1
|
McGraw CM, Poduri A. Machine learning enables high-throughput, low-replicate screening for novel anti-seizure targets and compounds using combined movement and calcium fluorescence in larval zebrafish. Eur J Pharmacol 2025; 991:177327. [PMID: 39914783 DOI: 10.1016/j.ejphar.2025.177327] [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: 09/24/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/12/2025]
Abstract
Identifying new anti-seizure medications (ASMs) is difficult due to limitations in animal-based assays. Zebrafish (Danio rerio) serve as a model for chemical and genetic seizures, but current methods for detecting anti-seizure responses are limited by incomplete detection of anti-seizure responses (locomotor assays) or low-throughput (electrophysiology, fluorescence microscopy). To overcome these challenges, we developed a novel high-throughput method using combined locomotor and calcium fluorescence data from unrestrained larval zebrafish in a 96-well plate reader. Custom software tracked fish movement and fluorescence changes (deltaF/F0) from high-speed time-series, and logistic classifiers trained with elastic net regression distinguished seizure-like activity in response to the GABAA receptor antagonist pentylenetetrazole (PTZ). A classifier using combined data ("PTZ M + F"; AUC-ROC: 0.98; F1: 0.912) outperformed movement-only ("PTZ M"; AUC-ROC: 0.9) and fluorescence-only classifiers ("PTZ F"; AUC-ROC 0.96). Seizure-like event rate increased in proportion to PTZ concentration, and was suppressed by valproic acid (VPA). Meanwhile, TGB selectively reduced events defined by the "PTZ M + F″ classifier, paralleling previous reports that TGB reduces electrographic but not locomotor seizures. Using bootstrap simulation, we calculated statistical power and demonstrated reliable detection of ASM effects with as few as N = 4 replicates. In a test screen, 4 out of 5 ASMs were detected. This high-throughput approach combines previously orthogonal assays for zebrafish ASM screening.
Collapse
Affiliation(s)
- Christopher Michael McGraw
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Neurology, Feinberg School of Medicine, Northwestern University, USA.
| | - Annapurna Poduri
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA; National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
2
|
Karimi-Sani I, Sharifi M, Abolpour N, Lotfi M, Atapour A, Takhshid MA, Sahebkar A. Drug repositioning for Parkinson's disease: An emphasis on artificial intelligence approaches. Ageing Res Rev 2025; 104:102651. [PMID: 39755176 DOI: 10.1016/j.arr.2024.102651] [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: 10/08/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
Parkinson's disease (PD) is one of the most incapacitating neurodegenerative diseases (NDDs). PD is the second most common NDD worldwide which affects approximately 1-2 percent of people over 65 years. It is an attractive pursuit for artificial intelligence (AI) to contribute to and evolve PD treatments through drug repositioning by repurposing existing drugs, shelved drugs, or even candidates that do not meet the criteria for clinical trials. A search was conducted in three databases Web of Science, Scopus, and PubMed. We reviewed the data related to the last years (1975-present) to identify those drugs currently being proposed for repositioning in PD. Moreover, we reviewed the present status of the computational approach, including AI/Machine Learning (AI/ML)-powered pharmaceutical discovery efforts and their implementation in PD treatment. It was found that the number of drug repositioning studies for PD has increased recently. Repositioning of drugs in PD is taking off, and scientific communities are increasingly interested in communicating its results and finding effective treatment alternatives for PD. A better chance of success in PD drug discovery has been made possible due to AI/ML algorithm advancements. In addition to the experimentation stage of drug discovery, it is also important to leverage AI in the planning stage of clinical trials to make them more effective. New AI-based models or solutions that increase the success rate of drug development are greatly needed.
Collapse
Affiliation(s)
- Iman Karimi-Sani
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehrdad Sharifi
- Emergency Medicine Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Nahid Abolpour
- Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehrzad Lotfi
- Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran; Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad-Ali Takhshid
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran; Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Srivastava V, Muralidharan A, Swaminathan A, Poulose A. Anxiety in aquatics: Leveraging machine learning models to predict adult zebrafish behavior. Neuroscience 2025; 565:577-587. [PMID: 39675692 DOI: 10.1016/j.neuroscience.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
Accurate analysis of anxiety behaviors in animal models is pivotal for advancing neuroscience research and drug discovery. This study compares the potential of DeepLabCut, ZebraLab, and machine learning models to analyze anxiety-related behaviors in adult zebrafish. Using a dataset comprising video recordings of unstressed and pre-stressed zebrafish, we extracted features such as total inactivity duration/immobility, time spent at the bottom, time spent at the top and turn angles (large and small). We observed that the data obtained using DeepLabCut and ZebraLab were highly correlated. Using this data, we annotated behaviors as anxious and not anxious and trained several machine learning models, including Logistic Regression, Decision Tree, K-Nearest Neighbours (KNN), Random Forests, Naive Bayes Classifiers, and Support Vector Machines (SVMs). The effectiveness of these machine learning models was validated and tested on independent datasets. We found that some machine learning models, such as Decision Tree and Random Forests, performed excellently to differentiate between anxious and non-anxious behavior, even in the control group, where the differences between subjects were more subtle. Our findings show that upcoming technologies, such as machine learning models, are able to effectively and accurately analyze anxiety behaviors in zebrafish and provide a cost-effective method to analyze animal behavior.
Collapse
Affiliation(s)
- Vartika Srivastava
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER TVM), Vithura, Thiruvananthapuram 695551, Kerala, India.
| | - Anagha Muralidharan
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER TVM), Vithura, Thiruvananthapuram 695551, Kerala, India.
| | - Amrutha Swaminathan
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER TVM), Vithura, Thiruvananthapuram 695551, Kerala, India.
| | - Alwin Poulose
- School of Data Science, Indian Institute of Science Education and Research Thiruvananthapuram (IISER TVM), Vithura, Thiruvananthapuram 695551, Kerala, India.
| |
Collapse
|
4
|
Rostad KO, Trognitz T, Frøyset AK, Bifulco E, Fladmark KE. Accelerated Sarcopenia Phenotype in the DJ-1/ Park7-Knockout Zebrafish. Antioxidants (Basel) 2024; 13:1509. [PMID: 39765837 PMCID: PMC11673048 DOI: 10.3390/antiox13121509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Age-dependent loss of muscle mass and function is associated with oxidative stress. DJ-1/Park7 acts as an antioxidant through multiple signalling pathways. DJ-1-knockout zebrafish show a decline in swimming performance and loss of weight gain between 6 and 9 months of age. Here, we address the degree to which this is associated with muscle degeneration and identify molecular changes preceding dysregulation of muscle performance. Loss of DJ-1 reduced the skeletal muscle fibre cross-section area. The highly mitochondrial-dependent red slow muscle was more affected than the white muscle, and degeneration of sub-sarcolemma red muscle mitochondria was observed. Using TandemMassTag-based quantitative proteomics, we identified a total of 3721 proteins in the multiplex sample of 4 and 12-month-old muscles. A total of 68 proteins, mainly associated with inflammation and mitochondrial function, were dysregulated in the young DJ-1-null adults, with Annexin A3, Sphingomyelin phosphodiesterase acid-like 3B, Complement C3a, and 2,4-dienoyl CoA reductase 1 being the most affected. The loss of DJ-1 also accelerated molecular features associated with sarcopenia, such as a decrease in the NAD+/NADH ratio and a reduction in Prostaglandin reductase 2 and Cytosolic glycerol-3-phosphate dehydrogenase levels. In view of the experimental power of zebrafish, the DJ-1-null zebrafish makes a valuable model for understanding the connection between oxidative stress and age-dependent muscle loss and function.
