1
|
Palaniselvam S, Narasimman V, Vijayashree R, Ramachandran S. Neuroprotective effect of nano-carboxymethyl chitosan from Doryteuthis sibogae against rotenone-induced Parkinson's disease in the zebrafish model. Behav Brain Res 2025; 485:115523. [PMID: 40064356 DOI: 10.1016/j.bbr.2025.115523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/01/2025] [Accepted: 03/02/2025] [Indexed: 03/16/2025]
Abstract
This study investigates the neuroprotective effects of N-carboxymethyl chitosan (N-CMC) against rotenone (ROT)-induced Parkinson's disease (PD) in a zebrafish (ZF) model. In vivo experiments revealed that ROT-exposed ZF larvae exhibited reduced locomotor activity, increased edge preference, and impaired touch response, while N-CMC treatment significantly improved these behavioral parameters. The reactive oxygen species (ROS) levels in ROT-exposed larvae were elevated (37.75 %) but decreased with N-CMC treatment (30.28 %). Apoptosis was also reduced from 38.87 % in ROT-exposed larvae to 16.52 % with N-CMC treatment. In vitro studies using the N2A cell line confirmed N-CMC's neuroprotective effects. In adult ZF, ROT exposure decreased locomotion, and N-CMC treatment reversed these effects, as demonstrated through ToxTrac analysis. Novel Tank and Light/Dark tests showed significant behavioral improvements with N-CMC. Neurotransmitter analysis indicated increased dopamine, GABA, and glutamate levels in ROT-exposed ZF, which were moderated with N-CMC treatment. Gene expression analysis of gap43, syn2a, and tuba1b showed upregulation in ROT-exposed ZF, while N-CMC treatment downregulated these genes. Antioxidant assays demonstrated that ROT decreased SOD, CAT, and GSH levels in the brain, while N-CMC treatment increased these antioxidant levels by 1.3-fold, 7.5-fold, and 1.3-fold, respectively. Histopathology revealed neuronal degeneration in ROT-exposed ZF brains, but N-CMC treatment protected the neuronal loss. This study is the first to explore the neuroprotective and antioxidant properties of N-CMC in a ZF model, indicating its potential therapeutic benefits over conventional ROT-based treatments for PD.
Collapse
Affiliation(s)
- Srinivasan Palaniselvam
- Native Medicine and Marine Pharmacology Laboratory, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu 603 103, India.
| | - Vignesh Narasimman
- Native Medicine and Marine Pharmacology Laboratory, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu 603 103, India; Department of Medical Biotechnology, Karpaga Vinayaga Institute of Medical Sciences and Research Centre, Mathuranthagam, Tamil Nadu 603308, India.
| | - R Vijayashree
- Department of Pathology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu 603103, India.
| | - Saravanan Ramachandran
- Native Medicine and Marine Pharmacology Laboratory, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu 603 103, India.
| |
Collapse
|
2
|
Abraham JN, Rawat D, Srikanth P, Sunny LP, Abraham NM. Alpha-synuclein pathology and Parkinson's disease-related olfactory dysfunctions: an update on preclinical models and therapeutic approaches. Mamm Genome 2025:10.1007/s00335-025-10128-w. [PMID: 40293510 DOI: 10.1007/s00335-025-10128-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 04/03/2025] [Indexed: 04/30/2025]
Abstract
Olfactory dysfunction (OD) is considered one of the early signs of Parkinson's disease (PD), affecting over 90% of PD patients. OD often appears several years before the onset of motor symptoms and is therefore considered an early biomarker of PD. Recent studies have shown that COVID-19 infection might lead to worsening of symptoms and acceleration of disease progression in neurodegenerative disorders, where OD is a common symptom to both. Hence, it is essential to accurately monitor olfactory fitness in clinical settings using any of the currently available olfactory function tests. Even after a quarter of a century of the discovery of α-synuclein (α-syn) pathogenesis in PD, many aspects related to the α-syn pathogenesis in OD remain unknown. Currently, there is no definitive cure for PD; the disease management options include dopaminergic medications, deep brain stimulations, stem cells, and immunotherapy. Generating reliable PD animal models is critical for understanding the molecular pathways and neural circuits affected by disease conditions. This might contribute to the development and validation of new therapeutic approaches. This review discusses the known mechanisms of α-syn aggregated forms causing neuronal death, the recent developments in the PD preclinical models with ODs, and the treatment strategies employed.
Collapse
Affiliation(s)
- Jancy Nixon Abraham
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India.
- Department of Life Sciences, Centre of Excellence in Epigenetics, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh, 201314, India.
| | - Devesh Rawat
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Priyadharshini Srikanth
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Lisni P Sunny
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Nixon M Abraham
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India.
| |
Collapse
|
3
|
Ye H, Li D, Zhang L, Wang Y, Wang C, Jin M, Lin H, Li P, Sun C, Li N. Epicoccin A Ameliorates PD-like Symptoms in Zebrafish: Enhancement of PINK1/Parkin-Dependent Mitophagy and Inhibition of Excessive Oxidative Stress. Mar Drugs 2025; 23:175. [PMID: 40278296 PMCID: PMC12028493 DOI: 10.3390/md23040175] [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: 03/12/2025] [Revised: 03/30/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder, yet effective agents for its prevention and therapy remain highly limited. Epicoccin A, a significant secondary metabolite from Exserohilum sp., demonstrates various biological activities; however, its neuroprotective effects have not been elucidated. Here, we investigated the therapeutic potential of epicoccin A for PD by evaluating its impact on neural phenotype, reactive oxygen species (ROS) generation, and locomotor activity in PD-like zebrafish. Transcriptomic analysis and molecular docking were conducted, with key gene expressions further verified using real-time qPCR. As a result, epicoccin A notably mitigated dopaminergic neuron loss, neural vasculature deficiency, nervous system injury, ROS accumulation, locomotor impairments, and abnormal expressions of hallmark genes associated with PD and oxidative stress. Underlying mechanism investigation indicated epicoccin A may alleviate PD-like symptoms by activating PINK1/Parkin-dependent mitophagy, as evidenced by the reversal of aberrant gene expressions related to the pink1/parkin pathway and its upstream mTOR/FoxO pathway following epicoccin A co-treatments. This finding was further confirmed by the robust interactions between epicoccin A and these mitophagy regulators. Our results suggest that epicoccin A relieves PD symptoms by activating pink1/parkin-dependent mitophagy and inhibiting excessive oxidative stress, highlighting its potential as a therapeutic approach for PD.
Collapse
Affiliation(s)
- Haicheng Ye
- Shandong Academy of Sciences & Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology, Jinan 250000, China; (H.Y.); (D.L.); (M.J.); (P.L.); (C.S.)
| | - Dan Li
- Shandong Academy of Sciences & Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology, Jinan 250000, China; (H.Y.); (D.L.); (M.J.); (P.L.); (C.S.)
| | - Lei Zhang
- Shandong Overseas Fisheries Association, Jinan 250000, China;
| | - Yufei Wang
- Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission & Guangxi Key Laboratory of Chemistry and Engineering of Forest Products/Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, Guangxi Minzu University, Nanning 530000, China; (Y.W.); (C.W.)
| | - Cong Wang
- Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission & Guangxi Key Laboratory of Chemistry and Engineering of Forest Products/Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, Guangxi Minzu University, Nanning 530000, China; (Y.W.); (C.W.)
| | - Meng Jin
- Shandong Academy of Sciences & Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology, Jinan 250000, China; (H.Y.); (D.L.); (M.J.); (P.L.); (C.S.)
| | - Houwen Lin
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China;
| | - Peihai Li
- Shandong Academy of Sciences & Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology, Jinan 250000, China; (H.Y.); (D.L.); (M.J.); (P.L.); (C.S.)
| | - Chen Sun
- Shandong Academy of Sciences & Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology, Jinan 250000, China; (H.Y.); (D.L.); (M.J.); (P.L.); (C.S.)
| | - Ning Li
- Shandong Academy of Sciences & Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology, Jinan 250000, China; (H.Y.); (D.L.); (M.J.); (P.L.); (C.S.)
