1
|
Wu J, Li Y, Tian S, Na S, Wei H, Wu Y, Yang Y, Shen Z, Ding J, Bao S, Liu S, Li L, Feng R, Zhu Y, He C, Yue J. CYP1B1 affects the integrity of the blood-brain barrier and oxidative stress in the striatum: An investigation of manganese-induced neurotoxicity. CNS Neurosci Ther 2024; 30:e14633. [PMID: 38429921 PMCID: PMC10907825 DOI: 10.1111/cns.14633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 01/19/2024] [Accepted: 01/25/2024] [Indexed: 03/03/2024] Open
Abstract
AIMS Excessive influx of manganese (Mn) into the brain across the blood-brain barrier induces neurodegeneration. CYP1B1 is involved in the metabolism of arachidonic acid (AA) that affects vascular homeostasis. We aimed to investigate the effect of brain CYP1B1 on Mn-induced neurotoxicity. METHOD Brain Mn concentrations and α-synuclein accumulation were measured in wild-type and CYP1B1 knockout mice treated with MnCl2 (30 mg/kg) and biotin (0.2 g/kg) for 21 continuous days. Tight junctions and oxidative stress were analyzed in hCMEC/D3 and SH-SY5Y cells after the treatment with MnCl2 (200 μM) and CYP1B1-derived AA metabolites (HETEs and EETs). RESULTS Mn exposure inhibited brain CYP1B1, and CYP1B1 deficiency increased brain Mn concentrations and accelerated α-synuclein deposition in the striatum. CYP1B1 deficiency disrupted the integrity of the blood-brain barrier (BBB) and increased the ratio of 3, 4-dihydroxyphenylacetic acid (DOPAC) to dopamine in the striatum. HETEs attenuated Mn-induced inhibition of tight junctions by activating PPARγ in endothelial cells. Additionally, EETs attenuated Mn-induced up-regulation of the KLF/MAO-B axis and down-regulation of NRF2 in neuronal cells. Biotin up-regulated brain CYP1B1 and reduced Mn-induced neurotoxicity in mice. CONCLUSIONS Brain CYP1B1 plays a critical role in both cerebrovascular and dopamine homeostasis, which might serve as a novel therapeutic target for the prevention of Mn-induced neurotoxicity.
Collapse
Affiliation(s)
- Juan Wu
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
- Department of PharmacyTaikang Tongji (Wuhan) HospitalWuhuChina
| | - Yueran Li
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
- Department of PharmacyThe First Affiliated Hospital of Wannan Medical CollegeWuhuChina
| | - Shuwei Tian
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Shufang Na
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan UniversityTransplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on TransplantationWuhanHubeiChina
| | - Hongyan Wei
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Yafei Wu
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Yafei Yang
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Zixia Shen
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Jiayue Ding
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Shenglan Bao
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Siqi Liu
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Lingyun Li
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Rongling Feng
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Yong Zhu
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Chunyan He
- Demonstration Center for Experimental Basic Medicine Education, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Jiang Yue
- Department of Pharmacology, School of Basic Medical SciencesWuhan UniversityWuhanChina
- Hubei Province Key Laboratory of Allergy and ImmunologyWuhanChina
| |
Collapse
|
2
|
Li B, Yu W, Verkhratsky A. Trace metals and astrocytes physiology and pathophysiology. Cell Calcium 2024; 118:102843. [PMID: 38199057 DOI: 10.1016/j.ceca.2024.102843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 01/12/2024]
Abstract
Several trace metals, including iron, copper, manganese and zinc are essential for normal function of the nervous system. Both deficiency and excessive accumulation of these metals trigger neuropathological developments. The central nervous system (CNS) is in possession of dedicated homeostatic system that removes, accumulates, stores and releases these metals to fulfil nervous tissue demand. This system is mainly associated with astrocytes that act as dynamic reservoirs for trace metals, these being a part of a global system of CNS ionostasis. Here we overview physiological and pathophysiological aspects of astrocyte-cantered trace metals regulation.
Collapse
Affiliation(s)
- Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China; Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, China; China Medical University Centre of Forensic Investigation, China
| | - Weiyang Yu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China; Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, China; China Medical University Centre of Forensic Investigation, China
| | - Alexei Verkhratsky
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, Ikerbasque, Bilbao 48011, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius LT-01102, Lithuania.
| |
Collapse
|
3
|
Latronico T, Rossano R, Miniero DV, Casalino E, Liuzzi GM. Neuroprotective Effect of Resveratrol against Manganese-Induced Oxidative Stress and Matrix Metalloproteinase-9 in an "In Vivo" Model of Neurotoxicity. Int J Mol Sci 2024; 25:2142. [PMID: 38396818 PMCID: PMC10888573 DOI: 10.3390/ijms25042142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Chronic exposure to manganese (Mn) leads to its accumulation in the central nervous system (CNS) and neurotoxicity with not well-known mechanisms. We investigated the involvement of matrix metalloproteinase (MMP)-2 and -9 in Mn neurotoxicity in an in vivo model of rats treated through an intraperitoneal injection, for 4 weeks, with 50 mg/kg of MnCl2 in the presence or in the absence of 30 mg/kg of resveratrol (RSV). A loss of weight was observed in Mn-treated rats compared with untreated and RSV-treated rats. A progressive recovery of body weight was detected in rats co-treated with Mn and RSV. The analysis of brain homogenates indicated that RSV counteracted the Mn-induced increase in MMP-9 levels and reactive oxygen species production as well as the Mn-induced decrease in superoxide dismutase activity and glutathione content. In conclusion, Mn exposure, resulting in MMP-9 induction with mechanisms related to oxidative stress, represents a risk factor for the development of CNS diseases.
Collapse
Affiliation(s)
- Tiziana Latronico
- Department of Biosciences, Biotechnologies and Environment, University of Bari “A. Moro”, 70126 Bari, Italy; (D.V.M.); (G.M.L.)
| | - Rocco Rossano
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy;
| | - Daniela Valeria Miniero
- Department of Biosciences, Biotechnologies and Environment, University of Bari “A. Moro”, 70126 Bari, Italy; (D.V.M.); (G.M.L.)
| | - Elisabetta Casalino
- Department of Veterinary Medicine, University of Bari “A. Moro”, 70010 Bari, Italy;
| | - Grazia Maria Liuzzi
- Department of Biosciences, Biotechnologies and Environment, University of Bari “A. Moro”, 70126 Bari, Italy; (D.V.M.); (G.M.L.)
| |
Collapse
|
4
|
Ng MG, Chan BJL, Koh RY, Ng KY, Chye SM. Prevention of Parkinson's Disease: From Risk Factors to Early Interventions. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:746-760. [PMID: 37326115 DOI: 10.2174/1871527322666230616092054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 05/15/2023] [Accepted: 05/19/2023] [Indexed: 06/17/2023]
Abstract
Parkinson's disease (PD) is a debilitating neurological disorder characterized by progressively worsening motor dysfunction. Currently, available therapies merely alleviate symptoms, and there are no cures. Consequently, some researchers have now shifted their attention to identifying the modifiable risk factors of PD, with the intention of possibly implementing early interventions to prevent the development of PD. Four primary risk factors for PD are discussed including environmental factors (pesticides and heavy metals), lifestyle (physical activity and dietary intake), drug abuse, and individual comorbidities. Additionally, clinical biomarkers, neuroimaging, biochemical biomarkers, and genetic biomarkers could also help to detect prodromal PD. This review compiled available evidence that illustrates the relationship between modifiable risk factors, biomarkers, and PD. In summary, we raise the distinct possibility of preventing PD via early interventions of the modifiable risk factors and early diagnosis.
Collapse
Affiliation(s)
- Ming Guan Ng
- School of Health Science, International Medical University, 57000 Kuala Lumpur, Malaysia
| | - Brendan Jun Lam Chan
- School of Health Science, International Medical University, 57000 Kuala Lumpur, Malaysia
| | - Rhun Yian Koh
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, Kuala Lumpur, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University, 47500 Selangor, Malaysia
| | - Soi Moi Chye
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
5
|
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
|
6
|
Dorman DC. The Role of Oxidative Stress in Manganese Neurotoxicity: A Literature Review Focused on Contributions Made by Professor Michael Aschner. Biomolecules 2023; 13:1176. [PMID: 37627240 PMCID: PMC10452838 DOI: 10.3390/biom13081176] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
This literature review focuses on the evidence implicating oxidative stress in the pathogenesis of manganese neurotoxicity. This review is not intended to be a systematic review of the relevant toxicologic literature. Instead, in keeping with the spirit of this special journal issue, this review highlights contributions made by Professor Michael Aschner's laboratory in this field of study. Over the past two decades, his laboratory has made significant contributions to our scientific understanding of cellular responses that occur both in vitro and in vivo following manganese exposure. These studies have identified molecular targets of manganese toxicity and their respective roles in mitochondrial dysfunction, inflammation, and cytotoxicity. Other studies have focused on the critical role astrocytes play in manganese neurotoxicity. Recent studies from his laboratory have used C. elegans to discover new facets of manganese-induced neurotoxicity. Collectively, his body of work has dramatically advanced the field and presents broader implications beyond metal toxicology.
Collapse
Affiliation(s)
- David C Dorman
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, 1052 William Moore Dr, Raleigh, NC 27606, USA
| |
Collapse
|
7
|
Puty B, Bittencourt LO, Lima LAO, Plaça JR, Dionizio A, Buzalaf MAR, Gomes BD, de Oliveira EHC, Lima RR. Unraveling molecular characteristic of fluoride neurotoxicity on U87 glial-like cells: insights from transcriptomic and proteomic approach. Front Cell Neurosci 2023; 17:1153198. [PMID: 37362003 PMCID: PMC10289037 DOI: 10.3389/fncel.2023.1153198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 05/19/2023] [Indexed: 06/28/2023] Open
Abstract
The potential of fluoride (F) as a neurotoxicant in humans is still controversial in the literature. However, recent studies have raised the debate by showing different mechanism of F-induced neurotoxicity, as oxidative stress, energy metabolism and inflammation in the central nervous system (CNS). In the present study, we investigated the mechanistic action of two F concentration (0.095 and 0.22 μg/ml) on gene and protein profile network using a human glial cell in vitro model over 10 days of exposure. A total of 823 genes and 2,084 genes were modulated after exposure to 0.095 and 0.22 μg/ml F, respectively. Among them, 168 were found to be modulated by both concentrations. The number of changes in protein expression induced by F were 20 and 10, respectively. Gene ontology annotations showed that the main terms were related to cellular metabolism, protein modification and cell death regulation pathways, such as the MAP kinase (MAPK) cascade, in a concentration independent manner. Proteomics confirmed the changes in energy metabolism and also provided evidence of F-induced changes in cytoskeleton components of glial cells. Our results not only reveal that F has the potential to modulate gene and protein profiles in human U87 glial-like cells overexposed to F, but also identify a possible role of this ion in cytoskeleton disorganization.
