1
|
Sirwani N, Hedtke SM, Grant K, McColl G, Grant WN. Levels of Amyloid Beta ( Aβ) Expression in the Caenorhabditis elegans Neurons Influence the Onset and Severity of Neuronally Mediated Phenotypes. Cells 2024; 13:1598. [PMID: 39329779 PMCID: PMC11430350 DOI: 10.3390/cells13181598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/16/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024] Open
Abstract
A characteristic feature of Alzheimer's disease (AD) is the formation of neuronal extracellular senile plaques composed of aggregates of fibrillar amyloid β (Aβ) peptides, with the Aβ1-42 peptide being the most abundant species. These Aβ peptides have been proposed to contribute to the pathophysiology of the disease; however, there are few tools available to test this hypothesis directly. In particular, there are no data that establish a dose-response relationship between Aβ peptide expression level and disease. We have generated a panel of transgenic Caenorhabditis elegans strains expressing the human Aβ1-42 peptide under the control of promoter regions of two pan-neuronal expressed genes, snb-1 and rgef-1. Phenotypic data show strong age-related defects in motility, subtle changes in chemotaxis, reduced median and maximum lifespan, changes in health span indicators, and impaired learning. The Aβ1-42 expression level of these strains differed as a function of promoter identity and transgene copy number, and the timing and severity of phenotypes mediated by Aβ1-42 were strongly positively correlated with expression level. The pan-neuronal expression of varying levels of human Aβ1-42 in a nematode model provides a new tool to investigate the in vivo toxicity of neuronal Aβ expression and the molecular and cellular mechanisms underlying AD progression in the absence of endogenous Aβ peptides. More importantly, it allows direct quantitative testing of the dose-response relationship between neuronal Aβ peptide expression and disease for the first time. These strains may also be used to develop screens for novel therapeutics to treat Alzheimer's disease.
Collapse
Affiliation(s)
- Neha Sirwani
- Department of Environment and Genetics, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3086, Australia
| | - Shannon M Hedtke
- Department of Environment and Genetics, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3086, Australia
| | - Kirsten Grant
- Department of Environment and Genetics, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3086, Australia
| | - Gawain McColl
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Warwick N Grant
- Department of Environment and Genetics, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3086, Australia
| |
Collapse
|
2
|
Romussi S, Giunti S, Andersen N, De Rosa MJ. C. elegans: a prominent platform for modeling and drug screening in neurological disorders. Expert Opin Drug Discov 2024; 19:565-585. [PMID: 38509691 DOI: 10.1080/17460441.2024.2329103] [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: 11/13/2023] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Human neurodevelopmental and neurodegenerative diseases (NDevDs and NDegDs, respectively) encompass a broad spectrum of disorders affecting the nervous system with an increasing incidence. In this context, the nematode C. elegans, has emerged as a benchmark model for biological research, especially in the field of neuroscience. AREAS COVERED The authors highlight the numerous advantages of this tiny worm as a model for exploring nervous system pathologies and as a platform for drug discovery. There is a particular focus given to describing the existing models of C. elegans for the study of NDevDs and NDegDs. Specifically, the authors underscore their strong applicability in preclinical drug development. Furthermore, they place particular emphasis on detailing the common techniques employed to explore the nervous system in both healthy and diseased states. EXPERT OPINION Drug discovery constitutes a long and expensive process. The incorporation of invertebrate models, such as C. elegans, stands as an exemplary strategy for mitigating costs and expediting timelines. The utilization of C. elegans as a platform to replicate nervous system pathologies and conduct high-throughput automated assays in the initial phases of drug discovery is pivotal for rendering therapeutic options more attainable and cost-effective.
Collapse
Affiliation(s)
- Stefano Romussi
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
| | - Sebastián Giunti
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Natalia Andersen
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - María José De Rosa
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| |
Collapse
|
3
|
Caldero-Escudero E, Romero-Sanz S, De la Fuente S. Using C. elegans as a model for neurodegenerative diseases: Methodology and evaluation. Methods Cell Biol 2024; 188:1-34. [PMID: 38880519 DOI: 10.1016/bs.mcb.2024.03.004] [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] [Indexed: 06/18/2024]
Abstract
Caenorhabditis elegans is a nematode that has been used as an animal model for almost 50years. It has primitive and simple tissues and organs, making it an ideal model for studying neurological pathways involved in neurodegenerative diseases like Alzheimer's disease (AD) and Parkinson's disease (PD). C. elegans has conserved neurological pathways and is able to mimic human diseases, providing valuable insights into the human disease phenotype. This methodological review presents current approaches to generate neurodegenerative-like models of AD and PD in C. elegans, and evaluates the experiments commonly used to validate the diseases. These experimental approaches include assessing survival, fertility, mobility, electropharyngeogram assays, confocal mitochondrial imaging, RNA extraction for qRT-PCR or RT-PCR, and rate of defecation. This review also summarizes the current knowledge acquired on AD and PD using the aforementioned experimental approaches. Additionally, gaps in knowledge and future directions for research are also discussed in the review.
Collapse
|
4
|
Zeng Y, Wu T, Liang F, Long S, Guo W, Huang Y, Pei Z. Expression of human Ras-related protein Rab39B variant T168K in Caenorhabditis elegans leads to motor dysfunction and dopaminergic neuron degeneration. Heliyon 2024; 10:e26902. [PMID: 38444482 PMCID: PMC10912484 DOI: 10.1016/j.heliyon.2024.e26902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
Human RAB39B gene is related to familial early-onset Parkinson disease. In early adulthood, men with the RAB39B c.503C > A (Thr168Lys, p. T168K) mutation develop typical tremor, bradykinesia, and alpha-synuclein accumulation. We investigated the pathological mechanism of RAB39B T168K in a Caenorhabditis elegans model. In early adult C. elegans, RAB39B T168K led to dopaminergic neuron degeneration that presented as disrupted dendrites and blunt neuronal cells. Abnormal dopamine secretion was inferred from a decline in motor function and a positive basal slowing phenotype. Dopamine-associated tests confirmed that synthesis and recycling of dopamine were normal. The RAB39B T168K mutation might impair dopamine vesicular transmission from the presynaptic membrane to the synaptic gap in dopaminergic neurons. The release-dependent feedback mechanism in neurotransmitters regulates the balance of receptor activities. Protein-protein interactions network analysis revealed that RAB39B may also function in lysosomal degradation and autophagy. Impaired disposal of misfolded α-synuclein eventually leads to protein aggregation. Thus, like other members of the Rab family, RAB39B may be involved in vesicular transport associated with dopamine secretion and α-synuclein clearance.
Collapse
Affiliation(s)
- Yixuan Zeng
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, China
| | - Tengteng Wu
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fengyin Liang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Simei Long
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenyuan Guo
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Huang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Pei
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Panska L, Nedvedova S, Vacek V, Krivska D, Konecny L, Knop F, Kutil Z, Skultetyova L, Leontovyc A, Ulrychova L, Sakanari J, Asahina M, Barinka C, Macurkova M, Dvorak J. Uncovering the essential roles of glutamate carboxypeptidase 2 orthologs in Caenorhabditis elegans. Biosci Rep 2024; 44:BSR20230502. [PMID: 38108122 PMCID: PMC10794815 DOI: 10.1042/bsr20230502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 12/19/2023] Open
Abstract
Human glutamate carboxypeptidase 2 (GCP2) from the M28B metalloprotease group is an important target for therapy in neurological disorders and an established tumor marker. However, its physiological functions remain unclear. To better understand general roles, we used the model organism Caenorhabditis elegans to genetically manipulate its three existing orthologous genes and evaluate the impact on worm physiology. The results of gene knockout studies showed that C. elegans GCP2 orthologs affect the pharyngeal physiology, reproduction, and structural integrity of the organism. Promoter-driven GFP expression revealed distinct localization for each of the three gene paralogs, with gcp-2.1 being most abundant in muscles, intestine, and pharyngeal interneurons, gcp-2.2 restricted to the phasmid neurons, and gcp-2.3 located in the excretory cell. The present study provides new insight into the unique phenotypic effects of GCP2 gene knockouts in C. elegans, and the specific tissue localizations. We believe that elucidation of particular roles in a non-mammalian organism can help to explain important questions linked to physiology of this protease group and in extension to human GCP2 involvement in pathophysiological processes.
Collapse
Affiliation(s)
- Lucie Panska
- Department of Zoology and Fisheries, Center of Infectious Animal Diseases, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamycka 129, Prague 165 00, Czech Republic
- Faculty of Environmental Sciences, Center of Infectious Animal Diseases, Czech University of Life Sciences in Prague, Kamycka 129, Prague 165 00, Czech Republic
| | - Stepanka Nedvedova
- Department of Zoology and Fisheries, Center of Infectious Animal Diseases, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamycka 129, Prague 165 00, Czech Republic
- Department of Chemistry, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamycka 129, Prague 165 00, Czech Republic
| | - Vojtech Vacek
- Department of Zoology and Fisheries, Center of Infectious Animal Diseases, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamycka 129, Prague 165 00, Czech Republic
| | - Daniela Krivska
- Department of Zoology and Fisheries, Center of Infectious Animal Diseases, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamycka 129, Prague 165 00, Czech Republic
- Department of Chemistry, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamycka 129, Prague 165 00, Czech Republic
| | - Lukas Konecny
- Department of Zoology and Fisheries, Center of Infectious Animal Diseases, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamycka 129, Prague 165 00, Czech Republic
- Department of Parasitology, Faculty of Science, Charles University, Vinicna 7, Prague 2 128 00, Czech Republic
| | - Filip Knop
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague 2 128 00, Czech Republic
| | - Zsofia Kutil
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, Vestec 252 50, Czech Republic
| | - Lubica Skultetyova
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, Vestec 252 50, Czech Republic
| | - Adrian Leontovyc
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Flemingovo n. 2, Prague 160 00, Czech Republic
| | - Lenka Ulrychova
- Department of Parasitology, Faculty of Science, Charles University, Vinicna 7, Prague 2 128 00, Czech Republic
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Flemingovo n. 2, Prague 160 00, Czech Republic
| | - Judy Sakanari
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 1700 4th Street, CA 94143, USA
| | - Masako Asahina
- Department of Physiology, University of California, San Francisco, 600 16th Street, CA 94143, U.S.A
| | - Cyril Barinka
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, Vestec 252 50, Czech Republic
| | - Marie Macurkova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague 2 128 00, Czech Republic
| | - Jan Dvorak
- Department of Zoology and Fisheries, Center of Infectious Animal Diseases, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamycka 129, Prague 165 00, Czech Republic
- Faculty of Environmental Sciences, Center of Infectious Animal Diseases, Czech University of Life Sciences in Prague, Kamycka 129, Prague 165 00, Czech Republic
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Flemingovo n. 2, Prague 160 00, Czech Republic
| |
Collapse
|
6
|
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
|
7
|
Rani N, Alam MM, Jamal A, Bin Ghaffar U, Parvez S. Caenorhabditis elegans: A transgenic model for studying age-associated neurodegenerative diseases. Ageing Res Rev 2023; 91:102036. [PMID: 37598759 DOI: 10.1016/j.arr.2023.102036] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
Neurodegenerative diseases (NDs) are a heterogeneous group of aging-associated ailments characterized by interrupting cellular proteostasic machinery and the misfolding of distinct proteins to form toxic aggregates in neurons. Neurodegenerative diseases, which include Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and others, are becoming an increasing threat to human health worldwide. The degeneration and death of certain specific groups of neurons are the hallmarks of these diseases. Over the past decades, Caenorhabditis eleganshas beenwidely used as a transgenic model to investigate biological processes related to health and disease. The nematode Caenorhabditis elegans (C. elegans) has developed as a powerful tool for studying disease mechanisms due to its ease of genetic handling and instant cultivation while providing a whole-animal system amendable to several molecular and biochemical techniques. In this review, we elucidate the potential of C. elegans as a versatile platform for systematic dissection of the molecular basis of human disease, focusing on neurodegenerative disorders, and may help better our understanding of the disease mechanisms and search for new therapeutics for these devastating diseases.
Collapse
Affiliation(s)
- Nisha Rani
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Mumtaz Alam
- Drug Design and Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Azfar Jamal
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Usama Bin Ghaffar
- Department of Basic Science, College of Medicine, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Suhel Parvez
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
8
|
Huang X, Wang C, Zhang T, Li R, Chen L, Leung KL, Lakso M, Zhou Q, Zhang H, Wong G. PIWI-interacting RNA expression regulates pathogenesis in a Caenorhabditis elegans model of Lewy body disease. Nat Commun 2023; 14:6137. [PMID: 37783675 PMCID: PMC10545829 DOI: 10.1038/s41467-023-41881-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 09/21/2023] [Indexed: 10/04/2023] Open
Abstract
PIWI-interacting RNAs (piRNAs) are small noncoding RNAs that regulate gene expression, yet their molecular functions in neurobiology are unclear. While investigating neurodegeneration mechanisms using human α-syn(A53T)Tg and AβTg;α-syn(A53T)Tg pan-neuronal overexpressing strains, we unexpectedly observed dysregulation of piRNAs. RNAi screening revealed that knock down of piRNA biogenesis genes improved thrashing behavior; further, a tofu-1 gene deletion ameliorated phenotypic deficits in α-syn(A53T)Tg and AβTg;α-syn(A53T)Tg transgenic strains. piRNA expression was extensively downregulated and H3K9me3 marks were decreased after tofu-1 deletion in α-syn(A53T)Tg and AβTg;α-syn(A53T)Tg strains. Dysregulated piRNAs targeted protein degradation genes suggesting that a decrease of piRNA expression leads to an increase of degradation ability in C. elegans. Finally, we interrogated piRNA expression in brain samples from PD patients. piRNAs were observed to be widely overexpressed at late motor stage. In this work, our results provide evidence that piRNAs are mediators in pathogenesis of Lewy body diseases and suggest a molecular mechanism for neurodegeneration in these and related disorders.
