1
|
Kumar J, Delgado SA, Sarma H, Narayan M. Caffeic acid recarbonization: A green chemistry, sustainable carbon nano material platform to intervene in neurodegeneration induced by emerging contaminants. ENVIRONMENTAL RESEARCH 2023; 237:116932. [PMID: 37598847 PMCID: PMC11285802 DOI: 10.1016/j.envres.2023.116932] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/12/2023] [Accepted: 08/18/2023] [Indexed: 08/22/2023]
Abstract
Environmental agents such as pesticides, weedicides and herbicides (collectively referred to as pesticides) are associated with the onset and pathogenesis of neurodegenerative disorders such as Parkinson's (PD) and Alzheimer's (AD) diseases. The development of blood-brain barrier (BBB)-penetrating therapeutic candidates to both prevent and treat the aforementioned xenotoxicant-induced neurodegenerative disorders remains an unmet need. Here, we examine whether caffeic-acid based Carbon Quantum Dots (CACQDs) can intervene in pesticide-associated onset and progress of the PD phenotype. Pulse-chase fluorescence analyses revealed that CACQDs intervene in the soluble-to-toxic transformation of the amyloid-forming protein model Hen Egg White Lysozyme (HEWL). The sp2-rich CACQDs also scavenged free radicals, a milestone along the PD trajectory. In-vitro, CACQDs introduced into a human neuroblastoma-derived cell line (SH-SY5Y) demonstrated negligible cytotoxicity up to 5 mg/mL and protected the cell line against oxidative stress-induced neuronal injury induced by the pesticide and potent neurotoxin, paraquat. Our findings suggest that the potentially BBB-penetrating CACQDs derived from caffeic acid hold promise for mitigating neurodegenerative disorders associated with environmental pesticides and xenobiotic neurotoxicants. Importantly, CACQDs sourced from coffee, coupled with their facile synthesis, represent a sustainable, green chemistry platform for generating interventional candidates in neurodegeneration.
Collapse
Affiliation(s)
- Jyotish Kumar
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, TX, 79968, United States
| | - Sofia A Delgado
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, TX, 79968, United States
| | - Hemen Sarma
- Department of Botany, Bodoland University, Rangalikhata, Deborgaon, Kokrajhar (BTR), Assam, 783370, India.
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, TX, 79968, United States.
| |
Collapse
|
2
|
Hou L, Liu J, Sun F, Huang R, Chang R, Ruan Z, Wang Y, Zhao J, Wang Q. Integrin Mac1 mediates paraquat and maneb-induced learning and memory impairments in mice through NADPH oxidase-NLRP3 inflammasome axis-dependent microglial activation. J Neuroinflammation 2023; 20:42. [PMID: 36804009 PMCID: PMC9938991 DOI: 10.1186/s12974-023-02732-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 02/13/2023] [Indexed: 02/20/2023] Open
Abstract
INTRODUCTION The mechanisms of cognitive impairments in Parkinson's disease (PD) remain unknown. Accumulating evidence revealed that brain neuroinflammatory response mediated by microglial cells contributes to cognitive deficits in neuropathological conditions and macrophage antigen complex-1 (Mac1) is a key factor in controlling microglial activation. OBJECTIVES To explore whether Mac1-mediated microglial activation participates in cognitive dysfunction in PD using paraquat and maneb-generated mouse PD model. METHODS Cognitive performance was measured in wild type and Mac1-/- mice using Morris water maze test. The role and mechanisms of NADPH oxidase (NOX)-NLRP3 inflammasome axis in Mac1-mediated microglial dysfunction, neuronal damage, synaptic degeneration and phosphorylation (Ser129) of α-synuclein were explored by immunohistochemistry, Western blot and RT-PCR. RESULTS Genetic deletion of Mac1 significantly ameliorated learning and memory impairments, neuronal damage, synaptic loss and α-synuclein phosphorylation (Ser129) caused by paraquat and maneb in mice. Subsequently, blocking Mac1 activation was found to mitigate paraquat and maneb-elicited microglial NLRP3 inflammasome activation in both in vivo and in vitro. Interestingly, stimulating activation of NOX by phorbol myristate acetate abolished the inhibitory effects of Mac1 blocking peptide RGD on paraquat and maneb-provoked NLRP3 inflammasome activation, indicating a key role of NOX in Mac1-mediated NLRP3 inflammasome activation. Furthermore, NOX1 and NOX2, two members of NOX family, and downstream PAK1 and MAPK pathways were recognized to be essential for NOX to regulate NLRP3 inflammasome activation. Finally, a NLRP3 inflammasome inhibitor glybenclamide abrogated microglial M1 activation, neurodegeneration and phosphorylation (Ser129) of α-synuclein elicited by paraquat and maneb, which were accompanied by improved cognitive capacity in mice. CONCLUSIONS Mac1 was involved in cognitive dysfunction in a mouse PD model through NOX-NLRP3 inflammasome axis-dependent microglial activation, providing a novel mechanistic basis of cognitive decline in PD.
Collapse
Affiliation(s)
- Liyan Hou
- grid.411971.b0000 0000 9558 1426Dalian Medical University Library, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China ,grid.411971.b0000 0000 9558 1426National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044 China
| | - Jianing Liu
- grid.411971.b0000 0000 9558 1426School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China
| | - Fuqiang Sun
- grid.411971.b0000 0000 9558 1426School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China
| | - Ruixue Huang
- grid.411971.b0000 0000 9558 1426School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China
| | - Rui Chang
- grid.411971.b0000 0000 9558 1426School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China
| | - Zhengzheng Ruan
- grid.411971.b0000 0000 9558 1426School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China
| | - Ying Wang
- grid.411971.b0000 0000 9558 1426School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China.
| | - Qingshan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China. .,School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China.
| |
Collapse
|
3
|
Brahadeeswaran S, Lateef M, Calivarathan L. An Insight into the Molecular Mechanism of Mitochondrial Toxicant-induced Neuronal Apoptosis in Parkinson's Disease. Curr Mol Med 2023; 23:63-75. [PMID: 35125081 DOI: 10.2174/1566524022666220203163631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 11/25/2021] [Accepted: 12/07/2021] [Indexed: 12/16/2022]
Abstract
Parkinson's disease (PD) is one of the most common progressive neurodegenerative disorders affecting approximately 1% of the world's population at the age of 50 and above. Majority of PD cases are sporadic and show symptoms after the age of 60 and above. At that time, most of the dopaminergic neurons in the region of substantia nigra pars compacta have been degenerated. Although in past decades, discoveries of genetic mutations linked to PD have significantly impacted our current understanding of the pathogenesis of this devastating disorder, it is likely that the environment also plays a critical role in the etiology of sporadic PD. Recent epidemiological and experimental studies indicate that exposure to environmental agents, including a number of agricultural and industrial chemicals, may contribute to the pathogenesis of several neurodegenerative disorders, including PD. Furthermore, there is a strong correlation between mitochondrial dysfunction and several forms of neurodegenerative disorders, including Alzheimer's disease (AD), Huntington's disease (HD), Amyotrophic lateral sclerosis (ALS) and PD. Interestingly, substantia nigra of patients with PD has been shown to have a mild deficiency in mitochondrial respiratory electron transport chain NADH dehydrogenase (Complex I) activity. This review discusses the role of mitochondrial toxicants in the selective degeneration of dopaminergic neurons targeting the electron transport system that leads to Parkinsonism.
Collapse
Affiliation(s)
- Subhashini Brahadeeswaran
- Molecular Pharmacology & Toxicology Laboratory, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Neelakudi Campus, Thiruvarur - 610005, India
| | - Mohammad Lateef
- Department of Animal Sciences, School of Life Sciences, Central University of Kashmir, Nunar Campus, Ganderbal - 191201, Jammu & Kashmir, India
| | - Latchoumycandane Calivarathan
- Molecular Pharmacology & Toxicology Laboratory, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Neelakudi Campus, Thiruvarur - 610005, India
| |
Collapse
|
4
|
Kvetkina A, Pislyagin E, Menchinskaya E, Yurchenko E, Kalina R, Kozlovskiy S, Kaluzhskiy L, Menshov A, Kim N, Peigneur S, Tytgat J, Ivanov A, Ayvazyan N, Leychenko E, Aminin D. Kunitz-Type Peptides from Sea Anemones Protect Neuronal Cells against Parkinson's Disease Inductors via Inhibition of ROS Production and ATP-Induced P2X7 Receptor Activation. Int J Mol Sci 2022; 23:ijms23095115. [PMID: 35563513 PMCID: PMC9103184 DOI: 10.3390/ijms23095115] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/29/2022] [Accepted: 04/29/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is a socially significant disease, during the development of which oxidative stress and inflammation play a significant role. Here, we studied the neuroprotective effects of four Kunitz-type peptides from Heteractis crispa and Heteractis magnifica sea anemones against PD inductors. The peptide HCIQ1c9, which was obtained for the first time, inhibited trypsin less than other peptides due to unfavorable interactions of Arg17 with Lys43 in the enzyme. Its activity was reduced by up to 70% over the temperature range of 60–100 °C, while HCIQ2c1, HCIQ4c7, and HMIQ3c1 retained their conformation and stayed active up to 90–100 °C. All studied peptides inhibited paraquat- and rotenone-induced intracellular ROS formation, in particular NO, and scavenged free radicals outside the cells. The peptides did not modulate the TRPV1 channels but they affected the P2X7R, both of which are considered therapeutic targets in Parkinson’s disease. HMIQ3c1 and HCIQ4c7 almost completely inhibited the ATP-induced uptake of YO-PRO-1 dye in Neuro-2a cells through P2X7 ion channels and significantly reduced the stable calcium response in these cells. The complex formation of the peptides with the P2X7R extracellular domain was determined via SPR analysis. Thus, these peptides may be considered promising compounds to protect neuronal cells against PD inductors, which act as ROS production inhibitors and partially act as ATP-induced P2X7R activation inhibitors.
