1
|
Friedman A, Schildroth S, Bauer JA, Coull BA, Smith DR, Placidi D, Cagna G, Krengel MH, Tripodis Y, White RF, Lucchini RG, Wright RO, Horton M, Austin C, Arora M, Claus Henn B. Early-life manganese exposure during multiple developmental periods and adolescent verbal learning and memory. Neurotoxicol Teratol 2023; 100:107307. [PMID: 37832858 PMCID: PMC10834060 DOI: 10.1016/j.ntt.2023.107307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/08/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND Manganese (Mn) is both an essential and toxic metal, and associations with neurodevelopment depend on exposure timing. Prospective data examining early life Mn with adolescent cognition are sparse. METHODS We enrolled 140 Italian adolescents (10-14 years old) from the Public Health Impact of Metals Exposure study. Mn in deciduous teeth was measured using laser ablation-mass spectrometry to represent prenatal, postnatal and early childhood exposure. The California Verbal Learning Test for Children (CVLT-C) was administered to assess adolescent verbal learning and memory. Multivariable regression models estimated changes in CVLT-C scores and the odds of making an error per doubling in dentine Mn in each exposure period. Multiple informant models tested for differences in associations across exposure periods. RESULTS A doubling in prenatal dentine Mn levels was associated with lower odds of making an intrusion error (OR = 0.23 [95% CI: 0.09, 0.61]). This beneficial association was not observed in other exposure periods. A doubling in childhood Mn was beneficially associated with short delay free recall: (ß = 0.47 [95% CI: -0.02, 0.97]), which was stronger in males (ß = 0.94 [95% CI: 0.05, 1.82]). Associations were null in the postnatal period. CONCLUSION Exposure timing is critical for understanding Mn-associated changes in cognitive function.
Collapse
Affiliation(s)
- Alexa Friedman
- Department of Environmental Health, Boston University School of Public Health, Boston, USA.
| | - Samantha Schildroth
- Department of Environmental Health, Boston University School of Public Health, Boston, USA
| | - Julia A Bauer
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Darmouth, USA
| | - Brent A Coull
- Department of Biostatistics, Harvard T. H Chan School of Public Health, Boston, USA
| | - Donald R Smith
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, Santa Cruz, USA
| | - Donatella Placidi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Giuseppa Cagna
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Maxine H Krengel
- Department of Neurology, Boston University School of Medicine, Boston, USA
| | - Yorghos Tripodis
- Department of Biostatistics, Boston University School of Public Health, Boston, USA
| | - Roberta F White
- Department of Environmental Health, Boston University School of Public Health, Boston, USA; Department of Neurology, Boston University School of Medicine, Boston, USA
| | - Roberto G Lucchini
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy; Department of Environmental Health Sciences, School of Public Health, Florida International University, Miami, USA
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Megan Horton
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Christine Austin
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Manish Arora
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Birgit Claus Henn
- Department of Environmental Health, Boston University School of Public Health, Boston, USA
| |
Collapse
|
2
|
Baj J, Flieger W, Barbachowska A, Kowalska B, Flieger M, Forma A, Teresiński G, Portincasa P, Buszewicz G, Radzikowska-Büchner E, Flieger J. Consequences of Disturbing Manganese Homeostasis. Int J Mol Sci 2023; 24:14959. [PMID: 37834407 PMCID: PMC10573482 DOI: 10.3390/ijms241914959] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
Manganese (Mn) is an essential trace element with unique functions in the body; it acts as a cofactor for many enzymes involved in energy metabolism, the endogenous antioxidant enzyme systems, neurotransmitter production, and the regulation of reproductive hormones. However, overexposure to Mn is toxic, particularly to the central nervous system (CNS) due to it causing the progressive destruction of nerve cells. Exposure to manganese is widespread and occurs by inhalation, ingestion, or dermal contact. Associations have been observed between Mn accumulation and neurodegenerative diseases such as manganism, Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. People with genetic diseases associated with a mutation in the gene associated with impaired Mn excretion, kidney disease, iron deficiency, or a vegetarian diet are at particular risk of excessive exposure to Mn. This review has collected data on the current knowledge of the source of Mn exposure, the experimental data supporting the dispersive accumulation of Mn in the brain, the controversies surrounding the reference values of biomarkers related to Mn status in different matrices, and the competitiveness of Mn with other metals, such as iron (Fe), magnesium (Mg), zinc (Zn), copper (Cu), lead (Pb), calcium (Ca). The disturbed homeostasis of Mn in the body has been connected with susceptibility to neurodegenerative diseases, fertility, and infectious diseases. The current evidence on the involvement of Mn in metabolic diseases, such as type 2 diabetes mellitus/insulin resistance, osteoporosis, obesity, atherosclerosis, and non-alcoholic fatty liver disease, was collected and discussed.
Collapse
Affiliation(s)
- Jacek Baj
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Wojciech Flieger
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Aleksandra Barbachowska
- Department of Plastic, Reconstructive and Burn Surgery, Medical University of Lublin, 21-010 Łęczna, Poland;
| | - Beata Kowalska
- Department of Water Supply and Wastewater Disposal, Lublin University of Technology, 20-618 Lublin, Poland;
| | - Michał Flieger
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | - Alicja Forma
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Grzegorz Teresiński
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | - Piero Portincasa
- Clinica Medica A. Murri, Department of Biomedical Sciences & Human Oncology, Medical School, University of Bari, 70124 Bari, Italy;
| | - Grzegorz Buszewicz
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | | | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
3
|
Hammond SL, Bantle CM, Popichak KA, Wright KA, Thompson D, Forero C, Kirkley KS, Damale PU, Chong EKP, Tjalkens RB. NF-κB Signaling in Astrocytes Modulates Brain Inflammation and Neuronal Injury Following Sequential Exposure to Manganese and MPTP During Development and Aging. Toxicol Sci 2021; 177:506-520. [PMID: 32692843 DOI: 10.1093/toxsci/kfaa115] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Chronic exposure to manganese (Mn) is associated with neuroinflammation and extrapyramidal motor deficits resembling features of Parkinson's disease. Activation of astrocytes and microglia is implicated in neuronal injury from Mn but it is not known whether early life exposure to Mn may predispose glia to more severe inflammatory responses during aging. We therefore examined astrocyte nuclear factor kappa B (NF-κB) signaling in mediating innate immune inflammatory responses during multiple neurotoxic exposures spanning juvenile development into adulthood. MnCl2 was given in drinking water for 30-day postweaning to both wildtype mice and astrocyte-specific knockout (KO) mice lacking I kappa B kinase 2, the central upstream activator of NF-κB. Following juvenile exposure to Mn, mice were subsequently administered 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) at 4 months of age. Animals were evaluated for behavioral alterations and brain tissue was analyzed for catecholamine neurotransmitters. Stereological analysis of neuronal and glial cell counts from multiple brain regions indicated that juvenile exposure to Mn amplified glial activation and neuronal loss from MPTP exposure in the caudate-putamen and globus pallidus, as well as increased the severity of neurobehavioral deficits in open field activity assays. These alterations were prevented in astrocyte-specific I kappa B kinase 2 KO mice. Juvenile exposure to Mn increased the number of neurotoxic A1 astrocytes expressing C3 as well as the number of activated microglia in adult mice following MPTP challenge, both of which were inhibited in KO mice. These results demonstrate that exposure to Mn during juvenile development heightens the innate immune inflammatory response in glia during a subsequent neurotoxic challenge through NF-κB signaling in astrocytes.
Collapse
Affiliation(s)
- Sean L Hammond
- Toxicology Program, Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences
| | - Collin M Bantle
- Toxicology Program, Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences
| | - Katriana A Popichak
- Toxicology Program, Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences
| | - Katie A Wright
- Toxicology Program, Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences
| | - Delaney Thompson
- Toxicology Program, Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences
| | - Catalina Forero
- Toxicology Program, Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences
| | - Kelly S Kirkley
- Toxicology Program, Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences
| | - Pranav U Damale
- Department of Electrical and Computer Engineering, College of Engineering, Colorado State University, Fort Collins, Colorado 80523-1680
| | - Edwin K P Chong
- Department of Electrical and Computer Engineering, College of Engineering, Colorado State University, Fort Collins, Colorado 80523-1680
| | - Ronald B Tjalkens
- Toxicology Program, Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences
| |
Collapse
|
4
|
Bantle CM, French CT, Cummings JE, Sadasivan S, Tran K, Slayden RA, Smeyne RJ, Tjalkens RB. Manganese exposure in juvenile C57BL/6 mice increases glial inflammatory responses in the substantia nigra following infection with H1N1 influenza virus. PLoS One 2021; 16:e0245171. [PMID: 33493177 PMCID: PMC7833173 DOI: 10.1371/journal.pone.0245171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/22/2020] [Indexed: 01/22/2023] Open
Abstract
Infection with Influenza A virus can lead to the development of encephalitis and subsequent neurological deficits ranging from headaches to neurodegeneration. Post-encephalitic parkinsonism has been reported in surviving patients of H1N1 infections, but not all cases of encephalitic H1N1 infection present with these neurological symptoms, suggesting that interactions with an environmental neurotoxin could promote more severe neurological damage. The heavy metal, manganese (Mn), is a potential interacting factor with H1N1 because excessive exposure early in life can induce long-lasting effects on neurological function through inflammatory activation of glial cells. In the current study, we used a two-hit model of neurotoxin-pathogen exposure to examine whether exposure to Mn during juvenile development would induce a more severe neuropathological response following infection with H1N1 in adulthood. To test this hypothesis, C57BL/6 mice were exposed to MnCl2 in drinking water (50 mg/kg/day) for 30 days from days 21–51 postnatal, then infected intranasally with H1N1 three weeks later. Analyses of dopaminergic neurons, microglia and astrocytes in basal ganglia indicated that although there was no significant loss of dopaminergic neurons within the substantia nigra pars compacta, there was more pronounced activation of microglia and astrocytes in animals sequentially exposed to Mn and H1N1, as well as altered patterns of histone acetylation. Whole transcriptome Next Generation Sequencing (RNASeq) analysis was performed on the substantia nigra and revealed unique patterns of gene expression in the dual-exposed group, including genes involved in antioxidant activation, mitophagy and neurodegeneration. Taken together, these results suggest that exposure to elevated levels of Mn during juvenile development could sensitize glial cells to more severe neuro-immune responses to influenza infection later in life through persistent epigenetic changes.
