1
|
Martins AC, Oliveira-Paula GH, Tinkov AA, Skalny AV, Tizabi Y, Bowman AB, Aschner M. Role of manganese in brain health and disease: Focus on oxidative stress. Free Radic Biol Med 2025; 232:306-318. [PMID: 40086492 PMCID: PMC11985276 DOI: 10.1016/j.freeradbiomed.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Manganese (Mn) is an essential trace element crucial for various physiological processes, but excessive exposure can lead to significant health concerns, particularly neurotoxicity. This review synthesizes current knowledge on Mn-induced oxidative stress and its role in cellular dysfunction and disease. We discuss how Mn promotes toxicity through multiple mechanisms, primarily through reactive oxygen species (ROS) generation, which leads to oxidative stress and disruption of cellular processes. The review examines key pathways affected by Mn toxicity, including mitochondrial dysfunction, endoplasmic reticulum stress, inflammasome activation, and epigenetic modifications. Recent studies have identified promising therapeutic compounds, including both synthetic and natural substances such as probucol, metformin, curcumin, resveratrol, and daidzein, which demonstrate protective effects through various mechanisms, including antioxidant enhancement, mitochondrial function preservation, and epigenetic pathway modulation. Understanding these mechanisms provides new insights into potential therapeutic strategies for Mn-induced disorders. This review also highlights future research directions, emphasizing the need for developing targeted therapies and investigating combination approaches to address multiple aspects of Mn toxicity simultaneously.
Collapse
Affiliation(s)
- Airton C Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Gustavo H Oliveira-Paula
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Alexey A Tinkov
- Institute of Bioelementology, Orenburg State University, Orenburg, 460000, Russia; IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003, Russia
| | - Anatoly V Skalny
- Institute of Bioelementology, Orenburg State University, Orenburg, 460000, Russia; IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003, Russia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington DC, 20059, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
2
|
Okeowo OM, Anadu VE, Ijomone OK, Aschner M, Ijomone OM. Combined Restraint Stress and Metal Exposure Paradigms in Rats: Unravelling Behavioural and Neurochemical Perturbations. Mol Neurobiol 2025; 62:4355-4376. [PMID: 39443350 DOI: 10.1007/s12035-024-04570-1] [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: 06/09/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Accumulation of heavy metals (Mn and Ni) and prolonged exposure to stress are associated with adverse health outcomes. Various studies have shown the impacts of stress and metal exposures on brain function. However, no study has examined the effects of co-exposure to stress, Mn, and Ni on the brain. This study addresses this gap by evaluating oxidative and glial responses, apoptotic activity, as well as cognitive processes in a rat model. Adult Wistar rats were exposed to vehicle (control), restraint stress, 25 mg/kg of manganese (Mn) or nickel (Ni), or combined restraint stress plus Mn or Ni. Following treatment, rats were subjected to several behavioural paradigms to assess cognitive function. Enzyme activity, as well as ATPase levels, were evaluated. Thereafter, an immunohistochemical procedure was utilised to evaluate neurochemical markers of glial function, myelination, oxidative stress, and apoptosis in the hippocampus, prefrontal cortex (PFC), and striatum. Results showed that stress and metal exposure increased oxidative stress markers and reduced antioxidant levels. Further, combined stress and metal exposure reduced various forms of learning and memory ability in rats. In addition, there were alterations in Iba1 activity and Nrf2 levels, reduced Olig2 and myelin basic protein (MBP) levels, and increased caspase-3 expression. These neurotoxic outcomes were mostly exacerbated by co-exposure to stress and metals. Overall, our findings establish that stress and metal exposures impaired cognitive performance, induced oxidative stress and apoptosis, and led to demyelination effects which were worsened by combined stress and metal exposure.
Collapse
Affiliation(s)
- Oritoke M Okeowo
- Department of Physiology, School of Basic Medical Sciences, Federal University of Technology, Akure, Nigeria
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Nigeria
| | - Victor E Anadu
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Nigeria
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Medical Sciences, Ondo, Nigeria
| | - Olayemi K Ijomone
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Nigeria
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Medical Sciences, Ondo, Nigeria
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Omamuyovwi M Ijomone
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Nigeria.
- Department of Human Anatomy, School of Basic Medical Sciences, Federal University of Technology, Akure, Nigeria.
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Medical Sciences, Ondo, Nigeria.
