt carbonyl complex, on malignant glioma cells. In the zebrafish exposed to Co-NMS ranging from 5 to 20 μM, cell death and heat shock protein 70 expression in the brain and neurobehavioral performance were evaluated. Our data showed that Co-NMS at 5 μM did not cause the appreciable neurotoxicity, and thereby was given as a novel radiation sensitizer in further study. In the U251 cells, Co-NMS combined with irradiation treatment resulted in significant inhibition of cell growth and clonogenic capability as well as remarkable increases of G2/M arrest and apoptotic cell population compared to the irradiation alone treatment. This demonstrated that the Co-NMS administration exerted a strong potential of sensitizing effect on the irradiated cells. With regard to the tumor radiosensitization of Co-NMS, it could be primarily attributed to the Co-NMS-derived mitochondrial impairment, reflected by the loss of mitochondrial membrane potential, the disruption of mitochondrial fusion and fission balance as well as redox homeostasis. Furthermore, the energy metabolism of the U251 cells was obviously suppressed by cotreatment with Co-NMS and irradiation through repressing mitochondrial function. Taken together, our findings suggested that Co-NMS could be a desirable drug to enhance the radiotherapeutic effects in glioblastoma patients.
Collapse
Affiliation(s)
- Yang Liu
- Medical Physics Division, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Medicine, Chinese Academy of Sciences, Lanzhou, China.,School of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Taofeng Zhang
- Institute of Radiochemistry, School of Nuclear Science and Technology, Lanzhou University, Lanzhou, China
| | - Guo Li
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Sirui Li
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Jili Li
- Institute of Medicinal Chemistry, School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Quanyi Zhao
- Institute of Medicinal Chemistry, School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Qingfen Wu
- Medical Physics Division, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Medicine, Chinese Academy of Sciences, Lanzhou, China.,School of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Dan Xu
- Medical Physics Division, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Medicine, Chinese Academy of Sciences, Lanzhou, China.,School of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoli Hu
- Medical Physics Division, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Medicine, Chinese Academy of Sciences, Lanzhou, China.,School of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Luwei Zhang
- Medical Physics Division, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Medicine, Chinese Academy of Sciences, Lanzhou, China.,School of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Qiang Li
- Medical Physics Division, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Medicine, Chinese Academy of Sciences, Lanzhou, China.,School of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Hong Zhang
- Medical Physics Division, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Medicine, Chinese Academy of Sciences, Lanzhou, China.,School of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Bin Liu
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| |
Collapse
|
48
|
Aponte A, Petrunich-Rutherford ML. Acute net stress of young adult zebrafish ( Danio rerio) is not sufficient to increase anxiety-like behavior and whole-body cortisol. PeerJ 2019; 7:e7469. [PMID: 31410315 PMCID: PMC6689218 DOI: 10.7717/peerj.7469] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/12/2019] [Indexed: 12/28/2022] Open
Abstract
In recent years, the zebrafish (Danio rerio) has become a popular model to study the mechanisms of physiological and behavioral effects of stress, due to the similarity in neural structures and biochemical pathways between zebrafish and mammals. Previous research in this vertebrate animal model has demonstrated an increase in whole-body cortisol resulting from an acute (30-second) net handling stress, but it remains unclear whether such a stressor will concomitantly increase anxiety-like behavior. In addition, as the previous study examined the effects of this acute stressor in adult zebrafish after a brief period of isolation, it is unclear whether this stressor would be effective in eliciting cortisol increases in younger aged subjects without isolation. In the current study, young adult zebrafish (approximately 90 days post-fertilization) were briefly exposed to a net handling stressor and were subsequently subjected to either the novel tank test or the light/dark preference test. The novel tank test was used to measure exploration and habituation in response to a novel environment, and the light/dark preference test was used to measure locomotor activity and scototaxis behavior. All subjects were sacrificed 15 minutes post-stressor and were analyzed for whole-body levels of cortisol. Contrary to expectations, there was no effect of acute net handling on cortisol levels. Similarly, acute net handling did not significantly induce anxiety-like behavior during the novel tank test or the light/dark preference test. Our findings demonstrate that there are possible developmental differences in response to an acute net handling stress, as we did not observe alterations in hormonal or behavioral measures of anxiety in young adult zebrafish. Alternatively, if zebrafish are not isolated before the stressor, they may be more resilient to a brief acute stressor. These results suggest the need for a different or more intense acute stressor in order further explore neuroendocrine mechanisms and anxiety-like behavior at this developmental stage in the zebrafish animal model.
Collapse
Affiliation(s)
- Amy Aponte
- Department of Psychology, Indiana University Northwest, Gary, IN, United States of America
| | | |
Collapse
|
49
|
Gawel K, Banono NS, Michalak A, Esguerra CV. A critical review of zebrafish schizophrenia models: Time for validation? Neurosci Biobehav Rev 2019; 107:6-22. [PMID: 31381931 DOI: 10.1016/j.neubiorev.2019.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/02/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022]
Abstract
Schizophrenia is a mental disorder that affects 1% of the population worldwide and is manifested as a broad spectrum of symptoms, from hallucinations to memory impairment. It is believed that genetic and/or environmental factors may contribute to the occurrence of this disease. Recently, the zebrafish has emerged as a valuable and attractive model for various neurological disorders including schizophrenia. In this review, we describe current pharmacological models of schizophrenia with special emphasis on providing insights into the pros and cons of using zebrafish as a behavioural model of this disease. Moreover, we highlight the advantages and utility of using zebrafish for elucidating the genetic mechanisms underlying this psychiatric disorder. We believe that the zebrafish has high potential also in the area of precision medicine and may complement the development of therapeutics, especially for pharmacoresistant patients.
