51
|
Vlasak T, Dujlovic T, Barth A. Manganese exposure and cognitive performance: A meta-analytical approach. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023:121884. [PMID: 37247766 DOI: 10.1016/j.envpol.2023.121884] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/09/2023] [Accepted: 05/23/2023] [Indexed: 05/31/2023]
Abstract
Occupational manganese exposure is associated with serious health concerns, ultimately leading to an illness called manganism. Competing meta-analytic results were published over a decade ago, ranging from undetectable to serious effects on cognitive performance among working adults. Novel studies and findings about the relationship between occupational manganese and cognitive functions have been proposed since. First of all, a systematic literature search was carried out until October 2022 via multiple electronic databases investigating the relationship between occupational manganese exposure and cognitive functions. Differences between the exposure and control groups in cognitive testing were synthesized by effect size Hedge's g. A random effects model was deployed with a restricted likelihood estimator using Hedges' invariance weighting. Publication bias, p-hacking and exposure-effect relationships were investigated. We included 18 studies with 75 effect sizes comparing n = 888 controls and 1092 exposed participants. After exclusion of outliers, we found significantly lower performances in processing speed, attention, working memory, reaction time, cognitive control and visual attention in workers exposed to manganese. Regression analysis revealed an indication of exposure-effect relationships between manganese exposure and cognitive functioning in exposed workers. We provide results of impaired cognitive functions for working adults exposed to manganese in processing speed, attention, working memory, reaction time and visual attention. Indications of quadratic exposure-effect relationships are discussed. We provide several recommendations for further studies to investigate possible exposure effects in the context of occupational health and safety.
Collapse
Affiliation(s)
- Thomas Vlasak
- Department of Psychology, Sigmund Freud Private University Linz, Linz, Austria
| | - Tanja Dujlovic
- Department of Psychology, Sigmund Freud Private University Linz, Linz, Austria
| | - Alfred Barth
- Department of Psychology, Sigmund Freud Private University Linz, Linz, Austria.
| |
Collapse
|
52
|
Meng CY, Ma XY, Xu MY, Pei SF, Liu Y, Hao ZL, Li QZ, Feng FM. Transcriptomics-based investigation of manganese dioxide nanoparticle toxicity in rats' choroid plexus. Sci Rep 2023; 13:8510. [PMID: 37231062 PMCID: PMC10213021 DOI: 10.1038/s41598-023-35341-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 05/16/2023] [Indexed: 05/27/2023] Open
Abstract
Manganese dioxide nanoparticles (MnO2-NPs) have a wide range of applications in biomedicine. Given this widespread usage, it is worth noting that MnO2-NPs are definitely toxic, especially to the brain. However, the damage caused by MnO2-NPs to the choroid plexus (CP) and to the brain after crossing CP epithelial cells has not been elucidated. Therefore, this study aims to investigate these effects and elucidate potential underlying mechanisms through transcriptomics analysis. To achieve this objective, eighteen SD rats were randomly divided into three groups: the control group (control), low-dose exposure group (low-dose) and high-dose exposure group (high-dose). Animals in the two treated groups were administered with two concentrations of MnO2-NPs (200 mg kg-1 BW and 400 mg kg-1 BW) using a noninvasive intratracheal injection method once a week for three months. Finally, the neural behavior of all the animals was tested using a hot plate tester, open-field test and Y-type electric maze. The morphological characteristics of the CP and hippocampus were observed by H&E stain, and the transcriptome of CP tissues was analysed by transcriptome sequencing. The representative differentially expressed genes were quantified by qRT-PCR. We found that treatment with MnO2-NPs could induce learning capacity and memory faculty decline and destroy the structure of hippocampal and CP cells in rats. High doses of MnO2-NPs had a more obvious destructive capacity. For transcriptomic analysis, we found that there were significant differences in the numbers and types of differential genes in CP between the low- and high-dose groups compared to the control. Through GO terms and KEGG analysis, high-dose MnO2-NPs significantly affected the expression of transporters, ion channel proteins, and ribosomal proteins. There were 17 common differentially expressed genes. Most of them were transporter and binding genes on the cell membrane, and some of them had kinase activity. Three genes, Brinp, Synpr and Crmp1, were selected for qRT-PCR to confirm their expression differences among the three groups. In conclusion, high-dose MnO2-NPs exposure induced abnormal neurobehaviour, impaired memory function, destroyed the structure of the CP and changed its transcriptome in rats. The most significant DEGs in the CP were within the transport system.
