51
|
Song Q, Meng B, Xu H, Mao Z. The emerging roles of vacuolar-type ATPase-dependent Lysosomal acidification in neurodegenerative diseases. Transl Neurodegener 2020; 9:17. [PMID: 32393395 PMCID: PMC7212675 DOI: 10.1186/s40035-020-00196-0] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/23/2020] [Indexed: 12/15/2022] Open
Abstract
Background Lysosomes digest extracellular material from the endocytic pathway and intracellular material from the autophagic pathway. This process is performed by the resident hydrolytic enzymes activated by the highly acidic pH within the lysosomal lumen. Lysosome pH gradients are mainly maintained by the vacuolar (H+) ATPase (or V-ATPase), which pumps protons into lysosomal lumen by consuming ATP. Dysfunction of V-ATPase affects lysosomal acidification, which disrupts the clearance of substrates and leads to many disorders, including neurodegenerative diseases. Main body As a large multi-subunit complex, the V-ATPase is composed of an integral membrane V0 domain involved in proton translocation and a peripheral V1 domain catalyzing ATP hydrolysis. The canonical functions of V-ATPase rely on its H+-pumping ability in multiple vesicle organelles to regulate endocytic traffic, protein processing and degradation, synaptic vesicle loading, and coupled transport. The other non-canonical effects of the V-ATPase that are not readily attributable to its proton-pumping activity include membrane fusion, pH sensing, amino-acid-induced activation of mTORC1, and scaffolding for protein-protein interaction. In response to various stimuli, V-ATPase complex can reversibly dissociate into V1 and V0 domains and thus close ATP-dependent proton transport. Dysregulation of pH and lysosomal dysfunction have been linked to many human diseases, including neurodegenerative disorders such as Alzheimer disease, Parkinson’s disease, amyotrophic lateral sclerosis as well as neurodegenerative lysosomal storage disorders. Conclusion V-ATPase complex is a universal proton pump and plays an important role in lysosome acidification in all types of cells. Since V-ATPase dysfunction contributes to the pathogenesis of multiple neurodegenerative diseases, further understanding the mechanisms that regulate the canonical and non-canonical functions of V-ATPase will reveal molecular details of disease process and help assess V-ATPase or molecules related to its regulation as therapeutic targets.
Collapse
Affiliation(s)
- Qiaoyun Song
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang, Hebei Province, 050051, People's Republic of China.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Bo Meng
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Haidong Xu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zixu Mao
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA. .,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
52
|
Magee CP, German CL, Siripathane YH, Curtis PS, Anderson DJ, Wilkins DG, Hanson GR, Fleckenstein AE. 3,4-Methylenedioxypyrovalerone: Neuropharmacological Impact of a Designer Stimulant of Abuse on Monoamine Transporters. J Pharmacol Exp Ther 2020; 374:273-282. [PMID: 32385092 DOI: 10.1124/jpet.119.264895] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/05/2020] [Indexed: 12/13/2022] Open
Abstract
Methylenedioxypyrovalerone (MDPV) is an abused synthetic cathinone, commonly referred to as a "bath salt." Because the dopamine (DA) transporter (DAT) and vesicular monoamine transporter-2 (VMAT-2) are key regulators of both the abuse and neurotoxic potential of structurally and behaviorally related agents, the impact of MDPV on these transporters was investigated. Results revealed that a single in vivo MDPV administration rapidly (within 1 hour) and reversibly increased both rat striatal DAT and VMAT-2 activity, as assessed via [3H]DA uptake in synaptosomes and synaptic vesicles, respectively, prepared from treated rats. There was no evidence of an MDPV-induced increase in plasmalemmal membrane DAT surface expression. Plasma concentrations of MDPV increased dose-dependently as assessed 1 hour after 2.5 and 5.0 mg/kg (s.c.) administration and returned to levels less than 10 ng/ml by 18 hours after 2.5 mg/kg (s.c.). Neither pretreatment with a D1 receptor (SCH23390), a D2 receptor (eticlopride), nor a nicotinic receptor (mecamylamine) antagonist attenuated the MDPV-induced increase in DAT activity. In contrast, eticlopride pretreatment attenuated both the MDPV-induced increase in VMAT-2-mediated DA uptake and an associated increase in cytoplasmic-associated vesicle VMAT-2 immunoreactivity. SCH23390 did not attenuate the MDPV-induced increase in VMAT-2 activity. Repeated MDPV injections did not cause persistent DAergic deficits, as assessed 7 to 8 days later. The impact of MDPV on striatal and hippocampal serotonergic assessments was minimal. Taken together, these data contribute to a growing pharmacological rubric for evaluating the ever-growing list of designer cathinone-related stimulants. The profile of MDPV compared with related psychostimulants is discussed. SIGNIFICANCE STATEMENT: Pharmacological characterization of the synthetic cathinone, 3,4-methylenedioxypyrovalerone (MDPV; commonly referred to as a "bath salt"), is critical for understanding the abuse liability and neurotoxic potential of this and related agents. Accordingly, the impact of MDPV on monoaminergic neurons is described and compared with that of related psychostimulants.
Collapse
Affiliation(s)
- Charlotte P Magee
- School of Dentistry (C.P.M., C.L.G., Y.H.S., P.S.C., G.R.H., A.E.F.), Interdepartmental Program in Neuroscience (C.P.M., C.L.G., G.R.H., A.E.F.), Center for Human Toxicology (D.J.A., D.G.W.), and Department of Pathology (D.G.W.), University of Utah, Salt Lake City, Utah
| | - Christopher L German
- School of Dentistry (C.P.M., C.L.G., Y.H.S., P.S.C., G.R.H., A.E.F.), Interdepartmental Program in Neuroscience (C.P.M., C.L.G., G.R.H., A.E.F.), Center for Human Toxicology (D.J.A., D.G.W.), and Department of Pathology (D.G.W.), University of Utah, Salt Lake City, Utah
| | - Yasmeen H Siripathane
- School of Dentistry (C.P.M., C.L.G., Y.H.S., P.S.C., G.R.H., A.E.F.), Interdepartmental Program in Neuroscience (C.P.M., C.L.G., G.R.H., A.E.F.), Center for Human Toxicology (D.J.A., D.G.W.), and Department of Pathology (D.G.W.), University of Utah, Salt Lake City, Utah
| | - Peter S Curtis
- School of Dentistry (C.P.M., C.L.G., Y.H.S., P.S.C., G.R.H., A.E.F.), Interdepartmental Program in Neuroscience (C.P.M., C.L.G., G.R.H., A.E.F.), Center for Human Toxicology (D.J.A., D.G.W.), and Department of Pathology (D.G.W.), University of Utah, Salt Lake City, Utah
| | - David J Anderson
- School of Dentistry (C.P.M., C.L.G., Y.H.S., P.S.C., G.R.H., A.E.F.), Interdepartmental Program in Neuroscience (C.P.M., C.L.G., G.R.H., A.E.F.), Center for Human Toxicology (D.J.A., D.G.W.), and Department of Pathology (D.G.W.), University of Utah, Salt Lake City, Utah
| | - Diana G Wilkins
- School of Dentistry (C.P.M., C.L.G., Y.H.S., P.S.C., G.R.H., A.E.F.), Interdepartmental Program in Neuroscience (C.P.M., C.L.G., G.R.H., A.E.F.), Center for Human Toxicology (D.J.A., D.G.W.), and Department of Pathology (D.G.W.), University of Utah, Salt Lake City, Utah
| | - Glen R Hanson
- School of Dentistry (C.P.M., C.L.G., Y.H.S., P.S.C., G.R.H., A.E.F.), Interdepartmental Program in Neuroscience (C.P.M., C.L.G., G.R.H., A.E.F.), Center for Human Toxicology (D.J.A., D.G.W.), and Department of Pathology (D.G.W.), University of Utah, Salt Lake City, Utah
| | - Annette E Fleckenstein
- School of Dentistry (C.P.M., C.L.G., Y.H.S., P.S.C., G.R.H., A.E.F.), Interdepartmental Program in Neuroscience (C.P.M., C.L.G., G.R.H., A.E.F.), Center for Human Toxicology (D.J.A., D.G.W.), and Department of Pathology (D.G.W.), University of Utah, Salt Lake City, Utah
| |
Collapse
|
53
|
Ding J, Hu S, Meng Y, Li C, Huang J, He Y, Qiu P. Alpha-Synuclein deficiency ameliorates chronic methamphetamine induced neurodegeneration in mice. Toxicology 2020; 438:152461. [PMID: 32278788 DOI: 10.1016/j.tox.2020.152461] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/13/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023]
Abstract
The α-Synuclein (α-syn) and tau have synergistic effects on neurodegenerative diseases induced by environmental factors or genetic mutation. Thus, we investigated the role of α-syn and tau in neurodegeneration induced by chronic methamphetamine (METH) exposure (1.0∼20.0 mg/kg/d body weight, for 14 consecutive days). Here, we present a mice model with evidences of α-syn and tau participating in toxicology in chronic METH. METH increased α-syn level in the stratum oriens, pyramidal layer, stratum radiatum and stratum moleculare of hippocampal CA1, CA2 and CA3, polymorph layer of hippocampal dentate gyrus (DG), and substantia nigra (SN). The subcellular locations of the upregulated α-syn were mainly found in mitochondria and axons. The METH upregulated α-syn may directly induce mitochondrial damage, myelin sheath destruction, and synaptic failure. Also, the excess α-syn might indirectly promote tau phosphorylation through tau kinase GSK3β and CDK5, leading to microtubule depolymerization and eventually fusion deficit of autophagosome and lysosome. In the in vitro experiment, the autophagic vacuoles failed to fuse with the lysosome. The neuropathology induced by both the direct and indirect effects of α-syn could be alleviated by α-syn knockout. Taking together, these results indicate that the α-syn mediates the neurodegenerative process induced by chronic METH and that reducing α-syn might be a potential approach to protect the toxic effects of METH and also be, to a broader view, of therapeutic value in neurodegenerative diseases.
Collapse
Affiliation(s)
- Jiuyang Ding
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Shanshan Hu
- Good Clinical Practice Center, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, Guizhou, China
| | - Yunle Meng
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Chen Li
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Jian Huang
- School of Forensic Medicine, Kunming Medical University, Kunming 650000, Yunnan, China
| | - Yitong He
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Pingming Qiu
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, Guangdong, China.
| |
Collapse
|
54
|
Temmingh HS, van den Brink W, Howells F, Sibeko G, Stein DJ. Methamphetamine Use and Antipsychotic-related Extrapyramidal Side-effects in Patients with Psychotic Disorders. J Dual Diagn 2020; 16:208-217. [PMID: 31984872 DOI: 10.1080/15504263.2020.1714099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Objective: Extrapyramidal side-effects (EPSE) are frequent in patients treated with antipsychotics and comorbid substance use disorders (SUDs). Methamphetamine has been shown to act as a dopaminergic neurotoxin. We aimed to determine whether EPSE occur more often in patients with psychotic disorders and co-occurring methamphetamine (MA) use disorders, and we examined the relationship between MA use, antipsychotic type, dose and EPSE. Methods: This study was a secondary analysis of data from three separate primary studies. Across all studies, psychiatric and SUD diagnoses were determined using the SCID-I for DSM-IV. EPSE were determined using the Simpson-Angus Scale (SAS) for Parkinsonism, the Barnes Akathisia Rating scale (BARS), and the Abnormal Involuntary Movement Scale (AIMS) for tardive dyskinesia. Participants were classified as having any EPSE if they scored above the cutoff on any of the EPSE scales (SAS, BARS, AIMS). We analyzed data using multivariable logistic regression analysis. Results: The sample included 102 patients with non-affective or affective psychotic disorders. Of the total sample, 65.7% were male, 54.9% had schizophrenia spectrum disorders, 20.5% bipolar type I disorder with psychotic features, 11.7% schizoaffective disorder and 12.7% had substance-induced psychosis. A diagnosis of a methamphetamine use disorder (abuse or dependence) was present in 25.5% of participants. EPSE occurred in 38.2% of patients and were significantly associated with MA use in the unadjusted and adjusted analysis, ORadj = 4.01, 95% CI [1.07, 14.98], p = .039. Patients with MA dependence and MA use >3 years were significantly more likely to have EPSE. We found a significant interaction effect between MA use disorders and standardized antipsychotic dose on the occurrence of EPSE, ORadj = 1.01, 95% CI [1.00, 1.01], p = .042, with MA users having a disproportionally higher likelihood of having EPSE compared to MA non-users as antipsychotic dosage increased. There were no significant associations of EPSE with comorbid alcohol, cannabis, or methaqualone use disorders. Conclusions: Patients with a MA use disorder were significantly more likely to have EPSE with evidence for a dose-response effect. Clinicians should carefully titrate antipsychotic dosage from lower to higher doses to avoid EPSE in patients with MA use disorders.