Collapse
Affiliation(s)
| | | | | | | | - Kari E. Fladmark
- Department of Biological Sciences, University of Bergen, 5020 Bergen, Norway (T.T.); (A.K.F.); (E.B.)
| |
Collapse
|
5
|
Muralidharan A, Swaminathan A, Poulose A. Deep learning dives: Predicting anxiety in zebrafish through novel tank assay analysis. Physiol Behav 2024; 287:114696. [PMID: 39293590 DOI: 10.1016/j.physbeh.2024.114696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/30/2024] [Accepted: 09/11/2024] [Indexed: 09/20/2024]
Abstract
Behavior is fundamental to neuroscience research, providing insights into the mechanisms underlying thoughts, actions and responses. Various model organisms, including mice, flies, and fish, are employed to understand these mechanisms. Zebrafish, in particular, serve as a valuable model for studying anxiety-like behavior, typically measured through the novel tank diving (NTD) assay. Traditional methods for analyzing NTD assays are either manually intensive or costly when using specialized software. To address these limitations, it is useful to develop methods for the automated analysis of zebrafish NTD assays using deep-learning models. In this study, we classified zebrafish based on their anxiety levels using DeepLabCut. Subsequently, based on a training dataset of image frames, we compared deep-learning models to identify the model best suited to classify zebrafish as anxious or non anxious and found that specific architectures, such as InceptionV3, are able to effectively perform this classification task. Our findings suggest that these deep learning models hold promise for automated behavioral analysis in zebrafish, offering an efficient and cost-effective alternative to traditional methods.
Collapse
Affiliation(s)
- Anagha Muralidharan
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER TVM), Vithura, Thiruvananthapuram 695551, Kerala, India.
| | - Amrutha Swaminathan
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER TVM), Vithura, Thiruvananthapuram 695551, Kerala, India.
| | - Alwin Poulose
- School of Data Science, Indian Institute of Science Education and Research Thiruvananthapuram (IISER TVM), Vithura, Thiruvananthapuram 695551, Kerala, India.
| |
Collapse
|
6
|
Gendelev L, Taylor J, Myers-Turnbull D, Chen S, McCarroll MN, Arkin MR, Kokel D, Keiser MJ. Deep phenotypic profiling of neuroactive drugs in larval zebrafish. Nat Commun 2024; 15:9955. [PMID: 39551797 PMCID: PMC11570628 DOI: 10.1038/s41467-024-54375-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024] Open
Abstract
Behavioral larval zebrafish screens leverage a high-throughput small molecule discovery format to find neuroactive molecules relevant to mammalian physiology. We screen a library of 650 central nervous system active compounds in high replicate to train deep metric learning models on zebrafish behavioral profiles. The machine learning initially exploited subtle artifacts in the phenotypic screen, necessitating a complete experimental re-run with rigorous physical well-wise randomization. These large matched phenotypic screening datasets (initial and well-randomized) provide a unique opportunity to quantify and understand shortcut learning in a full-scale, real-world drug discovery dataset. The final deep metric learning model substantially outperforms correlation distance-the canonical way of computing distances between profiles-and generalizes to an orthogonal dataset of diverse drug-like compounds. We validate predictions by prospective in vitro radio-ligand binding assays against human protein targets, achieving a hit rate of 58% despite crossing species and chemical scaffold boundaries. These neuroactive compounds exhibit diverse chemical scaffolds, demonstrating that zebrafish phenotypic screens combined with metric learning achieve robust scaffold hopping capabilities.
Collapse
Affiliation(s)
- Leo Gendelev
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Jack Taylor
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- UCSF Weill Institute for Neurosciences Memory and Aging Center, University of California, San Francisco, CA, USA
| | - Douglas Myers-Turnbull
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Steven Chen
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Matthew N McCarroll
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Michelle R Arkin
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - David Kokel
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
| | - Michael J Keiser
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA.
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA.
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
Wang R, Wang B, Chen A. Application of machine learning in the study of development, behavior, nerve, and genotoxicity of zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 358:124473. [PMID: 38945191 DOI: 10.1016/j.envpol.2024.124473] [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: 03/01/2024] [Revised: 05/26/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024]
Abstract
Machine learning (ML) as a novel model-based approach has been used in studying aquatic toxicology in the environmental field. Zebrafish, as an ideal model organism in aquatic toxicology research, has been widely used to study the toxic effects of various pollutants. However, toxicity testing on organisms may cause significant harm, consume considerable time and resources, and raise ethical concerns. Therefore, ML is used in related research to reduce animal experiments and assist researchers in conducting toxicological research. Although ML techniques have matured in various fields, research on ML-based aquatic toxicology is still in its infancy due to the lack of comprehensive large-scale toxicity databases for environmental pollutants and model organisms. Therefore, to better understand the recent research progress of ML in studying the development, behavior, nerve, and genotoxicity of zebrafish, this review mainly focuses on using ML modeling to assess and predict the toxic effects of zebrafish exposure to different toxic chemicals. Meanwhile, the opportunities and challenges faced by ML in the field of toxicology were analyzed. Finally, suggestions and perspectives were proposed for the toxicity studies of ML on zebrafish in future applications.
Collapse
Affiliation(s)
- Rui Wang
- Key Laboratory of Karst Georesources and Environment, Ministry of Education, (Guizhou University), Guiyang, Guizhou, 550025, China
| | - Bing Wang
- Key Laboratory of Karst Georesources and Environment, Ministry of Education, (Guizhou University), Guiyang, Guizhou, 550025, China; College of Resources and Environmental Engineering, Guizhou University, Guiyang, Guizhou, 550025, China.
| | - Anying Chen
- College of Resources and Environmental Engineering, Guizhou University, Guiyang, Guizhou, 550025, China
| |
Collapse
|
8
|
He S, Ru Q, Chen L, Xu G, Wu Y. Advances in animal models of Parkinson's disease. Brain Res Bull 2024; 215:111024. [PMID: 38969066 DOI: 10.1016/j.brainresbull.2024.111024] [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: 05/12/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
Parkinson's disease is a complex neurodegenerative disease characterized by progressive movement impairments. Predominant symptoms encompass resting tremor, bradykinesia, limb rigidity, and postural instability. In addition, it also includes a series of non-motor symptoms such as sleep disorders, hyposmia, gastrointestinal dysfunction, autonomic dysfunction and cognitive impairment. Pathologically, the disease manifests through dopaminergic neuronal loss and the presence of Lewy bodies. At present, no significant breakthrough has been achieved in clinical Parkinson's disease treatment. Exploring treatment modalities necessitate the establishment of scientifically sound animal models. In recent years, researchers have focused on replicating the symptoms of human Parkinson's disease, resulting in the establishment of various experimental animal models primarily through drugs and transgenic methods to mimic relevant pathologies and identify more effective treatments. This review examines traditional neurotoxin and transgenic animal models as well as α-synuclein pre-formed fibrils models, non-human primate models and non-mammalian specie models. Additionally, it introduces emerging models, including models based on optogenetics, induced pluripotent stem cells, and gene editing, aiming to provide a reference for the utilization of experimental animal models and clinical research for researchers in this field.