Collapse
Affiliation(s)
- Aleksandra Kvetkina
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Evgeny Pislyagin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Ekaterina Menchinskaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Ekaterina Yurchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Rimma Kalina
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Sergei Kozlovskiy
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Leonid Kaluzhskiy
- V.N. Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya St., 119121 Moscow, Russia; (L.K.); (A.I.)
| | - Alexander Menshov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Natalia Kim
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Steve Peigneur
- Toxicology and Pharmacology, Campus Gasthuisberg O&N2, University of Leuven (KU Leuven), Herestraat 49, P.O. Box 922, B-3000 Leuven, Belgium; (S.P.); (J.T.)
| | - Jan Tytgat
- Toxicology and Pharmacology, Campus Gasthuisberg O&N2, University of Leuven (KU Leuven), Herestraat 49, P.O. Box 922, B-3000 Leuven, Belgium; (S.P.); (J.T.)
| | - Alexis Ivanov
- V.N. Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya St., 119121 Moscow, Russia; (L.K.); (A.I.)
| | - Naira Ayvazyan
- L.A. Orbeli Institute of Physiology, National Academy of Sciences of Armenia, Yerevan 0028, Armenia;
| | - Elena Leychenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Dmitry Aminin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
- Correspondence:
| |
Collapse
|
5
|
Klonarakis M, De Vos M, Woo E, Ralph L, Thacker JS, Gil-Mohapel J. The three sisters of fate: Genetics, pathophysiology and outcomes of animal models of neurodegenerative diseases. Neurosci Biobehav Rev 2022; 135:104541. [DOI: 10.1016/j.neubiorev.2022.104541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 11/28/2021] [Accepted: 01/13/2022] [Indexed: 02/07/2023]
|
6
|
Johnson AM, Ou ZYA, Gordon R, Saminathan H. Environmental neurotoxicants and inflammasome activation in Parkinson's disease - A focus on the gut-brain axis. Int J Biochem Cell Biol 2022; 142:106113. [PMID: 34737076 DOI: 10.1016/j.biocel.2021.106113] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 10/25/2021] [Accepted: 10/29/2021] [Indexed: 12/26/2022]
Abstract
Inflammasomes are multi-protein complexes expressed in immune cells that function as intracellular sensors of environmental, metabolic and cellular stress. Inflammasome activation in the brain, has been shown to drive neuropathology and disease progression by multiple mechanisms, making it one of the most attractive therapeutic targets for disease modification in Parkinson's Disease (PD). Extensive inflammasome activation is evident in the brains of people with PD at the sites of dopaminergic degeneration and synuclein aggregation. While substantial progress has been made on validating inflammasome activation as a therapeutic target for PD, the mechanisms by which inflammasome activation is triggered and sustained over the disease course remain poorly understood. A growing body of evidence point to environmental and occupational chemical exposures as possible triggers of inflammasome activation in PD. The involvement of the gastrointestinal system and gut microbiota in PD pathophysiology is beginning to be elucidated, especially the profound link between gut dysbiosis and immune activation. While large cohort studies confirmed specific changes in the gut microbiota in PD patients compared to age-matched healthy controls, recent research suggest that synuclein pathology could be initiated in the gastrointestinal tract. In this review, we present a summarized perspective on current understanding on inflammasome activation and the gut-brain-axis link during PD pathophysiology. We discuss multiple environmental toxicants that are implicated as the etiological agents in causing idiopathic PD and their mechanistic underpinnings during neuroinflammatory events. We additionally present future directions that needs to address the research questions related to the gut-microbiome-brain mechanisms in PD.
Collapse
Affiliation(s)
- Aishwarya M Johnson
- Department of Veterinary Medicine, College of Food and Agriculture, United Arab Emirates University, Al Ain, UAE
| | - Zhen-Yi Andy Ou
- Translational Neuroscience Laboratory, UQ Centre for Clinical Research, The University of Queensland, Australia; School of Biomedical Sciences, University of Queensland, Australia
| | - Richard Gordon
- Translational Neuroscience Laboratory, UQ Centre for Clinical Research, The University of Queensland, Australia; School of Biomedical Sciences, University of Queensland, Australia
| | - Hariharan Saminathan
- Department of Veterinary Medicine, College of Food and Agriculture, United Arab Emirates University, Al Ain, UAE.
| |
Collapse
|
7
|
Wen S, Aki T, Unuma K, Uemura K. Chemically Induced Models of Parkinson's Disease: History and Perspectives for the Involvement of Ferroptosis. Front Cell Neurosci 2020; 14:581191. [PMID: 33424553 PMCID: PMC7786020 DOI: 10.3389/fncel.2020.581191] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022] Open
Abstract
Ferroptosis is a newly discovered form of necrotic cell death characterized by its dependency on iron and lipid peroxidation. Ferroptosis has attracted much attention recently in the area of neurodegeneration since the involvement of ferroptosis in Parkinson’s disease (PD), a major neurodegenerative disease, has been indicated using animal models. Although PD is associated with both genetic and environmental factors, sporadic forms of PD account for more than 90% of total PD. Following the importance of environmental factors, various neurotoxins are used as chemical inducers of PD both in vivo and in vitro. In contrast to other neurodegenerative diseases such as Alzheimer’s and Huntington’s diseases (AD and HD), many of the characteristics of PD can be reproduced in vivo by the use of specific neurotoxins. Given the indication of ferroptosis in PD pathology, several studies have been conducted to examine whether ferroptosis plays role in the loss of dopaminergic neurons in PD. However, there are still few reports showing an authentic form of ferroptosis in neuronal cells during exposure to the neurotoxins used as PD inducers. In this review article, we summarize the history of the uses of chemicals to create PD models in vivo and in vitro. Besides, we also survey recent reports examining the possible involvement of ferroptosis in chemical models of PD.
Collapse
Affiliation(s)
- Shuheng Wen
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshihiko Aki
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kana Unuma
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koichi Uemura
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
8
|
Vaccari C, El Dib R, Gomaa H, Lopes LC, de Camargo JL. Paraquat and Parkinson's disease: a systematic review and meta-analysis of observational studies. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2019; 22:172-202. [PMID: 31476981 DOI: 10.1080/10937404.2019.1659197] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
This investigation aimed to conduct a systematic review of the literature and meta-analysis to determine whether exposure to the herbicide paraquat was associated with the development of Parkinson's disease (PD). Observational studies that enrolled adults exposed to paraquat with PD as the outcome of interest were searched in the PubMed, Embase, LILACS, TOXNET, and Web of Science databases up to May 2019. Two authors independently selected relevant studies, extracted data, and assessed methodological quality. The evidence certainty was assessed by the GRADE approach, which served as basis for a tentative causality assessment, supplemented by the Bradford Hill criteria when necessary. Results from nine case-control studies indicated that PD occurrence was 25% higher in participants exposed to paraquat. The only cohort investigation included demonstrated a non-significant OR of 1.08. Results from subgroup analyses also indicated higher PD frequency in participants that were exposed to paraquat for longer periods or individuals co-exposed with paraquat and any other dithiocarbamate. Data indicate apositive association between exposure to paraquat and PD occurrence, but the weight-of-evidence does not enable one to assume an indisputable cause-effect relationship between these two conditions. Better designed studies are needed to increase confidence in results. Systematic Review Registration: PROSPERO CRD42017069994.
Collapse
Affiliation(s)
- Carolina Vaccari
- Department of Pathology, São Paulo State University (UNESP) , Botucatu , Brazil
| | - Regina El Dib
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP) , São Paulo , Brazil
- McMaster Institute of Urology, St. Joseph's Healthcare, McMaster University , Hamilton , Canada
- Department of Community Health and Epidemiology, Dalhousie University , Halifax , Canada
| | - Huda Gomaa
- Department of Bio-statistics, High Institute of Public Health, Alexandria University , Alexandria , Egypt
- Drug Information Center, Tanta Chest Hospital, Ministry of Health , Tanta , Egypt
| | - Luciane C Lopes
- Department of Pharmaceutical Sciences, University of Sorocaba (UNISO) , Sorocaba , Brazil
| | | |
Collapse
|
9
|
Ferreira-Junior NC, Campos AC, Guimarães FS, Del-Bel E, Zimmermann PMDR, Brum Junior L, Hallak JE, Crippa JA, Zuardi AW. Biological bases for a possible effect of cannabidiol in Parkinson's disease. ACTA ACUST UNITED AC 2019; 42:218-224. [PMID: 31314869 PMCID: PMC7115443 DOI: 10.1590/1516-4446-2019-0460] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/08/2019] [Indexed: 01/10/2023]
Abstract
Current pharmacotherapy of Parkinson’s disease (PD) is palliative and unable to modify the progression of neurodegeneration. Treatments that can improve patients’ quality of life with fewer side effects are needed, but not yet available. Cannabidiol (CBD), the major non-psychotomimetic constituent of cannabis, has received considerable research attention in the last decade. In this context, we aimed to critically review the literature on potential therapeutic effects of CBD in PD and discuss clinical and preclinical evidence supporting the putative neuroprotective mechanisms of CBD. We searched MEDLINE (via PubMed) for indexed articles published in English from inception to 2019. The following keywords were used: cannabis; cannabidiol and neuroprotection; endocannabinoids and basal ganglia; Parkinson’s animal models; Parkinson’s history; Parkinson’s and cannabidiol. Few studies addressed the biological bases for the purported effects of CBD on PD. Six preclinical studies showed neuroprotective effects, while three targeted the antidyskinetic effects of CBD. Three human studies have tested CBD in patients with PD: an open-label study, a case series, and a randomized controlled trial. These studies reported therapeutic effects of CBD on non-motor symptoms. Additional research is needed to elucidate the potential effectiveness of CBD in PD and the underlying mechanisms involved.
Collapse
Affiliation(s)
- Nilson C Ferreira-Junior
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto (FMRP), Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Alline C Campos
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto (FMRP), Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Francisco S Guimarães
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto (FMRP), Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Elaine Del-Bel
- Departamento de Morfologia, Fisiologia e Patologia Básica, Faculdade de Odontologia de Ribeirão Preto (FORP), USP, Ribeirão Preto, SP, Brazil
| | | | | | - Jaime E Hallak
- Departamento de Neurociências e Ciências do Comportamento, FMRP, USP, Ribeirão Preto, SP, Brazil
| | - José A Crippa
- Departamento de Neurociências e Ciências do Comportamento, FMRP, USP, Ribeirão Preto, SP, Brazil
| | - Antonio W Zuardi
- Departamento de Neurociências e Ciências do Comportamento, FMRP, USP, Ribeirão Preto, SP, Brazil
| |
Collapse
|
10
|
Neuroprotective effects of coenzyme Q10 on paraquat-induced Parkinson's disease in experimental animals. Behav Pharmacol 2019; 29:79-86. [PMID: 28902670 DOI: 10.1097/fbp.0000000000000342] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Parkinson's disease (PD) affects ∼1-2% of the elderly population. Development of a neuroprotective therapy that may be initiated early in the course of the disease to retard/prevent disease progression is highly desirable. This study aimed to investigate prophylactic treatment with coenzyme Q10 (CoQ10) before paraquat (PQ) exposure, a herbicide known to increase the risk for PD, to attain neuroprotection. In addition, therapeutic intervention with CoQ10 in mice already exposed to PQ (24 h) might halt ongoing neurodegeneration and behavioural deterioration. PD was induced experimentally in mice by an injection of PQ (10 mg/kg, intraperitoneal), twice a week for 3 consecutive weeks, either before or after the initiation of treatment with CoQ10 (200 mg/kg). The results of the sustained supplementation with CoQ10, prophylactically and therapeutically, were compared with L-DOPA (100 mg/kg). A battery of behavioural tests was performed, in addition to estimation of protein carbonyl in the brain. CoQ10 elicited a remarkable improvement in most of the behavioural tests and decreased protein carbonyl content in the brain, particularly when it was initiated before rather than after PQ induction of PD. Therefore, CoQ10, which protects against mitochondrial damage, may be beneficial in slowing the progression of PD, particularly when initiated as prophylactic treatment.