Collapse
Affiliation(s)
- Collin M. Bantle
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - C. Tenley French
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jason E. Cummings
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Shankar Sadasivan
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Kevin Tran
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Richard A. Slayden
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Richard J. Smeyne
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Ronald B. Tjalkens
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- * E-mail:
| |
Collapse
|
5
|
Manganese Accumulation in the Brain via Various Transporters and Its Neurotoxicity Mechanisms. Molecules 2020; 25:molecules25245880. [PMID: 33322668 PMCID: PMC7763224 DOI: 10.3390/molecules25245880] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
Manganese (Mn) is an essential trace element, serving as a cofactor for several key enzymes, such as glutamine synthetase, arginase, pyruvate decarboxylase, and mitochondrial superoxide dismutase. However, its chronic overexposure can result in a neurological disorder referred to as manganism, presenting symptoms similar to those inherent to Parkinson’s disease. The pathological symptoms of Mn-induced toxicity are well-known, but the underlying mechanisms of Mn transport to the brain and cellular toxicity leading to Mn’s neurotoxicity are not completely understood. Mn’s levels in the brain are regulated by multiple transporters responsible for its uptake and efflux, and thus, dysregulation of these transporters may result in Mn accumulation in the brain, causing neurotoxicity. Its distribution and subcellular localization in the brain and associated subcellular toxicity mechanisms have also been extensively studied. This review highlights the presently known Mn transporters and their roles in Mn-induced neurotoxicity, as well as subsequent molecular and cellular dysregulation upon its intracellular uptakes, such as oxidative stress, neuroinflammation, disruption of neurotransmission, α-synuclein aggregation, and amyloidogenesis.
Collapse
|
6
|
Ivleva I, Pestereva N, Zubov A, Karpenko M. Intranasal exposure of manganese induces neuroinflammation and disrupts dopamine metabolism in the striatum and hippocampus. Neurosci Lett 2020; 738:135344. [PMID: 32889006 DOI: 10.1016/j.neulet.2020.135344] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/18/2020] [Accepted: 08/27/2020] [Indexed: 01/26/2023]
Abstract
Prolonged exposure to manganese (Mn) may lead to toxic effects on the central nervous system (CNS). The mechanisms underlying neuronal death from exposure to Mn are not well understood but undoubtedly involve inflammatory processes. The aim of this study was to explore the effects of long-lasting intranasal Mn exposure in rats focusing on inflammatory processes and catecholamine (dopamine, norepinephrine) levels in the striatum and hippocampus. It was found that intranasal administration by instillation of MnCl2 solution once a day for 90 days leads to impaired movement and gait. We also observed that Mn concentration increased in the hippocampus (by 30 %) and in the striatum (by 220 %), dopamine (24 %) and DOPAC (35 %) were reduced in the striatum, and dopamine (190 %) and DOPAC (220 %) levels increased with simultaneously norepinephrine reduction (30 %) in the hippocampus. Observation of cytokine mRNA revealed increased expression of both assayed cytokines (IL-1β and TNF-α) in the hippocampus. There was a 3-fold increase in the expression of IBA-1 mRNA, 2-fold increase in NFκB mRNA, and dramatic reduction in IkB mRNA in the striatum. Taken together, intranasal exposure to a high dose of MnCl2 induces neuroinflammation and neurotransmission disturbance, but the effects are specific for each studied brain region.
Collapse
Affiliation(s)
- Irina Ivleva
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia.
| | - Nina Pestereva
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia
| | - Alexander Zubov
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia
| | - Marina Karpenko
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia; Institute of Physics, Nanotechnology and Telecommunications, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| |
Collapse
|
7
|
Conley TE, Beaudin SA, Lasley SM, Fornal CA, Hartman J, Uribe W, Khan T, Strupp BJ, Smith DR. Early postnatal manganese exposure causes arousal dysregulation and lasting hypofunctioning of the prefrontal cortex catecholaminergic systems. J Neurochem 2020; 153:631-649. [PMID: 31811785 PMCID: PMC7261255 DOI: 10.1111/jnc.14934] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022]
Abstract
Studies have reported associations between environmental manganese (Mn) exposure and impaired cognition, attention, impulse control, and fine motor function in children. Our recent rodent studies established that elevated Mn exposure causes these impairments. Here, rats were exposed orally to 0, 25, or 50 mg Mn kg-1 day-1 during early postnatal life (PND 1-21) or lifelong to determine whether early life Mn exposure causes heightened behavioral reactivity in the open field, lasting changes in the catecholaminergic systems in the medial prefrontal cortex (mPFC), altered dendritic spine density, and whether lifelong exposure exacerbates these effects. We also assessed astrocyte reactivity (glial fibrillary acidic protein, GFAP), and astrocyte complement C3 and S100A10 protein levels as markers of A1 proinflammatory or A2 anti-inflammatory reactive astrocytes. Postnatal Mn exposure caused heightened behavioral reactivity during the first 5-10 min intervals of daily open field test sessions, consistent with impairments in arousal regulation. Mn exposure reduced the evoked release of norepinephrine (NE) and caused decreased protein levels of tyrosine hydroxylase (TH), dopamine (DA) and NE transporters, and DA D1 receptors, along with increased DA D2 receptors. Mn also caused a lasting increase in reactive astrocytes (GFAP) exhibiting increased A1 and A2 phenotypes, with a greater induction of the A1 proinflammatory phenotype. These results demonstrate that early life Mn exposure causes broad lasting hypofunctioning of the mPFC catecholaminergic systems, consistent with the impaired arousal regulation, attention, impulse control, and fine motor function reported in these animals, suggesting that mPFC catecholaminergic dysfunction may underlie similar impairments reported in Mn-exposed children.
Collapse
Affiliation(s)
- Travis E. Conley
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA
| | - Stephane A. Beaudin
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA
| | - Stephen M. Lasley
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, 61605, USA
| | - Casimir A. Fornal
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, 61605, USA
| | - Jasenia Hartman
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA
| | - Walter Uribe
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA
| | - Tooba Khan
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA
| | - Barbara J. Strupp
- Division of Nutritional Sciences and Department of Psychology, Cornell University, Ithaca, NY, 14853, USA
| | - Donald R. Smith
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA
| |
Collapse
|
8
|
Microglial Function in the Effects of Early-Life Stress on Brain and Behavioral Development. J Clin Med 2020; 9:jcm9020468. [PMID: 32046333 PMCID: PMC7074320 DOI: 10.3390/jcm9020468] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
The putative effects of early-life stress (ELS) on later behavior and neurobiology have been widely investigated. Recently, microglia have been implicated in mediating some of the effects of ELS on behavior. In this review, findings from preclinical and clinical literature with a specific focus on microglial alterations induced by the exposure to ELS (i.e., exposure to behavioral stressors or environmental agents and infection) are summarized. These studies were utilized to interpret changes in developmental trajectories based on the time at which the stress occurred, as well as the paradigm used. ELS and microglial alterations were found to be associated with a wide array of deficits including cognitive performance, memory, reward processing, and processing of social stimuli. Four general conclusions emerged: (1) ELS interferes with microglial developmental programs, including their proliferation and death and their phagocytic activity; (2) this can affect neuronal and non-neuronal developmental processes, which are dynamic during development and for which microglial activity is instrumental; (3) the effects are extremely dependent on the time point at which the investigation is carried out; and (4) both pre- and postnatal ELS can prime microglial reactivity, indicating a long-lasting alteration, which has been implicated in behavioral abnormalities later in life.
Collapse
|
9
|
Verbal Memory and Learning in Schoolchildren Exposed to Manganese in Mexico. Neurotox Res 2019; 36:827-835. [PMID: 31148117 DOI: 10.1007/s12640-019-00037-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 12/20/2022]
Abstract
Manganese (Mn) is an essential nutrient for cellular function, but in high concentrations, it is neurotoxic. Environmental exposure to Mn has been associated with cognitive effects in children. This study aimed to assess the effect of environmental exposure to Mn on verbal memory and learning in schoolchildren residents from two municipalities in the state of Hidalgo, Mexico. Cross-sectional studies were conducted in 2006 and 2013 with a total of 265 schoolchildren of 7 to 11 years old. Children's Auditory Verbal Learning Test-2 (CAVLT-2) was used to assess verbal memory and learning. Mn exposure tertiles were defined according to hair manganese (MnH) levels determined by atomic absorption spectrophotometry. Linear regression models were used to estimate the association between MnH levels and CAVLT-2 scores. The models were adjusted by potential confounders. The lowest and highest exposure tertiles were defined below and above MnH levels of ≤ 0.72 and ≥ 3.96 μg/g, respectively. Mn exposure was significantly associated with an average of 5- to 9-point decrease in learning curves and summary CAVLT-2 scores in the highest tertile. This study adds to the evidence of decreased verbal memory and learning in schoolchildren environmentally exposed to manganese.