Collapse
Affiliation(s)
- Bruna Puty
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Laboratory of Cell Culture and Cytogenetics, Environmental Section, Evandro Chagas Institute, Ananindeua, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Leidiane Alencar Oliveira Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Jéssica Rodrigues Plaça
- National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, Centro de Pesquisa, Inovacão e Desenvolvimento/Fundacão de Amparo á Pesuisa do Estado de São Paulo (CEPID/FAPESP), Ribeirão Preto, Brazil
| | - Aline Dionizio
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | | | - Bruno Duarte Gomes
- Laboratory of Neurophysiology Eduardo Oswaldo Cruz, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | | | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| |
Collapse
|
8
|
Agarrayua DA, Silva AC, Saraiva NR, Soares AT, Aschner M, Avila DS. Neurotoxicology of metals and metallic nanoparticles in Caenorhabditis elegans. ADVANCES IN NEUROTOXICOLOGY 2023; 9:107-148. [PMID: 37384197 PMCID: PMC10306323 DOI: 10.1016/bs.ant.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Affiliation(s)
- Danielle Araujo Agarrayua
- Graduate Program in Biochemistry, Laboratory of Biochemistry and Toxicology in Caenorhabditis elegans, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Aline Castro Silva
- Graduate Program in Biochemistry, Laboratory of Biochemistry and Toxicology in Caenorhabditis elegans, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Nariani Rocha Saraiva
- Graduate Program in Biochemistry, Laboratory of Biochemistry and Toxicology in Caenorhabditis elegans, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Ana Thalita Soares
- Graduate Program in Biochemistry, Laboratory of Biochemistry and Toxicology in Caenorhabditis elegans, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Daiana Silva Avila
- Graduate Program in Biochemistry, Laboratory of Biochemistry and Toxicology in Caenorhabditis elegans, Federal University of Pampa, Uruguaiana, RS, Brazil
- Graduate Program in Biological Sciences- Toxicological Biochemistry, Federal University of Santa Maria, RS, Brazil
| |
Collapse
|
9
|
Aaseth JO, Nurchi VM. Chelation Combination-A Strategy to Mitigate the Neurotoxicity of Manganese, Iron, and Copper? Biomolecules 2022; 12:1713. [PMID: 36421727 PMCID: PMC9687779 DOI: 10.3390/biom12111713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/13/2022] [Accepted: 11/16/2022] [Indexed: 01/19/2024] Open
Abstract
The chelating thiol dimercaptosuccinate (DMSA) and the traditional agent D-penicillamine (PSH) are effective in enhancing the urinary excretion of copper (Cu) and lead (Pb) in poisoned individuals. However, DMSA, PSH, EDTA (ethylenediamine tetraacetate), and deferoxamine (DFOA) are water-soluble agents with limited access to the central nervous system (CNS). Strategies for mobilization of metals such as manganese (Mn), iron (Fe), and Cu from brain deposits may require the combined use of two agents: one water-soluble agent to remove circulating metal into urine, in addition to an adjuvant shuttler to facilitate the brain-to-blood mobilization. The present review discusses the chemical basis of metal chelation and the ligand exchange of metal ions. To obtain increased excretion of Mn, Cu, and Fe, early experiences showed promising results for CaEDTA, PSH, and DFOA, respectively. Recent experiments have indicated that p-amino salicylate (PAS) plus CaEDTA may be a useful combination to remove Mn from binding sites in CNS, while the deferasirox-DFOA and the tetrathiomolybdate-DMSA combinations may be preferable to promote mobilization of Fe and Cu, respectively, from the CNS. Further research is requested to explore benefits of chelator combinations.
Collapse
Affiliation(s)
- Jan O. Aaseth
- Department of Research, Innlandet Hospital Trust, P.O. Box 104, N-2381 Brumunddal, Norway
- Faculty of Health and Social Sciences, Inland Norway University of Applied Sciences, P.O. Box 104, N-2418 Elverum, Norway
| | - Valeria M. Nurchi
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy
| |
Collapse
|
10
|
Nabi M, Tabassum N. Role of Environmental Toxicants on Neurodegenerative Disorders. FRONTIERS IN TOXICOLOGY 2022; 4:837579. [PMID: 35647576 PMCID: PMC9131020 DOI: 10.3389/ftox.2022.837579] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/22/2022] [Indexed: 12/22/2022] Open
Abstract
Neurodegeneration leads to the loss of structural and functioning components of neurons over time. Various studies have related neurodegeneration to a number of degenerative disorders. Neurological repercussions of neurodegeneration can have severe impacts on the physical and mental health of patients. In the recent past, various neurodegenerative ailments such as Alzheimer’s and Parkinson’s illnesses have received global consideration owing to their global occurrence. Environmental attributes have been regarded as the main contributors to neural dysfunction-related disorders. The majority of neurological diseases are mainly related to prenatal and postnatal exposure to industrially produced environmental toxins. Some neurotoxic metals, like lead (Pb), aluminium (Al), Mercury (Hg), manganese (Mn), cadmium (Cd), and arsenic (As), and also pesticides and metal-based nanoparticles, have been implicated in Parkinson’s and Alzheimer’s disease. The contaminants are known for their ability to produce senile or amyloid plaques and neurofibrillary tangles (NFTs), which are the key features of these neurological dysfunctions. Besides, solvent exposure is also a significant contributor to neurological diseases. This study recapitulates the role of environmental neurotoxins on neurodegeneration with special emphasis on major neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease.
Collapse
Affiliation(s)
- Masarat Nabi
- Department of Environmental Science, University of Kashmir, Srinagar, India
- *Correspondence: Masarat Nabi, , orcid.org/0000-0003-1677-6498; Nahida Tabassum,
| | - Nahida Tabassum
- Department of Pharmaceutical Sciences, University of Kashmir, Srinagar, India
- *Correspondence: Masarat Nabi, , orcid.org/0000-0003-1677-6498; Nahida Tabassum,
| |
Collapse
|
11
|
Marwah PK, Paik G, Issa CJ, Jemison CC, Qureshi MB, Hanna TM, Palomino E, Maddipati KR, Njus D. Manganese-stimulated redox cycling of dopamine derivatives: Implications for manganism. Neurotoxicology 2022; 90:10-18. [PMID: 35217070 DOI: 10.1016/j.neuro.2022.02.007] [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: 12/07/2021] [Revised: 02/06/2022] [Accepted: 02/17/2022] [Indexed: 10/19/2022]
Abstract
Manganism, the condition caused by chronic exposure to high levels of manganese, selectively targets the dopamine-rich basal ganglia causing a movement disorder with symptoms similar to Parkinson's disease. While the basis for this specific targeting is unknown, we hypothesize that it may involve complexation of Mn by dopamine derivatives. At micromolar concentrations, MnCl2 accelerates the two-equivalent redox cycling of a dopamine-derived benzothiazine (dopathiazine) by an order of magnitude. In the process, O2 is reduced to superoxide and hydrogen peroxide. This effect is unique to Mn and is not shared by Fe, Cu, Zn, Co, Ca or Mg. Notably, the effect of Mn requires the presence of inorganic phosphate, suggesting that phosphate may stabilize a Mn/catecholate complex, which reacts readily with O2. This or similar endogenous dopamine derivatives may exacerbate Mn-dependent oxidative stress accounting for the neurological selectivity of manganism.
Collapse
Affiliation(s)
- Praneet Kaur Marwah
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Gijong Paik
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Christopher J Issa
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | | | - Muhammad B Qureshi
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Tareq M Hanna
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Eduardo Palomino
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA; Walker Cancer Research Institute, 5047 Gullen Mall, Detroit, MI 48202, USA
| | - Krishna Rao Maddipati
- Department of Pathology, Wayne State Univ. School of Medicine, Detroit, MI 48201, USA
| | - David Njus
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA.
| |
Collapse
|
12
|
Ivleva IS, Ivlev AP, Pestereva NS, Tyutyunnik TV, Karpenko MN. Protective effect of calpain inhibitors against manganese-induced toxicity in rats. Metab Brain Dis 2022; 37:1003-1013. [PMID: 35089484 DOI: 10.1007/s11011-022-00916-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/17/2022] [Indexed: 10/19/2022]
Abstract
Development of manganism is a major complication of manganese exposure in which neurological dysfunction is linked to accumulation of metal in the brain. Current therapies do not prevent progression of the disease. Therefore, development of effective therapeutic strategies for treatment of manganism is of utmost importance. Since the hyperactivation of calpain family proteases in CNS during manganism in an animal model is observed, we assumed that inhibition of calpains can suppress the development of Mn-induced neurological disturbances. The goal of this study is to delineate protective effect and the mechanism of neuroprotection of calpain inhibitor in rat model of Mn-induced neurological symptoms. Using the Gait analysis test, we found that chronic intranasal administration of the calpain inhibitor Cast (184-210) (peptide, which is corresponding to the 184-210 amino acid of the endogenous inhibitor of calpains-human calpastatin) to Mn-treated rats contributed to a significant decrease in the severity of gait disorders, although it did not lead to a decrease in the Mn deposition in the striatum and hippocampus. Accordingly to the results of PCR-RT, this effect was accompanied by a partial reduction in the content of neuro-inflammatory markers (IL-1β, TNF-α, NFκB mRNA in the hippocampus and, additionally, IBA-1 mRNA in the striatum), as well as normalization of the content of dopamine and its metabolites in the hippocampus and striatum, which was assessed by HPLC. In striatum cells, the application of Cast (184-210) also led to a significant increase in the production of tyrosine hydroxylase, which was analyzed by immunoblotting method. These findings suggest that calpain inhibitors may be a valid therapeutic agent in manganism.
Collapse
Affiliation(s)
- I S Ivleva
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia.
| | - A P Ivlev
- Sechenov Institute of Evolutionary Physiology and Biochemistry Russian Academy of Sciences, St. Petersburg, Russia
| | - N S Pestereva
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia
| | - T V Tyutyunnik
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia
| | - M N Karpenko
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia
| |
Collapse
|
13
|
Sarkar S. Mechanism of Gene-Environment Interactions Driving Glial Activation in Parkinson's Diseases. Curr Environ Health Rep 2021; 8:203-211. [PMID: 34043217 DOI: 10.1007/s40572-021-00320-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE OF REVIEW Parkinson's disease (PD) is the most prevalent motor disorder and is characterized by loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) region of the brain. Though the pathology of PD is well established, the cause of this neuronal loss is not well understood. Approximately 90% of PD cases are sporadic, and the environment plays a significant role in disease pathogenesis. The etiology of PD is highly complex, with neuroinflammation being one of the most critical factors implicated in PD. However, the signaling mechanisms underlying neuroinflammation and its interaction with environmental factors are unclear. RECENT FINDINGS Astroglia and microglia are the two principal cells that play an essential role in maintaining neuronal health in many ways, including through immunological means. Exposure to environmental stressors from various sources affects these glial cells leading to chronic and sustained inflammation. Recent epidemiological studies have identified an interaction among environmental factors and glial genes in Parkinson's disease. Mechanistic studies have shown that exposure to pesticides like rotenone and paraquat, neurotoxic metals like manganese and lead, and even diesel exhaust fumes induce glial activation by regulating various key inflammatory pathways, including the inflammasomes, NOX pathways, and others. This review aims to discuss the recent advances in understanding the mechanism of glial induction in response to environmental stressors and discuss the potential role of gene-environment interaction in driving glial activation.
