1
|
Tang L, Liu M, Tao Y, Ranson JM, Napolioni V, Wang H, Huang J. Association of aging acceleration with serum neurofilament light chain levels: Implications for the roles of modifiable aging factors. J Affect Disord 2025; 372:481-490. [PMID: 39638062 DOI: 10.1016/j.jad.2024.12.023] [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: 08/13/2024] [Revised: 10/27/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Neurofilament light chain (NfL) is a specific biomarker of neuroaxonal damage and related neurodegenerative diseases. Aging acceleration, which reflects the impact of modifiable factors on the aging process, is increasingly recognized for its relevance. While normal aging is known to contribute substantially to neuroaxonal damage and many neurodegenerative diseases, the effects of aging acceleration warrant further investigation. This study aimed to investigate the association and causality between aging acceleration and serum NfL levels. METHODS We conducted a cross-sectional study involving 1695 adult participants from NHANES 2013-2014 to evaluate the association, dose-response relationship, and interaction network between aging acceleration and serum NfL levels. And we used Mendelian randomization (MR) to assess the causal effects between serum NfL levels and aging acceleration. RESULTS Significant positive associations were observed between aging acceleration and serum NfL levels. In linear regression, the regression coefficients were 0.016 (95 % CI: 0.011-0.021) for biological age acceleration and 0.020 (95 % CI: 0.012-0.028) for phenotypic age acceleration. In logistic regression, the odds ratios were 1.052 (95 % CI: 1.029-1.076) and 1.093 (95 % CI: 1.064-1.123), respectively. Restricted cubic spline regression identified significant positive dose-response relationships, and bidirectional MR analyses demonstrated forward causal effects. CONCLUSION Our study indicates that aging acceleration is significantly associated with serum NfL levels, with higher levels of aging acceleration linked to an increased risk of neuroaxonal damage. These findings provide robust evidence that aging acceleration affects the risk of neuroaxonal damage and highlight the importance of modifiable aging factors.
Collapse
Affiliation(s)
- Liwei Tang
- Department of School of Public Health and Emergency Medicine, Southern University of Science and Technology, No. 1088 Xueyuan Avenue, Shenzhen 518055, China
| | - Min Liu
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, No. 1068 Xueyuan Avenue, Shenzhen, Guangdong 518055, China
| | - Yifan Tao
- School of Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Janice M Ranson
- University of Exeter Medical School, Heavitree Road, Exeter EX12LU, UK
| | - Valerio Napolioni
- School of Biosciences and Veterinary Medicine, University of Camerino, Piazza Cavour 19/f, Camerino 62032, Italy
| | - Haidong Wang
- Department of School of Public Health and Emergency Medicine, Southern University of Science and Technology, No. 1088 Xueyuan Avenue, Shenzhen 518055, China
| | - Jie Huang
- Department of School of Public Health and Emergency Medicine, Southern University of Science and Technology, No. 1088 Xueyuan Avenue, Shenzhen 518055, China.
| |
Collapse
|
2
|
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
|
3
|
Chen N, Zhou H, He B, Peng S, Ding F, Liu QH, Ma Z, Liu W, Xu B. Melatonin promotes cell cycle progression of neural stem cells subjected to manganese via Nurr1. ENVIRONMENTAL TOXICOLOGY 2024; 39:3883-3896. [PMID: 38563506 DOI: 10.1002/tox.24258] [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: 11/21/2023] [Revised: 03/04/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
Excessive exposure to manganese (Mn) through drinking water and food during pregnancy significantly heightens the likelihood of neurodevelopmental damage in offspring. Multiple studies have indicated that melatonin (Mel) may help to relieve neurodevelopmental disorders caused by Mn, but potential mechanisms underlying this effect require further exploration. Here, we utilized primary neural stem cells (NSCs) as a model to elucidate the molecular mechanism underlying the protective function of Mel on Mn-induced cell proliferation dysfunction and cycle arrest. Our results showed that Mn disrupted the cell cycle in NSCs by suppressing positive regulatory proteins (CDK2, Cyclin A, Cyclin D1, and E2F1) and enhancing negative ones (p27KIP1 and p57KIP2), leading to cell proliferation dysfunction. Mel inhibited the Mn-dependent changes to these proteins and the cell cycle through nuclear receptor-related protein 1 (Nurr1), thus alleviating the proliferation dysfunction. Knockdown of Nurr1 using lentivirus-expressed shRNA in NSCs resulted in a diminished protective effect of Mel. We concluded that Mel mitigated Mn-induced proliferation dysfunction and cycle arrest in NSCs through Nurr1.
Collapse
Affiliation(s)
- Nan Chen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Han Zhou
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Bin He
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Sen Peng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Feng Ding
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Qi-Hao Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Zhuo Ma
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
- Key laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, PR China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
- Key laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, PR China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
- Key laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, PR China
| |
Collapse
|
4
|
Najafi N, Barangi S, Moosavi Z, Aghaee-Bakhtiari SH, Mehri S, Karimi G. Melatonin Attenuates Arsenic-Induced Neurotoxicity in Rats Through the Regulation of miR-34a/miR-144 in Sirt1/Nrf2 Pathway. Biol Trace Elem Res 2024; 202:3163-3179. [PMID: 37853305 DOI: 10.1007/s12011-023-03897-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 09/29/2023] [Indexed: 10/20/2023]
Abstract
Arsenic (As) exposure is known to cause several neurological disorders through various molecular mechanisms such as oxidative stress, apoptosis, and autophagy. In the current study, we assessed the effect of melatonin (Mel) on As-induced neurotoxicity. Thirty male Wistar rat were treated daily for 28 consecutive days. As (15 mg/kg, gavage) and Mel (10 and 20 mg/kg, i.p.) were administered to rats. Morris water maze test was done to evaluate learning and memory impairment in training days and probe trial. Oxidative stress markers including MDA and GSH levels, SOD activity, and HO-1 levels were measured. Besides, the levels of apoptosis (caspase 3, Bax/Bcl2 ratio) and autophagy markers (Sirt1, Beclin-1, and LC3 II/I ratio) as well as the expression of miR-144 and miR-34a in cortex tissue were determined. As exposure disturbed learning and memory in animals and Mel alleviated these effects. Also, Mel recovered cortex pathological damages and oxidative stress induced by As. Furthermore, As increased the levels of apoptosis and autophagy proteins in cortex, while Mel (20 mg/kg) decreased apoptosis and autophagy. Also, Mel increased the expression of miR-144 and miR-34a which inhibited by As. In conclusion, Mel administration attenuated As-induced neurotoxicity through anti-oxidative, anti-apoptotic, and anti-autophagy mechanisms, which may be recommended as a therapeutic target for neurological disorders.
Collapse
Affiliation(s)
- Nahid Najafi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samira Barangi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Moosavi
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyed Hamid Aghaee-Bakhtiari
- Bioinformatics Research Group, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soghra Mehri
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Lu M, Deng P, Yang L, Wang X, Mei X, Zhou C, Chen M, Zhou Z, Pi H, Wu L, Yu Z. Manganese overexposure induces Parkinson-like symptoms, altered lipid signature and oxidative stress in C57BL/6 J mouse. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 263:115238. [PMID: 37441952 DOI: 10.1016/j.ecoenv.2023.115238] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/02/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023]
Abstract
Although adequate intake of manganese (Mn) is essential to humans, Mn in excess is neurotoxic. Exposure to extremely high doses of Mn results in "manganism", a condition that exhibits Parkinson-like symptoms. However, the mechanisms underlying its neurotoxic effects in Mn-induced parkinsonism pathogenesis are unclear. In this study, 8-week-old male C57BL/6 J mice were injected intraperitoneally with saline and 50 mg/kg MnCl2 respectively once daily for 14 days to produce an acute Mn neurotoxicity model. Accumulation of Mn in the midbrain, motor dysfunction and loss of dopaminergic neurons in the substantia nigra evidenced Mn neurotoxicity. Untargeted lipidomic analysis demonstrated that Mn overexposure altered lipidome profiles. A significant modulation of 12 lipid subclasses belonging to 5 different categories were found in the midbrain and among the most abundant lipids were sphingolipids, glycerophospholipids, and glycerides. The levels of sphingomyelin (SM) were significantly decreased after Mn treatment. The expression of SM biosynthesis genes was decreased dramatically while sphingomyelinase was up-regulated. In addition, we observed oxidative stress in both the midbrain of mice and MN9D cells, indicated by the increase of MDA level, the decrease of reduced GSH level and the inhibition of SOD and GPx enzyme activities. There was a correlation between these changes and motor dysfunctions. Overall, our study is the first to use lipidomics techniques to explore the pathogenesis of Mn-induced parkinsonism in C57BL/6 J mice. Mn induced molecular events in the midbrain, such as lipid metabolism disorders, oxidative stress and dopaminergic neurons injury, may mechanistically play important roles in the pathogenesis of Parkinson-like symptoms. Moreover, these findings emphasize the necessity for reducing the health risk of environmental neurotoxic pollutants in relation to parkinsonism.