Collapse
Affiliation(s)
- Sui He
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Guodong Xu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
9
|
Luo Q, Yang Y, Xian C, Zhou P, Zhang H, Lv Z, Liu J. Nicotinamide riboside ameliorates survival time and motor dysfunction in an MPTP-Induced Parkinson's disease zebrafish model through effects on glucose metabolism and endoplasmic reticulum stress. Chem Biol Interact 2024; 399:111118. [PMID: 38925209 DOI: 10.1016/j.cbi.2024.111118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Nicotinamide riboside (NR) is a precursor and exogenous supplement of nicotinamide adenine dinucleotide (NAD+). NR has been shown to play a beneficial role in a variety of neurodegenerative diseases. A phase 1 clinical trial identified NR as a potential neuroprotective therapy for Parkinson's disease (PD). However, the mechanism of action of NR in PD has not been fully elucidated. Therefore, the present study aimed to investigate the potential effects of NR on a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD model in zebrafish and its underlying mechanisms. The results showed that NR improved motor dysfunction, survival time, dopamine neurons, and peripheral neurons, as well as the NAD+ levels in the MPTP-affected PD zebrafish model. In addition, transcriptome sequencing analysis revealed that, after NR treatment, differentially expressed genes were significantly enriched in the glucose metabolism and protein processing pathways in the endoplasmic reticulum (ER). Quantitative PCR (qPCR) revealed that the mRNA levels of the glycoheterotrophic enzyme (involved in glucose metabolism) were significantly decreased, and the glycolytic enzyme mRNA expression levels were significantly increased. The results of the non-targeted metabolomic analysis showed that NR treatment significantly increased the levels of metabolites such as nicotinic acid ,nicotinamide, d-glucose (from the gluconeogenesis and glycolysis metabolism pathways) and some glucogenic amino acids, such as glutamine. Importantly, NR ameliorated MPTP-induced endoplasmic reticulum stress (ERS) in the PD zebrafish model through the Perk-Eif2α-Atf4-Chop pathway. These results highlight the neuroprotective effect of NR in the present PD zebrafish model through modulation of glucose metabolism and ERS via the Perk-Eif2α-Atf4-Chop pathway and provide valuable mechanistic insights into the treatment of PD.
Collapse
Affiliation(s)
- Qing Luo
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, 25 Taiping Street, Luzhou, Sichuan, 646000, China
| | - Yanmei Yang
- Department of Neurology, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, Sichuan, 646000, China
| | - Chunyan Xian
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, 25 Taiping Street, Luzhou, Sichuan, 646000, China
| | - Pan Zhou
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, 25 Taiping Street, Luzhou, Sichuan, 646000, China
| | - Hui Zhang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, 25 Taiping Street, Luzhou, Sichuan, 646000, China
| | - Zhiyu Lv
- Department of Neurology, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, Sichuan, 646000, China.
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, 25 Taiping Street, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
10
|
Linsley JW, Reisine T, Finkbeiner S. Three dimensional and four dimensional live imaging to study mechanisms of progressive neurodegeneration. J Biol Chem 2024; 300:107433. [PMID: 38825007 PMCID: PMC11261153 DOI: 10.1016/j.jbc.2024.107433] [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: 08/30/2023] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Neurodegenerative diseases are complex and progressive, posing challenges to their study and understanding. Recent advances in microscopy imaging technologies have enabled the exploration of neurons in three spatial dimensions (3D) over time (4D). When applied to 3D cultures, tissues, or animals, these technologies can provide valuable insights into the dynamic and spatial nature of neurodegenerative diseases. This review focuses on the use of imaging techniques and neurodegenerative disease models to study neurodegeneration in 4D. Imaging techniques such as confocal microscopy, two-photon microscopy, miniscope imaging, light sheet microscopy, and robotic microscopy offer powerful tools to visualize and analyze neuronal changes over time in 3D tissue. Application of these technologies to in vitro models of neurodegeneration such as mouse organotypic culture systems and human organoid models provide versatile platforms to study neurodegeneration in a physiologically relevant context. Additionally, use of 4D imaging in vivo, including in mouse and zebrafish models of neurodegenerative diseases, allows for the investigation of early dysfunction and behavioral changes associated with neurodegeneration. We propose that these studies have the power to overcome the limitations of two-dimensional monolayer neuronal cultures and pave the way for improved understanding of the dynamics of neurodegenerative diseases and the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Jeremy W Linsley
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, California, USA; Operant Biopharma, San Francisco, California, USA
| | - Terry Reisine
- Independent Scientific Consultant, Santa Cruz, California, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, California, USA; Operant Biopharma, San Francisco, California, USA; Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, San Francisco, California, USA; Departments of Neurology and Physiology, University of California, San Francisco, California, USA; Neuroscience Graduate Program, University of California, San Francisco, California, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, California, USA.
| |
Collapse
|
11
|
Shan L, Heusinkveld HJ, Paul KC, Hughes S, Darweesh SKL, Bloem BR, Homberg JR. Towards improved screening of toxins for Parkinson's risk. NPJ Parkinsons Dis 2023; 9:169. [PMID: 38114496 PMCID: PMC10730534 DOI: 10.1038/s41531-023-00615-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023] Open
Abstract
Parkinson's disease (PD) is a chronic, progressive and disabling neurodegenerative disorder. The prevalence of PD has risen considerably over the past decades. A growing body of evidence suggest that exposure to environmental toxins, including pesticides, solvents and heavy metals (collectively called toxins), is at least in part responsible for this rapid growth. It is worrying that the current screening procedures being applied internationally to test for possible neurotoxicity of specific compounds offer inadequate insights into the risk of developing PD in humans. Improved screening procedures are therefore urgently needed. Our review first substantiates current evidence on the relation between exposure to environmental toxins and the risk of developing PD. We subsequently propose to replace the current standard toxin screening by a well-controlled multi-tier toxin screening involving the following steps: in silico studies (tier 1) followed by in vitro tests (tier 2), aiming to prioritize agents with human relevant routes of exposure. More in depth studies can be undertaken in tier 3, with whole-organism (in)vertebrate models. Tier 4 has a dedicated focus on cell loss in the substantia nigra and on the presumed mechanisms of neurotoxicity in rodent models, which are required to confirm or refute the possible neurotoxicity of any individual compound. This improved screening procedure should not only evaluate new pesticides that seek access to the market, but also critically assess all pesticides that are being used today, acknowledging that none of these has ever been proven to be safe from a perspective of PD. Importantly, the improved screening procedures should not just assess the neurotoxic risk of isolated compounds, but should also specifically look at the cumulative risk conveyed by exposure to commonly used combinations of pesticides (cocktails). The worldwide implementation of such an improved screening procedure, would be an essential step for policy makers and governments to recognize PD-related environmental risk factors.