Collapse
Affiliation(s)
- Souvarish Sarkar
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| |
Collapse
|
14
|
Zhang C, Xing Z, Tan M, Wu Y, Zeng W. Roflumilast Ameliorates Isoflurane-Induced Inflammation in Astrocytes via the CREB/BDNF Signaling Pathway. ACS OMEGA 2021; 6:4167-4174. [PMID: 33644540 PMCID: PMC7906587 DOI: 10.1021/acsomega.0c04799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/29/2020] [Indexed: 06/12/2023]
Abstract
Background and purpose: Astrocyte-mediated neuroinflammation plays an important role in anesthetic isoflurane-induced cognitive impairment. Roflumilast, a selective inhibitor of phosphodiesterase-4 (PDE-4) used for the treatment of chronic obstructive pulmonary disease (COPD), has displayed a wide range of anti-inflammatory capacity in different types of cells and tissues. In the current study, we aimed to investigate whether roflumilast possesses a protective effect against isoflurane-induced insults in mouse primary astrocytes. Methods: Primary astrocytes were isolated from the cerebral cortices of immature rats. The production of NO was determined using DAF-FM DA staining assay. QRT-PCR and western blot were used to evaluate the expression levels of iNOS, COX-2, and BDNF in the astrocytes treated with different therapies. The gene expressions and concentrations of IL-6 and MCP-1 released by the astrocytes were detected using qRT-PCR and ELISA, respectively. The expression levels of phosphorylated CREB and PGE2 were determined using western blot and ELISA, respectively. H89 was introduced to evaluate the function of CREB. Recombinant human BDNF and ANA-12 were used to verify the role of BDNF. Results: The upregulated iNOS, excessive production of NO, IL-6, and MCP-1, and activated COX-2/PGE2 signaling pathways in the astrocytes induced by isoflurane were significantly reversed by the introduction of roflumilast, in a dose-dependent manner. Subsequently, we found that BDNF could be upregulated by roflumilast, which was verified to be related to the activation of CREB and blocked by H89 (a CREB inhibitor). In addition, the COX-2/PGE2 signaling pathway activated by isoflurane can be inactivated by recombinant human BDNF. Finally, the regulatory effect of roflumilast against the isoflurane-activated COX-2/PGE2 signaling pathway was significantly blocked by ANA-12, which is a BDNF inhibitor. Conclusion: Roflumilast might ameliorate isoflurane-induced inflammation in astrocytes via the CREB/BDNF signaling pathway.
Collapse
|
15
|
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
|
16
|
Cheng JJ, Guo Q, Wu XG, Ma S, Gao Y, Ya-Zhen S. Scutellaria barbata flavonoids improve the composited Aβ-induced abnormal changes of glial cells in rats' brain. Comb Chem High Throughput Screen 2020; 25:64-76. [PMID: 33297910 DOI: 10.2174/1386207323666201209092358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/21/2020] [Accepted: 11/15/2020] [Indexed: 11/22/2022]
Abstract
AIM It has been reported that glial cells are involved in Alzheimer's disease (AD). According to our previous research, Scutellaria barbata flavonoids (SBFs) can protect the neuronal disorder and memory impairment for AD-like rats, while the effect of SBFs on the glial cells disorder in AD-like rats has been less well studied. The effects of SBFs on astrocytes(ASs), microglial cells (MGs) and oligodendrocytes (Ols), as well as heat shock proteins 70 (Hsp70) and apolipoprotein E (ApoE) were investigated in the present study. METHODS The successful model rats, screened by Morris water maze, were daily orally administrated with 35, 70 and 140 mg/kg SBFs for 36 d. The numbers of brain's astrocytes (ASs), microglial cells (MGs) and oligodendrocytes (Ols) were examined by immunohistochemistry. The cortical glial fibrillary acidic protein (GFAP), leukocyte common antigen (LCA) (CD45), Claudin 11 and heat shock proteins 70 (Hsp70) protein expression were assayed by Western blotting, and apolipoprotein E (ApoE) mRNA expression was analyzed by real-time quantitative polymerase chain reaction (qPCR). RESULTS Compared with the sham-operated group, the numbers of ASs and MGs in the brain were significantly increased in the model group (P<0.05, P<0.01), and accompanied with increases of GFAP, CD45 and Hsp70 protein and ApoE mRNA expression (P<0.05, P<0.01). Both Ols number and Claudin 11 protein expression decreased in the brain in the model group (P<0.05, P<0.01). However, the above abnormal changes induced by composited Aβ were differently reversed by treatment of SBFs at three doses of 35, 70 and 140 mg/kg (P<0.05, P<0.01). CONCLUSIONS SBFs can dramatically improve the abnormal changes of glial cells in rats' brain induced by composited Aβ, which may be a helpful treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jian-Jun Cheng
- Institute of Traditional Chinese Medicine, Chengde Medical College, Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia, Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development, Chengde, Hebei 067000. China
| | - Qing Guo
- The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011. China
| | - Xiao-Guang Wu
- Institute of Traditional Chinese Medicine, Chengde Medical College, Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia, Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development, Chengde, Hebei 067000. China
| | - Shuai Ma
- Institute of Traditional Chinese Medicine, Chengde Medical College, Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia, Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development, Chengde, Hebei 067000. China
| | - Yang Gao
- Institute of Traditional Chinese Medicine, Chengde Medical College, Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia, Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development, Chengde, Hebei 067000. China
| | - Shang Ya-Zhen
- Institute of Traditional Chinese Medicine, Chengde Medical College, Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia, Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development, Chengde, Hebei 067000. China
| |
Collapse
|
17
|
Zhan X, Wen G, Jiang E, Li F, Wu X, Pang H. Secretogranin III upregulation is involved in parkinsonian toxin-mediated astroglia activation. J Toxicol Sci 2020; 45:271-280. [PMID: 32404559 DOI: 10.2131/jts.45.271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Environmental neurotoxins such as paraquat (PQ), manganese, and 1-1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) are associated with a higher risk of Parkinson's disease (PD). These parkinsonian toxins exert certain common toxicological effects on astroglia; however, their role in the regulatory functions of astroglial secretory proteins remains unclear. In a previous study, we observed that secretogranin II (SCG2) and secretogranin III (SCG3), which are important components of the regulated secretory pathway, were elevated in PQ-activated U118 astroglia. In the current study, we used the parkinsonian toxins dopamine (DA), active metabolite of MPTP (MPP+), MnCl2, and lipopolysaccharide (LPS) as inducers, and studied the potential regulation of SCG2 and SCG3. Our results showed that all the parkinsonian toxins except LPS affected astroglial viability but did not cause apoptosis. Exposure to DA, MPP+, and MnCl2 upregulated glial fibrillary acidic protein (GFAP), a marker for astrocyte activation, and stimulated the levels of several astrocytic-derived factors. Further, DA, MPP+, and MnCl2 exposure impeded astroglial cell cycle progression. Moreover, the expression of SCG3 was elevated, while its exosecretion was inhibited in astroglia activated by parkinsonian toxins. The level of SCG2 remained unchanged. In combination with our previous findings, the results of this study indicate that SCG3 may act as a cofactor in astrocyte activation stimulated by various toxins, and the regulation of SCG3 could be involved in the toxicological mechanism by which parkinsonian toxins affect astroglia.
Collapse
Affiliation(s)
- Xiaoni Zhan
- Department of Forensic Genetics and Biology, School of Forensic Medicine, China Medical University, China
| | - Gehua Wen
- Department of Forensic Clinical Medicine, School of Forensic Medicine, China Medical University, China
| | - Enzhu Jiang
- Department of Forensic Genetics and Biology, School of Forensic Medicine, China Medical University, China
| | | | - Xu Wu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, China
| | - Hao Pang
- Department of Forensic Genetics and Biology, School of Forensic Medicine, China Medical University, China
| |
Collapse
|
18
|
Zhang Z, Yan J, Bowman AB, Bryan MR, Singh R, Aschner M. Dysregulation of TFEB contributes to manganese-induced autophagic failure and mitochondrial dysfunction in astrocytes. Autophagy 2020; 16:1506-1523. [PMID: 31690173 PMCID: PMC7469609 DOI: 10.1080/15548627.2019.1688488] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 10/09/2019] [Accepted: 10/30/2019] [Indexed: 01/16/2023] Open
Abstract
Epidemiological and clinical studies have long shown that exposure to high levels of heavy metals are associated with increased risks of neurodegenerative diseases. It is widely accepted that autophagic dysfunction is involved in pathogenesis of various neurodegenerative disorders; however, the role of heavy metals in regulation of macroautophagy/autophagy is unclear. Here, we show that manganese (Mn) induces a decline in nuclear localization of TFEB (transcription factor EB), a master regulator of the autophagy-lysosome pathway, leading to autophagic dysfunction in astrocytes of mouse striatum. We further show that Mn exposure suppresses autophagic-lysosomal degradation of mitochondria and induces accumulation of unhealthy mitochondria. Activation of autophagy by rapamycin or TFEB overexpression ameliorates Mn-induced mitochondrial respiratory dysfunction and reactive oxygen species (ROS) generation in astrocytes, suggesting a causal relation between autophagic failure and mitochondrial dysfunction in Mn toxicity. Taken together, our data demonstrate that Mn inhibits TFEB activity, leading to impaired autophagy that is causally related to mitochondrial dysfunction in astrocytes. These findings reveal a previously unappreciated role for Mn in dysregulation of autophagy and identify TFEB as a potential therapeutic target to mitigate Mn toxicity. ABBREVIATIONS BECN1: beclin 1; CTSD: cathepsin D; DMEM: Dulbecco's Modified Eagle Medium; GFAP: glial fibrillary acid protein; GFP: green fluorescent protein; HBSS: hanks balanced salt solution; LAMP: lysosomal-associated membrane protein; LDH: lactate dehydrogenase; Lys Inh: lysosomal inhibitors; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MAPK: mitogen-activated protein kinase; Mn: manganese; MTOR: mechanistic target of rapamycin kinase; OCR: oxygen consumption rate; PBS: phosphate-buffered saline; PFA: paraformaldehyde; PI: propidium iodide; ROS: reactive oxygen species; s.c.: subcutaneous; SQSTM1/p62: sequestosome 1; TEM: transmission electron microscopy; TFEB: transcription factor EB.