Collapse
|
10
|
Sarkar S, Rokad D, Malovic E, Luo J, Harischandra DS, Jin H, Anantharam V, Huang X, Lewis M, Kanthasamy A, Kanthasamy AG. Manganese activates NLRP3 inflammasome signaling and propagates exosomal release of ASC in microglial cells. Sci Signal 2019; 12:12/563/eaat9900. [PMID: 30622196 DOI: 10.1126/scisignal.aat9900] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Chronic, sustained inflammation underlies many pathological conditions, including neurodegenerative diseases. Divalent manganese (Mn2+) exposure can stimulate neurotoxicity by increasing inflammation. In this study, we examined whether Mn2+ activates the multiprotein NLRP3 inflammasome complex to promote neuroinflammation. Exposing activated mouse microglial cells to Mn2+ substantially augmented NLRP3 abundance, caspase-1 cleavage, and maturation of the inflammatory cytokine interleukin-1β (IL-1β). Exposure of mice to Mn2+ had similar effects in brain microglial cells. Furthermore, Mn2+ impaired mitochondrial ATP generation, basal respiratory rate, and spare capacity in microglial cells. These data suggest that Mn-induced mitochondrial defects drove the inflammasome signal amplification. We found that Mn induced cell-to-cell transfer of the inflammasome adaptor protein ASC in exosomes. Furthermore, primed microglial cells exposed to exosomes from Mn-treated mice released more IL-1β than did cells exposed to exosomes from control-treated animals. We also observed that welders exposed to manganese-containing fumes had plasma exosomes that contained more ASC than did those from a matched control group. Together, these results suggest that the divalent metal manganese acts as a key amplifier of NLRP3 inflammasome signaling and exosomal ASC release.
Collapse
Affiliation(s)
- Souvarish Sarkar
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Dharmin Rokad
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Emir Malovic
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Jie Luo
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Dilshan S Harischandra
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Huajun Jin
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Xuemei Huang
- Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Mechelle Lewis
- Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Anumantha G Kanthasamy
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA.
| |
Collapse
|
11
|
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
|
12
|
Adhikari A, Das M, Mondal S, Darbar S, Das AK, Bhattacharya SS, Pal D, Pal SK. Manganese neurotoxicity: nano-oxide compensates for ion-damage in mammals. Biomater Sci 2019; 7:4491-4502. [DOI: 10.1039/c9bm01039d] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Here, we have shown that citrate functionalized Mn3O4nanoparticles can ameliorate Mn-induced neurotoxicity (Parkinson's-like syndrome) through the chelation of excess Mn ions and subsequent reduction of oxidative damage.
Collapse
Affiliation(s)
- Aniruddha Adhikari
- Department of Chemical
- Biological and Macromolecular Sciences
- SN Bose National Centre for Basic Sciences
- Kolkata-700106
- India
| | - Monojit Das
- Department of Zoology
- Uluberia College
- University of Calcutta
- Uluberia
- India
| | - Susmita Mondal
- Department of Chemical
- Biological and Macromolecular Sciences
- SN Bose National Centre for Basic Sciences
- Kolkata-700106
- India
| | - Soumendra Darbar
- Research & Development Division
- Dey's Medical Stores (Mfg.) Ltd
- Kolkata-700019
- India
| | - Anjan Kumar Das
- Department of Pathology
- Calcutta National Medical College and Hospital
- Kolkata-700014
- India
| | | | - Debasish Pal
- Department of Zoology
- Uluberia College
- University of Calcutta
- Uluberia
- India
| | - Samir Kumar Pal
- Department of Chemical
- Biological and Macromolecular Sciences
- SN Bose National Centre for Basic Sciences
- Kolkata-700106
- India
| |
Collapse
|
13
|
Popichak KA, Afzali MF, Kirkley KS, Tjalkens RB. Glial-neuronal signaling mechanisms underlying the neuroinflammatory effects of manganese. J Neuroinflammation 2018; 15:324. [PMID: 30463564 PMCID: PMC6247759 DOI: 10.1186/s12974-018-1349-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/29/2018] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Exposure to increased manganese (Mn) causes inflammation and neuronal injury in the cortex and basal ganglia, resulting in neurological symptoms resembling Parkinson's disease. The mechanisms underlying neuronal death from exposure to Mn are not well understood but involve inflammatory activation of microglia and astrocytes. Expression of neurotoxic inflammatory genes in glia is highly regulated through the NF-κB pathway, but factors modulating neurotoxic glial-glial and glial-neuronal signaling by Mn are not well understood. METHODS We examined the role of NF-κB in Mn-induced neurotoxicity by exposing purified microglia, astrocytes (from wild-type and astrocyte-specific IKK knockout mice), and mixed glial cultures to varying Mn concentrations and then treating neurons with the conditioned media (GCM) of each cell type. We hypothesized that mixed glial cultures exposed to Mn (0-100 μM) would enhance glial activation and neuronal death compared to microglia, wild-type astrocytes, or IKK-knockout astrocytes alone or in mixed cultures. RESULTS Mixed glial cultures treated with 0-100 μM Mn for 24 h showed the most pronounced effect of increased expression of inflammatory genes including inducible nitric oxide synthase (Nos2), Tnf, Ccl5, Il6, Ccr2, Il1b, and the astrocyte-specific genes, C3 and Ccl2. Gene deletion of IKK2 in astrocytes dramatically reduced cytokine release in Mn-treated mixed glial cultures. Measurement of neuronal viability and apoptosis following exposure to Mn-GCM demonstrated that mixed glial cultures induced greater neuronal death than either cell type alone. Loss of IKK in astrocytes also decreased neuronal death compared to microglia alone, wild-type astrocytes, or mixed glia. CONCLUSIONS This suggests that astrocytes are a critical mediator of Mn neurotoxicity through enhanced expression of inflammatory cytokines and chemokines, including those most associated with a reactive phenotype such as CCL2 but not C3.
Collapse
Affiliation(s)
- Katriana A. Popichak
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 1680 Campus Delivery, Physiology Building, Room 101, Fort Collins, CO 80523-1680 USA
| | - Maryam F. Afzali
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO USA
| | - Kelly S. Kirkley
- Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO USA
| | - Ronald B. Tjalkens
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 1680 Campus Delivery, Physiology Building, Room 101, Fort Collins, CO 80523-1680 USA
| |
Collapse
|
14
|
Langley MR, Ghaisas S, Ay M, Luo J, Palanisamy BN, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Manganese exposure exacerbates progressive motor deficits and neurodegeneration in the MitoPark mouse model of Parkinson's disease: Relevance to gene and environment interactions in metal neurotoxicity. Neurotoxicology 2018; 64:240-255. [PMID: 28595911 PMCID: PMC5736468 DOI: 10.1016/j.neuro.2017.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/02/2017] [Accepted: 06/02/2017] [Indexed: 10/19/2022]
Abstract
Parkinson's disease (PD) is now recognized as a neurodegenerative condition caused by a complex interplay of genetic and environmental influences. Chronic manganese (Mn) exposure has been implicated in the development of PD. Since mitochondrial dysfunction is associated with PD pathology as well as Mn neurotoxicity, we investigated whether Mn exposure augments mitochondrial dysfunction and neurodegeneration in the nigrostriatal dopaminergic system using a newly available mitochondrially defective transgenic mouse model of PD, the MitoPark mouse. This unique PD model recapitulates key features of the disease including progressive neurobehavioral changes and neuronal degeneration. We exposed MitoPark mice to a low dose of Mn (10mg/kg, p.o.) daily for 4 weeks starting at age 8 wks and then determined the behavioral, neurochemical and histological changes. Mn exposure accelerated the rate of progression of motor deficits in MitoPark mice when compared to the untreated MitoPark group. Mn also worsened olfactory function in this model. Most importantly, Mn exposure intensified the depletion of striatal dopamine and nigral TH neuronal loss in MitoPark mice. The neurodegenerative changes were accompanied by enhanced oxidative damage in the striatum and substantia nigra (SN) of MitoPark mice treated with Mn. Furthermore, Mn-treated MitoPark mice had significantly more oligomeric protein and IBA-1-immunoreactive microglia cells, suggesting Mn augments neuroinflammatory processes in the nigrostriatal pathway. To further confirm the direct effect of Mn on impaired mitochondrial function, we also generated a mitochondrially defective dopaminergic cell model by knocking out the TFAM transcription factor by using a CRISPR-Cas9 gene-editing method. Seahorse mitochondrial bioenergetic analysis revealed that Mn decreases mitochondrial basal and ATP-linked respiration in the TFAM KO cells. Collectively, our results reveal that Mn can augment mitochondrial dysfunction to exacerbate nigrostriatal neurodegeneration and PD-related behavioral symptoms. Our study also demonstrates that the MitoPark mouse is an excellent model to study the gene-environment interactions associated with mitochondrial defects in the nigral dopaminergic system as well as to evaluate the contribution of potential environmental toxicant interactions in a slowly progressive model of Parkinsonism.