Collapse
Affiliation(s)
- Henk S Temmingh
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Wim van den Brink
- Department of Psychiatry, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Fleur Howells
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa.,Psychiatry and Mental Health, Neuroscience Institute of South Africa, University of Cape Town, Cape Town, South Africa
| | - Goodman Sibeko
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Dan J Stein
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa.,Psychiatry and Mental Health, Neuroscience Institute of South Africa, University of Cape Town, Cape Town, South Africa.,South African MRC Unit on Risk & Resilience in Mental Disorders, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
55
|
Differential Responses of LINE-1 in the Dentate Gyrus, Striatum and Prefrontal Cortex to Chronic Neurotoxic Methamphetamine: A Study in Rat Brain. Genes (Basel) 2020; 11:genes11040364. [PMID: 32231019 PMCID: PMC7230251 DOI: 10.3390/genes11040364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/19/2020] [Accepted: 03/25/2020] [Indexed: 11/17/2022] Open
Abstract
Methamphetamine (METH) is a widely abused psychostimulant with the potential to cause a broad range of severe cognitive deficits as well as neurobehavioral abnormalities when abused chronically, particularly at high doses. Cognitive deficits are related to METH neurotoxicity in the striatum and hippocampus. The activation of transposable Long INterspersed Nuclear Element 1 (LINE-1) is associated with several neurological diseases and drug abuse, but there are very limited data regarding the effects of high-dose METH on the activity of LINE-1 in the adult brain. Using real-time quantitative PCR, the present study demonstrates that the chronic administration of neurotoxic METH doses results in the increased expression of LINE-1-encoded Open Reading Frame 1 (ORF-1) in rat striatum shortly after the last dose of the drug and decreased ORF-1 expression during METH withdrawal, with dentate gyrus potentially developing "tolerance" to these METH effects. LINE-1 activation may be a new factor mediating the neurotoxic effects of chronic METH in the striatum and, therefore, a new drug target against METH-induced psychomotor impairments in chronic METH users.
Collapse
|
56
|
Costa G, De Luca MA, Piras G, Marongiu J, Fattore L, Simola N. Neuronal and peripheral damages induced by synthetic psychoactive substances: an update of recent findings from human and animal studies. Neural Regen Res 2020; 15:802-816. [PMID: 31719240 PMCID: PMC6990793 DOI: 10.4103/1673-5374.268895] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Preclinical and clinical studies indicate that synthetic psychoactive substances, in addition to having abuse potential, may elicit toxic effects of varying severity at the peripheral and central levels. Nowadays, toxicity induced by synthetic psychoactive substances poses a serious harm for health, since recreational use of these substances is on the rise among young and adult people. The present review summarizes recent findings on the peripheral and central toxicity elicited by “old” and “new” synthetic psychoactive substances in humans and experimental animals, focusing on amphetamine derivatives, hallucinogen and dissociative drugs and synthetic cannabinoids.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Maria Antonietta De Luca
- Department of Biomedical Sciences; National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| | - Gessica Piras
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Jacopo Marongiu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Liana Fattore
- National Research Council of Italy, Institute of Neuroscience, Cagliari, Italy
| | - Nicola Simola
- Department of Biomedical Sciences; National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| |
Collapse
|
57
|
Nolan RA, Reeb KL, Rong Y, Matt SM, Johnson HS, Runner K, Gaskill PJ. Dopamine activates NF-κB and primes the NLRP3 inflammasome in primary human macrophages. Brain Behav Immun Health 2019; 2. [PMID: 33665636 PMCID: PMC7929492 DOI: 10.1016/j.bbih.2019.100030] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Induction of innate immune genes in the brain is thought to be a major factor in the development of addiction to substances of abuse. As the major component of the innate immune system in the brain, aberrant activation of myeloid cells such as macrophages and microglia due to substance use may mediate neuroinflammation and contribute to the development of addiction. All addictive drugs modulate the dopaminergic system and our previous studies have identified dopamine as a pro-inflammatory modulator of macrophage function. However, the mechanism that mediates this effect is currently unknown. Inflammatory activation of macrophages and induction of cytokine production is often mediated by the transcription factor NF-κB, and prior studies have shown that dopamine can modulate NF-κB activity in T-cells and other non-immune cell lines. Here we demonstrated that dopamine can activate NF-κB in primary human macrophages, resulting in the induction of its downstream targets including the NLRP3 inflammasome and the inflammatory cytokine IL-1β. These data also indicate that dopamine primes but does not activate the NLRP3 inflammasome in human macrophages. Activation of NF-κB was required for dopamine-mediated increases in IL-1β, as an inhibitor of NF-κB was able to abrogate the effects of dopamine on production of these cytokines. Connecting an increase in extracellular dopamine to NF-κB activation and inflammation suggests specific intracellular targets that could be used to ameliorate the inflammatory impact of dopamine in neuroinflammatory conditions associated with myeloid cell activation such as addiction. Dopamine exposure primes, but does not activate the NLRP3 inflammasome. Inflammasome priming can be mediated, at least partially, by a dopamine-induced increase in the activation and nuclear translocation of NF-κB in primary human macrophages. Dopamine additively increases the impact of cytomegalovirus on NF-κB activation in macrophages. Dopamine priming increases IL-1β release in response to inflammasome activation.
Collapse
Affiliation(s)
- R A Nolan
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - K L Reeb
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - Y Rong
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - S M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - H S Johnson
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - K Runner
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - P J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| |
Collapse
|
58
|
Darvishzadeh H, Mirzaee M, Jahani Y, Sharifi H. Age of Onset of Methamphetamine Consumption among the Iranian Youth Aged 19-29: A Cross-sectional Study. ADDICTION & HEALTH 2019; 11:138-147. [PMID: 31839911 PMCID: PMC6904974 DOI: 10.22122/ahj.v11i3.231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Background Around the world, one of the main concerns and risky behaviors among youths is methamphetamine consumption. Since the age of onset of methamphetamine use is decreasing, therefore, it is important to identify the effective factors on the age of onset of methamphetamine consumption. In the present study, factors affecting the age of onset of methamphetamine consumption was studied in Iranian youths aged 19-29 years. Methods In this cross-sectional nationwide study, individuals aged 19-29 years were selected through multistage cluster sampling and convenience sampling method from 13 provinces in Iran. People completed the questionnaire pertaining to knowledge, attitude, and history of methamphetamine consumption. In order to investigate the factors effective on the age of onset of methamphetamine consumption, we used the Weibull parametric model for data with doubly censored characteristic. Findings 3246 people participated in this study, half of whom were men and mostly single (60.3%), university student or graduate (50.2%), and unemployed (58.1%). Nearly 6% of participants have ever used methamphetamine. Mean and standard deviation (SD) of age of onset of methamphetamine use was 20.3 ± 3.3 years. Data analysis indicated that the variables of gender, marital status, education, knowing a methamphetamine consumer, knowing an ecstasy consumer, ecstasy consumption, illegitimate sex, attitude towards methamphetamine, and age group were the factors affecting the age of onset of methamphetamine consumption. Conclusion The results can contribute to the policy-makers to take the necessary interventions on the factors affecting the age of onset of methamphetamine consumption to reduce the methamphetamine consumption, especially in the critical young ages.
Collapse
Affiliation(s)
- Hadiseh Darvishzadeh
- HIV/STI Surveillance Research Center, and WHO Collaborating Center for HIV Surveillance, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Moghaddameh Mirzaee
- Modeling in Health Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Yunes Jahani
- Modeling in Health Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Sharifi
- HIV/STI Surveillance Research Center, and WHO Collaborating Center for HIV Surveillance, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
59
|
Foroughi K, Jahanbani S, Khaksari M, Shayannia A. Obestatin attenuated methamphetamine-induced PC12 cells neurotoxicity via inhibiting autophagy and apoptosis. Hum Exp Toxicol 2019; 39:301-310. [PMID: 31726888 DOI: 10.1177/0960327119886036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Methamphetamine (METH) is an illicit dopaminergic neurotoxin and is an extremely addictive psychostimulant drug that influences monoamine neurotransmitter system of the brain and is responsible for enhancing energy and satisfaction and feelings of alertness. Long-lasting exposure to METH causes psychosis and increases the risk of Parkinson's disease. Studies have revealed that obestatin (OB) is a novel endogenous ligand, which may have neuroprotective effects. Hence, we hypothesized that OB might appropriately limit METH-induced neurotoxicity via the control of apoptotis and autophagy. In the current study, PC12 cells were exposed to both METH (0.5, 1, 2, 3, 4, and 6 mmol/L) and pretreatment OB (1, 10, 100, and 200 nmol/L) in vitro for 24 h to determine appropriate dose, and then downstream pathways were measured to investigate apoptosis and autophagy. The results have shown that OB reduced the apoptotic response post-METH exposure in PC12 cells by developing cell viability and diminishing apoptotic rates. Furthermore, the study has exhibited OB decreased gene expression of Beclin-1 by real-time polymerase chain reaction and LC3-II by Western blotting in METH-induced PC12 cells, which demonstrated that autophagy is reduced. The study is proposed that OB is useful in reducing oxidative stress, which may also play an essential role in the regulation of METH-triggered apoptotic response. So these data indicate that OB could potentially alleviate METH-induced neurotoxicity via the reduction of apoptotic and autophagy responses.
Collapse
Affiliation(s)
- K Foroughi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - S Jahanbani
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - M Khaksari
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - A Shayannia
- Department of Medical Biotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
60
|
Lappin JM, Sara GE. Psychostimulant use and the brain. Addiction 2019; 114:2065-2077. [PMID: 31321819 DOI: 10.1111/add.14708] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/01/2019] [Accepted: 06/03/2019] [Indexed: 12/18/2022]
Abstract
Psychostimulant users are typically young adults. We have conducted a narrative review of neuropsychiatric harms associated with the psychostimulants methamphetamine/amphetamine, cocaine and 3,4-methylenedioxymethamphetamine (MDMA), focusing on epidemiological factors, common clinical presentations, underlying causal mechanisms and treatment options. The major neuropsychiatric harms of psychostimulant use are stroke, neurocognitive impairment, Parkinson's disease, seizures and psychotic illness. These arise through a combination of acute monoamine release, longer-term neurotransmitter effects and indirect effects. These effects are moderated by factors in the individual and in the pattern of substance use. Neuropsychiatric harms associated with psychostimulant use can thus lead to severe long-term impairment.
Collapse
Affiliation(s)
- Julia M Lappin
- National Drug and Alcohol Research Centre (NDARC), University of New South Wales, Sydney, Australia.,School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Grant E Sara
- InforMH, NSW Ministry of Health, North Ryde, NSW, Australia.,Northern Clinical School, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
61
|
Illés A, Balicza P, Molnár V, Bencsik R, Szilvási I, Molnar MJ. Dynamic interaction of genetic risk factors and cocaine abuse in the background of Parkinsonism - a case report. BMC Neurol 2019; 19:260. [PMID: 31660902 PMCID: PMC6816197 DOI: 10.1186/s12883-019-1496-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 10/13/2019] [Indexed: 11/16/2022] Open
Abstract
Background Parkinsonism is a complex multifactorial neurodegenerative disorder, in which genetic and environmental risk factors may both play a role. Among environmental risk factors cocaine was earlier ambiguously linked to Parkinsonism. Former single case reports described Parkinsonism in chronic cocaine users, but an epidemiological study did not confirm an increased risk of Parkinson’s disease. Here we report a patient, who developed Parkinsonism in young age after chronic cocaine use, in whom a homozygous LRRK2 risk variant was also detected. Case presentation The patient was investigated because of hand tremor, which started after a 1.5-year period of cocaine abuse. Neurological examination suggested Parkinsonism, and asymmetrical pathology was confirmed by the dopamine transporter imaging study. The genetic investigations revealed a homozygous risk allele in the LRRK2 gene. After a period of cocaine abstinence, the patient’s symptoms spontaneously regressed, and the dopamine transporter imaging also returned to near-normal. Conclusions This case report suggests that cocaine abuse indeed might be linked to secondary Parkinsonism and serves as an example of a potential gene-environmental interaction between the detected LRRK2 risk variant and cocaine abuse. The reversible nature of the DaTscan pathology is a unique feature of this case, and needs further evaluation, whether this is incidental or can be a feature of cocaine related Parkinsonism.