Collapse
Affiliation(s)
- Ziyan Zhang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jingqi Yan
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Miles R. Bryan
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology and Biochemistry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rajat Singh
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine
- Diabetes Research Center
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
19
|
Naz Z, Moin ST, Hofer TS. Hydration of Closely Related Manganese and Magnesium Porphyrins in Aqueous Solutions: Ab Initio Quantum Mechanical Charge Field Molecular Dynamics Simulation Study. J Phys Chem B 2019; 123:10769-10779. [PMID: 31738566 DOI: 10.1021/acs.jpcb.9b07639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
To the best of our knowledge, the current study based on ab initio quantum mechanical charge field molecular dynamics (QMCF-MD) is the first to explore the difference in the hydration behavior between Mn(II)- and Mg(II)-associated porphyrins (Mn(II)-POR and Mg(II)-POR) in aqueous solution. The simulation study highlights similar and dissimilar characteristics of the structural, dynamical, and thermodynamical properties of these closely related metals bound to porphyrins in aqueous solution. The structural analysis is based on radial and angular distribution functions, coordination number distributions, and angular-radial distributions. Both hydrated systems demonstrate similar pentacoordinated structures formed via the axial coordination of one water molecule to the metal ion in addition to the four nitrogen atoms of the porphyrin ring. However, in the case of Mn(II)-POR, the formation of a distorted square pyramidal geometry was observed. It was envisaged as a weak coordination of the water molecule to the Mn(II) atom and thus higher atomic fluctuation for all atoms in contrast to that for the hydrated Mg(II)-POR. The dynamical data in terms of the mean residence times, velocity autocorrelation function, free energy, and other parameters revealed the difference in the metal binding effect because the Mn(II) atom was observed to inhibit H-bond formation more than the presence of Mg(II) atoms in the core of the porphyrin. The current study thus highlights the significant differences in the structural and dynamical properties of Mn(II)- and Mg(II)-associated porphyrin systems.
Collapse
Affiliation(s)
- Zobia Naz
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences , University of Karachi , Karachi 75270 , Pakistan
| | - Syed Tarique Moin
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences , University of Karachi , Karachi 75270 , Pakistan
| | - Thomas S Hofer
- Theoretical Chemistry Division, Institute of General, Inorganic and Theoretical Chemistry , University of Innsbruck , Innrain 80-82 , A-6020 Innsbruck , Austria
| |
Collapse
|
20
|
Rizor A, Pajarillo E, Johnson J, Aschner M, Lee E. Astrocytic Oxidative/Nitrosative Stress Contributes to Parkinson's Disease Pathogenesis: The Dual Role of Reactive Astrocytes. Antioxidants (Basel) 2019; 8:antiox8080265. [PMID: 31374936 PMCID: PMC6719180 DOI: 10.3390/antiox8080265] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/23/2019] [Accepted: 07/30/2019] [Indexed: 12/20/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease worldwide; it is characterized by dopaminergic neurodegeneration in the substantia nigra pars compacta, but its etiology is not fully understood. Astrocytes, a class of glial cells in the central nervous system (CNS), provide critical structural and metabolic support to neurons, but growing evidence reveals that astrocytic oxidative and nitrosative stress contributes to PD pathogenesis. As astrocytes play a critical role in the production of antioxidants and the detoxification of reactive oxygen and nitrogen species (ROS/RNS), astrocytic oxidative/nitrosative stress has emerged as a critical mediator of the etiology of PD. Cellular stress and inflammation induce reactive astrogliosis, which initiates the production of astrocytic ROS/RNS and may lead to oxidative/nitrosative stress and PD pathogenesis. Although the cause of aberrant reactive astrogliosis is unknown, gene mutations and environmental toxicants may also contribute to astrocytic oxidative/nitrosative stress. In this review, we briefly discuss the physiological functions of astrocytes and the role of astrocytic oxidative/nitrosative stress in PD pathogenesis. Additionally, we examine the impact of PD-related genes such as α-synuclein, protein deglycase DJ-1( DJ-1), Parkin, and PTEN-induced kinase 1 (PINK1) on astrocytic function, and highlight the impact of environmental toxicants, such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), rotenone, manganese, and paraquat, on astrocytic oxidative/nitrosative stress in experimental models.
Collapse
Affiliation(s)
- Asha Rizor
- Department of Pharmaceutical Sciences, College of Pharmacy Florida A&M University, Tallahassee, FL 32301, USA
| | - Edward Pajarillo
- Department of Pharmaceutical Sciences, College of Pharmacy Florida A&M University, Tallahassee, FL 32301, USA
| | - James Johnson
- Department of Pharmaceutical Sciences, College of Pharmacy Florida A&M University, Tallahassee, FL 32301, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine Bronx, New York, NY 10461, USA
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy Florida A&M University, Tallahassee, FL 32301, USA.
| |
Collapse
|
21
|
Sachse B, Kolbaum AE, Ziegenhagen R, Andres S, Berg K, Dusemund B, Hirsch-Ernst KI, Kappenstein O, Müller F, Röhl C, Lindtner O, Lampen A, Schäfer B. Dietary Manganese Exposure in the Adult Population in Germany-What Does it Mean in Relation to Health Risks? Mol Nutr Food Res 2019; 63:e1900065. [PMID: 31216097 DOI: 10.1002/mnfr.201900065] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/29/2019] [Indexed: 11/10/2022]
Abstract
Manganese is both an essential nutrient and a potential neurotoxicant. Therefore, the question arises whether the dietary manganese intake in the German population is on the low or high side. Results from a pilot total diet study in Germany presented here reveal that the average dietary manganese intake in the general population in Germany aged 14-80 years is about 2.8 mg day-1 for a person of 70 kg body weight. This exposure level is within the intake range of 2-5 mg per person and day as recommended by the societies for nutrition in Germany, Austria, and Switzerland. No information on the dietary exposure of children in Germany can be provided so far. Although reliable information on health effects related to oral manganese exposure is limited, there is no indication from the literature that these dietary intake levels are associated with adverse health effects either by manganese deficiency or excess. However, there is limited evidence that manganese taken up as a highly bioavailable bolus, for example, uptake via drinking water or food supplements, could pose a potential risk to human health-particularly in certain subpopulations-when certain intake amounts, which are currently not well defined, are exceeded.
Collapse
Affiliation(s)
- Benjamin Sachse
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| | - Anna Elena Kolbaum
- German Federal Institute for Risk Assessment (BfR), Department of Exposure, 12277, Berlin, Germany
| | - Rainer Ziegenhagen
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| | - Susanne Andres
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| | - Katharina Berg
- German Federal Institute for Risk Assessment (BfR), Department of Exposure, 12277, Berlin, Germany
| | - Birgit Dusemund
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| | - Karen Ildico Hirsch-Ernst
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| | - Oliver Kappenstein
- German Federal Institute for Risk Assessment (BfR), Department of Chemicals and Product Safety, 10589, Berlin, Germany
| | - Frederic Müller
- German Federal Institute for Risk Assessment (BfR), Department of Chemicals and Product Safety, 10589, Berlin, Germany
| | - Claudia Röhl
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany.,State Agency for Social Services Schleswig-Holstein (LAsD), Department of Environmental Health Protection, 24105, Kiel, Germany
| | - Oliver Lindtner
- German Federal Institute for Risk Assessment (BfR), Department of Exposure, 12277, Berlin, Germany
| | - Alfonso Lampen
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| | - Bernd Schäfer
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| |
Collapse
|
22
|
Ammonia-induced mitochondrial impairment is intensified by manganese co-exposure: relevance to the management of subclinical hepatic encephalopathy and cirrhosis-associated brain injury. Clin Exp Hepatol 2019; 5:109-117. [PMID: 31501786 PMCID: PMC6728860 DOI: 10.5114/ceh.2019.85071] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 10/29/2018] [Indexed: 12/30/2022] Open
Abstract
Aim of the study Hepatic encephalopathy (HE) is a neuropsychiatric syndrome ensuing from liver failure. The liver is the major site of ammonia detoxification in the human body. Hence, acute and chronic liver dysfunction can lead to hyperammonemia. Manganese (Mn) is a trace element incorporated in several physiological processes in the human body. Mn is excreted through bile. It has been found that cirrhosis is associated with hyperammonemia as well as body Mn accumulation. The brain is the primary target organ for both ammonia and Mn toxicity. On the other hand, brain mitochondria impairment is involved in the mechanism of Mn and ammonia neurotoxicity. Material and methods The current study was designed to evaluate the effect of Mn and ammonia and their combination on mitochondrial indices of functionality in isolated brain mitochondria. Isolated brain mitochondria were exposed to increasing concentrations of ammonia and Mn alone and/or in combination and several mitochondrial indices were assessed. Results The collapse of mitochondrial membrane potential, increased mitochondrial permeabilization, reactive oxygen species formation, and a significant decrease in mitochondrial dehydrogenase activity and ATP content were evident in Mn-exposed (0.005-1 mM) brain mitochondria. On the other hand, ammonia (0.005-0.5 mM) caused no significant changes in brain mitochondrial function. It was found that co-exposure of the brain mitochondria to Mn and ammonia causes more evident mitochondrial impairment in comparison with Mn and/or ammonia alone. Conclusions These data indicate additive toxicity of ammonia and Mn in isolated brain mitochondria exposed to these neurotoxins.
Collapse
|
23
|
Kanthasamy A, Jin H, Charli A, Vellareddy A, Kanthasamy A. Environmental neurotoxicant-induced dopaminergic neurodegeneration: a potential link to impaired neuroinflammatory mechanisms. Pharmacol Ther 2019; 197:61-82. [PMID: 30677475 PMCID: PMC6520143 DOI: 10.1016/j.pharmthera.2019.01.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
With the increased incidence of neurodegenerative diseases worldwide, Parkinson's disease (PD) represents the second-most common neurodegenerative disease. PD is a progressive multisystem neurodegenerative disorder characterized by a marked loss of nigrostriatal dopaminergic neurons and the formation of Lewy pathology in diverse brain regions. Although the mechanisms underlying dopaminergic neurodegeneration remain poorly characterized, data from animal models and postmortem studies have revealed that heightened inflammatory responses mediated via microglial and astroglial activation and the resultant release of proinflammatory factors may act as silent drivers of neurodegeneration. In recent years, numerous studies have demonstrated a positive association between the exposure to environmental neurotoxicants and the etiology of PD. Although it is unclear whether neuroinflammation drives pesticide-induced neurodegeneration, emerging evidence suggests that the failure to dampen neuroinflammatory mechanisms may account for the increased vulnerability to pesticide neurotoxicity. Furthermore, recent studies provide additional evidence that shifts the focus from a neuron-centric view to glial-associated neurodegeneration following pesticide exposure. In this review, we propose to summarize briefly the possible factors that regulate neuroinflammatory processes during environmental neurotoxicant exposure with a focus on the potential roles of mitochondria-driven redox mechanisms. In this context, a critical discussion of the data obtained from experimental research and possible epidemiological studies is included. Finally, we hope to provide insights on the pivotal role of exosome-mediated intercellular transmission of aggregated proteins in microglial activation response and the resultant dopaminergic neurodegeneration after exposure to pesticides. Collectively, an improved understanding of glia-mediated neuroinflammatory signaling might provide novel insights into the mechanisms that contribute to neurodegeneration induced by environmental neurotoxicant exposure.