| |
Collapse
|
4
|
Green CS, Morris JM, Magnuson JT, Leads RR, Lay CR, Gielazyn M, Rosman L, Schlenk D, Roberts AP. Exposure to the Polychlorinated biphenyl mixture Aroclor 1254 elicits neurological and cardiac developmental effects in early life stage zebrafish (Danio rerio). CHEMOSPHERE 2025; 371:144023. [PMID: 39724984 DOI: 10.1016/j.chemosphere.2024.144023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/25/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
The goal of this study was to compare the bioaccumulation of the PCB mixture Aroclor 1254 in zebrafish to cardiac and neurologic outcomes. The establishment of effect concentrations (ECs) for cardiac and neurotoxic effects of PCBs in early life stage fish is challenging due to a lack of measured PCB concentrations in test media (e.g., fish tissue), the lack of standard exposure methods, and the propensity of PCBs to adsorb to test glassware and materials resulting in discrepancies in ECs from different studies with similar endpoints. Reporting tissue concentrations in test organisms will allow for standardization across different tests and thus may improve estimations of effect thresholds. Early life stage zebrafish (Danio rerio) are a common environmental toxicological model well represented within the literature, making them ideal for comparisons across multiple studies. Embryos were exposed at 6 h post fertilization (hpf) to aqueous Aroclor 1254 for 96 h with or without renewal in addition to a PCB 126 positive control for cardiotoxicity. PCB concentrations were measured in both exposure solutions and tissue samples. Measured concentrations of Aroclor 1254 in test solutions ranged from 8.7% to 870% of nominal concentrations. Heart rate, pericardial edema, and neurological endpoints (eye tremors) were measured in 102 hpf larvae. Pericardial edema was not present in Aroclor 1254-treated zebrafish but was observed in those exposed to PCB-126. Concentration-dependent bradycardia was observed in zebrafish exposed to Aroclor 1254 and PCB-126. Similarly, a concentration-dependent increase in eye tremor behavior was observed in embryos exposed to Aroclor 1254. Data produced by this study demonstrate novel toxicological effects of Aroclor 1254 and highlight the importance of measuring PCBs in both exposure and receptor media.
Collapse
Affiliation(s)
- Corey S Green
- Eastern New Mexico University, Department of Biological Sciences, 1500 Ave. K, Portales, NM, 88130, USA.
| | | | - Jason T Magnuson
- U.S. Geological Survey, Columbia Environmental Research Center, Columbia, MO, 65201, USA.
| | - Rachel R Leads
- Michigan State University, Department of Fisheries and Wildlife, East Lansing, MI, 48825, USA.
| | | | - Michel Gielazyn
- National Oceanic and Atmospheric Administration, Assessment and Restoration Division, St. Petersburg, FL, 33701, USA.
| | - Lisa Rosman
- National Oceanic and Atmospheric Administration, Assessment and Restoration Division, New York, NY, 10278, USA.
| | - Daniel Schlenk
- University of California Riverside, Department of Environmental Science, Riverside, CA, 92521, USA.
| | - Aaron P Roberts
- University of North Texas, Department of Biological Sciences and Advanced Environmental Research Institute, Denton, TX, 76203, USA.
| |
Collapse
|
5
|
Liu X, Gong Q, Deng X, Li L, Luo R, Li X, Guo D, Deng F. UHPLC-Q/Orbitrap HRMS combined with spectrum-effect relationship and network pharmacology to discovery the gastrointestinal motility-promoting material basis in Citri Sarcodactylis Fructus. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118926. [PMID: 39393559 DOI: 10.1016/j.jep.2024.118926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The prevalence of gastrointestinal motility disorders (GMD) is increasing and is characterized by long-term recurrence. Citri Sarcodactylis Fructus (CSF), as a traditional Chinese medicine (TCM) known in "regulating qi and harmonizing the stomach", has therapeutic effects on GMD. However, the material basis of its efficacy is not clear. AIM OF THE STUDY The aim of this study was to evaluate the gastrointestinal motility-promoting activity of CSF extracts and to screen their active ingredients and to perform a preliminary validation. METHODS The chemical composition spectrum of different extracts of CSF were established by ultra high-performance liquid chromatography coupled with quadrupole orbitrap high-resolution mass spectrometry (UHPLC-Q/Orbitrap HRMS). The gastrointestinal motility-promoting activities of CSF were investigated by determining the intestinal propulsion rate, gastric emptying rate, acetylcholinesterase activity, and motilin content in L-arginine-induced GMD mice. Spectrum-effect relationship and network pharmacology analysis were used for the screening of potential active ingredients. A zebrafish gastrointestinal motility model traced with Nile Red was established to validate the active ingredients. Molecular docking prediction was used to explore the mechanism of action of the active ingredient. Finally, Western blotting and TUNEL staining were performed to validate the molecular docking predictions. RESULTS In total, 42 shared components were identified. The main active fraction of CSF to promote gastrointestinal motility was 70% ethanol elution fraction. Eleven potential active ingredients were screened by grey correlation analysis, orthogonal partial least squares analysis, and "active ingredient-target" network. Six compounds were confirmed as the pharmacodynamic substances of CSF by zebrafish gastrointestinal motility model, namely, quercetin, kaempferol, isorhamnetin, diosmetin, hesperetin, and 5,7,3'-trihydroxy-6,4',5'-trimethoxyflavone. Molecular docking predictions and Western blotting assays indicated that CSF may act on AKT and MMP9 targets to exert gastrointestinal motility-promoting activity. CONCLUSION This study provided a foundation for elucidating the gastrointestinal motility-promoting activity of CSF and its material basis by integrating spectrum-effect relationship and network pharmacology. It also provided a theoretical basis for quality control of CSF and a new idea for the discovery and validation of pharmacodynamic substances in TCM.
Collapse
Affiliation(s)
- Xin Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qianqian Gong
- State Key Laboratory of Southwestern Chinese Medicine Resources, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xianglan Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Longxuan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ruiyi Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xuemin Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dale Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Fang Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
6
|
Ranasinghe T, Seo Y, Park HC, Choe SK, Cha SH. Rotenone exposure causes features of Parkinson`s disease pathology linked with muscle atrophy in developing zebrafish embryo. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136215. [PMID: 39461288 DOI: 10.1016/j.jhazmat.2024.136215] [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: 07/31/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024]
Abstract
Parkinson's disease (PD) is associated with both genetic and environmental factors; however, sporadic forms of PD account for > 90 % of cases, and PD prevalence has doubled in the past 25 years. Depending on the importance of the environmental factors, various neurotoxins are used to induce PD both in vivo and in vitro. Unlike other neurodegenerative diseases, PD can be induced in vivo using specific neurotoxic chemicals. However, no chemically induced PD model is available because of the sporadic nature of PD. Rotenone is a pesticide that accelerates the induction of PD and exhibits the highest toxicity in fish, unlike other pesticides. Therefore, in this study, we aimed to establish a model exhibiting PD pathologies such as dysfunction of DArgic neuron, aggregation of ɑ-synuclein, and behavioral abnormalities, which are known features of PD pathology, by rotenone exposure at an environmentally relevant concentration (30 nM) in developing zebrafish embryos. Our results provide direct evidence for the association between PD and muscle degeneration by confirming rotenone-induced muscle atrophy. Therefore, we conclude that the rotenone-induced model presents non-motor and motor defects with extensive studies related to muscle atrophy.
Collapse
Affiliation(s)
- Thilini Ranasinghe
- Department of Marine Bio and Medical Sciences, Hanseo University, Seosan-si 31962, Republic of Korea
| | - Yongbo Seo
- Department of Biomedical Sciences, Korea University, Ansan 15328, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, Korea University, Ansan 15328, Republic of Korea
| | - Seong-Kyu Choe
- Department of Microbiology, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea; Sacopenia Total Solution Center, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea
| | - Seon-Heui Cha
- Department of Marine Bio and Medical Sciences, Hanseo University, Seosan-si 31962, Republic of Korea; Department of Aquatic Life Medicine, Hanseo University, Seosan-si 31962, Republic of Korea; Institute for International Fisheries Science, Hanseo University, Seosan-si 31962, Republic of Korea.
| |
Collapse
|
7
|
Siddiqui S, Liu F, Kanthasamy AG, McGrail M. Stat3 mediates Fyn kinase-driven dopaminergic neurodegeneration and microglia activation. Dis Model Mech 2024; 17:dmm052011. [PMID: 39641161 PMCID: PMC11646115 DOI: 10.1242/dmm.052011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/17/2024] [Indexed: 12/07/2024] Open
Abstract
The Alzheimer's disease and Parkinson's disease risk locus FYN kinase is implicated in neurodegeneration and inflammatory signaling. To investigate in vivo mechanisms of Fyn-driven neurodegeneration, we built a zebrafish neural-specific Gal4:UAS model of constitutively active FynY531F signaling. Using in vivo live imaging, we demonstrated that neural FynY531F expression leads to dopaminergic neuron loss and mitochondrial aggregation in 5 day larval brain. Dopaminergic loss coincided with microglia activation and induction of tnfa, il1b and il12a inflammatory cytokine expression. Transcriptome analysis revealed Stat3 signaling as a potential Fyn target. Chemical inhibition experiments confirmed Fyn-driven dopaminergic neuron loss, and the inflammatory response was dependent upon activation of Stat3 and NF-κB pathways. Dual chemical inhibition demonstrated that Stat3 acts synergistically with NF-κB in dopaminergic neuron degeneration. These results identify Stat3 as a novel downstream effector of Fyn signaling in neurodegeneration and inflammation.