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
3
|
Aschner M, Martins AC, Oliveira-Paula GH, Skalny AV, Zaitseva IP, Bowman AB, Kirichuk AA, Santamaria A, Tizabi Y, Tinkov AA. Manganese in autism spectrum disorder and attention deficit hyperactivity disorder: The state of the art. Curr Res Toxicol 2024; 6:100170. [PMID: 38737010 PMCID: PMC11088232 DOI: 10.1016/j.crtox.2024.100170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/27/2024] [Accepted: 04/23/2024] [Indexed: 05/14/2024] Open
Abstract
The objective of the present narrative review was to synthesize existing clinical and epidemiological findings linking manganese (Mn) exposure biomarkers to autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD), and to discuss key pathophysiological mechanisms of neurodevelopmental disorders that may be affected by this metal. Existing epidemiological data demonstrated both direct and inverse association between Mn body burden and ASD, or lack of any relationship. In contrast, the majority of studies revealed significantly higher Mn levels in subjects with ADHD, as well as direct relationship between Mn body burden with hyperactivity and inattention scores in children, although several studies reported contradictory results. Existing laboratory studies demonstrated that impaired attention and hyperactivity in animals following Mn exposure was associated with dopaminergic dysfunction and neuroinflammation. Despite lack of direct evidence on Mn-induced neurobiological alterations in patients with ASD and ADHD, a plethora of studies demonstrated that neurotoxic effects of Mn overexposure may interfere with key mechanisms of pathogenesis inherent to these neurodevelopmental disorders. Specifically, Mn overload was shown to impair not only dopaminergic neurotransmission, but also affect metabolism of glutamine/glutamate, GABA, serotonin, noradrenaline, thus affecting neuronal signaling. In turn, neurotoxic effects of Mn may be associated with its ability to induce oxidative stress, apoptosis, and neuroinflammation, and/or impair neurogenesis. Nonetheless, additional detailed studies are required to evaluate the association between environmental Mn exposure and/or Mn body burden and neurodevelopmental disorders at a wide range of concentrations to estimate the potential dose-dependent effects, as well as environmental and genetic factors affecting this association.
Collapse
Affiliation(s)
- Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | | | - Anatoly V. Skalny
- Department of Medical Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
| | - Irina P. Zaitseva
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl 150003, Russia
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, USA
| | - Anatoly A. Kirichuk
- Department of Medical Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - Abel Santamaria
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Cuidado de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City 04960, Mexico
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059, USA
| | - Alexey A. Tinkov
- Department of Medical Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl 150003, Russia
| |
Collapse
|
4
|
Overexpressed miRNA-nov-1 promotes manganese-induced apoptosis in N27 cells by regulating Dhrs3 to activate mTOR signaling pathway. Toxicology 2023; 489:153472. [PMID: 36868551 DOI: 10.1016/j.tox.2023.153472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/20/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
Environmental and occupational chronic manganese exposure can cause neurotoxicity and apoptosis. Moreover, microRNAs (miRNAs) are extensively involved in the process of neuronal apoptosis. Therefore, it is crucial to study the mechanism of miRNA in manganese-induced neuronal apoptosis and to find potential targets. In the present study, we found that the expression of miRNA-nov-1 was increased after N27 cells were exposed to MnCl2. Then, seven different cell groups were constructed by lentiviral infection of cells, and the overexpression of miRNA-nov-1 promoted the apoptosis process of N27 cells. Further studies showed a negative regulatory relationship between miRNA-nov-1 and dehydrogenase/reductase 3 (Dhrs3). The up-regulation of miRNA-nov-1 reduced the protein level of Dhrs3 in N27 cells exposed to manganese, increased the expression of a caspase-3 protein, activated the rapamycin (mTOR) signaling pathway, and increased cell apoptosis. Furthermore, we found that the expression of the Caspase-3 protein was decreased after the low expression of miRNA-nov-1, the mTOR signaling pathway was inhibited, and reduced cell apoptosis. However, these effects were reversed by the knockdown of Dhrs3. Taken together, these results suggested that overexpression of miRNA-nov-1 can promote manganese-induced apoptosis in N27 cells by activating the mTOR signaling pathway and negatively regulating Dhrs3.