Collapse
|
11
|
Anselmi L, Bove C, Coleman FH, Le K, Subramanian MP, Venkiteswaran K, Subramanian T, Travagli RA. Ingestion of subthreshold doses of environmental toxins induces ascending Parkinsonism in the rat. NPJ Parkinsons Dis 2018; 4:30. [PMID: 30302391 PMCID: PMC6160447 DOI: 10.1038/s41531-018-0066-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 08/20/2018] [Accepted: 08/28/2018] [Indexed: 01/08/2023] Open
Abstract
Increasing evidence suggests that environmental neurotoxicants or misfolded α-synuclein generated by such neurotoxicants are transported from the gastrointestinal tract to the central nervous system via the vagus nerve, triggering degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and causing Parkinson's disease (PD). We tested the hypothesis that gastric co-administration of subthreshold doses of lectins and paraquat can recreate the pathology and behavioral manifestations of PD in rats. A solution containing paraquat + lectin was administered daily for 7 days via gastric gavage, followed by testing for Parkinsonian behavior and gastric dysmotility. At the end of the experiment, brainstem and midbrain tissues were analyzed for the presence of misfolded α-synuclein and neuronal loss in the SNpc and in the dorsal motor nucleus of the vagus (DMV). Misfolded α-synuclein was found in DMV and SNpc neurons. A significant decrease in tyrosine hydroxylase positive dopaminergic neurons was noted in the SNpc, conversely there was no apparent loss of cholinergic neurons of the DMV. Nigrovagally-evoked gastric motility was impaired in treated rats prior to the onset of parkinsonism, the motor deficits of which were improved by l-dopa treatment. Vagotomy prevented the development of parkinsonian symptoms and constrained the appearance of misfolded α-synuclein to myenteric neurons. These data demonstrate that co-administration of subthreshold doses of paraquat and lectin induces progressive, l-dopa-responsive parkinsonism that is preceded by gastric dysmotility. This novel preclinical model of environmentally triggered PD provides functional support for Braak's staging hypothesis of idiopathic PD.
Collapse
Affiliation(s)
- L. Anselmi
- Department of Neural and Behavioral Sciences, Penn State—College of Medicine, Hershey, PA USA
| | - C. Bove
- Department of Neural and Behavioral Sciences, Penn State—College of Medicine, Hershey, PA USA
| | - F. H. Coleman
- Department of Neural and Behavioral Sciences, Penn State—College of Medicine, Hershey, PA USA
| | - K. Le
- Department of Neurology, Penn State—College of Medicine, Hershey, PA USA
| | - M. P. Subramanian
- Department of Neural and Behavioral Sciences, Penn State—College of Medicine, Hershey, PA USA
| | - K. Venkiteswaran
- Department of Neural and Behavioral Sciences, Penn State—College of Medicine, Hershey, PA USA
- Department of Neurology, Penn State—College of Medicine, Hershey, PA USA
| | - T. Subramanian
- Department of Neural and Behavioral Sciences, Penn State—College of Medicine, Hershey, PA USA
- Department of Neurology, Penn State—College of Medicine, Hershey, PA USA
| | - R. A. Travagli
- Department of Neural and Behavioral Sciences, Penn State—College of Medicine, Hershey, PA USA
| |
Collapse
|
12
|
Revisiting the Paraquat-Induced Sporadic Parkinson's Disease-Like Model. Mol Neurobiol 2018; 56:1044-1055. [PMID: 29862459 DOI: 10.1007/s12035-018-1148-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 05/23/2018] [Indexed: 02/05/2023]
Abstract
Parkinson's disease (PD) is a major neurodegenerative disorder that affects 1-2% of the total global population. Despite its high prevalence and publication of several studies focused on understanding its pathology, an effective treatment that stops and/or reverses the damage to dopaminergic neurons is unavailable. Similar to other neurodegenerative disorders, PD etiology may be linked to several factors, including genetic susceptibility and environmental elements. Regarding environmental factors, several neurotoxic pollutants, including 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), have been identified. Moreover, some pesticides/herbicides, such as rotenone, paraquat (PQ), maneb (MB), and mancozeb (MZ), cause neurotoxicity and induce a PD-like pathology. Based on these findings, several in vitro and in vivo PD-like models have been developed to understand the pathophysiology of PD and evaluate different therapeutic strategies to fight dopaminergic neurodegeneration. 6-OHDA and MPTP are common models used in PD research, and pesticide-based approaches have become secondary models of study. However, some herbicides, such as PQ, are commonly used by farming laborers in developing countries. Thus, the present review summarizes the relevant scientific background regarding the use and effects of chronic exposure to PQ in the context of PD. Similarly, we discuss the relevance of PD-like models developed using this agrochemical compound.
Collapse
|
13
|
Vaz RL, Outeiro TF, Ferreira JJ. Zebrafish as an Animal Model for Drug Discovery in Parkinson's Disease and Other Movement Disorders: A Systematic Review. Front Neurol 2018; 9:347. [PMID: 29910763 PMCID: PMC5992294 DOI: 10.3389/fneur.2018.00347] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/30/2018] [Indexed: 12/21/2022] Open
Abstract
Movement disorders can be primarily divided into hypokinetic and hyperkinetic. Most of the hypokinetic syndromes are associated with the neurodegenerative disorder Parkinson’s disease (PD). By contrast, hyperkinetic syndromes encompass a broader array of diseases, including dystonia, essential tremor, or Huntington’s disease. The discovery of effective therapies for these disorders has been challenging and has also involved the development and characterization of accurate animal models for the screening of new drugs. Zebrafish constitutes an alternative vertebrate model for the study of movement disorders. The neuronal circuitries involved in movement in zebrafish are well characterized, and most of the associated molecular mechanisms are highly conserved. Particularly, zebrafish models of PD have contributed to a better understanding of the role of several genes implicated in the disease. Furthermore, zebrafish is a vertebrate model particularly suited for large-scale drug screenings. The relatively small size of zebrafish, optical transparency, and lifecycle, are key characteristics that facilitate the study of multiple compounds at the same time. Several transgenic, knockdown, and mutant zebrafish lines have been generated and characterized. Therefore, it is central to critically analyze these zebrafish lines and understand their suitability as models of movement disorders. Here, we revise the pathogenic mechanisms, phenotypes, and responsiveness to pharmacotherapies of zebrafish lines of the most common movement disorders. A systematic review of the literature was conducted by including all studies reporting the characterization of zebrafish models of the movement disorders selected from five bibliographic databases. A total of 63 studies were analyzed, and the most relevant data within the scope of this review were gathered. The majority (62%) of the studies were focused in the characterization of zebrafish models of PD. Overall, the zebrafish models included display conserved biochemical and neurobehavioral features of the phenomenology in humans. Nevertheless, in light of what is known for all animal models available, the use of zebrafish as a model for drug discovery requires further optimization. Future technological developments alongside with a deeper understanding of the molecular bases of these disorders should enable the development of novel zebrafish lines that can prove useful for drug discovery for movement disorders.
Collapse
Affiliation(s)
- Rita L Vaz
- TechnoPhage, SA, Lisboa, Portugal.,Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany.,Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,CEDOC, Chronic Diseases Research Centre, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal.,The Medical School, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Joaquim J Ferreira
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal.,Laboratory of Clinical Pharmacology and Therapeutics, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,CNS-Campus Neurológico Sénior, Torres Vedras, Portugal
| |
Collapse
|
14
|
Jiang P, Dickson DW. Parkinson's disease: experimental models and reality. Acta Neuropathol 2018; 135:13-32. [PMID: 29151169 PMCID: PMC5828522 DOI: 10.1007/s00401-017-1788-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is a chronic, progressive movement disorder of adults and the second most common neurodegenerative disease after Alzheimer's disease. Neuropathologic diagnosis of PD requires moderate-to-marked neuronal loss in the ventrolateral substantia nigra pars compacta and α-synuclein (αS) Lewy body pathology. Nigrostriatal dopaminergic neurodegeneration correlates with the Parkinsonian motor features, but involvement of other peripheral and central nervous system regions leads to a wide range of non-motor features. Nigrostriatal dopaminergic neurodegeneration is shared with other parkinsonian disorders, including some genetic forms of parkinsonism, but many of these disorders do not have Lewy bodies. An ideal animal model for PD, therefore, should exhibit age-dependent and progressive dopaminergic neurodegeneration, motor dysfunction, and abnormal αS pathology. Rodent models of PD using genetic or toxin based strategies have been widely used in the past several decades to investigate the pathogenesis and therapeutics of PD, but few recapitulate all the major clinical and pathologic features of PD. It is likely that new strategies or better understanding of fundamental disease processes may facilitate development of better animal models. In this review, we highlight progress in generating rodent models of PD based on impairments of four major cellular functions: mitochondrial oxidative phosphorylation, autophagy-lysosomal metabolism, ubiquitin-proteasome protein degradation, and endoplasmic reticulum stress/unfolded protein response. We attempt to evaluate how impairment of these major cellular systems contribute to PD and how they can be exploited in rodent models. In addition, we review recent cell biological studies suggesting a link between αS aggregation and impairment of nuclear membrane integrity, as observed during cellular models of apoptosis. We also briefly discuss the role of incompetent phagocytic clearance and how this may be a factor to consider in developing new rodent models of PD.
Collapse
Affiliation(s)
- Peizhou Jiang
- Neuropathology Laboratory, Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Dennis W Dickson
- Neuropathology Laboratory, Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
15
|
Smeyne RJ, Breckenridge CB, Beck M, Jiao Y, Butt MT, Wolf JC, Zadory D, Minnema DJ, Sturgess NC, Travis KZ, Cook AR, Smith LL, Botham PA. Assessment of the Effects of MPTP and Paraquat on Dopaminergic Neurons and Microglia in the Substantia Nigra Pars Compacta of C57BL/6 Mice. PLoS One 2016; 11:e0164094. [PMID: 27788145 PMCID: PMC5082881 DOI: 10.1371/journal.pone.0164094] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 09/20/2016] [Indexed: 12/21/2022] Open
Abstract
The neurotoxicity of paraquat dichloride (PQ) was assessed in two inbred strains of 9- or 16-week old male C57BL/6 mice housed in two different laboratories and compared to the effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). PQ was administered by intraperitoneal injections; either once (20 mg/kg) or twice (10 mg/kg) weekly for 3 weeks, while MPTP-HCl was injected 4 times on a single day (20 mg/kg/dose). Brains were collected 8, 16, 24, 48, 96 or 168 hours after the last PQ treatment, and 48 or 168 hours after MPTP treatment. Dopamine neurons in the substantia nigra pars compacta (SNpc) were identified by antibodies to tyrosine hydroxylase (TH+) and microglia were identified using Iba-1 immunoreactivity. The total number of TH+ neurons and the number of resting and activated microglia in the SNpc at 168 hours after the last dose were estimated using model- or design-based stereology, with investigators blinded to treatment. In a further analysis, a pathologist, also blinded to treatment, evaluated the SNpc and/or striatum for loss of TH+ neurons (SNpc) or terminals (striatum), cell death (as indicated by amino cupric silver uptake, TUNEL and/or caspase 3 staining) and neuroinflammation (as indicated by Iba-1 and/or GFAP staining). PQ, administered either once or twice weekly to 9- or 16-week old mice from two suppliers, had no effect on the number of TH+ neurons or microglia in the SNpc, as assessed by two groups, each blinded to treatment, using different stereological methods. PQ did not induce neuronal cell loss or degeneration in the SNpc or striatum. Additionally, there was no evidence of apoptosis, microgliosis or astrogliosis. In MPTP-treated mice, the number of TH+ neurons in the SNpc was significantly decreased and the number of activated microglia increased. Histopathological assessment found degenerating neurons/terminals in the SNpc and striatum but no evidence of apoptotic cell death. MPTP activated microglia in the SNpc and increased the number of astrocytes in the SNpc and striatum.
Collapse
Affiliation(s)
- Richard Jay Smeyne
- St. Jude Children’s Research Hospital, Dept. of Developmental Neurobiology, 262 Danny Thomas Place, Memphis, TN 38105, United States of America
- * E-mail:
| | - Charles B. Breckenridge
- Syngenta Crop Protection, LLC, P.O. Box 18300, Greensboro, NC 27419–8300, United States of America
| | - Melissa Beck
- WIL Research Laboratories, LLC., Ashland, OH 44805, United States of America
| | - Yun Jiao
- St. Jude Children’s Research Hospital, Dept. of Developmental Neurobiology, 262 Danny Thomas Place, Memphis, TN 38105, United States of America
| | - Mark T. Butt
- Tox Path Specialists, LLC, 8747 Chestnut Grove Road, Frederick, MD 21701–2607, United States of America
| | - Jeffrey C. Wolf
- Experimental Pathology Laboratories, Inc., 45600 Terminal Drive, Sterling, VA 20166, United States of America
| | - Dan Zadory
- Experimental Pathology Laboratories, Inc., 45600 Terminal Drive, Sterling, VA 20166, United States of America
| | - Daniel J. Minnema
- Syngenta Crop Protection, LLC, P.O. Box 18300, Greensboro, NC 27419–8300, United States of America
| | - Nicholas C. Sturgess
- Syngenta Limited, Jealott's Hill International Research Centre, Bracknell, Berkshire, RG42 6EY, United Kingdom
| | - Kim Z. Travis
- Syngenta Limited, Jealott's Hill International Research Centre, Bracknell, Berkshire, RG42 6EY, United Kingdom
| | - Andrew R. Cook
- Syngenta Limited, Jealott's Hill International Research Centre, Bracknell, Berkshire, RG42 6EY, United Kingdom
| | - Lewis L. Smith
- University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Philip A. Botham
- Syngenta Limited, Jealott's Hill International Research Centre, Bracknell, Berkshire, RG42 6EY, United Kingdom
| |
Collapse
|
16
|
Parkinson's Disease: The Mitochondria-Iron Link. PARKINSONS DISEASE 2016; 2016:7049108. [PMID: 27293957 PMCID: PMC4886095 DOI: 10.1155/2016/7049108] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 12/14/2022]
Abstract
Mitochondrial dysfunction, iron accumulation, and oxidative damage are conditions often found in damaged brain areas of Parkinson's disease. We propose that a causal link exists between these three events. Mitochondrial dysfunction results not only in increased reactive oxygen species production but also in decreased iron-sulfur cluster synthesis and unorthodox activation of Iron Regulatory Protein 1 (IRP1), a key regulator of cell iron homeostasis. In turn, IRP1 activation results in iron accumulation and hydroxyl radical-mediated damage. These three occurrences-mitochondrial dysfunction, iron accumulation, and oxidative damage-generate a positive feedback loop of increased iron accumulation and oxidative stress. Here, we review the evidence that points to a link between mitochondrial dysfunction and iron accumulation as early events in the development of sporadic and genetic cases of Parkinson's disease. Finally, an attempt is done to contextualize the possible relationship between mitochondria dysfunction and iron dyshomeostasis. Based on published evidence, we propose that iron chelation-by decreasing iron-associated oxidative damage and by inducing cell survival and cell-rescue pathways-is a viable therapy for retarding this cycle.
Collapse
|
17
|
Multifactorial theory applied to the neurotoxicity of paraquat and paraquat-induced mechanisms of developing Parkinson's disease. J Transl Med 2016; 96:496-507. [PMID: 26829122 DOI: 10.1038/labinvest.2015.161] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 10/07/2015] [Accepted: 10/12/2015] [Indexed: 11/08/2022] Open
Abstract
Laboratory studies involving repeated exposure to paraquat (PQ) in different animal models can induce many of the pathological features of Parkinson's disease (PD), such as the loss of dopaminergic neurons in the nigrostriatal dopamine system. Epidemiological studies identify an increased risk of developing PD in human populations living in areas where PQ exposure is likely to occur and among workers lacking appropriate protective equipment. The mechanisms involved in developing PD may not be due to any single cause, but rather a multifactorial situation may exist where PQ exposure may cause PD in some circumstances. Multifactorial theory is adopted into this review that includes a number of sub-cellular mechanisms to explain the pathogenesis of PD. The theory is placed into an environmental context of chronic low-dose exposure to PQ that consequently acts as an oxidative stress inducer. Oxidative stress and the metabolic processes of PQ-inducing excitotoxicity, α-synuclein aggregate formation, autophagy, alteration of dopamine catabolism, and inactivation of tyrosine hydroxylase are positioned as causes for the loss of dopaminergic cells. The environmental context and biochemistry of PQ in soils, water, and organisms is also reviewed to identify potential routes that can lead to chronic rates of low-dose exposure that would replicate the type of response that is observed in animal models, epidemiological studies, and other types of laboratory investigations involving PQ exposure. The purpose of this review is to synthesize key relations and summarize hypotheses linking PD to PQ exposure by using the multifactorial approach. Recommendations are given to integrate laboratory methods to the environmental context as a means to improve on experimental design. The multifactorial approach is necessary for conducting valid tests of causal relations, for understanding of potential relations between PD and PQ exposure, and may prevent further delay in solving what has proven to be an evasive etiological problem.
Collapse
|
18
|
Jagmag SA, Tripathi N, Shukla SD, Maiti S, Khurana S. Evaluation of Models of Parkinson's Disease. Front Neurosci 2016; 9:503. [PMID: 26834536 PMCID: PMC4718050 DOI: 10.3389/fnins.2015.00503] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/21/2015] [Indexed: 12/01/2022] Open
Abstract
Parkinson's disease is one of the most common neurodegenerative diseases. Animal models have contributed a large part to our understanding and therapeutics developed for treatment of PD. There are several more exhaustive reviews of literature that provide the initiated insights into the specific models; however a novel synthesis of the basic advantages and disadvantages of different models is much needed. Here we compare both neurotoxin based and genetic models while suggesting some novel avenues in PD modeling. We also highlight the problems faced and promises of all the mammalian models with the hope of providing a framework for comparison of various systems.
Collapse
Affiliation(s)
- Shail A Jagmag
- Department of Biology, Indian Institute of Science Education and Research Kolkata, India
| | - Naveen Tripathi
- Department of Biology, Indian Institute of Science Education and Research Kolkata, India
| | - Sunil D Shukla
- Department of Zoology, Government Meera Girl's College Udaipur, India
| | - Sankar Maiti
- Department of Biology, Indian Institute of Science Education and Research Kolkata, India
| | - Sukant Khurana
- Department of Biology, Indian Institute of Science Education and Research Kolkata, India
| |
Collapse
|
19
|
Dietary administration of diquat for 13 weeks does not result in a loss of dopaminergic neurons in the substantia nigra of C57BL/6J mice. Regul Toxicol Pharmacol 2015; 75:81-8. [PMID: 26683030 DOI: 10.1016/j.yrtph.2015.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 11/24/2022]
Abstract
Male and female C57BL/6J mice were administered diquat dibromide (DQ∙Br2) in their diets at concentrations of 0 (control), 12.5 and 62.5 ppm for 13 weeks to assess the potential effects of DQ on the nigrostriatal dopaminergic system. Achieved dose levels at 62.5 ppm were 6.4 and 7.6 mg DQ (ion)/kg bw/day for males and females, respectively. A separate group of mice was administered 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) ip as a positive control. The comparative effects of DQ and MPTP on the substantia nigra pars compacta (SNpc) and/or striatum were assessed using neurochemical, neuropathological and stereological endpoints. Morphological and stereological assessments were performed by investigators who were "blinded" to dose group. DQ had no effect on striatal dopamine concentration or dopamine turnover. There was no evidence of neuronal degeneration, astrocytic or microglial activation, or a reduction in the number of tyrosine hydroxylase positive (TH(+)) neurons in the SNpc or neuronal processes in the striatum of DQ-treated mice. These results are consistent with the rapid clearance of DQ from the brain following a single dose of radiolabeled DQ. In contrast, MPTP-treated mice exhibited decreased striatal dopamine concentration, reduced numbers of TH(+) neurons in the SNpc, and neuropathological changes, including neuronal necrosis, as well as astrocytic and microglial activation in the striatum and SNpc.
Collapse
|
20
|
Gubellini P, Kachidian P. Animal models of Parkinson's disease: An updated overview. Rev Neurol (Paris) 2015; 171:750-61. [PMID: 26343921 DOI: 10.1016/j.neurol.2015.07.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/20/2015] [Accepted: 07/22/2015] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder whose etiology, besides a minority of genetic cases, is still largely unknown. Animal models have contributed to elucidate PD etiology and pathogenesis, as well as its cellular and molecular mechanisms, leading to the general hypothesis that this neurological disorder is due to complex interactions between environmental and genetic factors. However, the full understanding of PD is still very far from being achieved, and new potential treatments need to be tested to further improve patients' quality of life and, possibly, slow down the neurodegenerative process. In this context, animal models of PD are required to address all these issues. "Classic" models are based on neurotoxins that selectively target catecholaminergic neurons (such as 6-hydroxydopamine, 1-methyl-1,2,3,6-tetrahydropiridine, agricultural pesticides, etc.), while more recent models employ genetic manipulations that either introduce mutations similar to those find in familial cases of PD (α-synuclein, DJ-1, PINK1, Parkin, etc.) or selectively disrupt nigrostriatal neurons (MitoPark, Pitx3, Nurr1, etc.). Each one of these models has its own advantages and limitations, thus some are better suited for studying PD pathogenesis, while others are more pertinent to test therapeutic treatments. Here, we provide a critical and updated review of the most used PD models.
Collapse
Affiliation(s)
- P Gubellini
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille (IBDM) UMR7288, case 907, parc scientifique de Luminy, 163, avenue de Luminy, 13009 Marseille, France
| | - P Kachidian
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille (IBDM) UMR7288, case 907, parc scientifique de Luminy, 163, avenue de Luminy, 13009 Marseille, France.
| |
Collapse
|
21
|
Comparative Proteomic Analysis of Carbonylated Proteins from the Striatum and Cortex of Pesticide-Treated Mice. PARKINSONS DISEASE 2015; 2015:812532. [PMID: 26345149 PMCID: PMC4546751 DOI: 10.1155/2015/812532] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 05/27/2015] [Accepted: 05/30/2015] [Indexed: 01/28/2023]
Abstract
Epidemiological studies indicate exposures to the herbicide paraquat (PQ) and fungicide maneb (MB) are associated with increased risk of Parkinson's disease (PD). Oxidative stress appears to be a premier mechanism that underlies damage to the nigrostriatal dopamine system in PD and pesticide exposure. Enhanced oxidative stress leads to lipid peroxidation and production of reactive aldehydes; therefore, we conducted proteomic analyses to identify carbonylated proteins in the striatum and cortex of pesticide-treated mice in order to elucidate possible mechanisms of toxicity. Male C57BL/6J mice were treated biweekly for 6 weeks with saline, PQ (10 mg/kg), MB (30 mg/kg), or the combination of PQ and MB (PQMB). Treatments resulted in significant behavioral alterations in all treated mice and depleted striatal dopamine in PQMB mice. Distinct differences in 4-hydroxynonenal-modified proteins were observed in the striatum and cortex. Proteomic analyses identified carbonylated proteins and peptides from the cortex and striatum, and pathway analyses revealed significant enrichment in a variety of KEGG pathways. Further analysis showed enrichment in proteins of the actin cytoskeleton in treated samples, but not in saline controls. These data indicate that treatment-related effects on cytoskeletal proteins could alter proper synaptic function, thereby resulting in impaired neuronal function and even neurodegeneration.
Collapse
|
22
|
Renaud J, Nabavi SF, Daglia M, Nabavi SM, Martinoli MG. Epigallocatechin-3-Gallate, a Promising Molecule for Parkinson's Disease? Rejuvenation Res 2015; 18:257-69. [PMID: 25625827 DOI: 10.1089/rej.2014.1639] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, and it is characterized by the loss of the neurotransmitter dopamine and neuronal degeneration in the substantia nigra pars compacta. Thus far, current therapeutic strategies have failed to address neuronal degeneration. It has been reported that overproduction of reactive oxygen species, resulting in oxidative stress, and neuroinflammation play an important role in neurodegenerative diseases through the induction of macromolecular oxidative damage and modulation of intracellular signaling pathways concurring to neuronal cell death. Indeed, anti-oxidant and anti-inflammatory drugs have been the subject of recommendation as a complementary therapy alongside an effective symptomatic treatment to hamper the progression of PD. Today, much attention is paid to polyphenols in light of their potent capacity to reduce oxidative stress and inflammation, while having much fewer side effects than most other drugs. Camellia sinensis L. is the most common ancient herbal tea prepared as a beverage worldwide and it possesses numerous beneficial effects on human health. Epigallocatechin-3-gallate is the best-known bioactive component of C. sinensis and is recognized to exert potent neuroprotective effects against oxidative stress, neuroinflammation, protein aggregation, autophagy, and neuronal cell death in vitro as well as in vivo. The present review appraises the available literature on the beneficial role of epigallocatechin-3-gallate pertaining to dopaminergic degeneration characteristic of PD with particular emphasis on its possible mechanisms of action.
Collapse
Affiliation(s)
- Justine Renaud
- 1 Department of Medical Biology and Research Group in Neuroscience, Université du Québec , Trois-Rivières, Québec, Canada
| | - Seyed Fazel Nabavi
- 2 Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences , Tehran, Iran
| | - Maria Daglia
- 3 Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia , Italy
| | - Seyed Mohammad Nabavi
- 2 Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences , Tehran, Iran
| | - Maria-Grazia Martinoli
- 1 Department of Medical Biology and Research Group in Neuroscience, Université du Québec , Trois-Rivières, Québec, Canada
- 4 Department of Psychiatry and Neuroscience, Université Laval and CHU Research Center , Québec, Canada
| |
Collapse
|
23
|
Huang CL, Chao CC, Lee YC, Lu MK, Cheng JJ, Yang YC, Wang VC, Chang WC, Huang NK. Paraquat Induces Cell Death Through Impairing Mitochondrial Membrane Permeability. Mol Neurobiol 2015; 53:2169-88. [DOI: 10.1007/s12035-015-9198-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 04/22/2015] [Indexed: 12/20/2022]
|
24
|
Abstract
The central nervous system's extrapyramidal system provides involuntary motor control to the muscles of the head, neck, and limbs. Toxicants that affect the extrapyramidal system are generally clinically characterized by impaired motor control, which is usually the result of basal ganglionic dysfunction. A variety of extrapyramidal syndromes are recognized in humans and include Parkinson's disease, secondary parkinsonism, other degenerative diseases of the basal ganglia, and clinical syndromes that result in dystonia, dyskinesia, essential tremor, and other forms of tremor and chorea. This chapter briefly reviews the anatomy of the extrapyramidal system and discusses several naturally occurring and experimental models that target the mammalian (nonhuman) extrapyramidal system. Topics discussed include extrapyramidal syndromes associated with antipsychotic drugs, carbon monoxide, reserpine, cyanide, rotenone, paraquat, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and manganese. In most cases, animals are used as experimental models to improve our understanding of the toxicity and pathogenesis of these agents. Another agent discussed in this chapter, yellowstar thistle poisoning in horses, however, represents an important spontaneous cause of parkinsonism that naturally occurs in animals. The central focus of the chapter is on animal models, especially the concordance between clinical signs, neurochemical changes, and neuropathology between animals and people.
Collapse
Affiliation(s)
- David Dorman
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
25
|
Blesa J, Przedborski S. Parkinson's disease: animal models and dopaminergic cell vulnerability. Front Neuroanat 2014; 8:155. [PMID: 25565980 PMCID: PMC4266040 DOI: 10.3389/fnana.2014.00155] [Citation(s) in RCA: 345] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/27/2014] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that affects about 1.5% of the global population over 65 years of age. A hallmark feature of PD is the degeneration of the dopamine (DA) neurons in the substantia nigra pars compacta (SNc) and the consequent striatal DA deficiency. Yet, the pathogenesis of PD remains unclear. Despite tremendous growth in recent years in our knowledge of the molecular basis of PD and the molecular pathways of cell death, important questions remain, such as: (1) why are SNc cells especially vulnerable; (2) which mechanisms underlie progressive SNc cell loss; and (3) what do Lewy bodies or α-synuclein reveal about disease progression. Understanding the variable vulnerability of the dopaminergic neurons from the midbrain and the mechanisms whereby pathology becomes widespread are some of the primary objectives of research in PD. Animal models are the best tools to study the pathogenesis of PD. The identification of PD-related genes has led to the development of genetic PD models as an alternative to the classical toxin-based ones, but does the dopaminergic neuronal loss in actual animal models adequately recapitulate that of the human disease? The selection of a particular animal model is very important for the specific goals of the different experiments. In this review, we provide a summary of our current knowledge about the different in vivo models of PD that are used in relation to the vulnerability of the dopaminergic neurons in the midbrain in the pathogenesis of PD.
Collapse
Affiliation(s)
- Javier Blesa
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
| | | |
Collapse
|
26
|
Villeneuve LM, Purnell PR, Boska MD, Fox HS. Early Expression of Parkinson's Disease-Related Mitochondrial Abnormalities in PINK1 Knockout Rats. Mol Neurobiol 2014; 53:171-186. [PMID: 25421206 DOI: 10.1007/s12035-014-8927-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/07/2014] [Indexed: 12/21/2022]
Abstract
PTEN-induced kinase 1 (PINK1) mutations are responsible for an autosomal recessive, familial form of Parkinson's disease. PINK1 protein is a Ser/Thr kinase localized to the mitochondrial membrane and is involved in many processes including mitochondrial trafficking, mitophagy, and proteasomal function. Using a new PINK1 knockout (PINK1 KO) rat model, we found altered brain metabolomic markers using magnetic resonance spectroscopy, identified changes in mitochondrial pathways with quantitative proteomics using sequential window acquisition of all theoretical spectra (SWATH) mass spectrometry, and demonstrated mitochondrial functional alterations through measurement of oxygen consumption and acidification rates. The observed alterations included reduced creatine, decreased levels of complex I of the electron transport chain, and increased proton leak in the electron transport chain in PINK1 KO rat brains. In conjunction, these results demonstrate metabolomic and mitochondrial alterations occur during the asymptomatic phase of Parkinson's disease in this model. These results indicate both potential early diagnostic markers and therapeutic pathways that can be used in PD.
Collapse
Affiliation(s)
- Lance M Villeneuve
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985800 Nebraska Medical Center-DRC1 3008, Omaha, NE, 68198-5800, USA
| | - Phillip R Purnell
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985800 Nebraska Medical Center-DRC1 3008, Omaha, NE, 68198-5800, USA
| | - Michael D Boska
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985800 Nebraska Medical Center-DRC1 3008, Omaha, NE, 68198-5800, USA.,Department of Radiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Howard S Fox
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985800 Nebraska Medical Center-DRC1 3008, Omaha, NE, 68198-5800, USA.
| |
Collapse
|
27
|
Jones BC, Huang X, Mailman RB, Lu L, Williams RW. The perplexing paradox of paraquat: the case for host-based susceptibility and postulated neurodegenerative effects. J Biochem Mol Toxicol 2014; 28:191-7. [PMID: 24599642 PMCID: PMC4677573 DOI: 10.1002/jbt.21552] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 01/20/2014] [Accepted: 02/01/2014] [Indexed: 12/13/2022]
Abstract
Paraquat is an herbicide used extensively in agriculture and has also been proposed to be a risk factor for Parkinson's disease. To date, experimental, clinical, and epidemiological data on paraquat neurotoxicity have been equivocal. In this short review, we discuss some technical and biological mechanisms that contribute to inconsistencies regarding paraquat neurotoxicity. We hypothesize that individual genetic variations in susceptibility generate major differences in neurotoxic risk and functional outcome. Identifying these heritable sources of variation in host susceptibility, and their role in complex gene-environment interactions, is crucial to identify risk biomarkers and to devise better prevention and treatment for those exposed to paraquat and other potential neurotoxicants.
Collapse
Affiliation(s)
- Byron C Jones
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, USA; Department of Pharmacology, The Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA, USA.
| | | | | | | | | |
Collapse
|
28
|
Minnema DJ, Travis KZ, Breckenridge CB, Sturgess NC, Butt M, Wolf JC, Zadory D, Beck MJ, Mathews JM, Tisdel MO, Cook AR, Botham PA, Smith LL. Dietary administration of paraquat for 13 weeks does not result in a loss of dopaminergic neurons in the substantia nigra of C57BL/6J mice. Regul Toxicol Pharmacol 2014; 68:250-8. [PMID: 24389362 DOI: 10.1016/j.yrtph.2013.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 12/23/2013] [Accepted: 12/25/2013] [Indexed: 10/25/2022]
Abstract
Several investigations have reported that mice administered paraquat dichloride (PQ·Cl2) by intraperitoneal injection exhibit a loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). In this study, male and female C57BL/6J mice were administered PQ·Cl2 in the diet at concentrations of 0 (control), 10, and 50ppm for a duration of 13weeks. A separate group of mice were administered 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) during week 12 as positive controls to produce a loss of dopaminergic neurons in the SNpc. The comparative effects of PQ and MPTP on the SNpc and/or striatum were assessed using neurochemical, neuropathological, and stereological endpoints. Morphological and stereological assessments were performed by investigators 'blinded' to the origin of the tissue. Neither dose of PQ·Cl2 (10 or 50 ppm in the diet) caused a loss of striatal dopamine or dopamine metabolite concentrations in the brains of mice. Pathological assessments of the SNpc and striatum showed no evidence of neuronal degeneration or astrocytic/microglial activation. Furthermore, the number of tyrosine hydroxylase-positive (TH(+)) neurons in the SNpc was not reduced in PQ-treated mice. In contrast, MPTP caused a decrease in striatal dopamine concentration, a reduction in TH(+) neurons in the SNpc, and significant pathological changes including astrocytic and microglial activation in the striatum and SNpc. The MPTP-induced effects were greater in males than in females. It is concluded that 13weeks of continuous dietary exposure of C57BL/6J mice to 50ppm PQ·Cl2 (equivalent to 10.2 and 15.6mg PQ ion/kg body weight/day for males and females, respectively) does not result in the loss of, or damage to, dopaminergic neurons in the SNpc.
Collapse
Affiliation(s)
- Daniel J Minnema
- Syngenta Crop Protection, LLC, P.O. Box 18300, Greensboro, NC 27419-8300, USA.
| | - Kim Z Travis
- Syngenta Limited, Jealott's Hill International Research Centre, Bracknell, Berkshire RG42 6EY, UK
| | | | - Nicholas C Sturgess
- Syngenta Limited, Jealott's Hill International Research Centre, Bracknell, Berkshire RG42 6EY, UK
| | - Mark Butt
- Tox Path Specialists, LLC, 8420 Gas House Pike, Frederick, MD 21701-2607, USA
| | - Jeffrey C Wolf
- Experimental Pathology Laboratories, Inc., 45600 Terminal Drive, Sterling, VA 20166, USA
| | - Dan Zadory
- Experimental Pathology Laboratories, Inc., 45600 Terminal Drive, Sterling, VA 20166, USA
| | | | - James M Mathews
- RTI International, 3040 Cornwallis Road, Research Triangle Park, NC 27709-2194, USA
| | - Merrill O Tisdel
- Syngenta Crop Protection, LLC, P.O. Box 18300, Greensboro, NC 27419-8300, USA
| | - Andrew R Cook
- Syngenta Limited, Jealott's Hill International Research Centre, Bracknell, Berkshire RG42 6EY, UK
| | - Philip A Botham
- Syngenta Limited, Jealott's Hill International Research Centre, Bracknell, Berkshire RG42 6EY, UK
| | - Lewis L Smith
- University of Leicester, University Road, Leicester, LE1 7RH, UK
| |
Collapse
|
29
|
Poliquin PO, Chen J, Cloutier M, Trudeau LÉ, Jolicoeur M. Metabolomics and in-silico analysis reveal critical energy deregulations in animal models of Parkinson's disease. PLoS One 2013; 8:e69146. [PMID: 23935941 PMCID: PMC3720533 DOI: 10.1371/journal.pone.0069146] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Accepted: 06/04/2013] [Indexed: 11/18/2022] Open
Abstract
Parkinson's disease (PD) is a multifactorial disease known to result from a variety of factors. Although age is the principal risk factor, other etiological mechanisms have been identified, including gene mutations and exposure to toxins. Deregulation of energy metabolism, mostly through the loss of complex I efficiency, is involved in disease progression in both the genetic and sporadic forms of the disease. In this study, we investigated energy deregulation in the cerebral tissue of animal models (genetic and toxin induced) of PD using an approach that combines metabolomics and mathematical modelling. In a first step, quantitative measurements of energy-related metabolites in mouse brain slices revealed most affected pathways. A genetic model of PD, the Park2 knockout, was compared to the effect of CCCP, a mitochondrial uncoupler [corrected]. Model simulated and experimental results revealed a significant and sustained decrease in ATP after CCCP exposure, but not in the genetic mice model. In support to data analysis, a mathematical model of the relevant metabolic pathways was developed and calibrated onto experimental data. In this work, we show that a short-term stress response in nucleotide scavenging is most probably induced by the toxin exposure. In turn, the robustness of energy-related pathways in the model explains how genetic perturbations, at least in young animals, are not sufficient to induce significant changes at the metabolite level.
Collapse
Affiliation(s)
- Pierre O. Poliquin
- Department of Chemical Engineering, École Polytechnique de Montréal, Montréal, Quebec, Canada
| | - Jingkui Chen
- Department of Chemical Engineering, École Polytechnique de Montréal, Montréal, Quebec, Canada
| | - Mathieu Cloutier
- GERAD and Department of Chemical Engineering, École Polytechnique de Montréal, Montréal, Quebec, Canada
| | - Louis-Éric Trudeau
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Mario Jolicoeur
- Department of Chemical Engineering, École Polytechnique de Montréal, Montréal, Quebec, Canada
- * E-mail:
| |
Collapse
|
30
|
Breckenridge CB, Sturgess NC, Butt M, Wolf JC, Zadory D, Beck M, Mathews JM, Tisdel MO, Minnema D, Travis KZ, Cook AR, Botham PA, Smith LL. Pharmacokinetic, neurochemical, stereological and neuropathological studies on the potential effects of paraquat in the substantia nigra pars compacta and striatum of male C57BL/6J mice. Neurotoxicology 2013; 37:1-14. [PMID: 23523781 DOI: 10.1016/j.neuro.2013.03.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 03/12/2013] [Accepted: 03/12/2013] [Indexed: 11/19/2022]
Abstract
The pharmacokinetics and neurotoxicity of paraquat dichloride (PQ) were assessed following once weekly administration to C57BL/6J male mice by intraperitoneal injection for 1, 2 or 3 weeks at doses of 10, 15 or 25 mg/kg/week. Approximately 0.3% of the administered dose was taken up by the brain and was slowly eliminated, with a half-life of approximately 3 weeks. PQ did not alter the concentration of dopamine (DA), homovanillic acid (HVA) or 3,4-dihydroxyphenylacetic acid (DOPAC), or increase dopamine turnover in the striatum. There was inconsistent stereological evidence of a loss of DA neurons, as identified by chromogenic or fluorescent-tagged antibodies to tyrosine hydroxylase in the substantia nigra pars compacta (SNpc). There was no evidence that PQ induced neuronal degeneration in the SNpc or degenerating neuronal processes in the striatum, as indicated by the absence of uptake of silver stain or reduced immunolabeling of tyrosine-hydroxylase-positive (TH(+)) neurons. There was no evidence of apoptotic cell death, which was evaluated using TUNEL or caspase 3 assays. Microglia (IBA-1 immunoreactivity) and astrocytes (GFAP immunoreactivity) were not activated in PQ-treated mice 4, 8, 16, 24, 48, 96 or 168 h after 1, 2 or 3 doses of PQ. In contrast, mice dosed with the positive control substance, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP; 10mg/kg/dose×4 doses, 2 h apart), displayed significantly reduced DA and DOPAC concentrations and increased DA turnover in the striatum 7 days after dosing. The number of TH(+) neurons in the SNpc was reduced, and there were increased numbers of degenerating neurons and neuronal processes in the SNpc and striatum. MPTP-mediated cell death was not attributed to apoptosis. MPTP activated microglia and astrocytes within 4 h of the last dose, reaching a peak within 48 h. The microglial response ended by 96 h in the SNpc, but the astrocytic response continued through 168 h in the striatum. These results bring into question previous published stereological studies that report loss of TH(+) neurons in the SNpc of PQ-treated mice. This study also suggests that even if the reduction in TH(+) neurons reported by others occurs in PQ-treated mice, this apparent phenotypic change is unaccompanied by neuronal cell death or by modification of dopamine levels in the striatum.
Collapse
|
31
|
Neurotoxin-based models of Parkinson's disease. Neuroscience 2012; 211:51-76. [DOI: 10.1016/j.neuroscience.2011.10.057] [Citation(s) in RCA: 360] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 10/27/2011] [Accepted: 10/28/2011] [Indexed: 12/21/2022]
|
32
|
Duty S, Jenner P. Animal models of Parkinson's disease: a source of novel treatments and clues to the cause of the disease. Br J Pharmacol 2012; 164:1357-91. [PMID: 21486284 DOI: 10.1111/j.1476-5381.2011.01426.x] [Citation(s) in RCA: 510] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Animal models of Parkinson's disease (PD) have proved highly effective in the discovery of novel treatments for motor symptoms of PD and in the search for clues to the underlying cause of the illness. Models based on specific pathogenic mechanisms may subsequently lead to the development of neuroprotective agents for PD that stop or slow disease progression. The array of available rodent models is large and ranges from acute pharmacological models, such as the reserpine- or haloperidol-treated rats that display one or more parkinsonian signs, to models exhibiting destruction of the dopaminergic nigro-striatal pathway, such as the classical 6-hydroxydopamine (6-OHDA) rat and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse models. All of these have provided test beds in which new molecules for treating the motor symptoms of PD can be assessed. In addition, the emergence of abnormal involuntary movements (AIMs) with repeated treatment of 6-OHDA-lesioned rats with L-DOPA has allowed for examination of the mechanisms responsible for treatment-related dyskinesia in PD, and the detection of molecules able to prevent or reverse their appearance. Other toxin-based models of nigro-striatal tract degeneration include the systemic administration of the pesticides rotenone and paraquat, but whilst providing clues to disease pathogenesis, these are not so commonly used for drug development. The MPTP-treated primate model of PD, which closely mimics the clinical features of PD and in which all currently used anti-parkinsonian medications have been shown to be effective, is undoubtedly the most clinically-relevant of all available models. The MPTP-treated primate develops clear dyskinesia when repeatedly exposed to L-DOPA, and these parkinsonian animals have shown responses to novel dopaminergic agents that are highly predictive of their effect in man. Whether non-dopaminergic drugs show the same degree of predictability of response is a matter of debate. As our understanding of the pathogenesis of PD has improved, so new rodent models produced by agents mimicking these mechanisms, including proteasome inhibitors such as PSI, lactacystin and epoximycin or inflammogens like lipopolysaccharide (LPS) have been developed. A further generation of models aimed at mimicking the genetic causes of PD has also sprung up. Whilst these newer models have provided further clues to the disease pathology, they have so far been less commonly used for drug development. There is little doubt that the availability of experimental animal models of PD has dramatically altered dopaminergic drug treatment of the illness and the prevention and reversal of drug-related side effects that emerge with disease progression and chronic medication. However, so far, we have made little progress in moving into other pharmacological areas for the treatment of PD, and we have not developed models that reflect the progressive nature of the illness and its complexity in terms of the extent of pathology and biochemical change. Only when this occurs are we likely to make progress in developing agents to stop or slow the disease progression. The overarching question that draws all of these models together in the quest for better drug treatments for PD is how well do they recapitulate the human condition and how predictive are they of successful translation of drugs into the clinic? This article aims to clarify the current position and highlight the strengths and weaknesses of available models.
Collapse
Affiliation(s)
- Susan Duty
- King's College London, Wolfson Centre for Age-Related Disease, London, UK.
| | | |
Collapse
|
33
|
Huang CL, Lee YC, Yang YC, Kuo TY, Huang NK. Minocycline prevents paraquat-induced cell death through attenuating endoplasmic reticulum stress and mitochondrial dysfunction. Toxicol Lett 2012; 209:203-10. [PMID: 22245251 DOI: 10.1016/j.toxlet.2011.12.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Revised: 12/19/2011] [Accepted: 12/21/2011] [Indexed: 12/31/2022]
Abstract
Paraquat (PQ) was demonstrated to induce dopaminergic neuron death and is used as a Parkinson's disease (PD) mimetic; however, its mechanism remains contradictory. Alternatively, minocycline is a second-generation tetracycline and is undergoing clinical trials for treating PD with an unresolved mechanism. We thus investigated the molecular mechanism of minocycline in preventing PQ-induced cytotoxicity. In this study, minocycline was effective in preventing PQ-induced apoptotic cell death, which involves the cleavages of poly (ADP-ribose) polymerase (PARP) and caspase 3 and increased fluorescence intensity of annexin V-FITC. In addition, PQ also quickly induced alterations of unfolded protein responses (UPRs) and subsequently dysfunction of the mitochondria (such as the decrease in membrane potential and increase in membrane permeability and superoxide formation). Finally, the mechanism of minocycline in preventing PQ-induced apoptosis might be mediated by attenuating endoplasmic reticulum (ER) stress and mitochondrial dysfunction, which respectively results in caspase-12 activation and the release of H2O2, HtrA2/Omi, and Smac/Diablo. Thus, minocycline could possibly be used to treat other neurodegenerative disorders with similar pathologic mechanisms.
Collapse
Affiliation(s)
- Chuen-Lin Huang
- Medical Research Center, Cardinal Tien Hospital, Hsintien, New Taipei City, Taiwan, ROC
| | | | | | | | | |
Collapse
|
34
|
Moretto A, Colosio C. Biochemical and toxicological evidence of neurological effects of pesticides: The example of Parkinson's disease. Neurotoxicology 2011; 32:383-91. [DOI: 10.1016/j.neuro.2011.03.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 03/03/2011] [Indexed: 12/21/2022]
|
35
|
Tanner CM, Kamel F, Ross GW, Hoppin JA, Goldman SM, Korell M, Marras C, Bhudhikanok GS, Kasten M, Chade AR, Comyns K, Richards MB, Meng C, Priestley B, Fernandez HH, Cambi F, Umbach DM, Blair A, Sandler DP, Langston JW. Rotenone, paraquat, and Parkinson's disease. ENVIRONMENTAL HEALTH PERSPECTIVES 2011; 119:866-72. [PMID: 21269927 PMCID: PMC3114824 DOI: 10.1289/ehp.1002839] [Citation(s) in RCA: 898] [Impact Index Per Article: 69.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2010] [Accepted: 01/26/2011] [Indexed: 05/18/2023]
Abstract
BACKGROUND Mitochondrial dysfunction and oxidative stress are pathophysiologic mechanisms implicated in experimental models and genetic forms of Parkinson's disease (PD). Certain pesticides may affect these mechanisms, but no pesticide has been definitively associated with PD in humans. OBJECTIVES Our goal was to determine whether pesticides that cause mitochondrial dysfunction or oxidative stress are associated with PD or clinical features of parkinsonism in humans. METHODS We assessed lifetime use of pesticides selected by mechanism in a case-control study nested in the Agricultural Health Study (AHS). PD was diagnosed by movement disorders specialists. Controls were a stratified random sample of all AHS participants frequency-matched to cases by age, sex, and state at approximately three controls:one case. RESULTS In 110 PD cases and 358 controls, PD was associated with use of a group of pesticides that inhibit mitochondrial complex I [odds ratio (OR)=1.7; 95% confidence interval (CI), 1.0-2.8] including rotenone (OR=2.5; 95% CI, 1.3-4.7) and with use of a group of pesticides that cause oxidative stress (OR = 2.0; 95% CI, 1.2-3.6), including paraquat (OR=2.5; 95% CI, 1.4-4.7). CONCLUSIONS PD was positively associated with two groups of pesticides defined by mechanisms implicated experimentally-those that impair mitochondrial function and those that increase oxidative stress-supporting a role for these mechanisms in PD pathophysiology.
Collapse
|
36
|
Bartlett RM, Murali D, Nickles RJ, Barnhart TE, Holden JE, DeJesus OT. Assessment of fetal brain uptake of paraquat in utero using in vivo PET/CT imaging. Toxicol Sci 2011; 122:551-6. [PMID: 21546347 DOI: 10.1093/toxsci/kfr104] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Prenatal in utero conditions are thought to play a role in the development of adult diseases including Parkinson's disease (PD). Paraquat is a common herbicide with chemical structure similar to 1-methyl-4-phenyl 1,2,3,6-tetrahydropyridine, a neurotoxin known to induce parkinsonism. In order to assess the role of in utero paraquat exposure in PD, uptake in maternal and fetal brains were measured using positron emission tomography (PET)/computed tomography (CT) imaging. Two anesthetized pregnant rhesus macaques in the late second trimester of pregnancy were given bolus iv injections of ¹¹C-paraquat, and whole-body PET/CT imaging was performed. Using maternal ventricular blood pool as the input function, the unidirectional influx rate constants (K(i)s), a measure of the irreversible transport of paraquat from plasma to brain, were calculated for the maternal and fetal brains using Patlak graphical analysis. Results indicate minimal uptake of paraquat by both maternal and fetal brains with average K(i)s of 0.0009 and 0.0016 per minute, respectively. The highest regional cerebral uptake in the maternal brain (0.0009% injected dose) was seen in the pineal gland, a structure known to lack a blood brain barrier. The finding of minimal paraquat uptake in maternal and fetal brains is similar to previous findings in adult male macaques and extends the contention that a single acute paraquat exposure, prenatally or postnatally, is unlikely to play a role in PD.
Collapse
Affiliation(s)
- Rachel M Bartlett
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | | | | | | | | | |
Collapse
|
37
|
Defranchi E, Novellino A, Whelan M, Vogel S, Ramirez T, van Ravenzwaay B, Landsiedel R. Feasibility Assessment of Micro-Electrode Chip Assay as a Method of Detecting Neurotoxicity in vitro. FRONTIERS IN NEUROENGINEERING 2011; 4:6. [PMID: 21577249 PMCID: PMC3088865 DOI: 10.3389/fneng.2011.00006] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Accepted: 04/03/2011] [Indexed: 11/18/2022]
Abstract
Detection and characterization of chemically induced toxic effects in the nervous system represent a challenge for the hazard assessment of chemicals. In vivo, neurotoxicological assessments exploit the fact that the activity of neurons in the central and peripheral nervous system has functional consequences. And so far, no in vitro method for evaluating the neurotoxic hazard has yet been validated and accepted for regulatory purpose. The micro-electrode array (MEA) assay consists of a culture chamber into which an integrated array of micro-electrodes is capable of measuring extracellular electrophysiology (spikes and bursts) from electro-active tissues. A wide variety of electrically excitable biological tissues may be placed onto the chips including primary cultures of nervous system tissue. Recordings from this type of in vitro cultured system are non-invasive, give label free evaluations and provide a higher throughput than conventional electrophysiological techniques. In this paper, 20 substances were tested in a blinded study for their toxicity and dose-response curves were obtained from fetal rat cortical neuronal networks coupled to MEAs. The experimental procedure consisted of evaluating the firing activity (spiking rate) and modification/reduction in response to chemical administration. Native/reference activity, 30 min of activity recording per dilution, plus the recovery points (after 24 h) were recorded. The preliminary data, using a set of chemicals with different mode-of-actions (13 known to be neurotoxic, 2 non-neuroactive and not toxic, and 5 non-neuroactive but toxic) show good predictivity (sensitivity: 0.77; specificity: 0.86; accuracy: 0.85). Thus, the MEA with a neuronal network has the potency to become an effective tool to evaluate the neurotoxicity of substances in vitro.
Collapse
Affiliation(s)
| | | | - Maurice Whelan
- Systems Toxicology Unit, Institute for Health and Consumer Protection, Joint Research CentreIspra, Varese, Italy
| | - Sandra Vogel
- Badische Anilin- und Soda-Fabrik Societas EuropaeaLudwigshafen, Germany
| | - Tzutzuy Ramirez
- Badische Anilin- und Soda-Fabrik Societas EuropaeaLudwigshafen, Germany
| | | | - Robert Landsiedel
- Badische Anilin- und Soda-Fabrik Societas EuropaeaLudwigshafen, Germany
| |
Collapse
|
38
|
Franco R, Panayiotidis MI. Cell death or survival: The double-edged sword of environmental and occupational toxicity. Chem Biol Interact 2010; 188:265-6. [PMID: 20553883 PMCID: PMC2943050 DOI: 10.1016/j.cbi.2010.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Rodrigo Franco
- Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences. University of Nebraska-Lincoln. Lincoln, NE 68583
| | - Mihalis I. Panayiotidis
- Department of Pathology, Medical School, University of Ioannina, University Campus 45110, Ioannina, Greece
- School of Community Health Sciences, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
39
|
Franco R, Li S, Rodriguez-Rocha H, Burns M, Panayiotidis MI. Molecular mechanisms of pesticide-induced neurotoxicity: Relevance to Parkinson's disease. Chem Biol Interact 2010; 188:289-300. [PMID: 20542017 PMCID: PMC2942983 DOI: 10.1016/j.cbi.2010.06.003] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 05/27/2010] [Accepted: 06/03/2010] [Indexed: 11/20/2022]
Abstract
Pesticides are widely used in agricultural and other settings, resulting in continued human exposure. Pesticide toxicity has been clearly demonstrated to alter a variety of neurological functions. Particularly, there is strong evidence suggesting that pesticide exposure predisposes to neurodegenerative diseases. Epidemiological data have suggested a relationship between pesticide exposure and brain neurodegeneration. However, an increasing debate has aroused regarding this issue. Paraquat is a highly toxic quaternary nitrogen herbicide which has been largely studied as a model for Parkinson's disease providing valuable insight into the molecular mechanisms involved in the toxic effects of pesticides and their role in the progression of neurodegenerative diseases. In this work, we review the molecular mechanisms involved in the neurotoxic action of pesticides, with emphasis on the mechanisms associated with the induction of neuronal cell death by paraquat as a model for Parkinsonian neurodegeneration.
Collapse
Affiliation(s)
- Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, 68583, United States.
| | | | | | | | | |
Collapse
|
40
|
González-Hernández T, Cruz-Muros I, Afonso-Oramas D, Salas-Hernandez J, Castro-Hernandez J. Vulnerability of mesostriatal dopaminergic neurons in Parkinson's disease. Front Neuroanat 2010; 4:140. [PMID: 21079748 PMCID: PMC2978035 DOI: 10.3389/fnana.2010.00140] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 09/24/2010] [Indexed: 12/21/2022] Open
Abstract
The term vulnerability was first associated with the midbrain dopaminergic neurons 85 years ago, before they were identified as monoaminergic neurons, when Foix and Nicolesco (1925) reported the loss of neuromelanin containing neurons in the midbrain of patients with post-encephalitic Parkinson's disease (PD). A few years later, Hassler (1938) showed that degeneration is more intense in the ventral tier of the substantia nigra compacta than in its dorsal tier and the ventral tegmental area (VTA), outlining the concept of differential vulnerability of midbrain dopaminergic (DA-) neurons. Nowadays, we know that other neuronal groups degenerate in PD, but the massive loss of nigral DA-cells is its pathological hallmark, having a pivotal position in the pathophysiology of the disease as it is responsible for the motor symptoms. Data from humans as well as cellular and animal models indicate that DA-cell degeneration is a complex process, probably precipitated by the convergence of different risk factors, mediated by oxidative stress, and involving pathogenic factors arising within the DA-neuron (intrinsic factors), and from its environment and distant interconnected brain regions (extrinsic factors). In light of current data, intrinsic factors seem to be preferentially involved in the first steps of the degenerative process, and extrinsic factors in its progression. A controversial issue is the relative weight of the impairment of common cell functions, such as energy metabolism and proteostasis, and specific dopaminergic functions, such as pacemaking activity and DA handling, in the pathogenesis of DA-cell degeneration. Here we will review the current knowledge about the relevance of these factors at the beginning and during the progression of PD, and in the differential vulnerability of midbrain DA-cells.
Collapse
|
41
|
Parkinson's disease: is it a toxic syndrome? Neurol Res Int 2010; 2010:103094. [PMID: 21152209 PMCID: PMC2989867 DOI: 10.1155/2010/103094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 06/05/2010] [Accepted: 07/26/2010] [Indexed: 02/05/2023] Open
Abstract
Parkinson's disease (PD) is one of the neurodegenerative diseases which we can by certainty identify its pathology, however, this confidence disappeares when talking about the cause. A long history of trials, suggestions, and theories tried linking PD to a specific causation. In this paper, a new suggestion is trying to find its way, could it be toxicology? Can we—in the future—look to PD as an occupational disease, in fact, many clues point to the possible toxic responsibility—either total or partial—in causing this disease. Searching for possible toxic causes for PD would help in designing perfect toxic models in animals.
Collapse
|
42
|
Glycogen synthase kinase 3β and its phosphorylated form (Y216) in the paraquat-induced model of parkinsonism. Neurotox Res 2010; 19:162-71. [PMID: 20143200 DOI: 10.1007/s12640-010-9153-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 01/08/2010] [Accepted: 01/18/2010] [Indexed: 12/21/2022]
Abstract
Parkinson's disease is a slowly progressing disease, due to a lesion of dopaminergic neurons in the substantia nigra and a dramatic loss of dopamine in the striatum. It is now accepted that several environmental agents including the herbicide paraquat (PQ) may contribute to its pathogenesis. However, till now nothing is known about the role of glycogen synthase kinase-3β (GSK-3β) in the PQ toxicity. Therefore, the aim of this study was to examine the influence of 37-week administration of PQ in rats on the immunohistochemically measured levels of the total GSK-3β and its active, tyrosine 216 (pY216)-phosphorylated form in subcellular fractions of the midbrain with pons, as well as of the striatum. The present results revealed that the long-term PQ administration increased the levels of total and active forms of GSK-3β in the midbrain with pons, whereas decreased them in the striatum. Examination of the lesion extent showed a decrease in the number of tyrosine-immunoreactive neurons in the substantia nigra pars compacta, ventral tegmental area, and locus coeruleus, as well as lower DOPAC/dopamine ratio and noradrenaline level in the striatum in rats treated with PQ. The long-term PQ administration disturbed also motor activity of rats. Summarizing, the present data indicate that the long-term exposure of rats to PQ, a commonly used herbicide, diversely alters levels of GSK-3β in different brain structures, which may be associated with their vulnerability to its toxicity.
Collapse
|
43
|
Bartlett RM, Holden JE, Nickles RJ, Murali D, Barbee DL, Barnhart TE, Christian BT, DeJesus OT. Paraquat is excluded by the blood brain barrier in rhesus macaque: An in vivo pet study. Brain Res 2008; 1259:74-9. [PMID: 19135428 DOI: 10.1016/j.brainres.2008.12.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 12/08/2008] [Accepted: 12/09/2008] [Indexed: 10/21/2022]
Abstract
Environmental factors have long been thought to have a role in the etiology of idiopathic Parkinson's disease (PD). Since the discovery of the selective neurotoxicity of MPTP to dopamine cells, suspicion has focused on paraquat, a common herbicide with chemical structure similar to 1-methyl-4-phenylpyridinium (MPP+), the MPTP metabolite responsible for its neurotoxicity. Although in vitro evidence for paraquat neurotoxicity to dopamine cells is well established, its in vivo effects have been ambiguous because paraquat is di-cationic in plasma, which raises questions about its ability to cross the blood brain barrier. This study assessed the brain uptake of [(11)C]-paraquat in adult male rhesus macaques using quantitative PET imaging. Results showed minimal uptake of [(11)C]-paraquat in the macaque brain. The highest concentrations of paraquat were seen in the pineal gland and the lateral ventricles. Global brain concentrations including those in known dopamine areas were consistent with the blood volume in those structures. This acute exposure study found that paraquat is excluded from the brain by the blood brain barrier and thus does not readily support the causative role of paraquat exposure in idiopathic Parkinson's disease.
Collapse
Affiliation(s)
- Rachel M Bartlett
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, 1005 Wisconsin Institutes of Medical Research, 1111 Highland Avenue, Madison, WI 53705, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Miller DB, O'Callaghan JP. Do early-life insults contribute to the late-life development of Parkinson and Alzheimer diseases? Metabolism 2008; 57 Suppl 2:S44-9. [PMID: 18803966 DOI: 10.1016/j.metabol.2008.07.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
How early-life events "set the stage" for adult disease has emerged as a research focus. Historically, the epidemiology of disease risk factors has centered on adult life, with little scrutiny of early-life events. Here we review the concept that events in early life may contribute to late-life neurodegenerative disease development, with a focus on Parkinson disease (PD) and Alzheimer disease (AD). Suspect events in early life include infections, stress, poor nutrition, and environmental factors such as chemical and pesticide exposure. Adiposity appears to contribute to both PD and AD; and because early-life events contribute to the development of obesity, linkages may exist between early determinants of obesity and the subsequent development of these neurologic diseases. Many now suggest a life-course approach for determining the relative contributions of genetic and environmental factors in any chronic disease. This requires determining when during the life course that a given exposure has its greatest effect and how exposures may accumulate over the life span. The data for PD and AD suggest that a number of insults occurring early in life may lead or contribute to these diseases. More definitive knowledge of the key risk factors involved will be needed to implement intervention and preventative strategies early in life to dampen or prevent any adverse late-life outcomes.
Collapse
Affiliation(s)
- Diane B Miller
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505, USA.
| | | |
Collapse
|
45
|
Jones DC, Miller GW. The effects of environmental neurotoxicants on the dopaminergic system: A possible role in drug addiction. Biochem Pharmacol 2008; 76:569-81. [DOI: 10.1016/j.bcp.2008.05.010] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Revised: 05/01/2008] [Accepted: 05/05/2008] [Indexed: 11/29/2022]
|
46
|
|
47
|
|