Collapse
Affiliation(s)
- Xiaobing Huang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, China
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, 999078, Macau, China
| | - Changliang Wang
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, 999078, Macau, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, 510799, China
| | - Tianjiao Zhang
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, 999078, Macau, China
| | - Rongzhen Li
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, 999078, Macau, China
| | - Liang Chen
- Department of Computer Science, College of Engineering, Shantou University, Shantou, 515063, China
| | - Ka Lai Leung
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, 999078, Macau, China
| | - Merja Lakso
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, 999078, Macau, China
| | - Qinghua Zhou
- Department of Anesthesiology, The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, China
| | - Hongjie Zhang
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, 999078, Macau, China
| | - Garry Wong
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, 999078, Macau, China.
| |
Collapse
|
9
|
Stover MA, Tinoco-Bravo B, Shults CA, Marouk S, Deole R, Manjarrez JR. Probiotic effects of Lactococcus lactis and Leuconostoc mesenteroides on stress and longevity in Caenorhabditis elegans. Front Physiol 2023; 14:1207705. [PMID: 37772058 PMCID: PMC10522913 DOI: 10.3389/fphys.2023.1207705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/23/2023] [Indexed: 09/30/2023] Open
Abstract
The short lifespan of Caenorhabditis elegans enables the efficient investigation of probiotic interventions affecting stress and longevity involving the potential therapeutic value of Lactococcus lactis and Leuconostoc mesenteroides isolated from organic basil. The lactic acid bacteria were cultured from the produce collected from a local grocery store in Tulsa, Oklahoma, and then identified through 16S rDNA sequencing and biochemical tests. To dive deep into this analysis for potential probiotic therapy, we used fluorescent reporters that allow us to assess the differential induction of multiple stress pathways such as oxidative stress and the cytoplasmic, endoplasmic reticulum, and the mitochondrial unfolded protein response. This is combined with the classic health span measurements of survival, development, and fecundity, allowing a wide range of organismal observations of the different communities of microbes supported by probiotic supplementation with Lactococcus lactis and Leuconostoc mesenteroides. These strains were initially assessed in relation to the Escherichia coli feeding strain OP50 and the C. elegans microbiome. The supplementation showed a reduction in the median lifespan of the worms colonized within the microbiome. This was unsurprising, as negative results are common when probiotics are introduced into healthy microbiomes. To further assess the supplementation potential of these strains on an unhealthy (undifferentiated) microbiome, the typical axenic C. elegans diet, OP50, was used to simulate this single-species biome. The addition of lactic acid bacteria to OP50 led to a significant improvement in the median and overall survival in simulated biomes, indicating their potential in probiotic therapy. The study analyzed the supplemented cultures in terms of C. elegans' morphology, locomotor behavior, reproduction, and stress responses, revealing unique characteristics and stress response patterns for each group. As the microbiome's influence on the health span gains interest, the study aims to understand the microbiome relationships that result in differential stress resistance and lifespans by supplementing microbiomes with Lactococcus lactis and Leuconostoc mesenteroides isolated from organic basil in C. elegans.
Collapse
Affiliation(s)
| | | | | | | | | | - Jacob R. Manjarrez
- Biochemistry and Microbiology Department, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| |
Collapse
|
10
|
Salgueiro WG, Soares MV, Martins CF, Paula FR, Rios-Anjos RM, Carrazoni T, Mori MA, Müller RU, Aschner M, Dal Belo CA, Ávila DS. Dopaminergic modulation by quercetin: In silico and in vivo evidence using Caenorhabditis elegans as a model. Chem Biol Interact 2023; 382:110610. [PMID: 37348670 PMCID: PMC10527449 DOI: 10.1016/j.cbi.2023.110610] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
Quercetin is a flavonol widely distributed in plants and has various described biological functions. Several studies have reported on its ability to restore neuronal function in a wide variety of disease models, including animal models of neurodegenerative disorders such as Parkinson's disease. Quercetin per se can act as a neuroprotector/neuromodulator, especially in diseases related to impaired dopaminergic neurotransmission. However, little is known about how quercetin interacts with the dopaminergic machinery. Here we employed the nematode Caenorhabditis elegans to study this putative interaction. After observing behavioral modulation, mutant analysis and gene expression in C. elegans upon exposure to quercetin at a concentration that does not protect against MPTP, we constructed a homology-based dopamine transporter protein model to conduct a docking study. This led to suggestive evidence on how quercetin may act as a dopaminergic modulator by interacting with C. elegans' dopamine transporter and alter the nematode's exploratory behavior. Consistent with this model, quercetin controls C. elegans behavior in a way dependent on the presence of both the dopamine transporter (dat-1), which is up-regulated upon quercetin exposure, and the dopamine receptor 2 (dop-2), which appears to be mandatory for dat-1 up-regulation. Our data propose an interaction with the dopaminergic machinery that may help to establish the effects of quercetin as a neuromodulator.
Collapse
Affiliation(s)
- Willian Goulart Salgueiro
- Research Group in Biochemistry and Toxicology in Caenorhabditis elegans (GBToxCe), Federal University of Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil; Department of Biochemistry and Tissue Biology, University of Campinas, Monteiro Lobato Avenue, 255, Campinas, São Paulo, 13083-862, Brazil; Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany; Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Marcell Valandro Soares
- Research Group in Biochemistry and Toxicology in Caenorhabditis elegans (GBToxCe), Federal University of Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil; Department of Biochemistry and Molecular Biology, Post-graduate Program in Biological Sciences, Federal University of Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Cassiano Fiad Martins
- Research Group in Biochemistry and Toxicology in Caenorhabditis elegans (GBToxCe), Federal University of Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil
| | - Fávero Reisdorfer Paula
- Laboratory for Development and Quality Control in Medicines (LDCQ), Federal University of Pampa, Uruguaiana, RS, Brazil
| | | | - Thiago Carrazoni
- Neurobiology and Toxinology Laboratory, (LANETOX), Federal University of Pampa - UNIPAMPA, CEP 97300-000, São Gabriel, RS, Brazil
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, University of Campinas, Monteiro Lobato Avenue, 255, Campinas, São Paulo, 13083-862, Brazil; Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, SP, Brazil; Experimental Medicine Research Cluster, University of Campinas, Campinas, SP, Brazil
| | - Roman-Ulrich Müller
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany; Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Cháriston André Dal Belo
- Department of Biochemistry and Molecular Biology, Post-graduate Program in Biological Sciences, Federal University of Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil; Neurobiology and Toxinology Laboratory, (LANETOX), Federal University of Pampa - UNIPAMPA, CEP 97300-000, São Gabriel, RS, Brazil; Multidisciplinar Department, Federal University of São Paulo (UNIFESP), Angelica Street, 100- CEP 06110295, Osasco, SP, Brazil
| | - Daiana Silva Ávila
- Research Group in Biochemistry and Toxicology in Caenorhabditis elegans (GBToxCe), Federal University of Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil; Department of Biochemistry and Molecular Biology, Post-graduate Program in Biological Sciences, Federal University of Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
11
|
Li H, Gao M, Chen Z, Zhou Z, Li W, Zhang X, Jiang X, Luo L, Li F, Wang G, Zhang Y, Huang X, Zhu J, Fan S, Wu X, Huang C. Hordenine improves Parkinsonian-like motor deficits in mice and nematodes by activating dopamine D2 receptor-mediated signaling. Phytother Res 2023; 37:3296-3308. [PMID: 36883794 DOI: 10.1002/ptr.7790] [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: 09/07/2022] [Revised: 02/12/2023] [Accepted: 02/17/2023] [Indexed: 03/09/2023]
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative disease characterized by selective loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the striatum, leading to dopamine (DA) deficiency in the striatum and typical motor symptoms. A small molecule as a dietary supplement for PD would be ideal for practical reasons. Hordenine (HOR) is a phenolic phytochemical marketed as a dietary supplement found in cereals and germinated barley, as well as in beer, a widely consumed beverage. This study was aimed to identify HOR as a dopamine D2 receptor (DRD2) agonist in living cells, and investigate the alleviative effect and mechanism of HOR on PD-like motor deficits in mice and nematodes. Our results firstly showed that HOR is an agonist of DRD2, but not DRD1, in living cells. Moreover, HOR could improve the locomotor dysfunction, gait, and postural imbalance in MPTP- or 6-OHDA-induced mice or Caenorhabditis elegans, and prevent α-synuclein accumulation via the DRD2 pathway in C. elegans. Our results suggested that HOR could activate DRD2 to attenuate the PD-like motor deficits, and provide scientific evidence for the safety and reliability of HOR as a dietary supplement.
Collapse
Affiliation(s)
- Hongli Li
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min Gao
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziyu Chen
- Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhenyu Zhou
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Physiology, and Institute for Brain Sciences, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xiaoyang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Physiology, and Institute for Brain Sciences, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xi Jiang
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lingling Luo
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fei Li
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Gaorui Wang
- Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xingxu Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jingning Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Physiology, and Institute for Brain Sciences, School of Life Sciences, Nanjing University, Nanjing, China
| | - Shengjie Fan
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng Huang
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Zhang J, Liu S, Yuan H, Yong R, Duan S, Li Y, Spencer J, Lim EG, Yu L, Song P. Deep Learning for Microfluidic-Assisted Caenorhabditis elegans Multi-Parameter Identification Using YOLOv7. MICROMACHINES 2023; 14:1339. [PMID: 37512650 PMCID: PMC10386376 DOI: 10.3390/mi14071339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/14/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023]
Abstract
The Caenorhabditis elegans (C. elegans) is an ideal model organism for studying human diseases and genetics due to its transparency and suitability for optical imaging. However, manually sorting a large population of C. elegans for experiments is tedious and inefficient. The microfluidic-assisted C. elegans sorting chip is considered a promising platform to address this issue due to its automation and ease of operation. Nevertheless, automated C. elegans sorting with multiple parameters requires efficient identification technology due to the different research demands for worm phenotypes. To improve the efficiency and accuracy of multi-parameter sorting, we developed a deep learning model using You Only Look Once (YOLO)v7 to detect and recognize C. elegans automatically. We used a dataset of 3931 annotated worms in microfluidic chips from various studies. Our model showed higher precision in automated C. elegans identification than YOLOv5 and Faster R-CNN, achieving a mean average precision (mAP) at a 0.5 intersection over a union (mAP@0.5) threshold of 99.56%. Additionally, our model demonstrated good generalization ability, achieving an mAP@0.5 of 94.21% on an external validation set. Our model can efficiently and accurately identify and calculate multiple phenotypes of worms, including size, movement speed, and fluorescence. The multi-parameter identification model can improve sorting efficiency and potentially promote the development of automated and integrated microfluidic platforms.
Collapse
Affiliation(s)
- Jie Zhang
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool L693BX, UK
| | - Shuhe Liu
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Hang Yuan
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Ruiqi Yong
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Sixuan Duan
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool L693BX, UK
| | - Yifan Li
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool L693BX, UK
| | - Joseph Spencer
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool L693BX, UK
| | - Eng Gee Lim
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool L693BX, UK
| | - Limin Yu
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool L693BX, UK
| | - Pengfei Song
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool L693BX, UK
| |
Collapse
|
13
|
Okoro NO, Odiba AS, Yu Q, He B, Liao G, Jin C, Fang W, Wang B. Polysaccharides Extracted from Dendrobium officinale Grown in Different Environments Elicit Varying Health Benefits in Caenorhabditis elegans. Nutrients 2023; 15:2641. [PMID: 37375545 DOI: 10.3390/nu15122641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Dendrobium officinale is one of the most widely used medicinal herbs, especially in Asia. In recent times, the polysaccharide content of D. officinale has garnered attention due to the numerous reports of its medicinal properties, such as anticancer, antioxidant, anti-diabetic, hepatoprotective, neuroprotective, and anti-aging activities. However, few reports of its anti-aging potential are available. Due to high demand, the wild D. officinale is scarce; hence, alternative cultivation methods are being employed. In this study, we used the Caenorhabditis elegans model to investigate the anti-aging potential of polysaccharides extracted from D. officinale (DOP) grown in three different environments; tree (TR), greenhouse (GH), and rock (RK). Our findings showed that at 1000 µg/mL, GH-DOP optimally extended the mean lifespan by 14% and the maximum lifespan by 25% (p < 0.0001). TR-DOP and RK-DOP did not extend their lifespan at any of the concentrations tested. We further showed that 2000 µg/mL TR-DOP, GH-DOP, or RK-DOP all enhanced resistance to H2O2-induced stress (p > 0.05, p < 0.01, and p < 0.01, respectively). In contrast, only RK-DOP exhibited resistance (p < 0.01) to thermal stress. Overall, DOP from the three sources all increased HSP-4::GFP levels, indicating a boost in the ability of the worms to respond to ER-related stress. Similarly, DOP from all three sources decreased α-synuclein aggregation; however, only GH-DOP delayed β-amyloid-induced paralysis (p < 0.0001). Our findings provide useful information on the health benefits of DOP and also provide clues on the best practices for cultivating D. officinale for maximum medicinal applications.
