1
|
Li S, Zhua Y, Liu X. Parkinsonism in liver diseases or dysfunction. Med Clin (Barc) 2024:S0025-7753(24)00356-7. [PMID: 38955605 DOI: 10.1016/j.medcli.2024.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 04/20/2024] [Indexed: 07/04/2024]
Abstract
Parkinsonism in liver diseases or dysfunction, mainly including neurological manifestations in hereditary liver diseases and neurological complications of advanced liver diseases, occur in isolation or in combination with other movement disorders, and progress along disease course. Prominent akinetic-rigidity syndrome, various onset and progression, poor levodopa response and metabolism abnormalities reflected by serum biomarkers and neuroimaging, make this atypical parkinsonism recognizable and notable in clinical practice. Different susceptibility of brain areas, especially in basal ganglia, to manganese, iron, copper, ammonia overload, together with subsequent oxidative stress, neurotransmitter alterations, disturbed glia-neuron homeostasis and eventually neurotoxicity, contribute to parkinsonism under the circumstances of insufficient liver clearance ability. These mechanisms are interrelated and may interact collectively, adding to the complexity of clinical manifestations and treatment responses. This review summarizes shared clinical features of parkinsonism in liver diseases or dysfunction, depicts their underlying mechanisms and suggests practical flowchart for differential diagnosis.
Collapse
Affiliation(s)
- Sichen Li
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxia Zhua
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xi Liu
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Harini K, Girigoswami K, Anand AV, Pallavi P, Gowtham P, Elboughdiri N, Girigoswami A. Nano-mediated Strategies for Metal Ion–Induced Neurodegenerative Disorders: Focus on Alzheimer’s and Parkinson’s Diseases. CURRENT PHARMACOLOGY REPORTS 2022; 8:450-463. [DOI: 10.1007/s40495-022-00307-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/09/2022] [Indexed: 11/29/2023]
|
3
|
Chougar L, Arsovic E, Gaurav R, Biondetti E, Faucher A, Valabrègue R, Pyatigorskaya N, Dupont G, Lejeune FX, Cormier F, Corvol JC, Vidailhet M, Degos B, Grabli D, Lehéricy S. Regional Selectivity of Neuromelanin Changes in the Substantia Nigra in Atypical Parkinsonism. Mov Disord 2022; 37:1245-1255. [PMID: 35347754 DOI: 10.1002/mds.28988] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Neurodegeneration in the substantia nigra pars compacta (SNc) in parkinsonian syndromes may affect the nigral territories differently. OBJECTIVE The objective of this study was to investigate the regional selectivity of neurodegenerative changes in the SNc in patients with Parkinson's disease (PD) and atypical parkinsonism using neuromelanin-sensitive magnetic resonance imaging (MRI). METHODS A total of 22 healthy controls (HC), 38 patients with PD, 22 patients with progressive supranuclear palsy (PSP), 20 patients with multiple system atrophy (MSA, 13 with the parkinsonian variant, 7 with the cerebellar variant), 7 patients with dementia with Lewy body (DLB), and 4 patients with corticobasal syndrome were analyzed. volume and signal-to-noise ratio (SNR) values of the SNc were derived from neuromelanin-sensitive MRI in the whole SNc. Analysis of signal changes was performed in the sensorimotor, associative, and limbic territories of the SNc. RESULTS SNc volume and corrected volume were significantly reduced in PD, PSP, and MSA versus HC. Patients with PSP had lower volume, corrected volume, SNR, and contrast-to-noise ratio than HC and patients with PD and MSA. Patients with PSP had greater SNR reduction in the associative region than HC and patients with PD and MSA. Patients with PD had reduced SNR in the sensorimotor territory, unlike patients with PSP. Patients with MSA did not differ from patients with PD. CONCLUSIONS This study provides the first MRI comparison of the topography of neuromelanin changes in parkinsonism. The spatial pattern of changes differed between PSP and synucleinopathies. These nigral topographical differences are consistent with the topography of the extranigral involvement in parkinsonian syndromes. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Lydia Chougar
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, DMU DIAMENT, Department of Neuroradiology, F-75013, Paris, France, Paris, France.,ICM, Centre de NeuroImagerie de Recherche-CENIR, Paris, France.,ICM, Team "Movement Investigations and Therapeutics" (MOV'IT), Paris, France
| | - Emina Arsovic
- ICM, Centre de NeuroImagerie de Recherche-CENIR, Paris, France.,ICM, Team "Movement Investigations and Therapeutics" (MOV'IT), Paris, France.,Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, DMU DIAMENT, Department of Neuroradiology, F-75013, Paris, France, Paris, France
| | - Rahul Gaurav
- ICM, Centre de NeuroImagerie de Recherche-CENIR, Paris, France.,ICM, Team "Movement Investigations and Therapeutics" (MOV'IT), Paris, France.,Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inserm, F-75013, Paris, France
| | - Emma Biondetti
- ICM, Centre de NeuroImagerie de Recherche-CENIR, Paris, France.,ICM, Team "Movement Investigations and Therapeutics" (MOV'IT), Paris, France.,Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inserm, F-75013, Paris, France
| | - Alice Faucher
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR7241/INSERM U1050, Université PSL, Paris, France.,Service de Neurologie, Hôpital Avicenne, Hôpitaux Universitaires de Paris Seine-Saint-Denis, APHP, Bobigny, France
| | - Romain Valabrègue
- ICM, Centre de NeuroImagerie de Recherche-CENIR, Paris, France.,Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inserm, F-75013, Paris, France
| | - Nadya Pyatigorskaya
- ICM, Centre de NeuroImagerie de Recherche-CENIR, Paris, France.,ICM, Team "Movement Investigations and Therapeutics" (MOV'IT), Paris, France.,Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, DMU DIAMENT, Department of Neuroradiology, F-75013, Paris, France, Paris, France
| | - Gwendoline Dupont
- Centre hospitalier universitaire François Mitterrand, Département de Neurologie, Université de Bourgogne, Dijon, France
| | - François-Xavier Lejeune
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inserm, F-75013, Paris, France.,ICM, Data and Analysis Core, Paris, France
| | - Florence Cormier
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inserm, F-75013, Paris, France.,Clinique des mouvements anormaux, Département de Neurologie, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jean-Christophe Corvol
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inserm, F-75013, Paris, France.,ICM, Centre d'Investigation Clinique Neurosciences, Paris, France
| | - Marie Vidailhet
- ICM, Team "Movement Investigations and Therapeutics" (MOV'IT), Paris, France.,Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inserm, F-75013, Paris, France.,Clinique des mouvements anormaux, Département de Neurologie, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Bertrand Degos
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR7241/INSERM U1050, Université PSL, Paris, France.,Service de Neurologie, Hôpital Avicenne, Hôpitaux Universitaires de Paris Seine-Saint-Denis, APHP, Bobigny, France
| | - David Grabli
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inserm, F-75013, Paris, France.,Clinique des mouvements anormaux, Département de Neurologie, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Stéphane Lehéricy
- ICM, Centre de NeuroImagerie de Recherche-CENIR, Paris, France.,ICM, Team "Movement Investigations and Therapeutics" (MOV'IT), Paris, France.,Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, DMU DIAMENT, Department of Neuroradiology, F-75013, Paris, France, Paris, France
| |
Collapse
|
4
|
Effects of Pentoxifylline in a Rat Model of Manganism: Evaluation of the Possible Toxicity. J CHEM-NY 2021. [DOI: 10.1155/2021/9926100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Objective. Manganese (Mn) has been reported, through dietary and occupational overexposure, to induce neurotoxicity named manganism. Pentoxifylline (PTX) administration attracts much attention considering the beneficial properties of PTX, as an anti-inflammatory and smooth muscle relaxation agent. This in vivo study aims to evaluate the effect of PTX on manganism in rat model. Materials and Methods. Thirty adult male Sprague Dawley rats received MnCl2 (100 mg/kg, i.p. on days 1, 3, and 7) during a week alone or in combination with PTX (300 mg/kg, i.p. every day for 8 consecutive days on manganism rat model). Several locomotor activity indices, as well as biomarkers of oxidative stress, were monitored in the brain tissue of Mn-exposed animals. Results. It was found that PTX supplementation (300 mg/kg, i.p.) deteriorated the Mn-induced locomotor deficit. This drug also increased the Mn brain accumulation as well as reactive oxygen species (ROS) and lipid peroxidation products in the manganism rat model. Moreover, the levels of total antioxidant capacity (TAC) and glutathione (GSH) were shown to be reduced significantly compared to the control group. Conclusion. The results of this study revealed that PTX at a high dose (300 mg/kg) might increase manganism complications. PTX lowers the blood viscosity, improves the tissue perfusion, and increases the Mn levels in the brain.
Collapse
|
5
|
[11C]dihydrotetrabenazine Positron Emission Tomography in Manganese-Exposed Workers. J Occup Environ Med 2021; 62:788-794. [PMID: 32472844 DOI: 10.1097/jom.0000000000001915] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To understand the neurotoxic effects of manganese (Mn) exposure on monoaminergic function, utilizing [C]dihydrotetrabenazine (DTBZ) positron emission tomography (PET) to measure vesicular monoamine transporter 2 (VMAT2). METHODS Basal ganglia and thalamic DTBZ binding potentials (BPND) were calculated on 56 PETs from 41 Mn-exposed workers. Associations between cumulative Mn exposure, regional BPND, and parkinsonism were examined by mixed linear regression. RESULTS Thalamic DTBZ BPND was inversely associated with exposure in workers with less than 3 mg Mn/m-yrs, but subsequently remained stable. Pallidal DTBZ binding increased in workers with less than 2 mg Mn/m-yrs of exposure, but decreased thereafter. Thalamic DTBZ binding was inversely associated with parkinsonism (P = 0.003). CONCLUSION Mn-dose-dependent associations with thalamic and pallidal DTBZ binding indicate direct effects on monoaminergic VMAT2. Thalamic DTBZ binding was also associated with parkinsonism, suggesting potential as an early biomarker of Mn neurotoxicity.
Collapse
|
6
|
Lee EY, Flynn MR, Du G, Lewis MM, Goldenberg M, Kong L, Mailman RB, Hong YS, Huang X. Nigral MRI features of asymptomatic welders. Parkinsonism Relat Disord 2021; 85:37-43. [PMID: 33691274 DOI: 10.1016/j.parkreldis.2021.02.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 02/05/2021] [Accepted: 02/14/2021] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Manganese (Mn)-induced parkinsonism involves motor symptoms similar to those observed in Parkinson's disease (PD). Previous literature suggests that chronic Mn- exposure may increase PD risk, although Mn-induced clinical syndromes are considered atypical for PD. This study investigated whether asymptomatic welders display differences in the substantia nigra (SN), the key pathological locus of PD. METHOD Brain MRI data and occupational exposure history were obtained in welders (N = 43) and matched controls (N = 31). Diffusion tensor imaging fractional anisotropy (FA; estimate of microstructural integrity) and R2* (estimate of iron and other PD-related brain differences) values in the SN pars compacta (SNc), SN reticulata (SNr), and globus pallidus (GP) were compared between the two groups. The MRI markers of the SN and GP within welders were related to exposure estimates. RESULTS Compared to controls, welders who had chronic, but low-level, Mn-exposure had similar FA and R2* values in both SN regions (p's > 0.082), but significantly lower FA (p = 0.0013), although not R2* (p = 0.553), in the GP. In welders, FA values in the SN and GP showed a second-order polynomial relationship with cumulative lifetime welding exposure (p's < 0.03). CONCLUSION Neurotoxic processes associated with Mn-exposure may be different from those in PD when the exposure-level is relatively low. Greater welding duration and level, however, were associated with FA differences in the GP and SN, indicating that welding exposures above a certain level may induce neurotoxicity in the SN, a finding that should be explored further in future studies.