Collapse
Affiliation(s)
- Muxue Lu
- School of Medicine, Guangxi University, Nanning 530004, Guangxi, China
| | - Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Lingling Yang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Xue Wang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Xiang Mei
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Chao Zhou
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China; 953 Hospital, Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Shigatse 857000, China
| | - Mengyan Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Zhou Zhou
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Lichuan Wu
- School of Medicine, Guangxi University, Nanning 530004, Guangxi, China.
| | - Zhengping Yu
- School of Medicine, Guangxi University, Nanning 530004, Guangxi, China.
| |
Collapse
|
6
|
Pajarillo E, Kim S, Digman A, Dutton M, Son DS, Aschner M, Lee E. The role of microglial LRRK2 kinase in manganese-induced inflammatory neurotoxicity via NLRP3 inflammasome and RAB10-mediated autophagy dysfunction. J Biol Chem 2023; 299:104879. [PMID: 37269951 PMCID: PMC10331485 DOI: 10.1016/j.jbc.2023.104879] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/12/2023] [Accepted: 05/20/2023] [Indexed: 06/05/2023] Open
Abstract
Chronic manganese (Mn) exposure can lead to manganism, a neurological disorder sharing common symptoms with Parkinson's disease (PD). Studies have shown that Mn can increase the expression and activity of leucine-rich repeat kinase 2 (LRRK2), leading to inflammation and toxicity in microglia. LRRK2 G2019S mutation also elevates LRRK2 kinase activity. Thus, we tested if Mn-increased microglial LRRK2 kinase is responsible for Mn-induced toxicity, and exacerbated by G2019S mutation, using WT and LRRK2 G2019S knock-in mice and BV2 microglia. Mn (30 mg/kg, nostril instillation, daily for 3 weeks) caused motor deficits, cognitive impairments, and dopaminergic dysfunction in WT mice, which were exacerbated in G2019S mice. Mn induced proapoptotic Bax, NLRP3 inflammasome, IL-1β, and TNF-α in the striatum and midbrain of WT mice, and these effects were more pronounced in G2019S mice. BV2 microglia were transfected with human LRRK2 WT or G2019S, followed by Mn (250 μM) exposure to better characterize its mechanistic action. Mn increased TNF-α, IL-1β, and NLRP3 inflammasome activation in BV2 cells expressing WT LRRK2, which was elevated further in G2019S-expressing cells, while pharmacological inhibition of LRRK2 mitigated these effects in both genotypes. Moreover, the media from Mn-treated G2019S-expressing BV2 microglia caused greater toxicity to the cath.a-differentiated (CAD) neuronal cells compared to media from microglia expressing WT. Mn-LRRK2 activated RAB10 which was exacerbated in G2019S. RAB10 played a critical role in LRRK2-mediated Mn toxicity by dysregulating the autophagy-lysosome pathway and NLRP3 inflammasome in microglia. Our novel findings suggest that microglial LRRK2 via RAB10 plays a critical role in Mn-induced neuroinflammation.
Collapse
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Sanghoon Kim
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Alexis Digman
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Matthew Dutton
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Deok-Soo Son
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, Tennessee, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Eunsook Lee
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA.
| |
Collapse
|
7
|
Pajarillo E, Kim SH, Digman A, Dutton M, Son DS, Aschner M, Lee E. The role of microglial LRRK2 in manganese-induced inflammatory neurotoxicity via NLRP3 inflammasome and RAB10-mediated autophagy dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535418. [PMID: 37066140 PMCID: PMC10103982 DOI: 10.1101/2023.04.03.535418] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Chronic exposure to manganese (Mn) can lead to manganism, a neurological disorder sharing common symptoms with Parkinson's disease (PD). Studies have shown that Mn can increase the expression and activity of leucine-rich repeat kinase 2 (LRRK2), leading to inflammation and toxicity in microglia. LRRK2 G2019S mutation also elevates LRRK2 kinase activity. Thus, we tested if Mn-increased microglial LRRK2 kinase is responsible for Mn-induced toxicity, and exacerbated by G2019S mutation, using WT and LRRK2 G2019S knock-in mice, and BV2 microglia. Mn (30 mg/kg, nostril instillation, daily for 3 weeks) caused motor deficits, cognitive impairments, and dopaminergic dysfunction in WT mice, which were exacerbated in G2019S mice. Mn induced proapoptotic Bax, NLRP3 inflammasome, IL-1β and TNF-α in the striatum and midbrain of WT mice, and these effects were exacerbated in G2019S mice. BV2 microglia were transfected with human LRRK2 WT or G2019S, followed by Mn (250 μM) exposure to better characterize its mechanistic action. Mn increased TNF-α, IL-1β, and NLRP3 inflammasome activation in BV2 cells expressing WT LRRK2, which was exacerbated in G2019S-expressing cells, while pharmacological inhibition of LRRK2 mitigated these effects in both genotypes. Moreover, the media from Mn-treated BV2 microglia expressing G2019S caused greater toxicity to cath.a-differentiated (CAD) neuronal cells compared to media from microglia expressing WT. Mn-LRRK2 activated RAB10, which was exacerbated in G2019S. RAB10 played a critical role in LRRK2-mediated Mn toxicity by dysregulating the autophagy-lysosome pathway, and NLRP3 inflammasome in microglia. Our novel findings suggest that microglial LRRK2 via RAB10 plays a critical role in Mn-induced neuroinflammation.
Collapse
|
8
|
Smit C, De Wet S, Barron T, Loos B. Rooibos tea-in the cross fire of ROS, mitochondrial dysfunction and loss of proteostasis-positioned for healthy aging. Biogerontology 2023; 24:149-162. [PMID: 36781516 DOI: 10.1007/s10522-022-10012-z] [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: 09/20/2022] [Accepted: 12/16/2022] [Indexed: 02/15/2023]
Abstract
Impaired mitochondrial function and loss of cellular proteostasis control are key hallmarks of aging and are implicated in the development of neurodegenerative diseases. A common denominator is the cell's inability to handle reactive oxygen species (ROS), leading to major downstream oxidative damage that exacerbates neuronal dysfunction. Although we have progressed in understanding the molecular defects associated with neuronal aging, many unanswered questions remain. How much ROS is required to serve cellular function before it becomes detrimental and how does the cell's oxidative status impact mitochondrial function and protein degradation through autophagy? How does ROS regulate autophagy? Aspalathus linearis, also commonly known as rooibos, is an endemic South African plant that is gaining globally acclaim for its antioxidant properties and its role as functional medicinal beverage. In this article we dissect the role of rooibos in the context of the cell's ROS handling capacity, mitochondrial function and autophagy activity. By addressing the dynamic relationship between these critical interconnected systems, and by evaluating the functional properties of rooibos, we unravel its position for preserving cell viability and promoting healthy aging.