Collapse
Affiliation(s)
- Nkwachukwu Oziamara Okoro
- Institute of Biological Sciences and Technology, Guangxi Academy of Sciences, Nanning 530007, China
- Department of Pharmaceutical and Medicinal Chemistry, University of Nigeria, Nsukka 410001, Nigeria
| | - Arome Solomon Odiba
- Institute of Biological Sciences and Technology, Guangxi Academy of Sciences, Nanning 530007, China
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qi Yu
- Institute of Biological Sciences and Technology, Guangxi Academy of Sciences, Nanning 530007, China
| | - Bin He
- School of Agriculture and Engineering, Guangxi Vocational and Technical College, Nanning 530226, China
| | - Guiyan Liao
- Institute of Biological Sciences and Technology, Guangxi Academy of Sciences, Nanning 530007, China
| | - Cheng Jin
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wenxia Fang
- Institute of Biological Sciences and Technology, Guangxi Academy of Sciences, Nanning 530007, China
| | - Bin Wang
- Institute of Biological Sciences and Technology, Guangxi Academy of Sciences, Nanning 530007, China
| |
Collapse
|
14
|
Starr LA, McKay LE, Peter KN, Seyfarth LM, Berkowitz LA, Caldwell KA, Caldwell GA. Attenuation of Dopaminergic Neurodegeneration in a C. elegans Parkinson's Model through Regulation of Xanthine Dehydrogenase (XDH-1) Expression by the RNA Editase, ADR-2. J Dev Biol 2023; 11:jdb11020020. [PMID: 37218814 DOI: 10.3390/jdb11020020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 05/24/2023] Open
Abstract
Differential RNA editing by adenosine deaminases that act on RNA (ADARs) has been implicated in several neurological disorders, including Parkinson's disease (PD). Here, we report results of a RNAi screen of genes differentially regulated in adr-2 mutants, normally encoding the only catalytically active ADAR in Caenorhabditis elegans, ADR-2. Subsequent analysis of candidate genes that alter the misfolding of human α-synuclein (α-syn) and dopaminergic neurodegeneration, two PD pathologies, reveal that reduced expression of xdh-1, the ortholog of human xanthine dehydrogenase (XDH), is protective against α-synuclein-induced dopaminergic neurodegeneration. Further, RNAi experiments show that WHT-2, the worm ortholog of the human ABCG2 transporter and a predicted interactor of XDH-1, is the rate-limiting factor in the ADR-2, XDH-1, WHT-2 system for dopaminergic neuroprotection. In silico structural modeling of WHT-2 indicates that the editing of one nucleotide in the wht-2 mRNA leads to the substitution of threonine with alanine at residue 124 in the WHT-2 protein, changing hydrogen bonds in this region. Thus, we propose a model where wht-2 is edited by ADR-2, which promotes optimal export of uric acid, a known substrate of WHT-2 and a product of XDH-1 activity. In the absence of editing, uric acid export is limited, provoking a reduction in xdh-1 transcription to limit uric acid production and maintain cellular homeostasis. As a result, elevation of uric acid is protective against dopaminergic neuronal cell death. In turn, increased levels of uric acid are associated with a decrease in ROS production. Further, downregulation of xdh-1 is protective against PD pathologies because decreased levels of XDH-1 correlate to a concomitant reduction in xanthine oxidase (XO), the form of the protein whose by-product is superoxide anion. These data indicate that modifying specific targets of RNA editing may represent a promising therapeutic strategy for PD.
Collapse
Affiliation(s)
- Lindsey A Starr
- Department of Biological Sciences, Center for Convergent Biomedicine, Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Luke E McKay
- Department of Biological Sciences, Center for Convergent Biomedicine, Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Kylie N Peter
- Department of Biological Sciences, Center for Convergent Biomedicine, Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Lena M Seyfarth
- Department of Biological Sciences, Center for Convergent Biomedicine, Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Laura A Berkowitz
- Department of Biological Sciences, Center for Convergent Biomedicine, Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Kim A Caldwell
- Department of Biological Sciences, Center for Convergent Biomedicine, Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35487, USA
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center of Excellence for the Basic Biology of Aging, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Guy A Caldwell
- Department of Biological Sciences, Center for Convergent Biomedicine, Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35487, USA
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center of Excellence for the Basic Biology of Aging, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
15
|
Use of invertebrates to model chemically induced parkinsonism-symptoms. Biochem Soc Trans 2023; 51:435-445. [PMID: 36645005 PMCID: PMC9987996 DOI: 10.1042/bst20221172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/17/2023]
Abstract
The prevalence of neurological diseases is currently growing due to the combination of several factor, including poor lifestyle and environmental imbalance which enhance the contribution of genetic factors. Parkinson's disease (PD), a chronic and progressive neurological condition, is one of the most prevalent neurodegenerative human diseases. Development of models may help to understand its pathophysiology. This review focuses on studies using invertebrate models to investigate certain chemicals that generate parkinsonian-like symptoms models. Additionally, we report some preliminary results of our own research on a crustacean (the crab Ucides cordatus) and a solitary ascidian (Styela plicata), used after induction of parkinsonism with 6-hydroxydopamine and the pesticide rotenone, respectively. We also discuss the advantages, limits, and drawbacks of using invertebrate models to study PD. We suggest prospects and directions for future investigations of PD, based on invertebrate models.
Collapse
|
16
|
Anti-Parkinson Effects of Holothuria leucospilota-Derived Palmitic Acid in Caenorhabditis elegans Model of Parkinson’s Disease. Mar Drugs 2023; 21:md21030141. [PMID: 36976190 PMCID: PMC10051922 DOI: 10.3390/md21030141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease which is still incurable. Sea cucumber-derived compounds have been reported to be promising candidate drugs for treating age-related neurological disorders. The present study evaluated the beneficial effects of the Holothuria leucospilota (H. leucospilota)-derived compound 3 isolated from ethyl acetate fraction (HLEA-P3) using Caenorhabditis elegans PD models. HLEA-P3 (1 to 50 µg/mL) restored the viability of dopaminergic neurons. Surprisingly, 5 and 25 µg/mL HLEA-P3 improved dopamine-dependent behaviors, reduced oxidative stress and prolonged lifespan of PD worms induced by neurotoxin 6-hydroxydopamine (6-OHDA). Additionally, HLEA-P3 (5 to 50 µg/mL) decreased α-synuclein aggregation. Particularly, 5 and 25 µg/mL HLEA-P3 improved locomotion, reduced lipid accumulation and extended lifespan of transgenic C. elegans strain NL5901. Gene expression analysis revealed that treatment with 5 and 25 µg/mL HLEA-P3 could upregulate the genes encoding antioxidant enzymes (gst-4, gst-10 and gcs-1) and autophagic mediators (bec-1 and atg-7) and downregulate the fatty acid desaturase gene (fat-5). These findings explained the molecular mechanism of HLEA-P3-mediated protection against PD-like pathologies. The chemical characterization elucidated that HLEA-P3 is palmitic acid. Taken together, these findings revealed the anti-Parkinson effects of H. leucospilota-derived palmitic acid in 6-OHDA induced- and α-synuclein-based models of PD which might be useful in nutritional therapy for treating PD.
Collapse
|
17
|
de los Ríos C, Viejo L, Carretero VJ, Juárez NH, Cruz-Martins N, Hernández-Guijo JM. Promising Molecular Targets in Pharmacological Therapy for Neuronal Damage in Brain Injury. Antioxidants (Basel) 2023; 12:118. [PMID: 36670980 PMCID: PMC9854812 DOI: 10.3390/antiox12010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023] Open
Abstract
The complex etiopathogenesis of brain injury associated with neurodegeneration has sparked a lot of studies in the last century. These clinical situations are incurable, and the currently available therapies merely act on symptoms or slow down the course of the diseases. Effective methods are being sought with an intent to modify the disease, directly acting on the properly studied targets, as well as to contribute to the development of effective therapeutic strategies, opening the possibility of refocusing on drug development for disease management. In this sense, this review discusses the available evidence for mitochondrial dysfunction induced by Ca2+ miscommunication in neurons, as well as how targeting phosphorylation events may be used to modulate protein phosphatase 2A (PP2A) activity in the treatment of neuronal damage. Ca2+ tends to be the catalyst for mitochondrial dysfunction, contributing to the synaptic deficiency seen in brain injury. Additionally, emerging data have shown that PP2A-activating drugs (PADs) suppress inflammatory responses by inhibiting different signaling pathways, indicating that PADs may be beneficial for the management of neuronal damage. In addition, a few bioactive compounds have also triggered the activation of PP2A-targeted drugs for this treatment, and clinical studies will help in the authentication of these compounds. If the safety profiles of PADs are proven to be satisfactory, there is a case to be made for starting clinical studies in the setting of neurological diseases as quickly as possible.
Collapse
Affiliation(s)
- Cristóbal de los Ríos
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
- Departamento de Ciencias Básicas de la Salud, University Rey Juan Carlos, Avda. Atenas s/n, 28922 Alcorcón, Spain
| | - Lucía Viejo
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Victoria Jiménez Carretero
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Natalia Hernández Juárez
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Natália Cruz-Martins
- Faculty of Medicine, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-319 Porto, Portugal
- Institute for Research and Advanced Training in Health Sciences and Technologies, Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal
| | - Jesús M. Hernández-Guijo
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
- Ramón y Cajal Institute for Health Research, IRYCIS, Hospital Ramón y Cajal, Ctra. de Colmenar Viejo, Km. 9,100, 28029 Madrid, Spain
| |
Collapse
|
18
|
Gonzales-Moreno C, Fernandez-Hubeid LE, Holgado A, Virgolini MB. Low-dose N-acetyl cysteine prevents paraquat-induced mortality in Caenorhabditis elegans. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000815. [PMID: 37065769 PMCID: PMC10101809 DOI: 10.17912/micropub.biology.000815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/27/2023] [Accepted: 03/21/2023] [Indexed: 04/18/2023]
Abstract
Exposure to the herbicide paraquat (PQ; 1,1'-dimethyl-4,4'-bipyridinium dichloride) affects the redox balance of the cell, an effect that can be restored by antioxidants, including N-acetyl cysteine (NAC). One hour of exposure to PQ (0 mM, 10 mM, 50 mM, or 100 mM) dose-dependently increased mortality in Caenorhabditis elegans after exposure (immediate toxicity), while this effect was more evident 24 hours thereafter (delayed toxicity). Importantly, pretreatment with NAC 0.5 mM for one hour partially prevented mortality in the immediate assay, while it had no effect in the delayed test, revealing the importance of long-term studies when evaluating toxicity.
Collapse
Affiliation(s)
- Candelaria Gonzales-Moreno
- Departamento de Farmacología Otto Orsingher. Facultad de Ciencias Químicas. Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Lucia E Fernandez-Hubeid
- IFEC-CONICET. Departamento de Farmacología Otto Orsingher. Facultad de Ciencias Químicas. Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Andrea Holgado
- Department of Biological Sciences, St. Edward's University, Austin, TX, USA
| | - Miriam B Virgolini
- IFEC-CONICET. Departamento de Farmacología Otto Orsingher. Facultad de Ciencias Químicas. Universidad Nacional de Córdoba, Córdoba, Argentina
- Correspondence to: Miriam B Virgolini (
)
| |
Collapse
|
19
|
Pham TD. Classification of Caenorhabditis Elegans Locomotion Behaviors With Eigenfeature-Enhanced Long Short-Term Memory Networks. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:206-216. [PMID: 35196241 DOI: 10.1109/tcbb.2022.3153668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The free-living nematode Caenorhabditis elegans is an ideal model for understanding behavior and networks of neurons. Experimental and quantitative analyses of neural circuits and behavior have led to system-level understanding of behavioral genetics and process of transformation from sensory integration in stimulus environments to behavioral outcomes. The ability to differentiate locomotion behavior between wild-type and mutant Caenorhabditis elegans strains allows precise inference on and gaining insights into genetic and environmental influences on behaviors. This paper presents an eigenfeature-enhanced deep-learning method for classifying the dynamics of locomotion behavior of wild-type and mutant Caenorhabditis elegans. Classification results obtained from public benchmark time-series data of eigenworms illustrate the superior performance of the new method over several existing classifiers. The proposed method has potential as a useful artificial-intelligence tool for automated identification of the nematode worm behavioral patterns aiming at elucidating molecular and genetic mechanisms that control the nervous system.