Collapse
Affiliation(s)
- Eun-Young Lee
- Department of Health Care and Science, Dong-A University, Busan, South Korea.
| | - Michael R Flynn
- Department of Kinesiology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Environmental Sciences, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Guangwei Du
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Mechelle M Lewis
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Pharmacology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Michael Goldenberg
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Lan Kong
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Richard B Mailman
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Pharmacology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Young-Seoub Hong
- Department of Preventive Medicine, Dong-A University College of Medicine, Busan, South Korea.
| | - Xuemei Huang
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Pharmacology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Radiology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Neurosurgery, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Kinesiology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Environmental Sciences, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
7
|
Apetauerova D, Hildebrand P, Scala S, Zani JW, Lipert L, Clark E, Fennell T, Gordon FD. A Prospective Study of the Prevalence of Parkinsonism in Patients With Liver Cirrhosis. Hepatol Commun 2021; 5:323-333. [PMID: 33553978 PMCID: PMC7850299 DOI: 10.1002/hep4.1624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/02/2020] [Accepted: 09/12/2020] [Indexed: 02/04/2023] Open
Abstract
Acquired hepatocerebral degeneration refers to a neurological syndrome consisting of various movement disorders and cognitive impairment in advanced liver cirrhosis or portosystemic shunt. Neurological signs and symptoms may be attributed to the accumulation of toxic substances in the brain. The most common neurological presentation of this is parkinsonism. Our prospective study aimed to investigate the prevalence of parkinsonism in patients with cirrhosis who were evaluated for liver transplant and to identify any correlation between findings on brain magnetic resonance imaging (MRI) and severity of parkinsonism. Of the 120 enrolled participants with liver cirrhosis, 62 (52%) exhibited signs of parkinsonism and all had MRI basal ganglia hyperintensity. Eighteen patients from this group were transplanted and showed statistically significant improvements in their Unified Parkinson's Disease Rating Scale (UPDRS) scores. Conclusion: The data suggest the reversibility of the neurological impairment seen in cirrhosis, and therefore the effectiveness of transplantation in improving parkinsonian symptoms. There was no correlation between severity of MRI findings and clinical motor UPDRS part III. Laboratory findings showed no correlation among the abnormal levels, MRI brain signal abnormality, or UPDRS scores.
Collapse
Affiliation(s)
| | | | | | - Janet W Zani
- Lahey Hospital and Medical CenterBurlingtonMAUSA
| | | | - Erin Clark
- The University of New England College of Osteopathic MedicineBiddefordMEUSA
| | | | | |
Collapse
|
8
|
Harischandra DS, Ghaisas S, Zenitsky G, Jin H, Kanthasamy A, Anantharam V, Kanthasamy AG. Manganese-Induced Neurotoxicity: New Insights Into the Triad of Protein Misfolding, Mitochondrial Impairment, and Neuroinflammation. Front Neurosci 2019; 13:654. [PMID: 31293375 PMCID: PMC6606738 DOI: 10.3389/fnins.2019.00654] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/06/2019] [Indexed: 12/21/2022] Open
Abstract
Occupational or environmental exposure to manganese (Mn) can lead to the development of "Manganism," a neurological condition showing certain motor symptoms similar to Parkinson's disease (PD). Like PD, Mn toxicity is seen in the central nervous system mainly affecting nigrostriatal neuronal circuitry and subsequent behavioral and motor impairments. Since the first report of Mn-induced toxicity in 1837, various experimental and epidemiological studies have been conducted to understand this disorder. While early investigations focused on the impact of high concentrations of Mn on the mitochondria and subsequent oxidative stress, current studies have attempted to elucidate the cellular and molecular pathways involved in Mn toxicity. In fact, recent reports suggest the involvement of Mn in the misfolding of proteins such as α-synuclein and amyloid, thus providing credence to the theory that environmental exposure to toxicants can either initiate or propagate neurodegenerative processes by interfering with disease-specific proteins. Besides manganism and PD, Mn has also been implicated in other neurological diseases such as Huntington's and prion diseases. While many reviews have focused on Mn homeostasis, the aim of this review is to concisely synthesize what we know about its effect primarily on the nervous system with respect to its role in protein misfolding, mitochondrial dysfunction, and consequently, neuroinflammation and neurodegeneration. Based on the current evidence, we propose a 'Mn Mechanistic Neurotoxic Triad' comprising (1) mitochondrial dysfunction and oxidative stress, (2) protein trafficking and misfolding, and (3) neuroinflammation.
Collapse
Affiliation(s)
- Dilshan S Harischandra
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| | - Shivani Ghaisas
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| | - Gary Zenitsky
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| | - Huajun Jin
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| | - Anumantha G Kanthasamy
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| |
Collapse
|
9
|
Bailey RA, Gutierrez A, Kyser TL, Hemmerle AM, Hufgard JR, Seroogy KB, Vorhees CV, Williams MT. Effects of Preweaning Manganese in Combination with Adult Striatal Dopamine Lesions on Monoamines, BDNF, TrkB, and Cognitive Function in Sprague-Dawley Rats. Neurotox Res 2019; 35:606-620. [PMID: 30612279 DOI: 10.1007/s12640-018-9992-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/19/2018] [Accepted: 12/18/2018] [Indexed: 01/25/2023]
Abstract
Manganese (Mn) is an essential nutrient especially during development, but Mn overexposure (MnOE) produces long-term cognitive deficits. Evidence of long-term changes in dopamine in the neostriatum was found in rats from developmental MnOE previously. To examine the relationship between MnOE and dopamine, we tested whether the effects of developmental MnOE would be exaggerated by dopamine reductions induced by 6-hydroxydopamine (6-OHDA) neostriatal infusion when the rats were adults. The experiment consisted of four groups of females and males: Vehicle/Sham, MnOE/Sham, Vehicle/6-OHDA, and MnOE/6-OHDA. Both MnOE/Sham and Vehicle/6-OHDA groups displayed egocentric and allocentric memory deficits, whereas MnOE+6-OHDA had additive effects on spatial memory in the Morris water maze and egocentric learning in the Cincinnati water maze. 6-OHDA reduced dopamine in the neostriatum and nucleus accumbens, reduced norepinephrine in the hippocampus, reduced TH+ cells and TrkB and TH expression in the substantia nigra pars compacta (SNpc), but increased TrkB in the neostriatum. MnOE alone had no effect on monoamines or TrkB in the neostriatum or hippocampus but reduced BDNF in the hippocampus. A number of sex differences were noted; however, only a few significant interactions were found for MnOE and/or 6-OHDA exposure. These data further implicate dopamine and BDNF in the cognitive deficits arising from developmental MnOE.
Collapse
Affiliation(s)
- Rebecca A Bailey
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Arnold Gutierrez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Tara L Kyser
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Ann M Hemmerle
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Jillian R Hufgard
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| | - Kim B Seroogy
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Charles V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Michael T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA.
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
10
|
Thompson KJ, Hein J, Baez A, Sosa JC, Wessling-Resnick M. Manganese transport and toxicity in polarized WIF-B hepatocytes. Am J Physiol Gastrointest Liver Physiol 2018; 315:G351-G363. [PMID: 29792530 PMCID: PMC6335010 DOI: 10.1152/ajpgi.00103.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Manganese (Mn) toxicity arises from nutritional problems, community and occupational exposures, and genetic risks. Mn blood levels are controlled by hepatobiliary clearance. The goals of this study were to determine the cellular distribution of Mn transporters in polarized hepatocytes, to establish an in vitro assay for hepatocyte Mn efflux, and to examine possible roles the Mn transporters would play in metal import and export. For these experiments, hepatocytoma WIF-B cells were grown for 12-14 days to achieve maximal polarity. Immunoblots showed that Mn transporters ZIP8, ZnT10, ferroportin (Fpn), and ZIP14 were present. Indirect immunofluorescence microscopy localized Fpn and ZIP14 to WIF-B cell basolateral domains whereas ZnT10 and ZIP8 associated with intracellular vesicular compartments. ZIP8-positive structures were distributed uniformly throughout the cytoplasm, but ZnT10-positive vesicles were adjacent to apical bile compartments. WIF-B cells were sensitive to Mn toxicity, showing decreased viability after 16 h exposure to >250 μM MnCl2. However, the hepatocytes were resistant to 4-h exposures of up to 500 μM MnCl2 despite 50-fold increased Mn content. Washout experiments showed time-dependent efflux with 80% Mn released after a 4 h chase period. Hepcidin reduced levels of Fpn in WIF-B cells, clearing Fpn from the cell surface, but Mn efflux was unaffected. The secretory inhibitor, brefeldin A, did block release of Mn from WIF-B cells, suggesting vesicle fusion may be involved in export. These results point to a possible role of ZnT10 to import Mn into vesicles that subsequently fuse with the apical membrane and empty their contents into bile. NEW & NOTEWORTHY Polarized WIF-B hepatocytes express manganese (Mn) transporters ZIP8, ZnT10, ferroportin (Fpn), and ZIP14. Fpn and ZIP14 localize to basolateral domains. ZnT10-positive vesicles were adjacent to apical bile compartments, and ZIP8-positive vesicles were distributed uniformly throughout the cytoplasm. WIF-B hepatocyte Mn export was resistant to hepcidin but inhibited by brefeldin A, pointing to an efflux mechanism involving ZnT10-mediated uptake of Mn into vesicles that subsequently fuse with and empty their contents across the apical bile canalicular membrane.