Collapse
|
5
|
Liu ZF, Liu K, Liu ZQ, Cong L, Lei MY, Li J, Ma Z, Deng Y, Liu W, Xu B. Melatonin attenuates manganese-induced mitochondrial fragmentation by suppressing the Mst1/JNK signaling pathway in primary mouse neurons. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 844:157134. [PMID: 35792268 DOI: 10.1016/j.scitotenv.2022.157134] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/18/2022] [Accepted: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Manganese (Mn) toxicity is mainly caused by excessive Mn content in drinking water and occupational exposure. Moreover, overexposure to Mn can impair mental, cognitive, memory, and motor capacities. Although melatonin (Mel) can protect against Mn-induced neuronal damage and mitochondrial fragmentation, the underlying mechanism remains elusive. Here, we examined the related molecular mechanisms underlying Mel attenuating Mn-induced mitochondrial fragmentation through the mammalian sterile 20-like kinase-1 (Mst1)/JNK signaling path. To test the role of Mst1 in mitochondrial fragmentation, we treated mouse primary neurons overexpressing Mst1 with Mel and Mn stimulation. In normal neurons, 10 μM Mel reduced the effects of Mn (200 μM) on Mst1 expression at the mRNA and protein levels and on phosphorylation of JNK and Drp1, Drp1 mitochondrial translocation, and mitochondrial fragmentation. Conversely, overexpression of Mst1 hindered the protective effect of Mel (10 μM) against Mn-induced mitochondrial fragmentation. Anisomycin (ANI), an activator of JNK signaling, was similarly found to inhibit the protective effect of Mel on mitochondria, while Mst1 levels were not significantly changed. Thus, our results demonstrated that 10 μM Mel negatively regulated the Mst1-JNK pathway, thereby reducing excessive mitochondrial fission, maintaining the mitochondrial network, and alleviating Mn-induced mitochondrial dysfunction.
Collapse
Affiliation(s)
- Zhuo-Fan Liu
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Kuan Liu
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Zhi-Qi Liu
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Lin Cong
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Meng-Yu Lei
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Jing Li
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Zhuo Ma
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, China.
| |
Collapse
|
6
|
Tan Y, Cheng H, Su C, Chen P, Yang X. PI3K/Akt Signaling Pathway Ameliorates Oxidative Stress-Induced Apoptosis upon Manganese Exposure in PC12 Cells. Biol Trace Elem Res 2022; 200:749-760. [PMID: 33772736 DOI: 10.1007/s12011-021-02687-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/21/2021] [Indexed: 12/16/2022]
Abstract
Manganese (Mn)-induced neurotoxicity has aroused public concerns for many years, but its precise mechanism is still poorly understood. Herein, we report the impacts of the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) signaling pathway in mediating neurological effects induced by manganese sulfate (MnSO4) exposure in PC12 cells. In this study, cells were treated with MnSO4 for 24 h in the absence or presence of LY294002 (a special inhibitor of PI3K). We investigated cell viability and apoptosis signals, as well as levels of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT), and malondialdehyde (MDA). The mRNA levels of B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax), and Caspase-3 were also quantified through real-time quantitative PCR (RT-qPCR); protein levels of serine/threonine protein kinase (Akt) and forkhead box O3A (Foxo3a) were determined by western blot. Increasing of MnSO4 doses led to decreased SOD, GSH-Px, and CAT activities, while the level of MDA was upregulated. Moreover, cell apoptosis was significantly increased, as the mRNA of Bcl-2 and Caspase-3 was significantly decreased, while Bax mRNA was increased. Phosphorylated Akt (p-Akt) and Foxo3a (p-Foxo3a) were upregulated in a dose-dependent manner. In addition, LY294002 pretreatment reduced the activity of SOD, GSH-Px, and CAT but elevated MDA levels. Meanwhile, LY294002 pretreatment also increased cell apoptosis given the upregulated Bax and Caspase-3 mRNAs and decreased Bcl-2 mRNA. In summary, the PI3K/Akt signaling pathway can be activated by MnSO4 exposure and mediate MnSO4-induced neurotoxicity.
Collapse
Affiliation(s)
- Yanli Tan
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi, China
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Hong Cheng
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi, China
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Cheng Su
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi, China
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Xiaobo Yang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi, China.
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, China.
- Department of Public Health, School of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi, China.