Collapse
|
20
|
Sanguanphun T, Sornkaew N, Malaiwong N, Chalorak P, Jattujan P, Niamnont N, Sobhon P, Meemon K. Neuroprotective effects of a medium chain fatty acid, decanoic acid, isolated from H. leucospilota against Parkinsonism in C. elegans PD model. Front Pharmacol 2022; 13:1004568. [PMID: 36582526 PMCID: PMC9792845 DOI: 10.3389/fphar.2022.1004568] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
Sea cucumbers are marine organism that have long been used for food and traditional medicine in Asian countries. Recently, we have shown that ethyl acetate fraction (HLEA) of the crude extract of the black sea cucumber, Holothuria leucospilota, could alleviate Parkinsonism in Caenorhabditis elegans PD models. In this study, we found that the effective neuroprotective activity is attributed to HLEA-P1 compound chemically isolated and identified in H. leucospilota ethyl acetate. We reported here that HLEA-P1 could attenuate DAergic neurodegeneration, improve DAergic-dependent behaviors, reduce oxidative stress in 6-OHDA-induced C. elegans. In addition, HLEA-P1 reduced α-synuclein aggregation, improved behavior deficit and recovered lipid deposition in transgenic C. elegans overexpressing α-synuclein. We also found that HLEA-P1 activates nuclear localization of DAF-16 transcription factor of insulin/IGF-1 signaling (IIS) pathway. Treatment with 25 μg/ml of HLEA-P1 upregulated transcriptional activity of DAF-16 target genes including anti-oxidant genes (such as sod-3) and small heat shock proteins (such as hsp16.1, hsp16.2, and hsp12.6) in 6-OHDA-induced worms. In α-synuclein-overexpressed C. elegans strain, treatment with 5 μg/ml of HLEA-P1 significantly activated mRNA expression of sod-3 and hsp16.2. Chemical analysis demonstrated that HLEA-P1 compound is decanoic acid/capric acid. Taken together, our findings revealed that decanoic acid isolated from H. leucospilota exerts anti-Parkinson effect in C. elegans PD models by partly modulating IIS/DAF-16 pathway.
Collapse
Affiliation(s)
- Tanatcha Sanguanphun
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Nilubon Sornkaew
- Department of Chemistry, Faculty of Science, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
| | - Nawaphat Malaiwong
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Pawanrat Chalorak
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand,Department of Radiological Technology and Medical Physics, Chulalongkorn University, Bangkok, Thailand
| | - Prapaporn Jattujan
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand,Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nakorn Niamnont
- Department of Chemistry, Faculty of Science, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
| | - Prasert Sobhon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Krai Meemon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand,Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, Thailand,*Correspondence: Krai Meemon,
| |
Collapse
|
21
|
Kitisin T, Muangkaew W, Sukphopetch P. Infections of Cryptococcus species induce degeneration of dopaminergic neurons and accumulation of α-Synuclein in Caenorhabditis elegans. Front Cell Infect Microbiol 2022; 12:1039336. [PMID: 36389163 PMCID: PMC9643722 DOI: 10.3389/fcimb.2022.1039336] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/11/2022] [Indexed: 11/27/2022] Open
Abstract
Cryptococcosis in the central nervous system (CNS) can present with motor declines described as Parkinsonism. Although several lines of evidence indicate that dopaminergic (DA) neuron degeneration and α-synuclein accumulation contribute to the hallmark of Parkinsonism and Parkinson’s disease (PD), little is known about cryptococcal infections associated with neuronal degeneration. In this study, the effects of Cryptococcus neoformans and C. gattii infections on dopaminergic neuron degeneration, α-synuclein accumulation, and lifespan in Caenorhabditis elegans were investigated. The results showed that cryptococcal infections significantly (P<0.05) induced DA neuron degeneration similar to a selective cathecholamine neurotoxin 6-hydroxydopamine (6-OHDA) in C. elegans (BZ555 strain) when compared to mock infected controls. Cryptococcal infections also significantly (P< 0.05) induced α-synuclein aggregation in C. elegans (NL5901 strain). Moreover, lifespan of the infected worms was significantly decreased (P<0.0001). In conclusion, DA neurodegeneration and α-synuclein accumulation are associated with lifespan reduction during cryptococcal infection in C elegans.
Collapse
|
22
|
Chauhan P, Wadhwa K, Singh G. Caenorhabditis elegans as a model system to evaluate neuroprotective potential of nano formulations. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2022.1018754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The impact of neurodegenerative illnesses on society is significant, but the mechanisms leading to neuronal malfunction and death in these conditions remain largely unknown despite identifying essential disease genes. To pinpoint the mechanisms behind the pathophysiology of neurodegenerative diseases, several researchers have turned to nematode C. elegans instead of using mammals. Since C. elegans is transparent, free-living, and amenable to culture, it has several benefits. As a result, all the neurons in C. elegans can be easily identified, and their connections are understood. Human proteins linked to Neurodegeneration can be made to express in them. It is also possible to analyze how C. elegans orthologs of the genes responsible for human neurodegenerative diseases function. In this article, we focused at some of the most important C. elegans neurodegeneration models that accurately represent many elements of human neurodegenerative illness. It has been observed that studies using the adaptable C. elegans have helped us in better understanding of human diseases. These studies have used it to replicate several aspects of human neurodegeneration. A nanotech approach involves engineering materials or equipments interacting with biological systems at the molecular level to trigger physiological responses by increasing stimulation, responding, and interacting with target sites while minimizing side effects, thus revolutionizing the treatment and diagnosis of neurodegenerative diseases. Nanotechnologies are being used to treat neurological disorders and deliver nanoscale drugs. This review explores the current and future uses of these nanotechnologies as innovative therapeutic modalities in treatment of neurodegenerative diseases using C elegans as an experimental model.
Collapse
|
23
|
Ma L, Li X, Liu C, Yan W, Ma J, Petersen RB, Peng A, Huang K. Modelling Parkinson's Disease in C. elegans: Strengths and Limitations. Curr Pharm Des 2022; 28:3033-3048. [PMID: 36111767 DOI: 10.2174/1381612828666220915103502] [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: 03/29/2022] [Accepted: 08/08/2022] [Indexed: 01/28/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease that affects the motor system and progressively worsens with age. Current treatment options for PD mainly target symptoms, due to our limited understanding of the etiology and pathophysiology of PD. A variety of preclinical models have been developed to study different aspects of the disease. The models have been used to elucidate the pathogenesis and for testing new treatments. These models include cell models, non-mammalian models, rodent models, and non-human primate models. Over the past few decades, Caenorhabditis elegans (C. elegans) has been widely adopted as a model system due to its small size, transparent body, short generation time and life cycle, fully sequenced genome, the tractability of genetic manipulation and suitability for large scale screening for disease modifiers. Here, we review studies using C. elegans as a model for PD and highlight the strengths and limitations of the C. elegans model. Various C. elegans PD models, including neurotoxin-induced models and genetic models, are described in detail. Moreover, met.
Collapse
Affiliation(s)
- Liang Ma
- Department of Pharmacy, Wuhan Mental Health Center, Wuhan, China.,Department of Pharmacy, Wuhan Hospital for Psychotherapy, Wuhan, China
| | - Xi Li
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengyu Liu
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanyao Yan
- Department of Pharmacy, Wuhan Fourth Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinlu Ma
- Human Resources Department, Wuhan Mental Health Center, Wuhan, China.,Human Resources Department, Wuhan Hospital for Psychotherapy, Wuhan, China
| | - Robert B Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mount Pleasant, MI, USA
| | - Anlin Peng
- Department of Pharmacy, The Third Hospital of Wuhan, Tongren Hospital of Wuhan University, Wuhan, China
| | - Kun Huang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
Albrecht PA, Fernandez-Hubeid LE, Deza-Ponzio R, Virgolini MB. The intertwining between lead and ethanol in the model organism Caenorhabditis elegans. FRONTIERS IN TOXICOLOGY 2022; 4:991787. [PMID: 36204698 PMCID: PMC9531147 DOI: 10.3389/ftox.2022.991787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Caenorhabditis elegans (C. elegans) is a model organism widely used to evaluate the mechanistic aspects of toxicants with the potential to predict responses comparable to those of mammals. We report here the consequences of developmental lead (Pb) exposure on behavioral responses to ethanol (EtOH) in C. elegans. In addition, we present data on morphological alterations in the dopamine (DA) synapse and DA-dependent behaviors aimed to dissect the neurobiological mechanisms that underlie the relationship between these neurotoxicants. Finally, the escalation to superior animals that parallels the observed effects in both experimental models with references to EtOH metabolism and oxidative stress is also discussed. Overall, the literature revised here underpins the usefulness of C. elegans to evidence behavioral responses to a combination of neurotoxicants in mechanistic-orientated studies.
Collapse
Affiliation(s)
- P A Albrecht
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Farmacología Experimental de Córdoba-Consejo Nacional de Investigaciones Científicas y Técnicas (IFEC-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - L E Fernandez-Hubeid
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Farmacología Experimental de Córdoba-Consejo Nacional de Investigaciones Científicas y Técnicas (IFEC-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - R Deza-Ponzio
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Farmacología Experimental de Córdoba-Consejo Nacional de Investigaciones Científicas y Técnicas (IFEC-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - M B Virgolini
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Farmacología Experimental de Córdoba-Consejo Nacional de Investigaciones Científicas y Técnicas (IFEC-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
25
|
Muhammad F, Liu Y, Wang N, Zhao L, Zhou Y, Yang H, Li H. Neuroprotective effects of cannabidiol on dopaminergic neurodegeneration and α-synuclein accumulation in C. elegans models of Parkinson's disease. Neurotoxicology 2022; 93:128-139. [PMID: 36108815 DOI: 10.1016/j.neuro.2022.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 08/23/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022]
Abstract
Parkinson disease (PD) is the second most progressive neurodegenerative disorder of the central nervous system (CNS) in the elderly, causing motor impediments and cognitive dysfunctions. Dopaminergic (DA) neuron degeneration and α-synuclein (α-Syn) accumulation in substantia nigra pars compacta (SNPc) are the major contributor to this disease. At present, the disease has no effective treatment. Many recent studies focus on identifying novel therapeutics that provide benefits to stop disease advancement in PD patients. Cannabidiol (CBD) is a cannabinoid derived from the Cannabis sativa plant and possesses anti-depressive, anti-inflammatory, and antioxidative effects. The present study aims to evaluate the neuroprotective effect of CBD in transgenic C. elegans PD models. We observed that CBD at 0.025 mM (24.66 %), 0.05 mM (52.41 %) and 0.1 mM (71.36 %) diminished DA neuron degenerations induced by 6-hydroxydopamine (6-OHDA), reduced (0.025, 27.1 %), (0.05, 38.9 %), (0.1, 51.3 %) food-sensing behavioural disabilities in BZ555, reduced 40.6 %, 56.3 %, 70.2 % the aggregative toxicity of α-Syn and expanded the nematodes' lifespan up to 11.5 %, 23.1 %, 28.8 %, dose-dependently. Moreover, CBD augmented the ubiquitin-like proteasomes 28.11 %, 43.27, 61.33 % and SOD-3 expressions by about 16.4 %, 21.2 %, 44.8 % in transgenic models. Further, we observed the antioxidative role of CBD by reducing 33.2 %, 41.4 %, 56.7 % reactive oxygen species in 6-OHDA intoxicated worms. Together, these findings supported CBD as an anti-parkinsonian drug and may exert its effects by raising lipid depositions to enhance proteasome activity and reduce oxidative stress via the antioxidative pathway.
Collapse
Affiliation(s)
- Fahim Muhammad
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China; School of Pharmacy, Lanzhou University, Donggang West Road No. 199, Lanzhou 730020, China; Clinical Center for Parkinson's Disease, Capital Medical University, Beijing 100053, China.
| | - Yan Liu
- School of Pharmacy, Lanzhou University, Donggang West Road No. 199, Lanzhou 730020, China.
| | - Ningbo Wang
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China.
| | - Longhe Zhao
- School of Pharmacy, Lanzhou University, Donggang West Road No. 199, Lanzhou 730020, China.
| | - Yangtao Zhou
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Clinical Center for Parkinson's Disease, Capital Medical University, Beijing 100053, China.
| | - Hui Yang
- Institute of Biology, Gansu Academy of Sciences, Lanzhou 730000, Gansu, China.
| | - Hongyu Li
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China; School of Pharmacy, Lanzhou University, Donggang West Road No. 199, Lanzhou 730020, China.
| |
Collapse
|
26
|
Wu X, Nagasawa S, Muto K, Ueda M, Suzuki C, Abe T, Higashitani A. Mitochonic Acid 5 Improves Duchenne Muscular Dystrophy and Parkinson's Disease Model of Caenorhabditis elegans. Int J Mol Sci 2022; 23:9572. [PMID: 36076995 PMCID: PMC9455831 DOI: 10.3390/ijms23179572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/09/2022] [Accepted: 08/23/2022] [Indexed: 11/24/2022] Open
Abstract
Mitochonic Acid 5 (MA-5) enhances mitochondrial ATP production, restores fibroblasts from mitochondrial disease patients and extends the lifespan of the disease model "Mitomouse". Additionally, MA-5 interacts with mitofilin and modulates the mitochondrial inner membrane organizing system (MINOS) in mammalian cultured cells. Here, we used the nematode Caenorhabditis elegans to investigate whether MA-5 improves the Duchenne muscular dystrophy (DMD) model. Firstly, we confirmed the efficient penetration of MA-5 in the mitochondria of C. elegans. MA-5 also alleviated symptoms such as movement decline, muscular tone, mitochondrial fragmentation and Ca2+ accumulation of the DMD model. To assess the effect of MA-5 on mitochondria perturbation, we employed a low concentration of rotenone with or without MA-5. MA-5 significantly suppressed rotenone-induced mitochondria reactive oxygen species (ROS) increase, mitochondrial network fragmentation and nuclear destruction in body wall muscles as well as endogenous ATP levels decline. In addition, MA-5 suppressed rotenone-induced degeneration of dopaminergic cephalic (CEP) neurons seen in the Parkinson's disease (PD) model. Furthermore, the application of MA-5 reduced mitochondrial swelling due to the immt-1 null mutation. These results indicate that MA-5 has broad mitochondrial homing and MINOS stabilizing activity in metazoans and may be a therapeutic agent for these by ameliorating mitochondrial dysfunction in DMD and PD.