Collapse
Affiliation(s)
- Catherine Smit
- Department of Physiological Sciences, Stellenbosch University, Merriman Avenue, Mike de Vries Building, Stellenbosch, 7600, South Africa
| | - Sholto De Wet
- Department of Physiological Sciences, Stellenbosch University, Merriman Avenue, Mike de Vries Building, Stellenbosch, 7600, South Africa
| | - Tamryn Barron
- Department of Physiological Sciences, Stellenbosch University, Merriman Avenue, Mike de Vries Building, Stellenbosch, 7600, South Africa
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Merriman Avenue, Mike de Vries Building, Stellenbosch, 7600, South Africa.
| |
Collapse
|
9
|
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
|
10
|
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: 18] [Impact Index Per Article: 6.0] [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
|
11
|
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
|
12
|
A review on neurodegenerative diseases associated with oxidative stress and mitochondria. Int J Health Sci (Qassim) 2022. [DOI: 10.53730/ijhs.v6ns1.6130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Alzheimer's disease, Parkinson's disease, and other neurological diseases afflict people of all ages. Neuronal loss and cognitive dysfunction are common symptoms of these disorders. Overproduction of reactive oxygen species has been demonstrated to aggravate disease progression in previous investigations (ROS). Because of the large quantities of polyunsaturated fatty acids in their membranes and their fast oxygen consumption rate, neurons are especially susceptible to oxidative damage. The molecular aetiology of neurodegeneration produced by changes in redox balance has not yet been established. New antioxidants have shown considerable potential in modifying disease characteristics. For the treatment of Alzheimer's disease and other neurodegenerative illnesses such as Parkinson's disease, ALS and spinocerebellar ataxia and Huntington's disease, antioxidant-based therapies are examined extensively in the literature.
Collapse
|
13
|
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: 32] [Impact Index Per Article: 10.7] [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
|
14
|
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
|
15
|
Alali S, Riazi G, Ashrafi-Kooshk MR, Meknatkhah S, Ahmadian S, Hooshyari Ardakani M, Hosseinkhani B. Cannabidiol Inhibits Tau Aggregation In Vitro. Cells 2021; 10:cells10123521. [PMID: 34944028 PMCID: PMC8700709 DOI: 10.3390/cells10123521] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/03/2021] [Accepted: 12/09/2021] [Indexed: 12/26/2022] Open
Abstract
A hallmark of Alzheimer’s disease (AD) is the accumulation of tau protein in the brain. Compelling evidence indicates that the presence of tau aggregates causes irreversible neuronal destruction, eventually leading to synaptic loss. So far, the inhibition of tau aggregation has been recognized as one of the most effective therapeutic strategies. Cannabidiol (CBD), a major component found in Cannabis sativa L., has antioxidant activities as well as numerous neuroprotective features. Therefore, we hypothesize that CBD may serve as a potent substance to hamper tau aggregation in AD. In this study, we aim to investigate the CBD effect on the aggregation of recombinant human tau protein 1N/4R isoform using biochemical methods in vitro and in silico. Using Thioflavin T (ThT) assay, circular dichroism (CD), and atomic force microscopy (AFM), we demonstrated that CBD can suppress tau fibrils formation. Moreover, by quenching assay, docking, and job’s plot, we further demonstrated that one molecule of CBD interacts with one molecule of tau protein through a spontaneous binding. Experiments performed by quenching assay, docking, and Thioflavin T assay further established that the main forces are hydrogen Van der Waals and some non-negligible hydrophobic forces, affecting the lag phase of tau protein kinetics. Taken together, this study provides new insights about a natural substance, CBD, for tau therapy which may offer new hope for the treatment of AD.
Collapse
Affiliation(s)
- Soha Alali
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 1417614335, Iran; (M.R.A.-K.); (S.M.)
- Correspondence: (S.A.); (G.R.)
| | - Gholamhossein Riazi
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 1417614335, Iran; (M.R.A.-K.); (S.M.)
- Correspondence: (S.A.); (G.R.)
| | - Mohammad Reza Ashrafi-Kooshk
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 1417614335, Iran; (M.R.A.-K.); (S.M.)
| | - Sogol Meknatkhah
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 1417614335, Iran; (M.R.A.-K.); (S.M.)
| | - Shahin Ahmadian
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614335, Iran;
| | - Mohammad Hooshyari Ardakani
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, G.C., Evin, Tehran 1983969411, Iran;
| | - Baharak Hosseinkhani
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500 Hasselt, Belgium;
| |
Collapse
|
16
|
Li J, Deng Y, Peng D, Zhao L, Fang Y, Zhu X, Li S, Aschner M, Ou S, Jiang Y. Sodium P-aminosalicylic Acid Attenuates Manganese-Induced Neuroinflammation in BV2 Microglia by Modulating NF-κB Pathway. Biol Trace Elem Res 2021; 199:4688-4699. [PMID: 33447908 DOI: 10.1007/s12011-021-02581-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
Exposure to high levels of manganese (Mn) leads to brain Mn accumulation, and a disease referred to as manganism. Activation of microglia plays an important role in Mn-induced neuroinflammation. Sodium p-aminosalicylic acid (PAS-Na) is a non-steroidal anti-inflammatory drug that inhibits Mn-induced neuroinflammation. The aim of the current study was to explore the role of NF-κB in the protective mechanism of PAS-Na on Mn-induced neuroinflammation in BV2 microglial experimental model. We treated BV2 microglia with 200 μM Mn for 24 h followed by 48 h treatment with graded concentrations of PAS-Na, using an NF-kB inhibitor, JSH-23, as a positive control. MTT results established that 200 and 400 μM PAS-Na treatment increased the Mn-induced cell viability reduction. NF-κB (P65) mRNA expression and the phosphorylation of p65 were increased in Mn-treated BV2 cell, and suppressed by PAS-Na, analogous to the effect of JSH-23 pretreatment. Furthermore, PAS-Na significantly reduced the contents of the inflammatory cytokine TNF-α and IL-1β, both of which were increased by Mn treatment. The current results show that PAS-Na attenuated Mn-induced inflammation by abrogating the activation of the NF-κB signaling pathways and reduced the release of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Junyan Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Yue Deng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Dongjie Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Lin Zhao
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Yuanyuan Fang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Xiaojuan Zhu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Shaojun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Shiyan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Yueming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| |
Collapse
|
17
|
Abdollahzade N, Majidinia M, Babri S. Melatonin: a pleiotropic hormone as a novel potent therapeutic candidate in arsenic toxicity. Mol Biol Rep 2021; 48:6603-6618. [PMID: 34453671 DOI: 10.1007/s11033-021-06669-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/18/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Arsenic is a natural element which exists in the environment in inorganic and organic forms. In humans, the main reason for the toxicity of arsenic is its uptake via water sources. As polluted water and the problems associated with it can be found in many countries. Therefore, considering all these positive effects of melatonin, this review is aimed at melatonin supplementation therapy on arsenic toxicity which seems to be a suitable therapeutic agent to eliminate the adverse effects of arsenic. METHODS AND RESULTS It is seen in previous studies that chronic exposure to arsenic could cause serious dys functions of organs and induce different degrees of toxicities that is one of the first hazardous materials in the classification of substances by the United States Environmental Protection Agency so leads to costly cleanup operations burdening the economy. Arsenic harmfulness degree depends on the bioavailability, chemical form, valence state, detoxification, and metabolism of human body. The oxidative stress has a major role in arsenic-induced toxicity; on the other hand, it was discovered that melatonin is a powerful scavenger for free radical and it's an extensive-spectrum antioxidant. CONCLUSION Due to its highly lipophilic and small size properties, melatonin accesses all intracellular organs by easily passing via the cell membrane and prevents protein, DNA damage, and lipid peroxidation. In particular, melatonin, by protecting and reducing oxidative stress in mitochondria, can normalize homeostasis and mitochondrial function and ultimately prevent apoptosis and cell death.