Collapse
Affiliation(s)
- Khristy J. Thompson
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Jennifer Hein
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Andrew Baez
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Jose Carlo Sosa
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Marianne Wessling-Resnick
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| |
Collapse
|
11
|
Foster ML, Bartnikas TB, Maresca-Fichter HC, Mercadante C, Dash M, Miller C, Dorman DC. Neonatal C57BL/6J and parkin mice respond differently following developmental manganese exposure: Result of a high dose pilot study. Neurotoxicology 2017; 64:291-299. [PMID: 29020610 DOI: 10.1016/j.neuro.2017.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 10/02/2017] [Accepted: 10/03/2017] [Indexed: 11/28/2022]
Abstract
It has been suggested that childhood exposure to neurotoxicants may increase the risk of Parkinson's disease (PD) or other neurodegenerative disease in adults. Some recessive forms of PD have been linked to loss-of-function mutations in the Park2 gene that encodes for parkin. The purpose of this pilot study was to evaluate whether responses to neonatal manganese (Mn) exposure differ in mice with a Park2 gene defect (parkin mice) when compared with a wildtype strain (C57BL/6J). Neonatal parkin and C57BL/6J littermates were randomly assigned to 0, 11, or 25mg Mn/kg-day dose groups with oral exposures occurring from postnatal day (PND) 1 through PND 28. Motor activity was measured on PND 19-22 and 29-32. Tissue Mn concentrations were measured in liver, femur, olfactory bulb, frontal cortex, and striatum on PND 29. Hepatic and frontal cortex gene expression of Slc11a2, Slc40a1, Slc30a10, Hamp (liver only), and Park2 were also measured on PND 29. Some strain differences were seen. As expected, decreased hepatic and frontal cortex Park2 expression was seen in the parkin mice when compared with C57BL/6J mice. Untreated parkin mice also had higher liver and femur Mn concentrations when compared with the C57BL/6J mice. Exposure to≥11mg Mn/kg-day was associated with increased brain Mn concentrations in all mice, no strain difference was observed. Manganese exposure in C57Bl6, but not parkin mice, was associated with a negative correlation between striatal Mn concentration and motor activity. Manganese exposure was not associated with changes in frontal cortex gene expression. Decreased hepatic Slc30a10, Slc40a1, and Hamp expression were seen in PND 29 C57BL/6J mice given 25mg Mn/kg-day. In contrast, Mn exposure was only associated with decreased Hamp expression in the parkin mice. Our results suggest that the Parkin gene defect did not increase the susceptibility of neonatal mice to adverse health effects associated with high-dose Mn exposure.
Collapse
Affiliation(s)
- Melanie L Foster
- North Carolina State University, College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, NC 27607, USA.
| | - Thomas B Bartnikas
- Brown University, Department of Pathology and Laboratory Medicine, 70 Ship St., Rm. 522, Providence, RI 02912, USA.
| | - Hailey C Maresca-Fichter
- North Carolina State University, College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, NC 27607, USA.
| | - Courtney Mercadante
- Brown University, Department of Pathology and Laboratory Medicine, 70 Ship St., Rm. 522, Providence, RI 02912, USA.
| | - Miriam Dash
- Brown University, Department of Pathology and Laboratory Medicine, 70 Ship St., Rm. 522, Providence, RI 02912, USA.
| | - Chelsea Miller
- Brown University, Department of Pathology and Laboratory Medicine, 70 Ship St., Rm. 522, Providence, RI 02912, USA.
| | - David C Dorman
- North Carolina State University, College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, NC 27607, USA.
| |
Collapse
|
12
|
Shin HW, Park HK. Recent Updates on Acquired Hepatocerebral Degeneration. Tremor Other Hyperkinet Mov (N Y) 2017; 7:463. [PMID: 28975044 PMCID: PMC5623760 DOI: 10.7916/d8tb1k44] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 07/24/2017] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND Acquired hepatocerebral degeneration (AHD) refers to a chronic neurological syndrome in patients with advanced hepatobiliary diseases. This comprehensive review focuses on the pathomechanism and neuroimaging findings in AHD. METHODS A PubMed search was performed using the terms "acquired hepatocerebral degeneration," "chronic hepatocerebral degeneration," "Non-Wilsonian hepatocerebral degeneration," "cirrhosis-related parkinsonism," and "manganese and liver disease." RESULTS Multiple mechanisms involving the accumulation of toxic substances such as ammonia or manganese and neuroinflammation may lead to widespread neurodegeneration in AHD. Clinical characteristics include movement disorders, mainly parkinsonism and ataxia-plus syndrome, as well as cognitive impairment with psychiatric features. Neuroimaging studies of AHD with parkinsonism show hyperintensity in the bilateral globus pallidus on T1-weighted magnetic resonance images, whereas molecular imaging of the presynaptic dopaminergic system shows variable findings. Ataxia-plus syndrome in AHD may demonstrate high-signal lesions in the middle cerebellar peduncles on T2-weighted images. DISCUSSION Future studies are needed to elucidate the exact pathomechanism and neuroimaging findings of this heterogeneous syndrome.
Collapse
Affiliation(s)
- Hae-Won Shin
- Department of Neurology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institute of Health, Bethesda, MD, USA
| | - Hee Kyung Park
- Department of Neurology, Inje University Ilsan-Paik Hospital, Goyang, Republic of Korea
- Movement Disorder Center, Department of Neurosciences, University of California San Diego, San Diego, CA, USA
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
13
|
Liu JP, Li J, Lu Y, Wang L, Chen G. Impulse control disorder, lysosomal malfunction and ATP13A2 insufficiency in Parkinsonism. Clin Exp Pharmacol Physiol 2016; 44:172-179. [PMID: 27997702 DOI: 10.1111/1440-1681.12714] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 12/14/2016] [Accepted: 12/15/2016] [Indexed: 12/27/2022]
Abstract
Lysosomal transport of cargos in neurons is essential for neuronal proteostasis, transmission and functional motors and behaviours. Lysosomal malfunction including storage disorders is involved in the pathogenesis of Parkinson's disease (PD). Given the unclear molecular mechanisms of diverse defects in PD phenotypes, especially behavioural deficits, this mini review explores the cellular contexts of PD impulse control disorders and the molecular aspects of lysosomal cross-membrane transports. Focuses are paid to trace metal involvements in α-synuclein assembly in Lewy bodies, the functions and molecular interactions of ATP13A2 as ATPase transporters in lysosomal membranes for cross-membrane trafficking and lysosomal homeostasis, and our current understandings of the neural circuits in ICD. Erroneously polarized distributions of cargos such as metals and lipids on each side of lysosomal membranes triggered by gene mutations and deregulated expression of ATP13A2 may thus instigate sensing protein structural changes such as aggregations, organelle degeneration, and specific neuronal ageing and death in Parkinsonism.
Collapse
Affiliation(s)
- Jun-Ping Liu
- School of Medicine, Institute of Ageing Research, Hangzhou Normal University, Hangzhou, Zhejiang Province, China.,Department of Molecular and Translational Science, Faculty of Medicine, Hudson Institute of Medical Research, Monash University, Clayton, VIC, Australia.,Department of Immunology, Faculty of Medicine, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Jianfeng Li
- School of Medicine, Institute of Ageing Research, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Yanhua Lu
- School of Medicine, Institute of Ageing Research, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Lihui Wang
- School of Medicine, Institute of Ageing Research, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Gang Chen
- School of Medicine, Institute of Ageing Research, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
14
|
Tarale P, Sivanesan S, Daiwile AP, Stöger R, Bafana A, Naoghare PK, Parmar D, Chakrabarti T, Kannan K. Global DNA methylation profiling of manganese-exposed human neuroblastoma SH-SY5Y cells reveals epigenetic alterations in Parkinson's disease-associated genes. Arch Toxicol 2016; 91:2629-2641. [PMID: 27913844 DOI: 10.1007/s00204-016-1899-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 11/24/2016] [Indexed: 01/28/2023]
Abstract
Manganese (Mn) is an essential trace element required for optimal functioning of cellular biochemical pathways in the central nervous system. Elevated exposure to Mn through environmental and occupational exposure can cause neurotoxic effects resulting in manganism, a condition with clinical symptoms identical to idiopathic Parkinson's disease. Epigenetics is now recognized as a biological mechanism involved in the etiology of various diseases. Here, we investigated the role of DNA methylation alterations induced by chronic Mn (100 µM) exposure in human neuroblastoma (SH-SY5Y) cells in relevance to Parkinson's disease. A combined analysis of DNA methylation and gene expression data for Parkinson's disease-associated genes was carried out. Whole-genome bisulfite conversion and sequencing indicate epigenetic perturbation of key genes involved in biological processes associated with neuronal cell health. Integration of DNA methylation data with gene expression reveals epigenetic alterations to PINK1, PARK2 and TH genes that play critical roles in the onset of Parkinsonism. The present study suggests that Mn-induced alteration of DNA methylation of PINK1-PARK2 may influence mitochondrial function and promote Parkinsonism. Our findings provide a basis to further explore and validate the epigenetic basis of Mn-induced neurotoxicity .
Collapse
Affiliation(s)
- Prashant Tarale
- Environmental Health Division, CSIR - National Environmental Engineering Research Institute (NEERI), Nagpur, 440020, India.,Schools of Biosciences, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
| | - Saravanadevi Sivanesan
- Environmental Health Division, CSIR - National Environmental Engineering Research Institute (NEERI), Nagpur, 440020, India.
| | - Atul P Daiwile
- Environmental Health Division, CSIR - National Environmental Engineering Research Institute (NEERI), Nagpur, 440020, India
| | - Reinhard Stöger
- Schools of Biosciences, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
| | - Amit Bafana
- Environmental Health Division, CSIR - National Environmental Engineering Research Institute (NEERI), Nagpur, 440020, India
| | - Pravin K Naoghare
- Environmental Health Division, CSIR - National Environmental Engineering Research Institute (NEERI), Nagpur, 440020, India
| | - Devendra Parmar
- Developmental Toxicology Division, CSIR-Indian Institute of Toxicology Research (IITR), Lucknow, 226001, India
| | - Tapan Chakrabarti
- Visvesvaraya National Institute of Technology (VNIT), Nagpur, 440010, India
| | - Krishnamurthi Kannan
- Environmental Health Division, CSIR - National Environmental Engineering Research Institute (NEERI), Nagpur, 440020, India
| |
Collapse
|
15
|
Disease-Toxicant Interactions in Parkinson's Disease Neuropathology. Neurochem Res 2016; 42:1772-1786. [PMID: 27613618 DOI: 10.1007/s11064-016-2052-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 08/25/2016] [Accepted: 08/29/2016] [Indexed: 12/15/2022]
Abstract
Human disease commonly manifests as a result of complex genetic and environmental interactions. In the case of neurodegenerative diseases, such as Parkinson's disease (PD), understanding how environmental exposures collude with genetic polymorphisms in the central nervous system to cause dysfunction is critical in order to develop better treatment strategies, therapies, and a more cohesive paradigm for future research. The intersection of genetics and the environment in disease etiology is particularly relevant in the context of their shared pathophysiological mechanisms. This review offers an integrated view of disease-toxicant interactions in PD. Particular attention is dedicated to how mutations in the genes SNCA, parkin, leucine-rich repeat kinase 2 (LRRK2) and DJ-1, as well as dysfunction of the ubiquitin proteasome system, may contribute to PD and how exposure to heavy metals, pesticides and illicit drugs may further the consequences of these mutations to exacerbate PD and PD-like disorders. Although the toxic effects induced by exposure to these environmental factors may not be the primary causes of PD, their mechanisms of action are critical for our current understanding of the neuropathologies driving PD. Elucidating how environment and genetics collude to cause pathogenesis of PD will facilitate the development of more effective treatments for the disease. Additionally, we discuss the neuroprotection exerted by estrogen and other compounds that may prevent PD and provide an overview of current treatment strategies and therapies.
