1
|
Hong H, Liu S, Yang T, Lin J, Luo K, Xu Y, Li T, Xi Y, Yang L, Lu YQ, Yuan W, Zhou Z. Manganese exposure induces parkinsonism-like symptoms by Serpina3n-TFEB-v/p-ATPase signaling mediated lysosomal dysfunction. Cell Biol Toxicol 2025; 41:34. [PMID: 39847159 PMCID: PMC11759460 DOI: 10.1007/s10565-025-09989-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 01/03/2025] [Indexed: 01/30/2025]
Abstract
Manganese (Mn) is a neurotoxin that has been etiologically linked to the development of neurodegenerative diseases in the case of overexposure. It is widely accepted that overexposure to Mn leads to manganism, which has clinical symptoms similar to Parkinson's disease (PD), and is referred to as parkinsonism. Astrocytes have been reported to scavenge and degrade extracellular α-synuclein (α-Syn) in the brain. However, the mechanisms of Mn-induced neurotoxicity associated with PD remain unclear. Serpina3n is highly expressed in astrocytes and has been implicated in several neuropathologies. The role Serpina3n plays in Mn neurotoxicity and PD pathogenesis is still unknown. Here, we used wild-type and Serpina3n knockout (KO) C57BL/6 J mice with i.p. injection of 32.5 mg/kg MnCl2 once a day for 6 weeks to elucidate the role of Serpina3n in Mn-caused neurotoxicity regarding parkinsonism pathogenesis. We performed behavioral tests (open field, suspension and pole-climbing tests) to observe Mn-induced motor changes, immunohistochemistry to detect Mn-induced midbrain changes, and Western blot to detect Mn-induced changes of protein expression. It was found that Serpina3n KO markedly alleviated Mn neurotoxicity in mice by attenuating midbrain dopaminergic neuron damage and ameliorating motor deficits. Furthermore, using immunofluorescence colocalization analysis, Western blot and quantitative real-time PCR on Mn-treated C8-D1A cells, we found that Serpina3n KO significantly improved astrocytic α-Syn clearance by suppressing Mn-induced lysosomal dysfunction. Reduced transcription factor EB (TFEB)-v/p-ATPase signaling is responsible for the impairment of the lysosomal acidic environment. These novel findings highlight Serpina3n as a detrimental factor in Mn neurotoxicity associated with parkinsonism, capture the novel role of Serpina3n in regulating lysosomal function, and provide a potential target for antagonizing Mn neurotoxicity and curing parkinsonism in humans.
Collapse
Affiliation(s)
- Huihui Hong
- Department of Environmental Medicine, School of Medicine, Chongqing University, Chongqing, China
| | - Sicheng Liu
- Department of Environmental Medicine, School of Medicine, Chongqing University, Chongqing, China
| | - Ting Yang
- Department of Otolaryngology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Jinxian Lin
- Department of Environmental Medicine, School of Medicine, Chongqing University, Chongqing, China
| | - Kun Luo
- Department of Environmental Medicine, School of Medicine, Chongqing University, Chongqing, China
| | - Yudong Xu
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Li
- Department of Emergency Medicine, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, Hangzhou, China
| | - Yu Xi
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, 100048, Beijing, China
| | - Lingling Yang
- Department of Occupational Health, Third Military Medical University, 400038, Chongqing, China
| | - Yuan-Qiang Lu
- Department of Emergency Medicine, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China.
- Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, Hangzhou, China.
| | - Wei Yuan
- Department of Otolaryngology, Chongqing General Hospital, Chongqing University, Chongqing, China.
| | - Zhou Zhou
- Department of Environmental Medicine, School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
2
|
Zheng XW, Fang YY, Lin JJ, Luo JJ, Li SJ, Aschner M, Jiang YM. Signal Transduction Associated with Mn-induced Neurological Dysfunction. Biol Trace Elem Res 2024; 202:4158-4169. [PMID: 38155332 DOI: 10.1007/s12011-023-03999-0] [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: 09/09/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
Manganese (Mn) is a heavy metal that occurs widely in nature and has a vital physiological role in growth and development. However, excessive exposure to Mn can cause neurological damage, especially cognitive dysfunction, such as learning disability and memory loss. Numerous studies on the mechanisms of Mn-induced nervous system damage found that this metal targets a variety of metabolic pathways, for example, endoplasmic reticulum stress, apoptosis, neuroinflammation, cellular signaling pathway changes, and neurotransmitter metabolism interference. This article reviews the latest research progress on multiple signaling pathways related to Mn-induced neurological dysfunction.
Collapse
Affiliation(s)
- Xiao-Wei Zheng
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China
| | - Yuan-Yuan Fang
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China
| | - Jun-Jie Lin
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China
| | - Jing-Jing Luo
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China
| | - Shao-Jun Li
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China.
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China.
| | - Michael Aschner
- The Department of Molecular Pharmacology at Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yue-Ming Jiang
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China.