Collapse
Affiliation(s)
- Monica R Langley
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Shivani Ghaisas
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Muhammet Ay
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Jie Luo
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Bharathi N Palanisamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Huajun Jin
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Vellareddy Anantharam
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Arthi Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Anumantha G Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
15
|
Dees WL, Hiney JK, Srivastava VK. Influences of manganese on pubertal development. J Endocrinol 2017; 235:R33-R42. [PMID: 28720645 PMCID: PMC5675006 DOI: 10.1530/joe-17-0237] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 07/18/2017] [Indexed: 11/08/2022]
Abstract
The onset of puberty is the result of complex neuroendocrine interactions within hypothalamic region of the brain, as well as from genetic and environmental influences. These interactions ultimately result in the increased synthesis and release of luteinizing hormone-releasing hormone (LHRH). Manganese (Mn) is an essential environmental element known for years to be involved in numerous mammalian physiological processes, including growth and reproductive function. Studies in recent years have shown the ability of Mn to cross the blood-brain barrier and act within the hypothalamus to influence the timing of puberty. This review will depict research showing the molecular and physiological actions of Mn in the control of prepubertal LHRH and discuss the potential for the element to cause either helpful or harmful outcomes on the developmental process depending upon the age and accumulation of Mn within the hypothalamus.
Collapse
Affiliation(s)
- William L Dees
- Veterinary Integrative BiosciencesCollege of Veterinary Medicine, Texas A&M University, College Station, Texas, USA
| | - Jill K Hiney
- Veterinary Integrative BiosciencesCollege of Veterinary Medicine, Texas A&M University, College Station, Texas, USA
| | - Vinod K Srivastava
- Veterinary Integrative BiosciencesCollege of Veterinary Medicine, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
16
|
Chi L, Gao B, Bian X, Tu P, Ru H, Lu K. Manganese-induced sex-specific gut microbiome perturbations in C57BL/6 mice. Toxicol Appl Pharmacol 2017; 331:142-153. [PMID: 28610994 PMCID: PMC5653225 DOI: 10.1016/j.taap.2017.06.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 06/07/2017] [Accepted: 06/10/2017] [Indexed: 12/26/2022]
Abstract
Overexposure to manganese (Mn) leads to toxic effects, such as promoting the development of Parkinson's-like neurological disorders. The gut microbiome is deeply involved in immune development, host metabolism, and xenobiotics biotransformation, and significantly influences central nervous system (CNS) via the gut-brain axis, i.e. the biochemical signaling between the gastrointestinal tract and the CNS. However, it remains unclear whether Mn can affect the gut microbiome and its metabolic functions, particularly those linked to neurotoxicity. In addition, sex-specific effects of Mn have been reported, with no mechanism being identified yet. Recently, we have shown that the gut microbiome is largely different between males and females, raising the possibility that differential gut microbiome responses may contribute to sex-selective toxicity of Mn. Here, we applied high-throughput sequencing and gas chromatography-mass spectrometry (GC-MS) metabolomics to explore how Mn2+ exposure affects the gut microbiome and its metabolism in C57BL/6 mice. Mn2+ exposure perturbed the gut bacterial compositions, functional genes and fecal metabolomes in a highly sex-specific manner. In particular, bacterial genes and/or key metabolites of neurotransmitter synthesis and pro-inflammatory mediators are significantly altered by Mn2+ exposure, which can potentially affect chemical signaling of gut-brain interactions. Likewise, functional genes involved in iron homeostasis, flagellar motility, quorum sensing, and Mn transportation/oxidation are also widely changed by Mn2+ exposure. Taken together, this study has demonstrated that Mn2+ exposure perturbs the gut microbiome and its metabolic functions, which highlights the potential role of the gut microbiome in Mn2+ toxicity, particularly its sex-specific toxic effects.
Collapse
Affiliation(s)
- Liang Chi
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, 27599, United States
| | - Bei Gao
- Department of Environmental Health Science, University of Georgia, Athens, GA 30602, United States
| | - Xiaoming Bian
- Department of Environmental Health Science, University of Georgia, Athens, GA 30602, United States
| | - Pengcheng Tu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, 27599, United States
| | - Hongyu Ru
- Department of Population Health and Pathobiology, North Carolina State University, Raleigh, NC 27607, United States
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, 27599, United States.
| |
Collapse
|
17
|
Fleming SM, Santiago NA, Mullin EJ, Pamphile S, Karkare S, Lemkuhl A, Ekhator OR, Linn SC, Holden JG, Aga DS, Roth JA, Liou B, Sun Y, Shull GE, Schultheis PJ. The effect of manganese exposure in Atp13a2-deficient mice. Neurotoxicology 2017; 64:256-266. [PMID: 28595912 PMCID: PMC10178982 DOI: 10.1016/j.neuro.2017.06.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 05/25/2017] [Accepted: 06/03/2017] [Indexed: 12/30/2022]
Abstract
Loss of function mutations in the P5-ATPase ATP13A2 are associated with Kufor-Rakeb Syndrome and Neuronal Ceroid Lipofuscinosis. While the function of ATP13A2 is unclear, in vitro studies suggest it is a lysosomal protein that interacts with the metals manganese (Mn) and zinc and the presynaptic protein alpha-synuclein. Loss of ATP13A2 function in mice causes sensorimotor deficits, enhanced autofluorescent storage material, and accumulation of alpha-synuclein. The present study sought to determine the effect of Mn administration on these same outcomes in ATP13A2-deficient mice. Wildtype and ATP13A2-deficient mice received saline or Mn at 5-9 or 12-19 months for 45days. Sensorimotor function was assessed starting at day 30. Autofluorescence was quantified in multiple brain regions and alpha-synuclein protein levels were determined in the ventral midbrain. Brain Mn, iron, zinc, and copper concentrations were measured in 5-9 month old mice. The results show Mn enhanced sensorimotor function, increased autofluorescence in the substantia nigra, and increased insoluble alpha-synuclein in the ventral midbrain in older ATP13A2-deficient mice. In addition, the Mn regimen used increased Mn concentration in the brain and levels were higher in Mn-treated mutants than controls. These results indicate loss of ATP13A2 function leads to increased sensitivity to Mn in vivo.
Collapse
Affiliation(s)
- Sheila M Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States.
| | - Nicholas A Santiago
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | | | - Shanta Pamphile
- Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Swagata Karkare
- Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Andrew Lemkuhl
- Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Osunde R Ekhator
- Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Stephen C Linn
- Department of Biological Sciences, Northern Kentucky University, Highland Heights, KY, United States
| | - John G Holden
- Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Diana S Aga
- Department of Chemistry, SUNY Buffalo, Buffalo, NY, United States
| | - Jerome A Roth
- Department of Pharmacology and Toxicology, SUNY Buffalo, Buffalo, NY, United States
| | - Benjamin Liou
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Ying Sun
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Gary E Shull
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Cincinnati, OH, United States
| | - Patrick J Schultheis
- Department of Biological Sciences, Northern Kentucky University, Highland Heights, KY, United States
| |
Collapse
|
18
|
Sarkar S, Malovic E, Harischandra DS, Ngwa HA, Ghosh A, Hogan C, Rokad D, Zenitsky G, Jin H, Anantharam V, Kanthasamy AG, Kanthasamy A. Manganese exposure induces neuroinflammation by impairing mitochondrial dynamics in astrocytes. Neurotoxicology 2017; 64:204-218. [PMID: 28539244 DOI: 10.1016/j.neuro.2017.05.009] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 12/21/2022]
Abstract
Chronic manganese (Mn) exposure induces neurotoxicity, which is characterized by Parkinsonian symptoms resulting from impairment in the extrapyramidal motor system of the basal ganglia. Mitochondrial dysfunction and oxidative stress are considered key pathophysiological features of Mn neurotoxicity. Recent evidence suggests astrocytes as a major target of Mn neurotoxicity since Mn accumulates predominantly in astrocytes. However, the primary mechanisms underlying Mn-induced astroglial dysfunction and its role in metal neurotoxicity are not completely understood. In this study, we examined the interrelationship between mitochondrial dysfunction and astrocytic inflammation in Mn neurotoxicity. We first evaluated whether Mn exposure alters mitochondrial bioenergetics in cultured astrocytes. Metabolic activity assessed by MTS assay revealed an IC50 of 92.68μM Mn at 24h in primary mouse astrocytes (PMAs) and 50.46μM in the human astrocytic U373 cell line. Mn treatment reduced mitochondrial mass, indicative of impaired mitochondrial function and biogenesis, which was substantiated by the significant reduction in mRNA of mitofusin-2, a protein that serves as a ubiquitination target for mitophagy. Furthermore, Mn increased mitochondrial circularity indicating augmented mitochondrial fission. Seahorse analysis of bioenergetics status in Mn-treated astrocytes revealed that Mn significantly impaired the basal mitochondrial oxygen consumption rate as well as the ATP-linked respiration rate. The effect of Mn on mitochondrial energy deficits was further supported by a reduction in ATP production. Mn-exposed primary astrocytes also exhibited a severely quiescent energy phenotype, which was substantiated by the inability of oligomycin to increase the extracellular acidification rate. Since astrocytes regulate immune functions in the CNS, we also evaluated whether Mn modulates astrocytic inflammation. Mn exposure in astrocytes not only stimulated the release of proinflammatory cytokines, but also exacerbated the inflammatory response induced by aggregated α-synuclein. The novel mitochondria-targeted antioxidant, mito-apocynin, significantly attenuated Mn-induced inflammatory gene expression, further supporting the role of mitochondria dysfunction and oxidative stress in mediating astrogliosis. Lastly, intranasal delivery of Mn in vivo elevated GFAP and depressed TH levels in the olfactory bulbs, clearly supporting the involvement of astrocytes in Mn-induced dopaminergic neurotoxicity. Collectively, our study demonstrates that Mn drives proinflammatory events in astrocytes by impairing mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Souvarish Sarkar
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA 50011, United States
| | - Emir Malovic
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA 50011, United States
| | - Dilshan S Harischandra