Collapse
Affiliation(s)
- Xintong Wu
- Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Satoi Nagasawa
- Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Kasumi Muto
- Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Maiko Ueda
- Biomedical Research Core, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan
| | - Chitose Suzuki
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan
| | - Takaaki Abe
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan
| | | |
Collapse
|
27
|
Huang ML, Yen PL, Chang CH, Liao VHC. Chronic di(2-ethylhexyl) phthalate exposure leads to dopaminergic neuron degeneration through mitochondrial dysfunction in C. elegans. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 307:119574. [PMID: 35671892 DOI: 10.1016/j.envpol.2022.119574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/23/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
The plasticizer di(2-ethylhexyl) phthalate (DEHP) is frequently detected in the environment due to the abundance of its use. These levels might be hazardous to human health and ecosystems. Phthalates have been associated with neurological disorders, yet whether chronic DEHP exposure plays a role in Parkinson's disease (PD) or its underlying mechanisms is unknown. We investigated the effects of chronic DEHP exposure less than an environmentally-relevant dose on PD hallmarks, using Caenorhabditis elegans as a model. We show that developmental stage and exposure timing influence DEHP-induced dopaminergic neuron degeneration. In addition, in response to chronic DEHP exposure at 5 mg/L, mitochondrial fragmentation became significantly elevated, reactive oxygen species (ROS) levels increased, and ATP levels decreased, suggesting that mitochondrial dysfunction occurs. Furthermore, the data show that mitochondrial complex I (nuo-1 and gas-1) and complex II (mev-1) are involved in DEHP-induced dopaminergic neuron toxicity. These results suggest that chronic exposure to DEHP at levels less than an environmentally-relevant dose causes dopaminergic neuron degeneration through mitochondrial dysfunction involving mitochondrial complex I and II. Considering the high level of genetic conservation between C. elegans and mammals, chronic DEHP exposure might elevate the risk of developing PD in humans.
Collapse
Affiliation(s)
- Mei-Lun Huang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Pei-Ling Yen
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Chun-Han Chang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Vivian Hsiu-Chuan Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
28
|
Chakravorty A, Sharma A, Sheeba V, Manjithaya R. Glutamatergic Synapse Dysfunction in Drosophila Neuromuscular Junctions Can Be Rescued by Proteostasis Modulation. Front Mol Neurosci 2022; 15:842772. [PMID: 35909443 PMCID: PMC9337869 DOI: 10.3389/fnmol.2022.842772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
Glutamate is the major excitatory neurotransmitter in the nervous system, and the Drosophila glutamatergic neuromuscular junctions (NMJs) offer a tractable platform to understand excitatory synapse biology both in health and disease. Synaptopathies are neurodegenerative diseases that are associated with synaptic dysfunction and often display compromised proteostasis. One such rare, progressive neurodegenerative condition, Spinocerebellar Ataxia Type 3 (SCA3) or Machado-Joseph Disease (MJD), is characterized by cerebellar ataxia, Parkinsonism, and degeneration of motor neuron synapses. While the polyQ repeat mutant protein ataxin-3 is implicated in MJD, it is unclear how it leads to impaired synaptic function. In this study, we indicated that a Drosophila model of MJD recapitulates characteristics of neurodegenerative disorders marked by motor neuron dysfunction. Expression of 78 polyQ repeats of mutant ataxin-3 protein in Drosophila motor neurons resulted in behavioral defects, such as impaired locomotion in both larval and adult stages. Furthermore, defects in eclosion and lifespan were observed in adult flies. Detailed characterization of larval glutamatergic neuromuscular junctions (NMJs) revealed defects in morphological features along with compromised NMJ functioning. Autophagy, one of the key proteostasis pathways, is known to be impaired in the case of several synaptopathies. Our study reveals that overexpression of the autophagy-related protein Atg8a rescued behavioral defects. Thus, we present a model for glutamatergic synapse dysfunction that recapitulates synaptic and behavioral deficits and show that it is an amenable system for carrying out genetic and chemical biology screens to identify potential therapeutic targets for synaptopathies.
Collapse
Affiliation(s)
- Anushka Chakravorty
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Ankit Sharma
- Chronobiology and Behavioural Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Vasu Sheeba
- Chronobiology and Behavioural Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
- *Correspondence: Vasu Sheeba
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
- Ravi Manjithaya
| |
Collapse
|
29
|
Palikaras K, SenGupta T, Nilsen H, Tavernarakis N. Assessment of dopaminergic neuron degeneration in a C. elegans model of Parkinson’s disease. STAR Protoc 2022; 3:101264. [PMID: 35403008 PMCID: PMC8983426 DOI: 10.1016/j.xpro.2022.101264] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Transgenic Caenorhabditis elegans that expresses the full-length wild-type human α-synuclein in dopaminergic neurons provides a well-established Parkinson’s disease (PD) nematode model. Here, we present a detailed protocol to monitor and dissect the molecular underpinnings of age-associated neurodegeneration using this PD nematode model. This protocol includes preparation of nematode growth media and bacterial food sources, as well as procedures for nematode growth, synchronization, and treatment. We then describe procedures to assess dopaminergic neuronal death in vivo using fluorescence imaging. For complete details on the use and execution of this protocol, please refer to SenGupta et al. (2021). A Parkinson’s disease nematode model to study α-synuclein-mediated neurotoxicity Comprehensive approach for scoring cell death of dopaminergic neurons in C. elegans Genetic tools to investigate the tissue specific effects on neurodegeneration
Collapse
|
30
|
Invited review: Unearthing the mechanisms of age-related neurodegenerative disease using Caenorhabditis elegans. Comp Biochem Physiol A Mol Integr Physiol 2022; 267:111166. [PMID: 35176489 DOI: 10.1016/j.cbpa.2022.111166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/11/2022]
Abstract
As human life expectancy increases, neurodegenerative diseases present a growing public health threat, for which there are currently few effective treatments. There is an urgent need to understand the molecular and genetic underpinnings of these disorders so new therapeutic targets can be identified. Here we present the argument that the simple nematode worm Caenorhabditis elegans is a powerful tool to rapidly study neurodegenerative disorders due to their short lifespan and vast array of genetic tools, which can be combined with characterization of conserved neuronal processes and behavior orthologous to those disrupted in human disease. We review how pre-existing C. elegans models provide insight into human neurological disease as well as an overview of current tools available to study neurodegenerative diseases in the worm, with an emphasis on genetics and behavior. We also discuss open questions that C. elegans may be particularly well suited for in future studies and how worms will be a valuable preclinical model to better understand these devastating neurological disorders.
Collapse
|
31
|
Hughes S, van Dop M, Kolsters N, van de Klashorst D, Pogosova A, Rijs AM. Using a Caenorhabditis elegans Parkinson's Disease Model to Assess Disease Progression and Therapy Efficiency. Pharmaceuticals (Basel) 2022; 15:512. [PMID: 35631338 PMCID: PMC9143865 DOI: 10.3390/ph15050512] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 12/12/2022] Open
Abstract
Despite Parkinson's Disease (PD) being the second most common neurodegenerative disease, treatment options are limited. Consequently, there is an urgent need to identify and screen new therapeutic compounds that slow or reverse the pathology of PD. Unfortunately, few new therapeutics are being produced, partly due to the low throughput and/or poor predictability of the currently used model organisms and in vivo screening methods. Our objective was to develop a simple and affordable platform for drug screening utilizing the nematode Caenorhabditis elegans. The effect of Levodopa, the "Gold standard" of PD treatment, was explored in nematodes expressing the disease-causing α-synuclein protein. We focused on two key hallmarks of PD: plaque formation and mobility. Exposure to Levodopa ameliorated the mobility defect in C. elegans, similar to people living with PD who take the drug. Further, long-term Levodopa exposure was not detrimental to lifespan. This C. elegans-based method was used to screen a selection of small-molecule drugs for an impact on α-synuclein aggregation and mobility, identifying several promising compounds worthy of further investigation, most notably Ambroxol. The simple methodology means it can be adopted in many labs to pre-screen candidate compounds for a positive impact on disease progression.
Collapse
Affiliation(s)
- Samantha Hughes
- HAN BioCentre, HAN University of Applied Sciences, Laan van Scheut 2, 6525 EM Nijmegen, The Netherlands; (M.v.D.); (N.K.); (D.v.d.K.); (A.P.)
- A-LIFE Amsterdam Institute for Life and Environment, Section Environmental Health and Toxicology, Vrije Univeristeit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Maritza van Dop
- HAN BioCentre, HAN University of Applied Sciences, Laan van Scheut 2, 6525 EM Nijmegen, The Netherlands; (M.v.D.); (N.K.); (D.v.d.K.); (A.P.)
| | - Nikki Kolsters
- HAN BioCentre, HAN University of Applied Sciences, Laan van Scheut 2, 6525 EM Nijmegen, The Netherlands; (M.v.D.); (N.K.); (D.v.d.K.); (A.P.)
| | - David van de Klashorst
- HAN BioCentre, HAN University of Applied Sciences, Laan van Scheut 2, 6525 EM Nijmegen, The Netherlands; (M.v.D.); (N.K.); (D.v.d.K.); (A.P.)
| | - Anastasia Pogosova
- HAN BioCentre, HAN University of Applied Sciences, Laan van Scheut 2, 6525 EM Nijmegen, The Netherlands; (M.v.D.); (N.K.); (D.v.d.K.); (A.P.)
| | - Anouk M. Rijs
- Division of BioAnalytical Chemistry, AIMMS Amsterdam Institute of Molecular and Life Sciences, Vrije Univeristeit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
32
|
Para-Hydroxybenzyl Alcohol Delays the Progression of Neurodegenerative Diseases in Models of Caenorhabditis elegans through Activating Multiple Cellular Protective Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8986287. [PMID: 35401930 PMCID: PMC8989581 DOI: 10.1155/2022/8986287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/01/2022] [Accepted: 03/15/2022] [Indexed: 11/23/2022]
Abstract
The traditional Chinese medicine Gastrodia elata (commonly called “Tianma” in Chinese) has been widely used in the treatment of rheumatism, epilepsy, paralysis, headache, and dizziness. Phenolic compounds, such as gastrodin, para-hydroxybenzyl alcohol (HBA), p-hydroxybenzaldehyde, and vanillin are the main bioactive components isolated from Gastrodia elata. These compounds not only are structurally related but also share similar pharmacological activities, such as antioxidative and anti-inflammatory activities, and effects on the treatment of aging-related diseases. Here, we investigated the effect of para-hydroxybenzyl alcohol (HBA) on neurodegenerative diseases and aging in models of Caenorhabditis elegans (C. elegans). Our results showed that HBA effectively delayed the progression of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and Huntington's disease in models of C. elegans. In addition, HBA could increase the average lifespan of N2 worms by more than 25% and significantly improve the age-related physiological functions of worms. Moreover, HBA improved the survival rate of worms under stresses of oxidation, heat, and pathogenic bacteria. Further mechanistic investigation revealed that HBA could activate FOXO/DAF-16 and SKN-1 to regulate antioxidative and xenobiotic metabolism pathway. HBA could also activate HSF-1 to regulate proteostasis maintenance pathway, mitochondrial unfolded stress response, endoplasmic stress response and autophagy pathways. The above results suggest that HBA activated multiple cellular protective pathways to increase stress resistance and protect against aging and aging-related diseases. Overall, our study indicates that HBA is a potential candidate for future development of antiaging pharmaceutical application.
Collapse
|
33
|
Widaad A, Zulkipli IN, Petalcorin MIR. Anthelmintic Effect of Leucaena leucocephala Extract and Its Active Compound, Mimosine, on Vital Behavioral Activities in Caenorhabditis elegans. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27061875. [PMID: 35335240 PMCID: PMC8950933 DOI: 10.3390/molecules27061875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/22/2022] [Accepted: 03/07/2022] [Indexed: 12/03/2022]
Abstract
Helminth infections continue to be a neglected global threat in tropical regions, and there have been growing cases of anthelmintic resistance reported towards the existing anthelmintic drugs. Thus, the search for a novel anthelmintic agent has been increasing, especially those derived from plants. Leucaena leucocephala (LL) is a leguminous plant that is known to have several pharmacological activities, including anthelmintic activity. It is widely known to contain a toxic compound called mimosine, which we believed could be a potential lead candidate that could exert a potent anthelmintic effect. Hence, this study aimed to validate the presence of mimosine in LL extract and to investigate the anthelmintic effect of LL extract and mimosine on head thrashing, egg-laying, and pharyngeal pumping activities using the animal model Caenorhabditis elegans (C. elegans). Mimosine content in LL extract was confirmed through an HPLC analysis of spiking LL extract with different mimosine concentrations, whereby an increasing trend in peak heights was observed at a retention time of 0.9 min. LL extract and mimosine caused a significant dose-dependent increase in the percentage of worm mortality, which produced LC50s of 73 mg/mL and 6.39 mg/mL, respectively. Exposure of C. elegans to different concentrations of LL extract and mimosine significantly decreased the head thrashing, egg-laying, and mean pump amplitude of pharyngeal pumping activity. We speculated that these behavioral changes are due to the inhibitory effect of LL extract and mimosine on an L-type calcium channel called EGL-19. Our findings provide evidential support for the potential of LL extract and its active compound, mimosine, as novel anthelmintic candidates. However, the underlying mechanism of the anthelmintic action has yet to be elucidated.