Collapse
Affiliation(s)
- Ling Shan
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands.
| | - Harm J Heusinkveld
- Centre for Health Protection, National Institute for Public Health and Environment (RIVM), Bilthoven, The Netherlands
| | - Kimberly C Paul
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Samantha Hughes
- A-LIFE Amsterdam Institute for Life and Environment, Section Environmental Health and Toxicology, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| | - Sirwan K L Darweesh
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Bastiaan R Bloem
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
12
|
Fan YL, Hsu FR, Wang Y, Liao LD. Unlocking the Potential of Zebrafish Research with Artificial Intelligence: Advancements in Tracking, Processing, and Visualization. Med Biol Eng Comput 2023; 61:2797-2814. [PMID: 37558927 DOI: 10.1007/s11517-023-02903-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
Zebrafish have become a widely accepted model organism for biomedical research due to their strong cortisol stress response, behavioral strain differences, and sensitivity to both drug treatments and predators. However, experimental zebrafish studies generate substantial data that must be analyzed through objective, accurate, and repeatable analysis methods. Recently, advancements in artificial intelligence (AI) have enabled automated tracking, image recognition, and data analysis, leading to more efficient and insightful investigations. In this review, we examine key AI applications in zebrafish research, including behavior analysis, genomics, and neuroscience. With the development of deep learning technology, AI algorithms have been used to precisely analyze and identify images of zebrafish, enabling automated testing and analysis. By applying AI algorithms in genomics research, researchers have elucidated the relationship between genes and biology, providing a better basis for the development of disease treatments and gene therapies. Additionally, the development of more effective neuroscience tools could help researchers better understand the complex neural networks in the zebrafish brain. In the future, further advancements in AI technology are expected to enable more extensive and in-depth medical research applications in zebrafish, improving our understanding of this important animal model. This review highlights the potential of AI technology in achieving the full potential of zebrafish research by enabling researchers to efficiently track, process, and visualize the outcomes of their experiments.
Collapse
Affiliation(s)
- Yi-Ling Fan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35, Keyan Road, Zhunan Town, Miaoli County, 35053, Taiwan
- Department of Information Engineering and Computer Science, Feng Chia University, Taichung, 407, Taiwan
| | - Fang-Rong Hsu
- Department of Information Engineering and Computer Science, Feng Chia University, Taichung, 407, Taiwan
| | - Yuhling Wang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35, Keyan Road, Zhunan Town, Miaoli County, 35053, Taiwan
- Department of Electrical Engineering, National United University, 2, Lien-Da, Nan-Shih Li, Miaoli, 360302, Taiwan
| | - Lun-De Liao
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35, Keyan Road, Zhunan Town, Miaoli County, 35053, Taiwan.
| |
Collapse
|
13
|
Stonadge A, Genzor AV, Russell A, Hamed MF, Romero N, Evans G, Pownall ME, Bekker-Jensen S, Blanco G. Myofibrillar myopathy hallmarks associated with ZAK deficiency. Hum Mol Genet 2023; 32:2751-2770. [PMID: 37427997 PMCID: PMC10789240 DOI: 10.1093/hmg/ddad113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023] Open
Abstract
The ZAK gene encodes two functionally distinct kinases, ZAKα and ZAKβ. Homozygous loss of function mutations affecting both isoforms causes a congenital muscle disease. ZAKβ is the only isoform expressed in skeletal muscle and is activated by muscle contraction and cellular compression. The ZAKβ substrates in skeletal muscle or the mechanism whereby ZAKβ senses mechanical stress remains to be determined. To gain insights into the pathogenic mechanism, we exploited ZAK-deficient cell lines, zebrafish, mice and a human biopsy. ZAK-deficient mice and zebrafish show a mild phenotype. In mice, comparative histopathology data from regeneration, overloading, ageing and sex conditions indicate that while age and activity are drivers of the pathology, ZAKβ appears to have a marginal role in myoblast fusion in vitro or muscle regeneration in vivo. The presence of SYNPO2, BAG3 and Filamin C (FLNC) in a phosphoproteomics assay and extended analyses suggested a role for ZAKβ in the turnover of FLNC. Immunofluorescence analysis of muscle sections from mice and a human biopsy showed evidence of FLNC and BAG3 accumulations as well as other myofibrillar myopathy markers. Moreover, endogenous overloading of skeletal muscle exacerbated the presence of fibres with FLNC accumulations in mice, indicating that ZAKβ signalling is necessary for an adaptive turnover of FLNC that allows for the normal physiological response to sustained mechanical stress. We suggest that accumulation of mislocalized FLNC and BAG3 in highly immunoreactive fibres contributes to the pathogenic mechanism of ZAK deficiency.
Collapse
Affiliation(s)
- Amy Stonadge
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5DD, UK
| | - Aitana V Genzor
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Alex Russell
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5DD, UK
| | - Mohamed F Hamed
- Department of Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Norma Romero
- Unité de Morphologie Neuromusculaire Institut de Myologie - Inserm Sorbonne Université - GHU Pitié-Salpêtrière 47- 83, boulevard de l’Hôpital F-75 651 Paris, Cedex 13, France
| | - Gareth Evans
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5DD, UK
| | - Mary Elizabeth Pownall
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5DD, UK
| | - Simon Bekker-Jensen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Gonzalo Blanco
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5DD, UK
| |
Collapse
|
14
|
Nakamae K, Bono H. DANGER analysis: risk-averse on/off-target assessment for CRISPR editing without a reference genome. BIOINFORMATICS ADVANCES 2023; 3:vbad114. [PMID: 37661945 PMCID: PMC10469126 DOI: 10.1093/bioadv/vbad114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/11/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023]
Abstract
Motivation The CRISPR-Cas9 system has successfully achieved site-specific gene editing in organisms ranging from humans to bacteria. The technology efficiently generates mutants, allowing for phenotypic analysis of the on-target gene. However, some conventional studies did not investigate whether deleterious off-target effects partially affect the phenotype. Results Herein, we present a novel phenotypic assessment of CRISPR-mediated gene editing: Deleterious and ANticipatable Guides Evaluated by RNA-sequencing (DANGER) analysis. Using RNA-seq data, this bioinformatics pipeline can elucidate genomic on/off-target sites on mRNA-transcribed regions related to expression changes and then quantify phenotypic risk at the gene ontology term level. We demonstrated the risk-averse on/off-target assessment in RNA-seq data from gene-edited samples of human cells and zebrafish brains. Our DANGER analysis successfully detected off-target sites, and it quantitatively evaluated the potential contribution of deleterious off-targets to the transcriptome phenotypes of the edited mutants. Notably, DANGER analysis harnessed de novo transcriptome assembly to perform risk-averse on/off-target assessments without a reference genome. Thus, our resources would help assess genome editing in non-model organisms, individual human genomes, and atypical genomes from diseases and viruses. In conclusion, DANGER analysis facilitates the safer design of genome editing in all organisms with a transcriptome. Availability and implementation The Script for the DANGER analysis pipeline is available at https://github.com/KazukiNakamae/DANGER_analysis. In addition, the software provides a tutorial on reproducing the results presented in this article on the Readme page. The Docker image of DANGER_analysis is also available at https://hub.docker.com/repository/docker/kazukinakamae/dangeranalysis/general.