Collapse
Affiliation(s)
- Anett Illés
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Péter Balicza
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Viktor Molnár
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Renáta Bencsik
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - István Szilvási
- Department of Nuclear Medicine, Hungarian Defence Force Medical Center, Budapest, Hungary
| | - Maria Judit Molnar
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
62
|
Blaker AL, Moore ER, Yamamoto BK. Serial exposure to ethanol drinking and methamphetamine enhances glutamate excitotoxicity. J Neurochem 2019; 151:749-763. [PMID: 31478210 DOI: 10.1111/jnc.14861] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/05/2019] [Accepted: 08/26/2019] [Indexed: 12/22/2022]
Abstract
A significant comorbidity exists between alcohol and methamphetamine (Meth) abuse but the neurochemical consequences of this co-abuse are unknown. Alcohol and Meth independently and differentially affect glutamatergic transmission but the unique effects of their serial exposure on glutamate signaling in mediating damage to dopamine neurons are unknown. Sprague-Dawley rats had intermittent voluntary access to 10% ethanol (EtOH) every other day and water over 28 days and were then administered a binge injection regimen of Meth or saline. EtOH drinking decreased the glutamate aspartate transporter and increased basal extracellular concentrations of glutamate within the striatum when measured after the last day of drinking. Ceftriaxone is known to increase the expression and/or activity of glutamate transporters in the brain and prevented both the decreases in glutamate aspartate transporter and the increases in basal extracellular glutamate when administered during EtOH drinking. EtOH drinking also exacerbated the acute increases in extracellular glutamate observed upon Meth exposure, the subsequent increases in spectrin proteolysis, and the long-term decreases in dopamine content in the striatum, all of which were attenuated by ceftriaxone administration during EtOH drinking only. These results implicate EtOH-induced increases in extracellular glutamate and corresponding decreases in glutamate uptake as mechanisms that contribute to the vulnerability produced by EtOH drinking and the unique neurotoxicity observed after serial exposure to Meth that is not observed with either drug alone. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Amanda L Blaker
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Elizabeth R Moore
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Bryan K Yamamoto
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
63
|
Asser A, Kõks S, Soomets U, Terasmaa A, Sauk M, Eltermaa M, Piip P, Ubhayasekera K, Bergquist J, Taba P. Acute effects of methcathinone and manganese in mice: A dose response study. Heliyon 2019; 5:e02475. [PMID: 31687570 PMCID: PMC6819833 DOI: 10.1016/j.heliyon.2019.e02475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 06/15/2019] [Accepted: 09/11/2019] [Indexed: 11/24/2022] Open
Abstract
An intravenously injectable illicit drug made by mixing pseudoephedrine, potassium permanganate, vinegar and water, yielding methcathinone (Mcat) and manganese (Mn), induces an extrapyramidal syndrome with parkinsonism, dystonia, gait and balance disorders similar to manganism. Although the cause of the syndrome is largely attributed to Mn, the interaction of the drug's individual components is not known and the role of Mcat is possibly underestimated. Aim of the present study was to analyze dose-dependent behavioral effects of the mixture and its two main active components Mcat and Mn in an acute setting and determine the lethal doses of each substance. Three groups of C57BL/6 mice were injected intraperitoneally with (1) the drug mixture containing 10, 25, 50, 100 or 150 mg of Mcat and respectively 1.6, 3.8, 6.9, 17.1 and 22.6 mg of Mn per kilogram of body weight; (2) 10, 25, 50, 100, 150, 200 or 300 mg of racemic Mcat/kg of body weight; (3) MnCl2 10, 25 or 50 mg/kg of body weight. Locomotor activity of the animals, various signs and time of death were recorded. Lower doses (10 and 25 mg/kg) of Mcat had a clear motor activity stimulating effect and this was clearly dose-dependent. High doses of Mcat produced epileptic seizures in 74% of the animals and became lethal with the highest doses. Similarly, the mixture had a clear dose-dependent stimulating effect and the higher doses became lethal. The LD50 of the pseudoephedrine mixture was 110.2 mg of Mcat/kg and for pure Mcat 201.7 mg/kg. Mn did not prove to be lethal in doses up to 50 mg/kg, but had a strong dose dependent inhibitory effect on the animals’ behavior. Our data reveal that both Mn and Mcat have a significant role in the toxicity of the mixture.
Collapse
Affiliation(s)
- Andres Asser
- Department of Neurology and Neurosurgery, University of Tartu, Puusepa 8, Tartu, 51014, Estonia
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, University of Western Australia, Perth, WA, Australia
| | - Ursel Soomets
- Department of Biochemistry, University of Tartu, Ravila 19, Tartu, 50411, Estonia
| | - Anton Terasmaa
- Department of Physiology, University of Tartu, Ravila 19, Tartu, 50411, Estonia
| | - Martin Sauk
- Institute of Molecular and Cell Biology, University of Tartu, Riia 23, Tartu, 51010, Estonia
| | - Mall Eltermaa
- Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, Tartu, 50411, Estonia
| | - Piret Piip
- Department of Neurology and Neurosurgery, University of Tartu, Puusepa 8, Tartu, 51014, Estonia
| | - Kumari Ubhayasekera
- Department of Chemistry, Biomedical Center, Uppsala University, SE-751 24, Uppsala, Sweden
| | - Jonas Bergquist
- Department of Chemistry, Biomedical Center, Uppsala University, SE-751 24, Uppsala, Sweden
| | - Pille Taba
- Department of Neurology and Neurosurgery, University of Tartu, Puusepa 8, Tartu, 51014, Estonia
| |
Collapse
|
64
|
Biagioni F, Ferese R, Limanaqi F, Madonna M, Lenzi P, Gambardella S, Fornai F. Methamphetamine persistently increases alpha-synuclein and suppresses gene promoter methylation within striatal neurons. Brain Res 2019; 1719:157-175. [DOI: 10.1016/j.brainres.2019.05.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 12/20/2022]
|
65
|
Sun L, Lian Y, Ding J, Meng Y, Li C, Chen L, Qiu P. The role of chaperone-mediated autophagy in neurotoxicity induced by alpha-synuclein after methamphetamine exposure. Brain Behav 2019; 9:e01352. [PMID: 31286692 PMCID: PMC6710200 DOI: 10.1002/brb3.1352] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/28/2019] [Accepted: 06/02/2019] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Chaperone-mediated autophagy (CMA) is an autophagy-lysosome pathway (ALP) that is different from the other two lysosomal pathways, namely, macroautophagy and microautophagy, and can selectively degrade cytosolic proteins in lysosomes without vesicle formation. CMA activity declines in neurodegenerative diseases such as Parkinson's disease, and similar neurotoxicity can occur after methamphetamine (METH) treatment. The relationship between CMA and METH-induced neurotoxicity is not clear. METHODS We detected changes in the chaperone protein Hsc70 and the lysosomal surface receptor Lamp-2a after METH treatment and then regulated these two proteins by small interfering RNA and DNA plasmid transfection to investigate how CMA influences METH-induced neurotoxicity. RESULTS We found that CMA activity is decreased after METH exposure in neurons and downregulated Lamp-2a can aggravate the neurotoxicity induced by α-Syn after METH exposure and that Hsc70 overexpression can relieve the abnormal levels of alpha-synuclein and its aggregate forms and the increase in cell apoptosis induced by METH. CONCLUSIONS The results provide in vivo evidence for CMA plays a pivotal role in METH-induced neurotoxicity, and upregulation of Hsc70 expression significantly protects neuronal cells against METH-induced toxicity. This research may pave the way for potential therapeutic approaches targeting CMA for METH abuse and neurodegenerative disorders.
Collapse
Affiliation(s)
- Leping Sun
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Yongling Lian
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Jiuyang Ding
- School of Forensic Medicine, Southern Medical University, Guangzhou, China.,Department of Anatomy, Zunyi Medical College, Zunyi, China
| | - Yunle Meng
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Chen Li
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Ling Chen
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Pingming Qiu
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
66
|
Huang E, Huang H, Guan T, Liu C, Qu D, Xu Y, Yang J, Yan L, Xiong Y, Liang T, Wang Q, Chen L. Involvement of C/EBPβ-related signaling pathway in methamphetamine-induced neuronal autophagy and apoptosis. Toxicol Lett 2019; 312:11-21. [PMID: 31059759 DOI: 10.1016/j.toxlet.2019.05.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 04/29/2019] [Accepted: 05/02/2019] [Indexed: 12/24/2022]
Abstract
Methamphetamine (METH) is a widely abused illicit psychoactive drug. Our previous study has shown that CCAAT-enhancer binding protein β (C/EBPβ) is an important regulator in METH-induced neuronal autophagy and apoptosis. However, the detailed molecular mechanisms underlying this process remain poorly understood. Previous studies have demonstrated that DNA damage-inducible transcript 4 (DDIT4), Trib3 (tribbles pseudo kinase 3), alpha-synuclein (α-syn) are involved in METH-induced dopaminergic neurotoxicity. We hypothesized that C/EBPβ is involved in METH-induced DDIT4-mediated neuronal autophagy and Trib3-mediated neuronal apoptosis. We tested our hypothesis by examining the effects of silencing C/EBPβ, DDIT4, Trib3 or α-syn with small interfering ribonucleic acid (siRNA) on METH-induced autophagy and apoptosis in the human neuroblastoma SH-SY5Y cells. We also measured the levels of phosphorylated tuberous sclerosis complex 2 (TSC2) protein and Parkin protein level in SH-SY5Y cells. Furthermore, we demonstrated the effect of silencing C/EBPβ on METH-caused neurotoxicity in the striatum of rats by injecting LV-shC/EBPβ lentivirus using a stereotaxic positioning system. The results showed that METH exposure increased C/EBPβ, DDIT4 protein expression. Elevated DDIT4 expression raised up p-TSC2/TSC2 protein expression ratio, inhibited mTOR signaling pathway, activating cell autophagy. We also found that METH exposure increased the expression of Trib3, α-syn, decreased the Parkin protein expression. Lowering levels of Parkin raised up α-syn expression, which initiated mitochondrial apoptosis by down-regulating anti-apoptotic Bcl-2, followed by up-regulation of pro-apoptotic Bax, resulting in translocation of cytochrome c (cyto c), an apoptogenic factor, from the mitochondria to cytoplasm and activation of caspase-dependent pathways. These findings were supported by data showing METH-induced autophagy and apoptosis was significantly inhibited by silencing C/EBPβ, DDIT4, Trib3 or α-syn, or by Parkin over-expression. Based on the present data, a novel of mechanism on METH-induced cell toxicity is proposed, METH exposure increased C/EBPβ protein expression, triggered DDIT4/TSC2/mTOR signaling pathway, and evoked Trib3/Parkin/α-syn-related mitochondrial apoptotic signaling pathway. Collectively, these results suggest that C/EBPβ plays an important role in METH-triggered autophagy and apoptosis and it may be a potential target for therapeutics in METH-caused neurotoxicity.
Collapse
Affiliation(s)
- Enping Huang
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Hongyan Huang
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Tianshan Guan
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Chao Liu
- Guangzhou Forensic Science Institute, Guangzhou 510030, People's Republic of China
| | - Dong Qu
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Yue Xu
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jiao Yang
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Lei Yan
- School of Basic Medicine Science, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Yahui Xiong
- Nanfang Hospital, Southern Medical University, The First Clinical Medicine School, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Ting Liang
- Nanfang Hospital, Southern Medical University, The First Clinical Medicine School, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Qi Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, People's Republic of China.
| | - Ling Chen
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, People's Republic of China.
| |
Collapse
|
67
|
Ball N, Teo WP, Chandra S, Chapman J. Parkinson's Disease and the Environment. Front Neurol 2019; 10:218. [PMID: 30941085 PMCID: PMC6433887 DOI: 10.3389/fneur.2019.00218] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 02/20/2019] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is a heterogeneous neurodegenerative disorder that affects an estimated 10 million sufferers worldwide. The two forms of PD include familial and sporadic, and while the etiology of PD is still largely unknown, the condition is likely to be multifactorial with genetic and environmental factors contributing to disease genesis. Diagnosis of the condition is attained through the observation of cardinal clinical manifestations including resting tremor, muscle rigidity, slowness or loss of movement, and postural instability. Unfortunately, by the time these features become apparent extensive neurological damage has already occurred. A cure for PD has not been identified and the current therapy options are pharmaceutical- and/or surgical-based interventions to treat condition symptoms. There is no specific test for PD and most diagnoses are confirmed by a combination of clinical symptoms and positive responses to dopaminergic drug therapies. The prevalence and incidence of PD vary worldwide influenced by several factors such as age, gender, ethnicity, genetic susceptibilities, and environmental exposures. Here, we will present environmental factors implicated in sporadic PD onset. By understanding the mechanisms in which environmental factors interact with, and affect the brain we can stride toward finding the underlying cause(s) of PD.