Collapse
Affiliation(s)
- Naseh Abdollahzade
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Physiology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| | - Shirin Babri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Physiology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
18
|
Zhang H, Yang T. FBXW7alpha Promotes the Recovery of Traumatic Spinal Cord. Curr Mol Med 2021; 20:494-504. [PMID: 31870261 DOI: 10.2174/1566524020666191223164916] [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: 08/15/2019] [Revised: 11/20/2019] [Accepted: 12/12/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND White matter damage and neuronal cell death are incurred by spinal cord injury (SCI). FBXW7α, an important mediator of cell division and growth was investigated to explore its role in repairing the traumatic spinal cord in rats. Underlying mechanisms such as oxidative stress and inflammasomes signaling were also studied. METHODS Spinal cord injury in rats was established by longitudinal surgical incision from the lower to mid-thoracic vertebrae on the backside, followed by 20-g weight placed on the exposed Th12 surface for 30 min. AAV-delivered FBXW7α and -sh-FBXW7α were intrathecally injected into the rat spinal cord. Indices of oxidation, neurotrophic factors, and pyroptosis were measured by Western blot, Elisa, and RT-PCR. RESULTS We found the overexpression of FBXW7α in spinal cord rescue neuronal death triggered by the injury. Specifically, the nutritional condition, oxidative stress, and pyroptosis were improved. A synchronization of BNDF and GDNF expression patterns in various groups indicated the secretion of neurotrophic factors affect the outcome of SCI. The SOD1, CAT, and GSH-px were suppressed after trauma but all restored in response to FBXW7α overexpression. Inflammasomes-activated pyroptosis was incurred after the injury, and relevant biomarkers such as GSDMD, caspase-1, caspase- 11, IL-1β, and IL-18 were down-regulated after the introduction of FBXW7α into the injured cord. Additionally, up-regulating FBXW7α also repaired the mitochondria dysfunction. CONCLUSION Our data indicate FBXW7α probably serves as an important molecular target for the therapy of spinal cord injury.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Trauma Center, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, 222061, China
| | - Tao Yang
- Department of Orthopedics, 4th (Xing Yuan) Hospital of Yulin, Yulin City, Shaanxi Province, 719000, China
| |
Collapse
|
19
|
Peng D, Li J, Deng Y, Zhu X, Zhao L, Zhang Y, Li Z, Ou S, Li S, Jiang Y. Sodium para-aminosalicylic acid inhibits manganese-induced NLRP3 inflammasome-dependent pyroptosis by inhibiting NF-κB pathway activation and oxidative stress. J Neuroinflammation 2020; 17:343. [PMID: 33203418 PMCID: PMC7670624 DOI: 10.1186/s12974-020-02018-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Background The activation of NOD-like receptor protein 3 (NLRP3) inflammasome-dependent pyroptosis has been shown to play a vital role in the pathology of manganese (Mn)-induced neurotoxicity. Sodium para-aminosalicylic acid (PAS-Na) has a positive effect on the treatment of manganism. However, the mechanism is still unclear. We hypothesized that PAS-Na might act through NLRP3. Methods The microglial cell line BV2 and male Sprague-Dawley rats were used to investigate the impacts of PAS-Na on Mn-induced NLRP3 inflammasome-dependent pyroptosis. The related protein of the NF-κB pathway and NLRP3-inflammasome-dependent pyroptosis was detected by western blot. The reactive oxygen species and mitochondrial membrane potential were detected by immunofluorescence staining and flow cytometry. The activation of microglia and the gasdermin D (GSDMD) were detected by immunofluorescence staining. Results Our results showed that Mn treatment induced oxidative stress and activated the NF-κB pathway by increasing the phosphorylation of p65 and IkB-α in BV2 cells and in the basal ganglia of rats. PAS-Na could alleviate Mn-induced oxidative stress damage by inhibiting ROS generation, increasing mitochondrial membrane potential and ATP levels, thereby reducing the phosphorylation of p65 and IkB-α. Besides, Mn treatment could activate the NLRP3 pathway and promote the secretion of IL-18 and IL-1β, mediating pyroptosis in BV2 cells and in the basal ganglia and hippocampus of rats. But an inhibitor of NF-κb (JSH-23) treatment could significantly reduce LDH release, the expression of NLRP3 and Cleaved CASP1 protein and IL-1β and IL-18 mRNA level in BV2 cells. Interestingly, the effect of PAS-Na treatment in Mn-treated BV2 cells is similar to those of JSH-23. Besides, immunofluorescence results showed that PAS-Na reduced the increase number of activated microglia, which stained positively for GSDMD. Conclusion PAS-Na antagonized Mn-induced NLRP3 inflammasome dependent pyroptosis through inhibiting NF-κB pathway activation and oxidative stress. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-020-02018-6.
Collapse
Affiliation(s)
- Dongjie Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Junyan Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Yue Deng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Xiaojuan Zhu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Lin Zhao
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Yuwen Zhang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Zhaocong Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Shiyan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Shaojun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China. .,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.
| | - Yueming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China. .,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.
| |
Collapse
|
20
|
Qi Z, Yang X, Sang Y, Liu Y, Li J, Xu B, Liu W, He M, Xu Z, Deng Y, Zhu J. Fluoxetine and Riluzole Mitigates Manganese-Induced Disruption of Glutamate Transporters and Excitotoxicity via Ephrin-A3/GLAST-GLT-1/Glu Signaling Pathway in Striatum of Mice. Neurotox Res 2020; 38:508-523. [PMID: 32472497 DOI: 10.1007/s12640-020-00209-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/09/2020] [Accepted: 04/14/2020] [Indexed: 01/05/2023]
Abstract
Manganese (Mn) is an essential element required for many biological processes and systems in the human body. Mn intoxication increases brain glutamate (Glu) levels causing neuronal damage. Recent studies have reported that ephrin-A3 regulates this glutamate transporter. However, none has explored the role of this crucial molecule in Mn-induced excitotoxicity. The present study investigated whether ephrin-A3/GLAST-GLT-1/Glu signaling pathway participates in Mn-induced excitotoxicity using astrocytes and Kunming mice. The mechanisms were explored using fluoxetine (ephrin-A3 inhibitor) and riluzole (a Glu release inhibitor). Firstly, we demonstrated that Mn exposure (500 μM or 50 mg/kg MnCl2) significantly increased Mn, ephrin-A3, and Glu levels, and inhibited Na+-K+ ATPase activity, as well as mRNA and protein levels of GLAST and GLT-1. Secondly, we found that astrocytes and mice pretreated with fluoxetine (100 μM or 15 mg/kg) and riluzole (100 μM or 32 μmol/kg) prior to Mn exposure had lower ephrin-A3 and Glu levels, but higher Na+-K+ ATPase activity, expression levels of GLAST and GLT-1 than those exposed to 500 μM or 50 mg/kg MnCl2. Moreover, the morphology of cells and the histomorphology of mice striatum were injured. Results showed that pretreatment with fluoxetine and riluzole attenuated the Mn-induced motor dysfunctions. Together, these results suggest that the ephrin-A3/GLAST-GLT-1/Glu signaling pathway participates in Mn-induced excitotoxicity, and fluoxetine and riluzole can mitigate the Mn-induced excitotoxicity in mice brain.
Collapse
Affiliation(s)
- Zhipeng Qi
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Xinxin Yang
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Yanqi Sang
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Yanan Liu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Jiashuo Li
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Miao He
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Zhaofa Xu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China.
| | - Jinghai Zhu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China.
| |
Collapse
|
21
|
Yang Y, Wei F, Wang J, Chen R, Zhang J, Li D, Gan D, Yang X, Zou Y. Manganese modifies Neurotrophin-3 (NT3) and its tropomyosin receptor kinase C (TrkC) in the cortex: Implications for manganese-induced neurotoxicity. Food Chem Toxicol 2019; 135:110925. [PMID: 31676349 DOI: 10.1016/j.fct.2019.110925] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 10/07/2019] [Accepted: 10/24/2019] [Indexed: 12/26/2022]
Abstract
Manganese (Mn), an essential micronutrient, has the potential to induce apoptosis. The NT3/TrkC ligand/receptor pair known as part of the classic neurotrophic theory plays a critical role in neuronal survival. However, whether the NT3/TrkC-mediated signaling pathways are involved in Mn-induced apoptosis of cortical neurons remains unknown. The present study was designed to investigate the interactions between NT3/TrkC-mediated signaling pathways and Mn-induced apoptosis in cortical neurons. This study showed that subacute Mn exposure significantly increased the levels of pro-apoptotic Bax while decreasing the levels of anti-apoptotic Bcl 2 in the cortex compared with the corresponding control. Markedly reduced NT3 and TrkC levels along with decreased Ras/MAPK and PI3/Akt signaling in the cortex were observed following subacute Mn exposure. We further found increased levels of Bax, cleaved caspase-3, and the total apoptosis rate, and decreased levels of Bcl 2, NT3, TrkC, and Ras/MAPK and PI3/Akt signaling in Mn-treated primary cortical neurons. Pretreatment with hNT3 or Z-VAD-FAM ameliorated Mn-induced apoptosis by increasing the levels of NT3 and TrkC and its Ras/MAPK and PI3/Akt signaling pathways. Taken together, our findings clearly indicate that NT3/TrkC and mediated Ras/MAPK and PI3/Akt signaling pathways play a crucial role in Mn-induced neurotoxicity.