Collapse
Affiliation(s)
- Kazuki Nakamae
- Laboratory of Bio-DX, Genome Editing Innovation Center, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-0046, Japan
- Research and Development Department, PtBio Inc., 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-0046, Japan
| | - Hidemasa Bono
- Laboratory of Bio-DX, Genome Editing Innovation Center, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-0046, Japan
- Laboratory of Genome Informatics, Graduate School of Integrated Sciences for Life, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-0046, Japan
| |
Collapse
|
15
|
Qu J, Liu N, Gao L, Hu J, Sun M, Yu D. Development of CRISPR Cas9, spin-off technologies and their application in model construction and potential therapeutic methods of Parkinson's disease. Front Neurosci 2023; 17:1223747. [PMID: 37483347 PMCID: PMC10359996 DOI: 10.3389/fnins.2023.1223747] [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: 05/16/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common degenerative diseases. It is most typically characterized by neuronal death following the accumulation of Lewis inclusions in dopaminergic neurons in the substantia nigra region, with clinical symptoms such as motor retardation, autonomic dysfunction, and dystonia spasms. The exact molecular mechanism of its pathogenesis has not been revealed up to now. And there is a lack of effective treatments for PD, which places a burden on patients, families, and society. CRISPR Cas9 is a powerful technology to modify target genomic sequence with rapid development. More and more scientists utilized this technique to perform research associated neurodegenerative disease including PD. However, the complexity involved makes it urgent to organize and summarize the existing findings to facilitate a clearer understanding. In this review, we described the development of CRISPR Cas9 technology and the latest spin-off gene editing systems. Then we focused on the application of CRISPR Cas9 technology in PD research, summarizing the construction of the novel PD-related medical models including cellular models, small animal models, large mammal models. We also discussed new directions and target molecules related to the use of CRISPR Cas9 for PD treatment from the above models. Finally, we proposed the view about the directions for the development and optimization of the CRISPR Cas9 technology system, and its application to PD and gene therapy in the future. All these results provided a valuable reference and enhanced in understanding for studying PD.
Collapse
Affiliation(s)
- Jiangbo Qu
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Na Liu
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Lu Gao
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Jia Hu
- School of Life Science and Technology, Weifang Medical University, Weifang, Shandong, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Dongyi Yu
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| |
Collapse
|
16
|
Otsuka T, Matsui H. Fish Models for Exploring Mitochondrial Dysfunction Affecting Neurodegenerative Disorders. Int J Mol Sci 2023; 24:ijms24087079. [PMID: 37108237 PMCID: PMC10138900 DOI: 10.3390/ijms24087079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/05/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Neurodegenerative disorders are characterized by the progressive loss of neuronal structure or function, resulting in memory loss and movement disorders. Although the detailed pathogenic mechanism has not been elucidated, it is thought to be related to the loss of mitochondrial function in the process of aging. Animal models that mimic the pathology of a disease are essential for understanding human diseases. In recent years, small fish have become ideal vertebrate models for human disease due to their high genetic and histological homology to humans, ease of in vivo imaging, and ease of genetic manipulation. In this review, we first outline the impact of mitochondrial dysfunction on the progression of neurodegenerative diseases. Then, we highlight the advantages of small fish as model organisms, and present examples of previous studies regarding mitochondria-related neuronal disorders. Lastly, we discuss the applicability of the turquoise killifish, a unique model for aging research, as a model for neurodegenerative diseases. Small fish models are expected to advance our understanding of the mitochondrial function in vivo, the pathogenesis of neurodegenerative diseases, and be important tools for developing therapies to treat diseases.
Collapse
Affiliation(s)
- Takayoshi Otsuka
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Hideaki Matsui
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| |
Collapse
|
17
|
Domínguez-Oliva A, Hernández-Ávalos I, Martínez-Burnes J, Olmos-Hernández A, Verduzco-Mendoza A, Mota-Rojas D. The Importance of Animal Models in Biomedical Research: Current Insights and Applications. Animals (Basel) 2023; 13:ani13071223. [PMID: 37048478 PMCID: PMC10093480 DOI: 10.3390/ani13071223] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/19/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023] Open
Abstract
Animal research is considered a key element in advance of biomedical science. Although its use is controversial and raises ethical challenges, the contribution of animal models in medicine is essential for understanding the physiopathology and novel treatment alternatives for several animal and human diseases. Current pandemics’ pathology, such as the 2019 Coronavirus disease, has been studied in primate, rodent, and porcine models to recognize infection routes and develop therapeutic protocols. Worldwide issues such as diabetes, obesity, neurological disorders, pain, rehabilitation medicine, and surgical techniques require studying the process in different animal species before testing them on humans. Due to their relevance, this article aims to discuss the importance of animal models in diverse lines of biomedical research by analyzing the contributions of the various species utilized in science over the past five years about key topics concerning human and animal health.
Collapse
Affiliation(s)
- Adriana Domínguez-Oliva
- Master’s Program in Agricultural and Livestock Sciences [Maestría en Ciencias Agropecuarias], Xochimilco Campus, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Ismael Hernández-Ávalos
- Clinical Pharmacology and Veterinary Anesthesia, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán 54714, Mexico
| | - Julio Martínez-Burnes
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Tamaulipas, Victoria City 87000, Mexico
| | - Adriana Olmos-Hernández
- Division of Biotechnology—Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación-Luis, Guillermo Ibarra Ibarra (INR-LGII), Mexico City 14389, Mexico
| | - Antonio Verduzco-Mendoza
- Division of Biotechnology—Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación-Luis, Guillermo Ibarra Ibarra (INR-LGII), Mexico City 14389, Mexico
| | - Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
- Correspondence:
| |
Collapse
|
18
|
Layton R, Layton D, Beggs D, Fisher A, Mansell P, Stanger KJ. The impact of stress and anesthesia on animal models of infectious disease. Front Vet Sci 2023; 10:1086003. [PMID: 36816193 PMCID: PMC9933909 DOI: 10.3389/fvets.2023.1086003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/09/2023] [Indexed: 02/05/2023] Open
Abstract
Stress and general anesthesia have an impact on the functional response of the organism due to the detrimental effects on cardiovascular, immunological, and metabolic function, which could limit the organism's response to an infectious event. Animal studies have formed an essential step in understanding and mitigating infectious diseases, as the complexities of physiology and immunity cannot yet be replicated in vivo. Using animals in research continues to come under increasing societal scrutiny, and it is therefore crucial that the welfare of animals used in disease research is optimized to meet both societal expectations and improve scientific outcomes. Everyday management and procedures in animal studies are known to cause stress, which can not only cause poorer welfare outcomes, but also introduces variables in disease studies. Whilst general anesthesia is necessary at times to reduce stress and enhance animal welfare in disease research, evidence of physiological and immunological disruption caused by general anesthesia is increasing. To better understand and quantify the effects of stress and anesthesia on disease study and welfare outcomes, utilizing the most appropriate animal monitoring strategies is imperative. This article aims to analyze recent scientific evidence about the impact of stress and anesthesia as uncontrolled variables, as well as reviewing monitoring strategies and technologies in animal models during infectious diseases.