Collapse
Affiliation(s)
- Nicole Ball
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD, Australia
| | - Wei-Peng Teo
- School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition, Deakin University, Melbourne, VIC, Australia.,Physical Education and Sports Science Academic Group, National Institute of Education, Nanyang Technological University, Singapore, Singapore
| | - Shaneel Chandra
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD, Australia
| | - James Chapman
- School of Science, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
68
|
Uhl GR. Dopamine compartmentalization, selective dopaminergic vulnerabilities in Parkinson's disease and therapeutic opportunities. Ann Clin Transl Neurol 2019; 6:406-415. [PMID: 30847375 PMCID: PMC6389739 DOI: 10.1002/acn3.707] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/29/2018] [Accepted: 11/05/2018] [Indexed: 12/13/2022] Open
Abstract
Progressive depletion of selected dopamine neurons is central to much Parkinson's disease (PD) disability. Although symptomatic treatments can ameliorate the disabilities that this neuronal depletion causes, no current strategy is documented to slow these losses. There is substantial evidence that dopamine in intracytoplasmic/extravesicular neuronal compartments can be toxic. Here, I review evidence that supports roles for dopamine compartmentalization, mediated largely by serial actions of plasma membrane SLC6A3/DAT and vesicular SLC18A2/VMAT2 transporters, in the selective patterns of dopamine neuronal loss found in PD brains. This compartmentalization hypothesis for the dopamine cell type specificity of PD lesions nominates available drugs for amelioration of damage arising from miscompartmentalized dopamine and raises cautions in using other drugs.
Collapse
Affiliation(s)
- George R. Uhl
- Neurology and Research ServicesNew Mexico VA HealthCare SystemAlbuquerqueNew Mexico87108
- Biomedical Research Institute of New MexicoAlbuquerqueNew Mexico87108
- Departments of Neurology, Neuroscience and Molecular Genetics and MicrobiologyUniversity of New MexicoAlbuquerqueNew Mexico87108
- Departments of Neurology, Neuroscience and Mental HealthJohns Hopkins Medical InstitutionsBaltimoreMaryland21287
| |
Collapse
|
69
|
Shin EJ, Dang DK, Hwang YG, Tran HQ, Sharma N, Jeong JH, Jang CG, Nah SY, Nabeshima T, Yoneda Y, Cadet JL, Kim HC. Significance of protein kinase C in the neuropsychotoxicity induced by methamphetamine-like psychostimulants. Neurochem Int 2019; 124:162-170. [PMID: 30654115 DOI: 10.1016/j.neuint.2019.01.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/27/2018] [Accepted: 01/14/2019] [Indexed: 02/06/2023]
Abstract
The abuse of methamphetamine (MA), an amphetamine (AMPH)-type stimulant, has been demonstrated to be associated with various neuropsychotoxicity, including memory impairment, psychiatric morbidity, and dopaminergic toxicity. Compelling evidence from preclinical studies has indicated that protein kinase C (PKC), a large family of serine/threonine protein kinases, plays an important role in MA-induced neuropsychotoxicity. PKC-mediated N-terminal phosphorylation of dopamine transporter has been identified as one of the prerequisites for MA-induced synaptic dopamine release. Consistently, it has been shown that PKC is involved in MA (or AMPH)-induced memory impairment and mania-like behaviors as well as MA drug dependence. Direct or indirect regulation of factors related to neuronal plasticity seemed to be critical for these actions of PKC. In addition, PKC-mediated mitochondrial dysfunction, oxidative stress or impaired antioxidant defense system has been suggested to play a role in psychiatric and cognitive disturbance induced by MA (or AMPH). In MA-induced dopaminergic toxicity, particularly PKCδ has been shown to trigger oxidative stress, mitochondrial dysfunction, pro-apoptotic changes, and neuroinflammation. Importantly, PKCδ may be a key mediator in the positive feedback loop composed of these detrimental events to potentiate MA-induced dopaminergic toxicity. This review outlines the role of PKC and its individual isozymes in MA-induced neuropsychotoxicity. Better understanding on the molecular mechanism of PKCs might provide a great insight for the development of potential therapeutic or preventive candidates for MA (or AMPH)-associated neuropsychotoxicity.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Duy-Khanh Dang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Young Gwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Hai-Quyen Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake 470-1192, Japan
| | - Yukio Yoneda
- Section of Prophylactic Pharmacology, Kanazawa University Venture Business Laboratory, Kanazawa, Ishikawa 920-1192, Japan
| | - Jean Lud Cadet
- NIDA Intramural Program, Molecular Neuropsychiatry Research Branch, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea.
| |
Collapse
|
70
|
Qiao HH, Zhu LN, Wang Y, Hui JL, Xie WB, Liu C, Chen L, Qiu PM. Implications of alpha-synuclein nitration at tyrosine 39 in methamphetamine-induced neurotoxicity in vitro and in vivo. Neural Regen Res 2019; 14:319-327. [PMID: 30531016 PMCID: PMC6301162 DOI: 10.4103/1673-5374.244795] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Methamphetamine is an amphetamine-type psychostimulant that can damage dopaminergic neurons and cause characteristic pathological changes similar to neurodegenerative diseases such as Parkinson’s disease. However, its specific mechanism of action is still unclear. In the present study, we established a Parkinson’s disease pathology model by exposing SH-SY5Y cells and C57BL/6J mice to methamphetamine. In vitro experiments were performed with 0, 0.5, 1.0, 1.5, 2.0 or 2.5 mM methamphetamine for 24 hours or 2.0 mM methamphetamine for 0-, 2-, 4-, 8-, 16-, and 24-hour culture of SH-SY5Y cells. Additional experimental groups of SH-SY5Y cells were administered a nitric oxide inhibitor, 0.1 mM N-nitro-L-arginine, 1 hour before exposure to 2.0 mM methamphetamine for 24 hours. In vivo experiments: C57BL/6J mice were intraperitoneally injected with N-nitro-L-arginine (8 mg/kg), eight times, at intervals of 12 hours. Methamphetamine 15 mg/kg was intraperitoneally injected eight times, at intervals of 12 hours, but 0.5-hour after each N-nitro-L-arginine injection in the combined group. Western blot assay was used to determine the expression of nitric oxide synthase, α-synuclein (α-Syn), 5G4, nitrated α-synuclein at the residue Tyr39 (nT39 α-Syn), cleaved caspase-3, and cleaved poly ADP-ribose polymerase (PARP) in cells and mouse brain tissue. Immunofluorescence staining was conducted to measure the positive reaction of NeuN, nT39 α-Syn and 5G4. Enzyme linked immunosorbent assay was performed to determine the dopamine levels in the mouse brain. After methamphetamine exposure, α-Syn expression increased; the aggregation of α-Syn 5G4 increased; nT39 α-Syn, nitric oxide synthase, cleaved caspase-3, and cleaved PARP expression increased in the cultures of SH-SY5Y cells and in the brains of C57BL/6J mice; and dopamine levels were reduced in the mouse brain. These changes were markedly reduced when N-nitro-L-arginine was administered with methamphetamine in both SH-SY5Y cells and C57BL/6J mice. These results suggest that nT39 α-Syn aggregation is involved in methamphetamine neurotoxicity.
Collapse
Affiliation(s)
- Hong-Hua Qiao
- School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lin-Nan Zhu
- School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yue Wang
- Kingmed Institute for Forensic Science, Guangzhou, Guangdong Province, China
| | - Jia-Liang Hui
- First Clinical Medicine College, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Wei-Bing Xie
- School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chao Liu
- Guangzhou Forensic Science Institute, Guangdong Province Key Laboratory of Forensic Genetics, Guangzhou, Guangdong Province, China
| | - Ling Chen
- School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ping-Ming Qiu
- School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
71
|
Schmitz F, Chao MV, Wyse ATS. Methylphenidate alters Akt-mTOR signaling in rat pheochromocytoma cells. Int J Dev Neurosci 2018; 73:10-18. [PMID: 30578823 DOI: 10.1016/j.ijdevneu.2018.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/13/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022] Open
Abstract
The exponential increase in methylphenidate (MPH) prescriptions in recent years has worried researchers about its misuse among individuals who do not meet the full diagnostic criteria for attention-deficit/hyperactivity disorder (ADHD) such as young children and students in search of cognitive improvement or for recreational reasons. The action of MPH is based mainly on inhibition of dopamine transporter, but the complete cellular effects are still unknown. Based upon prior studies, we attempted to determine whether the treatment with MPH (1μM) influences protein kinase B-mammalian target of rapamycin complex 1 signaling pathways (Akt-mTOR), including translation repressor protein (4E-BP1) and mitogen activated protein kinase (S6K), in rat pheochromocytoma cells (PC12), a well characterized cellular model, in a long or short term. MPH effects on the Akt substrates [cAMP response element-binding protein (CREB), forkhead box protein O1 (FoxO1), and glycogen synthase kinase 3 beta (GSK-3β)] were also evaluated. Whereas short term MPH treatment decreased the pAkt/Akt, pmTOR/mTOR and pS6K/S6K ratios, as well as pFoxO1 immunocontent in PC12 cells, long term treatment increased pAkt/Akt, pmTOR/mTOR and pGSK-3β/GSK-3β ratio. Phosphorylation levels of 4E-BP1 were decreased at 15 and 30 min and increased at 1 and 6 h by MPH. pCREB/CREB ratio was decreased. This study shows that the Akt-mTOR pathway, as well as other important Akt substrates which have been described as important regulators of protein synthesis, as well as being implicated in cellular survival, synaptic plasticity and memory consolidation, was affected by MPH in PC12 cells, representing an important step in exploring the MPH effects.
Collapse
Affiliation(s)
- Felipe Schmitz
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, USA; Laboratory of Neuroprotection and Metabolic Diseases, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Moses V Chao
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, USA
| | - Angela T S Wyse
- Laboratory of Neuroprotection and Metabolic Diseases, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Programa de Pós‑Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
72
|
Curtin K, Fleckenstein AE, Keeshin BR, Yurgelun-Todd DA, Renshaw PF, Smith KR, Hanson GR. Increased risk of diseases of the basal ganglia and cerebellum in patients with a history of attention-deficit/hyperactivity disorder. Neuropsychopharmacology 2018; 43:2548-2555. [PMID: 30209407 PMCID: PMC6224615 DOI: 10.1038/s41386-018-0207-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 08/10/2018] [Accepted: 08/28/2018] [Indexed: 12/13/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is marked by an ongoing pattern of inattention and/or hyperactivity and involves dysregulated dopaminergic pathways. Dopaminergic agents (i.e., amphetamine and methylphenidate) are thus prescribed to treat ADHD. As little is known regarding long-term consequences of either ADHD or its treatment, the objective of this study was to determine if either alters the risk of diseases of the basal ganglia and cerebellum, including Parkinson's disease. Statewide medical records from 1996 to 2016 were retrieved from the Utah Population Database to conduct a retrospective cohort study. Participants included ADHD patients (International Classification of Diseases, 9th revision (ICD-9) diagnosis codes 314.0-314.2, 314.8, 314.9) and 5:1 random sex-matched and age-matched subjects with no ADHD diagnosis history. Both patients and non-ADHD subjects met the following eligibility criteria: (1) no prior diagnosis of Parkinson's disease, secondary parkinsonism, basal ganglia disease, or essential tremor (ICD-9 codes 332.0, 332.1, 333.0, 333.1), (2) born in 1950 or later and age ≥20 years at last follow-up, and (3) no history of substance abuse (illicit drugs or alcohol). Outcomes were measured as time to diagnosis of diseases of the basal ganglia and cerebellum, death, or study-end. A Cox model incorporating a competing risk of death was used to provide hazard ratio estimates. Patients with ADHD (N = 31,769) had a 2.4-fold increased risk of basal ganglia and cerebellum diseases (95% confidence interval (CI): 2.0-3.0; P < 0.0001) compared with 158,790 non-ADHD persons, after controlling for sex and age and adjusting for tobacco use and psychotic conditions. In 4960 ADHD patients prescribed psychostimulants, risk of basal ganglia and cerebellum diseases between ages 21 and 49 years was especially pronounced, at 8.6-fold (95% CI: 4.8-15.6; P < 0001). The association of ADHD patients prescribed psychostimulants with higher risk of diseases of the basal ganglia and cerebellum may reflect a more severe ADHD phenotype rather than a direct association between prescribed stimulant use and basal ganglia or cerebellum disorders. Future studies to assess and stratify patient risk so as to inform treatment are warranted.