Collapse
|
16
|
Hubbs-Tait L, Nation JR, Krebs NF, Bellinger DC. Neurotoxicants, Micronutrients, and Social Environments. Psychol Sci Public Interest 2016; 6:57-121. [DOI: 10.1111/j.1529-1006.2005.00024.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
SUMMARY—Systematic research evaluating the separate and interacting impacts of neurotoxicants, micronutrients, and social environments on children's cognition and behavior has only recently been initiated. Years of extensive human epidemiologic and animal experimental research document the deleterious impact of lead and other metals on the nervous system. However, discrepancies among human studies and between animal and human studies underscore the importance of variations in child nutrition as well as social and behavioral aspects of children's environments that mitigate or exacerbate the effects of neurotoxicants. In this monograph, we review existing research on the impact of neurotoxic metals, nutrients, and social environments and interactions across the three domains. We examine the literature on lead, mercury, manganese, and cadmium in terms of dispersal, epidemiology, experimental animal studies, effects of social environments, and effects of nutrition. Research documenting the negative impact of lead on cognition and behavior influenced reductions by the Center for Disease Control in child lead-screening guidelines from 30 micrograms per deciliter (μg/dL) in 1975 to 25 μg/dL in 1985 and to 10 μg/dL in 1991. A further reduction is currently being considered. Experimental animal research documents lead's alteration of glutamate-neurotransmitter (particularly N-methyl-D-aspartate) activity vital to learning and memory. In addition, lead induces changes in cholinergic and dopaminergic activity. Elevated lead concentrations in the blood are more common among children living in poverty and there is some evidence that socioeconomic status influences associations between lead and child outcomes. Micronutrients that influence the effects of lead include iron and zinc. Research documenting the negative impact of mercury on children (as well as adults) has resulted in a reference dose (RfD) of 0.1 microgram per kilogram of body weight per day (μg/kg/day). In animal studies, mercury interferes with glutamatergic, cholinergic, and dopaminergic activity. Although evidence for interactions of mercury with children's social contexts is minimal, researchers are examining interactions of mercury with several nutrients. Research on the effects of cadmium and manganese on child cognition and behavior is just beginning. Experimental animal research links cadmium to learning deficits, manganese to behaviors characteristic of Parkinson's disease, and both to altered dopaminergic functioning. We close our review with a discussion of policy implications, and we recommend interdisciplinary research that will enable us to bridge gaps within and across domains.
Collapse
Affiliation(s)
- Laura Hubbs-Tait
- Department of Human Development and Family Science, Oklahoma State University
| | | | - Nancy F. Krebs
- Department of Pediatrics, University of Colorado School of Medicine
| | - David C. Bellinger
- Department of Neurology, Harvard Medical School; Department of Environmental Health, Harvard School of Public Health; and Children's Hospital Boston
| |
Collapse
|
17
|
Potential Role of Epigenetic Mechanism in Manganese Induced Neurotoxicity. BIOMED RESEARCH INTERNATIONAL 2016; 2016:2548792. [PMID: 27314012 PMCID: PMC4899583 DOI: 10.1155/2016/2548792] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/08/2016] [Indexed: 02/07/2023]
Abstract
Manganese is a vital nutrient and is maintained at an optimal level (2.5–5 mg/day) in human body. Chronic exposure to manganese is associated with neurotoxicity and correlated with the development of various neurological disorders such as Parkinson's disease. Oxidative stress mediated apoptotic cell death has been well established mechanism in manganese induced toxicity. Oxidative stress has a potential to alter the epigenetic mechanism of gene regulation. Epigenetic insight of manganese neurotoxicity in context of its correlation with the development of parkinsonism is poorly understood. Parkinson's disease is characterized by the α-synuclein aggregation in the form of Lewy bodies in neuronal cells. Recent findings illustrate that manganese can cause overexpression of α-synuclein. α-Synuclein acts epigenetically via interaction with histone proteins in regulating apoptosis. α-Synuclein also causes global DNA hypomethylation through sequestration of DNA methyltransferase in cytoplasm. An individual genetic difference may also have an influence on epigenetic susceptibility to manganese neurotoxicity and the development of Parkinson's disease. This review presents the current state of findings in relation to role of epigenetic mechanism in manganese induced neurotoxicity, with a special emphasis on the development of Parkinson's disease.
Collapse
|
18
|
Miranda M, Bustamante ML, Mena F, Lees A. Original footage of the Chilean miners with manganism published in Neurology in 1967. Neurology 2016; 85:2166-9. [PMID: 26668239 DOI: 10.1212/wnl.0000000000002223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Manganism has captured the imagination of neurologists for more than a century because of its similarities to Parkinson disease and its indirect but seminal role in the "l-dopa miracle." We present unpublished footage of the original case series reported in Neurology® in 1967 by Mena and Cotzias depicting the typical neurologic signs of manganism in 4 Chilean miners and their response to high doses of l-dopa.
Collapse
Affiliation(s)
- Marcelo Miranda
- From the Departments of Neurology (M.M.) and Radiology (F.M.), Clinica Las Condes; the Program of Human Genetics, Biomedical Sciences Institute, and Department of Psychiatry and Mental Health, North Division, Faculty of Medicine (M.L.B.), and Biomedical Neurosciences Institute, Faculty of Medicine (M.L.B.), Universidad de Chile, Santiago; and Reta Lila Weston Institute of Neurological Sciences (A.L.), Institute of Neurology, University College London, UK.
| | - M Leonor Bustamante
- From the Departments of Neurology (M.M.) and Radiology (F.M.), Clinica Las Condes; the Program of Human Genetics, Biomedical Sciences Institute, and Department of Psychiatry and Mental Health, North Division, Faculty of Medicine (M.L.B.), and Biomedical Neurosciences Institute, Faculty of Medicine (M.L.B.), Universidad de Chile, Santiago; and Reta Lila Weston Institute of Neurological Sciences (A.L.), Institute of Neurology, University College London, UK
| | - Francisco Mena
- From the Departments of Neurology (M.M.) and Radiology (F.M.), Clinica Las Condes; the Program of Human Genetics, Biomedical Sciences Institute, and Department of Psychiatry and Mental Health, North Division, Faculty of Medicine (M.L.B.), and Biomedical Neurosciences Institute, Faculty of Medicine (M.L.B.), Universidad de Chile, Santiago; and Reta Lila Weston Institute of Neurological Sciences (A.L.), Institute of Neurology, University College London, UK
| | - Andrew Lees
- From the Departments of Neurology (M.M.) and Radiology (F.M.), Clinica Las Condes; the Program of Human Genetics, Biomedical Sciences Institute, and Department of Psychiatry and Mental Health, North Division, Faculty of Medicine (M.L.B.), and Biomedical Neurosciences Institute, Faculty of Medicine (M.L.B.), Universidad de Chile, Santiago; and Reta Lila Weston Institute of Neurological Sciences (A.L.), Institute of Neurology, University College London, UK
| |
Collapse
|
19
|
Menon AV, Chang J, Kim J. Mechanisms of divalent metal toxicity in affective disorders. Toxicology 2015; 339:58-72. [PMID: 26551072 DOI: 10.1016/j.tox.2015.11.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/19/2015] [Accepted: 11/03/2015] [Indexed: 01/01/2023]
Abstract
Metals are required for proper brain development and play an important role in a number of neurobiological functions. The divalent metal transporter 1 (DMT1) is a major metal transporter involved in the absorption and metabolism of several essential metals like iron and manganese. However, non-essential divalent metals are also transported through this transporter. Therefore, altered expression of DMT1 can modify the absorption of toxic metals and metal-induced toxicity. An accumulating body of evidence has suggested that increased metal stores in the brain are associated with elevated oxidative stress promoted by the ability of metals to catalyze redox reactions, resulting in abnormal neurobehavioral function and the progression of neurodegenerative diseases. Metal overload has also been implicated in impaired emotional behavior, although the underlying mechanisms are not well understood with limited information. The current review focuses on psychiatric dysfunction associated with imbalanced metabolism of metals that are transported by DMT1. The investigations with respect to the toxic effects of metal overload on behavior and their underlying mechanisms of toxicity could provide several new therapeutic targets to treat metal-associated affective disorders.
Collapse
Affiliation(s)
| | - JuOae Chang
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Jonghan Kim
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Manganese-Induced Parkinsonism and Parkinson's Disease: Shared and Distinguishable Features. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:7519-40. [PMID: 26154659 PMCID: PMC4515672 DOI: 10.3390/ijerph120707519] [Citation(s) in RCA: 226] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/12/2014] [Accepted: 01/06/2015] [Indexed: 11/30/2022]
Abstract
Manganese (Mn) is an essential trace element necessary for physiological processes that support development, growth and neuronal function. Secondary to elevated exposure or decreased excretion, Mn accumulates in the basal ganglia region of the brain and may cause a parkinsonian-like syndrome, referred to as manganism. The present review discusses the advances made in understanding the essentiality and neurotoxicity of Mn. We review occupational Mn-induced parkinsonism and the dynamic modes of Mn transport in biological systems, as well as the detection and pharmacokinetic modeling of Mn trafficking. In addition, we review some of the shared similarities, pathologic and clinical distinctions between Mn-induced parkinsonism and Parkinson’s disease. Where possible, we review the influence of Mn toxicity on dopamine, gamma aminobutyric acid (GABA), and glutamate neurotransmitter levels and function. We conclude with a survey of the preventive and treatment strategies for manganism and idiopathic Parkinson’s disease (PD).
Collapse
|
21
|
Chen P, Chakraborty S, Mukhopadhyay S, Lee E, Paoliello MMB, Bowman AB, Aschner M. Manganese homeostasis in the nervous system. J Neurochem 2015; 134:601-10. [PMID: 25982296 DOI: 10.1111/jnc.13170] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 04/30/2015] [Accepted: 05/04/2015] [Indexed: 12/31/2022]
Abstract
Manganese (Mn) is an essential heavy metal that is naturally found in the environment. Daily intake through dietary sources provides the necessary amount required for several key physiological processes, including antioxidant defense, energy metabolism, immune function and others. However, overexposure from environmental sources can result in a condition known as manganism that features symptomatology similar to Parkinson's disease (PD). This disorder presents with debilitating motor and cognitive deficits that arise from a neurodegenerative process. In order to maintain a balance between its essentiality and neurotoxicity, several mechanisms exist to properly buffer cellular Mn levels. These include transporters involved in Mn uptake, and newly discovered Mn efflux mechanisms. This review will focus on current studies related to mechanisms underlying Mn import and export, primarily the Mn transporters, and their function and roles in Mn-induced neurotoxicity. Though and essential metal, overexposure to manganese may result in neurodegenerative disease analogous to Parkinson's disease. Manganese homeostasis is tightly regulated by transporters, including transmembrane importers (divalent metal transporter 1, transferrin and its receptor, zinc transporters ZIP8 and Zip14, dopamine transporter, calcium channels, choline transporters and citrate transporters) and exporters (ferroportin and SLC30A10), as well as the intracellular trafficking proteins (SPCA1 and ATP12A2). A manganese-specific sensor, GPP130, has been identified, which affords means for monitoring intracellular levels of this metal.