| |
Collapse
|
7
|
Morcillo P, Cordero H, Ijomone OM, Ayodele A, Bornhorst J, Gunther L, Macaluso FP, Bowman AB, Aschner M. Defective Mitochondrial Dynamics Underlie Manganese-Induced Neurotoxicity. Mol Neurobiol 2021; 58:3270-3289. [PMID: 33666854 PMCID: PMC9009155 DOI: 10.1007/s12035-021-02341-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/23/2021] [Indexed: 12/17/2022]
Abstract
Perturbations in mitochondrial dynamics have been observed in most neurodegenerative diseases. Here, we focus on manganese (Mn)-induced Parkinsonism-like neurodegeneration, a disorder associated with the preferential of Mn in the basal ganglia where the mitochondria are considered an early target. Despite the extensive characterization of the clinical presentation of manganism, the mechanism by which Mn mediated mitochondrial toxicity is unclear. In this study we hypothesized whether Mn exposure alters mitochondrial activity, including axonal transport of mitochondria and mitochondrial dynamics, morphology, and network. Using primary neuron cultures exposed to 100 μM Mn (which is considered the threshold of Mn toxicity in vitro) and intraperitoneal injections of MnCl2 (25mg/kg) in rat, we observed that Mn increased mitochondrial fission mediated by phosphorylation of dynamin-related protein-1 at serine 616 (p-s616-DRP1) and decreased mitochondrial fusion proteins (MFN1 and MFN2) leading to mitochondrial fragmentation, defects in mitochondrial respiratory capacity, and mitochondrial ultrastructural damage in vivo and in vitro. Furthermore, Mn exposure impaired mitochondrial trafficking by decreasing dynactin (DCTN1) and kinesin-1 (KIF5B) motor proteins and increasing destabilization of the cytoskeleton at protein and gene levels. In addition, mitochondrial communication may also be altered by Mn exposure, increasing the length of nanotunnels to reach out distal mitochondria. These findings revealed an unrecognized role of Mn in dysregulation of mitochondrial dynamics providing a potential explanation of early hallmarks of the disorder, as well as a possible common pathway with neurological disorders arising upon chronic Mn exposure.
Collapse
Affiliation(s)
- Patricia Morcillo
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Hector Cordero
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Omamuyovwi M Ijomone
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- The Neuro-Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Akinyemi Ayodele
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Julia Bornhorst
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Leslie Gunther
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Frank P Macaluso
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
8
|
Tinkov AA, Paoliello MMB, Mazilina AN, Skalny AV, Martins AC, Voskresenskaya ON, Aaseth J, Santamaria A, Notova SV, Tsatsakis A, Lee E, Bowman AB, Aschner M. Molecular Targets of Manganese-Induced Neurotoxicity: A Five-Year Update. Int J Mol Sci 2021; 22:4646. [PMID: 33925013 PMCID: PMC8124173 DOI: 10.3390/ijms22094646] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/22/2021] [Accepted: 04/25/2021] [Indexed: 12/16/2022] Open
Abstract
Understanding of the immediate mechanisms of Mn-induced neurotoxicity is rapidly evolving. We seek to provide a summary of recent findings in the field, with an emphasis to clarify existing gaps and future research directions. We provide, here, a brief review of pertinent discoveries related to Mn-induced neurotoxicity research from the last five years. Significant progress was achieved in understanding the role of Mn transporters, such as SLC39A14, SLC39A8, and SLC30A10, in the regulation of systemic and brain manganese handling. Genetic analysis identified multiple metabolic pathways that could be considered as Mn neurotoxicity targets, including oxidative stress, endoplasmic reticulum stress, apoptosis, neuroinflammation, cell signaling pathways, and interference with neurotransmitter metabolism, to name a few. Recent findings have also demonstrated the impact of Mn exposure on transcriptional regulation of these pathways. There is a significant role of autophagy as a protective mechanism against cytotoxic Mn neurotoxicity, yet also a role for Mn to induce autophagic flux itself and autophagic dysfunction under conditions of decreased Mn bioavailability. This ambivalent role may be at the crossroad of mitochondrial dysfunction, endoplasmic reticulum stress, and apoptosis. Yet very recent evidence suggests Mn can have toxic impacts below the no observed adverse effect of Mn-induced mitochondrial dysfunction. The impact of Mn exposure on supramolecular complexes SNARE and NLRP3 inflammasome greatly contributes to Mn-induced synaptic dysfunction and neuroinflammation, respectively. The aforementioned effects might be at least partially mediated by the impact of Mn on α-synuclein accumulation. In addition to Mn-induced synaptic dysfunction, impaired neurotransmission is shown to be mediated by the effects of Mn on neurotransmitter systems and their complex interplay. Although multiple novel mechanisms have been highlighted, additional studies are required to identify the critical targets of Mn-induced neurotoxicity.
Collapse
Affiliation(s)
- Alexey A. Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia;
- Laboratory of Molecular Dietetics, Department of Neurological Diseases and Neurosurgery, Department of Analytical and Forensic Toxicology, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (O.N.V.); (J.A.); (A.T.)
| | - Monica M. B. Paoliello
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.M.B.P.); (A.C.M.)