Collapse
Affiliation(s)
- Karen Curtin
- 0000 0001 2193 0096grid.223827.eDepartment of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT USA
| | - Annette E. Fleckenstein
- 0000 0001 2193 0096grid.223827.eUniversity of Utah School of Dentistry, Salt Lake City, UT USA
| | - Brooks R. Keeshin
- 0000 0001 2193 0096grid.223827.eDepartment of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT USA ,0000 0004 0442 6404grid.415178.eDepartment of Pediatric Psychiatry, Intermountain Healthcare Primary Children’s Hospital, Salt Lake City, UT USA
| | - Deborah A. Yurgelun-Todd
- 0000 0001 2193 0096grid.223827.eDepartment of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT USA
| | - Perry F. Renshaw
- 0000 0001 2193 0096grid.223827.eDepartment of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT USA
| | - Ken R. Smith
- 0000 0001 2193 0096grid.223827.eHuman Development and Family Studies, University of Utah, Salt Lake City, UT USA
| | - Glen R. Hanson
- 0000 0001 2193 0096grid.223827.eUniversity of Utah School of Dentistry, Salt Lake City, UT USA
| |
Collapse
|
73
|
Kuo SC, Yeh YW, Chen CY, Huang CC, Ho PS, Liang CS, Lin CL, Yeh TC, Tsou CC, Yang BZ, Lu RB, Huang SY. Differential effect of the DRD3 genotype on inflammatory cytokine responses during abstinence in amphetamine-dependent women. Psychoneuroendocrinology 2018; 97:37-46. [PMID: 30005280 DOI: 10.1016/j.psyneuen.2018.06.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 05/25/2018] [Accepted: 06/29/2018] [Indexed: 11/19/2022]
Abstract
Amphetamine exposure impacts on innate and adaptive immunity and DRD3 may modulate the effect of amphetamine on the immune response. We assessed the immune-cytokine markers in 72 female patients with amphetamine dependence (AD) at baseline and after 4-week drug abstinence and in 51 healthy women. Multiplex magnetic bead assay was used to measure the plasma cytokine expression level simultaneously in all participants and DRD3 rs6280 polymorphism was genotyped in patients. We demonstrated an increase of the T helper 1 (Th1) cytokines (IL-2), Th2 cytokines (IL-4, IL-5, IL-6 and IL-10) and other cytokines (IL-1β) in the entire AD cohort. A similar cytokine pattern, along with a significantly decreased IL-8 and IL-10 levels was observed after 4-week abstinence. Among AD patients with DRD3 rs6280 TT genotype, the cytokine expression profile was consistent with total AD cohort at baseline and revealed a significant down-regulated plasma level of the Th1, Th2, and other cytokines except for IL-6 after 4-week abstinence. In AD group with DRD3 rs6280 C allele carrier, we found IL-2 level was significantly higher than healthy controls at baseline and remained higher, accompanied with a borderline increase in IL-4, IL-6 and IL-1β levels after 4-week abstinence. Our results suggest that chronic use of amphetamine increased both pro- and anti-inflammatory cytokines in AD patients, indicating the immune imbalance that may persist for 4 weeks or more. Besides, DRD3 rs6280 TT genotype may be associated with favorable recovery in general inflammatory cytokines during period of abstinence.
Collapse
Affiliation(s)
- Shin-Chang Kuo
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yi-Wei Yeh
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chun-Yen Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chang-Chih Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Psychiatry, Buddhist Tzu Chi General Hospital, Taipei Branch, Taipei, Taiwan, ROC
| | - Pei-Shen Ho
- Department of Psychiatry, Taichung Armed Forces General Hospital, Taichung, Taiwan, ROC
| | - Chih-Sung Liang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chun-Long Lin
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Psychiatry, Hsinchu Branch, Taoyuan Armed Forces General Hospital, Hsinchu, Taiwan, ROC
| | - Ta-Chuan Yeh
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chang-Chih Tsou
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Bao-Zhu Yang
- Department of Psychiatry, Division of Human Genetics, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Ru-Band Lu
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - San-Yuan Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
74
|
Abstract
Methamphetamine has the second highest prevalence of drug abuse after cannabis, with estimates of 35 million users worldwide. The ( S)-(+)-enantiomer is the illicit drug, active neurostimulant, and eutomer, while the ( R)-(-)-enantiomer is contained in over the counter decongestants. While designated a schedule II drug in 1970, ( S)-(+)-methamphetamine is available by prescription for the treatment of attention-deficit disorder and obesity. The illicit use of ( S)-(+)-methamphetamine results in the sudden "rush" of stimulation to the motivation, movement, pleasure, and reward centers in the brain, caused by rapid release of dopamine. In this review, we will provide an overview of the synthesis, pharmacology, adverse effects, and drug metabolism of this widely abused psychostimulant that distinguish it as a DARK classic in Chemical Neuroscience.
Collapse
Affiliation(s)
- Thomas J. Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Paul C. Trippier
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| |
Collapse
|
75
|
Krill oil protects PC12 cells against methamphetamine-induced neurotoxicity by inhibiting apoptotic response and oxidative stress. Nutr Res 2018; 58:84-94. [DOI: 10.1016/j.nutres.2018.07.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 07/04/2018] [Accepted: 07/09/2018] [Indexed: 01/05/2023]
|
76
|
Flack A, Persons AL, Kousik SM, Celeste Napier T, Moszczynska A. Self-administration of methamphetamine alters gut biomarkers of toxicity. Eur J Neurosci 2018; 46:1918-1932. [PMID: 28661099 DOI: 10.1111/ejn.13630] [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: 03/09/2016] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 12/13/2022]
Abstract
Methamphetamine (METH) is a highly abused psychostimulant that is associated with an increased risk for developing Parkinson's disease (PD). This enhanced vulnerability likely relates to the toxic effects of METH that overlap with PD pathology, for example, aberrant functioning of α-synuclein and parkin. In PD, peripheral factors are thought to contribute to central nervous system (CNS) degeneration. For example, α-synuclein levels in the enteric nervous system (ENS) are elevated, and this precedes the onset of motor symptoms. It remains unclear whether neurons of the ENS, particularly catecholaminergic neurons, exhibit signs of METH-induced toxicity as seen in the CNS. The aim of this study was to determine whether self-administered METH altered the levels of α-synuclein, parkin, tyrosine hydroxylase (TH), and dopamine-β-hydroxylase (DβH) in the myenteric plexus of the distal colon ENS. Young adult male Sprague-Dawley rats self-administered METH for 3 h per day for 14 days and controls were saline-yoked. Distal colon tissue was collected at 1, 14, or 56 days after the last operant session. Levels of α-synuclein were increased, while levels of parkin, TH, and DβH were decreased in the myenteric plexus in the METH-exposed rats at 1 day following the last operant session and returned to the control levels after 14 or 56 days of forced abstinence. The changes were not confined to neurofilament-positive neurons. These results suggest that colon biomarkers may provide early indications of METH-induced neurotoxicity, particularly in young chronic METH users who may be more susceptible to progression to PD later in life.
Collapse
Affiliation(s)
- Amanda Flack
- Department of Pharmaceutical Sciences, Wayne State University, Eugene Applebaum College of Pharmaceutical Sciences, Detroit, MI, 48201, USA
| | - Amanda L Persons
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, USA.,Department of Pharmacology, Rush University Medial Center, Chicago, IL, USA
| | - Sharanya M Kousik
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, USA.,Department of Pharmacology, Rush University Medial Center, Chicago, IL, USA
| | - T Celeste Napier
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, USA.,Department of Pharmacology, Rush University Medial Center, Chicago, IL, USA.,Department of Psychiatry, Rush University Medical Center, Chicago, IL, USA
| | - Anna Moszczynska
- Department of Pharmaceutical Sciences, Wayne State University, Eugene Applebaum College of Pharmaceutical Sciences, Detroit, MI, 48201, USA
| |
Collapse
|
77
|
Epigenetic Effects Induced by Methamphetamine and Methamphetamine-Dependent Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4982453. [PMID: 30140365 PMCID: PMC6081569 DOI: 10.1155/2018/4982453] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/10/2018] [Indexed: 12/21/2022]
Abstract
Methamphetamine is a widely abused drug, which possesses neurotoxic activity and powerful addictive effects. Understanding methamphetamine toxicity is key beyond the field of drug abuse since it allows getting an insight into the molecular mechanisms which operate in a variety of neuropsychiatric disorders. In fact, key alterations produced by methamphetamine involve dopamine neurotransmission in a way, which is reminiscent of spontaneous neurodegeneration and psychiatric schizophrenia. Thus, understanding the molecular mechanisms operated by methamphetamine represents a wide window to understand both the addicted brain and a variety of neuropsychiatric disorders. This overlapping, which is already present when looking at the molecular and cellular events promoted immediately after methamphetamine intake, becomes impressive when plastic changes induced in the brain of methamphetamine-addicted patients are considered. Thus, the present manuscript is an attempt to encompass all the molecular events starting at the presynaptic dopamine terminals to reach the nucleus of postsynaptic neurons to explain how specific neurotransmitters and signaling cascades produce persistent genetic modifications, which shift neuronal phenotype and induce behavioral alterations. A special emphasis is posed on disclosing those early and delayed molecular events, which translate an altered neurotransmitter function into epigenetic events, which are derived from the translation of postsynaptic noncanonical signaling into altered gene regulation. All epigenetic effects are considered in light of their persistent changes induced in the postsynaptic neurons including sensitization and desensitization, priming, and shift of neuronal phenotype.
Collapse
|
78
|
Salvatore MF, Nejtek VA, Khoshbouei H. Prolonged increase in ser31 tyrosine hydroxylase phosphorylation in substantia nigra following cessation of chronic methamphetamine. Neurotoxicology 2018; 67:121-128. [PMID: 29782882 PMCID: PMC6088751 DOI: 10.1016/j.neuro.2018.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/14/2018] [Accepted: 05/16/2018] [Indexed: 12/13/2022]
Abstract
Methamphetamine (MA) exposure may increase the risk of motor or cognitive impairments similar to Parkinson's disease (PD) by middle age. Although damage to nigrostriatal or mesoaccumbens dopamine (DA) neurons may occur during or early after MA exposure, overt PD-like symptoms at a younger age may not manifest due to compensatory mechanisms to maintain DA neurotransmission. One possible compensatory mechanism is increased tyrosine hydroxylase (TH) phosphorylation. In the rodent PD 6-OHDA model, nigrostriatal lesion decreases TH protein in both striatum and substantia nigra (SN). However, DA loss in the SN is significantly less than that in the striatum. An increase in ser31 TH phosphorylation in the SN may increase TH activity in response to TH loss. To determine if similar compensatory mechanisms may be engaged in young mice after MA exposure, TH expression, phosphorylation, and DA tissue content were evaluated, along with dopamine transporter expression, 21 days after cessation of MA (24 mg/kg, daily, 14 days). DA tissue content was unaffected by the MA regimen in striatum, nucleus accumbens, SN, or ventral tegmental area (VTA), despite decreased TH protein in SN and VTA. In the SN, but not striatum, ser31 phosphorylation increased over 2-fold. This suggests that increased ser31 TH phosphorylation may be an inherent compensatory mechanism to attenuate DA loss against TH loss, similar to that in an established PD model. These results also indicate the somatodendritic compartments of DA neurons are more vulnerable to TH protein loss than terminal fields following MA exposure.