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA 50011, United States
| | - Hilary A Ngwa
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA 50011, United States
| | - Anamitra Ghosh
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA 50011, United States
| | - Colleen Hogan
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA 50011, United States
| | - Dharmin Rokad
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA 50011, United States
| | - Gary Zenitsky
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA 50011, United States
| | - Huajun Jin
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA 50011, United States
| | - Vellareddy Anantharam
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA 50011, United States
| | - Anumantha G Kanthasamy
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA 50011, United States
| | - Arthi Kanthasamy
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
19
|
Microglia amplify inflammatory activation of astrocytes in manganese neurotoxicity. J Neuroinflammation 2017; 14:99. [PMID: 28476157 PMCID: PMC5418760 DOI: 10.1186/s12974-017-0871-0] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 04/22/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND As the primary immune response cell in the central nervous system, microglia constantly monitor the microenvironment and respond rapidly to stress, infection, and injury, making them important modulators of neuroinflammatory responses. In diseases such as Parkinson's disease, Alzheimer's disease, multiple sclerosis, and human immunodeficiency virus-induced dementia, activation of microglia precedes astrogliosis and overt neuronal loss. Although microgliosis is implicated in manganese (Mn) neurotoxicity, the role of microglia and glial crosstalk in Mn-induced neurodegeneration is poorly understood. METHODS Experiments utilized immunopurified murine microglia and astrocytes using column-free magnetic separation. The effect of Mn on microglia was investigated using gene expression analysis, Mn uptake measurements, protein production, and changes in morphology. Additionally, gene expression analysis was used to determine the effect Mn-treated microglia had on inflammatory responses in Mn-exposed astrocytes. RESULTS Immunofluorescence and flow cytometric analysis of immunopurified microglia and astrocytes indicated cultures were 97 and 90% pure, respectively. Mn treatment in microglia resulted in a dose-dependent increase in pro-inflammatory gene expression, transition to a mixed M1/M2 phenotype, and a de-ramified morphology. Conditioned media from Mn-exposed microglia (MCM) dramatically enhanced expression of mRNA for Tnf, Il-1β, Il-6, Ccl2, and Ccl5 in astrocytes, as did exposure to Mn in the presence of co-cultured microglia. MCM had increased levels of cytokines and chemokines including IL-6, TNF, CCL2, and CCL5. Pharmacological inhibition of NF-κB in microglia using Bay 11-7082 completely blocked microglial-induced astrocyte activation, whereas siRNA knockdown of Tnf in primary microglia only partially inhibited neuroinflammatory responses in astrocytes. CONCLUSIONS These results provide evidence that NF-κB signaling in microglia plays an essential role in inflammatory responses in Mn toxicity by regulating cytokines and chemokines that amplify the activation of astrocytes.
Collapse
|
20
|
Inflammatory Activation of Microglia and Astrocytes in Manganese Neurotoxicity. ADVANCES IN NEUROBIOLOGY 2017; 18:159-181. [PMID: 28889267 DOI: 10.1007/978-3-319-60189-2_8] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurotoxicity due to excessive exposure to manganese (Mn) has been described as early as 1837 (Couper, Br Ann Med Pharm Vital Stat Gen Sci 1:41-42, 1837). Extensive research over the past two decades has revealed that Mn-induced neurological injury involves complex pathophysiological signaling mechanisms between neurons and glial cells. Glial cells are an important target of Mn in the brain, both for sequestration of the metal, as well as for activating inflammatory signaling pathways that damage neurons through overproduction of numerous reactive oxygen and nitrogen species and inflammatory cytokines. Understanding how these pathways are regulated in glial cells during Mn exposure is critical to determining the mechanisms underlying permanent neurological dysfunction stemming from excess exposure. The subject of this review will be to delineate mechanisms by which Mn interacts with glial cells to perturb neuronal function, with a particular emphasis on neuroinflammation and neuroinflammatory signaling between distinct populations of glial cells.
Collapse
|
21
|
Srivastava VK, Hiney JK, Dees WL. Manganese-Stimulated Kisspeptin Is Mediated by the IGF-1/Akt/Mammalian Target of Rapamycin Pathway in the Prepubertal Female Rat. Endocrinology 2016; 157:3233-41. [PMID: 27309941 PMCID: PMC4967113 DOI: 10.1210/en.2016-1090] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 06/10/2016] [Indexed: 11/19/2022]
Abstract
Low-dose administration of manganese chloride (MnCl2) causes release of hypothalamic LH-releasing hormone (LHRH) and advances puberty in rat. Recently, this element was shown to up-regulate mammalian target of rapamycin (mTOR), kisspeptin gene (KiSS-1), and LHRH gene expressions in the brain preoptic area (POA)/anteroventral periventricular (AVPV) nucleus. Because these genes are critical for puberty, this study was conducted to identify the upstream mechanism by which Mn activates the mTOR/KiSS-1 pathway. On day 12, immature female rats began receiving a daily supplemental dose of 10 mg/kg of MnCl2 or saline by gavage, and POA/AVPV tissues were collected on day 29 for specific protein assessments. Another experiment assessed in vitro IGF-1 release in response to Mn and assessed signal transduction pathways in the POA/AVPV region after Mn delivery into the third ventricle. Chronic Mn exposure increased (P < .05) basal expressions of mTOR and kisspeptin proteins. Mn increased protein kinase B (Akt) and Ras homolog enriched in brain, both capable of activating mTOR. Central Mn delivery increased expressions of phosphorylated IGF-1 receptor (IGF-1R) (P < .05) and Akt (P < .01) in the POA/AVPV region. The previous central delivery of JB1, an IGF-1R antagonist, blocked Mn-induced expressions of both phosphorylated IGF-1R and Akt. Downstream to Akt, centrally administered Mn increased tuberous sclerosis complex 2 (P < .05), Ras homolog enriched in brain (P < .01), mTOR (P < .05), and kisspeptin (P < .05). Finally, we observed that the early puberty induced by Mn was blocked by the administration of an mTOR inhibitor. These results suggest that Mn acts, at least in part, through the IGF-1/Akt/mTOR pathway to influence prepubertal kisspeptin and LHRH.
Collapse
Affiliation(s)
- Vinod K Srivastava
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843-4458
| | - Jill K Hiney
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843-4458
| | - William L Dees
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843-4458
| |
Collapse
|
22
|
Parmalee NL, Aschner M. Manganese and aging. Neurotoxicology 2016; 56:262-268. [PMID: 27293182 DOI: 10.1016/j.neuro.2016.06.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 06/04/2016] [Accepted: 06/05/2016] [Indexed: 12/11/2022]
Abstract
Manganese (Mn) is an essential metal that is required as a cofactor for many enzymes and is necessary for optimal biological function. Mn is abundant in the earth's crust and is present in soil and well water. Mn is also found in industrial settings, including mining, welding, and battery manufacture. Mn is also present in infant formula, parenteral nutrition, as well as pesticides and gasoline additives. A sufficient amount of Mn is obtained from most diets, and Mn deficiency is exceedingly rare. Excessive exposure to Mn in high doses can result in a condition known as manganism that results in psychological and emotional disturbances and motor symptoms that are reminiscent of Parkinson's disease, including gait disturbance, tremor, rigidity, and bradykinesia. Treatment for manganism is to remove the patient from Mn exposure, though symptoms are generally irreversible. The effects of exposure to Mn at lower doses are less clear. Little work has been done to evaluate the effects of chronic exposure to subclinical levels of Mn, especially in regard to lifelong exposures and the effects on the aging process. Mn is known to have effects on some of the same mechanistic processes that are altered in aging. This review will describe the general effects of Mn exposure and will focus on how Mn may be related to some of the mechanism of aging: neurogenesis, oxidative stress, and microglial activation and inflammation.
Collapse
Affiliation(s)
- Nancy L Parmalee
- Albert Einstein College of Medicine, Department of Molecular Pharmacology, 1300 Morris Park Avenue, Bronx, NY, United States.
| | - Michael Aschner
- Albert Einstein College of Medicine, Department of Molecular Pharmacology, 1300 Morris Park Avenue, Bronx, NY, United States.
| |
Collapse
|
23
|
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
|
24
|
Kraft AD. The use of glial data in human health assessments of environmental contaminants. Toxicology 2015; 333:127-136. [PMID: 25912087 DOI: 10.1016/j.tox.2015.04.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 04/10/2015] [Accepted: 04/21/2015] [Indexed: 11/27/2022]
Abstract
Central nervous system (CNS) glia (i.e., astrocytes, microglia, and oligodendrocytes) are essential for maintaining neuronal homeostasis, and they orchestrate an organized cellular response to CNS injury. In addition to their beneficial roles, studies have demonstrated that disrupted glial function can have disastrous consequences on neuronal health. While effects on neuron-supportive glia are important to consider when evaluating neurotoxicity risk, interpreting glial changes is not always straightforward, particularly when attempting to discern pro-neurotoxic phenotypes from homeostatic processes or adaptive responses. To better understand how glia have been characterized and used in human health assessments of environmental contaminants (e.g., chemicals), an evaluation of all finalized assessments conducted by the U.S. Environmental Protection Agency's influential Integrated Risk Information System (IRIS) program between 1987 and 2013 was performed. Human health assessments to date have placed a clear emphasis on the neuronal cell response to potential toxicants, although more recent assessments increasingly include descriptions of glial changes. However, these descriptions are generally brief and non-specific, and they primarily consist of documenting gliosis following overt neuronal injury. As research interest in this topic continues to increase, methods for evaluating changes in glia continue to be expanded and refined, and assessors' confidence in the reliability of these data is likely to rise. Thus, glial data are anticipated to have an increasingly influential impact on the interpretation of neurotoxicity risk and underlying mechanisms. As our understanding of the complex roles these cells play grows, this knowledge is expected to support the inclusion of more extensive and specific descriptions of glial changes, including informed interpretations of the potential impact on CNS health, in future human health assessments.