Collapse
Affiliation(s)
- Chun-Yan Meng
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, People's Republic of China
| | - Xin-Yi Ma
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, People's Republic of China
| | - Ming-Yan Xu
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, People's Republic of China
| | - Sheng-Fei Pei
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, People's Republic of China
| | - Yang Liu
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, People's Republic of China
| | - Zhuo-Lu Hao
- School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Qing-Zhao Li
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, People's Republic of China
| | - Fu-Min Feng
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, People's Republic of China.
- College of Life Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, People's Republic of China.
| |
Collapse
|
53
|
Zhou S, Chai P, Dong X, Liang Z, Yang Z, Li J, Teng G, Sun S, Xu M, Zheng ZJ, Wang J, Zhang Z, Chen K. Drinking water quality and inflammatory bowel disease: a prospective cohort study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023:10.1007/s11356-023-27460-w. [PMID: 37160856 DOI: 10.1007/s11356-023-27460-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/02/2023] [Indexed: 05/11/2023]
Abstract
Environmental factors, such as drinking water and diets, play an important role in the development of inflammatory bowel disease (IBD). This study aimed to investigate the associations of metal elements and disinfectants in drinking water with the risk of inflammatory bowel disease (IBD) and to assess whether diet influences these associations. We conducted a prospective cohort study including 22,824 participants free from IBD from the Yinzhou cohort study in the 2016-2022 period with an average follow-up of 5.24 years. The metal and disinfectant concentrations were measured in local pipeline terminal tap water samples. Cox regression models adjusted for multi-level covariates were used to estimate adjusted hazard ratios (aHRs) and 95% confidence intervals (95% CIs). During an average follow-up period of 5.24 years, 46 cases of IBD were identified. For every 1 standard deviation (SD) increase in the concentration of manganese, mercury, selenium, sulfur tetraoxide (SO4), chlorine, and nitrate nitrogen (NO3_N) were associated with a higher risk of IBD with the HRs of 1.45 (95% CI: 1.14 to 1.84), 1.51 (95% CI: 1.24-1.82), 1.29 (95% CI: 1.03-1.61), 1.52 (95% CI: 1.26-1.83), 1.26 (95% CI: 1.18-1.34), and 1.66 (95% CI: 1.32-2.09), whereas zinc and fluorine were inversely associated with IBD with the HRs of 0.42 (95% CI: 0.24 to 0.73) and 0.68 (95% CI: 0.54-0.84), respectively. Stronger associations were observed in females, higher income groups, low education groups, former drinkers, and participants who never drink tea. Diets have a moderating effect on the associations of metal and nonmetal elements with the risk of IBD. We found significant associations between exposure to metals and disinfectants and IBD. Diets regulated the associations to some extent.