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China.
| |
Collapse
|
3
|
Pajarillo E, Nyarko-Danquah I, Digman A, Multani HK, Kim S, Gaspard P, Aschner M, Lee E. Mechanisms of manganese-induced neurotoxicity and the pursuit of neurotherapeutic strategies. Front Pharmacol 2022; 13:1011947. [PMID: 36605395 PMCID: PMC9808094 DOI: 10.3389/fphar.2022.1011947] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/01/2022] [Indexed: 01/07/2023] Open
Abstract
Chronic exposure to elevated levels of manganese via occupational or environmental settings causes a neurological disorder known as manganism, resembling the symptoms of Parkinson's disease, such as motor deficits and cognitive impairment. Numerous studies have been conducted to characterize manganese's neurotoxicity mechanisms in search of effective therapeutics, including natural and synthetic compounds to treat manganese toxicity. Several potential molecular targets of manganese toxicity at the epigenetic and transcriptional levels have been identified recently, which may contribute to develop more precise and effective gene therapies. This review updates findings on manganese-induced neurotoxicity mechanisms on intracellular insults such as oxidative stress, inflammation, excitotoxicity, and mitophagy, as well as transcriptional dysregulations involving Yin Yang 1, RE1-silencing transcription factor, transcription factor EB, and nuclear factor erythroid 2-related factor 2 that could be targets of manganese neurotoxicity therapies. This review also features intracellular proteins such as PTEN-inducible kinase 1, parkin, sirtuins, leucine-rich repeat kinase 2, and α-synuclein, which are associated with manganese-induced dysregulation of autophagy/mitophagy. In addition, newer therapeutic approaches to treat manganese's neurotoxicity including natural and synthetic compounds modulating excitotoxicity, autophagy, and mitophagy, were reviewed. Taken together, in-depth mechanistic knowledge accompanied by advances in gene and drug delivery strategies will make significant progress in the development of reliable therapeutic interventions against manganese-induced neurotoxicity.
Collapse
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Ivan Nyarko-Danquah
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Alexis Digman
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Harpreet Kaur Multani
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL, United States
| | - Sanghoon Kim
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Patric Gaspard
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY, United States
| | - Eunsook Lee
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| |
Collapse
|
4
|
Qi Z, Wang S, Li J, Wen Y, Cui R, Zhang K, Liu Y, Yang X, Zhang L, Xu B, Liu W, Xu Z, Deng Y. Protective role of mRNA demethylase FTO on axon guidance molecules of nigro-striatal projection system in manganese-induced parkinsonism. JOURNAL OF HAZARDOUS MATERIALS 2022; 426:128099. [PMID: 34954437 DOI: 10.1016/j.jhazmat.2021.128099] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/02/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
One of the major environmental factors that induce PD is Manganese (Mn). Cellular and molecular mechanism of parkinsonism caused by Mn has not been explored clearly. The results of in vivo and in vitro experiments showed that Mn exposure caused abnormal projection of dopaminergic neurons and decreased mRNA expression and protein levels of FTO. This is due to Mn-induced the upregulation of Foxo3a. Using the cell model of overexpression of FTO, we found that FTO could antagonize Mn-induced the down-regulation of axon guidance molecule ephrin-B2 through RNA-seq, MeRIP-qPCR, and RT-qPCR experiments. Through RIP-seq and actinomycin D experiments, it was found that FTO can up-regulate the mRNA m6A level of ephrin-B2, which can be recognized by YTHDF2 and degraded. Finally, it is proved that Mn induces dopaminergic neurons projection injury and motor dysfunction through Foxo3a/FTO/m6A/ephrin-B2/YTHDF2 signal pathway.
Collapse
Affiliation(s)
- Zhipeng Qi
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China.
| | - Shuang Wang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China.
| | - Jiashuo Li
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China.
| | - Yi Wen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China.
| | - Rong Cui
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China.
| | - Ke Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.
| | - Yanan Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China; Department of Preventive Health, Zhuhai People's Hospital, Zhuhai, Guangdong, China.
| | - Xinxin Yang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China; School of Public Health, Xuzhou Medical University, Xuzhou 221004, China.
| | - Lei Zhang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China.
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China.
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China.
| | - Zhaofa Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China.
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China.
| |
Collapse
|
5
|
Critical Involvement of Glial Cells in Manganese Neurotoxicity. BIOMED RESEARCH INTERNATIONAL 2021; 2021:1596185. [PMID: 34660781 PMCID: PMC8514895 DOI: 10.1155/2021/1596185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022]
Abstract
Over the years, most of the research concerning manganese exposure was restricted to the toxicity of neuronal cells. Manganese is an essential trace element that in high doses exerts neurotoxic effects. However, in the last two decades, efforts have shifted toward a more comprehensive approach that takes into account the involvement of glial cells in the development of neurotoxicity as a brain insult. Glial cells provide structural, trophic, and metabolic support to neurons. Nevertheless, these cells play an active role in adult neurogenesis, regulation of synaptogenesis, and synaptic plasticity. Disturbances in glial cell function can lead to neurological disorders, including neurodegenerative diseases. This review highlights the pivotal role that glial cells have in manganese-induced neurotoxicity as well as the most sounding mechanisms involved in the development of this phenomenon.
Collapse
|
6
|
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
|