Collapse
Affiliation(s)
- Yiping Yang
- 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
| | - Fu Wei
- Center for Reproductive Medicine and Genetics, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Jian Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Rui Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Jie Zhang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Danni Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Dong Gan
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, China.
| | - Yunfeng Zou
- 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.
| |
Collapse
|
22
|
Peng DJ, Zhang YW, Li ZC, Li SJ, Cai M, Qin WX, Ou SY, Huang XW, Yuan ZX, Jiang YM. Preventive impacts of PAS-Na on the slow growth and activated inflammatory responses in Mn-exposed rats. J Trace Elem Med Biol 2019; 54:134-141. [PMID: 31109603 DOI: 10.1016/j.jtemb.2019.04.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/26/2019] [Accepted: 04/23/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Sodium para-aminosalicylic acid (PAS-Na), an anti-tuberculosis drug, has been demonstrated its function in facilitating the Mn elimination in manganism patients and Mn-exposed models in vivo and improving the symptoms of Mn poisoning. But whether it can improve the growth retardation and inflammatory responses induced by Mn have not been reported. OBJECTIVES This study was designed to investigate the preventive effects of PAS-Na on the development of retardation and inflammatory responses in Mn-exposed rats. METHODS Male Sprague Dawley (SD) rats (8 weeks old, weighing 180 ± 20 g) were randomly divided into normal control group and Mn-exposed group in the 4 weeks experiment observation and normal control group, Mn-exposed group, PAS-Na preventive group and PAS-Na control group in the 8 weeks experiment observation. The Mn-exposed group received an intraperitoneal injection (i.p.) of 15 mg/kg MnCl2 and the normal control group i.p. physiological Saline in the same volume once a day for 4 or 8 weeks, 5 days per week. The PAS-Na preventive group i.p. 15 mg/kg MnCl2 along with back subcutaneous (s.c.) injection of 240 mg/kg PAS-Na once a day for 8 weeks, 5 days per week. PAS-Na control group received s.c. injection of 240 mg/kg PAS-Na along with i.p. injection of saline once daily. The body weight was determined once a week until the end of the experiment. The manganese contents in the blood were detected by graphite furnace atomic absorption spectrometry. The inflammatory factor levels (TNF-α, IL-1β, IL-6, and PGE2) in the blood were detected by using enzyme-linked immunosorbent assay (Elisa) and each organ taking from rats were weighed and recorded. RESULTS Mn exposure significantly suppressed the growth in rats and increased heart, liver, spleen and kidney coefficients as compared with the control group. The whole blood Mn level and serum levels of IL-1β, IL-6, PGE2, and TNF-α in sub-chronic Mn-exposure group were markedly higher than those in the control group. However, preventive treatment with PAS-Na obviously reduced the whole blood Mn level, the spleen and liver coefficients of the Mn-exposed rats. And serum levels of IL-1β and TNF-α were significantly reduced by 33.9% and 14.7% respectively in PAS-Na prevention group. CONCLUSIONS PAS-Na could improve the growth retardation and alleviate inflammatory responses in Mn-exposed rats.
Collapse
Affiliation(s)
- Dong-Jie Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Yu-Wen Zhang
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Zhao-Cong Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Shao-Jun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Mei Cai
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China.
| | - Wen-Xia Qin
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Department of Child Health, Guilin Women and Children Hospital, Guilin, China.
| | - Shi-Yan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Xiao-Wei Huang
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Zong-Xiang Yuan
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, China.
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| |
Collapse
|
23
|
Lan H, Su Y, Liu Y, Deng C, Wang J, Chen T, Jules KED, Masau JF, Li H, Wei X. Melatonin protects circulatory death heart from ischemia/reperfusion injury via the JAK2/STAT3 signalling pathway. Life Sci 2019; 228:35-46. [DOI: 10.1016/j.lfs.2019.04.057] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023]
|
24
|
Durappanavar PN, Nadoor P, Waghe P, Pavithra BH, Jayaramu GM. Melatonin Ameliorates Neuropharmacological and Neurobiochemical Alterations Induced by Subchronic Exposure to Arsenic in Wistar Rats. Biol Trace Elem Res 2019; 190:124-139. [PMID: 30306420 DOI: 10.1007/s12011-018-1537-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023]
Abstract
An experimental study was conducted in Wistar rats to characterize the arsenic ("As")-induced alterations in neurobiochemistry in brain and its impact on neuropharmacological activities with or without the melatonin (MLT) as an antioxidant given exogenously. Male Wistar rats were randomly divided in to four groups of six each. Group I served as untreated control, while group II received As [sodium (meta) arsenite; NaAsO2] at 10 mg/kg bw (p.o.) for a period of 56 days. Experimental rats in group III received treatment similar to group II but in addition received MLT at 10 mg/kg bw (p.o.) from day 32 onwards. Rats in group IV received MLT alone from day 32 onwards similar to group III. Sub-chronic exposure to As (group II) significantly reduced both voluntary locomotor and forced motor activities and melatonin supplementation (group III) showed a significant improvement in motor activities, when subjected to test on day 42 or 56. Rats exposed to As showed a significant increase in anxiety level and a marginal nonsignificant reduction in pain latency. Sub-chronic administration of As induced (group II) significant increase in the levels of thiobarbituric acid reactive substance (TBARS) called malondialdehyde (MDA) in the brain tissue (5.55 ± 0.57 nmol g-1), and their levels were significantly reduced by MLT supplementation (group III 3.96 ± 0.15 nmol g-1). The increase in 3-nitrotyrosine (3-NT) levels in As-exposed rats indicated nitrosative stress due to the formation of peroxynitrite (ONOO-). However, exogenously given MLT significantly reduced the 3-NT formation as well as prostaglandin (PGE2) levels in the brain. Similarly, MLT administration have suppressed the release of pro-inflammatory cytokines (viz., IL-1β, IL-6, and TNF-α) and amyloid-β1-40 (Aβ) deposition in the brain tissues of experimental rats. To conclude, exogenous administration of melatonin can overcome the sub-chronic As-induced oxidative and nitrosative stress in the CNS, suppressed pro-inflammatory cytokines, and restored certain disturbed neuropharmacological activities in Wistar rats.
Collapse
Affiliation(s)
- Prasada Ningappa Durappanavar
- Department of Veterinary Pharmacology and Toxicology; Karnataka Veterinary, Animal and Fisheries Sciences University; Veterinary College, Vinobanagar, Shivamogga, Karnataka, 577 204, India
| | - Prakash Nadoor
- Department of Veterinary Pharmacology and Toxicology; Karnataka Veterinary, Animal and Fisheries Sciences University, Veterinary College, Veterinary College, Hebbal, Bengaluru, Karnataka, 560 024, India.