Collapse
|
34
|
Jeong H, Park JY, Lee JH, Baik JH, Kim CY, Cho JY, Driscoll M, Paik YK. Deficiency in RCAT-1 Function Causes Dopamine Metabolism Related Behavioral Disorders in Caenorhabditis elegans. Int J Mol Sci 2022; 23:ijms23042393. [PMID: 35216508 PMCID: PMC8879058 DOI: 10.3390/ijms23042393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/13/2022] [Accepted: 02/18/2022] [Indexed: 02/04/2023] Open
Abstract
When animals are faced with food depletion, food search-associated locomotion is crucial for their survival. Although food search-associated locomotion is known to be regulated by dopamine, it has yet to investigate the potential molecular mechanisms governing the regulation of genes involved in dopamine metabolism (e.g., cat-1, cat-2) and related behavioral disorders. During the studies of the pheromone ascaroside, a signal of starvation stress in C. elegans, we identified R02D3.7, renamed rcat-1 (regulator of cat genes-1), which had previously been shown to bind to regulatory sequences of both cat-1 and cat-2 genes. It was found that RCAT-1 (R02D3.7) is expressed in dopaminergic neurons and functions as a novel negative transcriptional regulator for cat-1 and cat-2 genes. When a food source becomes depleted, the null mutant, rcat-1(ok1745), exhibited an increased frequency of high-angled turns and intensified area restricted search behavior compared to the wild-type animals. Moreover, rcat-1(ok1745) also showed defects in state-dependent olfactory adaptation and basal slowing response, suggesting that the mutants are deficient in either sensing food or locomotion toward food. However, rcat-1(ok1745) has normal cuticular structures and locomotion genes. The discovery of rcat-1 not only identifies a new subtype of dopamine-related behaviors but also provides a potential therapeutic target in Parkinson’s disease.
Collapse
Affiliation(s)
- Haelim Jeong
- Department of Biochemistry, College of Life Sciences and Biotechnology, Yonsei University, Seoul 03722, Korea; (H.J.); (J.-H.L.)
- Yonsei Proteome Research Center, Yonsei University, Seoul 03722, Korea; (J.Y.P.); (C.-Y.K.); (J.-Y.C.)
| | - Jun Young Park
- Yonsei Proteome Research Center, Yonsei University, Seoul 03722, Korea; (J.Y.P.); (C.-Y.K.); (J.-Y.C.)
| | - Ji-Hyun Lee
- Department of Biochemistry, College of Life Sciences and Biotechnology, Yonsei University, Seoul 03722, Korea; (H.J.); (J.-H.L.)
| | - Ja-Hyun Baik
- Department of Life Sciences, Korea University, Seoul 02841, Korea;
| | - Chae-Yeon Kim
- Yonsei Proteome Research Center, Yonsei University, Seoul 03722, Korea; (J.Y.P.); (C.-Y.K.); (J.-Y.C.)
- Interdisciplinary Program in Integrative Omics for Biomedical Science, Yonsei University, Seoul 03722, Korea
| | - Jin-Young Cho
- Yonsei Proteome Research Center, Yonsei University, Seoul 03722, Korea; (J.Y.P.); (C.-Y.K.); (J.-Y.C.)
- Interdisciplinary Program in Integrative Omics for Biomedical Science, Yonsei University, Seoul 03722, Korea
| | - Monica Driscoll
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08855, USA;
| | - Young-Ki Paik
- Department of Biochemistry, College of Life Sciences and Biotechnology, Yonsei University, Seoul 03722, Korea; (H.J.); (J.-H.L.)
- Yonsei Proteome Research Center, Yonsei University, Seoul 03722, Korea; (J.Y.P.); (C.-Y.K.); (J.-Y.C.)
- Interdisciplinary Program in Integrative Omics for Biomedical Science, Yonsei University, Seoul 03722, Korea
- Correspondence: ; Tel.: +82-2-2123-4242
| |
Collapse
|
35
|
Orientin Prolongs the Longevity of Caenorhabditis elegans and Postpones the Development of Neurodegenerative Diseases via Nutrition Sensing and Cellular Protective Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8878923. [PMID: 35237385 PMCID: PMC8885179 DOI: 10.1155/2022/8878923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/02/2022] [Indexed: 12/16/2022]
Abstract
Age is the major risk factor for most of the deadliest diseases. Developing small molecule drugs with antiaging effects could improve the health of aged people and retard the onset and progress of aging-associated disorders. Bioactive secondary metabolites from medicinal plants are the main source for development of medication. Orientin is a water-soluble flavonoid monomer compound widely found in many medicinal plants. Orientin inhibits fat production, antioxidation, and anti-inflammatory activities. In this study, we explored whether orientin could affect the aging of C. elegans. We found that orientin improved heat, oxidative, and pathogenic stress resistances through activating stress responses, including HSF-1-mediated heat shock response, SKN-1-mediated xenobiotic and oxidation response, mitochondria unfolded responses, endoplasmic unfolded protein response, and increased autophagy activity. Orientin also could activate key regulators of the nutrient sensing pathway, including AMPK and insulin downstream transcription factor FOXO/DAF-16 to further improve the cellular health status. The above effects of orientin reduced the accumulation of toxic proteins (α-synuclein, β-amyloid, and poly-Q) and delayed the onset of neurodegenerative disorders in AD, PD, and HD models of C. elegans and finally increased the longevity and health span of C. elegans. Our results suggest that orientin has promising antiaging effects and could be a potential natural source for developing novel therapeutic drugs for aging and its related diseases.
Collapse
|
36
|
Chang CH, Wei CC, Ho CT, Liao VHC. N-γ-(L-glutamyl)-L-selenomethionine shows neuroprotective effects against Parkinson's disease associated with SKN-1/Nrf2 and TRXR-1 in Caenorhabditis elegans. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153733. [PMID: 34537465 DOI: 10.1016/j.phymed.2021.153733] [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: 02/06/2021] [Revised: 07/08/2021] [Accepted: 09/01/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is a common neurodegenerative disease, yet fundamental treatments for the disease remain sparse. Thus, the search for potentially efficacious compounds from medicinal plants that can be used in the treatment of PD has gained significant interest. PURPOSE In many medicinal plants, selenium is primarily found in an organic form. We investigated the neuroprotective potential of an organic form of selenium, N-γ-(L-glutamyl)-L-selenomethionine (Glu-SeMet) in a Caenorhabditis elegans PD model and its possible molecular mechanisms. METHODS We used a C. elegans pharmacological PD strain (BZ555) that specifically expresses green fluorescent protein (GFP) in dopaminergic neurons and a transgenic PD strain (NL5901) that expresses human α-synuclein (α-syn) in muscle cells to investigate the neuroprotective potential of Glu-SeMet against PD. RESULTS We found that Glu-SeMet significantly ameliorated 6-hydroxydopamine (6-OHDA)-induced dopaminergic neuron damage in the transgenic BZ555 strain, with corresponding improvements in slowing behavior and intracellular ROS levels. In addition, compared with clinical PD drugs (L-DOPA and selegiline), Glu-SeMet demonstrated stronger ameliorated effects on 6-OHDA-induced toxicity. Glu-SeMet also triggered the nuclear translocation of SKN-1/Nrf2 and significantly increased SKN-1, GST-4, and GCS-1 mRNA levels in the BZ555 strain. However, Glu-SeMet did not increase mRNA levels or ameliorate the damage to dopaminergic neurons when the BZ555 strain was subjected to skn-1 RNA interference (RNAi). Glu-SeMet also upregulated the mRNA levels of the selenoprotein TRXR-1 in both the BZ555 and BZ555; skn-1 RNAi strains and significantly decreased α-syn accumulation in the NL5901 strain, although this was not observed in the NL5901; trxr-1 strain. CONCLUSION We found that Glu-SeMet has a neuroprotective effect against PD in a C. elegans PD model and that the anti-PD effects of Glu-SeMet were associated with SKN-1/Nrf2 and TRXR-1. Glu-SeMet may thus have the potential for use in therapeutic applications or supplements to slow the progression of PD.
Collapse
Affiliation(s)
- Chun-Han Chang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, No. 1 Roosevelt Road, Sec. 4, Taipei 106, Taiwan
| | - Chia-Cheng Wei
- Institute of Food Safety and Health, National Taiwan University, No. 17, Xuzhou Rd., Taipei 100, Taiwan; Department of Public Health, National Taiwan University, No. 17, Xuzhou Rd., Taipei, 100, Taiwan
| | - Chi-Tang Ho
- Department of Food Science, School of Environmental and Biological Sciences, Rutgers, the State University of New Jersey, 65 Dudley Rd., New Brunswick, NJ 08901-8520, United States
| | - Vivian Hsiu-Chuan Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, No. 1 Roosevelt Road, Sec. 4, Taipei 106, Taiwan.
| |
Collapse
|
37
|
Wei CC, Yang NC, Huang CW. Zearalenone Induces Dopaminergic Neurodegeneration via DRP-1-Involved Mitochondrial Fragmentation and Apoptosis in a Caenorhabditis elegans Parkinson's Disease Model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:12030-12038. [PMID: 34586801 DOI: 10.1021/acs.jafc.1c05836] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The contamination of mycotoxin zearalenone (ZEN) in foods has been reported worldwide, resulting in potential risks to food safety. However, the toxic mechanism of ZEN on neurodegenerative diseases has not been fully elucidated. Therefore, this study conducted in vivo ZEN neurotoxicity assessment on Parkinson's disease (PD)-related dopaminergic neurodegeneration and mitochondrial dysfunction using Caenorhabditis elegans. The results demonstrated that dopaminergic neuron damage was induced by ZEN exposure (1.25, 10, and 50 μM), and dopaminergic neuron-related behaviors were adversely affected subsequently. Additionally, the mitochondrial fragmentation was significantly increased by ZEN exposure. Moreover, upregulated expression of mitochondrial fission and cell apoptosis-related genes (drp-1, egl-1, ced-4, and ced-3) revealed the crucial role of DRP-1 on ZEN-induced neurotoxicity, which was further confirmed by drp-1 mutant and RNAi assays. In conclusion, our study indicates ZEN-induced dopaminergic neurodegeneration via DRP-1-involved mitochondrial fragmentation and apoptosis, which might cause harmful effects on PD-related symptoms.
Collapse
Affiliation(s)
- Chia-Cheng Wei
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, No. 17, Xuzhou Rd., Taipei 100, Taiwan
- Department of Public Health, College of Public Health, National Taiwan University, No. 17, Xuzhou Rd., Taipei 100, Taiwan
| | - Nien-Chieh Yang
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, No. 17, Xuzhou Rd., Taipei 100, Taiwan
| | - Chi-Wei Huang
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, No. 17, Xuzhou Rd., Taipei 100, Taiwan
| |
Collapse
|
38
|
Chandler R, Cogo S, Lewis P, Kevei E. Modelling the functional genomics of Parkinson's disease in Caenorhabditis elegans: LRRK2 and beyond. Biosci Rep 2021; 41:BSR20203672. [PMID: 34397087 PMCID: PMC8415217 DOI: 10.1042/bsr20203672] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/03/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
For decades, Parkinson's disease (PD) cases have been genetically categorised into familial, when caused by mutations in single genes with a clear inheritance pattern in affected families, or idiopathic, in the absence of an evident monogenic determinant. Recently, genome-wide association studies (GWAS) have revealed how common genetic variability can explain up to 36% of PD heritability and that PD manifestation is often determined by multiple variants at different genetic loci. Thus, one of the current challenges in PD research stands in modelling the complex genetic architecture of this condition and translating this into functional studies. Caenorhabditis elegans provide a profound advantage as a reductionist, economical model for PD research, with a short lifecycle, straightforward genome engineering and high conservation of PD relevant neural, cellular and molecular pathways. Functional models of PD genes utilising C. elegans show many phenotypes recapitulating pathologies observed in PD. When contrasted with mammalian in vivo and in vitro models, these are frequently validated, suggesting relevance of C. elegans in the development of novel PD functional models. This review will discuss how the nematode C. elegans PD models have contributed to the uncovering of molecular and cellular mechanisms of disease, with a focus on the genes most commonly found as causative in familial PD and risk factors in idiopathic PD. Specifically, we will examine the current knowledge on a central player in both familial and idiopathic PD, Leucine-rich repeat kinase 2 (LRRK2) and how it connects to multiple PD associated GWAS candidates and Mendelian disease-causing genes.