Collapse
Affiliation(s)
- Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sudipta Chakraborty
- Neuroscience Graduate Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology & Toxicology, College of Pharmacy; Institute for Cellular & Molecular Biology; and Institute for Neuroscience, The University of Texas at Austin, Austin, Texas, USA
| | - Eunsook Lee
- Department of Physiology, Meharry Medical College, Nashville, Tennessee, USA
| | - Monica M B Paoliello
- Graduate Program in Public Health, Department of Pathology, Clinical and Toxicological Analysis, Center of Health Science, State University of Londrina, Parana, Brazil
| | - Aaron B Bowman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA.,Neuroscience Graduate Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
22
|
Huang CY, Liu CH, Tsao E, Hsieh CJ, Weng YH, Hsiao IT, Yen TC, Lin KJ, Huang CC. Chronic manganism: A long-term follow-up study with a new dopamine terminal biomarker of 18F-FP-(+)-DTBZ (18F-AV-133) brain PET scan. J Neurol Sci 2015; 353:102-6. [PMID: 25936253 DOI: 10.1016/j.jns.2015.04.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 10/23/2022]
Abstract
Recent experimental studies revealed that dopamine neuron dysfunction in chronic manganism may be due to a reduced capacity of dopamine release in the striatum. The findings imposed further difficulty in the differential diagnosis between manganism and IPD. We conducted a long-term clinical follow-up study of 4 manganism patients, applying a new tracer (18)F-9-fluoropropyl-(+)-dihydrotetrabenazine ((18)F-AV-133) with positron emission tomography (PET). Twenty age-matched subjects including 4 manganism patients, 8 idiopathic Parkinson's disease (IPD) patients, and 8 healthy controls were enrolled for comparison. Volumes of interest of the bilateral putamen, caudate nuclei and occipital cortex as the reference region were delineated from individual magnetic resonance images. The clinical features of the manganism patients still progressed, with increased scores on the Unified Parkinson Disease Rating Scale. The (18)F-AV-133 uptake in the IPD patients decreased at the bilateral striatum, compared with the healthy controls. In the manganism patients, there was no decreased uptake of radioactivity involving the bilateral striatum, except Patient 4, who had a stroke with decreased uptake in the right posterior putamen. The (18)F-AV-133 PET finding reveals that nigrostriatum neurons are not degenerated in chronic manganism and can provide a useful neuroimage biomarker in the differential diagnosis.
Collapse
Affiliation(s)
- Chu-Yun Huang
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chi-Hung Liu
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, Division of Medical Education, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Eusden Tsao
- Department of Family Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Environmental and Occupational Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chia-Ju Hsieh
- Molecular Imaging Center and Nuclear Medicine, Chang Gung University, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Healthy Aging Research Center Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Hsin Weng
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Neurology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ing-Tsung Hsiao
- Molecular Imaging Center and Nuclear Medicine, Chang Gung University, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Healthy Aging Research Center Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tzu-Chen Yen
- Molecular Imaging Center and Nuclear Medicine, Chang Gung University, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Healthy Aging Research Center Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kun-Ju Lin
- Molecular Imaging Center and Nuclear Medicine, Chang Gung University, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Healthy Aging Research Center Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chin-Chang Huang
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Neurology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
23
|
Abstract
In recent years, the contribution of exposure to environmental toxicants has been recognized as a significant contributor to the etiopathogenesis of parkinsonism. Of these toxicants, exposure to pesticides, metals, solvents used in manufacturing processes, as well as flame-retardant chemicals used in consumer and commercial products, has received the greatest attention as possible risk factors. Related to this, individuals who are exposed to these compounds at high concentrations or for prolonged periods of time in an occupational setting appear to be one of the more vulnerable populations to these effects. Our understanding of which compounds are involved and the potential molecular pathways that are susceptible to these chemicals and may underlie the pathogenesis has greatly improved. However, there are still hundreds of chemicals that we are exposed to in the environment for which we do not have any information on their potential neurotoxicity on the nigrostriatal dopamine system. Thus, using our past accomplishments as a blueprint, future endeavors should focus on elaborating upon these initial findings in order to identify specific and relevant chemical toxicants in our environment that can impact the risk of parkinsonism and work towards a means to attenuate or abolish their effects on the human population.
Collapse
Affiliation(s)
- W Michael Caudle
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
24
|
Racette BA. Manganism in the 21st century: the Hanninen lecture. Neurotoxicology 2014; 45:201-7. [PMID: 24148923 PMCID: PMC3992192 DOI: 10.1016/j.neuro.2013.09.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/27/2013] [Accepted: 09/22/2013] [Indexed: 11/24/2022]
Abstract
Since the original description of the health effects of inhaled occupational manganese (Mn) by Couper in 1837, an extensive literature details the clinical syndrome and pathophysiology of what was thought to be a rare condition. In the last decade, conventional wisdom regarding the clinicopathological effects of Mn has been challenged. Past exposures to Mn were an order of magnitude higher than modern exposures in developed countries; therefore, the clinical syndrome seen in the time of Couper is no longer typical of modern Mn exposed workers. Parkinsonism (rigidity, bradykinesia, rest tremor, and postural instability) is present in 15% of Mn-exposed workers in welding industries, and these parkinsonian signs are associated with reduced health status and quality of life. These parkinsonian signs also overlap considerably with the clinical findings seen in early stages of Parkinson's disease (PD); although, molecular imaging suggests that Mn-exposed workers have dopaminergic dysfunction in a pattern unique from PD. Furthermore, geographic information system studies demonstrate that regions of the US with high industrial Mn emissions have an increased incidence of PD and increased PD associated mortality. This review will contrast historical, descriptive human studies in Mn-exposed subjects with more recent data and will suggest a research agenda for the 21st century.
Collapse
Affiliation(s)
- Brad A Racette
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO 63110, USA; University of the Witwatersrand, School of Public Health, Faculty of Health Sciences, Johannesburg, South Africa.
| |
Collapse
|
25
|
Correlation between the biochemical pathways altered by mutated parkinson-related genes and chronic exposure to manganese. Neurotoxicology 2014; 44:314-25. [DOI: 10.1016/j.neuro.2014.08.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 08/11/2014] [Accepted: 08/11/2014] [Indexed: 01/02/2023]
|
26
|
Zaitout Z, Romanowski C, Karunasaagarar K, Connolly D, Batty R. A review of pathologies associated with high T1W signal intensity in the basal ganglia on Magnetic Resonance Imaging. Pol J Radiol 2014; 79:126-30. [PMID: 24900164 PMCID: PMC4043538 DOI: 10.12659/pjr.890043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 12/03/2013] [Indexed: 11/09/2022] Open
Abstract
With several functions and a fundamental influence over cognition and motor functions, the basal ganglia are the cohesive centre of the brain. There are several conditions which affect the basal ganglia and these have various clinical and radiological manifestations. Nevertheless, on magnetic resonance imaging there is a limited differential diagnosis for those conditions presenting with T1 weighted spin echo hyperintensity within the central nervous system in general and the basal ganglia in particular. The aim of our review is to explore some of these basal ganglia pathologies and provide image illustrations.
Collapse
Affiliation(s)
- Zahia Zaitout
- Department of Radiology, St Bartholomew Hospital, London, U.K
| | | | | | - Daniel Connolly
- Department of Radiology, Royal Hallamshire Hospital, Sheffield, U.K
| | - Ruth Batty
- Department of Radiology, Royal Hallamshire Hospital, Sheffield, U.K
| |
Collapse
|
27
|
Koksal A, Keskinkilic C, Sozmen MV, Dirican AC, Aysal F, Altunkaynak Y, Baybas S. Evaluation of Cognitive Characteristics of Patients Developing Manifestations of Parkinsonism Secondary to Long-Term Ephedrone Use. Eur Neurol 2014; 71:208-12. [DOI: 10.1159/000356509] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/20/2013] [Indexed: 11/19/2022]
|
28
|
Li P, Zhu J, Kong Q, Jiang B, Wan X, Yue J, Li M, Jiang H, Li J, Gao Z. The ethylene bis-dithiocarbamate fungicide Mancozeb activates voltage-gated KCNQ2 potassium channel. Toxicol Lett 2013; 219:211-7. [DOI: 10.1016/j.toxlet.2013.03.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 03/15/2013] [Accepted: 03/20/2013] [Indexed: 10/27/2022]
|
29
|
The effect of manganese on dopamine toxicity and dopamine transporter (DAT) in control and DAT transfected HEK cells. Neurotoxicology 2013; 35:121-8. [PMID: 23313730 DOI: 10.1016/j.neuro.2013.01.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/02/2013] [Accepted: 01/04/2013] [Indexed: 11/23/2022]
Abstract
Chronic exposure to Mn results in the development of a neurological disorder known as manganism characterized by neurological deficits resembling that seen in Parkinsonism. Although dopaminergic neurons within the nigrostriatal pathway appear intact, Mn-induced irregularities in DA transmission have been observed including decreased amphetamine-induced DA release and loss of the dopamine transporter (DAT). Results of studies to evaluate the effect of Mn and DA on cell viability in control and DAT-transfected HEK cells reveal that Mn is equally toxic to both cell lines whereas DA was only toxic to cells containing DAT. DA toxicity was saturable suggesting that transport may be rate limiting. When Mn and DA were added simultaneously to the media, cell toxicity was similar to that produced by Mn alone suggesting that Mn may suppress DA uptake in the DAT containing cells. Preincubation of DA prior to the addition of Mn resulted in cell death which was essentially additive with that produced independently by the two agents. Mn was also shown to decrease DA uptake and amphetamine-induced DA efflux in DAT containing cells. Time-lapsed confocal microscopy indicates that Mn can promote trafficking of cell surface DAT into intracellular compartments which may account for the decrease in DA uptake and DA efflux in these cells. Mn-induced internalization of DAT may provide an explanation for disruption in DA transmission previously reported in the striatum.
Collapse
|
30
|
Abstract
The review addresses issues pertinent to Mn accumulation and its mechanisms of transport, its neurotoxicity and mechanisms of neurodegeneration. The role of mitochondria and glia in this process is emphasized. We also discuss gene x environment interactions, focusing on the interplay between genes linked to Parkinson's disease (PD) and sensitivity to Mn.
Collapse
Affiliation(s)
- Jerome Roth
- Department of Pharmacology and Toxicology, University at Buffalo School of Medicine, 11 Cary Hall, Buffalo, NY, 14214, USA
| | | | | |
Collapse
|
31
|
Abstract
This article schematically reviews the clinical features, diagnostic approaches to, and toxicological implications of toxic encephalopathy. The review will focus on the most significant occupational causes of toxic encephalopathy. Chronic toxic encephalopathy, cerebellar syndrome, parkinsonism, and vascular encephalopathy are commonly encountered clinical syndromes of toxic encephalopathy. Few neurotoxins cause patients to present with pathognomonic neurological syndromes. The symptoms and signs of toxic encephalopathy may be mimicked by many psychiatric, metabolic, inflammatory, neoplastic, and degenerative diseases of the nervous system. Thus, the importance of good history-taking that considers exposure and a comprehensive neurological examination cannot be overemphasized in the diagnosis of toxic encephalopathy. Neuropsychological testing and neuroimaging typically play ancillary roles. The recognition of toxic encephalopathy is important because the correct diagnosis of occupational disease can prevent others (e.g., workers at the same worksite) from further harm by reducing their exposure to the toxin, and also often provides some indication of prognosis. Physicians must therefore be aware of the typical signs and symptoms of toxic encephalopathy, and close collaborations between neurologists and occupational physicians are needed to determine whether neurological disorders are related to occupational neurotoxin exposure.