- Graduate Program in Public Health, Center of Health Sciences, State University of Londrina, Londrina, PR 86038-350, Brazil
| | - Aksana N. Mazilina
- Department of Medical Elementology, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia;
| | - Anatoly V. Skalny
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia;
- Laboratory of Medical Elementology, KG Razumovsky Moscow State University of Technologies and Management, 109004 Moscow, Russia
| | - Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.M.B.P.); (A.C.M.)
| | - Olga N. Voskresenskaya
- Laboratory of Molecular Dietetics, Department of Neurological Diseases and Neurosurgery, Department of Analytical and Forensic Toxicology, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (O.N.V.); (J.A.); (A.T.)
| | - Jan Aaseth
- Laboratory of Molecular Dietetics, Department of Neurological Diseases and Neurosurgery, Department of Analytical and Forensic Toxicology, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (O.N.V.); (J.A.); (A.T.)
- Research Department, Innlandet Hospital Trust, P.O. Box 104, 2381 Brumunddal, Norway
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, SSA, Mexico City 14269, Mexico;
| | - Svetlana V. Notova
- Institute of Bioelementology, Orenburg State University, 460018 Orenburg, Russia;
- Federal Research Centre of Biological Systems and Agro-technologies of the Russian Academy of Sciences, 460000 Orenburg, Russia
| | - Aristides Tsatsakis
- Laboratory of Molecular Dietetics, Department of Neurological Diseases and Neurosurgery, Department of Analytical and Forensic Toxicology, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (O.N.V.); (J.A.); (A.T.)
- Laboratory of Toxicology, Medical School, University of Crete, Voutes, 700 13 Heraklion, Greece
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47906, USA;
| | - Michael Aschner
- Laboratory of Molecular Dietetics, Department of Neurological Diseases and Neurosurgery, Department of Analytical and Forensic Toxicology, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (O.N.V.); (J.A.); (A.T.)
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.M.B.P.); (A.C.M.)
| |
Collapse
|
9
|
Pajarillo E, Nyarko-Danquah I, Adinew G, Rizor A, Aschner M, Lee E. Neurotoxicity mechanisms of manganese in the central nervous system. ADVANCES IN NEUROTOXICOLOGY 2021; 5:215-238. [PMID: 34263091 DOI: 10.1016/bs.ant.2020.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Ivan Nyarko-Danquah
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Getinet Adinew
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Asha Rizor
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| |
Collapse
|
10
|
Sun Q, Kang RR, Chen KG, Liu K, Ma Z, Liu C, Deng Y, Liu W, Xu B. Sirtuin 3 is required for the protective effect of Resveratrol on Manganese-induced disruption of mitochondrial biogenesis in primary cultured neurons. J Neurochem 2020; 156:121-135. [PMID: 32426865 DOI: 10.1111/jnc.15095] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/14/2022]
Abstract
Chronic manganese (Mn) exposure can disturb mitochondrial homeostasis leading to mitochondrial dysfunction, which is involved in Mn-induced neurodegenerative diseases. Resveratrol (RSV), as a promoter of mitochondrial biogenesis, plays a significant role against mitochondrial dysfunction. However, whether RSV can relieve Mn-induced neuronal injury and mitochondrial dysfunction remains unknown. Sirtuin 3 (SIRT3), a main mitochondrial sirtuin, is an important regulator of mitochondria to maintain mitochondrial homeostasis. Therefore, this study investigated whether SIRT3 was required for RSV alleviating Mn-induced mitochondrial dysfunction in primary cultured neurons from C57BL/6 mice. Here, we showed that Mn (100 and 200 μM) exposure for 24 hr caused significant neuronal damage and mitochondrial dysfunction through increasing mitochondrial ROS, reducing mitochondrial membrane potential and adenosine triphosphate level, and leading to mitochondrial network fragmentation, which could be ameliorated by RSV pretreatment in primary cultured neurons. Additionally, our results also indicated that RSV could activate the SIRT1/PGC-1α signaling pathway and alleviate Mn-induced disruption of mitochondrial biogenesis by increasing SIRT1 expression and activity, enhancing deacetylation of PGC-1α. Furthermore, SIRT3 over-expression increased deacetylation of mitochondrial transcription factor A and mitochondrial DNA (mtDNA) copy number. Oppositely, silencing SIRT3 increased acetylation of mitochondrial transcription factor A and decreased mtDNA copy number. Our results showed SIRT3 was required for the protective effect of RSV in mitochondrial biogenesis. In conclusion, our findings demonstrated that RSV could ameliorate Mn-induced neuronal injury and mitochondrial dysfunction in primary cultured neurons through activating the SIRT1/ PGC-1α signaling pathway, and that SIRT3 is required for promoting mitochondrial biogenesis and attenuating Mn-induced mitochondrial dysfunction.