Collapse
Affiliation(s)
- Shuduo Zhou
- Department of Global Health, School of Public Health, Peking University, 38 Xue Yuan Road, Haidian District, Beijing, 100191, China
- Institute for Global Health and Development, Peking University, Beijing, China
| | - Pengfei Chai
- The Center for Disease Control and Prevention of Yinzhou District, Ningbo, Zhejiang, China
| | - Xuejie Dong
- Department of Global Health, School of Public Health, Peking University, 38 Xue Yuan Road, Haidian District, Beijing, 100191, China
- Institute for Global Health and Development, Peking University, Beijing, China
| | - Zhisheng Liang
- Department of Global Health, School of Public Health, Peking University, 38 Xue Yuan Road, Haidian District, Beijing, 100191, China
| | - Zongming Yang
- Department of Epidemiology and Biostatistics, Zhejiang University School of Public Health, Hangzhou, 310058, China
| | - Junxia Li
- Department of Gastroenterology, Peking University First Hospital, Beijing, 100034, China
| | - Guigen Teng
- Department of Gastroenterology, Peking University First Hospital, Beijing, 100034, China
| | - Shengzhi Sun
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Ming Xu
- Department of Global Health, School of Public Health, Peking University, 38 Xue Yuan Road, Haidian District, Beijing, 100191, China
- Institute for Global Health and Development, Peking University, Beijing, China
| | - Zhi-Jie Zheng
- Department of Global Health, School of Public Health, Peking University, 38 Xue Yuan Road, Haidian District, Beijing, 100191, China
- Institute for Global Health and Development, Peking University, Beijing, China
| | - Jianbing Wang
- Department of Epidemiology and Biostatistics, Zhejiang University School of Public Health, Hangzhou, 310058, China
- Department of Epidemiology and Biostatistics, and National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zhenyu Zhang
- Department of Global Health, School of Public Health, Peking University, 38 Xue Yuan Road, Haidian District, Beijing, 100191, China.
- Institute for Global Health and Development, Peking University, Beijing, China.
| | - Kun Chen
- Department of Epidemiology and Biostatistics, Zhejiang University School of Public Health, Hangzhou, 310058, China
- Department of Epidemiology and Biostatistics, and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
54
|
Chen X, Xing L, Li X, Chen N, Liu L, Wang J, Zhou X, Liu S. Manganese Ion-Induced Amyloid Fibrillation Kinetics of Hen Egg White-Lysozyme in Thermal and Acidic Conditions. ACS OMEGA 2023; 8:16439-16449. [PMID: 37179629 PMCID: PMC10173442 DOI: 10.1021/acsomega.3c01531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023]
Abstract
As manganese ions (Mn2+) are identified as an environmental risk factor for neurodegenerative diseases, uncovering their action mechanism on protein amyloid fibril formation is crucial for related disease treatments. Herein, we performed a combined study of Raman spectroscopy, atomic force microscopy (AFM), thioflavin T (ThT) fluorescence, and UV-vis absorption spectroscopy assays, in which the distinctive effect of Mn2+ on the amyloid fibrillation kinetics of hen egg white-lysozyme (HEWL) was clarified at the molecular level. With thermal and acid treatments, the unfolding of protein tertiary structures is efficiently accelerated by Mn2+ to form oligomers, as indicated by two Raman markers for the Trp residues on protein side chains: the FWHM at 759 cm-1 and the I1340/I1360 ratio. Meanwhile, the inconsistent evolutionary kinetics of the two indicators, as well as AFM images and UV-vis absorption spectroscopy assays, validate the tendency of Mn2+ toward the formation of amorphous aggregates instead of amyloid fibrils. Moreover, Mn2+ plays an accelerator role in the secondary structure transition from α-helix to organized β-sheet structures, as indicated by the N-Cα-C intensity at 933 cm-1 and the amide I position of Raman spectroscopy and ThT fluorescence assays. Notably, the more significant promotion effect of Mn2+ on the formation of amorphous aggregates provides credible clues to understand the fact that excess exposure to manganese is associated with neurological diseases.