Collapse
Affiliation(s)
- Michael F Salvatore
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, United States; Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, United States.
| | - Vicki A Nejtek
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, FL, United States; Center for Addiction Research & Education, University of Florida, Gainesville, FL, United States
| |
Collapse
|
79
|
Methamphetamine toxicity-induced calcineurin activation, nuclear translocation of nuclear factor of activated T-cells and elevation of cyclooxygenase 2 levels are averted by calpastatin overexpression in neuroblastoma SH-SY5Y cells. Neurotoxicology 2018; 67:287-295. [DOI: 10.1016/j.neuro.2018.06.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/17/2018] [Accepted: 06/22/2018] [Indexed: 12/17/2022]
|
80
|
Granado N, Ares-Santos S, Tizabi Y, Moratalla R. Striatal Reinnervation Process after Acute Methamphetamine-Induced Dopaminergic Degeneration in Mice. Neurotox Res 2018; 34:627-639. [PMID: 29934756 DOI: 10.1007/s12640-018-9925-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/28/2018] [Accepted: 06/07/2018] [Indexed: 01/03/2023]
Abstract
Methamphetamine (METH), an amphetamine derivate, may increase the risk of developing Parkinson's disease (PD). Human and animal studies have shown that METH produces persistent dopaminergic neurotoxicity in the nigrostriatal pathway, despite initial partial recovery. To determine the processes leading to early compensation, we studied the detailed morphology and distribution of tyrosine hydroxylase immunoreactive fibers (TH-ir) classified by their thickness (types I-IV) before and after METH. Applying three established neurotoxic regimens of METH: single high dose (1 × 30 mg/kg), multiple lower doses (3 × 5 mg/kg) or (3 × 10 mg/kg), we show that METH primarily damages type I fibers (the thinner ones), and to a much lesser extend types II-IV fibers including sterile axons. The striatal TH terminal partial recovery process, consisting of a progressive regrowth increases in types II, III, and IV fibers, demonstrated by co-localization of GAP-43, a sprouting marker, was observed 3 days post-METH treatment. In addition, we demonstrate the presence of growth-cone-like TH-ir structures, indicative of new terminal generation as well as improvement in motor functions after 3 days. A temporal relationship was observed between decreases in TH-expression and increases in silver staining, a marker of degeneration. Striatal regeneration was associated with an increase in astroglia and decrease in microglia expression, suggesting a possible role for the neuroimmune system in regenerative processes. Identification of regenerative compensatory mechanisms in response to neurotoxic agents could point to novel mechanisms in countering the neurotoxicity and/or enhancing the regenerative processes.
Collapse
Affiliation(s)
- Noelia Granado
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Avda Dr Arce 37, 28002, Madrid, Spain.,CIBERNED, ISCIII, Madrid, Spain
| | - Sara Ares-Santos
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Avda Dr Arce 37, 28002, Madrid, Spain.,CIBERNED, ISCIII, Madrid, Spain
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington DC, USA
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Avda Dr Arce 37, 28002, Madrid, Spain. .,CIBERNED, ISCIII, Madrid, Spain.
| |
Collapse
|
81
|
Yang X, Wang Y, Li Q, Zhong Y, Chen L, Du Y, He J, Liao L, Xiong K, Yi CX, Yan J. The Main Molecular Mechanisms Underlying Methamphetamine- Induced Neurotoxicity and Implications for Pharmacological Treatment. Front Mol Neurosci 2018; 11:186. [PMID: 29915529 PMCID: PMC5994595 DOI: 10.3389/fnmol.2018.00186] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/14/2018] [Indexed: 01/07/2023] Open
Abstract
Methamphetamine (METH) is a popular new-type psychostimulant drug with complicated neurotoxicity. In spite of mounting evidence on METH-induced damage of neural cell, the accurate mechanism of toxic effect of the drug on central nervous system (CNS) has not yet been completely deciphered. Besides, effective treatment strategies toward METH neurotoxicity remain scarce and more efficacious drugs are to be developed. In this review, we summarize cellular and molecular bases that might contribute to METH-elicited neurotoxicity, which mainly include oxidative stress, excitotoxicity, and neuroinflammation. We also discuss some drugs that protect neural cells suffering from METH-induced neurotoxic consequences. We hope more in-depth investigations of exact details that how METH produces toxicity in CNS could be carried out in future and the development of new drugs as natural compounds and immunotherapies, including clinic trials, are expected.
Collapse
Affiliation(s)
- Xue Yang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yong Wang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Qiyan Li
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yaxian Zhong
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Liangpei Chen
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yajun Du
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Jing He
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Lvshuang Liao
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
82
|
Sambo DO, Lebowitz JJ, Khoshbouei H. The sigma-1 receptor as a regulator of dopamine neurotransmission: A potential therapeutic target for methamphetamine addiction. Pharmacol Ther 2018; 186:152-167. [PMID: 29360540 PMCID: PMC5962385 DOI: 10.1016/j.pharmthera.2018.01.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Methamphetamine (METH) abuse is a major public health issue around the world, yet there are currently no effective pharmacotherapies for the treatment of METH addiction. METH is a potent psychostimulant that increases extracellular dopamine levels by targeting the dopamine transporter (DAT) and alters neuronal activity in the reward centers of the brain. One promising therapeutic target for the treatment of METH addiction is the sigma-1 receptor (σ1R). The σ1R is an endoplasmic reticulum-localized chaperone protein that is activated by cellular stress, and, unique to this chaperone, its function can also be induced or inhibited by different ligands. Upon activation of this unique "chaperone receptor", the σ1R regulates a variety of cellular functions and possesses neuroprotective activity in the brain. Interestingly, a variety of σ1R ligands modulate dopamine neurotransmission and reduce the behavioral effects of METH in animal models of addictive behavior, suggesting that the σ1R may be a viable therapeutic target for the treatment of METH addiction. In this review, we provide background on METH and the σ1R as well as a literature review regarding the role of σ1Rs in modulating both dopamine neurotransmission and the effects of METH. We aim to highlight the complexities of σ1R pharmacology and function as well as the therapeutic potential of the σ1R as a target for the treatment of METH addiction.
Collapse
Affiliation(s)
- Danielle O Sambo
- University of Florida, College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States
| | - Joseph J Lebowitz
- University of Florida, College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States
| | - Habibeh Khoshbouei
- University of Florida, College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States.
| |
Collapse
|
83
|
Lappin JM, Darke S, Farrell M. Methamphetamine use and future risk for Parkinson's disease: Evidence and clinical implications. Drug Alcohol Depend 2018; 187:134-140. [PMID: 29665491 DOI: 10.1016/j.drugalcdep.2018.02.032] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/21/2018] [Accepted: 02/24/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Methamphetamine use has been posited to be a risk factor for the development of Parkinson's disease (PD) and parkinsonism. The clinical implications of a potential association between methamphetamine use and PD are considered. METHODS A review of methamphetamine and PD and parkinsonism was conducted, including evidence from animal models, clinical and population studies. RESULTS There is biological plausibility to a link between methamphetamine use and PD. Though clinical and epidemiological evidence in this area is scant, a number of studies suggest that methamphetamine is associated with a moderately increased risk of PD and parkinsonism, and may also lead to premature onset of PD. The long lag time between exposure to methamphetamine and onset of PD, the potential for recovery from neurotoxic effects, and tobacco smoking each may attenuate the association. Individual and drug use characteristics that may modulate a user's risk remain poorly understood. CONCLUSIONS The use of methamphetamine may be an initiating event in the development of PD and parkinsonism, in addition to other risk factors that a given individual may hold. Clinicians should be vigilant to signs of prodromal and emerging PD among methamphetamine users. In individuals with premature onset illness, information on current or prior exposure to methamphetamine should be sought.
Collapse
Affiliation(s)
- Julia M Lappin
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, Australia; School of Psychiatry, University of New South Wales, Sydney, Australia.
| | - Shane Darke
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, Australia
| | - Michael Farrell
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, Australia
| |
Collapse
|
84
|
Li L, Chen S, Wang Y, Yue X, Xu J, Xie W, Qiu P, Liu C, Wang A, Wang H. Role of GSK3β/α-synuclein axis in methamphetamine-induced neurotoxicity in PC12 cells. Toxicol Res (Camb) 2018; 7:221-234. [PMID: 30090577 PMCID: PMC6062219 DOI: 10.1039/c7tx00189d] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 12/22/2017] [Indexed: 12/22/2022] Open
Abstract
Methamphetamine (METH) is well-known as a potent psychostimulant of abuse worldwide. METH administration can cause neurotoxicity and neurodegenerative injury, which are similar to the two prevalent neurodegenerative disorders Alzheimer's disease (AD) and Parkinson's disease (PD). Recent results suggested that METH exposure increased the level of α-synuclein (α-syn) that could be a possible cause of neurotoxicity. However, the mechanism of METH-induced neurodegeneration remains unclear. This study was aimed at examining the effects of glycogen synthase kinase3β (GSK3β), α-syn, and tau on METH-induced neurotoxicity. Our results indicated that P-GSK3β (Tyr216), P-Tau (Ser396), α-syn, and P-α-syn (Ser129) levels were increased after METH administration in dose- and time-dependent manners. Upon inhibiting the GSK3β activity with LiCl or GSK3β-siRNA, these protein expressions were significantly decreased. We observed that LiCl protected the cells from METH-caused cytotoxicity by weakening the cell morphological damage and preventing cell apoptosis and death. We also found that P-GSK3β colocalized with P-Tau and α-syn by the immunofluorescence method. Further, METH disrupted the cellular autophagy by upregulation of LC3-II and P62 proteins, and the cellular autophagy was restored by LiCl and GSK3β-siRNA. The expressions of the α-syn-specific degradative enzyme glucocerebrosidase (GCase) with its regulator lysosomal integral membrane protein type-2 (LIMP-2) decreased inversely with the doses of METH treatment. The GCase inhibitor conduritol-β-epoxide (CβE) increased the α-syn levels, and LiCl restored GCase and LIMP-2 expressions disrupted by the METH treatment. In summary, we conclude that GSK3β plays key roles in METH-induced neurotoxicity and neurodegenerative injury by promoting abnormal protein phosphorylation and α-syn accumulation, blocking the autophagy-lysosomal degradation pathway, and finally leading to cell apoptosis and death. GSK3β may be a potential target to prevent METH-induced neurodegeneration.
Collapse
Affiliation(s)
- Lizeng Li
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Si Chen
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Yue Wang
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Xia Yue
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Jingtao Xu
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Weibing Xie
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Pingming Qiu
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Chao Liu
- Guangzhou Forensic Science Institute , Guangzhou 510030 , People's Republic of China
| | - AiFeng Wang
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Huijun Wang
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| |
Collapse
|
85
|
Inflammasome Activation by Methamphetamine Potentiates Lipopolysaccharide Stimulation of IL-1β Production in Microglia. J Neuroimmune Pharmacol 2018; 13:237-253. [PMID: 29492824 DOI: 10.1007/s11481-018-9780-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/19/2018] [Indexed: 01/09/2023]
Abstract
Methamphetamine (Meth) is an addictive psychostimulant abused worldwide. Ample evidence indicate that chronic abuse of Meth induces neurotoxicity via microglia-associated neuroinflammation and the activated microglia present in both Meth-administered animals and human abusers. The development of anti-neuroinflammation as a therapeutic strategy against Meth dependence promotes research to identify inflammatory pathways that are specifically tied to Meth-induced neurotoxicity. Currently, the exact mechanisms for Meth-induced microglia activation are largely unknown. NLRP3 is a well-studied cytosolic pattern recognition receptor (PRR), which promotes the assembly of the inflammasome in response to the danger-associated molecular patterns (DAMPs). It is our hypothesis that Meth activates NLRP3 inflammasome in microglia and promotes the processing and release of interleukin (IL)-1β, resulting in neurotoxic activity. To test this hypothesis, we studied the effects of Meth on IL-1β maturation and release from rat cortical microglial cultures. Incubation of microglia with physiologically relevant concentrations of Meth after lipopolysaccharide (LPS) priming produced an enhancement on IL-1β maturation and release. Meth treatment potentiated aggregation of inflammasome adaptor apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), induced activation of the IL-1β converting enzyme caspase-1 and produced lysosomal and mitochondrial impairment. Blockade of capase-1 activity, lysosomal cathepsin B activity or mitochondrial ROS production by their specific inhibitors reversed the effects of Meth, demonstrating an involvement of inflammasome in Meth-induced microglia activation. Taken together, our results suggest that Meth triggers microglial inflammasome activation in a manner dependent on both mitochondrial and lysosomal danger-signaling pathways.