Collapse
Affiliation(s)
- Rachel Layton
- Australian Centre for Disease Preparedness, CSIRO, Geelong, VIC, Australia,*Correspondence: Rachel Layton ✉
| | - Daniel Layton
- Australian Centre for Disease Preparedness, CSIRO, Geelong, VIC, Australia
| | - David Beggs
- Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew Fisher
- Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, University of Melbourne, Melbourne, VIC, Australia
| | - Peter Mansell
- Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, University of Melbourne, Melbourne, VIC, Australia
| | - Kelly J. Stanger
- Australian Centre for Disease Preparedness, CSIRO, Geelong, VIC, Australia
| |
Collapse
|
19
|
Mohamed WMY, Ekker M. Editorial: Zebrafish as a translational neuroscience model: today's science and tomorrow's success. Front Physiol 2023; 14:1202198. [PMID: 37153225 PMCID: PMC10154676 DOI: 10.3389/fphys.2023.1202198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 05/09/2023] Open
Affiliation(s)
- Wael M. Y. Mohamed
- Basic Medical Science Department, Kulliyyah of Medicine, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
- *Correspondence: Wael M. Y. Mohamed,
| | - Marc Ekker
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
20
|
Li D, Li X, Wang Q, Hao Y. Advanced Techniques for the Intelligent Diagnosis of Fish Diseases: A Review. Animals (Basel) 2022; 12:2938. [PMID: 36359061 PMCID: PMC9656208 DOI: 10.3390/ani12212938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 10/15/2023] Open
Abstract
Aquatic products, as essential sources of protein, have attracted considerable concern by producers and consumers. Precise fish disease prevention and treatment may provide not only healthy fish protein but also ecological and economic benefits. However, unlike intelligent two-dimensional diagnoses of plants and crops, one of the most serious challenges confronted in intelligent aquaculture diagnosis is its three-dimensional space. Expert systems have been applied to diagnose fish diseases in recent decades, allowing for restricted diagnosis of certain aquaculture. However, this method needs aquaculture professionals and specialists. In addition, diagnosis speed and efficiency are limited. Therefore, developing a new quick, automatic, and real-time diagnosis approach is very critical. The integration of image-processing and computer vision technology intelligently allows the diagnosis of fish diseases. This study comprehensively reviews image-processing technology and image-based fish disease detection methods, and analyzes the benefits and drawbacks of each diagnostic approach in different environments. Although it is widely acknowledged that there are many approaches for disease diagnosis and pathogen identification, some improvements in detection accuracy and speed are still needed. Constructing AR 3D images of fish diseases, standard and shared datasets, deep learning, and data fusion techniques will be helpful in improving the accuracy and speed of fish disease diagnosis.
Collapse
Affiliation(s)
- Daoliang Li
- National Innovation Center for Digital Fishery, China Agricultural University, 17 Tsinghua East Road, Beijing 100083, China
- College of Information and Electrical Engineering, China Agricultural University, Beijing 100083, China
- Beijing Engineering and Technology Research Centre for Internet of Things in Agriculture, China Agriculture University, Beijing 100083, China
- Key Laboratory of Agricultural Information Acquisition Technology, Ministry of Agriculture, Beijing 100083, China
| | - Xin Li
- National Innovation Center for Digital Fishery, China Agricultural University, 17 Tsinghua East Road, Beijing 100083, China
- College of Information and Electrical Engineering, China Agricultural University, Beijing 100083, China
- Beijing Engineering and Technology Research Centre for Internet of Things in Agriculture, China Agriculture University, Beijing 100083, China
- Key Laboratory of Agricultural Information Acquisition Technology, Ministry of Agriculture, Beijing 100083, China
| | - Qi Wang
- National Innovation Center for Digital Fishery, China Agricultural University, 17 Tsinghua East Road, Beijing 100083, China
- College of Information and Electrical Engineering, China Agricultural University, Beijing 100083, China
- Beijing Engineering and Technology Research Centre for Internet of Things in Agriculture, China Agriculture University, Beijing 100083, China
- Key Laboratory of Agricultural Information Acquisition Technology, Ministry of Agriculture, Beijing 100083, China
| | - Yinfeng Hao
- National Innovation Center for Digital Fishery, China Agricultural University, 17 Tsinghua East Road, Beijing 100083, China
- College of Information and Electrical Engineering, China Agricultural University, Beijing 100083, China
- Beijing Engineering and Technology Research Centre for Internet of Things in Agriculture, China Agriculture University, Beijing 100083, China
- Key Laboratory of Agricultural Information Acquisition Technology, Ministry of Agriculture, Beijing 100083, China
| |
Collapse
|
21
|
Fu CW, Horng JL, Chou MY. Fish Behavior as a Neural Proxy to Reveal Physiological States. Front Physiol 2022; 13:937432. [PMID: 35910555 PMCID: PMC9326089 DOI: 10.3389/fphys.2022.937432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Behaviors are the integrative outcomes of the nervous system, which senses and responds to the internal physiological status and external stimuli. Teleosts are aquatic organisms which are more easily affected by the surrounding environment compared to terrestrial animals. To date, behavioral tests have been widely used to assess potential environmental risks using fish as model animals. In this review, we summarized recent studies regarding the effects of internal and external stimuli on fish behaviors. We concluded that behaviors reflect environmental and physiological changes, which have possible implications for environmental and physiological assessments.