Collapse
Affiliation(s)
| | - Susanna Cogo
- School of Biological Sciences, University of Reading, Reading, RG6 6AH, U.K
- Department of Biology, University of Padova, Padova, Via Ugo Bassi 58/B, 35121, Italy
| | - Patrick A. Lewis
- Royal Veterinary College, University of London, London, NW1 0TU, U.K
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, U.K
| | - Eva Kevei
- School of Biological Sciences, University of Reading, Reading, RG6 6AH, U.K
| |
Collapse
|
39
|
Shadrina M, Slominsky P. Modeling Parkinson's Disease: Not Only Rodents? Front Aging Neurosci 2021; 13:695718. [PMID: 34421573 PMCID: PMC8377290 DOI: 10.3389/fnagi.2021.695718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/29/2021] [Indexed: 01/12/2023] Open
Abstract
Parkinson’s disease (PD) is a common chronic progressive multifactorial neurodegenerative disease. In most cases, PD develops as a sporadic idiopathic disease. However, in 10%–15% of all patients, Mendelian inheritance of the disease is observed in an autosomal dominant or autosomal recessive manner. To date, mutations in seven genes have been convincingly confirmed as causative in typical familial forms of PD, i.e., SNCA, LRRK2, VPS35, PRKN, PINK1, GBA, and DJ-1. Family and genome-wide association studies have also identified a number of candidate disease genes and a common genetic variability at 90 loci has been linked to risk for PD. The analysis of the biological function of both proven and candidate genes made it possible to conclude that mitochondrial dysfunction, lysosomal dysfunction, impaired exosomal transport, and immunological processes can play important roles in the development of the pathological process of PD. The mechanisms of initiation of the pathological process and its earliest stages remain unclear. The study of the early stages of the disease (before the first motor symptoms appear) is extremely complicated by the long preclinical period. In addition, at present, the possibility of performing complex biochemical and molecular biological studies familial forms of PD is limited. However, in this case, the analysis of the state of the central nervous system can only be assessed by indirect signs, such as the level of metabolites in the cerebrospinal fluid, peripheral blood, and other biological fluids. One of the potential solutions to this problem is the analysis of disease models, in which it is possible to conduct a detailed in-depth study of all aspects of the pathological process, starting from its earliest stages. Many modeling options are available currently. An analysis of studies published in the 2000s suggests that toxic models in rodents are used in the vast majority of cases. However, interesting and important data for understanding the pathogenesis of PD can be obtained from other in vivo models. Within the framework of this review, we will consider various models of PD that were created using various living organisms, from unicellular yeast (Saccharomyces cerevisiae) and invertebrate (Nematode and Drosophila) forms to various mammalian species.
Collapse
Affiliation(s)
- Maria Shadrina
- Laboratory of Molecular Genetics of Hereditary Diseases, Institute of Molecular Genetics of National Research Centre "Kurchatov Institute", Moscow, Russia
| | - Petr Slominsky
- Laboratory of Molecular Genetics of Hereditary Diseases, Institute of Molecular Genetics of National Research Centre "Kurchatov Institute", Moscow, Russia
| |
Collapse
|
40
|
Wang Z, Zheng P, Xie Y, Chen X, Solowij N, Green K, Chew YL, Huang XF. Cannabidiol regulates CB1-pSTAT3 signaling for neurite outgrowth, prolongs lifespan, and improves health span in Caenorhabditis elegans of Aβ pathology models. FASEB J 2021; 35:e21537. [PMID: 33817834 DOI: 10.1096/fj.202002724r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022]
Abstract
Cannabidiol (CBD), a phytocannabinoid from the Cannabis sativa plant, exhibits a broad spectrum of potential therapeutic properties for neurodegenerative diseases. An accumulation of amyloid-β (Aβ) protein is one of the most important neuropathology in neurodegenerative diseases like Alzheimer's disease (AD). Data on the effect of CBD on the amelioration of Aβ-induced neurite degeneration and its consequences of life and health spans is sparse. This study aimed to investigate the effects of CBD on neurite outgrowth in cells and lifespan and health span in Caenorhabditis elegans (C. elegans). In human SH-SY5Y neuronal cells, CBD prevented neurite lesion induced by Aβ1-42 and increased the expression of fatty acid amide hydrolase (FAAH) and cannabinoid receptor 1 (CB1R). Furthermore, CBD both protected the reduction of dendritic spine density and rescued the activity of synaptic Ca2+ /calmodulin-dependent protein kinase II (CaMKII) from Aβ1-42 toxicity in primary hippocampal neurons. In C. elegans, we used the transgenic CL2355 strain of C. elegans, which expresses the human Aβ peptide throughout the nervous system and found that CBD treatment extended lifespan and improved health span. The neuroprotective effect of CBD was further explored by observing the dopaminergic neurons using transgenic dat-1: GFP strains using the confocal microscope. This study shows that CBD prevents the neurite degeneration induced by Aβ, by a mechanism involving CB1R activation, and extends lifespan and improves health span in Aβ-overexpressing worms. Our findings support the potential therapeutic approach of CBD for the treatment of AD patients.
Collapse
Affiliation(s)
- Zhizhen Wang
- Australian Centre for Cannabinoid Clinical and Research Excellence (ACRE), New Lambton Heights, NSW, Australia.,Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Peng Zheng
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Yuanyi Xie
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Xi Chen
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Nadia Solowij
- Australian Centre for Cannabinoid Clinical and Research Excellence (ACRE), New Lambton Heights, NSW, Australia.,Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia.,School of Psychology, University of Wollongong, Wollongong, NSW, Australia
| | - Katrina Green
- Australian Centre for Cannabinoid Clinical and Research Excellence (ACRE), New Lambton Heights, NSW, Australia.,Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia.,Molecular Horizons, School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Yee Lian Chew
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia.,Molecular Horizons, School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Xu-Feng Huang
- Australian Centre for Cannabinoid Clinical and Research Excellence (ACRE), New Lambton Heights, NSW, Australia.,Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia.,Molecular Horizons, School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
41
|
Ijomone OM, Gubert P, Okoh COA, Varão AM, Amara LDO, Aluko OM, Aschner M. Application of Fluorescence Microscopy and Behavioral Assays to Demonstrating Neuronal Connectomes and Neurotransmitter Systems in C. elegans. NEUROMETHODS 2021; 172:399-426. [PMID: 34754139 PMCID: PMC8575032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The nematode Caenorhabditis elegans (C. elegans) is a prevailing model which is commonly utilized in a variety of biomedical research arenas, including neuroscience. Due to its transparency and simplicity, it is becoming a choice model organism for conducting imaging and behavioral assessment crucial to understanding the intricacies of the nervous system. Here, the methods required for neuronal characterization using fluorescent proteins and behavioral tasks are described. These are simplified protocols using fluorescent microscopy and behavioral assays to examine neuronal connections and associated neurotransmitter systems involved in normal physiology and aberrant pathology of the nervous system. Our aim is to make available to readers some streamlined and replicable procedures using C. elegans models as well as highlighting some of the limitations.
Collapse
Affiliation(s)
- Omamuyovwi M. Ijomone
- The Neuro- Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
- Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Priscila Gubert
- Department of Biochemistry, Laboratório de Imunopatologia Keizo Asami, LIKA, Federal University of Pernambuco, Recife, Brazil
- Postgraduate Program in Pure and Applied Chemistry, Federal University of Western of Bahia, Bahia, Brazil
| | - Comfort O. A. Okoh
- The Neuro- Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Alexandre M. Varão
- Postgraduate Program in Pure and Applied Chemistry, Federal University of Western of Bahia, Bahia, Brazil
| | - Leandro de O. Amara
- Postgraduate Program in Pure and Applied Chemistry, Federal University of Western of Bahia, Bahia, Brazil
| | - Oritoke M. Aluko
- The Neuro- Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
- Department of Physiology, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Michael Aschner
- Departments of Molecular Pharmacology and Neurosciences, Albert Einstein College of Medicine, NY, USA
| |
Collapse
|
42
|
Huang X, Wang C, Chen L, Zhang T, Leung KL, Wong G. Human amyloid beta and α-synuclein co-expression in neurons impair behavior and recapitulate features for Lewy body dementia in Caenorhabditis elegans. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166203. [PMID: 34146705 DOI: 10.1016/j.bbadis.2021.166203] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022]
Abstract
Amyloid β (Aβ), a product of APP, and SNCA (α-synuclein (α-syn)) are two of the key proteins found in lesions associated with the age-related neurodegenerative disorders Alzheimer's disease (AD) and Parkinson's disease (PD), respectively. Previous clinical studies uncovered Aβ and α-syn co-expression in the brains of patients, which lead to Lewy body dementia (LBD), a disease encompassing Dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD). To explore the pathogenesis and define the relationship between Aβ and α-syn for LBD, we established a C. elegans model which co-expresses human Aβ and α-syn with alanine 53 to threonine mutant (α-syn(A53T)) in pan-neurons. Compared to α-syn(A53T) single transgenic animals, pan-neuronal Aβ and α-syn(A53T) co-expression further enhanced the thrashing, egg laying, serotonin and cholinergic signaling deficits, and dopaminergic neuron damage in C. elegans. In addition, Aβ increased α-syn expression in transgenic animals. Transcriptome analysis of both Aβ;α-syn(A53T) strains and DLB patients showed common downregulation in lipid metabolism and lysosome function genes, suggesting that a decrease of lysosome function may reduce the clearance ability in DLB, and this may lead to the further pathogenic protein accumulation. These findings suggest that our model can recapitulate some features in LBD and provides a mechanism by which Aβ may exacerbate α-syn pathogenesis.
Collapse
Affiliation(s)
- Xiaobing Huang
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Changliang Wang
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Liang Chen
- Department of Computer Science, College of Engineering, Shantou University, Shantou 515063, China; Key Laboratory of Intelligent Manufacturing Technology of Ministry of Education, Shantou University, Shantou 515063, China
| | - Tianjiao Zhang
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Ka Lai Leung
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Garry Wong
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China.
| |
Collapse
|
43
|
Murphy D, Patel H, Wimalasena K. Caenorhabditis elegans Model Studies Show MPP + Is a Simple Member of a Large Group of Related Potent Dopaminergic Toxins. Chem Res Toxicol 2021; 34:1275-1285. [PMID: 33496570 PMCID: PMC8931847 DOI: 10.1021/acs.chemrestox.0c00422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Although the causes of Parkinson's disease (PD) are not fully understood, the consensus is that a combination of genetic and environmental factors plays a major role. The discovery that the synthetic chemical, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-derived N-methyl-4-phenylpyridinium (MPP+), recapitulates major pathophysiological characteristics of PD in humans and other mammals has provided the strongest support for this possibility; however, several key aspects of the mechanism remain unclear. In contrast to the widely accepted view that MPP+ is structurally unique and optimal for selective dopaminergic toxicity, previous in vitro studies have suggested that MPP+ is most likely a simple member of a large group of related dopaminergic toxins. Here we provide first in vivo evidence to support the above possibility using Caenorhabditis elegans PD models. We also provide in vivo evidence to show that the inherent predisposition of dopaminergic neurons to produce high oxidative stress and related downstream effects when exposed to MPP+ and related mitochondrial toxins is responsible for their selective vulnerability to these toxins. More significantly, present findings suggest that if this broad group of MPP+ related dopaminergic toxins are present in work places or in the environment, they could cause far-reaching public health consequences.
Collapse
Affiliation(s)
- David Murphy
- Department of Chemistry, Wichita State University, Wichita, KS 67260
| | - Harshil Patel
- Department of Chemistry, Wichita State University, Wichita, KS 67260
| | | |
Collapse
|
44
|
Wellenberg A, Weides L, Kurzke J, Hennecke T, Bornhorst J, Crone B, Karst U, Brinkmann V, Fritz G, Honnen S. Use of C. elegans as a 3R-compliant in vivo model for the chemoprevention of cisplatin-induced neurotoxicity. Exp Neurol 2021; 341:113705. [PMID: 33753139 DOI: 10.1016/j.expneurol.2021.113705] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023]
Abstract
Anticancer therapeutics can provoke severe side effects that impair the patient's quality of life. A frequent dose-limiting side effect of platinum-based anticancer therapy is neurotoxicity. Its pathophysiology is poorly understood, and effective preventive or therapeutic measures are missing. Therefore, elucidation of the molecular mechanism of platinating drug-induced neurotoxicity and the development of preventive strategies is urgently needed. To this end, we aim to use C. elegans as a 3R-compliant in vivo model. The 3R principles were conceived for animal welfare in science concerning animal experiments, which should be replaced, reduced or refined. We can analytically demonstrate dose-dependent uptake of cisplatin (CisPt) in C. elegans, as well as genotoxic and cytotoxic effects based on DNA adduct formation (i.e., 1,2-GpG intrastrand crosslinks), induction of apoptosis, and developmental toxicity. Measuring the impairment of pharyngeal pumping as a marker of neurotoxicity, we found that especially CisPt reduces the pumping frequency at concentrations where basal and touch-provoked movement were not yet affected. CisPt causes glutathione (GSH) depletion and RNAi-mediated knockdown of the glutamate-cysteine ligase GCS-1 aggravates the CisPt-induced inhibition of pharyngeal pumping. Moreover, N-acetylcysteine (NAC) mitigated CisPt-triggered toxicity, indicating that GSH depletion contributes to the CisPt-induced pharyngeal damage. In addition to NAC, amifostine (WR1065) also protected the pharynx of C. elegans from the toxic effects of CisPt. Measuring pharyngeal activity by the electrophysiological recording of neurotransmission in the pharynx, we confirmed that CisPt is neurotoxic in C. elegans and that NAC is neuroprotective in the nematode. The data support the hypothesis that monitoring the pharyngeal activity of C. elegans is a useful surrogate marker of CisPt-induced neurotoxicity. In addition, a low GSH pool reduces the resistance of neurons to CisPt treatment, and both NAC and WR1065 are capable of attenuating platinum-induced neurotoxicity during post-incubation in C. elegans. Overall, we propose C. elegans as a 3R-compliant in vivo model to study the molecular mechanisms of platinum-induced neurotoxicity and to explore novel neuroprotective therapeutic strategies to alleviate respective side effects of platinum-based cancer therapy.