| | - Prashantkumar Waghe
- Department of Veterinary Pharmacology and Toxicology Veterinary College, Nandinagar, Bidar, Karnataka, 585401, India
| | - B H Pavithra
- Department of Veterinary Pharmacology and Toxicology; Karnataka Veterinary, Animal and Fisheries Sciences University, Veterinary College, Veterinary College, Hebbal, Bengaluru, Karnataka, 560 024, India
| | - G M Jayaramu
- Department of Veterinary Pathology, Karnataka Veterinary, Animal and Fisheries Sciences University, Veterinary College, Vinobanagar, Shivamogga, Karnataka, 577 204, India
| |
Collapse
|
25
|
Ommati MM, Heidari R, Ghanbarinejad V, Aminian A, Abdoli N, Niknahad H. The neuroprotective properties of carnosine in a mouse model of manganism is mediated via mitochondria regulating and antioxidative mechanisms. Nutr Neurosci 2019; 23:731-743. [PMID: 30856059 DOI: 10.1080/1028415x.2018.1552399] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective(s): Manganese (Mn) is an essential trace element physiologically incorporated in the structure of several vital enzymes. Despite its essentiality, excessive Mn exposure is toxic with brain tissue as the primary target organ. There is no specific and clinically available therapeutic/preventive option against Mn neurotoxicity. Carnosine is a neuropeptide with several physiological roles. The neuroprotective properties of this peptide have been evaluated in different experimental models. The current study was designed to investigate the effect of carnosine supplementation and its potential mechanisms of action in an animal model of Mn-induced neurotoxicity. Materials and Methods: Male C57BL/6 mice received Mn (100 mg/kg, s.c) alone and/or in combination with carnosine (10, 50, and 100 mg/kg, i.p). Several locomotor activity indices were monitored. Moreover, biomarkers of oxidative stress and mitochondrial function were assessed in the brain tissue of Mn-exposed animals. Results: Significant locomotor dysfunction was revealed in Mn-exposed animals. Furthermore, brain tissue biomarkers of oxidative stress were significantly increased, and mitochondrial indices of functionality were impaired in Mn-treated animals. It was found that carnosine supplementation (10, 50, and 100 mg/kg, i.p) alleviated the Mn-induced locomotor deficit. Moreover, this peptide mitigated oxidative stress biomarkers and preserved brain tissue mitochondrial functionality in the animal model of manganism. Conclusion: These data indicate that carnosine is a potential neuroprotective agent against Mn neurotoxicity. Antioxidative and mitochondria protecting effects of carnosine might play a fundamental role in its neuroprotective properties against Mn toxicity.
Collapse
Affiliation(s)
- Mohammad Mehdi Ommati
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Bioinformatics, College of Life Sciences, Shanxi Agricultural University, Taigu, Peoples' Republic of China
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Ghanbarinejad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmadreza Aminian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Abdoli
- Iran Food and Drug Administration (IFDA), Ministry of Health, Tehran, Iran
| | - Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
26
|
Doğanlar ZB, Uzun M, Ovali MA, Dogan A, Ongoren G, Doğanlar O. Melatonin attenuates caspase-dependent apoptosis in the thoracic aorta by regulating element balance and oxidative stress in pinealectomised rats. Appl Physiol Nutr Metab 2019; 44:153-163. [DOI: 10.1139/apnm-2018-0205] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The aim of this study was to explain the possible mechanisms by which melatonin deficiency results in cardiovascular injury and to investigate the effects of melatonin administration on important signalling pathways and element equilibrium in the thoracic aorta (TA). For this purpose, we analysed the cellular and molecular effects of melatonin deficiency or administration on oxidative stress, DNA damage, molecular chaperone response, and apoptosis induction in TA tissues of pinealectomised rats using ELISA, RAPD, qRT-PCR, and Western blot assays. The results showed that melatonin deficiency led to an imbalance in essential element levels, unfolded or misfolded proteins, increased lipid peroxidation, and selectively induced caspase-dependent apoptosis in TA tissues without significantly affecting the Bcl-2/BAX ratio (2.28 in pinealectomised rats, 2.73 in pinealectomised rats treated with melatonin). In pinealectomised rats, the genomic template stability (80.22%) was disrupted by the significantly increased oxidative stress, and heat shock protein 70 (20.96-fold), TNF-α (1.73-fold), caspase-8 (2.03-fold), and caspase-3 (2.87-fold) were markedly overexpressed compared with the sham group. Melatonin treatment was protective against apoptosis and inhibited oxidative damage. In addition, melatonin increased the survivin level and improved the regulation of element equilibrium in TA tissues. The results of the study indicate that melatonin deficiency induces TNF-α-related extrinsic apoptosis signals and that the administration of pharmacological doses of melatonin attenuates cardiovascular toxicity by regulating the increase in the rate of apoptosis caused by melatonin deficiency in TA tissue of Sprague–Dawley rats.
Collapse
Affiliation(s)
- Zeynep Banu Doğanlar
- Department of Medical Biology, Faculty of Medicine, Trakya University, 22030 Edirne, Turkey
| | - Metehan Uzun
- Department of Physiology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Çanakkale 17020, Turkey
| | - Mehmet Akif Ovali
- Department of Physiology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Çanakkale 17020, Turkey
| | - Ayten Dogan
- Department of Medical Biology, Faculty of Medicine, Trakya University, 22030 Edirne, Turkey
| | - Gulin Ongoren
- Department of Medical Biology, Faculty of Medicine, Trakya University, 22030 Edirne, Turkey
| | - Oğuzhan Doğanlar
- Department of Medical Biology, Faculty of Medicine, Trakya University, 22030 Edirne, Turkey
| |
Collapse
|
27
|
Cheng H, Xia B, Su C, Chen K, Chen X, Chen P, Zou Y, Yang X. PI3K/Akt signaling pathway and Hsp70 activate in hippocampus of rats with chronic manganese sulfate exposure. J Trace Elem Med Biol 2018; 50:332-338. [PMID: 30262300 DOI: 10.1016/j.jtemb.2018.07.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/15/2018] [Accepted: 07/23/2018] [Indexed: 12/11/2022]
Abstract
Manganese (Mn) has come to the forefront of environmental concerns due to its neurotoxicity. However, the toxic effect of Mn is not fully understood. The purpose of this study is to investigate the impacts of chronic manganese sulfate (MnSO4) exposure in regulating the phosphatidylinositol 3 kinase/protein kinase B (PI3K/Akt) signaling pathway in rats. In this study, rats were treated with 0, 5.0, 10.0, and 20.0 mg/kg MnSO4•H2O five days a week for 24 weeks via intraperitoneal injection. At the end of the exposure period, the levels of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT), malondialdehyde (MDA), and heat shock protein (Hsp70) in rats' plasma were quantified; the mRNA expression levels of caspase-3, B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax), serine-threonine protein kinase (Akt-1), and forkhead box O3a (FoxO3a) were measured through real-time quantitative PCR (RT-PCR); and the levels of protein Hsp70 and Akt were assessed by western blot. With an increasing dose of MnSO4, the organ coefficients of all tested organs were significantly increased, except the testis. Compared with the control group, the activities of plasma SOD, GSH-Px, and CAT in MnSO4-exposed groups were significantly decreased, while the concentrations of plasma MDA and Hsp70 were significantly increased. Moreover, the hippocampal mRNA levels of Bcl-2, caspase-3, Akt-1, and FoxO3a in MnSO4-exposed groups were downregulated, but the level of Bax was upregulated. Meanwhile, the level of phosphorylation of Akt (p-Akt) and Hsp70 proteins tends to be upregulated by increasing MnSO4 exposure (P < 0.05). The plasma Hsp70 level was negatively associated with SOD, CAT, and GSH-Px activities (P < 0.05), and positively associated with blood MDA concentration and hippocampal Hsp70 levels (P < 0.05). Chronic MnSO4 exposure can result in apoptosis of central nerve cells, activate the PI3K/Akt signaling pathway in rats' hippocampus, and upregulate Hsp70 transcription and translation.
Collapse
Affiliation(s)
- Hong Cheng
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Bing Xia
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Cheng Su
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Kangcheng Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiang Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, United States
| | - Yunfeng Zou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, Guangxi, China; Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| |
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: 3.1] [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
|
Ma Z, Xin Z, Di W, Yan X, Li X, Reiter RJ, Yang Y. Melatonin and mitochondrial function during ischemia/reperfusion injury. Cell Mol Life Sci 2017; 74:3989-3998. [PMID: 28795196 PMCID: PMC11107672 DOI: 10.1007/s00018-017-2618-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023]
Abstract
Ischemia/reperfusion (IR) injury occurs in many organs and tissues, and contributes to morbidity and mortality worldwide. Melatonin, an endogenously produced indolamine, provides a strong defense against IR injury. Mitochondrion, an organelle for ATP production and a decider for cell fate, has been validated to be a crucial target for melatonin to exert its protection against IR injury. In this review, we first clarify the mechanisms underlying mitochondrial dysfunction during IR and melatonin's protection of mitochondria under this condition. Thereafter, special focus is placed on the protective actions of melatonin against IR injury in brain, heart, liver, and others. Finally, we explore several potential future directions of research in this area. Collectively, the information compiled here will serve as a comprehensive reference for the actions of melatonin in IR injury identified to date and will hopefully aid in the design of future research and increase the potential of melatonin as a therapeutic agent.