Collapse
Affiliation(s)
- Arthi Kanthasamy
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA.
| | - Huajun Jin
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Adhithiya Charli
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Anantharam Vellareddy
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Anumantha Kanthasamy
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
24
|
Heidari R. Brain mitochondria as potential therapeutic targets for managing hepatic encephalopathy. Life Sci 2019; 218:65-80. [DOI: 10.1016/j.lfs.2018.12.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/08/2018] [Accepted: 12/16/2018] [Indexed: 02/07/2023]
|
25
|
Verkhratsky A, Parpura V, Rodriguez-Arellano JJ, Zorec R. Astroglia in Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1175:273-324. [PMID: 31583592 DOI: 10.1007/978-981-13-9913-8_11] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease is the most common cause of dementia. Cellular changes in the brains of the patients suffering from Alzheimer's disease occur well in advance of the clinical symptoms. At the cellular level, the most dramatic is a demise of neurones. As astroglial cells carry out homeostatic functions of the brain, it is certain that these cells are at least in part a cause of Alzheimer's disease. Historically, Alois Alzheimer himself has recognised this at the dawn of the disease description. However, the role of astroglia in this disease has been understudied. In this chapter, we summarise the various aspects of glial contribution to this disease and outline the potential of using these cells in prevention (exercise and environmental enrichment) and intervention of this devastating disease.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK. .,Faculty of Health and Medical Sciences, Center for Basic and Translational Neuroscience, University of Copenhagen, 2200, Copenhagen, Denmark. .,Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain.
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA.,University of Rijeka, Rijeka, Croatia
| | - Jose Julio Rodriguez-Arellano
- BioCruces Health Research Institute, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.,Department of Neuroscience, The University of the Basque Country UPV/EHU, Plaza de Cruces 12, 48903, Barakaldo, Bizkaia, Spain
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia.,Celica BIOMEDICAL, Ljubljana, Slovenia
| |
Collapse
|
26
|
Sarkar S, Malovic E, Jin H, Kanthasamy A, Kanthasamy AG. The role of manganese in neuroinflammation. ROLE OF INFLAMMATION IN ENVIRONMENTAL NEUROTOXICITY 2019. [DOI: 10.1016/bs.ant.2018.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
27
|
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
|
28
|
Mn Inhibits GSH Synthesis via Downregulation of Neuronal EAAC1 and Astrocytic xCT to Cause Oxidative Damage in the Striatum of Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4235695. [PMID: 30228854 PMCID: PMC6136513 DOI: 10.1155/2018/4235695] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/24/2018] [Accepted: 07/12/2018] [Indexed: 11/17/2022]
Abstract
Excessive manganese (Mn) can accumulate in the striatum of the brain following overexposure. Oxidative stress is a well-recognized mechanism in Mn-induced neurotoxicity. It has been proven that glutathione (GSH) depletion is a key factor in oxidative damage during Mn exposure. However, no study has focused on the dysfunction of GSH synthesis-induced oxidative stress in the brain during Mn exposure. The objective of the present study was to explore the mechanism of Mn disruption of GSH synthesis via EAAC1 and xCT in vitro and in vivo. Primary neurons and astrocytes were cultured and treated with different doses of Mn to observe the state of cells and levels of GSH and reactive oxygen species (ROS) and measure mRNA and protein expression of EAAC1 and xCT. Mice were randomly divided into seven groups, which received saline, 12.5, 25, and 50 mg/kg MnCl2, 500 mg/kg AAH (EAAC1 inhibitor) + 50 mg/kg MnCl2, 75 mg/kg SSZ (xCT inhibitor) + 50 mg/kg MnCl2, and 100 mg/kg NAC (GSH rescuer) + 50 mg/kg MnCl2 once daily for two weeks. Then, levels of EAAC1, xCT, ROS, GSH, malondialdehyde (MDA), protein sulfhydryl, carbonyl, 8-hydroxy-2-deoxyguanosine (8-OHdG), and morphological and ultrastructural features in the striatum of mice were measured. Mn reduced protein levels, mRNA expression, and immunofluorescence intensity of EAAC1 and xCT. Mn also decreased the level of GSH, sulfhydryl, and increased ROS, MDA, 8-OHdG, and carbonyl in a dose-dependent manner. Injury-related pathological and ultrastructure changes in the striatum of mice were significantly present. In conclusion, excessive exposure to Mn disrupts GSH synthesis through inhibition of EAAC1 and xCT to trigger oxidative damage in the striatum.
Collapse
|
29
|
Johnson J, Pajarillo E, Karki P, Kim J, Son DS, Aschner M, Lee E. Valproic acid attenuates manganese-induced reduction in expression of GLT-1 and GLAST with concomitant changes in murine dopaminergic neurotoxicity. Neurotoxicology 2018; 67:112-120. [PMID: 29778792 PMCID: PMC6441963 DOI: 10.1016/j.neuro.2018.05.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 12/31/2022]
Abstract
Exposure to elevated levels of manganese (Mn) causes manganism, a neurological disorder with similar characteristics to those of Parkinson's disease (PD). Valproic acid (VPA), an antiepileptic, is known to inhibit histone deacetylases and exert neuroprotective effects in many experimental models of neurological disorders. In the present study, we investigated if VPA attenuated Mn-induced dopaminergic neurotoxicity and the possible mechanisms involved in VPA's neuroprotection, focusing on modulation of astrocytic glutamate transporters (glutamate aspartate transporter, GLAST and glutamate transporter 1, GLT-1) and histone acetylation in H4 astrocyte culture and mouse models. The results showed that VPA increased promoter activity, mRNA/protein levels of GLAST/GLT-1 and glutamate uptake, and reversed Mn-reduced GLAST/GLT-1 in in vitro astrocyte cultures. VPA also attenuated Mn-induced reduction of GLAST and GLT-1 mRNA/protein levels in midbrain and striatal regions of the mouse brain when VPA (200 mg/kg, i.p., daily, 21 d) was administered 30 min prior to Mn exposure (30 mg/kg, intranasal instillation, daily, 21 d). Importantly, VPA attenuated Mn-induced dopaminergic neuronal damage by reversing Mn-induced decrease of tyrosine hydroxylase (TH) mRNA/protein levels in the nigrostriatal regions. VPA also reversed Mn-induced reduction of histone acetylation in astrocytes as well as mouse brain tissue. Taken together, VPA exerts attenuation against Mn-induced decrease of astrocytic glutamate transporters parallel with reversing Mn-induced dopaminergic neurotoxicity and Mn-reduced histone acetylation. Our findings suggest that VPA could serve as a potential neuroprotectant against Mn neurotoxicity as well as other neurodegenerative diseases associated with excitotoxicity and impaired astrocytic glutamate transporters.
Collapse
Affiliation(s)
- James Johnson
- Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, United States
| | - Edward Pajarillo
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, 32301, United States
| | - Pratap Karki
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, 32301, United States
| | - Judong Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, 32301, United States
| | - Deok-Soo Son
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN, 37208, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, United States
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, 32301, United States.
| |
Collapse
|
30
|
Perfluorooctanesulfonate induces neuroinflammation through the secretion of TNF-α mediated by the JAK2/STAT3 pathway. Neurotoxicology 2018. [DOI: 10.1016/j.neuro.2018.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
31
|
Sharma K, Sharma D, Sharma M, Sharma N, Bidve P, Prajapati N, Kalia K, Tiwari V. Astaxanthin ameliorates behavioral and biochemical alterations in in-vitro and in-vivo model of neuropathic pain. Neurosci Lett 2018; 674:162-170. [PMID: 29559419 DOI: 10.1016/j.neulet.2018.03.030] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/03/2018] [Accepted: 03/16/2018] [Indexed: 12/15/2022]
Abstract
Despite considerable advances in understanding mechanisms involved in chronic pain, effective treatment remains limited. Astaxanthin, a marine natural drug, having potent anti-oxidant and anti-inflammatory activities is known to possess neuroprotective effects. However, effects of astaxanthin against nerve injury induce chronic pain remains unknown. Overactivity of glutamatergic NMDARs results in excitotoxicity which may participate in astrocytic and microglial activation during pathology which further contribute to the development of neuropathic pain. In this study, we investigate the effects of astaxanthin on oxido-inflammatory and NMDA receptor down-regulation pathway by using in-silico, in-vitro and in-vivo models of neuropathic pain. In-silico molecular docking study ascertained the binding affinity of astaxanthin to NMDA receptors and showed antagonistic effects. Data from in-vitro studies suggest that astaxanthin significantly reduces the oxidative stress induced by the lipopolysaccharides in C6 glial cells. In male Sprague dawley rats, a significant attenuation of neuropathic pain behavior was observed in Hargreaves test and von Frey hair test after astaxanthin treatment. Findings from the current study suggest that astaxanthin can be used as potential alternative in the treatment of chronic neuropathic pain. However, more detailed investigations are required to further probe the in-depth mechanism of action of astaxanthin.
Collapse
Affiliation(s)
- Kuhu Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Dilip Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Monika Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Nishant Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Pankaj Bidve
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Namrata Prajapati
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Vinod Tiwari
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
32
|
Role of LRRK2 in manganese-induced neuroinflammation and microglial autophagy. Biochem Biophys Res Commun 2018; 498:171-177. [PMID: 29408508 DOI: 10.1016/j.bbrc.2018.02.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 02/02/2018] [Indexed: 12/22/2022]
Abstract
Overexposure to manganese (Mn) leads to manganism and neurotoxicity induced by Mn is the focus of recent research. Microglia play a vital role in Mn-induced neurotoxicity, and our previous studies firstly showed that Mn could stimulate activation of microglia, leading to the neuroinflammation, and inhibition of microglial inflammation effectively attenuated Mn-induced death of dopamine neurons. However, the detailed mechanism of manganese-induced neuroinflammation is still unclear. Leucine rich repeat kinase 2 (LRRK2) is a key molecule in the pathogenesis of many neurodegenerative disorders. Recent studies have indicated that LRRK2, which is highly expressed in microglia, plays a specific role in microglia and autophagy process. In this paper, we try to find the effect of LRRK2 on Mn-triggered neuroinflammation and its possible mechanism in vivo and in vitro. By establishing a Mn exposure animal model, our studies found that Mn exposure could induce dopaminergic neurons damage and activate microglia. Activated microglia triggered neuroinflammation by releasing multiple inflammatory cytokines, and the expression of LRRK2 was upregulated in vivo and in vitro. We also found that Mn exposure induced autophagy dysfunction in vivo and in vitro. Next, we used LRRK2 siRNA and LRRK2-IN-1 to inhibit the expression of LRRK2, and found that inhibition of LRRK2 could not only decrease the expression of inflammatory cytokines, but also recover autophagic function of microglia. Our investigation not only reveals the role of LRRK2 in Mn-induced neuroinflammation but also sheds light on the prevention and protection of manganism.