Collapse
Affiliation(s)
- Kinga Gawel
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway, University of Oslo, Gaustadalléen 21, 0349, Oslo, Norway; Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego St. 8b, 20-090, Lublin, Poland.
| | - Nancy Saana Banono
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway, University of Oslo, Gaustadalléen 21, 0349, Oslo, Norway
| | - Agnieszka Michalak
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki St. 4A, 20-093, Lublin, Poland
| | - Camila V Esguerra
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway, University of Oslo, Gaustadalléen 21, 0349, Oslo, Norway; Department of Pharmacy, University of Oslo, Oslo, Norway.
| |
Collapse
|
50
|
Franco MB, Andrade TS, Sousa-Moura D, Lopes da Silva M, Machado Ferraz IB, Camargo NS, Domingues I, Oliveira R, Grisolia CK. Exposure to dilute concentrations of bupropion affects zebrafish early life stages. CHEMOSPHERE 2019; 222:175-183. [PMID: 30708151 DOI: 10.1016/j.chemosphere.2019.01.141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/21/2019] [Accepted: 01/24/2019] [Indexed: 06/09/2023]
Abstract
Psychiatric pharmaceuticals are one of the most prescribed active substances globally. Bupropion (BPP) is an antidepressant that acts via inhibition of norepinephrine and dopamine reuptake. It has been found in various water matrices, and thus its effects on aquatic organisms must be studied. The present study aimed to evaluate the acute toxic effects of BPP on zebrafish (Danio rerio) early life stages. For developmental analysis, organisms were exposed for 168 h to concentrations ranging from 0 to 82000 μg/L. Two other experiments were performed by exposing embryos to a wide range of concentrations (from 0 to 50000 μg/L) in order to evaluate BPP effects on embryonic behavior, using the Zebrabox and testing at the biochemical level (acetylcholinesterase, glutathione-S-transferase, lactate dehydrogenase and catalase). Developmental analysis indicated that BPP had low acute toxicity with a calculated 168 h-LC50 of 50346 μg/L. Concentrations equal to or above 44800 μg/L elicited several effects such as hatching delay, edemas and tail deformities. However, concentrations from 7300 μg/L upwards elicited equilibrium alteration. Behavioral analysis showed that BPP affected zebrafish locomotor behavior by decreasing activity at 0.6 μg/L, increasing activity at 8.8 and 158 μg/L, and decreasing activity at 50000 μg/L. Biochemical analysis showed an increase of AChE activity at 158 and 2812 μg/L, an increase in GST at the highest concentrations, CAT alteration and increase of LDH at 0.6, 2812 and 50000 μg/L. We can conclude that BPP affects zebrafish early life stages at environmental concentrations.
Collapse
Affiliation(s)
- Mariana Bernardes Franco
- Laboratório de Genética Toxicológica, Departamento de Genética e Morfologia, Instituto de Ciências Biológicas, Universidade de Brasília, UnB, 70910-900, Brasília, Distrito Federal, Brazil; Faculdade da Ceilândia, Universidade de Brasília, 72220-90, Brasília, Distrito Federal, Brazil.
| | - Thayres S Andrade
- Laboratório de Genética Toxicológica, Departamento de Genética e Morfologia, Instituto de Ciências Biológicas, Universidade de Brasília, UnB, 70910-900, Brasília, Distrito Federal, Brazil; Universidade Federal do Ceará, UFC, Campus de Crateús, 63700-000, Crateús, Ceará, Brazil.
| | - Diego Sousa-Moura
- Laboratório de Genética Toxicológica, Departamento de Genética e Morfologia, Instituto de Ciências Biológicas, Universidade de Brasília, UnB, 70910-900, Brasília, Distrito Federal, Brazil.
| | - Muriel Lopes da Silva
- Laboratório de Genética Toxicológica, Departamento de Genética e Morfologia, Instituto de Ciências Biológicas, Universidade de Brasília, UnB, 70910-900, Brasília, Distrito Federal, Brazil.
| | - Irvin Bryan Machado Ferraz
- Laboratório de Genética Toxicológica, Departamento de Genética e Morfologia, Instituto de Ciências Biológicas, Universidade de Brasília, UnB, 70910-900, Brasília, Distrito Federal, Brazil.
| | - Níchollas Serafim Camargo
- Laboratório de Genética Toxicológica, Departamento de Genética e Morfologia, Instituto de Ciências Biológicas, Universidade de Brasília, UnB, 70910-900, Brasília, Distrito Federal, Brazil; Faculdade da Ceilândia, Universidade de Brasília, 72220-90, Brasília, Distrito Federal, Brazil.
| | - Inês Domingues
- Departamento de Biologia & CESAM, Universidade de Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Rhaul Oliveira
- Laboratório de Genética Toxicológica, Departamento de Genética e Morfologia, Instituto de Ciências Biológicas, Universidade de Brasília, UnB, 70910-900, Brasília, Distrito Federal, Brazil; Faculdade de Tecnologia, Universidade Estadual de Campinas, UNICAMP, 13484-332, Limeira, São Paulo, Brazil; Programa de Pós-graduação em Toxicologia e Análises Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, USP, 05508-000, Butantã, São Paulo, Brazil.
| | - Cesar Koppe Grisolia
- Laboratório de Genética Toxicológica, Departamento de Genética e Morfologia, Instituto de Ciências Biológicas, Universidade de Brasília, UnB, 70910-900, Brasília, Distrito Federal, Brazil.
| |
Collapse
|