Collapse
Affiliation(s)
- Xiaodong Chen
- Department
of Chemical Physics, University of Science
and Technology of China, Hefei 230026, China
| | - Lei Xing
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing 210023, China
| | - Xinfei Li
- Department
of Chemical Physics, University of Science
and Technology of China, Hefei 230026, China
| | - Ning Chen
- Department
of Chemical Physics, University of Science
and Technology of China, Hefei 230026, China
| | - Liming Liu
- Department
of Chemical Physics, University of Science
and Technology of China, Hefei 230026, China
| | - Jionghan Wang
- Department
of Chemical Physics, University of Science
and Technology of China, Hefei 230026, China
| | - Xiaoguo Zhou
- Department
of Chemical Physics, University of Science
and Technology of China, Hefei 230026, China
| | - Shilin Liu
- Department
of Chemical Physics, University of Science
and Technology of China, Hefei 230026, China
| |
Collapse
|
55
|
Alikunju M, Misiriyyah N, Sayeed Iqbal S, Khan M. Manganese Neurotoxicity as a Stroke Mimic: A Case Report. Cureus 2023; 15:e37247. [PMID: 37168188 PMCID: PMC10166376 DOI: 10.7759/cureus.37247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2023] [Indexed: 05/13/2023] Open
Abstract
Manganese (Mn)-induced cerebral toxicity is a rare neurological condition that can present as a stroke mimic in high-risk populations. We present a case of a 40-year-old male with no known comorbidities who was brought to the emergency department with complaints of nonprogressive slurred speech and left facial weakness for eight days. Further history revealed that he had been working as a welder in a steel factory for the past seven years without using proper personal protective equipment (PPE). On physical examination, an upper motor neuron (UMN) type weakness on the left side of his face and spastic dysarthria could be appreciated. Following a brain computed tomography (CT) scan that showed ill-defined hypodensities in the basal ganglia without any signs of a hemorrhage, he was admitted to the stroke unit for conservative management and further investigations. A magnetic resonance imaging (MRI) scan of the brain done later showed features of manganese deposition and absorption in the globus pallidus and corticospinal tracts, indicating a diagnosis of manganese-induced cerebral toxicity. His serum manganese levels obtained during admission were normal. He was managed conservatively with intravenous rehydration and was discharged after symptomatic improvement. He was counseled and educated regarding the importance of wearing protective equipment while at work to reduce further exposure to the metal. During his follow-up visit, his symptoms had considerably improved with proper adherence to workplace safety measures.
Collapse
|
56
|
Obsekov V, Ghassabian A, Mukhopadhyay S, Trasande L. Manganese and thyroid function in the national health and nutrition examination survey, 2011-2012. ENVIRONMENTAL RESEARCH 2023; 222:115371. [PMID: 36709872 DOI: 10.1016/j.envres.2023.115371] [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: 11/28/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
CONTEXT Manganese (Mn) exposure is prevalent, as it is found naturally as ionized trace elements and released into the environment as a byproduct of manufacturing and waste disposal. Animal and human studies have suggested variable effects on thyroid function, but the association of Mn exposure with thyroid function has not been evaluated in a national sample. OBJECTIVE To investigate the associations between serum and urinary Mn levels and serum thyroid hormone concentrations in a nationally representative sample. DESIGN, SETTING, PARTICIPANTS, AND INTERVENTION This was a cross-sectional analysis of data from the 2011-2012 National Health and Nutrition Examination Survey among 1360 participants. MAIN OUTCOME MEASURES Serum thyroid stimulating hormone (TSH), total triiodothyronine (T3), total thyroxine (T4), free T3, and free T4. RESULTS Serum Mn levels were positively associated with increasing total T4, free T3, and total T3 in the whole cohort (p < 0.01). Urinary Mn levels were not associated with thyroid hormone levels. When subgroup analyses were performed by gender, only males had total T4 associated with serum Mn [β = 0.01, p < 0.01, confidence interval (CI): 0.004-0.018]. In individuals under 22 years old, serum Mn was significantly associated with total T4 (β = 0.02, p = 0.002, CI: 0.008-0.029). Serum Mn was positively associated with Free T3 in both genders (β = 0.07, p < 0.001). CONCLUSION While our findings do not suggest clinical thyroid dysfunction, there is an association between serum Mn and subclinical changes in thyroid function that warrant further studies. Regulatory action should be considered as Mn-based organometallic compounds are being considered as replacements for lead in gasoline and may pose future risks to human health.