Collapse
|
33
|
Cervantes-Yépez S, López-Zepeda LS, Fortoul TI. Vanadium inhalation induces retinal Müller glial cell (MGC) alterations in a murine model. Cutan Ocul Toxicol 2017; 37:200-206. [PMID: 29157004 DOI: 10.1080/15569527.2017.1392560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Vanadium (V) is a transition metal adhered to suspended particles. Previous studies demonstrated that V inhalation causes oxidative stress in the ependymal epithelium, the choroid plexus on brain lateral ventricles and in the retina. Inhaled-V reaches the eye´s retina through the systemic circulation; however, its effect on the retina has not been widely studied. The Müller glial cell provides support and structure to the retina, facilitates synapses and regulates the microenvironment and neuronal metabolism. Hence, it is of great interest to study the effect of V exposure on the expression and localization of specific biomarkers on this cell. METHODS Male CD-1 mice were exposed to V inhalation 1 h/twice/week for 4 and 8-Wk. Expression changes in the retina of Glial fibrillary acidic protein, highly expressed in Müller glial cell when retina is damaged, and Glutamine synthetase, important in preventing excitotoxicity in the retina, were analysed by immunohistochemistry. RESULTS Glial fibrillary acidic protein expression increased at 4-Wk of V inhalation compared to the control and decreased at 8-Wk of exposure. A time-dependent gradual reduction in glutamine synthetase expression was observed. CONCLUSION Changes in glial fibrillary acidic protein expression induced by V suggest retinal damage, whereas glutamine synthetase gradual reduction might indicate that photoreceptors, which produce most of the glutamine synthetase substrate in the retina, are degenerating, probably as a consequence of the oxidative stress induced by V.
Collapse
Affiliation(s)
- Silvana Cervantes-Yépez
- a Departamento de Biología Celular y Tisular, Facultad de Medicina , Universidad Nacional Autónoma de México (UNAM) , México City , CP , México
| | - Lorena Sofía López-Zepeda
- a Departamento de Biología Celular y Tisular, Facultad de Medicina , Universidad Nacional Autónoma de México (UNAM) , México City , CP , México
| | - Teresa I Fortoul
- a Departamento de Biología Celular y Tisular, Facultad de Medicina , Universidad Nacional Autónoma de México (UNAM) , México City , CP , México
| |
Collapse
|
34
|
Lee S, Nam Y, Jang J, Na GH, Kim DG, Shin NY, Choi HS, Jung SL, Ahn KJ, Kim BS. Deep gray matter iron measurement in patients with liver cirrhosis using quantitative susceptibility mapping: Relationship with pallidal T1
hyperintensity. J Magn Reson Imaging 2017; 47:1342-1349. [DOI: 10.1002/jmri.25841] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/02/2017] [Indexed: 01/12/2023] Open
Affiliation(s)
- Song Lee
- Department of Radiology; Seoul St. Mary's Hospital, School of Medicine, Catholic University of Korea; Seoul Korea
| | - Yoonho Nam
- Department of Radiology; Seoul St. Mary's Hospital, School of Medicine, Catholic University of Korea; Seoul Korea
| | - Jinhee Jang
- Department of Radiology; Seoul St. Mary's Hospital, School of Medicine, Catholic University of Korea; Seoul Korea
| | - Gun Hyung Na
- Department of Surgery; Seoul St. Mary's Hospital, School of Medicine, Catholic University of Korea; Seoul Korea
- Department of Surgery; Bucheon St. Mary's Hospital, School of Medicine, Catholic University of Korea; Bucheon Korea
| | - Dong Goo Kim
- Department of Surgery; Seoul St. Mary's Hospital, School of Medicine, Catholic University of Korea; Seoul Korea
| | - Na-Young Shin
- Department of Radiology; Seoul St. Mary's Hospital, School of Medicine, Catholic University of Korea; Seoul Korea
| | - Hyun Seok Choi
- Department of Radiology; Seoul St. Mary's Hospital, School of Medicine, Catholic University of Korea; Seoul Korea
| | - So-Lyung Jung
- Department of Radiology; Seoul St. Mary's Hospital, School of Medicine, Catholic University of Korea; Seoul Korea
| | - Kook-Jin Ahn
- Department of Radiology; Seoul St. Mary's Hospital, School of Medicine, Catholic University of Korea; Seoul Korea
| | - Bum-soo Kim
- Department of Radiology; Seoul St. Mary's Hospital, School of Medicine, Catholic University of Korea; Seoul Korea
| |
Collapse
|
35
|
Abstract
Although an essential nutrient, manganese (Mn) can be toxic at high doses. There is, however, uncertainty regarding the effects of chronic low-level Mn-exposure. This review provides an overview of Mn-related brain and functional changes based on studies of a cohort of asymptomatic welders who had lower Mn-exposure than in most previous work. In welders with low-level Mn-exposure, we found: 1) Mn may accumulate in the brain in a non-linear fashion: MRI R1 (1/T1) signals significantly increased only after a critical level of exposure was reached (e.g., ≥300 welding hours in the past 90days prior to MRI). Moreover, R1 may be a more sensitive marker to capture short-term dynamic changes in Mn accumulation than the pallidal index [T1-weighted intensity ratio of the globus pallidus vs. frontal white matter], a traditional marker for Mn accumulation; 2) Chronic Mn-exposure may lead to microstructural changes as indicated by lower diffusion tensor fractional anisotropy values in the basal ganglia (BG), especially when welding years exceeded more than 30 years; 3) Mn-related subtle motor dysfunctions can be captured sensitively by synergy metrics (indices for movement stability), whereas traditional fine motor tasks failed to detect any significant differences; and 4) Iron (Fe) also may play a role in welding-related neurotoxicity, especially at low-level Mn-exposure, evidenced by higher R2* values (an estimate for brain Fe accumulation) in the BG. Moreover, higher R2* values were associated with lower phonemic fluency performance. These findings may guide future studies and the development of occupation- and public health-related polices involving Mn-exposure.
Collapse
|
36
|
Li SJ, Qin WX, Peng DJ, Yuan ZX, He SN, Luo YN, Aschner M, Jiang YM, Liang DY, Xie BY, Xu F. Sodium P-aminosalicylic acid inhibits sub-chronic manganese-induced neuroinflammation in rats by modulating MAPK and COX-2. Neurotoxicology 2017; 64:219-229. [PMID: 28651968 DOI: 10.1016/j.neuro.2017.06.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 06/18/2017] [Accepted: 06/21/2017] [Indexed: 02/08/2023]
Abstract
Excessive manganese (Mn) accumulation in the brain may induce an extrapyramidal disorder known as manganism. Inflammatory processes play a critical role in neurodegenerative diseases. Therapeutically, non-steroidal anti-inflammatory drugs or analogous anti-inflammatory therapies have neuroprotective effects. As a non-steroidal anti-inflammatory drug, p-aminosalicylic acid (PAS) has anti-inflammatory effects, which are mediated by decreased prostaglandins E2 (PGE2) levels. The aim of the current study was to investigate whether PAS-Na treatment prevents Mn-induced behavioral changes and neuroinflammation in vivo. Male Sprague-Dawley rats were intraperitoneally (i.p.) injected with MnCl2·4H2O (15mg/kg) for 12 weeks, followed by 6 weeks PAS-Na treatment. Sub-chronic Mn exposure increased Mn levels in the whole blood, cortex, hippocampus and thalamus, and induced learning and memory deficits, concomitant with astrocytes activation in the cortex, hippocampus and thalamus. Moreover inflammatory cytokine levels in serum and brain of Mn-treated group were increased, including IL-1β, IL-6, TNF-αand PGE2, especially in the hippocampus and thalamus. Furthermore, sub-chronic Mn exposure also increased inflammatory cytokines and COX-2 in transcription levels concomitant with increased MAPK signaling and COX-2 in the same selected brain regions. PAS-Na treatment at the highest doses also decreased Mn levels in the whole blood and selected brain tissues, and reversed the Mn-induced learning and memory deficits. PAS-Na inhibited astrocyte activation as well as the Mn-induced increase in inflammatory cytokine levels, reducing p38, ERK MAPK pathway and COX-2 activity. In contrast PAS-Na had no effects on the JNK MAPK pathway. These data establish the efficacy of PAS-Na not only as a chelating agent to mobilize whole blood Mn, but also as an anti-inflammatory agent.
Collapse
Affiliation(s)
- Shao-Jun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Wen-Xia Qin
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Dong-Jie Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Zong-Xiang Yuan
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Sheng-Nan He
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Yi-Ni Luo
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China.
| | - Dian-Yin Liang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Bing-Yan Xie
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Fang Xu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| |
Collapse
|
37
|
Wang C, Xu B, Ma Z, Liu C, Deng Y, Liu W, Xu ZF. Inhibition of Calpains Protects Mn-Induced Neurotransmitter release disorders in Synaptosomes from Mice: Involvement of SNARE Complex and Synaptic Vesicle Fusion. Sci Rep 2017. [PMID: 28623313 PMCID: PMC5473846 DOI: 10.1038/s41598-017-04017-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Overexposure to manganese (Mn) could disrupt neurotransmitter release via influencing the formation of SNARE complex, but the underlying mechanisms are still unclear. A previous study demonstrated that SNAP-25 is one of substrate of calpains. The current study investigated whether calpains were involved in Mn-induced disorder of SNARE complex. After mice were treated with Mn for 24 days, Mn deposition increased significantly in basal nuclei in Mn-treated and calpeptin pre-treated groups. Behaviorally, less time spent in the center of the area and decreased average velocity significantly in an open field test after 24 days of Mn exposure. With the increase in MnCl2 dosage, intracellular Ca2+ increased significantly, but pretreatment with calpeptin caused a dose-dependent decrease in calpains activity. There were fragments of N-terminal of SNAP-25 protein appearance in Mn-treated groups, but it is decreased with pretreatment of calpeptin. FM1-43-labeled synaptic vesicles also provided evidence that the treatment with Mn resulted in increasing first and then decreasing, which was consistent with Glu release and the 80 kDa protein levels of SNARE complexes. In summary, Mn induced the disorder of neurotransmitter release through influencing the formation of SNARE complex via cleaving SNAP-25 by overactivation of calpains in vivo.
Collapse
Affiliation(s)
- Can Wang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China.
| | - Zhuo Ma
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China
| | - Chang Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China
| | - Zhao-Fa Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China
| |
Collapse
|
38
|
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
|
39
|
Li SJ, Luo YN, Li Y, Chen JW, Mo YH, Yuan ZX, Ou SY, Ou CY, Jiang YM, Deng XF. Sodium para-aminosalicylate protected cultured basal ganglia astrocytes from manganese-induced DNA damages and alteration of amino acid neurotransmitter levels. J Toxicol Sci 2017; 41:573-81. [PMID: 27665767 DOI: 10.2131/jts.41.573] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Sodium para-aminosalicylate (PAS-Na) was first applied successfully in clinical treatment of two manganism patients with good prognosis. However, the mechanism of how PAS-Na protects against Mn-induced neurotoxicity is still elusive. The current study was conducted to explore the effects of PAS-Na on Mn-induced basal ganglia astrocyte injury, and the involvement of amino acid neurotransmitter in vitro. Basal ganglia astrocytes were exposed to 500 μM manganese chloride (MnCl2) for 24 hr, following by 50, 150, or 450 μM PAS-Na treatment for another 24 hr. MnCl2 significantly decreased viability of astrocytes and induced DNA damages via increasing the percentage of tail DNA and Olive tail moment of DNA. Moreover, Mn interrupted amino acid neurotransmitters by decreasing Gln levels and increasing Glu, Gly levels. In contrast, PAS-Na treatment reversed the aforementioned Mn-induced toxic effects on basal ganglia astrocytes. Taken together, our results demonstrated that excessive Mn exposure may induce toxic effects on basal ganglia astrocytes, while PAS-Na could protect basal ganglia astrocytes from Mn-induced neurotoxicity.