Collapse
Affiliation(s)
- Andrew D Kraft
- National Center for Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, 1200 Pennsylvania Ave., N.W., Mail Code 8623P, Washington, D.C. 20460, United States.
| |
Collapse
|
25
|
Singh V, Mitra S, Sharma AK, Gera R, Ghosh D. Isolation and Characterization of Microglia from Adult Mouse Brain: Selected Applications for ex Vivo Evaluation of Immunotoxicological Alterations Following in Vivo Xenobiotic Exposure. Chem Res Toxicol 2014; 27:895-903. [DOI: 10.1021/tx500046k] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Vikas Singh
- Immunotoxicology
Division, CSIR-Indian Institute of Toxicology Research, Lucknow-226001, Uttar Pradesh, India
- Academy
of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2
Rafi Marg, New Delhi-110
001, India
| | - Sumonto Mitra
- Immunotoxicology
Division, CSIR-Indian Institute of Toxicology Research, Lucknow-226001, Uttar Pradesh, India
| | - Anuj Kumar Sharma
- Immunotoxicology
Division, CSIR-Indian Institute of Toxicology Research, Lucknow-226001, Uttar Pradesh, India
| | - Ruchi Gera
- Immunotoxicology
Division, CSIR-Indian Institute of Toxicology Research, Lucknow-226001, Uttar Pradesh, India
- Academy
of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2
Rafi Marg, New Delhi-110
001, India
| | - Debabrata Ghosh
- Immunotoxicology
Division, CSIR-Indian Institute of Toxicology Research, Lucknow-226001, Uttar Pradesh, India
| |
Collapse
|
26
|
Astiz M, Acaz-Fonseca E, Garcia-Segura LM. Sex Differences and Effects of Estrogenic Compounds on the Expression of Inflammatory Molecules by Astrocytes Exposed to the Insecticide Dimethoate. Neurotox Res 2013; 25:271-85. [DOI: 10.1007/s12640-013-9417-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 07/26/2013] [Accepted: 08/03/2013] [Indexed: 12/31/2022]
|
27
|
Anwar K, Ejaz S, Ashraf M, Ahmad N, Javeed A. Monitoring trace elements generated by automobiles: air pollutants with possible health impacts. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2013; 20:4574-4586. [PMID: 23263758 DOI: 10.1007/s11356-012-1383-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 11/29/2012] [Indexed: 06/01/2023]
Abstract
Major transformations in the environmental composition are principally attributable to the combustion of fuels by automobiles. Motorized gasoline-powered two-stroke auto-rickshaws (TSA) and compressed natural gas (CNG)-powered four-stroke auto-rickshaws (FSA) are potential source of air pollution in south Asia and produce toxic amount of particulate matter (PM) to the environment. In this study, we attempted to characterize elemental pollutants from the PM of TSA and FSA using proton-induced X-ray emission (PIXE) analysis. The observations of the existing investigation recognized significant increase in Al (P < 0.05), P (P < 0.01), and Zn (P < 0.01) from the PM samples of FSA. In addition, the concentrations of Cu, Fe, K, Mg, Na and S were also observed exceeding the recommended National Institute for Environmental Studies limits. On the contrary, increased concentration of Sr and V were observed in the PM samples from TSA. It is generally believed that FSA generates smaller amount of PM but data obtained from FSA are clearly describing that emissions from FSA comprised potentially more toxic substances than TSA. The current research is specific to metropolitan population and has evidently revealed an inconsistent burden of exposure to air pollutants engendered by FSA in urban communities, which could lead to the disruption of several biological activities and may cause severe damage to entire ecological system.
Collapse
Affiliation(s)
- Khaleeq Anwar
- Department of Pharmacology and Toxicology, University of Veterinary and Animal Sciences, Abdul Qadir Jilani Road, Lahore, Pakistan 54600
| | | | | | | | | |
Collapse
|
28
|
Cordova FM, Aguiar AS, Peres TV, Lopes MW, Gonçalves FM, Pedro DZ, Lopes SC, Pilati C, Prediger RDS, Farina M, Erikson KM, Aschner M, Leal RB. Manganese-exposed developing rats display motor deficits and striatal oxidative stress that are reversed by Trolox. Arch Toxicol 2013; 87:1231-44. [PMID: 23385959 DOI: 10.1007/s00204-013-1017-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 01/22/2013] [Indexed: 01/05/2023]
Abstract
While manganese (Mn) is essential for proper central nervous system (CNS) development, excessive Mn exposure may lead to neurotoxicity. Mn preferentially accumulates in the basal ganglia, and in adults it may cause Parkinson's disease-like disorder. Compared to adults, younger individuals accumulate greater Mn levels in the CNS and are more vulnerable to its toxicity. Moreover, the mechanisms mediating developmental Mn-induced neurotoxicity are not completely understood. The present study investigated the developmental neurotoxicity elicited by Mn exposure (5, 10 and 20 mg/kg; i.p.) from postnatal day 8 to PN27 in rats. Neurochemical analyses were carried out on PN29, with a particular focus on striatal alterations in intracellular signaling pathways (MAPKs, Akt and DARPP-32), oxidative stress generation and cell death. Motor alterations were evaluated later in life at 3, 4 or 5 weeks of age. Mn exposure (20 mg/kg) increased p38(MAPK) and Akt phosphorylation, but decreased DARPP-32-Thr-34 phosphorylation. Mn (10 and 20 mg/kg) increased caspase activity and F2-isoprostane production (a biological marker of lipid peroxidation). Paralleling the changes in striatal biochemical parameters, Mn (20 mg/kg) also caused motor impairment, evidenced by increased falling latency in the rotarod test, decreased distance traveled and motor speed in the open-field test. Notably, the antioxidant Trolox™ reversed the Mn (20 mg/kg)-dependent augmentation in p38(MAPK) phosphorylation and reduced the Mn (20 mg/kg)-induced caspase activity and F2-isoprostane production. Trolox™ also reversed the Mn-induced motor coordination deficits. These findings are the first to show that long-term exposure to Mn during a critical period of neurodevelopment causes motor coordination dysfunction with parallel increment in oxidative stress markers, p38(MAPK) phosphorylation and caspase activity in the striatum. Moreover, we establish Trolox™ as a potential neuroprotective agent given its efficacy in reversing the Mn-induced neurodevelopmental effects.
Collapse
Affiliation(s)
- Fabiano M Cordova
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
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
|
30
|
Abstract
Autism spectrum disorders (ASDs) including classic autism is a group of complex developmental disabilities with core deficits of impaired social interactions, communication difficulties and repetitive behaviors. Although the neurobiology of ASDs has attracted much attention in the last two decades, the role of microglia has been ignored. Existing data are focused on their recognized role in neuroinflammation, which only covers a small part of the pathological repertoire of microglia. This review highlights recent findings on the broader roles of microglia, including their active surveillance of brain microenvironments and regulation of synaptic connectivity, maturation of brain circuitry and neurogenesis. Emerging evidence suggests that microglia respond to pre- and postnatal environmental stimuli through epigenetic interface to change gene expression, thus acting as effectors of experience-dependent synaptic plasticity. Impairments of these microglial functions could substantially contribute to several major etiological factors of autism, such as environmental toxins and cortical underconnectivity. Our recent study on Rett syndrome, a syndromic autistic disorder, provides an example that intrinsic microglial dysfunction due to genetic and epigenetic aberrations could detrimentally affect the developmental trajectory without evoking neuroinflammation. We propose that ASDs provide excellent opportunities to study the influence of microglia on neurodevelopment, and this knowledge could lead to novel therapies.