Collapse
Affiliation(s)
- Vladislav Obsekov
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Akhgar Ghassabian
- Department of Pediatrics, Division of Environmental Pediatrics, NYU Grossman School of Medicine, New York, NY, USA; Departments of Population Health and Environmental Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Leonardo Trasande
- Department of Pediatrics, Division of Environmental Pediatrics, NYU Grossman School of Medicine, New York, NY, USA; Departments of Population Health and Environmental Medicine, NYU Grossman School of Medicine, New York, NY, USA; NYU Wagner School of Public Service, New York, NY, USA; NYU College of Global Public Health, New York, NY, USA
| |
Collapse
|
57
|
Foster D, Larsen J. Polymeric Metal Contrast Agents for T 1-Weighted Magnetic Resonance Imaging of the Brain. ACS Biomater Sci Eng 2023; 9:1224-1242. [PMID: 36753685 DOI: 10.1021/acsbiomaterials.2c01386] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Imaging plays an integral role in diagnostics and treatment monitoring for conditions affecting the brain; enhanced brain imaging capabilities will improve upon both while increasing the general understanding of how the brain works. T1-weighted magnetic resonance imaging is the preferred modality for brain imaging. Commercially available contrast agents, which are often required to render readable brain images, have considerable toxicity concerns. In recent years, much progress has been made in developing new contrast agents based on the magnetic features of gadolinium, iron, or magnesium. Nanotechnological approaches for these systems allow for the protected integration of potentially harmful metals with added benefits like reduced dosage and improved transport. Polymeric enhancement of each design further improves biocompatibility while allowing for specific brain targeting. This review outlines research on polymeric nanomedicine designs for T1-weighted contrast agents that have been evaluated for performance in the brain.
Collapse
|
58
|
Aureliano M, De Sousa-Coelho AL, Dolan CC, Roess DA, Crans DC. Biological Consequences of Vanadium Effects on Formation of Reactive Oxygen Species and Lipid Peroxidation. Int J Mol Sci 2023; 24:ijms24065382. [PMID: 36982458 PMCID: PMC10049017 DOI: 10.3390/ijms24065382] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Lipid peroxidation (LPO), a process that affects human health, can be induced by exposure to vanadium salts and compounds. LPO is often exacerbated by oxidation stress, with some forms of vanadium providing protective effects. The LPO reaction involves the oxidation of the alkene bonds, primarily in polyunsaturated fatty acids, in a chain reaction to form radical and reactive oxygen species (ROS). LPO reactions typically affect cellular membranes through direct effects on membrane structure and function as well as impacting other cellular functions due to increases in ROS. Although LPO effects on mitochondrial function have been studied in detail, other cellular components and organelles are affected. Because vanadium salts and complexes can induce ROS formation both directly and indirectly, the study of LPO arising from increased ROS should include investigations of both processes. This is made more challenging by the range of vanadium species that exist under physiological conditions and the diverse effects of these species. Thus, complex vanadium chemistry requires speciation studies of vanadium to evaluate the direct and indirect effects of the various species that are present during vanadium exposure. Undoubtedly, speciation is important in assessing how vanadium exerts effects in biological systems and is likely the underlying cause for some of the beneficial effects reported in cancerous, diabetic, neurodegenerative conditions and other diseased tissues impacted by LPO processes. Speciation of vanadium, together with investigations of ROS and LPO, should be considered in future biological studies evaluating vanadium effects on the formation of ROS and on LPO in cells, tissues, and organisms as discussed in this review.
Collapse
Affiliation(s)
- Manuel Aureliano
- Faculdade de Ciências e Tecnologia (FCT), Universidade do Algarve, 8005-139 Faro, Portugal
- CCMar, Universidade do Algarve, 8005-139 Faro, Portugal
- Correspondence: (M.A.); (D.C.C.); Tel.: +351-289-900-805 (M.A.)
| | - Ana Luísa De Sousa-Coelho
- Escola Superior de Saúde, Universidade do Algarve (ESSUAlg), 8005-139 Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), 8005-139 Faro, Portugal
| | - Connor C. Dolan
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | - Deborah A. Roess
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Debbie C. Crans
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
- Cellular and Molecular Biology Program, Colorado State University, Fort Collins, CO 80523, USA
- Correspondence: (M.A.); (D.C.C.); Tel.: +351-289-900-805 (M.A.)