Collapse
|
32
|
Roth JA, Ganapathy B, Ghio AJ. Manganese-induced toxicity in normal and human B lymphocyte cell lines containing a homozygous mutation in parkin. Toxicol In Vitro 2012; 26:1143-9. [PMID: 22841634 DOI: 10.1016/j.tiv.2012.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 07/10/2012] [Accepted: 07/12/2012] [Indexed: 01/01/2023]
Abstract
Mutations in the parkin gene are linked to development of juvenile onset of Parkinson's disease and recent studies have reported that parkin can protect against increased oxidative stress and mitochondrial dysfunction caused by a variety of oxidative and toxic insults. Overexpression of parkin has also been reported to selectively protect dopaminergic neurons from Mn toxicity. Accordingly, in this paper we compare the effect that mutations in parkin have on Mn toxicity and associated apoptotic signals in normal and human B lymphocyte cell lines containing a homozygous mutation in the gene. Results of these studies reveal that Mn toxicity was similar in both control and mutant parkin lymphocyte cells indicating that cell death caused by Mn was not altered in cells devoid of parkin activity. In contrast, Mn did inhibit mitochondrial function to a greater extent in cells devoid of active parkin as indicated by a decrease in ATP production although mitochondrial membrane potential was essentially unaffected. Consistent with inactive parkin influencing the Mn response is the observation of increased activity in the down-stream apoptotic signal, caspase 3. In summary, results reported in this paper demonstrate that mutations in parkin can lead to functional changes in potential signaling processes known to provoke Mn toxicity. The selectivity and magnitude of this response, however, does not necessarily lead to cell death in lymphocytes which are devoid of dopamine.
Collapse
Affiliation(s)
- Jerome A Roth
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, USA.
| | | | | |
Collapse
|
33
|
Roth JA, Sridhar S, Singleton ST. Effect of glutamate and riluzole on manganese-induced apoptotic cell signaling in neuronally differentiated mouse P19 Cells. Neurochem Int 2012; 61:25-33. [PMID: 22543103 DOI: 10.1016/j.neuint.2012.04.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Revised: 04/11/2012] [Accepted: 04/13/2012] [Indexed: 12/21/2022]
Abstract
Excess exposure to Mn causes a neurological disorder known as manganism which is similar to dystonic movements associated with Parkinson's disease. Manganism is largely restricted to occupations in which high atmospheric levels are prevalent which include Mn miners, welders and those employed in the ferroalloy processing or related industrial settings. T1 weighted MRI images reveal that Mn is deposited to the greatest extent in the globus pallidus, an area of the brain that is presumed to be responsible for the major CNS associated symptoms. Neurons within the globus pallidus receive glutamatergic input from the subthalamic nuclei which has been suggested to be involved in the toxic actions of Mn. The neurotoxic actions of Mn and glutamate are similar in that they both affect calcium accumulation in the mitochondria leading to apoptotic cell death. In this paper, we demonstrate that the combination of Mn and glutamate potentiates toxicity of neuronally differentiated P19 cells over that observed with either agent alone. Apoptotic signals ROS, caspase 3 and JNK were increased in an additive fashion when the two neurotoxins were combined. The anti-glutamatergic drug, riluzole, was shown to attenuate these apoptotic signals and prevent P19 cell death. Results of this study confirm, for the first time, that Mn toxicity is potentiated in the presence of glutamate and that riluzole is an effective antioxidant which protects against both Mn and glutamate toxicity.
Collapse
Affiliation(s)
- Jerome A Roth
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, USA.
| | | | | |
Collapse
|
34
|
Negga R, Stuart JA, Machen ML, Salva J, Lizek AJ, Richardson SJ, Osborne AS, Mirallas O, McVey KA, Fitsanakis VA. Exposure to glyphosate- and/or Mn/Zn-ethylene-bis-dithiocarbamate-containing pesticides leads to degeneration of γ-aminobutyric acid and dopamine neurons in Caenorhabditis elegans. Neurotox Res 2012; 21:281-90. [PMID: 21922334 PMCID: PMC3288661 DOI: 10.1007/s12640-011-9274-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 08/18/2011] [Accepted: 08/31/2011] [Indexed: 12/14/2022]
Abstract
Previous studies demonstrate a positive correlation between pesticide usage and Parkinson's disease (PD), which preferentially targets dopaminergic (DAergic) neurons. In order to examine the potential relationship between two common pesticides and specific neurodegeneration, we chronically (24 h) or acutely (30 min) exposed two Caenorhabditis elegans (C. elegans) strains to varying concentrations (LC(25), LC(50) or LC(75)) of TouchDown(®) (TD) as percent active ingredient (glyphosate), or Mancozeb(®) (MZ) as percent active ingredient (manganese/zinc ethylene-bis-dithiocarbamate). Furthermore, to more precisely model environmental exposure, worms were also exposed to TD for 30 min, followed by 30-min incubation with varying MZ concentrations. Previous data from out lab suggested general neuronal degeneration using the worm strain NW1229 (pan-neuronal//green fluorescent protein (GFP) construct). To determine whether distinct neuronal groups were preferentially affected, we specifically used EG1285 (GABAergic neurons//GFP construct) and BZ555 (DAergic neurons//GFP construct) worms to verify GABAergic and DAergic neurodegeneration, respectively. Results indicated a statistically significant decrease, when compared to controls (CN), in number of green pixels associated with GABAergic neurons in both chronic (*P < 0.05) and acute (*P < 0.05) treatment paradigms. Analysis of the BZ555 worms indicated a statistically significant decrease (*P < 0.05) in number of green pixels associated with DAergic neurons in both treatment paradigms (chronic and acute) when compared to CN. Taken together, our data suggest that exposure to TD and/or MZ promotes neurodegeneration in both GABAergic and DAergic neurons in the model organism C. elegans.
Collapse
Affiliation(s)
- Rekek Negga
- King College, Department of Biology, 1350 King College Road, Bristol, TN 37620 USA
| | - J Andrew Stuart
- King College, Department of Biology, 1350 King College Road, Bristol, TN 37620 USA
| | - Morgan L Machen
- King College, Department of Biology, 1350 King College Road, Bristol, TN 37620 USA
| | - Joel Salva
- King College, Department of Biology, 1350 King College Road, Bristol, TN 37620 USA
| | - Amanda J Lizek
- King College, Department of Biology, 1350 King College Road, Bristol, TN 37620 USA
| | - S Jayne Richardson
- King College, Department of Biology, 1350 King College Road, Bristol, TN 37620 USA
| | - Amanda S Osborne
- King College, Department of Biology, 1350 King College Road, Bristol, TN 37620 USA
| | - Oriol Mirallas
- King College, Department of Biology, 1350 King College Road, Bristol, TN 37620 USA
| | - Kenneth A McVey
- King College, Department of Biology, 1350 King College Road, Bristol, TN 37620 USA
| | - Vanessa A Fitsanakis
- King College, Department of Biology, 1350 King College Road, Bristol, TN 37620 USA
| |
Collapse
|
35
|
Industrial toxicants and Parkinson's disease. Neurotoxicology 2012; 33:178-88. [PMID: 22309908 DOI: 10.1016/j.neuro.2012.01.010] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 01/19/2012] [Accepted: 01/23/2012] [Indexed: 11/22/2022]
Abstract
The exposure of the human population to environmental contaminants is recognized as a significant contributing factor for the development of Parkinson's disease (PD) and other forms of parkinsonism. While pesticides have repeatedly been identified as risk factors for PD, these compounds represent only a subset of environmental toxicants that we are exposed to on a regular basis. Thus, non-pesticide contaminants, such as metals, solvents, and other organohalogen compounds have also been implicated in the clinical and pathological manifestations of these movement disorders and it is these non-pesticide compounds that are the subject of this review. As toxic exposures to these classes of compounds can result in a spectrum of PD or PD-related disorders, it is imperative to appreciate shared clinico-pathological characteristics or mechanisms of action of these compounds in order to further delineate the resultant disorders as well as identify improved preventive strategies or therapeutic interventions.
Collapse
|
36
|
Pathophysiology of manganese-associated neurotoxicity. Neurotoxicology 2011; 33:881-6. [PMID: 22202748 DOI: 10.1016/j.neuro.2011.12.010] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 12/01/2011] [Accepted: 12/13/2011] [Indexed: 11/22/2022]
Abstract
Manganese (Mn) is a well established neurotoxin associated with specific damage to the basal ganglia in humans. The phenotype associated with Mn neurotoxicity was first described in two workers with occupational exposure to Mn oxide (Couper, 1837). Although the description did not use modern clinical terminology, a parkinsonian illness characterized by slowness of movement (bradykinesia), masked facies, and gait impairment (postural instability) appears to have predominated. Nearly 100 years later an outbreak of an atypical parkinsonian illness in a Chilean Mn mine provided a phenotypic description of a fulminant neurologic disorder with parkinsonism, dystonia, and neuropsychiatric symptoms (Rodier, 1955). Exposures associated with this syndrome were massive and an order of magnitude greater than modern exposures (Rodier, 1955; Hobson et al., 2011). The clinical syndrome associated with Mn neurotoxicity has been called manganism. Modern exposures to Mn occur primarily through occupations in the steel industry and welding. These exposures are often chronic and varied, occurring over decades in the healthy workforce. Although the severe neurologic disorder described by Rodier and Couper are no longer seen, several reports have suggested a possible increased risk of neurotoxicity in these workers (Racette et al., 2005b; Bowler et al., 2007; Harris et al., 2011). Based upon limited prior imaging and pathologic investigations into the pathophysiology of neurotoxicity in Mn exposed workers (Huang et al., 2003), many investigators have concluded that the syndrome spares the dopamine system distinguishing manganism from Parkinson disease (PD), the most common cause of parkinsonism in the general population, and a disease with characteristic degenerative changes in the dopaminergic system (Jankovic, 2005). The purpose of this symposium was to highlight recent advances in the understanding of the pathophysiology of Mn associated neurotoxicity from Caenorhabditis elegans to humans. Dr. Aschner's presentation discussed mechanisms of dopaminergic neuronal toxicity in C. elegans and demonstrates a compelling potential role of Mn in dopaminergic degeneration. Dr. Guilarte's experimental, non-human primate model of Mn neurotoxicity suggests that Mn decreases dopamine release in the brain without loss of neuronal integrity markers, including dopamine. Dr. Racette's presentation demonstrates a unique pattern of dopaminergic dysfunction in active welders with chronic exposure to Mn containing welding fumes. Finally, Dr. Dydak presented novel magnetic resonance (MR) spectroscopy data in Mn exposed smelter workers and demonstrated abnormalities in the thalamus and frontal cortex for those workers. This symposium provided some converging evidence of the potential neurotoxic impact of Mn on the dopaminergic system and challenged existing paradigms on the pathophysiology of Mn in the central nervous system.