Collapse
|
31
|
Cordova FM, Aguiar AS, Peres TV, Lopes MW, Gonçalves FM, Remor AP, Lopes SC, Pilati C, Latini AS, Prediger RDS, Erikson KM, Aschner M, Leal RB. In vivo manganese exposure modulates Erk, Akt and Darpp-32 in the striatum of developing rats, and impairs their motor function. PLoS One 2012; 7:e33057. [PMID: 22427945 PMCID: PMC3302787 DOI: 10.1371/journal.pone.0033057] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2011] [Accepted: 02/06/2012] [Indexed: 11/30/2022] Open
Abstract
Manganese (Mn) is an essential metal for development and metabolism. However, exposures to high Mn levels may be toxic, especially to the central nervous system (CNS). Neurotoxicity is commonly due to occupational or environmental exposures leading to Mn accumulation in the basal ganglia and a Parkinsonian-like disorder. Younger individuals are more susceptible to Mn toxicity. Moreover, early exposure may represent a risk factor for the development of neurodegenerative diseases later in life. The present study was undertaken to investigate the developmental neurotoxicity in an in vivo model of immature rats exposed to Mn (5, 10 and 20 mg/kg; i.p.) from postnatal day 8 (PN8) to PN12. Neurochemical analysis was carried out on PN14. We focused on striatal alterations in intracellular signaling pathways, oxidative stress and cell death. Moreover, motor alterations as a result of early Mn exposure (PN8-12) were evaluated later in life at 3-, 4- and 5-weeks-of-age. Mn altered in a dose-dependent manner the activity of key cell signaling elements. Specifically, Mn increased the phosphorylation of DARPP-32-Thr-34, ERK1/2 and AKT. Additionally, Mn increased reactive oxygen species (ROS) production and caspase activity, and altered mitochondrial respiratory chain complexes I and II activities. Mn (10 and 20 mg/kg) also impaired motor coordination in the 3rd, 4th and 5th week of life. Trolox™, an antioxidant, reversed several of the Mn altered parameters, including the increased ROS production and ERK1/2 phosphorylation. However, Trolox™ failed to reverse the Mn (20 mg/kg)-induced increase in AKT phosphorylation and motor deficits. Additionally, Mn (20 mg/kg) decreased the distance, speed and grooming frequency in an open field test; Trolox™ blocked only the decrease of grooming frequency. Taken together, these results establish that short-term exposure to Mn during a specific developmental window (PN8-12) induces metabolic and neurochemical alterations in the striatum that may modulate later-life behavioral changes. Furthermore, some of the molecular and behavioral events, which are perturbed by early Mn exposure are not directly related to the production of oxidative stress.
Collapse
Affiliation(s)
- Fabiano M. Cordova
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
- Centro de Ciência Animal, Universidade Federal do Tocantins, Araguaína, Brazil
| | - Aderbal S. Aguiar
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Tanara V. Peres
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Mark W. Lopes
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Filipe M. Gonçalves
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Aline P. Remor
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Samantha C. Lopes
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Célso Pilati
- Centro de Ciências Agroveterinárias, Universidade do Estado de Santa Catarina, Lages, Brazil
| | - Alexandra S. Latini
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Rui D. S. Prediger
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Keith M. Erikson
- Department of Nutrition, University of North Carolina, Greensboro, North Carolina, United States of America
| | - Michael Aschner
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Rodrigo B. Leal
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
- * E-mail:
| |
Collapse
|
32
|
Streifel KM, Moreno JA, Hanneman WH, Legare ME, Tjalkens RB. Gene deletion of nos2 protects against manganese-induced neurological dysfunction in juvenile mice. Toxicol Sci 2012; 126:183-92. [PMID: 22174044 PMCID: PMC3289496 DOI: 10.1093/toxsci/kfr335] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 12/02/2011] [Indexed: 01/15/2023] Open
Abstract
The mechanisms underlying cognitive and neurobehavioral abnormalities associated with childhood exposure to manganese (Mn) are not well understood but may be influenced by neuroinflammatory activation of microglia and astrocytes that results in nitrosative stress due to expression of inducible nitric oxide synthase (iNOS/NOS2). We therefore postulated that gene deletion of NOS2 would protect against the neurotoxic effects of Mn in vivo and in vitro. Juvenile NOS2 knockout (NOS2(-/-)) mice were orally exposed to 50 mg/kg of MnCl₂ by intragastric gavage from days 21 to 34 postnatal. Results indicate that NOS2(-/-) mice exposed to Mn were protected against neurobehavioral alterations, despite histopathological activation of astrocytes and microglia in Mn-treated mice in both genotypes. NOS2(-/-) mice had decreased Mn-induced formation of 3-nitrotyrosine protein adducts within neurons in the basal ganglia that correlated with protection against Mn-induced neurobehavioral defects. Primary striatal astrocytes from wildtype mice caused apoptosis in cocultured striatal neurons following treatment with MnCl₂ and tumor necrosis factor-α, whereas NOS2(-/-) astrocytes failed to cause any increase in markers of apoptosis in striatal neurons. Additionally, scavenging nitric oxide (NO) with 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (PTIO) prevented the ability of Mn- and cytokine-treated wildtype astrocytes to cause apoptosis in cocultured striatal neurons. These data demonstrate that NO plays a crucial role in Mn-induced neurological dysfunction in juvenile mice and that NOS2 expression in activated glia is an important mediator of neuroinflammatory injury during Mn exposure.
Collapse
Affiliation(s)
| | | | | | | | - Ronald B. Tjalkens
- Center for Environmental Medicine, Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado 80523-1680
| |
Collapse
|
33
|
Harry GJ, Kraft AD. Microglia in the developing brain: a potential target with lifetime effects. Neurotoxicology 2012; 33:191-206. [PMID: 22322212 DOI: 10.1016/j.neuro.2012.01.012] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 01/24/2012] [Accepted: 01/25/2012] [Indexed: 12/15/2022]
Abstract
Microglia are a heterogenous group of monocyte-derived cells serving multiple roles within the brain, many of which are associated with immune and macrophage like properties. These cells are known to serve a critical role during brain injury and to maintain homeostasis; yet, their defined roles during development have yet to be elucidated. Microglial actions appear to influence events associated with neuronal proliferation and differentiation during development, as well as, contribute to processes associated with the removal of dying neurons or cellular debris and management of synaptic connections. These long-lived cells display changes during injury and with aging that are critical to the maintenance of the neuronal environment over the lifespan of the organism. These processes may be altered by changes in the colonization of the brain or by inflammatory events during development. This review addresses the role of microglia during brain development, both structurally and functionally, as well as the inherent vulnerability of the developing nervous system. A framework is presented considering microglia as a critical nervous system-specific cell that can influence multiple aspects of brain development (e.g., vascularization, synaptogenesis, and myelination) and have a long term impact on the functional vulnerability of the nervous system to a subsequent insult, whether environmental, physical, age-related, or disease-related.
Collapse
Affiliation(s)
- G Jean Harry
- National Toxicology Program Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| | | |
Collapse
|
34
|
Filipov NM, Dodd CA. Role of glial cells in manganese neurotoxicity. J Appl Toxicol 2011; 32:310-7. [PMID: 22120544 DOI: 10.1002/jat.1762] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 09/27/2011] [Accepted: 09/27/2011] [Indexed: 11/06/2022]
Abstract
The objectives of this focused review are to (i) provide a systematic overview of recent advances pertaining to the role of glia, namely microglia and astrocytes, in the neuropathology associated with excessive exposure to manganese (Mn), (ii) highlight possible mechanisms and factors involved in Mn-modulated, glia-derived neuroinflammation, and (iii) discuss the implications of excessive neuroinflammation on neuronal injury within the context of Mn overexposure. As this is not meant to be a comprehensive review on the topic of Mn neurotoxicity, the reader may wish to refer to several broader and more comprehensive reviews. After a brief introduction to Mn neurotoxicity, we first discuss the role of glial cells in neurodegeneration. Next, we review existing in vitro and in vivo studies that implicate Mn as a modulator of glial activation and ensuing neuroinflammation. This is followed by an examination of recognized and potential mechanisms that are involved in the modulation of glial inflammatory output by Mn; here the common pathways activated by Mn in glial and neuronal cells, including outcomes of such activation, are also addressed. We finish with a discussion of the implications of Mn-modulated glial activation for neuronal survival and with a list of data gaps in the topic that need to be filled in the future.
Collapse
Affiliation(s)
- Nikolay M Filipov
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA.
| | | |
Collapse
|
35
|
Chtourou Y, Fetoui H, Garoui EM, Boudawara T, Zeghal N. Improvement of cerebellum redox states and cholinergic functions contribute to the beneficial effects of silymarin against manganese-induced neurotoxicity. Neurochem Res 2011; 37:469-79. [PMID: 22033861 DOI: 10.1007/s11064-011-0632-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 08/03/2011] [Accepted: 10/05/2011] [Indexed: 12/17/2022]
Abstract
Manganese (Mn) is a potent neurotoxin involved in the initiation and progression of various cognitive disorders. Oxidative stress is reported as one of accepted mechanisms of Mn toxicity. The present study was designed to explore the effects of silymarin, a natural antioxidant, in attenuating the toxicity induced by Mn in rat cerebellum. In this investigation, rats were treated orally with MnCl₂ (20 mg/ml) for 30 days, subsets of these animals were treated intraperitoneally daily with silymarin (100 mg/kg) along with respective controls. Mn exposure caused a marked oxidative stress in cerebellum as indicated by a significant decrease in the activities of enzymatic antioxidants like superoxide dismutase, catalase and glutathione peroxidase and in the levels of non-enzymatic antioxidants like reduced glutathione (GSH), total thiols and vitamin C. Conversely an increase was obtained in lipid and protein markers such as thiobarbituric reactive acid substances, lipid hydroperoxide and protein carbonyl products contents. A significant increase in acetylcholinesterase and a decrease in Na⁺/K⁺-ATPase activities were also shown, with a substantial rise in the expression of acetylcholinesterase and inducible nitric oxide synthase (iNOS), and nitric oxide levels. The potential effect of SIL to prevent Mn induced neurotoxicity was also reflected by histopathological observations. Rats exposed to Mn showed a reduced number and morphological alterations of cerebellar Purkinje cells. These phenomenons were completely reversed by SIL co-treatment. We concluded that silymarin may protect against Mn-induced oxidative stress in cerebellum by inhibiting both lipid and protein oxidation and by activating acetylcholinesterase and inducible nitric oxide synthase (iNOS) gene expression.