Collapse
Affiliation(s)
- Qian Sun
- Department of Environmental Health, School of Public Health, China Medical University, Liaoning, China
| | - Run-Run Kang
- Department of Environmental Health, School of Public Health, China Medical University, Liaoning, China
| | - Kai-Ge Chen
- Department of Environmental Health, School of Public Health, China Medical University, Liaoning, China
| | - Kuan Liu
- Department of Environmental Health, School of Public Health, China Medical University, Liaoning, China
| | - Zhuo Ma
- Department of Environmental Health, School of Public Health, China Medical University, Liaoning, China
| | - Chang Liu
- Department of Environmental Health, School of Public Health, China Medical University, Liaoning, China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Liaoning, China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, Liaoning, China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Liaoning, China
| |
Collapse
|
11
|
Lin G, Sun Y, Long J, Sui X, Yang J, Wang Q, Wang S, He H, Luo Y, Qiu Z, Wang Y. Involvement of the Nrf2-Keap1 signaling pathway in protection against thallium-induced oxidative stress and mitochondrial dysfunction in primary hippocampal neurons. Toxicol Lett 2020; 319:66-73. [DOI: 10.1016/j.toxlet.2019.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 10/25/2022]
|
12
|
Tavakoli P, Ghaffarifar F, Delavari H, Shahpari N. Efficacy of manganese oxide (Mn 2O 3) nanoparticles against Leishmania major in vitro and in vivo. J Trace Elem Med Biol 2019; 56:162-168. [PMID: 31473559 DOI: 10.1016/j.jtemb.2019.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 07/28/2019] [Accepted: 08/12/2019] [Indexed: 01/25/2023]
Abstract
BACKGROUND The pentavalent antimonial compounds are the first drug of choice for leishmania infection, but have several side effects that cause some restriction for use. Extension of nanoparticle use in biological research and proven effectiveness of manganese nanoparticles on fungi and bacteria, along with the lack of information about its antileishmanial effects, have motivated this study. Manganese can induce cell apoptosis by increasing FOXO3a-Bim/PUMA mRNA activation and activating of caspase-3 pathway. METHODS This study was aimed to examine the efficacy of manganese oxide nanoparticles againstLeishmania major (MRHO/IR/75/ER) in vitro and in vivo. To evaluate the antileishmanial activity of NPs, light microscopic observation was used to determine the number of remaining parasites in each well. The MTT test was used to determine the cytotoxicity effects of Mn2O3 NPs against L. major promastigotes and macrophage cells. The effect of nanoparticles on cultured amastigotes under in vitro conditions was also investigated. The possible apoptosis of L. major by Mn2O3 NPs was evaluated with flow cytometry assay. Additionally, the preventive and therapeutic effects of Mn2O3 NPs in BALB/c mice following cutaneous L. major infection was tested. The effect of Mn2O3 NPs on promastigotes and amastigotes were proven by MTT assay and amastigote assay, respectively. RESULTS The IC50 value of Mn2O3 NPs against L. major promastigotes and macrophages was 15 and 40 μg ml-1 respectively. The results of flow cytometry showed about 57% of the promastigotes were induced to apoptosis with Mn2O3 NPs. In in vivo studies, the size of the ulcers were significantly reduced, and the survival rate of the mice, in comparison with the control group, was increased. CONCLUSION Mn2O3 NPs has a beneficial effect on L. major promastigotes in vitro and in vivo and could be considered as a candidate for the treatment of this infection.
Collapse
Affiliation(s)
- Pooya Tavakoli
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Fatemeh Ghaffarifar
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Hamid Delavari
- Nanomaterials Group, Department of Materials Engineering, Tarbiat Modares University, Tehran, Iran.