Collapse
Affiliation(s)
- Shao-Jun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Li SJ, Ou CY, He SN, Huang XW, Luo HL, Meng HY, Lu GD, Jiang YM, Vieira Peres T, Luo YN, Deng XF. Sodium p-Aminosalicylic Acid Reverses Sub-Chronic Manganese-Induced Impairments of Spatial Learning and Memory Abilities in Rats, but Fails to Restore γ-Aminobutyric Acid Levels. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14040400. [PMID: 28394286 PMCID: PMC5409601 DOI: 10.3390/ijerph14040400] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 02/06/2023]
Abstract
Excessive manganese (Mn) exposure is not only a health risk for occupational workers, but also for the general population. Sodium para-aminosalicylic acid (PAS-Na) has been successfully used in the treatment of manganism, but the involved molecular mechanisms have yet to be determined. The present study aimed to investigate the effects of PAS-Na on sub-chronic Mn exposure-induced impairments of spatial learning and memory, and determine the possible involvements of γ-aminobutyric acid (GABA) metabolism in vivo. Sprague-Dawley male rats received daily intraperitoneal injections MnCl2 (as 6.55 mg/kg Mn body weight, five days per week for 12 weeks), followed by daily subcutaneous injections of 100, 200, or 300 mg/kg PAS-Na for an additional six weeks. Mn exposure significantly impaired spatial learning and memory ability, as noted in the Morris water maze test, and the following PAS-Na treatment successfully restored these adverse effects to levels indistinguishable from controls. Unexpectedly, PAS-Na failed to recover the Mn-induced decrease in the overall GABA levels, although PAS-Na treatment reversed Mn-induced alterations in the enzyme activities directly responsible for the synthesis and degradation of GABA (glutamate decarboxylase and GABA-transaminase, respectively). Moreover, Mn exposure caused an increase of GABA transporter 1 (GAT-1) and decrease of GABA A receptor (GABAA) in transcriptional levels, which could be reverted by the highest dose of 300 mg/kg PAS-Na treatment. In conclusion, the GABA metabolism was interrupted by sub-chronic Mn exposure. However, the PAS-Na treatment mediated protection from sub-chronic Mn exposure-induced neurotoxicity, which may not be dependent on the GABA metabolism.
Collapse
Affiliation(s)
- Shao-Jun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China.
| | - Chao-Yan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China.
- Department of Toxicology, School of Public Health, Guilin Medical University, Guilin 541004, China.
| | - Sheng-Nan He
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China.
| | - Xiao-Wei Huang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China.
| | - Hai-Lan Luo
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China.
| | - Hao-Yang Meng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China.
| | - Guo-Dong Lu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning 530021, China.
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China.
| | - Tanara Vieira Peres
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer, 209, 1300 Morris Park Ave, Bronx, NY 10461, USA.
| | - Yi-Ni Luo
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China.
| | - Xiang-Fa Deng
- Department of Anatomy, School of Pre-Clinical Medicine, Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
41
|
Ou CY, Luo YN, He SN, Deng XF, Luo HL, Yuan ZX, Meng HY, Mo YH, Li SJ, Jiang YM. Sodium P-Aminosalicylic Acid Improved Manganese-Induced Learning and Memory Dysfunction via Restoring the Ultrastructural Alterations and γ-Aminobutyric Acid Metabolism Imbalance in the Basal Ganglia. Biol Trace Elem Res 2017; 176:143-153. [PMID: 27491492 DOI: 10.1007/s12011-016-0802-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/05/2016] [Indexed: 11/29/2022]
Abstract
Excessive intake of manganese (Mn) may cause neurotoxicity. Sodium para-aminosalicylic acid (PAS-Na) has been used successfully in the treatment of Mn-induced neurotoxicity. The γ-aminobutyric acid (GABA) is related with learning and memory abilities. However, the mechanism of PAS-Na on improving Mn-induced behavioral deficits is unclear. The current study was aimed to investigate the effects of PAS-Na on Mn-induced behavioral deficits and the involvement of ultrastructural alterations and γ-aminobutyric acid (GABA) metabolism in the basal ganglia of rats. Sprague-Dawley rats received daily intraperitoneally injections of 15 mg/kg MnCl2.4H2O, 5d/week for 4 weeks, followed by a daily back subcutaneously (sc.) dose of PAS-Na (100 and 200 mg/kg), 5 days/week for another 3 or 6 weeks. Mn exposure for 4 weeks and then ceased Mn exposure for 3 or 6 weeks impaired spatial learning and memory abilities, and these effects were long-lasting. Moreover, Mn exposure caused ultrastructural alterations in the basal ganglia expressed as swollen neuronal with increasing the electron density in the protrusions structure and fuzzed the interval of neuropil, together with swollen, focal hyperplasia, and hypertrophy of astrocytes. Additionally, the results also indicated that Mn exposure increased Glu/GABA values as by feedback loops controlling GAT-1, GABAA mRNA and GABAA protein expression through decreasing GABA transporter 1(GAT-1) and GABA A receptor (GABAA) mRNA expression, and increasing GABAA protein expression in the basal ganglia. But Mn exposure had no effects on GAT-1 protein expression. PAS-Na treatment for 3 or 6 weeks effectively restored the above-mentioned adverse effects induced by Mn. In conclusion, these findings suggest the involvement of GABA metabolism and ultrastructural alterations of basal ganglia in PAS-Na's protective effects on the spatial learning and memory abilities.
Collapse
Affiliation(s)
- Chao-Yan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd, Nanning, Guangxi, 530021, China
- Department of Toxicology, School of Public Health, Guilin Medical University, Guilin, 541004, China
| | - Yi-Ni Luo
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd, Nanning, Guangxi, 530021, China
| | - Sheng-Nan He
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd, Nanning, Guangxi, 530021, China
| | - Xiang-Fa Deng
- Department of Anatomy, School of Pre-clinical Medicine, Guangxi Medical University, Nanning, 530021, China
| | - Hai-Lan Luo
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd, Nanning, Guangxi, 530021, China
| | - Zong-Xiang Yuan
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd, Nanning, Guangxi, 530021, China
| | - Hao-Yang Meng
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd, Nanning, Guangxi, 530021, China
| | - Yu-Huan Mo
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd, Nanning, Guangxi, 530021, China
| | - Shao-Jun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd, Nanning, Guangxi, 530021, China.
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd, Nanning, Guangxi, 530021, China.
| |
Collapse
|
42
|
van Thriel C, Quetscher C, Pesch B, Lotz A, Lehnert M, Casjens S, Weiss T, Van Gelder R, Plitzke K, Brüning T, Beste C. Are multitasking abilities impaired in welders exposed to manganese? Translating cognitive neuroscience to neurotoxicology. Arch Toxicol 2017; 91:2865-2877. [DOI: 10.1007/s00204-017-1932-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/12/2017] [Indexed: 01/03/2023]
|
43
|
Sawikr Y, Yarla NS, Peluso I, Kamal MA, Aliev G, Bishayee A. Neuroinflammation in Alzheimer's Disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2017; 108:33-57. [DOI: 10.1016/bs.apcsb.2017.02.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
44
|
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
|
45
|
Magnetic Resonance Spectroscopy for Evaluating Portal-Systemic Encephalopathy in Patients with Chronic Hepatic Schistosomiasis Japonicum. PLoS Negl Trop Dis 2016; 10:e0005232. [PMID: 27977668 PMCID: PMC5199111 DOI: 10.1371/journal.pntd.0005232] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/29/2016] [Accepted: 12/05/2016] [Indexed: 12/02/2022] Open
Abstract
Portal-systemic encephalopathy (PSE) is classified as type B hepatic encephalopathy. Portal-systemic shunting rather than liver dysfunction is the main cause of PSE in chronic hepatic schistosomiasis japonicum (HSJ) patients. Owing to lack of detectable evidence of intrinsic liver disease, chronic HSJ patients with PSE are frequently clinically undetected or misdiagnosed, especially chronic HSJ patients with covert PSE (subclinical encephalopathy). In this study, we investigated whether magnetic resonance spectroscopy (MRS) could be a useful tool for diagnosing PSE in chronic HSJ patients. Magnetic resonance (MR) T1-weighted imaging, diffusion-weighted imaging, and MRS were performed in 41 chronic HSJ patients with suspected PSE and in 21 age-matched controls. The T1 signal intensity index (T1SI) and apparent diffusion coefficient (ADC) value were obtained in the Globus pallidus. Liver function was also investigated via serum ammonia and liver function tests. Higher T1SI and ADC values, increased lactate and glutamine levels, and decreased myo-inositol were found in the bilateral Globus pallidus in chronic HSJ patients with PSE. No significantly abnormal serum ammonia or liver function tests were observed in chronic HSJ patients with PSE. On the basis of these findings, we propose a diagnostic procedure for PSE in chronic HSJ patients. This study reveals that MRS can be useful for diagnosing PSE in chronic HSJ patients. Portal-systemic encephalopathy (PSE) is classified as type B hepatic encephalopathy. Portal-systemic shunting rather than liver dysfunction is the main cause of PSE in chronic hepatic schistosomiasis japonicum (HSJ) patients. Chronic HSJ patients with PSE are frequently misdiagnosed as suffering from dementia, depression, and other psychiatric disorders. However, there are few studies of this type of encephalopathy. In this study, magnetic resonance spectroscopy (MRS) was used as a noninvasive tool to detect abnormalities in brain metabolism in chronic HSJ patient with PSE. The results demonstrate that MRS would be useful for the diagnosis of PSE in chronic HSJ patients. Based on the results, a diagnostic procedure for PSE has also been proposed.