Collapse
Affiliation(s)
- Yassine Chtourou
- Animal Physiology Laboratory, Life Sciences Department, UR/08-73, Sfax Faculty of Sciences, University of Sfax, BP1171, 3000 Sfax, Tunisia
| | | | | | | | | |
Collapse
|
36
|
Blaylock RL, Maroon J. Immunoexcitotoxicity as a central mechanism in chronic traumatic encephalopathy-A unifying hypothesis. Surg Neurol Int 2011; 2:107. [PMID: 21886880 PMCID: PMC3157093 DOI: 10.4103/2152-7806.83391] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 06/06/2011] [Indexed: 12/17/2022] Open
Abstract
Some individuals suffering from mild traumatic brain injuries, especially repetitive mild concussions, are thought to develop a slowly progressive encephalopathy characterized by a number of the neuropathological elements shared with various neurodegenerative diseases. A central pathological mechanism explaining the development of progressive neurodegeneration in this subset of individuals has not been elucidated. Yet, a large number of studies indicate that a process called immunoexcitotoxicity may be playing a central role in many neurodegenerative diseases including chronic traumatic encephalopathy (CTE). The term immunoexcitotoxicity was first coined by the lead author to explain the evolving pathological and neurodevelopmental changes in autism and the Gulf War Syndrome, but it can be applied to a number of neurodegenerative disorders. The interaction between immune receptors within the central nervous system (CNS) and excitatory glutamate receptors trigger a series of events, such as extensive reactive oxygen species/reactive nitrogen species generation, accumulation of lipid peroxidation products, and prostaglandin activation, which then leads to dendritic retraction, synaptic injury, damage to microtubules, and mitochondrial suppression. In this paper, we discuss the mechanism of immunoexcitotoxicity and its link to each of the pathophysiological and neurochemical events previously described with CTE, with special emphasis on the observed accumulation of hyperphosphorylated tau.
Collapse
Affiliation(s)
- Russell L Blaylock
- Theoretical Neurosciences, LLC Visiting Professor of Biology, Belhaven University, Jackson, MS 315 Rolling Meadows Rd, Ridgeland, MS 39157, USA
| | | |
Collapse
|
37
|
Moreno JA, Streifel KM, Sullivan KA, Hanneman WH, Tjalkens RB. Manganese-induced NF-kappaB activation and nitrosative stress is decreased by estrogen in juvenile mice. Toxicol Sci 2011; 122:121-33. [PMID: 21512103 DOI: 10.1093/toxsci/kfr091] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Manganese toxicity can cause a neurodegenerative disorder affecting cortical and basal ganglia structures with a neurological presentation resembling features of Parkinson's disease. Children are more sensitive to Mn-induced neurological dysfunction than adults, and recent studies from our laboratory revealed a marked sensitivity of male juvenile mice to neuroinflammatory injury from Mn, relative to females. To determine the role of estrogen (E2) in mediating sex-dependent vulnerability to Mn-induced neurotoxicity, we exposed transgenic mice expressing an NF-κB-driven enhanced green fluorescent protein (EGFP) reporter construct (NF-κB-EGFP mice) to Mn, postulating that supplementing male mice with E2 during juvenile development would attenuate neuroinflammatory changes associated with glial activation, including expression of inducible nitric oxide synthase (NOS2) and neuronal protein nitration. Juvenile NF-κB-EGFP mice were separated in groups composed of females, males, and males surgically implanted with Silastic capsules containing 25 μg of 17-β-estradiol (E2) or vehicle control. Mice were then treated with 0 or 100 mg/Kg MnCl(2) by intragastric gavage from postnatal days 21-34. Manganese treatment caused alterations in levels of striatal dopamine, as well as increases in NF-κB reporter activity and NOS2 expression in both microglia and astrocytes that were prevented by supplementation with E2. E2 also decreased neuronal protein nitration in Mn-treated mice and inhibited apoptosis in striatal neurons cocultured with Mn-treated astrocytes in vitro. These data indicate that E2 protects against Mn-induced neuroinflammation in developing mice and that NF-κB is an important regulator of neuroinflammatory gene expression in glia associated with nitrosative stress in the basal ganglia during Mn exposure.
Collapse
Affiliation(s)
- Julie A Moreno
- Center for Environmental Medicine, Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado 80523-1680, USA
| | | | | | | | | |
Collapse
|
38
|
Paris I, Segura-Aguilar J. The role of metal ions in dopaminergic neuron degeneration in Parkinsonism and Parkinson’s disease. MONATSHEFTE FUR CHEMIE 2011. [DOI: 10.1007/s00706-011-0478-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
39
|
Martinez-Finley EJ, Avila DS, Chakraborty S, Aschner M. Insights from Caenorhabditis elegans on the role of metals in neurodegenerative diseases. Metallomics 2011; 3:271-9. [PMID: 21210060 PMCID: PMC3172965 DOI: 10.1039/c0mt00064g] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neurodegeneration is characterized by the cell death or loss of structure and/or function of neurons. Many neurodegenerative diseases including Parkinson's disease (PD) and Alzheimer's disease (AD) are the result of neurodegenerative processes. Metals are essential for many life processes, but they are also culpable for several neurodegenerative mechanisms. In this review, we discuss the role of metals in neurodegenerative diseases with emphasis on the utility of Caenorhabditis elegans (C. elegans) genetic models in deciphering mechanisms associated with the etiology of PD and AD.
Collapse
Affiliation(s)
- Ebany J. Martinez-Finley
- Division of Clinical Pharmacology and Pediatric Toxicology, Vanderbilt University Medical Center, 11425 MRB IV, 2215-B Garland Ave., Nashville, TN 37232-0414, USA; Tel: 615-322-8024
- Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daiana Silva Avila
- Division of Clinical Pharmacology and Pediatric Toxicology, Vanderbilt University Medical Center, 11425 MRB IV, 2215-B Garland Ave., Nashville, TN 37232-0414, USA; Tel: 615-322-8024
| | - Sudipta Chakraborty
- Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Molecular Neuroscience, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael Aschner
- Division of Clinical Pharmacology and Pediatric Toxicology, Vanderbilt University Medical Center, 11425 MRB IV, 2215-B Garland Ave., Nashville, TN 37232-0414, USA; Tel: 615-322-8024
- Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Molecular Neuroscience, Vanderbilt University Medical Center, Nashville, TN, USA
- The Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Division of Pediatric Toxicology, Nashville, TN, USA
| |
Collapse
|
40
|
Leifer CA, Dietert RR. Early life environment and developmental immunotoxicity in inflammatory dysfunction and disease. TOXICOLOGICAL AND ENVIRONMENTAL CHEMISTRY 2011; 93:1463-1485. [PMID: 26146439 PMCID: PMC4486307 DOI: 10.1080/02772248.2011.586114] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Components of the innate immune system such as macrophages and dendritic cells are instrumental in determining the fate of immune responses and are, also, among the most sensitive targets of early life environmental alterations including developmental immunotoxicity (DIT). DIT can impede innate immune cell maturation, disrupt tissue microenvironment, alter immune responses to infectious challenges, and disrupt regulatory responses. Dysregulation of inflammation, such as that observed with DIT, has been linked with an increased risk of chronic inflammatory diseases in both children and adults. In this review, we discuss the relationship between early-life risk factors for innate immune modulation and promotion of dysregulated inflammation associated with chronic inflammatory disease. The health risks from DIT-associated inflammation may extend beyond primary immune dysfunction to include an elevated risk of several later-life, inflammatory-mediated diseases that target a wide range of physiological systems and organs. For this reason, determination of innate immune status should be an integral part of drug and chemical safety evaluation.
Collapse
Affiliation(s)
- Cynthia A. Leifer
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Rodney R. Dietert
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
41
|
Tjalkens RB, Carbone DL, Wu G. Detection of nitric oxide formation in primary neural cells and tissues. Methods Mol Biol 2011; 758:267-77. [PMID: 21815072 DOI: 10.1007/978-1-61779-170-3_18] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nitric oxide (NO) is a free radical molecule with a short half-life (<5 s). Because its synthesis from L-arginine by constitutive NO synthase (NOS) is low in many cell types, including neurons and endothelial cells, direct detection of NO in biological systems is a difficult task. During pathological conditions in the CNS, the inducible form of NOS (iNOS or NOS2) is expressed in activated astrocytes and microglial cells and can result in higher levels of NO. However, it may still be difficult to detect NO in these cell types using typical spectrophotometric methods. Of particular note, NO is readily oxidized to nitrite and nitrate (relatively stable products) in cells and medium, which can be measured as a valid indicator of NO synthesis. The conversion of NO to peroxynitrite leads to the formation of stable protein adducts that can be detected by immunohistochemical or immunofluorescence methods. Additionally, intracellular levels of NO can be detected in real time using fluorescence imaging and NO-specific, cell permeable indicator dyes.
Collapse
Affiliation(s)
- Ronald B Tjalkens
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.
| | | | | |
Collapse
|
42
|
Winder BS, Salmon AG, Marty MA. Inhalation of an essential metal: Development of reference exposure levels for manganese. Regul Toxicol Pharmacol 2010; 57:195-9. [DOI: 10.1016/j.yrtph.2010.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 01/29/2010] [Accepted: 02/16/2010] [Indexed: 10/19/2022]
|