| | - Nima Shahpari
- Department of Electrical Engineering, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
13
|
Dong H, Zhou W, Xin J, Shi H, Yao X, He Z, Wang Z. Salvinorin A moderates postischemic brain injury by preserving endothelial mitochondrial function via AMPK/Mfn2 activation. Exp Neurol 2019; 322:113045. [DOI: 10.1016/j.expneurol.2019.113045] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 08/07/2019] [Accepted: 08/23/2019] [Indexed: 10/26/2022]
|
14
|
Yang Y, Ma S, Wei F, Liang G, Yang X, Huang Y, Wang J, Zou Y. Pivotal role of cAMP-PKA-CREB signaling pathway in manganese-induced neurotoxicity in PC12 cells. ENVIRONMENTAL TOXICOLOGY 2019; 34:1052-1062. [PMID: 31161640 DOI: 10.1002/tox.22776] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/05/2019] [Accepted: 05/09/2019] [Indexed: 06/09/2023]
Abstract
Manganese (Mn) plays a critical role in individual growth and development, yet excessive exposure can result in neurotoxicity, especially cognitive impairment. Neuronal apoptosis is considered as one of the mechanisms of Mn-induced neurotoxicity. Recent evidence suggests that cAMP-PKA-CREB signaling regulates apoptosis and is associated with cognitive function. However, whether this pathway participates in Mn-induced neurotoxicity is not completely understood. To fill this gap, in vitro cultures of PC12 cells were exposed to 0, 400, 500, and 600 μmol/L Mn for 24 hours, respectively. Another group of cells were pretreated with 10.0 μmol/L rolipram (a phosphodiesterase-4 [PDE4] inhibitor) for 1 hour followed by 500 μmol/L Mn exposure for 24 hours. Flow cytometry, immunofluorescence staining, enzyme-linked immunosorbent assay, and Western blot analysis were used to detect the apoptosis rate, protein levels of PDE4, cAMP signaling, and apoptosis-associated proteins, respectively. We found that Mn exposure significantly inhibited cAMP signaling and protein expression of Bcl-2, while increasing apoptosis rate, protein levels of PDE4, Bax, activated caspase-3, and activated caspase-8 in PC12 cells. Pretreatment of rolipram ameliorated Mn-induced deficits in cAMP signaling and apoptosis. These findings demonstrate that cAMP-PKA-CREB signaling pathway-induced apoptosis is involved in Mn-induced neurotoxicity.
Collapse
Affiliation(s)
- Yiping Yang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Shuyan Ma
- Department of Toxicology, Shaanxi Provincial Center for Disease Control and Prevention, Xi'an, China
| | - Fu Wei
- Center for Reproductive Medicine and Genetics, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Guiqiang Liang
- Department of Preventive Medicine, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, China
| | - Yuman Huang
- Guangxi Center for Disease Prevention and Control, Nanning, China
| | - Jian Wang
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, China
| | - Yunfeng Zou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| |
Collapse
|
15
|
Zhu G, Liu Y, Zhi Y, Jin Y, Li J, Shi W, Liu Y, Han Y, Yu S, Jiang J, Zhao X. PKA- and Ca 2+-dependent p38 MAPK/CREB activation protects against manganese-mediated neuronal apoptosis. Toxicol Lett 2019; 309:10-19. [PMID: 30951808 DOI: 10.1016/j.toxlet.2019.04.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/24/2019] [Accepted: 04/01/2019] [Indexed: 11/27/2022]
Abstract
Although manganese (Mn) is an essential trace element, its excessive consumption may lead to neuronal death and neurodegenerative disorders. Human cells launch adaptive responses to attenuate Mn-induced neurotoxicity. However, the regulation of the responsive proteins and their function during Mn-stimulated neurotoxicity remain largely unknown. We report the role of cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB) in Mn-induced neuronal apoptosis. Mn increased CREB phosphorylation and cellular apoptosis in both PC12 cells and mouse brain tissue. Furthermore, downregulation of CREB with shRNA plasmid transfection significantly worsened the PC12 cell apoptosis by decreasing mRNA and protein expression of brain-derived neurotrophic factor (BDNF). Moreover, Mn enhanced protein kinase A (PKA) activation and activation of the p38 MAPK and JNK pathways. Inhibition of p38 MAPK rather than JNK effectively reduced the CREB phosphorylation. Subsequent analysis showed that a PKA inhibitor blocked p38 MAPK and CREB phosphorylation. Moreover, the intracellular Ca2+ chelator BAPTA-AM decreased the phosphorylation of p38 MAPK and CREB but failed to reduce PKA activation. In summary, p38 MAPK/CREB activation via PKA activation and increased cellular Ca2+ helped to alleviate Mn-induced neuronal apoptosis via BDNF regulation. These findings improve our understanding of Mn-induced neurotoxicity and the molecular targets to antagonise it.
Collapse
Affiliation(s)
- Ganlin Zhu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Yiming Liu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Ye Zhi
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Yang Jin
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Jinlong Li
- School of Pharmacy, Nangtong University, Nantong 226001, China.
| | - Weiwei Shi
- Nantong Hospital of Traditional Chinese Medicine, Nantong 226001, China
| | - Yuting Liu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Yu Han
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Shali Yu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Junkang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China.