Collapse
Affiliation(s)
- Sahiba Siddiqui
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, USA
- Interdepartmental Genetics and Genomics Graduate Program (IGG), Iowa State University, Ames, IA 50011, USA
| | - Fang Liu
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, USA
| | - Anumantha G. Kanthasamy
- Center for Brain Science and Neurodegenerative Diseases, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA
| | - Maura McGrail
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, USA
- Interdepartmental Genetics and Genomics Graduate Program (IGG), Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
8
|
von Hellfeld R, Gade C, Leist M, Braunbeck T. Rearing conditions (isolated versus group rearing) affect rotenone-induced changes in the behavior of zebrafish (Danio rerio) embryos in the coiling assay. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:55624-55635. [PMID: 39240433 DOI: 10.1007/s11356-024-34870-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
Under regulations such as REACH, testing of novel and established compounds for their (neuro)toxic potential is a legal requirement in many countries. These are largely based on animal-, cost-, and time-intensive in vivo models, not in line with the 3 Rs' principle of animal experimentation. Thus, the development of alternative test methods has also received increasing attention in neurotoxicology. Such methods focus either on physiological alterations in brain development and neuronal pathways or on behavioral changes. An example of a behavioral developmental neurotoxicity (DNT) assay is the zebrafish (Danio rerio) embryo coiling assay, which quantifies effects of compounds on the development of spontaneous movement of zebrafish embryos. While the importance of embryo-to-embryo contact prior to hatching in response to environmental contaminants or natural threats has been documented for many other clutch-laying fish species, little is known about the relevance of intra-clutch contacts for zebrafish. Here, the model neurotoxin rotenone was used to assess the effect of grouped versus separate rearing of the embryos on the expression of the coiling behavior. Some group-reared embryos reacted with hyperactivity to the exposure, to an extent that could not be recorded effectively with the utilized software. Separately reared embryos showed reduced activity, compared with group-reared individuals when assessing. However, even the control group embryos of the separately reared cohort showed reduced activity, compared with group-reared controls. Rotenone could thus be confirmed to induce neurotoxic effects in zebrafish embryos, yet modifying one parameter in an otherwise well-established neurotoxicity assay such as the coiling assay may lead to changes in behavior influenced by the proximity between individual embryos. This indicates a complex dependence of the outcome of behavior assays on a multitude of environmental parameters.
Collapse
Affiliation(s)
- Rebecca von Hellfeld
- Centre for Organismal Studies, Aquatic Ecology and Toxicology, University of Heidelberg, 69120, Heidelberg, Germany.
- School of Biological Sciences, University of Aberdeen, Aberdeen, AB24 3UU, UK.
| | - Christoph Gade
- Centre for Organismal Studies, Aquatic Ecology and Toxicology, University of Heidelberg, 69120, Heidelberg, Germany
- School of Biological Sciences, University of Aberdeen, Aberdeen, AB24 3UU, UK
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated By the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457, Constance, Germany
- CAAT Europe, University of Konstanz, 78457, Constance, Germany
| | - Thomas Braunbeck
- Centre for Organismal Studies, Aquatic Ecology and Toxicology, University of Heidelberg, 69120, Heidelberg, Germany
| |
Collapse
|
9
|
Lai Y, Ay M, Hospital CD, Miller GW, Sarkar S. Seminar: Functional Exposomics and Mechanisms of Toxicity-Insights from Model Systems and NAMs. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:94201. [PMID: 39230330 PMCID: PMC11373422 DOI: 10.1289/ehp13120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/22/2024] [Accepted: 08/12/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Significant progress has been made over the past decade in measuring the chemical components of the exposome, providing transformative population-scale frameworks in probing the etiologic link between environmental factors and disease phenotypes. While the analytical technologies continue to evolve with reams of data being generated, there is an opportunity to complement exposome-wide association studies (ExWAS) with functional analyses to advance etiologic search at organismal, cellular, and molecular levels. OBJECTIVES Exposomics is a transdisciplinary field aimed at enabling discovery-based analysis of the nongenetic factors that contribute to disease, including numerous environmental chemical stressors. While advances in exposure assessment are enhancing population-based discovery of exposome-wide effects and chemical exposure agents, functional screening and elucidation of biological effects of exposures represent the next logical step toward precision environmental health and medicine. In this work, we focus on the use, strategies, and prospects of alternative approaches and model systems to enhance the current human exposomics framework in biomarker search and causal understanding, spanning from bench-based nonmammalian organisms and cell culture to computational new approach methods (NAMs). DISCUSSION We visit the definition of the functional exposome and exposomics and discuss a need to leverage alternative models as opposed to mammalian animals for delineating exposome-wide health effects. Under the "three Rs" principle of reduction, replacement, and refinement, model systems such as roundworms, fruit flies, zebrafish, and induced pluripotent stem cells (iPSCs) are advantageous over mammals (e.g., rodents or higher vertebrates). These models are cost-effective, and cell-specific genetic manipulations in these models are easier and faster, compared to mammalian models. Meanwhile, in silico NAMs enhance hazard identification and risk assessment in humans by bridging the translational gaps between toxicology data and etiologic inference, as represented by in vitro to in vivo extrapolation (IVIVE) and integrated approaches to testing and assessment (IATA) under the adverse outcome pathway (AOP) framework. Together, these alternatives offer a strong toolbox to support functional exposomics to study toxicity and causal mediators underpinning exposure-disease links. https://doi.org/10.1289/EHP13120.
Collapse
Affiliation(s)
- Yunjia Lai
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Muhammet Ay
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Carolina Duarte Hospital
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Gary W. Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Souvarish Sarkar
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, USA
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
10
|
Romero A, Sanchez A, Jones JD, Ledesma K, El-Halawany MS, Hamouda AK, Bill BR. Optimization of Zebrafish Larvae 6-OHDA Exposure for Neurotoxin Induced Dopaminergic Marker Reduction. Zebrafish 2024; 21:287-293. [PMID: 38608227 PMCID: PMC11876810 DOI: 10.1089/zeb.2023.0098] [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] [Indexed: 04/14/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that is clinically assessed by motor symptoms associated with the loss of midbrain dopaminergic neurons affecting the quality of life for over 8.5 million people worldwide. The neurotoxin 6-hydroxydopamine (6-OHDA) has been used to chemically induce a PD-like state in zebrafish larvae by several laboratories; however, highly variable concentration, methodology, and reagents have resulted in conflicting results suggesting a need to investigate these issues of reproducibility. We propose a protocol that addresses the differences in methodology and induces changes in 6 days postfertilization (dpf) larvae utilizing a 24-h exposure at 3 dpf with 30 μM 6-OHDA. Despite ∼50% lethality, no morphological or development differences in surviving fish are observed. Definition of our model is defined by downregulation of the expression of th1 by reverse transcriptase-quantitative polymerase chain reaction, a marker for dopaminergic neurons and a reduction in movement. Additionally, we observed a downregulation of pink1 and an upregulation of sod1 and sod2, indicators of mitochondrial dysfunction and response to reactive oxygen species, respectively.