Collapse
Affiliation(s)
- Zhiqiang Ma
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, UT Health San Antonio, 229 Taibai North Road, Xi'an, 710069, China
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710038, China
| | - Zhenlong Xin
- Department of Biomedical Engineering, Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Wencheng Di
- Department of Cardiology, Affiliated Drum Tower Hospital, Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710038, China
| | - Xiaofei Li
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710038, China
| | - Russel J Reiter
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, UT Health San Antonio, 229 Taibai North Road, Xi'an, 710069, China.
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, UT Health San Antonio, 229 Taibai North Road, Xi'an, 710069, China.
- Department of Cellular and Structural Biology, UT Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
| |
Collapse
|
30
|
Clinical effects of chemical exposures on mitochondrial function. Toxicology 2017; 391:90-99. [PMID: 28757096 DOI: 10.1016/j.tox.2017.07.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/07/2017] [Accepted: 07/17/2017] [Indexed: 12/16/2022]
Abstract
Mitochondria are critical for the provision of ATP for cellular energy requirements. Tissue and organ functions are dependent on adequate ATP production, especially when energy demand is high. Mitochondria also play a role in a vast array of important biochemical pathways including apoptosis, generation and detoxification of reactive oxygen species, intracellular calcium regulation, steroid hormone and heme synthesis, and lipid metabolism. The complexity of mitochondrial structure and function facilitates its diverse roles but also enhances its vulnerability. Primary disorders of mitochondrial bioenergetics, or Primary Mitochondrial Diseases (PMD) are due to inherited genetic defects in the nuclear or mitochondrial genomes that result in defective oxidative phosphorylation capacity and cellular energy production. Secondary mitochondrial dysfunction is observed in a wide range of diseases such as Alzheimer's and Parkinson's disease. Several lines of evidence suggest that environmental exposures cause substantial mitochondrial dysfunction. Whereby literature from experimental and human studies on exposures associated with Alzheimer's and Parkinson's diseases exist, the significance of exposures as potential triggers in Primary Mitochondrial Disease (PMD) is an emerging clinical question that has not been systematically studied.
Collapse
|
31
|
Smith MR, Fernandes J, Go YM, Jones DP. Redox dynamics of manganese as a mitochondrial life-death switch. Biochem Biophys Res Commun 2017; 482:388-398. [PMID: 28212723 PMCID: PMC5382988 DOI: 10.1016/j.bbrc.2016.10.126] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/25/2016] [Accepted: 10/27/2016] [Indexed: 12/16/2022]
Abstract
Sten Orrenius, M.D., Ph.D., pioneered many areas of cellular and molecular toxicology and made seminal contributions to our knowledge of oxidative stress and glutathione (GSH) metabolism, organellar functions and Ca+2-dependent mechanisms of cell death, and mechanisms of apoptosis. On the occasion of his 80th birthday, we summarize current knowledge on redox biology of manganese (Mn) and its role in mechanisms of cell death. Mn is found in all organisms and has critical roles in cell survival and death mechanisms by regulating Mn-containing enzymes such as manganese superoxide dismutase (SOD2) or affecting expression and activity of caspases. Occupational exposures to Mn cause "manganism", a Parkinson's disease-like condition of neurotoxicity, and experimental studies show that Mn exposure leads to accumulation of Mn in the brain, especially in mitochondria, and neuronal cell death occurs with features of an apoptotic mechanism. Interesting questions are why a ubiquitous metal that is essential for mitochondrial function would accumulate to excessive levels, cause increased H2O2 production and lead to cell death. Is this due to the interactions of Mn with other essential metals, such as iron, or with toxic metals, such as cadmium? Why is the Mn loading in the human brain so variable, and why is there such a narrow window between dietary adequacy and toxicity? Are non-neuronal tissues similarly vulnerable to insufficiency and excess, yet not characterized? We conclude that Mn is an important component of the redox interface between an organism and its environment and warrants detailed studies to understand the role of Mn as a mitochondrial life-death switch.
Collapse
Affiliation(s)
- Matthew Ryan Smith
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Jolyn Fernandes
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Young-Mi Go
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
32
|
Xia B, Chen K, Lv Y, Huang D, Liu J, Liang G, Zhang L, Wang F, Su C, Zou Y, Yang X. Increased oxidative stress and plasma Hsp70 levels among gasoline filling station attendants. Toxicol Ind Health 2016; 33:171-181. [PMID: 26792678 DOI: 10.1177/0748233715616554] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Methylcyclopentadienyl manganese tricarbonyl (MMT) is an organic derivative of manganese (Mn) and is used as an antiknock agent and octane enhancer in gasoline. In this article, we tested the oxidative stress and heat stress protein (Hsp) 70 levels of gasoline station attendants to explore potential plasma biomarkers. Furthermore, the dose-response relationship was also identified. METHODS A total of 144 workers, including 96 petrol fillers and 48 cashiers, participated in the study. Ambient concentrations of benzene, toluene, ethylbenzene, and xylene (BTEX) and Mn were monitored at nine filling stations. During the measuring process, the individual cumulative exposure index was calculated. Plasma oxidative stress and Hsp70 levels were also analysed using enzyme-linked immunosorbent assay. RESULTS The BTEX time-weighted average in office areas was significantly lower than in refuelling areas ( p < 0.05). In refuelling areas, the content of Mn ranged from 6.44 μg/m3 to 127.34 μg/m3, which was much higher than that in office areas (3.16-7.22 μg/m3; p < 0.05). Exposed workers had significantly different plasma oxidative stress indicators compared with the control group, respectively: superoxide dismutase (SOD), 39.18 ± 6.05 U/mL versus 52.84 ± 3.87 U/mL; glutathione peroxidase (GSH-Px), 186.07 ± 15.63 U versus 194.38 ± 10.42 U; and malondialdehyde (MDA), 1.68 ± 0.52 nmol/L versus 1.43 ± 0.64 nmol/L (in all comparisons, p < 0.05). Plasma Hsp70 level in the exposed group (2.77 ± 0.64 ng/mL) was significantly higher than in the control group (2.32 ± 0.87 ng/mL; p < 0.05). Furthermore, Hsp70 levels were inversely correlated with the activities of SOD ( r = -0.305) and GSH-Px ( r = -0.302) in the exposed group ( p < 0.05). Moreover, a positive correlation ( r = 0.653) was found between plasma Hsp70 levels and plasma MDA levels ( p < 0.05). CONCLUSION Exposure to MMT-containing gasoline may result in increasing reactive oxygen stress among filling station attendants. Plasma Hsp70 levels could be used as a sensitive responsive biomarker for exposed workers.
Collapse
Affiliation(s)
- Bing Xia
- 1 Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Kangcheng Chen
- 1 Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Yingnan Lv
- 1 Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Damin Huang
- 1 Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Jing Liu
- 1 Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Guiqiang Liang
- 2 Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Li'e Zhang
- 2 Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Fenfen Wang
- 1 Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Cheng Su
- 1 Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Yunfeng Zou
- 2 Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaobo Yang
- 1 Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China.,3 Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
33
|
Li SJ, Li Y, Chen JW, Yuan ZX, Mo YH, Lu GD, Jiang YM, Ou CY, Wang F, Huang XW, Luo YN, Ou SY, Huang YN. Sodium Para-aminosalicylic Acid Protected Primary Cultured Basal Ganglia Neurons of Rat from Manganese-Induced Oxidative Impairment and Changes of Amino Acid Neurotransmitters. Biol Trace Elem Res 2016; 170:357-65. [PMID: 26286965 DOI: 10.1007/s12011-015-0472-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/07/2015] [Indexed: 12/22/2022]
Abstract
Manganese (Mn), an essential trace metal for protein synthesis and particularly neurotransmitter metabolism, preferentially accumulates in basal ganglia. However, excessive Mn accumulation may cause neurotoxicity referred to as manganism. Sodium para-aminosalicylic acid (PAS-Na) has been used to treat manganism with unclear molecular mechanisms. Thus, we aim to explore whether PAS-Na can inhibit Mn-induced neuronal injury in basal ganglia in vitro. We exposed basal ganglia neurons with 50 μM manganese chloride (MnCl2) for 24 h and then replaced with 50, 150, and 450 μM PAS-Na treatment for another 24 h. MnCl2 significantly decreased cell viability but increased leakage rate of lactate dehydrogenase and DNA damage (as shown by increasing percentage of DNA tail and Olive tail moment). Mechanically, Mn reduced glutathione peroxidase and catalase activity and interrupted amino acid neurotransmitter balance. However, PAS-Na treatment reversed the aforementioned Mn-induced toxic effects. Taken together, these results showed that PAS-Na could protect basal ganglia neurons from Mn-induced neurotoxicity.