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China.
| |
Collapse
|
16
|
Sarkar S, Rokad D, Malovic E, Luo J, Harischandra DS, Jin H, Anantharam V, Huang X, Lewis M, Kanthasamy A, Kanthasamy AG. Manganese activates NLRP3 inflammasome signaling and propagates exosomal release of ASC in microglial cells. Sci Signal 2019; 12:12/563/eaat9900. [PMID: 30622196 DOI: 10.1126/scisignal.aat9900] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Chronic, sustained inflammation underlies many pathological conditions, including neurodegenerative diseases. Divalent manganese (Mn2+) exposure can stimulate neurotoxicity by increasing inflammation. In this study, we examined whether Mn2+ activates the multiprotein NLRP3 inflammasome complex to promote neuroinflammation. Exposing activated mouse microglial cells to Mn2+ substantially augmented NLRP3 abundance, caspase-1 cleavage, and maturation of the inflammatory cytokine interleukin-1β (IL-1β). Exposure of mice to Mn2+ had similar effects in brain microglial cells. Furthermore, Mn2+ impaired mitochondrial ATP generation, basal respiratory rate, and spare capacity in microglial cells. These data suggest that Mn-induced mitochondrial defects drove the inflammasome signal amplification. We found that Mn induced cell-to-cell transfer of the inflammasome adaptor protein ASC in exosomes. Furthermore, primed microglial cells exposed to exosomes from Mn-treated mice released more IL-1β than did cells exposed to exosomes from control-treated animals. We also observed that welders exposed to manganese-containing fumes had plasma exosomes that contained more ASC than did those from a matched control group. Together, these results suggest that the divalent metal manganese acts as a key amplifier of NLRP3 inflammasome signaling and exosomal ASC release.
Collapse
Affiliation(s)
- Souvarish Sarkar
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Dharmin Rokad
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Emir Malovic
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Jie Luo
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Dilshan S Harischandra
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Huajun Jin
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Xuemei Huang
- Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Mechelle Lewis
- Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Anumantha G Kanthasamy
- Department of Biomedical Sciences, Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA.
| |
Collapse
|
17
|
Zhao X, Liu Y, Zhu G, Liang Y, Liu B, Wu Y, Han M, Sun W, Han Y, Chen G, Jiang J. SIRT1 downregulation mediated Manganese-induced neuronal apoptosis through activation of FOXO3a-Bim/PUMA axis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 646:1047-1055. [PMID: 30235590 DOI: 10.1016/j.scitotenv.2018.07.363] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/21/2018] [Accepted: 07/25/2018] [Indexed: 06/08/2023]
Abstract
Manganese (Mn) is an essential trace element. Excessive exposure to Mn may lead to neuronal death and neurodegenerative disorders. Accumulating evidence has shown that silent mating type information regulation 2 homolog 1 (SIRT1) plays a vital role in brain damage. However, whether aberrant SIRT1 levels contribute to Mn-induced neurotoxicity remains unknown. In this study, we report the important role of SIRT1 downregulation during Mn-induced neuronal apoptosis. Mn was found to downregulate SIRT1 protein levels in the rat pheochromocytoma (PC12) cells and mouse brain tissues. Mn enhanced SIRT1 protein degradation and downregulated its gene expression. Furthermore, Mn induced cell apoptosis in a dose-dependent manner both in vitro and in vivo, and resulted in an increase in forkhead box O (FOXO) 3a expression and acetylation. SIRT1 activation by resveratrol clearly attenuated Mn-triggered apoptosis and FOXO3a activation. Mn markedly increased the expression of Bcl-2 interacting mediator of cell death (Bim) and p53-up-regulated modulator of apoptosis (PUMA), whereas downregulation of FOXO3a significantly inhibited their upregulation and subsequent apoptosis. In summary, we determined that Mn downregulated SIRT1 by multiple mechanisms, thus led to apoptosis via activation of the FOXO3a-Bim/PUMA axis in PC12 cells. These findings on the impact of Mn on SIRT1 may lead to an improved understanding of Mn-induced neurotoxicity and provide a molecular target to antagonise Mn-associated neuronal damage.
Collapse
Affiliation(s)
- Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Yiming Liu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Ganlin Zhu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Yuanyuan Liang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Bo Liu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Yifan Wu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Muxi Han
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Wenxing Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong 226019, China
| | - Yu Han
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Gang Chen
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Junkang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China.
| |
Collapse
|