Collapse
|
37
|
Khalid M, Aoun RA, Mathews TA. Altered striatal dopamine release following a sub-acute exposure to manganese. J Neurosci Methods 2011; 202:182-91. [PMID: 21740928 DOI: 10.1016/j.jneumeth.2011.06.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Revised: 05/05/2011] [Accepted: 06/20/2011] [Indexed: 12/30/2022]
Abstract
Certain metals that are necessary for regulating biological function at trace levels hold the potential to become neurotoxic when in excess. Specifically, chronic exposure to high levels of manganese leads to manganism, a neurological disorder that exhibits both motor and learning deficits similar to Parkinson's disease. Since Parkinson's disease symptomatology is primarily attributed to dopamine neurodegeneration in the striatum, dopamine system dysfunction has been implicated in the onset of manganism. In this study, dopamine system function in the dorsal striatum was evaluated in C57Bl/6 mice, 1, 7, and 21 days following repeated injections of manganese(II) chloride (50 mg/kg, subcutaneous) intermittently for 7 days. Tissue content analysis confirmed the presence of persistent accumulation of manganese in the striatum up to 21 days after cessation of treatment. In vitro fast scan cyclic voltammetry examined the effect of sub-acute manganese on electrically stimulated dopamine release and uptake in the striatum. While no difference was observed in uptake rates following manganese treatment, dopamine release was attenuated on days 7 and 21, compared to control levels. Basal levels of extracellular dopamine determined by the zero net flux microdialysis method were significantly lower in manganese-treated mice at 7 days post-treatment. On the other hand, potassium stimulated increases in extracellular dopamine were attenuated at all three time points. Together, these findings indicate that repeated manganese exposure has long-term effects on the regulation of exocytotic dopamine release in the striatum, which may be involved in the mechanism underlying manganese toxicity.
Collapse
Affiliation(s)
- Madiha Khalid
- Department of Chemistry, Wayne State University, 5101 Cass Ave., Detroit, MI 48202, USA
| | | | | |
Collapse
|
38
|
Madison JL, Wegrzynowicz M, Aschner M, Bowman AB. Gender and manganese exposure interactions on mouse striatal neuron morphology. Neurotoxicology 2011; 32:896-906. [PMID: 21641932 DOI: 10.1016/j.neuro.2011.05.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 05/19/2011] [Accepted: 05/19/2011] [Indexed: 12/15/2022]
Abstract
Gender differences in sensitivity and toxicokinetics of multiple metals have been identified in humans. A recent study suggested that young girls performed worse on intellectual exams than young boys exposed to manganese (Mn) in the environment. Animal studies have shown that Mn exposure causes differential effects on behavior in male compared to female mice. We hypothesized that in response to Mn exposure striatal Mn accumulation and/or striatal medium spiny neuron (MSN) morphology show gender-dependent effects. We evaluated the contribution of gender to neuropathology by examining striatal MSN morphology in male and female mice exposed to Mn. We found that gender played a significant role in alterations of striatal MSN morphology in mice exposed to Mn. Gender-dependent changes were strongest when striatal Mn levels were elevated 24h following the final Mn exposure. Nevertheless, gender-dependent alterations in neuron morphology were still present 3 weeks after the final Mn exposure. Gender differences in neuron morphology were not due to differential striatal Mn accumulation between genders. We conclude that although gender does not affect striatal Mn accumulation, MSN morphology is differentially sensitive to elevated Mn levels.
Collapse
Affiliation(s)
- Jennifer L Madison
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | | | | | | |
Collapse
|
39
|
Verina T, Kiihl SF, Schneider JS, Guilarte TR. Manganese exposure induces microglia activation and dystrophy in the substantia nigra of non-human primates. Neurotoxicology 2011; 32:215-26. [PMID: 21112353 PMCID: PMC3057349 DOI: 10.1016/j.neuro.2010.11.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Revised: 11/18/2010] [Accepted: 11/19/2010] [Indexed: 11/19/2022]
Abstract
Chronic manganese (Mn) exposure produces neurological deficits including a form of parkinsonism that is different from Parkinson's disease (PD). In chronic Mn exposure, dopamine neurons in the substantia nigra (SN) do not degenerate but they appear to be dysfunctional. Further, previous studies have suggested that the substantia nigra pars reticulata (SNr) is affected by Mn. In the present study, we investigated whether chronic Mn exposure induces microglia activation in the substantia nigra pars compacta (SNc) and SNr in Cynomolgus macaques. Animals were exposed to different weekly doses of Mn (3.3-5.0, 5.0-6.7, 8.3-10 mg Mn/kg body weight) and microglia were examined in the substantia nigra using LN3 immunohistochemistry. We observed that in control animals, LN3 labeled microglia were characterized by a resting phenotype. However, in Mn-treated animals, microglia increased in number and displayed reactive changes with increasing Mn exposure. This effect was more prominent in the SNr than in the SNc. In the SNr of animals administered the highest Mn dose, microglia activation was the most advanced and included dystrophic changes. Reactive microglia expressed increased iNOS, L-ferritin, and intracellular ferric iron which were particularly prominent in dystrophic compartments. Our observations indicate that moderate Mn exposure produces structural changes on microglia, which may have significant consequences on their function.
Collapse
Affiliation(s)
- Tatyana Verina
- Department of Environmental Health Sciences, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Samara F Kiihl
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Jay S Schneider
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA
| | - Tomás R Guilarte
- Department of Environmental Health Sciences, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
40
|
McDougall SA, Der-Ghazarian T, Britt CE, Varela FA, Crawford CA. Postnatal manganese exposure alters the expression of D2L and D2S receptor isoforms: relationship to PKA activity and Akt levels. Synapse 2010; 65:583-91. [PMID: 21484877 DOI: 10.1002/syn.20877] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2010] [Accepted: 09/27/2010] [Indexed: 01/04/2023]
Abstract
Postnatal manganese chloride (Mn) exposure causes persistent changes in presynaptic dopamine (DA) functioning (e.g., Mn reduces DA transporter levels and DA uptake), but evidence that Mn affects postsynaptic DA receptors and their associated second messenger systems is equivocal. Therefore, a goal of the present study was to determine whether exposing rats to Mn on postnatal days (PD) 1-21 would cause long-term alterations in D2 long (D2L) and D2 short (D2S) receptors that were detectible in adulthood (i.e., on PD 90). Signaling systems associated with D2 receptors were also assessed. Specifically, we measured protein kinase A (PKA) activity in the dorsal striatum and prefrontal cortex (PFC), whereas immunoblotting was used to quantify phosphorylated Akt (p-Akt) and phosphorylated ERK. Results showed that early Mn exposure caused a persistent elevation of D2L and D2S protein expression in the dorsal striatum, as well as an increase in the number of D2 binding sites. Conversely, Mn reduced D2 specific binding in the PFC on PD 90. PKA activity of Mn-treated rats was enhanced in both the dorsal striatum and PFC, whereas p-Akt levels were elevated in the dorsal striatum. When considered together, these results suggest that postnatal Mn exposure either directly or indirectly alters the functioning of postsynaptic DA receptors. One possibility is that early Mn exposure depresses presynaptic dopaminergic functioning and reduces DA levels, thereby causing an up-regulation of D2 receptors and a dysregulation of DA-associated signaling pathways. An alternative explanation is that early Mn exposure affects D2 receptors and PKA/p-Akt levels via independent mechanisms.
Collapse
Affiliation(s)
- Sanders A McDougall
- Department of Psychology, California State University, San Bernardino, California 92407, USA.
| | | | | | | | | |
Collapse
|
41
|
Guilarte TR. Manganese and Parkinson's disease: a critical review and new findings. ENVIRONMENTAL HEALTH PERSPECTIVES 2010; 118:1071-80. [PMID: 20403794 PMCID: PMC2920085 DOI: 10.1289/ehp.0901748] [Citation(s) in RCA: 216] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 04/19/2010] [Indexed: 05/17/2023]
Abstract
BACKGROUND Excess accumulation of manganese (Mn) in the brain results in a neurological syndrome with cognitive, psychiatric, and movement abnormalities. The highest concentrations of Mn in the brain are achieved in the basal ganglia, which may precipitate a form of parkinsonism with some clinical features that are similar and some that are different to those in Parkinson's disease (PD). Recently, scientists have debated the possibility that Mn may have an etiological role in PD or that it may accelerate the expression of PD. OBJECTIVE The goal of this review was to examine whether chronic Mn exposure produces dopamine neuron degeneration and PD or whether it has a distinct neuropathology and clinical presentation. DATA SOURCE I reviewed available clinical, neuroimaging, and neuropathological studies in humans and nonhuman primates exposed to Mn or other human conditions that result in elevated brain Mn concentrations. DATA EXTRACTION Human and nonhuman primate literature was examined to compare clinical, neuroimaging, and neuropathological changes associated with Mn-induced parkinsonism. DATA SYNTHESIS Clinical, neuroimaging, and neuropathological evidence was used to examine whether Mn-induced parkinsonism involves degeneration of the nigrostriatal dopaminergic system as is the case in PD. CONCLUSIONS The overwhelming evidence shows that Mn-induced parkinsonism does not involve degeneration of midbrain dopamine neurons and that l-dopa is not an effective therapy. New evidence is presented on a putative mechanism by which Mn may produce movement abnormalities. Confirmation of this hypothesis in humans is essential to make rational decisions about treatment, devise effective therapeutic strategies, and set regulatory guidelines.
Collapse
Affiliation(s)
- Tomás R Guilarte
- Neurotoxicology and Molecular Imaging Laboratory, Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
| |
Collapse
|
42
|
Kim JM, Kim JS, Jeong SH, Kim YK, Kim SE, Kim SH, Kim Y. Dopaminergic neuronal integrity in parkinsonism associated with liver cirrhosis. Neurotoxicology 2010; 31:351-5. [DOI: 10.1016/j.neuro.2010.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 04/11/2010] [Accepted: 05/02/2010] [Indexed: 11/28/2022]
|
43
|
Kim Y, Jeong KS, Yun YH, Oh MS. Occupational neurologic disorders in Korea. J Clin Neurol 2010; 6:64-72. [PMID: 20607045 PMCID: PMC2895226 DOI: 10.3988/jcn.2010.6.2.64] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 04/23/2010] [Accepted: 04/23/2010] [Indexed: 11/17/2022] Open
Abstract
This article presents a schematic review of the clinical manifestations of occupational neurologic disorders in Korea and discusses the toxicologic implications of these conditions. Vascular encephalopathy, parkinsonism, chronic toxic encephalopathy, cerebellar dysfunction, peripheral neuropathy, and neurodegenerative diseases are common presentations of occupational neurotoxic syndromes in Korea. Few neurotoxins cause patients to present with pathognomic neurologic syndrome. Detailed neurologic examinations and categorization of the clinical manifestations of neurologic disorders will improve the clinical management of occupational neurologic diseases. Physicians must be aware of the typical signs and symptoms of possible exposure to neurotoxins, and they should also pay attention to less-typical, rather-vague symptoms and signs in workers because the toxicologic characteristics of occupational neurologic diseases in Korea have changed from typical patterns to less-typical or equivocal patterns. This shift is likely to be due to several years of low-dose exposure, perhaps combined with the effects of aging, and new types of possibly toxicant-related neurodegenerative diseases. Close collaboration between neurologists and occupational physicians is needed to determine whether neurologic disorders are work-related.