Collapse
Affiliation(s)
- Shao-Jun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Yong Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Jing-Wen Chen
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Zong-Xiang Yuan
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Yu-Huan Mo
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Guo-Dong Lu
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| | - Chao-Yan Ou
- Department of Toxicology, School of Public Health, Guilin Medical University, Guilin, 541004, China
| | - Fang Wang
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Xiao-Wei Huang
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Yi-Ni Luo
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Shi-Yan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Yan-Ni Huang
- Department of Toxicology, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| |
Collapse
|
34
|
Paulose JK, Wright JM, Patel AG, Cassone VM. Human Gut Bacteria Are Sensitive to Melatonin and Express Endogenous Circadian Rhythmicity. PLoS One 2016; 11:e0146643. [PMID: 26751389 PMCID: PMC4709092 DOI: 10.1371/journal.pone.0146643] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/21/2015] [Indexed: 12/29/2022] Open
Abstract
Circadian rhythms are fundamental properties of most eukaryotes, but evidence of biological clocks that drive these rhythms in prokaryotes has been restricted to Cyanobacteria. In vertebrates, the gastrointestinal system expresses circadian patterns of gene expression, motility and secretion in vivo and in vitro, and recent studies suggest that the enteric microbiome is regulated by the host's circadian clock. However, it is not clear how the host's clock regulates the microbiome. Here, we demonstrate at least one species of commensal bacterium from the human gastrointestinal system, Enterobacter aerogenes, is sensitive to the neurohormone melatonin, which is secreted into the gastrointestinal lumen, and expresses circadian patterns of swarming and motility. Melatonin specifically increases the magnitude of swarming in cultures of E. aerogenes, but not in Escherichia coli or Klebsiella pneumoniae. The swarming appears to occur daily, and transformation of E. aerogenes with a flagellar motor-protein driven lux plasmid confirms a temperature-compensated circadian rhythm of luciferase activity, which is synchronized in the presence of melatonin. Altogether, these data demonstrate a circadian clock in a non-cyanobacterial prokaryote and suggest the human circadian system may regulate its microbiome through the entrainment of bacterial clocks.
Collapse
Affiliation(s)
- Jiffin K. Paulose
- Department of Biology, University of Kentucky, Lexington, KY, 40506, United States of America
| | - John M. Wright
- Department of Biology, University of Kentucky, Lexington, KY, 40506, United States of America
| | - Akruti G Patel
- Department of Biology, University of Kentucky, Lexington, KY, 40506, United States of America
| | - Vincent M. Cassone
- Department of Biology, University of Kentucky, Lexington, KY, 40506, United States of America
| |
Collapse
|
35
|
Manganese-Disrupted Interaction of Dopamine D1 and NMDAR in the Striatum to Injury Learning and Memory Ability of Mice. Mol Neurobiol 2015; 53:6745-6758. [PMID: 26660110 DOI: 10.1007/s12035-015-9602-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 12/01/2015] [Indexed: 10/22/2022]
Abstract
Manganese (Mn) is widely regarded as a neurotoxic heavy metal that causes learning and memory deficits. Recently, it has been proved that the striatum is related to memory and learning ability. However, no previous study focused on the effect of Mn-induced learning and memory deficits on the striatum. This study aims to investigate the probable interaction of dopamine D1 receptor (DR1) and N-methyl-D-aspartate receptor (NMDAR), two cognition-related receptors in the striatum during Mn exposure. Mice are randomly divided into four groups, including control group, 12.5 mg/kg MnCl2 group, 25 mg/kg MnCl2 group, and 50 mg/kg MnCl2 group. The mice receive intraperitoneal injections of 0, 12.5, 25, and 50 mg/kg MnCl2 once daily for 2 weeks. Then, learning and memory ability, pathological changes, expression, and interaction of DR1 and NMDAR are determined. It has been found that Mn disrupted spatial learning and memory ability of mice by Morris water maze test and the passive avoidance test. Pathological and ultrastructure were injured. Mn decreased the immunohistochemical activities, protein levels, and messenger RNA (mRNA) expression of DR1, NR1, and NR2A. Mn exposure inhibited interaction between DR1 and NMDAR in striatum by double immunofluorescent staining and co-immunoprecipitation. In conclusion, our study illustrated that Mn caused learning and memory dysfunction via injury of striatum and inhibition of interaction between DR1 and NMDAR in striatum.
Collapse
|
36
|
|
37
|
Tsujii S, Ishisaka M, Hara H. Modulation of endoplasmic reticulum stress in Parkinson's disease. Eur J Pharmacol 2015; 765:154-6. [DOI: 10.1016/j.ejphar.2015.08.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 10/23/2022]
|
38
|
Pomara C, Neri M, Bello S, Fiore C, Riezzo I, Turillazzi E. Neurotoxicity by synthetic androgen steroids: oxidative stress, apoptosis, and neuropathology: A review. Curr Neuropharmacol 2015; 13:132-45. [PMID: 26074748 PMCID: PMC4462038 DOI: 10.2174/1570159x13666141210221434] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/30/2014] [Accepted: 10/25/2014] [Indexed: 12/25/2022] Open
Abstract
Anabolic-androgenic steroids (AAS) are synthetic substances derived from testosterone that are largely employed due to their trophic effect on muscle tissue of athletes at all levels. Since a great number of organs and systems are a target of AAS, their adverse effects are primarily on the following systems: reproductive, hepatic, musculoskeletal, endocrine, renal, immunological, infectious, cardiovascular, cerebrovascular, and hematological. Neuropsychiatric and behavioral effects as a result of AAS abuse are well known and described in the literature. Mounting evidence exists suggesting that in addition to psychiatric and behavioral effects, non-medical use of AAS carries neurodegenerative potential. Although, the nature of this association remains largely unexplored, recent animal studies have shown the recurrence of this AAS effect, ranging from neurotrophin unbalance to increased neuronal susceptibility to apoptotic stimuli. Experimental and animal studies strongly suggest that apoptotic mechanisms are at least in part involved in AAS-induced neurotoxicity. Furthermore, a great body of evidence is emerging suggesting that increased susceptibility to cellular oxidative stress could play a pivotal role in the pathogenesis of many neurodegenerative disorders and cognitive impairment. As in other drug-evoked encephalopathies, the key mechanisms involved in AAS - induced neuropathology could represent a target for future neuroprotective strategies. Progress in the understanding of these mechanisms will provide important insights into the complex pathophysiology of AAS-induced neurodegeneration, and will pave the way for forthcoming studies. Supplementary to abandoning the drug abuse that represents the first step in reducing the possibility of irreversible brain damage in AAS abusers, neuroprotective strategies have to be developed and implemented in future.
Collapse
Affiliation(s)
- Cristoforo Pomara
- Institute of Legal Medicine, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
- Department of Anatomy, University of Malta. Msida, Malta
| | - Margherita Neri
- Institute of Legal Medicine, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Stefania Bello
- Institute of Legal Medicine, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Carmela Fiore
- Institute of Legal Medicine, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Irene Riezzo
- Institute of Legal Medicine, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Emanuela Turillazzi
- Institute of Legal Medicine, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| |
Collapse
|