Collapse
Affiliation(s)
- Anna Wellenberg
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Lea Weides
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Jennifer Kurzke
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| | - Till Hennecke
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| | - Julia Bornhorst
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, D-14558 Nuthetal, Germany; Faculty of Mathematics and Natural Sciences, Food Chemistry, University of Wuppertal, Gaußstr. 20, D-42119 Wuppertal, Germany.
| | - Barbara Crone
- Institute of Inorganic and Analytical Chemistry, University of Muenster, Corrensstraße 30, D-48149 Muenster, Germany.
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Muenster, Corrensstraße 30, D-48149 Muenster, Germany.
| | - Vanessa Brinkmann
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Sebastian Honnen
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| |
Collapse
|
45
|
Koch SC, Nelson A, Hartenstein V. Structural aspects of the aging invertebrate brain. Cell Tissue Res 2021; 383:931-947. [PMID: 33409654 PMCID: PMC7965346 DOI: 10.1007/s00441-020-03314-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/28/2020] [Indexed: 11/26/2022]
Abstract
Aging is characterized by a decline in neuronal function in all animal species investigated so far. Functional changes are accompanied by and may be in part caused by, structurally visible degenerative changes in neurons. In the mammalian brain, normal aging shows abnormalities in dendrites and axons, as well as ultrastructural changes in synapses, rather than global neuron loss. The analysis of the structural features of aging neurons, as well as their causal link to molecular mechanisms on the one hand, and the functional decline on the other hand is crucial in order to understand the aging process in the brain. Invertebrate model organisms like Drosophila and C. elegans offer the opportunity to apply a forward genetic approach to the analysis of aging. In the present review, we aim to summarize findings concerning abnormalities in morphology and ultrastructure in invertebrate brains during normal aging and compare them to what is known for the mammalian brain. It becomes clear that despite of their considerably shorter life span, invertebrates display several age-related changes very similar to the mammalian condition, including the retraction of dendritic and axonal branches at specific locations, changes in synaptic density and increased accumulation of presynaptic protein complexes. We anticipate that continued research efforts in invertebrate systems will significantly contribute to reveal (and possibly manipulate) the molecular/cellular pathways leading to neuronal aging in the mammalian brain.
Collapse
Affiliation(s)
- Sandra C Koch
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Annie Nelson
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Volker Hartenstein
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA.
| |
Collapse
|
46
|
Pandey T, Shukla A, Trivedi M, Khan F, Pandey R. Swertiamarin from Enicostemma littorale, counteracts PD associated neurotoxicity via enhancement α-synuclein suppressive genes and SKN-1/NRF-2 activation through MAPK pathway. Bioorg Chem 2021; 108:104655. [PMID: 33548732 DOI: 10.1016/j.bioorg.2021.104655] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 11/30/2022]
Abstract
The elusive targets and the multifactorial etiology of Parkinson's disease (PD) have hampered the discovery of a potent drug for PD. Furthermore, the presently available medications provide only symptomatic relief and have failed to mitigate the pathogenesis associated with PD. Therefore, the current study was aimed to evaluate the prospective of swertiamarin (SW), a secoiridoid glycoside isolated from a traditional medicinal plant, Enicostemma littorale Blume to ameliorate the characteristic features of PD in Caenorhabditis elegans. SW (25 μM) administration decreased the α-synuclein (α-syn) deposition, inhibited apoptosis and increased dopamine level mediated through upregulating the expression of genes linked to ceramide synthesis, mitochondrial morphology and function regulation, fatty acid desaturase genes along with stress responsive MAPK (mitogen-activated protein kinase) pathway genes. The neuroprotective effect of SW was evident from the robust reduction of 6-hydroxydopamine (6-OHDA) induced dopaminergic neurodegeneration independent of dopamine transporter (dat-1). SW mediated translational regulation of MAPK pathway genes was observed through increase expression of SKN-1 and GST-4. Further, in-silico molecular docking analysis of SW with C. elegans MEK-1 showed a promising binding affinity affirming the in-vivo results. Overall, these novel finding supports that SW is a possible lead for drug development against the multi- factorial PD pathologies.
Collapse
Affiliation(s)
- Taruna Pandey
- Aging Biology Lab, Microbial Technology and Nematology Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Aparna Shukla
- Department of Molecular and Structural Biology, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Mashu Trivedi
- Aging Biology Lab, Microbial Technology and Nematology Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Feroz Khan
- Department of Molecular and Structural Biology, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Rakesh Pandey
- Aging Biology Lab, Microbial Technology and Nematology Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India.
| |
Collapse
|
47
|
Tran J, Taylor SK, Gupta A, Amin N, Pant H, Gupta BP, Mishra RK. Therapeutic effects of TP5, a Cdk5/p25 inhibitor, in in vitro and in vivo models of Parkinson’s disease. CURRENT RESEARCH IN NEUROBIOLOGY 2021; 2:100006. [PMID: 36246507 PMCID: PMC9559888 DOI: 10.1016/j.crneur.2021.100006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 02/03/2021] [Accepted: 02/23/2021] [Indexed: 11/26/2022] Open
Abstract
Parkinson’s Disease (PD) is a chronic progressive neurodegenerative disease. Current treatments for PD are symptomatic and only increase striatal dopamine levels. Proactive neuroprotective approaches that slow the progression of PD and maintain appropriate dopamine neuron populations are needed to treat the disease. One suggested mechanism contributing to the pathology of PD involves the binding of cyclin-dependent kinase 5 (Cdk5) to p25, creating a hyperactivated complex to induce cell death. The objective of this study is to investigate the neuroprotective and neurorestorative properties of Truncated Peptide 5 (TP5), a derivative of the p35 activator involved in Cdk5 regulation, via the inhibition of Cdk5/p25 complex function. SH-SY5Y cell line and the nematode Caenorhabditis elegans were exposed to paraquat (PQ), an oxidative stressor, to induce Parkinsonian phenotypes. TP5 was administered prior to PQ exposure to determine its neuroprotective effects and, in further experiments, after PQ exposure to examine its neurorestorative effects. In the SH-SY5Y cell line, TP5 was found to have neuroprotective effects using a cell viability assay and demonstrated neuroprotective and neurorestorative effects in C. elegans by examining dopaminergic neurons and dopamine-dependent behaviour. TP5 decreased elevated Cdk5 activation in worms that were exposed to PQ. TP5’s inhibition of Cdk5/p25 hyperactivity led to the protection of dopamine neurons in these PD models. This suggests that TP5 can act as a potential therapeutic drug towards PD. Truncated Peptide 5 (TP5) is tested in SH-SY5Y culture cells and the worm C. elegans. TP5 protects and/or restores dopaminergic neurons in both Parkinson’s disease models. TP5 shows promising therapeutic effects in the worm system. Beneficial effects of TP5 are likely due to the reduced Cdk5/p25 hyperactivity.
Collapse
|
48
|
Li H, Feng Y, Chen Z, Jiang X, Zhou Z, Yuan J, Li F, Zhang Y, Huang X, Fan S, Wu X, Huang C. Pepper component 7-ethoxy-4-methylcoumarin, a novel dopamine D2 receptor agonist, ameliorates experimental Parkinson's disease in mice and Caenorhabditis elegans. Pharmacol Res 2021; 163:105220. [PMID: 33007422 DOI: 10.1016/j.phrs.2020.105220] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/22/2020] [Accepted: 09/22/2020] [Indexed: 02/09/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease resulting from the degeneration of dopaminergic (DAergic) neurons in the substantia nigra pars compacta (SNpc) and subsequent deficit of dopamine in the striatum. PD is inversely associated with consumption of peppers; however, the constituent and the underlying mechanism remain unclear. This study aimed to investigate the effects of 7-ethoxy-4-methylcoumarin (EMC), a pepper constituent, on PD-like disorders in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mice and 6-hydroxydopamine (6-OHDA)-exposed C. elegans. In this study, EMC was identified as an agonist of dopamine D2 receptor (DRD2) and increased the expression of P-CREB and BDNF in SH-SY5Y cells. In MPTP-treated PD mice, EMC was shown to apparently ameliorate the motor and gait disorders, and restore the depressed TH expression in SNpc and striatum. Meanwhile, it recovered the locomotor deficit caused by 6-OHDA in wild type N2 and CAT-2-transgenic UA57 of C. elegans, and relieved the degeneration of DAergic neurons resulting from 6-OHDA or with ageing. Moreover, EMC inhibited α-synuclein accumulation in C. elegans strain NL5901 overexpressing human α-synuclein gene. Taken together, EMC was identified as a novel DRD2 agonist and improved experimental PD in mice and C. elegans. These findings suggest that EMC may be beneficial to PD patients, further supporting that the consumption of peppers may have favorable effect on PD progression.
Collapse
Affiliation(s)
- Hongli Li
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yaru Feng
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziyu Chen
- Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xi Jiang
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhenyu Zhou
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinfeng Yuan
- Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fei Li
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Yu Zhang
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Xingxu Huang
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Shengjie Fan
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Cheng Huang
- Drug Discovery Laboratory, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
49
|
Prasad EM, Hung SY. Behavioral Tests in Neurotoxin-Induced Animal Models of Parkinson's Disease. Antioxidants (Basel) 2020; 9:E1007. [PMID: 33081318 PMCID: PMC7602991 DOI: 10.3390/antiox9101007] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023] Open
Abstract
Currently, neurodegenerative diseases are a major cause of disability around the world. Parkinson's disease (PD) is the second-leading cause of neurodegenerative disorder after Alzheimer's disease. In PD, continuous loss of dopaminergic neurons in the substantia nigra causes dopamine depletion in the striatum, promotes the primary motor symptoms of resting tremor, bradykinesia, muscle rigidity, and postural instability. The risk factors of PD comprise environmental toxins, drugs, pesticides, brain microtrauma, focal cerebrovascular injury, aging, and hereditary defects. The pathologic features of PD include impaired protein homeostasis, mitochondrial dysfunction, nitric oxide, and neuroinflammation, but the interaction of these factors contributing to PD is not fully understood. In neurotoxin-induced PD models, neurotoxins, for instance, 6-hydroxydopamine (6-OHDA), 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), 1-Methyl-4-phenylpyridinium (MPP+), paraquat, rotenone, and permethrin mainly impair the mitochondrial respiratory chain, activate microglia, and generate reactive oxygen species to induce autooxidation and dopaminergic neuronal apoptosis. Since no current treatment can cure PD, using a suitable PD animal model to evaluate PD motor symptoms' treatment efficacy and identify therapeutic targets and drugs are still needed. Hence, the present review focuses on the latest scientific developments in different neurotoxin-induced PD animal models with their mechanisms of pathogenesis and evaluation methods of PD motor symptoms.
Collapse
Affiliation(s)
- E. Maruthi Prasad
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, No.91, Hsueh-Shih Road, Taichung 40402, Taiwan;
| | - Shih-Ya Hung
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, No.91, Hsueh-Shih Road, Taichung 40402, Taiwan;
- Department of Medical Research, China Medical University Hospital, No. 2, Yude Road, Taichung 40447, Taiwan
| |
Collapse
|
50
|
Krum BN, Martins AC, Queirós L, Ferrer B, Milne GL, Soares FAA, Fachinetto R, Aschner M. Haloperidol Interactions with the dop-3 Receptor in Caenorhabditis elegans. Mol Neurobiol 2020; 58:304-316. [PMID: 32935232 DOI: 10.1007/s12035-020-02124-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/05/2020] [Indexed: 11/29/2022]
Abstract
Haloperidol is a typical antipsychotic drug commonly used to treat a broad range of psychiatric disorders related to dysregulations in the neurotransmitter dopamine (DA). DA modulates important physiologic functions and perturbations in Caenorhabditis elegans (C. elegans) and, its signaling have been associated with alterations in behavioral, molecular, and morphologic properties in C. elegans. Here, we evaluated the possible involvement of dopaminergic receptors in the onset of these alterations followed by haloperidol exposure. Haloperidol increased lifespan and decreased locomotor behavior (basal slowing response, BSR, and locomotion speed via forward speed) of the worms. Moreover, locomotion speed recovered to basal conditions upon haloperidol withdrawal. Haloperidol also decreased DA levels, but it did not alter neither dop-1, dop-2, and dop-3 gene expression, nor CEP dopaminergic neurons' morphology. These effects are likely due to haloperidol's antagonism of the D2-type DA receptor, dop-3. Furthermore, this antagonism appears to affect mechanistic pathways involved in the modulation and signaling of neurotransmitters such as octopamine, acetylcholine, and GABA, which may underlie at least in part haloperidol's effects. These pathways are conserved in vertebrates and have been implicated in a range of disorders. Our novel findings demonstrate that the dop-3 receptor plays an important role in the effects of haloperidol.
Collapse
Affiliation(s)
- Bárbara Nunes Krum
- Departamento de Fisiologia e Farmacologia, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Camobi, Santa Maria, RS, 97105-900, Brazil.,Department of Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University, Forccheimer 209, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Airton C Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University, Forccheimer 209, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Libânia Queirós
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University, Forccheimer 209, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.,Department of Biology and CESAM (Centre for Environmental and Marine Studies), University of Aveiro, 3810-193, Aveiro, Portugal
| | - Beatriz Ferrer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University, Forccheimer 209, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Ginger L Milne
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37240, USA
| | - Félix Alexandre Antunes Soares
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University, Forccheimer 209, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.,Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - Roselei Fachinetto
- Departamento de Fisiologia e Farmacologia, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University, Forccheimer 209, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| |
Collapse
|