Collapse
Affiliation(s)
- Adrian Romero
- The University of Texas at Tyler College of Arts and Sciences Department of Biology, Tyler, Texas, USA
- The University of Texas Tyler Ben and Maytee Fisch College of Pharmacy, Tyler, Texas, USA
| | - Armando Sanchez
- The University of Texas at Tyler College of Arts and Sciences Department of Biology, Tyler, Texas, USA
| | - Jocelyn D. Jones
- The University of Texas at Tyler College of Arts and Sciences Department of Biology, Tyler, Texas, USA
| | - Kristel Ledesma
- The University of Texas at Tyler College of Arts and Sciences Department of Biology, Tyler, Texas, USA
| | - Medhat S. El-Halawany
- The University of Texas Tyler Ben and Maytee Fisch College of Pharmacy, Tyler, Texas, USA
| | - Ayman K. Hamouda
- The University of Texas Tyler Ben and Maytee Fisch College of Pharmacy, Tyler, Texas, USA
| | - Brent R. Bill
- The University of Texas at Tyler College of Arts and Sciences Department of Biology, Tyler, Texas, USA
| |
Collapse
|
11
|
Nies YH, Yahaya MF, Lim WL, Teoh SL. Microarray-based Analysis of Differential Gene Expression Profile in Rotenone-induced Parkinson's Disease Zebrafish Model. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:761-772. [PMID: 37291778 DOI: 10.2174/1871527322666230608122552] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 05/04/2023] [Accepted: 05/04/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND & OBJECTIVES Despite much clinical and laboratory research that has been performed to explore the mechanisms of Parkinson's disease (PD), its pathogenesis remains elusive to date. Therefore, this study aimed to identify possible regulators of neurodegeneration by performing microarray analysis of the zebrafish PD model's brain following rotenone exposure. METHODS A total of 36 adult zebrafish were divided into two groups: control (n = 17) and rotenonetreated (n = 19). Fish were treated with rotenone water (5 μg/L water) for 28 days and subjected to locomotor behavior analysis. Total RNA was extracted from the brain tissue after rotenone treatment. The cDNA synthesized was subjected to microarray analysis and subsequently validated by qPCR. RESULTS Administration of rotenone has significantly reduced locomotor activity in zebrafish (p < 0.05), dysregulated dopamine-related gene expression (dat, th1, and th2, p < 0.001), and reduced dopamine level in the brain (p < 0.001). In the rotenone-treated group, genes involved in cytotoxic T lymphocytes (gzm3, cd8a, p < 0.001) and T cell receptor signaling (themis, lck, p < 0.001) were upregulated significantly. Additionally, gene expression involved in microgliosis regulation (tyrobp, p < 0.001), cellular response to IL-1 (ccl34b4, il2rb, p < 0.05), and regulation of apoptotic process (dedd1, p < 0.001) were also upregulated significantly. CONCLUSION The mechanisms of T cell receptor signaling, microgliosis regulation, cellular response to IL-1, and apoptotic signaling pathways have potentially contributed to PD development in rotenonetreated zebrafish.
Collapse
Affiliation(s)
- Yong Hui Nies
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Wei Ling Lim
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Selangor, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
12
|
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
|
13
|
Gao X, Zhang B, Zheng Y, Liu X, Rostyslav P, Finiuk N, Sik A, Stoika R, Liu K, Jin M. Neuroprotective effect of chlorogenic acid on Parkinson's disease like symptoms through boosting the autophagy in zebrafish. Eur J Pharmacol 2023; 956:175950. [PMID: 37544423 DOI: 10.1016/j.ejphar.2023.175950] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/19/2023] [Accepted: 08/01/2023] [Indexed: 08/08/2023]
Abstract
Parkinson's disease (PD) is characterized by both motor and non-motor symptoms, including hypokinesia, postural instability, dopaminergic (DA) neurons loss, and α-synuclein (α-syn) accumulation. A growing number of patients show negative responses towards the current therapies. Thus, preventative or disease-modifying treatment agents are worth to further research. In recent years, compounds extracted from natural sources become promising candidates to treat PD. Chlorogenic acid (CGA) is a phenolic compound appearing in coffee, honeysuckle, and eucommia that showed their potential as antioxidants and neuroprotectors. In this study, we investigated the anti-PD activity of CGA by testing its effect on 1-methyl-4-phenyl-1-1,2,3,6-tetrahydropyridine (MPTP) zebrafish model of PD. It was shown that CGA relieved MPTP-induced PD-like symptoms including DA neurons and blood vessel loss, locomotion reduction, and apoptosis events in brain. Moreover, CGA modulated the expression of PD- and autophagy-related genes (α-syn, lc3b, p62, atg5, atg7, and ulk1b), showing its ability to promote the autophagy which was interrupted in the PD pathology. The unblocked effect of CGA on autophagy was further verified in 6-hydroxydopamine (6-OHDA)-modeled SHSY5Y cells. Our findings indicated that CGA might relieve PD by boosting the autophagy in neuronal cells that makes CGA a potential candidate for anti-PD treatment.
Collapse
Affiliation(s)
- Xin Gao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Baoyue Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Yuanteng Zheng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Xuchang Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Shandong Provincial Hospital Affiliated to Shandong First Medical University, 16766 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Panchuk Rostyslav
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, National Academy of Sciences of Ukraine, Lviv, 79005, Ukraine
| | - Nataliya Finiuk
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, National Academy of Sciences of Ukraine, Lviv, 79005, Ukraine
| | - Attila Sik
- Institute of Physiology, Medical School, University of Pecs, Pecs, H-7624, Hungary; Szentagothai Research Centre, University of Pecs, Pecs, H-7624, Hungary; Institute of Clinical Sciences, Medical School, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Rostyslav Stoika
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, National Academy of Sciences of Ukraine, Lviv, 79005, Ukraine
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China.
| |
Collapse
|
14
|
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: 61] [Impact Index Per Article: 30.5] [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
|
15
|
da Cruz Guedes E, Erustes AG, Leão AHFF, Carneiro CA, Abílio VC, Zuardi AW, Hallak JEC, Crippa JA, Bincoletto C, Smaili SS, Reckziegel P, Pereira GJS. Cannabidiol Recovers Dopaminergic Neuronal Damage Induced by Reserpine or α-synuclein in Caenorhabditis elegans. Neurochem Res 2023:10.1007/s11064-023-03905-z. [PMID: 36964823 DOI: 10.1007/s11064-023-03905-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/26/2023]
Abstract
Progressive neurodegenerative disorders such as Parkinson Disease (PD) lack curative or long-term treatments. At the same time, the increase of the worldwide elderly population and, consequently, the extension in the prevalence of age-related diseases have promoted research interest in neurodegenerative disorders. Caenorhabditis elegans is a free-living nematode widely used as an animal model in studies of human diseases. Here we evaluated cannabidiol (CBD) as a possible neuroprotective compound in PD using the C. elegans models exposed to reserpine. Our results demonstrated that CBD reversed the reserpine-induced locomotor alterations and this response was independent of the NPR-19 receptors, an orthologous receptor for central cannabinoid receptor type 1. Morphological alterations of cephalic sensilla (CEP) dopaminergic neurons indicated that CBD also protects neurons from reserpine-induced degeneration. That is, CBD attenuates the reserpine-induced increase of worms with shrunken soma and dendrites loss, increasing the number of worms with intact CEP neurons. Finally, we found that CBD also reduced ROS formation and α-syn protein accumulation in mutant worms. Our findings collectively provide new evidence that CBD acts as neuroprotector in dopaminergic neurons, reducing neurotoxicity and α-syn accumulation highlighting its potential in the treatment of PD.
Collapse
Affiliation(s)
- Erika da Cruz Guedes
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Três de Maio Street, 100, São Paulo, SP, 04044-020, Brazil
| | - Adolfo Garcia Erustes
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Três de Maio Street, 100, São Paulo, SP, 04044-020, Brazil
| | - Anderson H F F Leão
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Três de Maio Street, 100, São Paulo, SP, 04044-020, Brazil
| | - César Alves Carneiro
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Três de Maio Street, 100, São Paulo, SP, 04044-020, Brazil
| | - Vanessa C Abílio
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Três de Maio Street, 100, São Paulo, SP, 04044-020, Brazil
- National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, Brazil
| | - Antonio W Zuardi
- National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, Brazil
- Department of Neuroscience and Behavior, Universidade de São Paulo, USP, Ribeirão Preto, Brazil
| | - Jaime Eduardo C Hallak
- National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, Brazil
- Department of Neuroscience and Behavior, Universidade de São Paulo, USP, Ribeirão Preto, Brazil
| | - José Alexandre Crippa
- National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, Brazil
- Department of Neuroscience and Behavior, Universidade de São Paulo, USP, Ribeirão Preto, Brazil
| | - Claudia Bincoletto
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Três de Maio Street, 100, São Paulo, SP, 04044-020, Brazil
| | - Soraya S Smaili
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Três de Maio Street, 100, São Paulo, SP, 04044-020, Brazil
| | - Patrícia Reckziegel
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Gustavo J S Pereira
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Três de Maio Street, 100, São Paulo, SP, 04044-020, Brazil.
| |
Collapse
|
16
|
Wang X, Shi X, Zheng S, Zhang Q, Peng J, Tan W, Wu K. Perfluorooctane sulfonic acid (PFOS) exposures interfere with behaviors and transcription of genes on nervous and muscle system in zebrafish embryos. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 848:157816. [PMID: 35931148 DOI: 10.1016/j.scitotenv.2022.157816] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/30/2022] [Accepted: 07/31/2022] [Indexed: 02/05/2023]
Abstract
Perfluorooctane sulfonic acid (PFOS) has been widely detected in environment and organisms. PFOS has been identified as the driving agent for the behavioral changes of zebrafish larvae, while the underlying molecular mechanism remains unclear. In this study, zebrafish embryos/larvae were exposed to 0, 0.04, 0.1, 0.4 and 1 μM PFOS for 166 h. The locomotor behaviors and the mRNA transcription of genes in neuromuscular system were detected. Exposure to PFOS did not affect the hatching/death rates and body length, but increased the heart beat rates and frequency of spontaneous tail coiling. Locomotor behavior in zebrafish larvae of 0.4 and 1 μM PFOS groups were increased in the light condition. Additionally, the levels of acetylcholine (Ach) in 0.4 μM PFOS group and dopamine (DA) in 0.1, 0.4 and 1 μM PFOS groups were found to be significantly increased. The expression of genes related to the synthesis and decomposition of ACh,the synthesis and receptor of DA, and fosab was increased in the different PFOS treatment groups, while the expression of all the other genes of the neuromuscular system were significantly reduced. The findings of this investigation demonstrated that PFOS exposure may alter the locomotor behavior of zebrafish through disrupting the expressions of genes in neuromuscular system. The disturbed process of neurotransmitter transmission and muscle contraction caused by PFOS may be the dominant mechanism of hyperactivity in zebrafish.
Collapse
Affiliation(s)
- Xin Wang
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China; Medical Record Statistics Office, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Xiaoling Shi
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Shukai Zheng
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Qiong Zhang
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Jiajun Peng
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Wei Tan
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Kusheng Wu
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China
| |
Collapse
|
17
|
Kim SS, Hwang KS, Kan H, Yang JY, Son Y, Shin DS, Lee BH, Chae CH, Bae MA. Neurotoxicological Profiling of Paraquat in Zebrafish Model. Neurochem Res 2022; 47:2294-2306. [PMID: 35562624 DOI: 10.1007/s11064-022-03615-y] [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: 01/14/2022] [Revised: 03/28/2022] [Accepted: 04/21/2022] [Indexed: 11/27/2022]
Abstract
Paraquat is a polar herbicide protecting plant products against invasive species, it requires careful manipulation and restricted usage because of its harmful potentials. Exposure to paraquat triggers oxidative damage in dopaminergic neurons and subsequently causes a behavioral defect in vivo. Thereby, persistent exposure to paraquat is known to increase Parkinson's disease risk by dysregulating dopaminergic systems in humans. Therefore, most studies have focused on the dopaminergic systems to elucidate the neurotoxicological mechanism of paraquat poisoning, and more comprehensive neurochemistry including histaminergic, serotonergic, cholinergic, and GABAergic systems has remained unclear. Therefore, in this study, we investigated the toxicological potential of paraquat poisoning using a variety of approaches such as toxicokinetic profiles, behavioral effects, neural activity, and broad-spectrum neurochemistry in zebrafish larvae after short-term exposure to paraquat and we performed the molecular modeling approach. Our results showed that paraquat was slowly absorbed in the brain of zebrafish after oral administration of paraquat. In addition, paraquat toxicity resulted in behavioral impairments, namely, reduced motor activity and led to abnormal neural activities in zebrafish larvae. This locomotor deficit came with a dysregulation of dopamine synthesis induced by the inhibition of tyrosine hydroxylase activity, which was also indirectly confirmed by molecular modeling studies. Furthermore, short-term exposure to paraquat also caused simultaneous dysregulation of other neurochemistry including cholinergic and serotonergic systems in zebrafish larvae. The present study suggests that this neurotoxicological profiling could be a useful tool for understanding the brain neurochemistry of neurotoxic agents that might be a potential risk to human and environmental health.
Collapse
Affiliation(s)
- Seong Soon Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Kyu-Seok Hwang
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Hyemin Kan
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Jung Yoon Yang
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Yuji Son
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Dae-Seop Shin
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Byung Hoi Lee
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Chong Hak Chae
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Myung Ae Bae
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea.
- Department of Medicinal Chemistry and Pharmacology, University of Science & Technology, Daejeon, Republic of Korea.
| |
Collapse
|
18
|
Ren Q, Jiang X, Paudel YN, Gao X, Gao D, Zhang P, Sheng W, Shang X, Liu K, Zhang X, Jin M. Co-treatment with natural HMGB1 inhibitor Glycyrrhizin exerts neuroprotection and reverses Parkinson's disease like pathology in Zebrafish. JOURNAL OF ETHNOPHARMACOLOGY 2022; 292:115234. [PMID: 35358621 DOI: 10.1016/j.jep.2022.115234] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 02/28/2022] [Accepted: 03/23/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Parkinson's disease (PD) is the second most devastating age-related neurodegenerative diseases after Alzheimer diseases (AD) and is characterized by the loss of dopaminergic (DA) neurons in the substantia nigra (SN) and aggregation of α-synuclein (α-syn). The precise etiology of PD is not yet fully understood and lacks the disease-modifying therapeutic strategies that could reverse the ongoing neurodegeneration. In the quest of exploring novel disease modifying therapeutic strategies, natural compounds from plant sources have gained much attention in recent days. Glycyrrhizin (GL) is the main active ingredient of the roots and rhizomes of licorice (Glycyrrhiza glabra L), which are generally used in the treatment of inflammatory diseases or as a tonifying herbal medicine. In Persia, GL is a conventional neuroprotective agent that are used to treat neurological disorders. The traditional use of GL in Japan is to treat chronic hepatitis B. In addition, GL is a natural inhibitor of high mobility group box 1 (HMGB1) which has exerted neuroprotective effect against several HMGB1 mediated pathological conditions. AIM OF THE STUDY The study is aimed to evaluate therapeutic effect of GL against PD in zebrafish. MATERIAL AND METHODS PD in zebrafish larvae is induced by administration of neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Apoptosis was assessed with TUNEL assay. Gene expression was performed to assess the modulation in genes related to neuroinflammatory and autophagy. RESULTS We observed that GL co-treatment increased the length of DA neurons, decreased the number of apoptotic cells in zebrafish brain, and inhibited the loss of vasculature and disorganized vasculature induced by MPTP. GL co-treatment relieved the MPTP-induced locomotor impairment in zebrafish. GL co-treatment suppressed MPTP-induced upregulated mRNA expression of inflammatory markers such as hmgb1a, tlr4b, nfκb, il1β, and il6. GL co-treatment suppressed the autophagy related genes α-syn and atg5 whereas increased the mRNA expression level of parkin and pink1. In addition, molecular docking study reveals that GL has binding interaction with HMGB1, TLR4, and RAGE. CONCLUSION Hence, the effect of GL co-treatment on MPTP-induced PD-like condition in zebrafish is to alleviate apoptosis and autophagy, as well as suppress inflammatory responses.
Collapse
Affiliation(s)
- Qingyu Ren
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; School of Psychology, North China University of Science and Technology, 21 Bohai Road, Tang'shan, 063210, Hebei Province, PR China
| | - Xin Jiang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, Ji'nan, 250353, Shandong Province, PR China
| | - Yam Nath Paudel
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500, Selangor, Malaysia
| | - Xin Gao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Daili Gao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Pengyu Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; School of Psychology, North China University of Science and Technology, 21 Bohai Road, Tang'shan, 063210, Hebei Province, PR China
| | - Wenlong Sheng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Xueliang Shang
- School of Psychology, North China University of Science and Technology, 21 Bohai Road, Tang'shan, 063210, Hebei Province, PR China
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Xiujun Zhang
- School of Psychology, North China University of Science and Technology, 21 Bohai Road, Tang'shan, 063210, Hebei Province, PR China.
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China.
| |
Collapse
|
19
|
Cerebroventricular Injection of Pgk1 Attenuates MPTP-Induced Neuronal Toxicity in Dopaminergic Cells in Zebrafish Brain in a Glycolysis-Independent Manner. Int J Mol Sci 2022; 23:ijms23084150. [PMID: 35456967 PMCID: PMC9025024 DOI: 10.3390/ijms23084150] [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: 02/02/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 12/26/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by the degeneration of dopaminergic neurons. While extracellular Pgk1 (ePgk1) is reported to promote neurite outgrowth, it remains unclear if it can affect the survival of dopaminergic cells. To address this, we employed cerebroventricular microinjection (CVMI) to deliver Pgk1 into the brain of larvae and adult zebrafish treated with methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) as a PD-like model. The number of dopamine-producing cells in ventral diencephalon clusters of Pgk1-injected, MPTP-treated embryos increased over that of MPTP-treated embryos. Swimming distances of Pgk1-injected, MPTP-treated larvae and adult zebrafish were much longer compared to MPTP-treated samples. The effect of injected Pgk1 on both dopamine-producing cells and locomotion was time- and dose-dependent. Indeed, injected Pgk1 could be detected, located on dopamine neurons. When the glycolytic mutant Pgk1, Pgk1-T378P, was injected into the brain of MPTP-treated zebrafish groups, the protective ability of dopaminergic neurons did not differ from that of normal Pgk1. Therefore, ePgk1 is functionally independent from intracellular Pgk1 serving as an energy supplier. Furthermore, when Pgk1 was added to the culture medium for culturing dopamine-like SH-SY5Y cells, it could reduce the ROS pathway and apoptosis caused by the neurotoxin MPP+. These results show that ePgk1 benefits the survival of dopamine-producing cells and decreases neurotoxin damage.
Collapse
|
20
|
Doyle JM, Croll RP. A Critical Review of Zebrafish Models of Parkinson's Disease. Front Pharmacol 2022; 13:835827. [PMID: 35370740 PMCID: PMC8965100 DOI: 10.3389/fphar.2022.835827] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/08/2022] [Indexed: 11/17/2022] Open
Abstract
A wide variety of human diseases have been modelled in zebrafish, including various types of cancer, cardiovascular diseases and neurodegenerative diseases like Alzheimer’s and Parkinson’s. Recent reviews have summarized the currently available zebrafish models of Parkinson’s Disease, which include gene-based, chemically induced and chemogenetic ablation models. The present review updates the literature, critically evaluates each of the available models of Parkinson’s Disease in zebrafish and compares them with similar models in invertebrates and mammals to determine their advantages and disadvantages. We examine gene-based models, including ones linked to Early-Onset Parkinson’s Disease: PARKIN, PINK1, DJ-1, and SNCA; but we also examine LRRK2, which is linked to Late-Onset Parkinson’s Disease. We evaluate chemically induced models like MPTP, 6-OHDA, rotenone and paraquat, as well as chemogenetic ablation models like metronidazole-nitroreductase. The article also reviews the unique advantages of zebrafish, including the abundance of behavioural assays available to researchers and the efficiency of high-throughput screens. This offers a rare opportunity for assessing the potential therapeutic efficacy of pharmacological interventions. Zebrafish also are very amenable to genetic manipulation using a wide variety of techniques, which can be combined with an array of advanced microscopic imaging methods to enable in vivo visualization of cells and tissue. Taken together, these factors place zebrafish on the forefront of research as a versatile model for investigating disease states. The end goal of this review is to determine the benefits of using zebrafish in comparison to utilising other animals and to consider the limitations of zebrafish for investigating human disease.
Collapse
Affiliation(s)
- Jillian M Doyle
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Roger P Croll
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
21
|
Thirugnanam T, Santhakumar K. Chemically induced models of Parkinson's disease. Comp Biochem Physiol C Toxicol Pharmacol 2022; 252:109213. [PMID: 34673252 DOI: 10.1016/j.cbpc.2021.109213] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/30/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022]
Abstract
Environmental toxins are harmful substances detrimental to humans. Constant exposure to these fatal neurotoxins can cause various neurodegenerative disorders. Although poisonous, specific neurotoxins at optimal concentrations mimic the clinical features of neurodegenerative diseases in several animal models. Such chemically-induced model systems are beneficial in deciphering the molecular mechanisms of neurodegeneration and drug screening for these disorders. One such neurotoxin is 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a widely used chemical that recapitulates Parkinsonian features in various animal models. Apart from MPTP, other neurotoxins like 6-hydroxydopamine (6-OHDA), paraquat, rotenone also induce specific clinical features of Parkinson's disease in animal models. These chemically-induced Parkinson's disease models are playing a crucial role in understanding Parkinson's disease onset, pathology, and novel therapeutics. In this review, we provide a concise overview of various neurotoxins that can recapitulate Parkinsonian features in different in vivo and in vitro model systems specifically focusing on the different treatment methodologies of neurotoxins.
Collapse
Affiliation(s)
- Thilaga Thirugnanam
- Zebrafish Genetics Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Kirankumar Santhakumar
- Zebrafish Genetics Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
22
|
Zebrafish, Medaka and Turquoise Killifish for Understanding Human Neurodegenerative/Neurodevelopmental Disorders. Int J Mol Sci 2022; 23:ijms23031399. [PMID: 35163337 PMCID: PMC8836067 DOI: 10.3390/ijms23031399] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/14/2022] [Accepted: 01/24/2022] [Indexed: 12/21/2022] Open
Abstract
In recent years, small fishes such as zebrafish and medaka have been widely recognized as model animals. They have high homology in genetics and tissue structure with humans and unique features that mammalian model animals do not have, such as transparency of embryos and larvae, a small body size and ease of experiments, including genetic manipulation. Zebrafish and medaka have been used extensively in the field of neurology, especially to unveil the mechanisms of neurodegenerative diseases such as Parkinson's and Alzheimer's disease, and recently, these fishes have also been utilized to understand neurodevelopmental disorders such as autism spectrum disorder. The turquoise killifish has emerged as a new and unique model animal, especially for ageing research due to its unique life cycle, and this fish also seems to be useful for age-related neurological diseases. These small fishes are excellent animal models for the analysis of human neurological disorders and are expected to play increasing roles in this field. Here, we introduce various applications of these model fishes to improve our understanding of human neurological disorders.
Collapse
|
23
|
Ren Q, Jiang X, Zhang S, Gao X, Paudel YN, Zhang P, Wang R, Liu K, Jin M. Neuroprotective effect of YIAEDAER peptide against Parkinson's disease like pathology in zebrafish. Biomed Pharmacother 2022; 147:112629. [PMID: 35030435 DOI: 10.1016/j.biopha.2022.112629] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/16/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic (DA) neurons in the substantia nigra (SN) and aggregation of α-synuclein (α-syn). Current PD therapies merely provide symptomatic relief, lacking the disease-modifying therapeutic strategies against that could reverse the ongoing neurodegeneration. In the quest of exploring novel disease modifying therapeutic strategies, compounds from natural sources have gained much attention in recent days. YIAEDAER (Tyr-Ile-Ala-Glu-Asp-Ala-Glu-Arg) peptide is a multi-functional peptide isolated and purified from the visceral mass extract of Neptunea arthritica cumingii (NAC) with plethora of pharmacological activities, however its neuroprotective effect against MPTP induced PD model is not yet reported. We found YIAEDAER peptide co-treatment could suppressed the MPTP-induced locomotor impairment in zebrafish, ameliorates the MPTP induced degeneration of DA neurons, inhibited the loss of vasculature and loss of cerebral vessels, suppressed α-syn levels. Moreover, YIAEDAER peptide modulates several genes related to autophagy (α-syn, pink1, parkin, atg5, atg7, beclin1, ulk1b, ulk2, and ambra1a), and oxidative stress (sod1, sod2, gss, gpx4a, gsto2, and cat). Hence, our finding suggests that YIAEDAER peptide might be a potential therapeutic candidate against MPTP-induced PD like condition.
Collapse
Affiliation(s)
- Qingyu Ren
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; School of Psychology, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, People's Republic of China
| | - Xin Jiang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Shanshan Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Xin Gao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Biological Engineering College, Qilu University of Technology (Shandong Academy of Sciences), Ji'nan 250056, Shandong Province, People's Republic of China
| | - Yam Nath Paudel
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| | - Pengyu Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; School of Psychology, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, People's Republic of China
| | - Rongchun Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China.
| |
Collapse
|
24
|
Jing W, Zhu M, Wang F, Zhao X, Dong S, Xu Y, Wang S, Yang J, Wang K, Liu W. Hyaluronic Acid-Melatonin Nanoparticles Improve the Dysregulated Intestinal Barrier, Microbiome and Immune Response in Mice with Dextran Sodium Sulfate-Induced Colitis. J Biomed Nanotechnol 2022; 18:175-184. [PMID: 35180910 DOI: 10.1166/jbn.2022.3232] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Although the cause of inflammatory bowel disease (IBD) is unclear, current studies have found that the main factors involved in its pathogenesis include imbalance of mucosal immune response, intestinal dysbiosis, and destruction of the intestinal barrier. We synthesized an amphiphilic conjugate of hyaluronic acid (HA) and melatonin (MT), which have established immunomodulatory and antioxidant properties, by stimulating their nano-aggregation. Inducing colitis by dextran sodium sulfate (DSS), HA-MT accumulated in the inflamed colon epithelium of colitis mice, and markedly improved the colitis symptoms, repaired the damaged intestinal barrier and inhibited colon inflammation. In addition, through bacterial 16S rDNA sequencing, it was found that HA-MT can restore the ratio of Firmicutes/Bacteroidetes by increasing the overall microbial richness and diversity, and alleviate the intestinal dysbiosis of mice with colitis. In the analysis of the intestinal flora at the species level, the abundance of Lactobacillus increased in colitis mice treated with HA-MT while that of Bacteroides, Blautia and Streptococcus decreased in the colitis mice treated with HA-MT. Our findings suggest that the HA-MT system is a promising prebiotic, which can relieve the symptoms of IBD by regulating the intestinal microflora and restoring intestinal homeostasis, inhibiting inflammation.
Collapse
Affiliation(s)
- Wanghui Jing
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Min Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Fushuo Wang
- School of Material Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300350, China
| | - Xinxin Zhao
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Sijing Dong
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yinyue Xu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Sicen Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jianhai Yang
- School of Material Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300350, China
| | - Ke Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Wenguang Liu
- School of Material Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300350, China
| |
Collapse
|
25
|
Kalyn M, Ekker M. Cerebroventricular Microinjections of MPTP on Adult Zebrafish Induces Dopaminergic Neuronal Death, Mitochondrial Fragmentation, and Sensorimotor Impairments. Front Neurosci 2021; 15:718244. [PMID: 34512252 PMCID: PMC8432913 DOI: 10.3389/fnins.2021.718244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/26/2021] [Indexed: 11/21/2022] Open
Abstract
Mitochondria are dynamic organelles that mediate the energetic supply to cells and mitigate oxidative stress through the intricate balance of fission and fusion. Mitochondrial dysfunction is a prominent feature within Parkinson disease (PD) etiologies. To date, there have been conflicting studies of neurotoxin impact on dopaminergic cell death, mitochondrial function and behavioral impairment using adult zebrafish. Here, we performed cerebroventricular microinjections (CVMIs) of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) on adult transgenic zebrafish that resulted in significant reductions in dopaminergic neurons within the telencephalon and olfactory bulbs (OB) of Tg(dat:eGFP) fish. Visualization of mCherry and mitochondrial gene expression analysis in Tg(dat:tom20 MLS:mCherry) fish reveal that MPTP induces mitochondrial fragmentation in dopaminergic neurons and the activation of the pink1/parkin pathway involved mitophagy. Moreover, the loss of dopaminergic neurons translated into a transient locomotor and olfactory phenotype. Taken together, these data can contribute to a better understanding of the mitochondrial impact on dopaminergic survivability.
Collapse
Affiliation(s)
- Michael Kalyn
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON, Canada
| | - Marc Ekker
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
26
|
Mustapha M, Taib CNM. MPTP-induced mouse model of Parkinson's disease: A promising direction of therapeutic strategies. Bosn J Basic Med Sci 2021; 21:422-433. [PMID: 33357211 PMCID: PMC8292858 DOI: 10.17305/bjbms.2020.5181] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/10/2020] [Indexed: 12/23/2022] Open
Abstract
Among the popular animal models of Parkinson's disease (PD) commonly used in research are those that employ neurotoxins, especially 1-methyl- 4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP). This neurotoxin exerts it neurotoxicity by causing a barrage of insults, such as oxidative stress, mitochondrial apoptosis, inflammation, excitotoxicity, and formation of inclusion bodies acting singly and in concert, ultimately leading to dopaminergic neuronal damage in the substantia nigra pars compacta and striatum. The selective neurotoxicity induced by MPTP in the nigrostriatal dopaminergic neurons of the mouse brain has led to new perspectives on PD. For decades, the MPTP-induced mouse model of PD has been the gold standard in PD research even though it does not fully recapitulate PD symptomatology, but it does have the advantages of simplicity, practicability, affordability, and fewer ethical considerations and greater clinical correlation than those of other toxin models of PD. The model has rejuvenated PD research and opened new frontiers in the quest for more novel therapeutic and adjuvant agents for PD. Hence, this review summarizes the role of MPTP in producing Parkinson-like symptoms in mice and the experimental role of the MPTP-induced mouse model. We discussed recent developments of more promising PD therapeutics to enrich our existing knowledge about this neurotoxin using this model.
Collapse
Affiliation(s)
- Musa Mustapha
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor (Darul Ehsan), Malaysia
- Department of Human Anatomy, Faculty of Basic Sciences, College of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Che Norma Mat Taib
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor (Darul Ehsan), Malaysia
| |
Collapse
|
27
|
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
|
28
|
Huang J, Sun L, Mennigen JA, Liu Y, Liu S, Zhang M, Wang Q, Tu W. Developmental toxicity of the novel PFOS alternative OBS in developing zebrafish: An emphasis on cilia disruption. JOURNAL OF HAZARDOUS MATERIALS 2021; 409:124491. [PMID: 33223314 DOI: 10.1016/j.jhazmat.2020.124491] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 06/11/2023]
Abstract
In recent years, sodium p-perfluorous nonenoxybenzene sulfonate (OBS) has emerged as a substitute for PFOS with large demand and application in the Chinese market. However, little is known about potential developmental effects of OBS. In this study, zebrafish embryos were acutely exposed to different concentrations of OBS and the positive control PFOS for a comparative developmental toxicity assessment. OBS caused hatching delays, body axis curvature, neurobehavioral inhibition and abnormal cardiovascular development. These organismal effects were accompanied by change of development related genes expression profile, in which some cases were similar to PFOS. Overall, the toxic effects induced by OBS were generally milder than that of PFOS. Further investigation suggested that both OBS and PFOS disrupted ciliogenesis, evidenced by the ciliary immunostaining, changes in gene expression of kinesin family, dynein arm family and tubulin family members, as well as downregulation of the abundance of motor proteins including KIF3C, DYNC1H1 and DYNC1LI1. The influence of PFOS was stronger than that of OBS on ciliary genes and proteins. Molecular docking analysis revealed that both OBS and PFOS fitted into the motor proteins tightly, but binding affinity between OBS and motor proteins was lower than PFOS. Collectively, OBS and PFOS may act on ciliary motor proteins to interfere with ciliogenesis, leading to ciliary dysfunction and providing a novel probable action mode linked to developmental toxicity. This raises concerns regarding the health risks of the novel PFOS alternative OBS.
Collapse
Affiliation(s)
- Jing Huang
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China; Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330012, China
| | - Liwei Sun
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China
| | | | - Yu Liu
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330012, China
| | - Shuai Liu
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330012, China
| | - Miao Zhang
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330012, China
| | - Qiyu Wang
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330012, China.
| | - Wenqing Tu
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330012, China.
| |
Collapse
|
29
|
Merhi R, Kalyn M, Zhu-Pawlowsky A, Ekker M. Loss of parla Function Results in Inactivity, Olfactory Impairment, and Dopamine Neuron Loss in Zebrafish. Biomedicines 2021; 9:205. [PMID: 33670667 PMCID: PMC7922472 DOI: 10.3390/biomedicines9020205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 01/02/2023] Open
Abstract
The presenilin-associated rhomboid-like (PARL) gene was found to contribute to mitochondrial morphology and function and was linked to familial Parkinson's disease (PD). The PARL gene product is a mitochondrial intramembrane cleaving protease that acts on a number of mitochondrial proteins involved in mitochondrial morphology, apoptosis, and mitophagy. To date, functional and genetic studies of PARL have been mainly performed in mammals. However, little is known about PARL function and its role in dopaminergic (DA) neuron development in vertebrates. The zebrafish genome comprises two PARL paralogs: parla and parlb. Here, we established a loss-of-function mutation in parla via CRISPR/Cas9-mediated mutagenesis. We examined DA neuron numbers in the adult brain and expression of genes associated with DA neuron function in larvae and adults. We show that loss of parla function results in loss of DA neurons, mainly in the olfactory bulb. Changes in the levels of tyrosine hydroxylase transcripts supported this neuronal loss. Expression of fis1, a gene involved in mitochondrial fission, was increased in parla mutants. Finally, we showed that loss of parla function translates into impaired olfaction and altered locomotion parameters. These results suggest a role for parla in the development and/or maintenance of DA neuron function in zebrafish.
Collapse
Affiliation(s)
| | | | | | - Marc Ekker
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (R.M.); (M.K.); (A.Z.-P.)
| |
Collapse
|
30
|
Christensen C, Þorsteinsson H, Maier VH, Karlsson KÆ. Multi-parameter Behavioral Phenotyping of the MPP+ Model of Parkinson's Disease in Zebrafish. Front Behav Neurosci 2021; 14:623924. [PMID: 33390914 PMCID: PMC7775599 DOI: 10.3389/fnbeh.2020.623924] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/30/2020] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease (PD) has been modeled in several animal species using the neurotoxins 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and its oxidized product 1-methyl-4-phenylpyridinium (MPP+). MPP+ selectively kills dopaminergic neurons in pars compacta of the substantia nigra, inducing parkinsonian symptoms in animals. Typically, neurotoxicity models of PD in zebrafish assess acute drug effects on locomotion. In the present study, we examined the lasting effects of MPP+ exposure and drug treatment in zebrafish larvae. Larvae were incubated in 500 μM MPP+, from 1 to 5 days post fertilization (dpf), followed by 24 h drug-free acclimation. At 6 dpf, the behavior was analyzed for locomotion, thigmotaxis, and sleep. Next, in separate assays we assessed the drug effects of brain injected glial cell-derived neurotrophic factor (GDNF) and 4-phenylbutyrate (PBA), co-incubated with MPP+. We show that MPP+ exposure consistently reduces swim distance, movement frequency, and cumulative time of movement; thus mimicking a parkinsonian phenotype of reduced movement. In contrast, MPP+ exposed larvae demonstrate reduced anxiety-like behavior and exhibit a sleep phenotype inconsistent with human PD: the larvae display longer sleep bouts, less sleep fragmentation, and more sleep. Previously reported rescuing effects of PBA were not replicated in this study. Moreover, whereas GDNF attenuated the sleep phenotype induced by MPP+, PBA augmented it. The current data suggest that MPP+ exposure generates a multifaceted phenotype in zebrafish and highlights that analyzing a narrow window of data can reveal effects that may be inconsistent with longer multi-parameter approaches. It further indicates that the model generally captures motor symptoms more faithfully than non-motor symptoms.
Collapse
Affiliation(s)
| | | | | | - Karl Ægir Karlsson
- 3Z Ehf, Reykjavik, Iceland.,Biomedical Center, University of Iceland, Reykjavik, Iceland.,Department of Engineering, School of Technology, Reykjavik University, Reykjavik, Iceland
| |
Collapse
|
31
|
Wasel O, Freeman JL. Chemical and Genetic Zebrafish Models to Define Mechanisms of and Treatments for Dopaminergic Neurodegeneration. Int J Mol Sci 2020; 21:ijms21175981. [PMID: 32825242 PMCID: PMC7503535 DOI: 10.3390/ijms21175981] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/14/2020] [Accepted: 08/16/2020] [Indexed: 01/08/2023] Open
Abstract
The zebrafish (Danio rerio) is routinely used in biological studies as a vertebrate model system that provides unique strengths allowing applications in studies of neurodevelopmental and neurodegenerative diseases. One specific advantage is that the neurotransmitter systems are highly conserved throughout vertebrate evolution, including between zebrafish and humans. Disruption of the dopaminergic signaling pathway is linked to multiple neurological disorders. One of the most common is Parkinson’s disease, a neurodegenerative disease associated with the loss of dopaminergic neurons, among other neuropathological characteristics. In this review, the development of the zebrafish’s dopaminergic system, focusing on genetic control of the dopaminergic system, is detailed. Second, neurotoxicant models used to study dopaminergic neuronal loss, including 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), the pesticides paraquat and rotenone, and 6-hydroxydopamine (6-OHDA), are described. Next, zebrafish genetic knockdown models of dj1, pink1, and prkn established for investigating mechanisms of Parkinson’s disease are discussed. Chemical modulators of the dopaminergic system are also highlighted to showcase the applicability of the zebrafish to identify mechanisms and treatments for neurodegenerative diseases such as Parkinson’s disease associated with the dopaminergic system.
Collapse
|
32
|
Barnhill LM, Murata H, Bronstein JM. Studying the Pathophysiology of Parkinson's Disease Using Zebrafish. Biomedicines 2020; 8:E197. [PMID: 32645821 PMCID: PMC7399795 DOI: 10.3390/biomedicines8070197] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease is a common neurodegenerative disorder leading to severe disability. The clinical features reflect progressive neuronal loss, especially involving the dopaminergic system. The causes of Parkinson's disease are slowly being uncovered and include both genetic and environmental insults. Zebrafish have been a valuable tool in modeling various aspects of human disease. Here, we review studies utilizing zebrafish to investigate both genetic and toxin causes of Parkinson's disease. They have provided important insights into disease mechanisms and will be of great value in the search for disease-modifying therapies.
Collapse
Affiliation(s)
| | | | - Jeff M. Bronstein
- David Geffen School of Medicine at UCLA, Department of Neurology and Molecular Toxicology Program, 710 Westwood Plaza, Los Angeles, CA 90095, USA; (L.M.B.); (H.M.)
| |
Collapse
|
33
|
Cellular Localization of gdnf in Adult Zebrafish Brain. Brain Sci 2020; 10:brainsci10050286. [PMID: 32403347 PMCID: PMC7288084 DOI: 10.3390/brainsci10050286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/04/2020] [Accepted: 05/08/2020] [Indexed: 12/15/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) was initially described as important for dopaminergic neuronal survival and is involved in many other essential functions in the central nervous system. Characterization of GDNF phenotype in mammals is well described; however, studies in non-mammalian vertebrate models are scarce. Here, we characterized the anatomical distribution of gdnf-expressing cells in adult zebrafish brain by means of combined in situ hybridization (ISH) and immunohistochemistry. Our results revealed that gdnf was widely dispersed in the brain. gdnf transcripts were co-localized with radial glial cells along the ventricular area of the telencephalon and in the hypothalamus. Interestingly, Sox2 positive cells expressed gdnf in the neuronal layer but not in the ventricular zone of the telencephalon. A subset of GABAergic precursor cells labeled with dlx6a-1.4kbdlx5a/6a: green fluorescence protein (GFP) in the pallium, parvocellular preoptic nucleus, and the anterior and dorsal zones of the periventricular hypothalamus also showed expression with gdnf mRNA. In addition, gdnf signals were detected in subsets of dopaminergic neurons, including those in the ventral diencephalon, similar to what is seen in mammalian brain. Our work extends our knowledge of gdnf action sites and suggests a potential role for gdnf in adult brain neurogenesis and regeneration.
Collapse
|