Collapse
Affiliation(s)
- Yangho Kim
- Department of Occupational and Environmental Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | | | | | | |
Collapse
|
44
|
Curtis JT, Hood AN, Chen Y, Cobb GP, Wallace DR. Chronic metals ingestion by prairie voles produces sex-specific deficits in social behavior: an animal model of autism. Behav Brain Res 2010; 213:42-9. [PMID: 20433873 DOI: 10.1016/j.bbr.2010.04.028] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 04/13/2010] [Accepted: 04/19/2010] [Indexed: 10/19/2022]
Abstract
We examined the effects of chronic metals ingestion on social behavior in the normally highly social prairie vole to test the hypothesis that metals may interact with central dopamine systems to produce the social withdrawal characteristic of autism. Relative to water-treated controls, 10 weeks of chronic ingestion of either Hg(++) or Cd(++) via drinking water significantly reduced social contact by male voles when they were given a choice between isolation or contact with an unfamiliar same-sex conspecific. The effects of metals ingestion were specific to males: no effects of metals exposure were seen in females. Metals ingestion did not alter behavior of males allowed to choose between isolation or their familiar cage-mates, rather than strangers. We also examined the possibility that metals ingestion affects central dopamine functioning by testing the voles' locomotor responses to peripheral administration of amphetamine. As with the social behavior, we found a sex-specific effect of metals on amphetamine responses. Males that consumed Hg(++) did not increase their locomotor activity in response to amphetamine, whereas similarly treated females and males that ingested only water significantly increased their locomotor activities. Thus, an ecologically relevant stimulus, metals ingestion, produced two of the hallmark characteristics of autism - social avoidance and a male-oriented bias. These results suggest that metals exposure may contribute to the development of autism, possibly by interacting with central dopamine function, and support the use of prairie voles as a model organism in which to study autism.
Collapse
Affiliation(s)
- J Thomas Curtis
- Department of Pharmacology & Physiology, Oklahoma State University Center for Health Sciences, 1111 W 17th Street, Tulsa, OK 74107, USA.
| | | | | | | | | |
Collapse
|
45
|
Williams JM, Milatovic D, Gore JC, Aschner M, Avison MJ. Chronic exposure to manganese alters brain responses to amphetamine: a pharmacological magnetic resonance imaging study. Toxicol Sci 2010; 114:310-22. [PMID: 20061340 DOI: 10.1093/toxsci/kfq002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The parkinsonian symptoms and increased Mn accumulation in dopaminergic (DAergic) neurons of the basal ganglia implicate impaired dopamine signaling in the neurotoxic effects of chronic manganese overexposure. Using blood oxygenation level-dependent (BOLD) pharmacological magnetic resonance imaging (phMRI), we mapped brain responses to acute amphetamine (AMPH; 3 mg/kg, ip), which stimulates midbrain DAergic systems, in male Sprague-Dawley rats following 6 weeks of chronic MnCl(2) (5 mg Mn/kg, one per week, iv) or saline treatment. Plasma Mn content, measured immediately following phMRI, was elevated twofold in Mn-treated animals (p < 0.05), but the twofold increase in mean striatal Mn content did not reach significance. In saline-treated animals, AMPH stimulated robust positive BOLD responses throughout the basal ganglia and their reciprocally innervated connections. In contrast, acute AMPH stimulated a negative BOLD response in many of these structures in the Mn-treated group, resulting in significant differences between saline- and Mn-treated AMPH-evoked BOLD responses within caudate putamen, globus pallidus, substantia nigra, mediodorsal thalamic nucleus, and somatosensory cortex. These results demonstrate the utility of AMPH-evoked phMRI as readout of the DAergic signaling in vivo and confirm the vulnerability of DAergic systems to Mn.
Collapse
Affiliation(s)
- Jason M Williams
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | | | |
Collapse
|
46
|
From manganism to manganese-induced parkinsonism: a conceptual model based on the evolution of exposure. Neuromolecular Med 2009; 11:311-21. [PMID: 20012385 DOI: 10.1007/s12017-009-8108-8] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Accepted: 11/19/2009] [Indexed: 10/20/2022]
Abstract
Manganism is a distinct medical condition from Parkinson's disease. Manganese exposure scenarios in the last century generally have changed from the acute, high-level exposure conditions responsible for the occurrence of manganism to chronic exposure to much lower levels. Such chronic exposures may progressively extend the site of manganese deposition and toxicity from the globus pallidus to the entire area of the basal ganglia, including the substantia nigra pars compacta involved in Parkinson's disease. The mechanisms of manganese neurotoxicity from chronic exposure to very low levels are not well understood, but promising information is based on the concept of susceptibility that may place individuals exposed to manganese at a higher risk for developing Parkinsonian disturbances. These conditions include mutations of genes which play important pathogenetic roles in both Parkinsonism and in the regulation of manganese transport and metabolism. Liver function is also important in manganese-related neurotoxicity and sub-clinical impairment may increase the risk of Parkinsonism. The purpose and scope of this report are to explore the literature concerning manganese exposure and potential subclinical effects and biological pathways, impairment, and development of diseases such as Parkinsonism and manganism. Inhalation and ingestion of manganese will be the focus of this report.
Collapse
|
47
|
Are there common biochemical and molecular mechanisms controlling manganism and parkisonism. Neuromolecular Med 2009; 11:281-96. [PMID: 19757210 DOI: 10.1007/s12017-009-8088-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Accepted: 08/25/2009] [Indexed: 02/08/2023]
Abstract
Over the past several decades there has been considerable progress in our basic knowledge as to the mechanisms and factors regulating Mn toxicity. The disorder known as manganism is associated with the preferential accumulation of Mn in the globus pallidus of the basal ganglia which is generally considered to be the major and initial site of injury. Because the area of the CNS comprising the basal ganglia is very complex and dependent on the precise function and balance of several neurotransmitters, it is not surprising that symptoms of manganism often overlap with that of Parkinson's disease. The fact that neurological symptoms and onset of Mn toxicity are quite broad and can vary unpredictably probably reflects specific genetic variance of the physiological and biochemical makeup within the basal ganglia in any individual. Differences in response to Mn overexposure are, thus, likely due to underlying genetic variability which ultimately presents in deviations in both susceptibility as well as the characteristics of the neurological lesions and symptoms expressed. Although chronic exposure to Mn is not the initial causative agent provoking Parkinsonism, there is evidence suggesting that persistent exposure can predispose an individual to acquire dystonic movements associated with Parkinson's disease. As noted in this review, there appears to be common threads between the two disorders, as mutations in the genes, parkin and ATP13A2, associated with early onset of Parkinsonism, may also predispose an individual to develop Mn toxicity. Mutations in both genes appear to effect transport of Mn into the cell. These genetic difference coupled with additional environmental or nutritional factors must also be considered as contributing to the severity and onset of manganism.
Collapse
|
48
|
Benedetto A, Au C, Aschner M. Manganese-Induced Dopaminergic Neurodegeneration: Insights into Mechanisms and Genetics Shared with Parkinson’s Disease. Chem Rev 2009; 109:4862-84. [DOI: 10.1021/cr800536y] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Alexandre Benedetto
- Department of Pediatrics, Center for Molecular Neuroscience, Department of Pharmacology, and the Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, Tennessee 37232-0414
| | - Catherine Au
- Department of Pediatrics, Center for Molecular Neuroscience, Department of Pharmacology, and the Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, Tennessee 37232-0414
| | - Michael Aschner
- Department of Pediatrics, Center for Molecular Neuroscience, Department of Pharmacology, and the Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, Tennessee 37232-0414
| |
Collapse
|
49
|
Aschner M, Erikson KM, Herrero Hernández E, Hernández EH, Tjalkens R. Manganese and its role in Parkinson's disease: from transport to neuropathology. Neuromolecular Med 2009; 11:252-66. [PMID: 19657747 PMCID: PMC4613768 DOI: 10.1007/s12017-009-8083-0] [Citation(s) in RCA: 216] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Accepted: 07/24/2009] [Indexed: 01/03/2023]
Abstract
The purpose of this review is to highlight recent advances in the neuropathology associated with Mn exposures. We commence with a discussion on occupational manganism and clinical aspects of the disorder. This is followed by novel considerations on Mn transport (see also chapter by Yokel, this volume), advancing new hypotheses on the involvement of several transporters in Mn entry into the brain. This is followed by a brief description of the effects of Mn on neurotransmitter systems that are putative modulators of dopamine (DA) biology (the primary target of Mn neurotoxicity), as well as its effects on mitochondrial dysfunction and disruption of cellular energy metabolism. Next, we discuss inflammatory activation of glia in neuronal injury and how disruption of synaptic transmission and glial-neuronal communication may serve as underlying mechanisms of Mn-induced neurodegeneration commensurate with the cross-talk between glia and neurons. We conclude with a discussion on therapeutic aspects of Mn exposure. Emphasis is directed at treatment modalities and the utility of chelators in attenuating the neurodegenerative sequelae of exposure to Mn. For additional reading on several topics inherent to this review as well as others, the reader may wish to consult Aschner and Dorman (Toxicological Review 25:147-154, 2007) and Bowman et al. (Metals and neurodegeneration, 2009).
Collapse
Affiliation(s)
- Michael Aschner
- Departments of Pediatrics and Pharmacology and the Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, 2215-B Garland Avenue, 11425 MRB IV, Nashville, TN, 37232-0414, USA.
| | | | | | | | | |
Collapse
|
50
|
Guilarte TR, Burton NC, McGlothan JL, Verina T, Zhou Y, Alexander M, Pham L, Griswold M, Wong DF, Syversen T, Schneider JS. Impairment of nigrostriatal dopamine neurotransmission by manganese is mediated by pre-synaptic mechanism(s): implications to manganese-induced parkinsonism. J Neurochem 2008; 107:1236-47. [PMID: 18808452 DOI: 10.1111/j.1471-4159.2008.05695.x] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The long-term consequences of chronic manganese (Mn) exposure on neurological health is a topic of great concern to occupationally-exposed workers and in populations exposed to moderate levels of Mn. We have performed a comprehensive assessment of Mn effects on dopamine (DA) synapse markers using positron emission tomography (PET) in the non-human primate brain. Young male Cynomolgus macaques were given weekly i.v. injections of 3.3-5.0 mg Mn/kg (n = 4), 5.0-6.7 mg Mn/kg (n = 5), or 8.3-10.0 mg Mn/kg (n = 3) for 7-59 weeks and received PET studies of various DA synapse markers before (baseline) and at one or two time points during the course of Mn exposure. We report that amphetamine-induced DA release measured by PET is markedly impaired in the striatum of Mn-exposed animals. The effect of Mn on DA release was present in the absence of changes in markers of dopamine terminal integrity determined in post-mortem brain tissue from the same animals. These findings provide compelling evidence that the effects of Mn on DA synapses in the striatum are mediated by inhibition of DA neurotransmission and are responsible for the motor deficits documented in these animals.
Collapse
Affiliation(s)
- Tomás R Guilarte
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|