| |
Collapse
|
59
|
Liu K, Liu Z, Liu Z, Ma Z, Jia Y, Deng Y, Liu W, Xu B. Manganese-induced PINK1 S-nitrosylation exacerbates nerve cell damage by promoting ZNF746 repression of mitochondrial biogenesis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 863:160985. [PMID: 36535484 DOI: 10.1016/j.scitotenv.2022.160985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 06/17/2023]
Abstract
Occupational exposure and non-occupational exposure to excessive levels of manganese (Mn) result in neuronal cell damage through mitochondrial dysfunction. The functional integrity of mitochondria is maintained by mitophagy and mitochondrial biogenesis. Although Mn-induced S-nitrosylation of PTEN-induced putative kinase 1 (PINK1) can interfere with mitophagy, its effect on mitochondrial biogenesis remains unclear. In this study, we established a rat model of Mn poisoning or "manganism" to examine the relationship between PINK1 S-nitrosylation and impairment of mitochondrial biogenesis, and found that treatment with 60 mg/kg Mn induced marked neurobehavioral abnormalities in rats and significantly increased the S-nitrosylation level of PINK1. We also found that the nuclear-encoded peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PPARGC1A)-mediated mitochondrial biogenesis was significantly upregulated in rats treated with 15 and 30 mg/kg Mn, and downregulated in rats treated with 60 mg/kg Mn. We further investigated the role of S-nitrosylated PINK1 and its molecular mechanism in the high-dose Mn-mediated impairment of mitochondrial biogenesis in primary cultured neurons treated with the nitric oxide synthase 2 (NOS2) inhibitor 1400 W. Our results revealed that the PPARGC1A-mediated mitochondrial biogenesis was upregulated in neurons treated with 100 μM, but downregulated in neurons treated with 200 μM Mn, which was similar to the in vivo results. However, treatment with 1400W could effectively prevent the 200 μM Mn-mediated impairment of mitochondrial biogenesis by suppressing nitric oxide (NO)-mediated PINK1 S-nitrosylation and rescuing Parkin-interacting substrate (PARIS, ZNF746) degradation, thereby upregulating mitochondrial biogenesis via PPARGC1A. These findings demonstrated that S-nitrosylation of PINK1 and subsequent prevention of ZNF746 degradation were crucial signaling processes involved in the Mn-mediated impairment of mitochondrial biogenesis, which might serve as an underlying mechanism of Mn-induced neurotoxicity. Furthermore, this study provided a reliable target for the prevention and treatment of manganism.
Collapse
Affiliation(s)
- Kuan Liu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, People's Republic of China
| | - Zhiqi Liu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, People's Republic of China
| | - Zhuofan Liu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, People's Republic of China
| | - Zhuo Ma
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, People's Republic of China
| | - Yunfei Jia
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 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 Province 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 Province 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 Province 110122, People's Republic of China.
| |
Collapse
|
60
|
Lodge EK, Martin CL, Fry RC, White AJ, Ward-Caviness CK, Galea S, Aiello AE. Objectively measured external building quality, Census housing vacancies and age, and serum metals in an adult cohort in Detroit, Michigan. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2023; 33:177-186. [PMID: 35577901 PMCID: PMC9666563 DOI: 10.1038/s41370-022-00447-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Residentially derived lead pollution remains a significant problem in urban areas across the country and globe. The risks of childhood residence in housing contaminated with lead-based paint are well-established, but less is known about the effects of housing quality on adult lead exposure. OBJECTIVE To evaluate the effects of residential-area housing age, vacancy status, and building quality on adult lead exposures. METHODS We evaluated the effect of Census block group housing vacancy proportion, block group housing age, and in-person survey evaluated neighborhood building quality on serum levels of lead, mercury, manganese, and copper among a representative cohort of adults in Detroit, Michigan, from 2008-2013 using generalized estimating equations. RESULTS Participants in Census block groups with higher proportions of vacant and aged housing had non-significantly elevated serum lead levels. We identified similar positive associations between residence in neighborhoods with poorer objectively measured building quality and serum lead. Associations between Census vacancies, housing age, objectively measured building quality, and serum lead were stronger among participants with a more stable residential history. SIGNIFICANCE Vacant, aged, and poorly maintained housing may contribute to widespread, low-level lead exposure among adult residents of older cities like Detroit, