1
|
da Silva S, da Costa CDL, Naime AA, Santos D, Farina M, Colle D. Mechanisms Mediating the Combined Toxicity of Paraquat and Maneb in SH-SY5Y Neuroblastoma Cells. Chem Res Toxicol 2024; 37:1269-1282. [PMID: 39058280 PMCID: PMC11337211 DOI: 10.1021/acs.chemrestox.3c00389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
Epidemiological and experimental studies have demonstrated that combined exposure to the pesticides paraquat (PQ) and maneb (MB) increases the risk of developing Parkinson's disease. However, the mechanisms mediating the toxicity induced by combined exposure to these pesticides are not well understood. The aim of this study was to investigate the mechanism(s) of neurotoxicity induced by exposure to the pesticides PQ and MB isolated or in association (PQ + MB) in SH-SY5Y neuroblastoma cells. PQ + MB exposure for 24 and 48 h decreased cell viability and disrupted cell membrane integrity. In addition, PQ + MB exposure for 12 h decreased the mitochondrial membrane potential. PQ alone increased reactive oxygen species (ROS) and superoxide anion generation and decreased the activity of mitochondrial complexes I and II at 12 h of exposure. MB alone increased ROS generation and depleted intracellular glutathione (GSH) within 6 h of exposure. In contrast, MB exposure for 12 h increased the GSH levels, the glutamate cysteine ligase (GCL, the rate-limiting enzyme in the GSH synthesis pathway) activity, and increased nuclear Nrf2 staining. Pretreatment with buthionine sulfoximine (BSO, a GCL inhibitor) abolished the MB-mediated GSH increase, indicating that MB increases GSH synthesis by upregulating GCL, probably by the activation of the Nrf2/ARE pathway. BSO pretreatment, which did not modify cell viability per se, rendered cells more sensitive to MB-induced toxicity. In contrast, treatment with the antioxidant N-acetylcysteine protected cells from MB-induced toxicity. These findings show that the combined exposure of SH-SY5Y cells to PQ and MB induced a cytotoxic effect higher than that observed when cells were subjected to individual exposures. Such a higher effect seems to be related to additive toxic events resulting from PQ and MB exposures. Thus, our study contributes to a better understanding of the toxicity of PQ and MB in combined exposures.
Collapse
Affiliation(s)
- Suzana da Silva
- Department
of Clinical Analyses, Federal University
of Santa Catarina, Florianopolis 88040-900 Santa Catarina, Brazil
| | - Carolina de Lima da Costa
- Department
of Clinical Analyses, Federal University
of Santa Catarina, Florianopolis 88040-900 Santa Catarina, Brazil
| | - Aline Aita Naime
- Department
of Biochemistry, Federal University of Santa
Catarina, Florianopolis 88040-900 Santa Catarina, Brazil
| | - Danúbia
Bonfanti Santos
- Department
of Biochemistry, Federal University of Santa
Catarina, Florianopolis 88040-900 Santa Catarina, Brazil
| | - Marcelo Farina
- Department
of Biochemistry, Federal University of Santa
Catarina, Florianopolis 88040-900 Santa Catarina, Brazil
| | - Dirleise Colle
- Department
of Clinical Analyses, Federal University
of Santa Catarina, Florianopolis 88040-900 Santa Catarina, Brazil
| |
Collapse
|
2
|
Ilieva NM, Hoffman EK, Ghalib MA, Greenamyre JT, De Miranda BR. LRRK2 kinase inhibition protects against Parkinson's disease-associated environmental toxicants. Neurobiol Dis 2024; 196:106522. [PMID: 38705492 PMCID: PMC11332574 DOI: 10.1016/j.nbd.2024.106522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024] Open
Abstract
Idiopathic Parkinson's disease (PD) is epidemiologically linked with exposure to toxicants such as pesticides and solvents, which comprise a wide array of chemicals that pollute our environment. While most are structurally distinct, a common cellular target for their toxicity is mitochondrial dysfunction, a key pathological trigger involved in the selective vulnerability of dopaminergic neurons. We and others have shown that environmental mitochondrial toxicants such as the pesticides rotenone and paraquat, and the organic solvent trichloroethylene (TCE) appear to be influenced by the protein LRRK2, a genetic risk factor for PD. As LRRK2 mediates vesicular trafficking and influences endolysosomal function, we postulated that LRRK2 kinase activity may inhibit the autophagic removal of toxicant damaged mitochondria, resulting in elevated oxidative stress. Conversely, we suspected that inhibition of LRRK2, which has been shown to be protective against dopaminergic neurodegeneration caused by mitochondrial toxicants, would reduce the intracellular production of reactive oxygen species (ROS) and prevent mitochondrial toxicity from inducing cell death. To do this, we tested in vitro if genetic or pharmacologic inhibition of LRRK2 (MLi2) protected against ROS caused by four toxicants associated with PD risk - rotenone, paraquat, TCE, and tetrachloroethylene (PERC). In parallel, we assessed if LRRK2 inhibition with MLi2 could protect against TCE-induced toxicity in vivo, in a follow up study from our observation that TCE elevated LRRK2 kinase activity in the nigrostriatal tract of rats prior to dopaminergic neurodegeneration. We found that LRRK2 inhibition blocked toxicant-induced ROS and promoted mitophagy in vitro, and protected against dopaminergic neurodegeneration, neuroinflammation, and mitochondrial damage caused by TCE in vivo. We also found that cells with the LRRK2 G2019S mutation displayed exacerbated levels of toxicant induced ROS, but this was ameliorated by LRRK2 inhibition with MLi2. Collectively, these data support a role for LRRK2 in toxicant-induced mitochondrial dysfunction linked to PD risk through oxidative stress and the autophagic removal of damaged mitochondria.
Collapse
Affiliation(s)
- Neda M Ilieva
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eric K Hoffman
- Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mohammed A Ghalib
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Briana R De Miranda
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
3
|
Soares ÉN, Costa ACDS, Ferrolho GDJ, Ureshino RP, Getachew B, Costa SL, da Silva VDA, Tizabi Y. Nicotinic Acetylcholine Receptors in Glial Cells as Molecular Target for Parkinson's Disease. Cells 2024; 13:474. [PMID: 38534318 PMCID: PMC10969434 DOI: 10.3390/cells13060474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease characterized by resting tremor, bradykinesia, rigidity, and postural instability that also includes non-motor symptoms such as mood dysregulation. Dopamine (DA) is the primary neurotransmitter involved in this disease, but cholinergic imbalance has also been implicated. Current intervention in PD is focused on replenishing central DA, which provides remarkable temporary symptomatic relief but does not address neuronal loss and the progression of the disease. It has been well established that neuronal nicotinic cholinergic receptors (nAChRs) can regulate DA release and that nicotine itself may have neuroprotective effects. Recent studies identified nAChRs in nonneuronal cell types, including glial cells, where they may regulate inflammatory responses. Given the crucial role of neuroinflammation in dopaminergic degeneration and the involvement of microglia and astrocytes in this response, glial nAChRs may provide a novel therapeutic target in the prevention and/or treatment of PD. In this review, following a brief discussion of PD, we focus on the role of glial cells and, specifically, their nAChRs in PD pathology and/or treatment.
Collapse
Affiliation(s)
- Érica Novaes Soares
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
| | - Ana Carla dos Santos Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
| | - Gabriel de Jesus Ferrolho
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
- Laboratory of Neurosciences, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
| | - Rodrigo Portes Ureshino
- Department of Biological Sciences, Universidade Federal de São Paulo, Diadema 09961-400, SP, Brazil
- Laboratory of Molecular and Translational Endocrinology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, SP, Brazil
| | - Bruk Getachew
- Department of Pharmacology, College of Medicine, Howard University, 520 W Street NW, Washington, DC 20059, USA
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
| | - Victor Diogenes Amaral da Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
- Laboratory of Neurosciences, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
| | - Yousef Tizabi
- Department of Pharmacology, College of Medicine, Howard University, 520 W Street NW, Washington, DC 20059, USA
| |
Collapse
|
4
|
Birla D, Kumar R M, Potale Y, Kumar S, Singh G, Kumar A. Drug induced Parkinson’s: A comprehensive review of the issues and measures required to tackle the same. BIO WEB OF CONFERENCES 2024; 86:01028. [DOI: 10.1051/bioconf/20248601028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Drug-inducedParkinsonism(DIP) closelyresemblesParkinson'sdisease(PD)inmotorsymptoms butiscausedbyspecificmedicationsdisruptingdopaminereceptorsandneurotransmitterbalance. PD involves a complex interplay of genetic, environmental, and biochemical factors resulting in the gradual degeneration of dopaminergic neurons. Environmental toxins and genetic mutations, such as LRRK2 and SNCA, contribute to the risk of developing PD. DIP primarily occurs due to the obstruction of dopamine receptors by certain drugs, notably antipsychotics and antiemetics, affecting dopamine transmission and causing Parkinsonian symptoms. Toxin-induced Parkinsonism(TIP)arisesfromexposuretosubstanceslikemanganese,herbicides,pesticides,and specific drugs, disrupting dopaminergic pathways and altering neurotransmission. This study examines various cases of DIP, emphasizing the significance of timely identification and intervention. A thorough understanding and proactive management of DIP are crucial for alleviatingsymptomsandimprovingpatientoutcomes.Healthcareprofessionalsneedtodiligently monitor patients using medications associated with DIP, adjust treatment plans, and educate patientsaboutpotentialsideeffects. Further researchisimperativetounravelthepathophysiology of DIP, considering genetic, environmental, and drug-related factors, to enhance clinical practices and optimize patient care. Addressing DIP requires a multifaceted approach, including early recognition, thoughtful management, and patient-centred care.
Collapse
|
5
|
Jain J, Hasan W, Jat D, Biswas P, Yadav RS. Delayed in sensorimotor reflex ontogeny, slow physical growth, and impairments in behaviour as well as dopaminergic neuronal death in mice offspring following prenatally rotenone administration. Int J Dev Neurosci 2023; 83:518-531. [PMID: 37337287 DOI: 10.1002/jdn.10282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/21/2023] [Accepted: 05/15/2023] [Indexed: 06/21/2023] Open
Abstract
The environment is varying day by day with the introduction of chemicals such as pesticides, most of which have not been effectively studied for their influence on a susceptible group of population involving infants and pregnant females. Rotenone is an organic pesticide used to prepare Parkinson's disease models. A lot of literature is available on the toxicity of rotenone on the adult brain, but to the best of our knowledge, effect of rotenone on prenatally exposed mice has never been investigated yet. Therefore, the recent work aims to evaluate the toxic effect of rotenone on mice, exposed prenatally. We exposed female mice to rotenone at the dose of 5 mg/Kg b.w. throughout the gestational period with oral gavage. We then investigated the effects of rotenone on neonate's central nervous systems as well as on postnatal day (PD) 35 offspring. In the rotenone group, we observed slow physical growth, delays in physical milestones and sensorimotor reflex in neonates and induction of anxiety and impairment in cognitive performances of offspring at PD-35. Additionally, immunohistochemical analysis revealed a marked reduction in TH-positive neurons in substantia nigra. Histological examination of the cerebellum revealed a decrease in Purkinje neurons in the rotenone exposed group as compared to the control. The data from the study showed that prenatally exposure to rotenone affects growth, physical milestones, neuronal population and behaviour of mice when indirectly exposed to the offspring through their mother. This study could provide a great contribution to researchers to find out the molecular mechanism and participating signalling pathway behind these outcomes.
Collapse
Affiliation(s)
- Juli Jain
- Neuroscience Research Lab, School of Biological Sciences, Department of Zoology, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, India
| | - Whidul Hasan
- Neurology Department, Harward Medical School, Harvard Medical School, Boston, USA
| | - Deepali Jat
- Neuroscience Research Lab, School of Biological Sciences, Department of Zoology, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, India
| | - Pronit Biswas
- Department of Life Sciences, Christ (Deemed-to-be University), Bangalore, India
| | - Rajesh Singh Yadav
- School of Forensic Science, National Forensic Sciences University, Bhopal, India
| |
Collapse
|
6
|
Kulikova O, Troshev D, Berezhnoy D, Stvolinsky S, Timoshina Y, Abaimov D, Muzychuk O, Latanov A, Fedorova T. Neuroprotective Efficacy of a Nanomicellar Complex of Carnosine and Lipoic Acid in a Rat Model of Rotenone-Induced Parkinson's Disease. Antioxidants (Basel) 2023; 12:1215. [PMID: 37371945 DOI: 10.3390/antiox12061215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Oxidative stress, accompanied by mitochondrial dysfunction, is a key mechanism involved in the pathogenesis of Parkinson's disease (PD). Both carnosine and lipoic acid are potent antioxidants, the applicability of which in therapy is hindered by their limited bioavailability. This study aimed to evaluate the neuroprotective properties of a nanomicellar complex of carnosine and lipoic acid (CLA) in a rotenone-induced rat model of PD. Parkinsonism was induced via the administration of 2 mg/kg rotenone over the course of 18 days. Two doses of intraperitoneal CLA (25 mg/kg and 50 mg/kg) were administered alongside rotenone to assess its neuroprotective effect. At 25 mg/kg CLA decreased muscle rigidity and partially restored locomotor activity in animals that received rotenone. Furthermore, it caused an overall increase in brain tissue antioxidant activity, accompanied by a 19% increase in neuron density in the substantia nigra and increased dopamine levels in the striatum relative to animals that only received rotenone. Based on the acquired results, it may be concluded that CLA have neuroprotective properties and could potentially be beneficial in PD treatment when used in conjunction with the base therapy.
Collapse
Affiliation(s)
- Olga Kulikova
- Laboratory of Translational and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia
| | - Dmitry Troshev
- Laboratory of Neural and Neuroendocrine Regulations, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Daniil Berezhnoy
- Laboratory of Translational and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia
| | - Sergey Stvolinsky
- Laboratory of Translational and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia
| | - Yulia Timoshina
- Laboratory of Translational and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia
- Department of Neurobiology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Denis Abaimov
- Laboratory of Translational and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia
| | - Olga Muzychuk
- Laboratory of Translational and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia
| | - Alexander Latanov
- Department of Neurobiology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Research Institute of Functional Brain Development and Peak Performance, Peoples' Friendship University of Russia, 117198 Moscow, Russia
| | - Tatiana Fedorova
- Laboratory of Translational and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia
| |
Collapse
|
7
|
Volta M. Lysosomal Pathogenesis of Parkinson's Disease: Insights From LRRK2 and GBA1 Rodent Models. Neurotherapeutics 2023; 20:127-139. [PMID: 36085537 PMCID: PMC10119359 DOI: 10.1007/s13311-022-01290-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2022] [Indexed: 01/18/2023] Open
Abstract
The discovery of mutations in LRRK2 and GBA1 that are linked to Parkinson's disease provided further evidence that autophagy and lysosome pathways are likely implicated in the pathogenic process. Their protein products are important regulators of lysosome function. LRRK2 has kinase-dependent effects on lysosome activity, autophagic efficacy and lysosomal Ca2+ signaling. Glucocerebrosidase (encoded by GBA1) is a hydrolytic enzyme contained in the lysosomes and contributes to the degradation of alpha-synuclein. PD-related mutations in LRRK2 and GBA1 slow the degradation of alpha-synuclein, thus directly implicating the dysfunction of the process in the neuropathology of Parkinson's disease. The development of genetic rodent models of LRRK2 and GBA1 provided hopes of obtaining reliable preclinical models in which to study pathogenic processes and perform drug validation studies. Here, I will review the extensive characterization of these models, their impact on understanding lysosome alterations in the course of Parkinson's disease and what novel insights have been obtained. In addition, I will discuss how these models fare with respect to the features of a "gold standard" animal models and what could be attempted in future studies to exploit LRRK2 and GBA1 rodent models in the fight against Parkinson's disease.
Collapse
Affiliation(s)
- Mattia Volta
- Institute for Biomedicine, Eurac Research - Affiliated Institute of the University of Lübeck, via Volta 21, Bolzano, 39100, Italy.
| |
Collapse
|
8
|
Yang Y, Wang Y, Wang C, Xu X, Liu C, Huang X. Identification of hub genes of Parkinson's disease through bioinformatics analysis. Front Neurosci 2022; 16:974838. [PMID: 36440267 PMCID: PMC9683342 DOI: 10.3389/fnins.2022.974838] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/15/2022] [Indexed: 12/03/2024] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease, and there is still a lack of effective diagnostic and treatment methods. This study aimed to search for hub genes that might serve as diagnostic or therapeutic targets for PD. All the analysis was performed in R software. The expression profile data of PD (number: GSE7621) was acquired from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) associated with PD were screened by the "Limma" package of the R software. Key genes associated with PD were screened by the "WGCNA" package of the R software. Target genes were screened by merging the results of "Limma" and "WGCNA." Enrichment analysis of target genes was performed by Gene Ontology (GO), Disease Ontology (DO), and Kyoto Enrichment of Genes and Genomes (KEGG). Machine learning algorithms were employed to screen for hub genes. Nomogram was constructed using the "rms" package. And the receiver operating characteristic curve (ROC) was plotted to detect and validate our prediction model sensitivity and specificity. Additional expression profile data of PD (number: GSE20141) was acquired from the GEO database to validate the nomogram. GSEA was used to determine the biological functions of the hub genes. Finally, RPL3L, PLEK2, PYCRL, CD99P1, LOC100133130, MELK, LINC01101, and DLG3-AS1 were identified as hub genes of PD. These findings can provide a new direction for the diagnosis and treatment of PD.
Collapse
Affiliation(s)
- Yajun Yang
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, China
- The First School of Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Yi Wang
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, China
- The First School of Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Ce Wang
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, China
- The First School of Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Xinjuan Xu
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, China
- The First School of Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Cai Liu
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, China
- The First School of Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Xintao Huang
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
9
|
Edgar JA, Molyneux RJ, Colegate SM. 1,2-Dehydropyrrolizidine Alkaloids: Their Potential as a Dietary Cause of Sporadic Motor Neuron Diseases. Chem Res Toxicol 2022; 35:340-354. [PMID: 35238548 DOI: 10.1021/acs.chemrestox.1c00384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Sporadic motor neuron diseases (MNDs), such as amyotrophic lateral sclerosis (ALS), can be caused by spontaneous genetic mutations. However, many sporadic cases of ALS and other debilitating neurodegenerative diseases (NDDs) are believed to be caused by environmental factors, subject to considerable debate and requiring intensive research. A common pathology associated with MND development involves progressive mitochondrial dysfunction and oxidative stress in motor neurons and glial cells of the central nervous system (CNS), leading to apoptosis. Consequent degeneration of skeletal and respiratory muscle cells can lead to death from respiratory failure. A significant number of MND cases present with cancers and liver and lung pathology. This Perspective explores the possibility that MNDs could be caused by intermittent, low-level dietary exposure to 1,2-dehydropyrrolizidine alkaloids (1,2-dehydroPAs) that are increasingly recognized as contaminants of many foods consumed throughout the world. Nontoxic, per se, 1,2-dehydroPAs are metabolized, by particular cytochrome P450 (CYP450) isoforms, to 6,7-dihydropyrrolizines that react with nucleophilic groups (-NH, -SH, -OH) on DNA, proteins, and other vital biochemicals, such as glutathione. Many factors, including aging, gender, smoking, and alcohol consumption, influence CYP450 isoform activity in a range of tissues, including glial cells and neurons of the CNS. Activation of 1,2-dehydroPAs in CNS cells can be expected to cause gene mutations and oxidative stress, potentially leading to the development of MNDs and other NDDs. While relatively high dietary exposure to 1,2-dehydroPAs causes hepatic sinusoidal obstruction syndrome, pulmonary venoocclusive disease, neurotoxicity, and diverse cancers, this Perspective suggests that, at current intermittent, low levels of dietary exposure, neurotoxicity could become the primary pathology that develops over time in susceptible individuals, along with a tendency for some of them to also display liver and lung pathology and diverse cancers co-occurring with some MND/NDD cases. Targeted research is recommended to investigate this proposal.
Collapse
Affiliation(s)
- John A Edgar
- CSIRO Agriculture and Food, 11 Julius Avenue, North Ryde, New South Wales 2113, Australia
| | - Russell J Molyneux
- Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, 200 West Kawili Street, Hilo, Hawaii 96720, United States
| | - Steven M Colegate
- Poisonous Plant Research Laboratory, ARS/USDA, 1150 East 1400 North, Logan, Utah 84341, United States
| |
Collapse
|
10
|
Protective Activity of Aspirin Eugenol Ester on Paraquat-Induced Cell Damage in SH-SY5Y Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6697872. [PMID: 34394831 PMCID: PMC8360752 DOI: 10.1155/2021/6697872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 07/23/2021] [Indexed: 11/28/2022]
Abstract
Aspirin eugenol ester (AEE) is a new pharmaceutical compound esterified by aspirin and eugenol, which has anti-inflammatory, antioxidant, and other pharmacological activities. The aim of this study was to investigate the protective effect of AEE on paraquat- (PQ-) induced cell damage of SH-SY5Y human neuroblastoma cells and its potential molecular mechanism. There was no significant change in cell viability when AEE was used alone. PQ treatment reduced cell viability in a concentration-dependent manner. However, AEE reduced the PQ-induced loss of cell viability. Flow cytometry, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and 4′6-diamidino-2-phenylindole (DAPI) staining were used to evaluate cell apoptosis. Compared with the PQ group, AEE pretreatment could significantly inhibit PQ-induced cell damage. AEE pretreatment could reduce the cell damage of SH-SY5Y cells induced by PQ via reducing superoxide anion, intracellular reactive oxygen species (ROS), and mitochondrial ROS (mtROS) and increasing the levels of mitochondrial membrane potential (ΔΨm). At the same time, AEE could increase the activity of glutathione peroxidase (GSH-Px), catalase (CAT), and superoxide dismutase (SOD) and decrease the activity of malondialdehyde (MDA). The results showed that compared with the control group, the expression of p-PI3K, p-Akt, and Bcl-2 was significantly decreased, while the expression of caspase-3 and Bax was significantly increased in the PQ group. In the AEE group, AEE pretreatment could upregulate the expression of p-PI3K, p-Akt, and Bcl-2 and downregulate the expression of caspase-3 and Bax in SH-SY5Y cells. PI3K inhibitor LY294002 and the silencing of PI3K by shRNA could weaken the protective effect of AEE on PQ-induced SH-SY5Y cells. Therefore, AEE has a protective effect on PQ-induced SH-SY5Y cells by regulating the PI3K/Akt signal pathway to inhibit oxidative stress.
Collapse
|
11
|
Zhao F, Austria Q, Wang W, Zhu X. Mfn2 Overexpression Attenuates MPTP Neurotoxicity In Vivo. Int J Mol Sci 2021; 22:ijms22020601. [PMID: 33435331 PMCID: PMC7827738 DOI: 10.3390/ijms22020601] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 12/16/2022] Open
Abstract
Mitochondrial dysfunction represents a critical event in the pathogenesis of Parkinson’s disease (PD). Increasing evidence demonstrates that disturbed mitochondrial dynamics and quality control play an important role in mitochondrial dysfunction in PD. Our previous study demonstrated that MPP+ induces mitochondrial fragmentation in vitro. In this study, we aimed to assess whether blocking MPTP-induced mitochondrial fragmentation by overexpressing Mfn2 affords neuroprotection in vivo. We found that the significant loss of dopaminergic neurons in the substantia nigra (SN) induced by MPTP treatment, as seen in wild-type littermate control mice, was almost completely blocked in mice overexpressing Mfn2 (hMfn2 mice). The dramatic reduction in dopamine neuronal fibers and dopamine levels in the striatum caused by MPTP administration was also partially inhibited in hMfn2 mice. MPTP-induced oxidative stress and inflammatory response in the SN and striatum were significantly alleviated in hMfn2 mice. The impairment of motor function caused by MPTP was also blocked in hMfn2 mice. Overall, our work demonstrates that restoration of mitochondrial dynamics by Mfn2 overexpression protects against neuronal toxicity in an MPTP-based PD mouse model, which supports the modulation of mitochondrial dynamics as a potential therapeutic target for PD treatment.
Collapse
|
12
|
|
13
|
Sim H, Seo JH, Kim J, Oh M, Lee JE, Baek A, Lee SY, Chung SK, Son MY, Chae JI, Jeon YJ, Kim J. Quantitative Proteomic Analysis of Primitive Neural Stem Cells from LRRK2 G2019S-Associated Parkinson's Disease Patient-Derived iPSCs. Life (Basel) 2020; 10:E331. [PMID: 33297425 PMCID: PMC7762312 DOI: 10.3390/life10120331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/27/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease, causing movement defects. The incidence of PD is constantly increasing and this disease is still incurable. Thus, understanding PD pathophysiology would be pivotal for the development of PD therapy, and various PD models have thus been already developed. Through recent advances in reprogramming techniques, a primitive neural stem cell (pNSC) derived from PD patient induced pluripotent stem cells (iPSCs) could be potentially used as a reproducible and reliable experimental system to analyze the effect of the leucine-rich repeat kinase 2 G2019S mutation (LK2GS) in neural cells. Here, we investigated the advantages of such a model system through quantitative proteomic analysis of pNSCs from normal control iPSCs and familial PD patient iPSCs harboring LK2GS. We confirmed that the expression of molecules known to be involved in PD pathogenesis, such as oxidative stress-, cell adhesion-, and cytoskeleton-related proteins, were altered in the LK2GS pNSC. In addition, we showed that down-regulation of Ku80, which was found in the proteomic analysis with LK2GS pNSCs, resulted in apoptosis induced by DNA damage response. Taken together, we suggest that pNSCs from PD iPSCs could provide a reliable and useful model system to study PD. Moreover, the highly expandable pNSC is suitable for multi-omics approaches to understand PD pathologies and discover therapeutic targets for PD.
Collapse
Affiliation(s)
- Hyuna Sim
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.S.); (M.O.); (J.-E.L.); (A.B.); (M.-Y.S.)
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon 34113, Korea
| | - Ji-Hye Seo
- Department of Dental Pharmacology, School of Dentistry, BK21 Plus, Jeonbuk National University, Jeonju 54896, Korea; (J.-H.S.); (J.K.); (J.-I.C.)
| | - Jumi Kim
- Department of Dental Pharmacology, School of Dentistry, BK21 Plus, Jeonbuk National University, Jeonju 54896, Korea; (J.-H.S.); (J.K.); (J.-I.C.)
| | - Minyoung Oh
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.S.); (M.O.); (J.-E.L.); (A.B.); (M.-Y.S.)
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon 34113, Korea
| | - Joo-Eun Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.S.); (M.O.); (J.-E.L.); (A.B.); (M.-Y.S.)
| | - Areum Baek
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.S.); (M.O.); (J.-E.L.); (A.B.); (M.-Y.S.)
| | - Seo-Young Lee
- Division of Herbal Medicine Research, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Korea;
| | - Sun-Ku Chung
- Division of Clinical Medicine, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Korea;
| | - Mi-Young Son
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.S.); (M.O.); (J.-E.L.); (A.B.); (M.-Y.S.)
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon 34113, Korea
| | - Jung-Il Chae
- Department of Dental Pharmacology, School of Dentistry, BK21 Plus, Jeonbuk National University, Jeonju 54896, Korea; (J.-H.S.); (J.K.); (J.-I.C.)
| | - Young-Joo Jeon
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.S.); (M.O.); (J.-E.L.); (A.B.); (M.-Y.S.)
| | - Janghwan Kim
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.S.); (M.O.); (J.-E.L.); (A.B.); (M.-Y.S.)
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon 34113, Korea
| |
Collapse
|
14
|
Parsafar S, Nayeri Z, Aliakbari F, Shahi F, Mohammadi M, Morshedi D. Multiple neuroprotective features of Scutellaria pinnatifida-derived small molecule. Heliyon 2020; 6:e04737. [PMID: 32913905 PMCID: PMC7472859 DOI: 10.1016/j.heliyon.2020.e04737] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/29/2020] [Accepted: 08/12/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD) is one of the most prevalent neurodegenerative disorders with no precise etiology. Multiple lines of evidence support that environmental factors, either neurotoxins or neuroinflammation, can induce Parkinsonism. In this study, we purified an active compound, neobaicalein (Skullcapflavone II), from the roots of Scutellaria pinnatifida (S. pinnatifida). Neobaicalein not only had protective impacts on rotenone-induced neurotoxicity but in glial cultures, it dampened the inflammatory response when stimulated with lipopolysaccharide (LPS). Neobaicalein had high antioxidant activity without any obvious toxicity. In addition, it could raise the cell viability, decrease early apoptosis, reduce the generation of reactive oxygen species (ROS), and keep the neurite's length normal in the treated SH-SY5Y cells. Pathway enrichment analysis (PEA) and target prediction provided insights into the PD related genes, protein-protein interaction (PPI) network, and the key proteins enriched in the signaling pathways. Furthermore, docking simulation (DS) on the proteins of the PD-PPI network revealed that neobaicalein might interact with the key proteins involved in PD pathology, including MAPK14, MAPK8, and CASP3. It also blocks the destructive processes, such as cell death, inflammation, and oxidative stress pathways. Our results demonstrate that neobaicalein alleviates pathological effects of factors related to PD, and may provide new insight into PD therapy.
Collapse
Affiliation(s)
- Soha Parsafar
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Zahra Nayeri
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Farhang Aliakbari
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Farshad Shahi
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mehdi Mohammadi
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Dina Morshedi
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
15
|
Ferreira C, Almeida C, Tenreiro S, Quintas A. Neuroprotection or Neurotoxicity of Illicit Drugs on Parkinson's Disease. Life (Basel) 2020; 10:life10060086. [PMID: 32545328 PMCID: PMC7344445 DOI: 10.3390/life10060086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/20/2022] Open
Abstract
Parkinson's Disease (PD) is currently the most rapid growing neurodegenerative disease and over the past generation, its global burden has more than doubled. The onset of PD can arise due to environmental, sporadic or genetic factors. Nevertheless, most PD cases have an unknown etiology. Chemicals, such as the anthropogenic pollutant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and amphetamine-type stimulants, have been associated with the onset of PD. Conversely, cannabinoids have been associated with the treatment of the symptoms'. PD and medical cannabis is currently under the spotlight, and research to find its benefits on PD is on-going worldwide. However, the described clinical applications and safety of pharmacotherapy with cannabis products are yet to be fully supported by scientific evidence. Furthermore, the novel psychoactive substances are currently a popular alternative to classical drugs of abuse, representing an unknown health hazard for young adults who may develop PD later in their lifetime. This review addresses the neurotoxic and neuroprotective impact of illicit substance consumption in PD, presenting clinical evidence and molecular and cellular mechanisms of this association. This research area is utterly important for contemporary society since illicit drugs' legalization is under discussion which may have consequences both for the onset of PD and for the treatment of its symptoms.
Collapse
Affiliation(s)
- Carla Ferreira
- Molecular Pathology and Forensic Biochemistry Laboratory, Centro de Investigação Interdisciplinar Egas Moniz, P-2825-084 Caparica, Portugal; (C.F.); (C.A.)
- Laboratório de Ciências Forenses e Psicológicas Egas Moniz, Campus Universitário–Quinta da Granja, Monte de Caparica, P-2825-084 Caparica, Portugal
- Faculty of Medicine of Porto University, Al. Prof. Hernâni Monteiro, P-4200–319 Porto, Portugal
| | - Catarina Almeida
- Molecular Pathology and Forensic Biochemistry Laboratory, Centro de Investigação Interdisciplinar Egas Moniz, P-2825-084 Caparica, Portugal; (C.F.); (C.A.)
| | - Sandra Tenreiro
- CEDOC–Chronic Diseases Research Center, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, P-1150-082 Lisboa, Portugal;
| | - Alexandre Quintas
- Molecular Pathology and Forensic Biochemistry Laboratory, Centro de Investigação Interdisciplinar Egas Moniz, P-2825-084 Caparica, Portugal; (C.F.); (C.A.)
- Laboratório de Ciências Forenses e Psicológicas Egas Moniz, Campus Universitário–Quinta da Granja, Monte de Caparica, P-2825-084 Caparica, Portugal
- Correspondence:
| |
Collapse
|
16
|
Bordoni L, Gabbianelli R, Fedeli D, Fiorini D, Bergheim I, Jin CJ, Marinelli L, Di Stefano A, Nasuti C. Positive effect of an electrolyzed reduced water on gut permeability, fecal microbiota and liver in an animal model of Parkinson's disease. PLoS One 2019; 14:e0223238. [PMID: 31600256 PMCID: PMC6786615 DOI: 10.1371/journal.pone.0223238] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 09/17/2019] [Indexed: 12/18/2022] Open
Abstract
There is growing awareness within the scientific community of the strong connection between the inflammation in the intestine and the pathogenesis of Parkinson’s disease (PD). In previous studies we developed a PD animal model exposing pup rats to permethrin (PERM) pesticide. Here, we intended to explore whether in our animal model there were changes in gut permeability, fecal microbiota and hepatic injury. Moreover, we tested if the co-treatment with an electrolyzed reduced (ERW) was effective to protect against alterations induced by PERM. Rats (from postnatal day 6 to 21) were gavaged daily with PERM, PERM+ERW or vehicle and gut, liver and feces were analyzed in 2-months-old rats. Increased gut permeability, measured by FITC-dextran assay, was detected in PERM group compared to control and PERM+ERW groups. In duodenum and ileum, concentration of occludin was higher in control group than those measured in PERM group, whereas only in duodenum ZO-1 was higher in control than those measured in PERM and PERM+ERW groups. Number of inflammatory focis and neutrophils as well as iNOS protein levels were higher in livers of PERM-treated rats than in those of PERM+ERW and control rats. Fecal microbiota analysis revealed that Lachnospira was less abundant and Defluviitaleaceae more abundant in the PERM group, whereas the co-treatment with ERW was protective against PERM treatment since the abundances in Lachnospira and Defluviitaleaceae were similar to those in the control group. Higher abundances of butyrate- producing bacteria such as Blautia, U.m. of Lachnospiraceae family, U.m. of Ruminococcaceae family, Papillibacter, Roseburia, Intestinimonas, Shuttleworthia together with higher butyric acid levels were detected in PERM+ERW group compared to the other groups. In conclusion, the PD animal model showed increased intestinal permeability together with hepatic inflammation correlated with altered gut microbiota. The positive effects of ERW co-treatment observed in gut, liver and brain of rats were linked to changes on gut microbiota.
Collapse
Affiliation(s)
- Laura Bordoni
- School of Pharmacy, Molecular Biology Unit, University of Camerino, Camerino, Italy
| | - Rosita Gabbianelli
- School of Pharmacy, Molecular Biology Unit, University of Camerino, Camerino, Italy
| | - Donatella Fedeli
- School of Pharmacy, Molecular Biology Unit, University of Camerino, Camerino, Italy
| | - Dennis Fiorini
- School of Science and Technology, Chemistry Unit, University of Camerino, Camerino, Italy
| | - Ina Bergheim
- Department of Nutritional Sciences, RF Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Cheng Jun Jin
- Institute of Nutritional Sciences, SD Model Systems of Molecular Nutrition, Friedrich-Schiller-University, Jena, Germany
| | - Lisa Marinelli
- Department of Pharmacy, University of "G. D’Annunzio", Chieti, Italy
| | | | - Cinzia Nasuti
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
- * E-mail:
| |
Collapse
|
17
|
Ren C, Ding Y, Wei S, Guan L, Zhang C, Ji Y, Wang F, Yin S, Yin P. G2019S Variation in LRRK2: An Ideal Model for the Study of Parkinson's Disease? Front Hum Neurosci 2019; 13:306. [PMID: 31551736 PMCID: PMC6738350 DOI: 10.3389/fnhum.2019.00306] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 08/19/2019] [Indexed: 12/17/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and has plagued humans for more than 200 years. The etiology and detailed pathogenesis of PD is unclear, but is currently believed to be the result of the interaction between genetic and environmental factors. Studies have found that PD patients with the LRRK2:G2019S variation have the typical clinical manifestations of PD, which may be familial or sporadic, and have age-dependent pathogenic characteristics. Therefore, the LRRK2:G2019S variation may be an ideal model to study the interaction of multiple factors such as genetic, environmental and natural aging factors in PD in the future. This article reviewed the progress of LRRK2:G2019S studies in PD research in order to provide new research ideas and directions for the pathogenesis and treatment of PD.
Collapse
Affiliation(s)
- Chao Ren
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yu Ding
- Institute of Neuroscience, Soochow University, Suzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shizhuang Wei
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Lina Guan
- Department of Neurosurgical Intensive Care Unit, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Caiyi Zhang
- Department of Emergency and Rescue Medicine, Xuzhou Medical University, Xuzhou, China
| | - Yongqiang Ji
- Department of Nephrology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Fen Wang
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Shaohua Yin
- Department of Nursing, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Peiyuan Yin
- Department of Blood Supply, Yantai Center Blood Station, Yantai, China
| |
Collapse
|
18
|
Colle D, Santos DB, Naime AA, Gonçalves CL, Ghizoni H, Hort MA, Farina M. Early Postnatal Exposure to Paraquat and Maneb in Mice Increases Nigrostriatal Dopaminergic Susceptibility to a Re-challenge with the Same Pesticides at Adulthood: Implications for Parkinson's Disease. Neurotox Res 2019; 37:210-226. [PMID: 31422567 DOI: 10.1007/s12640-019-00097-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 07/10/2019] [Accepted: 08/06/2019] [Indexed: 02/13/2023]
Abstract
Exposure to environmental contaminants represents an important etiological factor in sporadic Parkinson's disease (PD). It has been reported that PD could arise from events that occur early in development and that lead to delayed adverse consequences in the nigrostriatal dopaminergic system at adult life. We investigated the occurrence of late nigrostriatal dopaminergic neurotoxicity induced by exposures to the pesticides paraquat (PQ) and maneb (MB) during the early postnatal period in mice, as well as whether the exposure to pesticides during development could enhance mice vulnerability to subsequent challenges. Male Swiss mice were exposed to a combination of 0.3 mg/kg PQ and 1.0 mg/kg MB (PQ + MB) from postnatal (PN) day 5 to 19. PN exposure to pesticides neither induced mortally nor modified motor-related parameters. However, PN pesticides exposure decreased the number of tyrosine hydroxylase (TH)- and dopamine transporter (DAT)-positive neurons in the substantia nigra pars compacta (SNpc), as well as reduced TH and DAT immunoreactivity in the striatum. A parallel group of animals developmentally exposed to the pesticides was re-challenged at 3 months of age with 10 mg/kg PQ plus 30 mg/kg MB (twice a week, 6 weeks). Mice exposed to pesticides at both periods (PN + adulthood) presented motor deficits and reductions in the number of TH- and DAT-positive neurons in the SNpc. These findings indicate that the exposure to PQ + MB during the early PN period can cause neurotoxicity in the mouse nigrostriatal dopaminergic system, rendering it more susceptible to a subsequent adult re-challenge with the same pesticides.
Collapse
Affiliation(s)
- Dirleise Colle
- Departamento de Análises Clínicas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil. .,Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| | - Danúbia Bonfanti Santos
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Aline Aita Naime
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Cinara Ludvig Gonçalves
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Heloisa Ghizoni
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Mariana Appel Hort
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande, Rio Grande do Sul, Brazil
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
19
|
Aivazidis S, Anderson CC, Roede JR. Toxicant-mediated redox control of proteostasis in neurodegeneration. CURRENT OPINION IN TOXICOLOGY 2019; 13:22-34. [PMID: 31602419 PMCID: PMC6785977 DOI: 10.1016/j.cotox.2018.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Disruption in redox signaling and control of cellular processes has emerged as a key player in many pathologies including neurodegeneration. As protein aggregations are a common hallmark of several neuronal pathologies, a firm understanding of the interplay between redox signaling, oxidative and free radical stress, and proteinopathies is required to sort out the complex mechanisms in these diseases. Fortunately, models of toxicant-induced neurodegeneration can be utilized to evaluate and report mechanistic alterations in the proteostasis network (PN). The epidemiological links between environmental toxicants and neurological disease gives further credence into characterizing the toxicant-mediated PN disruptions observed in these conditions. Reviewed here are examples of mechanistic interaction between oxidative or free radical stress and PN alterations. Additionally, investigations into toxicant-mediated PN disruptions, specifically focusing on environmental metals and pesticides, are discussed. Finally, we emphasize the need to distinguish whether the presence of protein aggregations are contributory to phenotypes related to neurodegeneration, or if they are a byproduct of PN deficiencies.
Collapse
Affiliation(s)
- Stefanos Aivazidis
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Colin C Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - James R Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
20
|
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: 68] [Impact Index Per Article: 9.7] [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
|
21
|
Pacific Ciguatoxin Induces Excitotoxicity and Neurodegeneration in the Motor Cortex Via Caspase 3 Activation: Implication for Irreversible Motor Deficit. Mol Neurobiol 2018; 55:6769-6787. [DOI: 10.1007/s12035-018-0875-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 01/07/2018] [Indexed: 12/14/2022]
|
22
|
More SV, Choi DK. Emerging preclinical pharmacological targets for Parkinson's disease. Oncotarget 2018; 7:29835-63. [PMID: 26988916 PMCID: PMC5045437 DOI: 10.18632/oncotarget.8104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/08/2016] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurological condition caused by the degeneration of dopaminergic neurons in the basal ganglia. It is the most prevalent form of Parkinsonism, categorized by cardinal features such as bradykinesia, rigidity, tremors, and postural instability. Due to the multicentric pathology of PD involving inflammation, oxidative stress, excitotoxicity, apoptosis, and protein aggregation, it has become difficult to pin-point a single therapeutic target and evaluate its potential application. Currently available drugs for treating PD provide only symptomatic relief and do not decrease or avert disease progression resulting in poor patient satisfaction and compliance. Significant amount of understanding concerning the pathophysiology of PD has offered a range of potential targets for PD. Several emerging targets including AAV-hAADC gene therapy, phosphodiesterase-4, potassium channels, myeloperoxidase, acetylcholinesterase, MAO-B, dopamine, A2A, mGlu5, and 5-HT-1A/1B receptors are in different stages of clinical development. Additionally, alternative interventions such as deep brain stimulation, thalamotomy, transcranial magnetic stimulation, and gamma knife surgery, are also being developed for patients with advanced PD. As much as these therapeutic targets hold potential to delay the onset and reverse the disease, more targets and alternative interventions need to be examined in different stages of PD. In this review, we discuss various emerging preclinical pharmacological targets that may serve as a new promising neuroprotective strategy that could actually help alleviate PD and its symptoms.
Collapse
Affiliation(s)
- Sandeep Vasant More
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| |
Collapse
|
23
|
Ortega-Arellano HF, Jimenez-Del-Rio M, Velez-Pardo C. Minocycline protects, rescues and prevents knockdown transgenic parkin Drosophila against paraquat/iron toxicity: Implications for autosomic recessive juvenile parkinsonism. Neurotoxicology 2017; 60:42-53. [PMID: 28284907 DOI: 10.1016/j.neuro.2017.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 02/03/2017] [Accepted: 03/06/2017] [Indexed: 01/03/2023]
Abstract
Autosomal recessive Juvenile Parkinsonism (AR-JP) is a chronic, progressive neurodegenerative disorder caused by mutation in the PARKIN gene, and invariably associated with dopaminergic (DAergic) neuronal loss and brain iron accumulation. Since current medical therapy is symptomatic and lacks significant disease-modifying effects, other treatment approaches are urgently needed it. In the present work, we investigate the role of minocycline (MC) in paraquat (PQ)/iron-induced neurotoxicity in the Drosophila TH>parkin-RNAi/+ (w[*]; UAS-parkin-RNAi; TH-GAL4) fly and have shown the following: (i) MC increased life span and restored the locomotor activity of knockdown (KD) transgenic parkin flies in comparison with the control (vehicle) group; (ii) MC at low (0.1 and 0.3mM) and middle (0.5mM) concentrations protected, rescued and prevented KD parkin Drosophila against PQ toxicity. However, MC at high (1mM) concentration aggravated the toxic effect of PQ; (iii) MC protected and rescued DAergic neurons against the PQ toxic effect according to tyrosine hydroxylase (TH)>green-fluorescent protein (GFP) reporter protein microscopy and anti-TH Western blotting analysis; (iv) MC protected DAergic neurons against PQ/iron toxicity; (v) MC significantly abridged lipid peroxidation (LPO) in the protection, rescue and prevention treatment in TH>parkin-RNAi/+ flies against PQ or iron alone or combined (PQ/iron)-induced neuronal oxidative stress (OS). Our results suggest that MC exerts neuroprotection against PQ/iron-induced OS in DAergic neurons most probably by the scavenging activity of reactive oxygen species (ROS), and by chelating iron. Therefore, MC might be a potential therapeutic drug to delay, revert, or prevent AR-JP.
Collapse
Affiliation(s)
- Hector Flavio Ortega-Arellano
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Building 1, Room 412, SIU, Medellin, Colombia
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Building 1, Room 412, SIU, Medellin, Colombia.
| | - Carlos Velez-Pardo
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Building 1, Room 412, SIU, Medellin, Colombia.
| |
Collapse
|
24
|
Rietdijk CD, Perez-Pardo P, Garssen J, van Wezel RJA, Kraneveld AD. Exploring Braak's Hypothesis of Parkinson's Disease. Front Neurol 2017; 8:37. [PMID: 28243222 PMCID: PMC5304413 DOI: 10.3389/fneur.2017.00037] [Citation(s) in RCA: 206] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 01/26/2017] [Indexed: 12/21/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder for which there is no cure. Most patients suffer from sporadic PD, which is likely caused by a combination of genetic and environmental factors. Braak’s hypothesis states that sporadic PD is caused by a pathogen that enters the body via the nasal cavity, and subsequently is swallowed and reaches the gut, initiating Lewy pathology (LP) in the nose and the digestive tract. A staging system describing the spread of LP from the peripheral to the central nervous system was also postulated by the same research group. There has been criticism to Braak’s hypothesis, in part because not all patients follow the proposed staging system. Here, we review literature that either supports or criticizes Braak’s hypothesis, focused on the enteric route, digestive problems in patients, the spread of LP on a tissue and a cellular level, and the toxicity of the protein αSynuclein (αSyn), which is the major constituent of LP. We conclude that Braak’s hypothesis is supported by in vitro, in vivo, and clinical evidence. However, we also conclude that the staging system of Braak only describes a specific subset of patients with young onset and long duration of the disease.
Collapse
Affiliation(s)
- Carmen D Rietdijk
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University , Utrecht , Netherlands
| | - Paula Perez-Pardo
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University , Utrecht , Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands; Nutricia Research, Utrecht, Netherlands
| | - Richard J A van Wezel
- Department of Biomedical Signals and Systems, MIRA, University of Twente, Enschede, Netherlands; Department of Biophysics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University , Utrecht , Netherlands
| |
Collapse
|
25
|
Del Tredici K, Braak H. Review: Sporadic Parkinson's disease: development and distribution of α-synuclein pathology. Neuropathol Appl Neurobiol 2016; 42:33-50. [PMID: 26662475 DOI: 10.1111/nan.12298] [Citation(s) in RCA: 286] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 12/04/2015] [Accepted: 12/13/2015] [Indexed: 12/17/2022]
Abstract
The development of α-synuclein immunoreactive aggregates in selectively vulnerable neuronal types of the human central, peripheral, and enteric nervous systems is crucial for the pathogenesis of sporadic Parkinson's disease. The presence of these lesions persists into the end phase of the disease, a process that is not subject to remission. The initial induction of α-synuclein misfolding and subsequent aggregation probably occurs in the olfactory bulb and/or the enteric nervous system. Each of these sites is exposed to potentially hostile environmental factors. Once formed, the aggregates appear to be capable of propagating trans-synaptically from nerve cell to nerve cell in a virtually self-promoting pathological process. A regional distribution pattern of aggregated α-synuclein emerges that entails the involvement of only a few types of susceptible and axonally interconnected projection neurons within the human nervous system. One major route of disease progression may originate in the enteric nervous system and retrogradely reach the dorsal motor nucleus of the vagal nerve in the lower brainstem. From there, the disease process proceeds chiefly in a caudo-rostral direction through visceromotor and somatomotor brainstem centres to the midbrain, forebrain, and cerebral cortex. Spinal cord centres may become involved by means of descending projections from involved lower brainstem nuclei as well as by sympathetic projections connecting the enteric nervous system with postganglionic peripheral ganglia and preganglionic nuclei of the spinal cord. The development of experimental cellular and animal models is helping to explain the mechanisms of how abnormal α-synuclein can undergo aggregation and how transmission along axonal connectivities can occur, thereby encouraging the initiation of potential disease-modifying therapeutic strategies for sporadic Parkinson's disease.
Collapse
Affiliation(s)
- K Del Tredici
- Clinical Neuroanatomy Section, Department of Neurology, Center for Biomedical Research, University of Ulm, Ulm, Germany
| | - H Braak
- Clinical Neuroanatomy Section, Department of Neurology, Center for Biomedical Research, University of Ulm, Ulm, Germany
| |
Collapse
|
26
|
Koh YQ, Peiris HN, Vaswani K, Reed S, Rice GE, Salomon C, Mitchell MD. Characterization of exosomal release in bovine endometrial intercaruncular stromal cells. Reprod Biol Endocrinol 2016; 14:78. [PMID: 27829441 PMCID: PMC5103490 DOI: 10.1186/s12958-016-0207-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 10/25/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cell-to-cell communication between the blastocyst and endometrium is critical for implantation. In recent years, evidence has emerged from studies in humans and several other animal species that exosomes are secreted from the endometrium and trophoblast cells and may play an important role in cell-to-cell communication maternal-fetal interface during early pregnancy. Exosomes are stable extracellular lipid bilayer vesicles that encapsulate proteins, miRNAs, and mRNAs, with the ability to deliver their cargo to near and distant sites, altering cellular function(s). Furthermore, the exosomal cargo can be altered in response to environmental cues (e.g. hypoxia). The current study aims to develop an in vitro system to evaluate maternal-embryo interactions via exosomes (and exosomal cargo) produced by bovine endometrial stromal cells (ICAR) using hypoxia as a known stimulus associated with the release of exosomes and alterations to biological responses (e.g. cell proliferation). METHODS ICAR cells cultured under 8 % O2 or 1 % O2 for 48 h and changes in cell function (i.e. migration, proliferation and apoptosis) were evaluated. Exosome release was determined following the isolation (via differential centrifugation) and characterization of exosomes from ICAR cell-conditioned media. Exosomal proteomic content was evaluated by mass spectrometry. RESULTS Under hypoxic conditions (i.e. 1 % O2), ICAR cell migration and proliferation was decreased (~20 and ~32 %, respectively) and apoptotic protein caspase-3 activation was increased (∼1.6 fold). Hypoxia increased exosome number by ~3.6 fold compared with culture at 8 % O2. Mass spectrometry analysis identified 128 proteins unique to exosomes of ICAR cultured at 1 % O2 compared with only 46 proteins unique to those of ICAR cultured at 8 % O2. Differential production of proteins associated with specific biological processes and molecular functions were identified, most notably ADAM10, pantetheinase and kininogen 2. CONCLUSIONS In summary, we have shown that a stimulus such as hypoxia can alter both the cellular function and exosome release of ICAR cells. Alterations to exosome release and exosomal content in response to stimuli may play a crucial role in maternal-fetal crosstalk and could also affect placental development.
Collapse
Affiliation(s)
- Yong Qin Koh
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland Australia
| | - Hassendrini N. Peiris
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland Australia
| | - Kanchan Vaswani
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland Australia
| | - Sarah Reed
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland Australia
| | - Gregory E. Rice
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland Australia
| | - Carlos Salomon
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland Australia
| | - Murray D. Mitchell
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland Australia
| |
Collapse
|
27
|
Zhang LM, Sun CC, Mo MS, Cen L, Wei L, Luo FF, Li Y, Li GF, Zhang SY, Yi L, Huang W, Liu ZL, Le WD, Xu PY. Dopamine Agonists Exert Nurr1-inducing Effect in Peripheral Blood Mononuclear Cells of Patients with Parkinson's Disease. Chin Med J (Engl) 2016; 128:1755-60. [PMID: 26112716 PMCID: PMC4733729 DOI: 10.4103/0366-6999.159349] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background: Nurr1 plays an essential role in the development, survival, and function maintenance of midbrain dopaminergic (DA) neurons, and it is a potential target for Parkinson's disease (PD). Nurr1 mRNA can be detected in peripheral blood mononuclear cells (PBMCs), but whether there is any association of altered Nurr1 expression in PBMC with the disease and DA drug treatments remains elusive. This study aimed to measure the Nurr1 mRNA level in PBMC and evaluate the effect of Nurr1 expression by DA agents in vivo and in vitro. Methods: The mRNA levels of Nurr1 in PBMC of four subgroups of 362 PD patients and 193 healthy controls (HCs) using real-time polymerase chain reaction were measured. The nonparametric Mann-Whitney U-test and Kruskal-Wallis test were performed to evaluate the differences between PD and HC, as well as the subgroups of PD. Multivariate linear regression analysis was used to evaluate the independent association of Nurr1 expression with Hoehn and Yahr scale, age, and drug treatments. Besides, the Nurr1 expression in cultured PBMC was measured to determine whether DA agonist pramipexole affects its mRNA level. Results: The relative Nurr1 mRNA levels in DA agonists treated subgroup were significant higher than those in recent-onset cases without any anti-PD treatments (de novo) (P < 0.001) and HC groups (P < 0.010), respectively. Furthermore, the increase in Nurr1 mRNA expression was seen in DA agonist and L-dopa group. Multivariate linear regression showed DA agonists, L-dopa, and DA agonists were independent predictors correlated with Nurr1 mRNA expression level in PBMC. In vitro, in the cultured PBMC treated with 10 μmol/L pramipexole, the Nurr1 mRNA levels were significantly increased by 99.61%, 71.75%, 73.16% in 2, 4, and 8 h, respectively (P < 0.001). Conclusions: DA agonists can induce Nurr1 expression in PBMC, and such effect may contribute to DA agonists-mediated neuroprotection on DA neurons.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Ping-Yi Xu
- Department of Neurology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080; Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong 510120, China
| |
Collapse
|
28
|
Alpha synuclein protein is involved in Aluminum-induced cell death and oxidative stress in PC12 cells. Brain Res 2016; 1635:153-60. [PMID: 26826584 DOI: 10.1016/j.brainres.2016.01.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 12/16/2015] [Accepted: 01/22/2016] [Indexed: 12/20/2022]
Abstract
Increased expression and aggregation of α-synuclein (α-syn) protein plays a critical role in mediating the toxic effects of a number of neurodegenerative substances including metals. Thus, knockdown expression of α-syn is proposed as a possible modality for treatment of Parkinson disease (PD). Aluminum (Al) is a neurotoxic metal that contributes to pathogenesis of PD. The aim of this study was to investigate the role of α-syn protein in mediating Al-induced toxicity in PC12 cells. Specific α-syn small interference RNA (siRNA) was applied to knockdown the expression of α-syn protein in PC12 cells. The effects of different concentrations of Al-maltolate (Almal) were then evaluated on cell viability and oxidative stress in the α-syn downregulated cells. The results showed that Almal dose dependently induced apoptosis and increased malondialdehyde (MDA) and catalase activity in PC12 cells. Downregulation of α-syn protein significantly increased cell viability and decreased oxidative markers in Almal-treated cells. These findings suggest that α-syn protein may mediate Al-induced apoptosis and oxidative stress in PC12 cells.
Collapse
|
29
|
Oliveira LMA, Falomir-Lockhart LJ, Botelho MG, Lin KH, Wales P, Koch JC, Gerhardt E, Taschenberger H, Outeiro TF, Lingor P, Schüle B, Arndt-Jovin DJ, Jovin TM. Elevated α-synuclein caused by SNCA gene triplication impairs neuronal differentiation and maturation in Parkinson's patient-derived induced pluripotent stem cells. Cell Death Dis 2015; 6:e1994. [PMID: 26610207 PMCID: PMC4670926 DOI: 10.1038/cddis.2015.318] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 09/23/2015] [Indexed: 12/20/2022]
Abstract
We have assessed the impact of α-synuclein overexpression on the differentiation potential and phenotypic signatures of two neural-committed induced pluripotent stem cell lines derived from a Parkinson's disease patient with a triplication of the human SNCA genomic locus. In parallel, comparative studies were performed on two control lines derived from healthy individuals and lines generated from the patient iPS-derived neuroprogenitor lines infected with a lentivirus incorporating a small hairpin RNA to knock down the SNCA mRNA. The SNCA triplication lines exhibited a reduced capacity to differentiate into dopaminergic or GABAergic neurons and decreased neurite outgrowth and lower neuronal activity compared with control cultures. This delayed maturation phenotype was confirmed by gene expression profiling, which revealed a significant reduction in mRNA for genes implicated in neuronal differentiation such as delta-like homolog 1 (DLK1), gamma-aminobutyric acid type B receptor subunit 2 (GABABR2), nuclear receptor related 1 protein (NURR1), G-protein-regulated inward-rectifier potassium channel 2 (GIRK-2) and tyrosine hydroxylase (TH). The differentiated patient cells also demonstrated increased autophagic flux when stressed with chloroquine. We conclude that a two-fold overexpression of α-synuclein caused by a triplication of the SNCA gene is sufficient to impair the differentiation of neuronal progenitor cells, a finding with implications for adult neurogenesis and Parkinson's disease progression, particularly in the context of bioenergetic dysfunction.
Collapse
Affiliation(s)
- L M A Oliveira
- Laboratory of Cellular Dynamics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, Göttingen, Germany
| | - L J Falomir-Lockhart
- Laboratory of Cellular Dynamics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, Göttingen, Germany
| | - M G Botelho
- Laboratory of Cellular Dynamics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, Göttingen, Germany
| | - K-H Lin
- Group of Membrane Biophysics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, Göttingen, Germany
| | - P Wales
- Department of Neurodegeneration and Restorative Research, University Medical Center Göttingen, Waldweg 33, Göttingen, Germany
| | - J C Koch
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, Göttingen, Germany
| | - E Gerhardt
- Department of Neurodegeneration and Restorative Research, University Medical Center Göttingen, Waldweg 33, Göttingen, Germany
| | - H Taschenberger
- Group of Membrane Biophysics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, Göttingen, Germany
- DFG-Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - T F Outeiro
- Department of Neurodegeneration and Restorative Research, University Medical Center Göttingen, Waldweg 33, Göttingen, Germany
- DFG-Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - P Lingor
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, Göttingen, Germany
- DFG-Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - B Schüle
- The Parkinson's Institute, 675 Almanor Ave., Sunnyvale, CA, USA
| | - D J Arndt-Jovin
- Laboratory of Cellular Dynamics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, Göttingen, Germany
| | - T M Jovin
- Laboratory of Cellular Dynamics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, Göttingen, Germany
- Laboratory of Cellular Dynamics, Max Planck Institute for Biophysical Chemistry, Am FaÃberg 11, Göttingen 37077, Germany. Tel: +49 551 201 1381; Fax: +49 551 201 1467; E-mail:
| |
Collapse
|
30
|
Abstract
More than 80 % of patients with Parkinson's disease (PD) develop dysphagia during the course of their disease. Swallowing impairment reduces quality of life, complicates medication intake and leads to malnutrition and aspiration pneumonia, which is a major cause of death in PD. Although the underlying pathophysiology is poorly understood, it has been shown that dopaminergic and non-dopaminergic mechanisms are involved in the development of dysphagia in PD. Clinical assessment of dysphagia in PD patients is challenging and often delivers unreliable results. A modified water test assessing maximum swallowing volume is recommended to uncover oropharyngeal dysphagia in PD. PD-specific questionnaires may also be useful to identify patients at risk for swallowing impairment. Fiberoptic endoscopic evaluation of swallowing and videofluoroscopic swallowing study are both considered to be the gold standard for evaluation of PD-related dysphagia. In addition, high-resolution manometry may be a helpful tool. These instrumental methods allow a reliable detection of aspiration events. Furthermore, typical patterns of impairment during the oral, pharyngeal and/or esophageal swallowing phase of PD patients can be identified. Therapy of dysphagia in PD consists of pharmacological interventions and swallowing treatment by speech and language therapists (SLTs). Fluctuating dysphagia with deterioration during the off-state should be treated by optimizing dopaminergic medication. The methods used during swallowing treatment by SLTs shall be selected according to the individual dysphagia pattern of each PD patient. A promising novel method is an intensive training of expiratory muscle strength. Deep brain stimulation does not seem to have a clinical relevant effect on swallowing function in PD. The goal of this review is giving an overview on current stages of epidemiology, pathophysiology, diagnosis, and treatment of PD-associated dysphagia, which might be helpful for neurologists, speech-language therapists, and other clinicians in their daily work with PD patients and associated swallowing difficulties. Furthermore areas with an urgent need for future clinical research are identified.
Collapse
|
31
|
Liang Y, Jing X, Zeng Z, Bi W, Chen Y, Wu X, Yang L, Liu J, Xiao S, Liu S, Lin D, Tao E. Rifampicin attenuates rotenone-induced inflammation via suppressing NLRP3 inflammasome activation in microglia. Brain Res 2015; 1622:43-50. [PMID: 26086368 DOI: 10.1016/j.brainres.2015.06.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 06/06/2015] [Accepted: 06/08/2015] [Indexed: 12/12/2022]
Abstract
A growing body of evidence has supported that environmental factors, such as exposure to heavy metal and pesticides, play an important role in the pathogenesis of Parkinson׳s disease (PD). Rotenone, the active ingredient in various pesticides, has been identified as an inducer of PD. It has been revealed that rotenone induces activation of microglia and generation of pro-inflammatory factors in PD. Our previous studies demonstrated that rifampicin possessed neural protective effect in PD. In this study, we aimed to study the effect of rifampicin on the inflammation induced by rotenone in microglia and the underlying mechanisms. Results demonstrated that rifampicin pretreatment significantly reduced rotenone-induced cytotoxicity and gene expression of IL-1β in BV2 microglia. Moreover, western blot analysis verified that rifampicin pretreatment suppressed NLRP3 inflammasome activation via inhibiting caspase-1 cleavage and protein expression of NLRP3. As it is indicated that reactive oxidative stress (ROS) is one of the activators for NLRP3 inflammasome, we further employed 2',7'-Dichlorodihydrofluorescein diacetate (DCFH-DA) staining and Rhodamine123 staining to detect intracellular ROS and mitochondrial membrane potential (MMP), respectively. Results confirmed that rifampicin obviously reduced intracellular ROS and reversed loss of MMP in BV2 cells treated by rotenone. Taken together, our data indicate that rifampicin pretreatment inhibits maturation of IL-1β and neuroinflammation induced by rotenone via attenuating NLRP3 inflammasome activation. Rifampicin might emerge as a promising candidate for modulating neuroinflammation in PD.
Collapse
Affiliation(s)
- Yanran Liang
- Department of Neurology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, No. 107, West Yanjiang Road, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Xiuna Jing
- Department of Neurology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, No. 107, West Yanjiang Road, Guangzhou 510120, China
| | - Zhifen Zeng
- Department of Neurology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, No. 107, West Yanjiang Road, Guangzhou 510120, China
| | - Wei Bi
- Department of Neurology, The First Affiliated Hospital of Jinan University, No. 613, West Huangpu Road, Guangzhou 510630, China
| | - Ying Chen
- Department of Neurology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, No. 107, West Yanjiang Road, Guangzhou 510120, China
| | - Xia Wu
- Department of Neurology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, No. 107, West Yanjiang Road, Guangzhou 510120, China
| | - Lianhong Yang
- Department of Neurology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, No. 107, West Yanjiang Road, Guangzhou 510120, China
| | - Jun Liu
- Department of Neurology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, No. 107, West Yanjiang Road, Guangzhou 510120, China
| | - Songhua Xiao
- Department of Neurology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, No. 107, West Yanjiang Road, Guangzhou 510120, China
| | - Shuqiong Liu
- Department of Neurology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, No. 107, West Yanjiang Road, Guangzhou 510120, China
| | - Danyu Lin
- Department of Neurology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, No. 107, West Yanjiang Road, Guangzhou 510120, China
| | - Enxiang Tao
- Department of Neurology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, No. 107, West Yanjiang Road, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
32
|
Investigating the role of Sirt1-modulated oxidative stress in relation to benign paroxysmal positional vertigo and Parkinson's disease. Neurobiol Aging 2015; 36:2607-16. [PMID: 26130063 DOI: 10.1016/j.neurobiolaging.2015.05.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/29/2015] [Accepted: 05/20/2015] [Indexed: 01/08/2023]
Abstract
Benign paroxysmal positional vertigo (BPPV) is one of the most frequently encountered primary complaints in dizziness clinics. The incidence of BPPV has been proven to increase with age. The relationship between BPPV and another neurodegenerative disease, Parkinson's disease (PD), has not been previously discussed. This study aimed to investigate the relationship of BPPV and PD with oxidative stress. A total of 30,811 subjects participated in our cohort study. The study cohort comprised 5057 BPPV patients and a comparison cohort of 25,754 nonBPPV patients. SIRT1 axis gene expression was investigated in BPPV patient blood samples and a PD cell model of 6-hydroxydopamine (6-OHDA)-treated PC-12 cells to elucidate the potential in vitro and in vivo mechanisms of degeneration in PD and BPPV. Our data suggest that BPPV patients with histories of head injuries show a significantly higher hazard to develop subsequent PD (hazard ratio, 3.942; confidence interval, 1.523-10.205, p = 0.005). We also observed that oxidative status is increased in blood samples from patients with BPPV. Our in vitro study suggests that SIRT1 function is inhibited by oxidative stress, which thereby promotes 6-hydroxydopamine-induced cell death. We conclude that BPPV is independently associated with an increased risk of PD. This finding may be attributed to oxidative stress-mediated inhibition of SIRT1 expression levels.
Collapse
|
33
|
Jellinger KA. Neuropathobiology of non-motor symptoms in Parkinson disease. J Neural Transm (Vienna) 2015; 122:1429-40. [PMID: 25976432 DOI: 10.1007/s00702-015-1405-5] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 04/30/2015] [Indexed: 01/23/2023]
Abstract
Parkinson disease (PD) is a multisystem disorder associated with α-synuclein aggregates throughout the central, autonomic, and peripheral nervous system, clinically characterized by motor and non-motor (NM) symptoms. The NMS in PD, many of which antedating motor dysfunction and representing a preclinical phase spanning 20 or more years, are linked to widespread distribution of α-synuclein pathology not restricted to the dopaminergic nigrostriatal system that is responsible for core motor features of PD. The pathologic substrate of NM manifestations such as olfactory, autonomic (gastrointestinal, urogenital, cardia, respiratory), sensory, skin, sleep, visual, neuropsychiatric dysfunctions (cognitive, mood, dementia), and others are critically reviewed. In addition to non-nigral brainstem nuclei, α-synuclein pathology involves sympathetic and parasympathetic, enteric, cardiac and pelvic plexuses, and many other organs indicating a topographical and chronological spread, particularly in the prodromal stages of the disease. Few animal models recapitulate NMS in PD. The relationship between regional α-synuclein/Lewy pathology, neurodegeneration and the corresponding clinical deficits awaits further elucidation. Controlled clinicopathologic studies will refine the correlations between presymptomatic and late-developing NM features of PD and neuropathology, and new premotor biomarkers will facilitate early diagnosis of PD as a basis for more effective preventive and therapeutic options of this devastating disease.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Kenyongasse 18, 1070, Vienna, Austria.
| |
Collapse
|