Collapse
|
46
|
Porto LCS, da Silva J, Sousa K, Ambrozio ML, de Almeida A, Dos Santos CEI, Dias JF, Allgayer MC, Dos Santos MS, Pereira P, Ferraz ABF, Picada JN. Evaluation of Toxicological Effects of an Aqueous Extract of Shells from the Pecan Nut Carya illinoinensis (Wangenh.) K. Koch and the Possible Association with Its Inorganic Constituents and Major Phenolic Compounds. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2016; 2016:4647830. [PMID: 27525021 PMCID: PMC4971311 DOI: 10.1155/2016/4647830] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/10/2016] [Accepted: 06/13/2016] [Indexed: 12/01/2022]
Abstract
Background. Industrial processing of the pecan nut Carya illinoinensis K. Koch generated a large amount of shells, which have been used to prepare nutritional supplements and medicinal products; however, the safe use of shells requires assessment. This study evaluated the toxic, genotoxic, and mutagenic effects of pecan shell aqueous extract (PSAE) and the possible contribution of phenolic compounds, ellagic and gallic acids, and inorganic elements present in PSAE to induce toxicity. Results. Levels of inorganic elements like K, P, Cl, and Rb quantified using the Particle-Induced X-Ray Emission method were higher in PSAE than in pecan shells, while Mg and Mn levels were higher in shells. Mice showed neurobehavioral toxicity when given high PSAE doses (200-2,000 mg kg(-1)). The LD50 was 1,166.3 mg kg(-1). However, PSAE (50-200 mg·kg(-1)) and the phenolic compounds (10-100 mg·kg(-1)) did not induce DNA damage or mutagenicity evaluated using the comet assay and micronucleus test. Treatment with ellagic acid (10-100 mg·kg(-1)) decreased triglyceride and glucose levels, while treatments with PSAE and gallic acid had no effect. Conclusion. Pecan shell toxicity might be associated with high concentrations of inorganic elements such as Mn, Al, Cu, and Fe acting on the central nervous system, besides phytochemical components, suggesting that the definition of the safe dose should take into account the consumption of micronutrients.
Collapse
Affiliation(s)
- Luiz Carlos S Porto
- Laboratory of Toxicological Genetics, Lutheran University of Brazil (ULBRA), Farroupilha Avenue 8001, 92425-900 Canoas, RS, Brazil; University of the Campaign Region (URCAMP), Tancredo Neves Avenue 210, 97670000 São Borja, RS, Brazil
| | - Juliana da Silva
- Laboratory of Toxicological Genetics, Lutheran University of Brazil (ULBRA), Farroupilha Avenue 8001, 92425-900 Canoas, RS, Brazil
| | - Karen Sousa
- Laboratory of Toxicological Genetics, Lutheran University of Brazil (ULBRA), Farroupilha Avenue 8001, 92425-900 Canoas, RS, Brazil
| | - Mariana L Ambrozio
- Laboratory of Toxicological Genetics, Lutheran University of Brazil (ULBRA), Farroupilha Avenue 8001, 92425-900 Canoas, RS, Brazil
| | - Aline de Almeida
- Laboratory of Toxicological Genetics, Lutheran University of Brazil (ULBRA), Farroupilha Avenue 8001, 92425-900 Canoas, RS, Brazil
| | - Carla Eliete I Dos Santos
- Physics, Statistics, and Mathematics Institute, Federal University of Rio Grande (FURG), Barão do Caí 125, 95500000 Santo Antônio da Patrulha, RS, Brazil
| | - Johnny F Dias
- Ion Implantation Laboratory, Physics Institute, Federal University of Rio Grande do Sul (UFRGS), Bento Gonçalves Avenue 9500, 91501970 Porto Alegre, RS, Brazil
| | - Mariangela C Allgayer
- Laboratory of Clinical Pathology, Veterinary Hospital, Lutheran University of Brazil (ULBRA), Farroupilha Avenue 8001, 92425-900 Canoas, RS, Brazil
| | - Marcela S Dos Santos
- Laboratory of Pharmacognosis and Phytochemistry, Lutheran University of Brazil (ULBRA), Farroupilha Avenue 8001, 92425-900 Canoas, RS, Brazil
| | - Patrícia Pereira
- Pharmacology Department, Institute of Basic Sciences of Health, Federal University of Rio Grande do Sul (UFRGS), Sarmento Leite Street 500/305, 90050-170 Porto Alegre, RS, Brazil
| | - Alexandre B F Ferraz
- Laboratory of Pharmacognosis and Phytochemistry, Lutheran University of Brazil (ULBRA), Farroupilha Avenue 8001, 92425-900 Canoas, RS, Brazil
| | - Jaqueline N Picada
- Laboratory of Toxicological Genetics, Lutheran University of Brazil (ULBRA), Farroupilha Avenue 8001, 92425-900 Canoas, RS, Brazil
| |
Collapse
|
47
|
Szpetnar M, Luchowska-Kocot D, Boguszewska-Czubara A, Kurzepa J. The Influence of Manganese and Glutamine Intake on Antioxidants and Neurotransmitter Amino Acids Levels in Rats' Brain. Neurochem Res 2016; 41:2129-39. [PMID: 27161372 PMCID: PMC4947112 DOI: 10.1007/s11064-016-1928-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/25/2016] [Accepted: 04/18/2016] [Indexed: 01/11/2023]
Abstract
Depending on the concentration, Mn can exert protective or toxic effect. Potential mechanism for manganese neurotoxicity is manganese-induced oxidative stress. Glutamine supplementation could reduce manganese-induced neurotoxicity and is able to influence the neurotransmission processes. The aim of this study was to investigate whether the long term administration of manganese (alone or in combination with glutamine) in dose and time dependent manner could affect the selected parameters of oxidative-antioxidative status (superoxide dismutase and glutathione peroxidase activities, concentrations of vitamin C and malonic dialdehyde) and concentrations of excitatory (Asp, Glu) and inhibitory amino acids (GABA, Gly) in the brain of rats. The experiments were carried out on 2-months-old albino male rats randomly divided into 6 group: Mn300 and Mn500—received solution of MnCl2 to drink (dose 300 and 500 mg/L, respectively), Gln group—solution of glutamine (4 g/L), Mn300-Gln and Mn500-Gln groups—solution of Mn at 300 and 500 mg/L and Gln at 4 g/L dose. The control group (C) received deionized water. Half of the animals were euthanized after three and the other half—after 6 weeks of experiment. The exposure of rats to Mn in drinking water contributes to diminishing of the antioxidant enzymes activity and the increase in level of lipid peroxidation. Glutamine in the diet admittedly increases SOD and GPx activity, but it is unable to restore the intracellular redox balance. The most significant differences in the examined amino acids levels in comparison to both control and Gln group were observed in the group of rats receiving Mn at 500 mg/L dose alone or with Gln. It seems that Gln is amino acid which could improve antioxidant status and affect the concentrations of the neurotransmitters.
Collapse
Affiliation(s)
- Maria Szpetnar
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093, Lublin, Poland
| | - Dorota Luchowska-Kocot
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093, Lublin, Poland.
| | - Anna Boguszewska-Czubara
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093, Lublin, Poland
| | - Jacek Kurzepa
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093, Lublin, Poland
| |
Collapse
|
48
|
Particulate Matter Facilitates C6 Glioma Cells Activation and the Release of Inflammatory Factors Through MAPK and JAK2/STAT3 Pathways. Neurochem Res 2016; 41:1969-81. [DOI: 10.1007/s11064-016-1908-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 03/09/2016] [Accepted: 04/05/2016] [Indexed: 11/27/2022]
|
49
|
Expression Profiles of Long Noncoding RNAs and Messenger RNAs in Mn-Exposed Hippocampal Neurons of Sprague-Dawley Rats Ascertained by Microarray: Implications for Mn-Induced Neurotoxicity. PLoS One 2016; 11:e0145856. [PMID: 26745496 PMCID: PMC4706437 DOI: 10.1371/journal.pone.0145856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 12/09/2015] [Indexed: 12/12/2022] Open
Abstract
Manganese (Mn) is an essential trace element, while excessive expose may induce neurotoxicity. Recently, lncRNAs have been extensively studied and it has been confirmed that lncRNAs participate in neural functions and aberrantly expressed lncRNAs are involved in neurological diseases. However, the pathological effects of lncRNAs on Mn-induced neurotoxicity remain unclear. In this study, the expression profiles of lncRNAs and messenger RNAs (mRNAs) were identified in Mn-treated hippocampal neurons and control neurons via microarray. Bioinformatic methods and intersection analysis were also employed. Results indicated that 566, 1161, and 1474 lncRNAs meanwhile 1848, 3228, and 4022 mRNAs were aberrantly expressed in low, intermediate, and high Mn-exposed groups compared with the control group, respectively. Go analysis determined that differentially expressed mRNAs were targeted to biological processes, cellular components, and molecular functions. Pathway analysis indicated that these mRNAs were enriched in insulin secretion, cell cycle, and DNA replication. Intersection analysis denominated that 135 lncRNAs and 373 mRNAs were consistently up-regulated while 150 lncRNAs and 560 mRNAs were consistently down-regulated. Meanwhile, lncRNA BC079195 was significantly up-regulated while lncRNAs uc.229- and BC089928 were significantly down-regulated in three comparison groups. The relative expression levels of 3 lncRNAs and 4 mRNAs were validated through qRT-PCR. To the best of our knowledge, this study is the first to identify the expression patterns of lncRNAs and mRNAs in hippocampal neurons of Sprague-Dawley rats. The results may provide evidence on underlying mechanisms of Mn-induced neurotoxicity, and aberrantly expressed lncRNAs/mRNAs may be useful in further investigations to detect early symptoms of Mn-induced neuropsychiatric disorders in the central nervous system.
Collapse
|
50
|
The Glutamine Transporters and Their Role in the Glutamate/GABA-Glutamine Cycle. ADVANCES IN NEUROBIOLOGY 2016; 13:223-257. [PMID: 27885631 DOI: 10.1007/978-3-319-45096-4_8] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Glutamine is a key amino acid in the CNS, playing an important role in the glutamate/GABA-glutamine cycle (GGC). In the GGC, glutamine is transferred from astrocytes to neurons, where it will replenish the inhibitory and excitatory neurotransmitter pools. Different transporters participate in this neural communication, i.e., the transporters responsible for glutamine efflux from astrocytes and influx into the neurons, such as the members of the SNAT, LAT, y+LAT, and ASC families of transporters. The SNAT family consists of the transporter isoforms SNAT3 and SNAT5 that are related to efflux from the astrocytic compartment, and SNAT1 and SNAT2 that are associated with glutamine uptake into the neuronal compartment. The isoforms SNAT7 and SNAT8 do not have their role completely understood, but they likely also participate in the GGC. The isoforms LAT2 and y+LAT2 facilitate the exchange of neutral amino acids and cationic amino acids (y+LAT2 isoform) and have been associated with glutamine efflux from astrocytes. ASCT2 is a Na+-dependent antiporter, the participation of which in the GGC also remains to be better characterized. All these isoforms are tightly regulated by transcriptional and translational mechanisms, which are induced by several determinants such as amino acid deprivation, hormones, pH, and the activity of different signaling pathways. Dysfunctional glutamine transporter activity has been associated with the pathophysiological mechanisms of certain neurologic diseases, such as Hepatic Encephalopathy and Manganism. However, there might also be other neuropathological conditions associated with an altered GGC, in which glutamine transporters are dysfunctional. Hence, it appears to be of critical importance that the physiological and pathological aspects of glutamine transporters are thoroughly investigated.
Collapse
|