Collapse
Affiliation(s)
- Chih-Wei Fu
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Jiun-Lin Horng
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Yi Chou
- Department of Life Science, National Taiwan University, Taipei, Taiwan
- *Correspondence: Ming-Yi Chou,
| |
Collapse
|
22
|
Wang J, Cao H. Zebrafish and Medaka: Important Animal Models for Human Neurodegenerative Diseases. Int J Mol Sci 2021; 22:10766. [PMID: 34639106 PMCID: PMC8509648 DOI: 10.3390/ijms221910766] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
Animal models of human neurodegenerative disease have been investigated for several decades. In recent years, zebrafish (Danio rerio) and medaka (Oryzias latipes) have become popular in pathogenic and therapeutic studies about human neurodegenerative diseases due to their small size, the optical clarity of embryos, their fast development, and their suitability to large-scale therapeutic screening. Following the emergence of a new generation of molecular biological technologies such as reverse and forward genetics, morpholino, transgenesis, and gene knockout, many human neurodegenerative disease models, such as Parkinson's, Huntington's, and Alzheimer's, were constructed in zebrafish and medaka. These studies proved that zebrafish and medaka genes are functionally conserved in relation to their human homologues, so they exhibit similar neurodegenerative phenotypes to human beings. Therefore, fish are a suitable model for the investigation of pathologic mechanisms of neurodegenerative diseases and for the large-scale screening of drugs for potential therapy. In this review, we summarize the studies in modelling human neurodegenerative diseases in zebrafish and medaka in recent years.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Donghu South Road 7#, Wuhan 430072, China;
- College of Advanced Agriculture Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong Cao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Donghu South Road 7#, Wuhan 430072, China;
- College of Advanced Agriculture Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
23
|
Wang X, Zhang JB, He KJ, Wang F, Liu CF. Advances of Zebrafish in Neurodegenerative Disease: From Models to Drug Discovery. Front Pharmacol 2021; 12:713963. [PMID: 34335276 PMCID: PMC8317260 DOI: 10.3389/fphar.2021.713963] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative disease (NDD), including Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis, are characterized by the progressive loss of neurons which leads to the decline of motor and/or cognitive function. Currently, the prevalence of NDD is rapidly increasing in the aging population. However, valid drugs or treatment for NDD are still lacking. The clinical heterogeneity and complex pathogenesis of NDD pose a great challenge for the development of disease-modifying therapies. Numerous animal models have been generated to mimic the pathological conditions of these diseases for drug discovery. Among them, zebrafish (Danio rerio) models are progressively emerging and becoming a powerful tool for in vivo study of NDD. Extensive use of zebrafish in pharmacology research or drug screening is due to the high conserved evolution and 87% homology to humans. In this review, we summarize the zebrafish models used in NDD studies, and highlight the recent findings on pharmacological targets for NDD treatment. As high-throughput platforms in zebrafish research have rapidly developed in recent years, we also discuss the application prospects of these new technologies in future NDD research.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jin-Bao Zhang
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Kai-Jie He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Fen Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Chun-Feng Liu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.,Department of Neurology, Suqian First Hospital, Suqian, China
| |
Collapse
|
24
|
Bastioli G, Regoni M, Cazzaniga F, De Luca CMG, Bistaffa E, Zanetti L, Moda F, Valtorta F, Sassone J. Animal Models of Autosomal Recessive Parkinsonism. Biomedicines 2021; 9:biomedicines9070812. [PMID: 34356877 PMCID: PMC8301401 DOI: 10.3390/biomedicines9070812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s disease (PD) is the most common neurodegenerative movement disorder. The neuropathological hallmark of the disease is the loss of dopamine neurons of the substantia nigra pars compacta. The clinical manifestations of PD are bradykinesia, rigidity, resting tremors and postural instability. PD patients often display non-motor symptoms such as depression, anxiety, weakness, sleep disturbances and cognitive disorders. Although, in 90% of cases, PD has a sporadic onset of unknown etiology, highly penetrant rare genetic mutations in many genes have been linked with typical familial PD. Understanding the mechanisms behind the DA neuron death in these Mendelian forms may help to illuminate the pathogenesis of DA neuron degeneration in the more common forms of PD. A key step in the identification of the molecular pathways underlying DA neuron death, and in the development of therapeutic strategies, is the creation and characterization of animal models that faithfully recapitulate the human disease. In this review, we outline the current status of PD modeling using mouse, rat and non-mammalian models, focusing on animal models for autosomal recessive PD.
Collapse
Affiliation(s)
- Guendalina Bastioli
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (G.B.); (M.R.); (L.Z.); (F.V.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Maria Regoni
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (G.B.); (M.R.); (L.Z.); (F.V.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Federico Cazzaniga
- Division of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (F.C.); (C.M.G.D.L.); (E.B.); (F.M.)
| | - Chiara Maria Giulia De Luca
- Division of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (F.C.); (C.M.G.D.L.); (E.B.); (F.M.)
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati, 34136 Trieste, Italy
| | - Edoardo Bistaffa
- Division of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (F.C.); (C.M.G.D.L.); (E.B.); (F.M.)
| | - Letizia Zanetti
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (G.B.); (M.R.); (L.Z.); (F.V.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Fabio Moda
- Division of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (F.C.); (C.M.G.D.L.); (E.B.); (F.M.)
| | - Flavia Valtorta
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (G.B.); (M.R.); (L.Z.); (F.V.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Jenny Sassone
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (G.B.); (M.R.); (L.Z.); (F.V.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Correspondence:
| |
Collapse
|
25
|
Yang P, Takahashi H, Murase M, Itoh M. Zebrafish behavior feature recognition using three-dimensional tracking and machine learning. Sci Rep 2021; 11:13492. [PMID: 34188116 PMCID: PMC8242018 DOI: 10.1038/s41598-021-92854-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/14/2021] [Indexed: 11/09/2022] Open
Abstract
In this work, we aim to construct a new behavior analysis method by using machine learning. We used two cameras to capture three-dimensional (3D) tracking data of zebrafish, which were analyzed using fuzzy adaptive resonance theory (FuzzyART), a type of machine learning algorithm, to identify specific behavioral features. The method was tested based on an experiment in which electric shocks were delivered to zebrafish and zebrafish swimming was tracked in 3D simultaneously to find electric shock-associated behaviors. By processing the obtained data with FuzzyART, we discovered that distinguishing behaviors were statistically linked to the electric shock based on the machine learning algorithm. Moreover, our system could accept user-supplied data for detection and quantitative analysis of the behavior features, such as the behavior features defined by the 3D tracking analysis above. This system could be applied to discover new distinct behavior features in mutant zebrafish and used for drug administration screening and cognitive ability tests of zebrafish in the future.