Collapse
|
86
|
Role of dopamine D1 receptor in 3-fluoromethamphetamine-induced neurotoxicity in mice. Neurochem Int 2018; 113:69-84. [DOI: 10.1016/j.neuint.2017.11.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/14/2017] [Accepted: 11/28/2017] [Indexed: 01/26/2023]
|
87
|
Dang DK, Shin EJ, Kim DJ, Tran HQ, Jeong JH, Jang CG, Ottersen OP, Nah SY, Hong JS, Nabeshima T, Kim HC. PKCδ-dependent p47phox activation mediates methamphetamine-induced dopaminergic neurotoxicity. Free Radic Biol Med 2018; 115:318-337. [PMID: 29269308 PMCID: PMC7074955 DOI: 10.1016/j.freeradbiomed.2017.12.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 11/29/2017] [Accepted: 12/15/2017] [Indexed: 12/11/2022]
Abstract
Protein kinase C (PKC) has been recognized to activate NADPH oxidase (PHOX). However, the interaction between PKC and PHOX in vivo remains elusive. Treatment with methamphetamine (MA) resulted in a selective increase in PKCδ expression out of PKC isoforms. PKCδ co-immunoprecipitated with p47phox, and facilitated phosphorylation and membrane translocation of p47phox. MA-induced increases in PHOX activity and reactive oxygen species were attenuated by knockout of p47phox or PKCδ. In addition, MA-induced impairments in the Nrf-2-related glutathione synthetic system were also mitigated by knockout of p47phox or PKCδ. Glutathione-immunoreactivity was co-localized in Iba-1-labeled microglial cells and in NeuN-labeled neurons, but not in GFAP-labeled astrocytes, reflecting the necessity for self-protection against oxidative stress by mainly microglia. Buthionine-sulfoximine, an inhibitor of glutathione biosynthesis, potentiated microglial activation and pro-apoptotic changes, leading to dopaminergic losses. These neurotoxic processes were attenuated by rottlerin, a pharmacological inhibitor of PKCδ, genetic inhibitions of PKCδ [i.e., PKCδ knockout mice (KO) and PKCδ antisense oligonucleotide (ASO)], or genetic inhibition of p47phox (i.e., p47phox KO or p47phox ASO). Rottlerin did not exhibit any additive effects against the protective activity offered by genetic inhibition of p47phox. Therefore, we suggest that PKCδ is a critical regulator for p47phox activation induced by MA, and that Nrf-2-dependent GSH induction via inhibition of PKCδ or p47phox, is important for dopaminergic protection against MA insult.
Collapse
Affiliation(s)
- Duy-Khanh Dang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Dae-Joong Kim
- Department of Anatomy and Cell Biology, Medical School, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Hai-Quyen Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ole Petter Ottersen
- Laboratory of Molecular Neuroscience, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jau-Shyong Hong
- Neuropharmacology Section, Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Toshitaka Nabeshima
- Nabeshima Laboratory, Graduate School of Pharmaceutical Sciences, Meijo University, Nagoya 468-8503, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea.
| |
Collapse
|
88
|
Gemechu JM, Sharma A, Yu D, Xie Y, Merkel OM, Moszczynska A. Characterization of Dopaminergic System in the Striatum of Young Adult Park2 -/- Knockout Rats. Sci Rep 2018; 8:1517. [PMID: 29367643 PMCID: PMC5784013 DOI: 10.1038/s41598-017-18526-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 11/20/2017] [Indexed: 12/13/2022] Open
Abstract
Mutations in parkin gene (Park2) are linked to early-onset autosomal recessive Parkinson's disease (PD) and young-onset sporadic PD. Park2 knockout (PKO) rodents; however, do not display neurodegeneration of the nigrostriatal pathway, suggesting age-dependent compensatory changes. Our goal was to examine dopaminergic (DAergic) system in the striatum of 2 month-old PKO rats in order to characterize compensatory mechanisms that may have occurred within the system. The striata form wild type (WT) and PKO Long Evans male rats were assessed for the levels of DAergic markers, for monoamine oxidase (MAO) A and B activities and levels, and for the levels of their respective preferred substrates, serotonin (5-HT) and ß-phenylethylamine (ß-PEA). The PKO rats displayed lower activities of MAOs and higher levels of ß-PEA in the striatum than their WT counterparts. Decreased levels of ß-PEA receptor, trace amine-associated receptor 1 (TAAR-1), and postsynaptic DA D2 (D2L) receptor accompanied these alterations. Drug-naive PKO rats displayed normal locomotor activity; however, they displayed decreased locomotor response to a low dose of psychostimulant methamphetamine, suggesting altered DAergic neurotransmission in the striatum when challenged with an indirect agonist. Altogether, our findings suggest that 2 month-old PKO male rats have altered DAergic and trace aminergic signaling.
Collapse
Affiliation(s)
- Jickssa M Gemechu
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
- Department of Biomedical Sciences, OUWB School of Medicine, Rochester, MI, USA
| | - Akhil Sharma
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
| | - Dongyue Yu
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Yuran Xie
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
- Boston Biomedical Inc., Allston, MA, USA
| | - Olivia M Merkel
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
- Department of Pharmacy, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Anna Moszczynska
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
89
|
Scott LL, Downing TG. Β-N-Methylamino-L-Alanine (BMAA) Toxicity Is Gender and Exposure-Age Dependent in Rats. Toxins (Basel) 2017; 10:E16. [PMID: 29280981 PMCID: PMC5793103 DOI: 10.3390/toxins10010016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/21/2017] [Accepted: 12/26/2017] [Indexed: 12/21/2022] Open
Abstract
Cyanobacterial β-N-methylamino-L-alanine (BMAA) has been suggested as a causative or contributory factor in the development of several neurodegenerative diseases. However, no BMAA animal model has adequately shown clinical or behavioral symptoms that correspond to those seen in either Alzheimer's Disease (AD), Amyotrophic Lateral Sclerosis (ALS) or Parkinson's Disease (PD). We present here the first data that show that when neonatal rats were exposed to BMAA on postnatal days 3, 4 and 5, but not on gestational day 14 or postnatally on days 7 or 10, several AD and/or PD-related behavioral, locomotor and cognitive deficits developed. Male rats exhibited severe unilateral hindlimb splay while whole body tremors could be observed in exposed female rats. BMAA-exposed rats failed to identify and discriminate a learned odor, an early non-motor symptom of PD, and exhibited decreased locomotor activity, decreased exploration and increased anxiety in the open field test. Alterations were also observed in the rats' natural passive defense mechanism, and potential memory deficits and changes to the rat's natural height avoidance behavior could be observed as early as PND 30. Spatial learning, short-term working, reference and long-term memory were also impaired in 90-day-old rats that had been exposed to a single dose of BMAA on PND 3-7. These data suggest that BMAA is a developmental neurotoxin, with specific target areas in the brain and spinal cord.
Collapse
Affiliation(s)
- Laura Louise Scott
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77 000, Port Elizabeth 6031, South Africa.
| | - Timothy Grant Downing
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77 000, Port Elizabeth 6031, South Africa.
| |
Collapse
|
90
|
Prior nicotine self-administration attenuates subsequent dopaminergic deficits of methamphetamine in rats: role of nicotinic acetylcholine receptors. Behav Pharmacol 2017; 27:422-30. [PMID: 26871405 DOI: 10.1097/fbp.0000000000000215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Preclinical studies have demonstrated that oral nicotine exposure attenuates long-term dopaminergic damage induced by toxins, including repeated, high doses of methamphetamine. It is suggested that alterations in nicotinic acetylcholine receptor (nAChR) expression, including α4β2* and α6β2* subtypes, likely contribute to this protection. The current study extended these findings by investigating whether nicotine self-administration in male, Sprague-Dawley rats (a) attenuates short-term dopaminergic damage induced by methamphetamine and (b) causes alterations in levels of α4β2* and α6β2* nAChR subtypes. The findings indicate that nicotine self-administration (0.032 mg/kg/infusion for 14 days) per se did not alter α4β2* and α6β2* nAChR expression or dopamine transporter (DAT) expression and function. Interestingly, prior nicotine self-administration attenuated methamphetamine-induced decreases in DAT function when assessed 24 h, but not 1 h, after methamphetamine treatment (4×7.5 mg/kg/injection). The ability of nicotine to attenuate the effects of methamphetamine on DAT function corresponded with increases in α4β2*, but not α6β2*, nAChR binding density. Understanding the role of nAChRs in methamphetamine-induced damage has the potential to elucidate mechanisms underlying the etiology of disorders involving dopaminergic dysfunction, as well as to highlight potential new therapeutic strategies for prevention or reduction of dopaminergic neurodegeneration.
Collapse
|
91
|
Rumpf JJ, Albers J, Fricke C, Mueller W, Classen J. Structural abnormality of substantia nigra induced by methamphetamine abuse. Mov Disord 2017; 32:1784-1788. [DOI: 10.1002/mds.27205] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/14/2017] [Accepted: 09/17/2017] [Indexed: 12/23/2022] Open
Affiliation(s)
| | - Jana Albers
- Department of Neurology; University of Leipzig; Leipzig Germany
| | | | - Wolf Mueller
- Department of Neuropathology; University of Leipzig; Leipzig Germany
| | - Joseph Classen
- Department of Neurology; University of Leipzig; Leipzig Germany
| |
Collapse
|
92
|
Moszczynska A, Callan SP. Molecular, Behavioral, and Physiological Consequences of Methamphetamine Neurotoxicity: Implications for Treatment. J Pharmacol Exp Ther 2017; 362:474-488. [PMID: 28630283 PMCID: PMC11047030 DOI: 10.1124/jpet.116.238501] [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: 10/22/2016] [Accepted: 05/09/2017] [Indexed: 04/28/2024] Open
Abstract
Understanding the relationship between the molecular mechanisms underlying neurotoxicity of high-dose methamphetamine (METH) and related clinical manifestations is imperative for providing more effective treatments for human METH users. This article provides an overview of clinical manifestations of METH neurotoxicity to the central nervous system and neurobiology underlying the consequences of administration of neurotoxic METH doses, and discusses implications of METH neurotoxicity for treatment of human abusers of the drug.
Collapse
Affiliation(s)
- Anna Moszczynska
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Sean Patrick Callan
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| |
Collapse
|
93
|
Methamphetamine-Induced Brain Injury and Alcohol Drinking. J Neuroimmune Pharmacol 2017; 13:53-63. [PMID: 28856500 DOI: 10.1007/s11481-017-9764-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/21/2017] [Indexed: 12/12/2022]
Abstract
A majority of methamphetamine (Meth) abusers also abuse alcohol but the neurochemical consequences of this co-abuse are unknown. Individually, alcohol and Meth cause inflammation and long-term alterations in dopamine and serotonin signaling within the brain. Experiments were conducted to identify if serial exposure to alcohol and Meth has neurochemical consequences that are greater than after either drug alone. Male Sprague Dawley rats voluntarily drank 10% ethanol (EtOH) every other day for 4 weeks and were then exposed to a binge injection regimen of Meth (10 mg/kg injected every 2 h, for a total of 4 injections). EtOH drinking and preference increased over the 4 weeks and caused inflammation evidenced by increases in serum and brain lipopolysaccharide (LPS) and brain cyclooxygenase-2 (COX-2) 24 h after the last day of drinking. Meth alone depleted dopamine and serotonin in the striatum, as well as serotonin in the prefrontal cortex when measured 1 week later. In contrast, EtOH drinking alone did not affect dopamine and serotonin content in the striatum and prefrontal cortex, but prior EtOH drinking followed by injections of Meth enhanced Meth-induced depletions of dopamine, serotonin, as well as dopamine and serotonin transporter immunoreactivities in a manner that was correlated with the degree of EtOH consumption. Cyclooxygenase inhibition by ketoprofen during EtOH drinking blocked the increases in LPS and COX-2 and the enhanced decreases in dopamine and serotonin produced by Meth. Therefore, prior EtOH drinking causes an increase in inflammatory mediators that mediate a synergistic interaction with Meth to cause an enhanced neurotoxicity.
Collapse
|
94
|
de Rus Jacquet A, Tambe MA, Ma SY, McCabe GP, Vest JHC, Rochet JC. Pikuni-Blackfeet traditional medicine: Neuroprotective activities of medicinal plants used to treat Parkinson's disease-related symptoms. JOURNAL OF ETHNOPHARMACOLOGY 2017; 206:393-407. [PMID: 28088492 PMCID: PMC6149223 DOI: 10.1016/j.jep.2017.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 12/29/2016] [Accepted: 01/04/2017] [Indexed: 05/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Parkinson's disease (PD) is a multifactorial neurodegenerative disorder affecting 5% of the population over the age of 85 years. Current treatments primarily involve dopamine replacement therapy, which leads to temporary relief of motor symptoms but fails to slow the underlying neurodegeneration. Thus, there is a need for safe PD therapies with neuroprotective activity. In this study, we analyzed contemporary herbal medicinal practices used by members of the Pikuni-Blackfeet tribe from Western Montana to treat PD-related symptoms, in an effort to identify medicinal plants that are affordable to traditional communities and accessible to larger populations. AIM OF THE STUDY The aims of this study were to (i) identify medicinal plants used by the Pikuni-Blackfeet tribe to treat individuals with symptoms related to PD or other CNS disorders, and (ii) characterize a subset of the identified plants in terms of antioxidant and neuroprotective activities in cellular models of PD. MATERIALS AND METHODS Interviews of healers and local people were carried out on the Blackfeet Indian reservation. Plant samples were collected, and water extracts were produced for subsequent analysis. A subset of botanical extracts was tested for the ability to induce activation of the Nrf2-mediated transcriptional response and to protect against neurotoxicity elicited by the PD-related toxins rotenone and paraquat. RESULTS The ethnopharmacological interviews resulted in the documentation of 26 medicinal plants used to treat various ailments and diseases, including symptoms related to PD. Seven botanical extracts (out of a total of 10 extracts tested) showed activation of Nrf2-mediated transcriptional activity in primary cortical astrocytes. Extracts prepared from Allium sativum cloves, Trifolium pratense flowers, and Amelanchier arborea berries exhibited neuroprotective activity against toxicity elicited by rotenone, whereas only the extracts prepared from Allium sativum and Amelanchier arborea alleviated PQ-induced dopaminergic cell death. CONCLUSIONS Our findings highlight the potential clinical utility of plants used for medicinal purposes over generations by the Pikuni-Blackfeet people, and they shed light on mechanisms by which the plant extracts could slow neurodegeneration in PD.
Collapse
Affiliation(s)
- Aurélie de Rus Jacquet
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA.
| | - Mitali Arun Tambe
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA.
| | - Sin Ying Ma
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA.
| | - George P McCabe
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA.
| | | | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
95
|
Xu X, Huang E, Tai Y, Zhao X, Chen X, Chen C, Chen R, Liu C, Lin Z, Wang H, Xie WB. Nupr1 Modulates Methamphetamine-Induced Dopaminergic Neuronal Apoptosis and Autophagy through CHOP-Trib3-Mediated Endoplasmic Reticulum Stress Signaling Pathway. Front Mol Neurosci 2017; 10:203. [PMID: 28694771 PMCID: PMC5483452 DOI: 10.3389/fnmol.2017.00203] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/08/2017] [Indexed: 12/13/2022] Open
Abstract
Methamphetamine (METH) is an illegal and widely abused psychoactive stimulant. METH exposure causes detrimental effects on multiple organ systems, primarily the nervous system, especially dopaminergic pathways, in both laboratory animals and humans. In this study, we hypothesized that Nuclear protein 1 (Nupr1/com1/p8) is involved in METH-induced neuronal apoptosis and autophagy through endoplasmic reticulum (ER) stress signaling pathway. To test this hypothesis, we measured the expression levels of Nupr1, ER stress protein markers CHOP and Trib3, apoptosis-related protein markers cleaved-caspase3 and PARP, as well as autophagy-related protein markers LC3 and Beclin-1 in brain tissues of adult male Sprague-Dawley (SD) rats, rat primary cultured neurons and the rat adrenal pheochromocytoma cells (PC12 cells) after METH exposure. We also determined the effects of METH exposure on the expression of these proteins after silencing Nupr1, CHOP, or Trib3 expression with synthetic small hairpin RNA (shRNA) or siRNA in vitro, and after silencing Nupr1 in the striatum of rats by injecting lentivirus containing shRNA sequence targeting Nupr1 gene to rat striatum. The results showed that METH exposure increased Nupr1 expression that was accompanied with increased expression of ER stress protein markers CHOP and Trib3, and also led to apoptosis and autophagy in rat primary neurons and in PC12 cells after 24 h exposure (3.0 mM), and in the prefrontal cortex and striatum of rats after repeated intraperitoneal injections (15 mg/kg × 8 injections at 12 h intervals). Silencing of Nupr1 expression partly reduced METH-induced apoptosis and autophagy in vitro and in vivo. These results suggest that Nupr1 plays an essential role in METH-caused neuronal apoptosis and autophagy at relatively higher doses and may be a potential therapeutic target in high-dose METH-induced neurotoxicity.
Collapse
Affiliation(s)
- Xiang Xu
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China.,School of Forensic Medicine, Wannan Medical CollegeWuhu, China
| | - Enping Huang
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| | - Yunchun Tai
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| | - Xu Zhao
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| | - Xuebing Chen
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| | - Chuanxiang Chen
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| | - Rui Chen
- Department of Forensic Medicine, Guangdong Medical UniversityDongguan, China
| | - Chao Liu
- Guangzhou Forensic Science InstituteGuangzhou, China
| | - Zhoumeng Lin
- Institute of Computational Comparative Medicine and Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State UniversityManhattan, KS, United States
| | - Huijun Wang
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| | - Wei-Bing Xie
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| |
Collapse
|
96
|
Costa G, Morelli M, Simola N. Progression and Persistence of Neurotoxicity Induced by MDMA in Dopaminergic Regions of the Mouse Brain and Association with Noradrenergic, GABAergic, and Serotonergic Damage. Neurotox Res 2017; 32:563-574. [PMID: 28597409 DOI: 10.1007/s12640-017-9761-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/17/2017] [Accepted: 05/24/2017] [Indexed: 02/06/2023]
Abstract
The amphetamine-related drug 3,4-methylenedioxymethamphetamine (MDMA) is known to induce neurotoxic damage in dopaminergic regions of the mouse brain. In order to characterize how the number of administrations influenced the severity of MDMA-induced dopaminergic damage and to describe the localization and persistence of this damage, we evaluated the changes in tyrosine hydroxylase (TH) and dopamine transporter (DAT) in different regions of the mouse brain. Moreover, we investigated whether dopaminergic damage was associated with noradrenergic, GABAergic, and serotonergic damage, by evaluating the changes in noradrenaline transporter (NET), glutamic acid decarboxylase-67 (GAD-67), and serotonin transporter (SERT). Mice received 14, 28, or 36 MDMA administrations (10 mg/kg twice a week) and were sacrificed at different time points (postnatal days 85, 110, 138, or 214) for immunohistochemical evaluation. Mice receiving 28 administrations showed reduced levels of DAT-positive fibers in caudate-putamen (CPu) and medial prefrontal cortex (mPFC) and reduced levels of TH-positive nigral neurons. These mice also displayed increased NET-positive hippocampal fibers, reduced GAD-67-positive neurons in CPu and hippocampus, and reduced GAD-67-positive fibers in mPFC. Similar effects of MDMA on DAT, TH, and GAD-67 were found in mice receiving 36 administrations, which also displayed reduced levels of striatal, cortical, and hippocampal TH-immunoreactive fibers. The reductions in dopaminergic markers and GAD-67 persisted at 3 months after MDMA discontinuation. Finally, MDMA never modified the levels of SERT. These results provide further insight into the localization and persistence of MDMA-induced dopaminergic damage and show that this effect may associate with GABAergic but not noradrenergic or serotonergic damage.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124, Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124, Cagliari, Italy. .,National Research Council of Italy, Neuroscience Institute, Cagliari, Italy.
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124, Cagliari, Italy
| |
Collapse
|
97
|
Shin EJ, Tran HQ, Nguyen PT, Jeong JH, Nah SY, Jang CG, Nabeshima T, Kim HC. Role of Mitochondria in Methamphetamine-Induced Dopaminergic Neurotoxicity: Involvement in Oxidative Stress, Neuroinflammation, and Pro-apoptosis-A Review. Neurochem Res 2017; 43:66-78. [PMID: 28589520 DOI: 10.1007/s11064-017-2318-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/25/2017] [Accepted: 05/30/2017] [Indexed: 01/08/2023]
Abstract
Methamphetamine (MA), an amphetamine-type psychostimulant, is associated with dopaminergic toxicity and has a high abuse potential. Numerous in vivo and in vitro studies have suggested that impaired mitochondria are critical in dopaminergic toxicity induced by MA. Mitochondria are important energy-producing organelles with dynamic nature. Evidence indicated that exposure to MA can disturb mitochondrial energetic metabolism by inhibiting the Krebs cycle and electron transport chain. Alterations in mitochondrial dynamic processes, including mitochondrial biogenesis, mitophagy, and fusion/fission, have recently been shown to contribute to dopaminergic toxicity induced by MA. Furthermore, it was demonstrated that MA-induced mitochondrial impairment enhances susceptibility to oxidative stress, pro-apoptosis, and neuroinflammation in a positive feedback loop. Protein kinase Cδ has emerged as a potential mediator between mitochondrial impairment and oxidative stress, pro-apoptosis, or neuroinflammation in MA neurotoxicity. Understanding the role and underlying mechanism of mitochondrial impairment could provide a molecular target to prevent or alleviate dopaminergic toxicity induced by MA.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Hai-Quyen Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Phuong-Tram Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake, 470-1192, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
98
|
Ashok AH, Mizuno Y, Volkow ND, Howes OD. Association of Stimulant Use With Dopaminergic Alterations in Users of Cocaine, Amphetamine, or Methamphetamine: A Systematic Review and Meta-analysis. JAMA Psychiatry 2017; 74:511-519. [PMID: 28297025 PMCID: PMC5419581 DOI: 10.1001/jamapsychiatry.2017.0135] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Importance Stimulant use disorder is common, affecting between 0.3% and 1.1% of the population, and costs more than $85 billion per year globally. There are no licensed treatments to date. Several lines of evidence implicate the dopamine system in the pathogenesis of substance use disorder. Therefore, understanding the nature of dopamine dysfunction seen in stimulant users has the potential to aid the development of new therapeutics. Objective To comprehensively review the in vivo imaging evidence for dopaminergic alterations in stimulant (cocaine, amphetamine, or methamphetamine) abuse or dependence. Data Sources The entire PubMed, EMBASE, and PsycINFO databases were searched for studies from inception date to May 14, 2016. Study Selection Case-control studies were identified that compared dopaminergic measures between stimulant users and healthy controls using positron emission tomography or single-photon emission computed tomography to measure striatal dopamine synthesis or release or to assess dopamine transporter availability or dopamine receptor availability. Data Extraction and Synthesis Demographic, clinical, and imaging measures were extracted from each study, and meta-analyses and sensitivity analyses were conducted for stimulants combined, as well as for cocaine and for amphetamine and methamphetamine separately if there were sufficient studies. Main Outcomes and Measures Differences were measured in dopamine release (assessed using change in the D2/D3 receptor availability after administration of amphetamine or methylphenidate), dopamine transporter availability, and dopamine receptor availability in cocaine users, amphetamine and methamphetamine users, and healthy controls. Results A total of 31 studies that compared dopaminergic measures between 519 stimulant users and 512 healthy controls were included in the final analysis. In most of the studies, the duration of abstinence varied from 5 days to 3 weeks. There was a significant decrease in striatal dopamine release in stimulant users compared with healthy controls: the effect size was -0.84 (95% CI, -1.08 to -0.60; P < .001) for stimulants combined and -0.87 (95% CI, -1.15 to -0.60; P < .001) for cocaine. In addition, there was a significant decrease in dopamine transporter availability: the effect size was -0.91 (95% CI, -1.50 to -0.32; P < .01) for stimulants combined and -1.47 (95% CI, -1.83 to -1.10; P < .001) for amphetamine and methamphetamine. There was also a significant decrease in D2/D3 receptor availability: the effect size was -0.76 (95% CI, -0.92 to -0.60; P < .001) for stimulants combined, -0.73 (95% CI, -0.94 to -0.53; P < .001) for cocaine, and -0.81 (95% CI, -1.12 to -0.49; P < .001) for amphetamine and methamphetamine. Consistent alterations were not found in vesicular monoamine transporter, dopamine synthesis, or D1 receptor studies. Conclusions and Relevance Data suggest that both presynaptic and postsynaptic aspects of the dopamine system in the striatum are down-regulated in stimulant users. The commonality and differences between these findings and the discrepancies with the preclinical literature and models of drug addiction are discussed, as well as their implications for future drug development.
Collapse
Affiliation(s)
- Abhishekh H Ashok
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences Centre (LMS), Du Cane Road, London, UK
- Psychiatric Imaging Group, Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Yuya Mizuno
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, Bethesda, USA
| | - Oliver D Howes
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences Centre (LMS), Du Cane Road, London, UK
- Psychiatric Imaging Group, Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| |
Collapse
|
99
|
Current understanding of methamphetamine-associated dopaminergic neurodegeneration and psychotoxic behaviors. Arch Pharm Res 2017; 40:403-428. [DOI: 10.1007/s12272-017-0897-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/02/2016] [Indexed: 12/21/2022]
|
100
|
|