Collapse
Affiliation(s)
- Peng Yang
- Graduate School of Pharmaceutical Science, Chiba University, Chiba, Japan
| | - Hiro Takahashi
- Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Masataka Murase
- Aichi Institute of Technology Technical College of Communications and Electronics, Aichi, Japan
| | - Motoyuki Itoh
- Graduate School of Pharmaceutical Science, Chiba University, Chiba, Japan.
| |
Collapse
|
26
|
Chin HY, Lardelli M, Collins-Praino L, Barthelson K. Loss of park7 activity has differential effects on expression of iron responsive element (IRE) gene sets in the brain transcriptome in a zebrafish model of Parkinson's disease. Mol Brain 2021; 14:83. [PMID: 34030724 PMCID: PMC8146209 DOI: 10.1186/s13041-021-00792-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/18/2021] [Indexed: 11/26/2022] Open
Abstract
Mutation of the gene PARK7 (DJ1) causes monogenic autosomal recessive Parkinson's disease (PD) in humans. Subsequent alterations of PARK7 protein function lead to mitochondrial dysfunction, a major element in PD pathology. Homozygous mutants for the PARK7-orthologous genes in zebrafish, park7, show changes to gene expression in the oxidative phosphorylation pathway, supporting that disruption of energy production is a key feature of neurodegeneration in PD. Iron is critical for normal mitochondrial function, and we have previously used bioinformatic analysis of IRE-bearing transcripts in brain transcriptomes to find evidence supporting the existence of iron dyshomeostasis in Alzheimer's disease. Here, we analysed IRE-bearing transcripts in the transcriptome data from homozygous park7-/- mutant zebrafish brains. We found that the set of genes with "high quality" IREs in their 5' untranslated regions (UTRs, the HQ5'IRE gene set) was significantly altered in these 4-month-old park7-/- brains. However, sets of genes with IREs in their 3' UTRs appeared unaffected. The effects on HQ5'IRE genes are possibly driven by iron dyshomeostasis and/or oxidative stress, but illuminate the existence of currently unknown mechanisms with differential overall effects on 5' and 3' IREs.
Collapse
Affiliation(s)
- Hui Yung Chin
- Alzheimer’s Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA 5005 Australia
| | - Michael Lardelli
- Alzheimer’s Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA 5005 Australia
| | - Lyndsey Collins-Praino
- Department of Biomedical Sciences, Adelaide Medical School, University of Adelaide, Frome Rd, Adelaide, SA 5005 Australia
| | - Karissa Barthelson
- Alzheimer’s Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA 5005 Australia
| |
Collapse
|
27
|
Vatansever S, Schlessinger A, Wacker D, Kaniskan HÜ, Jin J, Zhou M, Zhang B. Artificial intelligence and machine learning-aided drug discovery in central nervous system diseases: State-of-the-arts and future directions. Med Res Rev 2021; 41:1427-1473. [PMID: 33295676 PMCID: PMC8043990 DOI: 10.1002/med.21764] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/30/2020] [Accepted: 11/20/2020] [Indexed: 01/11/2023]
Abstract
Neurological disorders significantly outnumber diseases in other therapeutic areas. However, developing drugs for central nervous system (CNS) disorders remains the most challenging area in drug discovery, accompanied with the long timelines and high attrition rates. With the rapid growth of biomedical data enabled by advanced experimental technologies, artificial intelligence (AI) and machine learning (ML) have emerged as an indispensable tool to draw meaningful insights and improve decision making in drug discovery. Thanks to the advancements in AI and ML algorithms, now the AI/ML-driven solutions have an unprecedented potential to accelerate the process of CNS drug discovery with better success rate. In this review, we comprehensively summarize AI/ML-powered pharmaceutical discovery efforts and their implementations in the CNS area. After introducing the AI/ML models as well as the conceptualization and data preparation, we outline the applications of AI/ML technologies to several key procedures in drug discovery, including target identification, compound screening, hit/lead generation and optimization, drug response and synergy prediction, de novo drug design, and drug repurposing. We review the current state-of-the-art of AI/ML-guided CNS drug discovery, focusing on blood-brain barrier permeability prediction and implementation into therapeutic discovery for neurological diseases. Finally, we discuss the major challenges and limitations of current approaches and possible future directions that may provide resolutions to these difficulties.
Collapse
Affiliation(s)
- Sezen Vatansever
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Avner Schlessinger
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Daniel Wacker
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - H. Ümit Kaniskan
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jian Jin
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ming‐Ming Zhou
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Bin Zhang
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
28
|
Razali K, Othman N, Mohd Nasir MH, Doolaanea AA, Kumar J, Ibrahim WN, Mohamed Ibrahim N, Mohamed WMY. The Promise of the Zebrafish Model for Parkinson's Disease: Today's Science and Tomorrow's Treatment. Front Genet 2021; 12:655550. [PMID: 33936174 PMCID: PMC8082503 DOI: 10.3389/fgene.2021.655550] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/23/2021] [Indexed: 11/29/2022] Open
Abstract
The second most prevalent neurodegenerative disorder in the elderly is Parkinson's disease (PD). Its etiology is unclear and there are no available disease-modifying medicines. Therefore, more evidence is required concerning its pathogenesis. The use of the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is the basis of most animal models of PD. MPTP is metabolized by monoamine oxidase B (MAO B) to MPP + and induces the loss of dopaminergic neurons in the substantia nigra in mammals. Zebrafish have been commonly used in developmental biology as a model organism, but owing to its perfect mix of properties, it is now emerging as a model for human diseases. Zebrafish (Danio rerio) are cheap and easy to sustain, evolve rapidly, breed transparent embryos in large amounts, and are readily manipulated by different methods, particularly genetic ones. Furthermore, zebrafish are vertebrate species and mammalian findings obtained from zebrafish may be more applicable than those derived from genetic models of invertebrates such as Drosophila melanogaster and Caenorhabditis elegans. The resemblance cannot be taken for granted, however. The goal of the present review article is to highlight the promise of zebrafish as a PD animal model. As its aminergic structures, MPTP mode of action, and PINK1 roles mimic those of mammalians, zebrafish seems to be a viable model for studying PD. The roles of zebrafish MAO, however, vary from those of the two types of MAO present in mammals. The benefits unique to zebrafish, such as the ability to perform large-scale genetic or drug screens, should be exploited in future experiments utilizing zebrafish PD models.
Collapse
Affiliation(s)
- Khairiah Razali
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia (IIUM), Kuantan, Malaysia
| | - Noratikah Othman
- Department of Basic Medical Sciences, Kulliyyah of Nursing, International Islamic University Malaysia (IIUM), Kuantan, Malaysia
| | - Mohd Hamzah Mohd Nasir
- Central Research and Animal Facility (CREAM), Kulliyyah of Science, International Islamic University Malaysia (IIUM), Kuantan, Malaysia
| | - Abd Almonem Doolaanea
- Department of Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia (IIUM), Kuantan, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, UKM Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| | - Wisam Nabeel Ibrahim
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | | | - Wael M. Y. Mohamed
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia (IIUM), Kuantan, Malaysia
- Clinical Pharmacology Department, Menoufia Medical School, Menoufia University, Menoufia, Egypt
| |
Collapse
|
29
|
First person – Gideon Hughes. Dis Model Mech 2020. [PMCID: PMC7578348 DOI: 10.1242/dmm.047415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
First Person is a series of interviews with the first authors of a selection of papers published in Disease Models & Mechanisms, helping early-career researchers promote themselves alongside their papers. Gideon Hughes is first author on ‘Machine learning discriminates a movement disorder in a zebrafish model of Parkinson's disease’, published in DMM. Gideon conducted the research described in this article while a PhD student in Betsy Pownall's lab at the University of York, York, UK. He is now a postdoc in the lab of Henry Roehl at the University of Sheffield, Sheffield, UK, using the zebrafish as a model organism to study human disease and tissue regeneration, combining his research with his interest in computer science.
Collapse
|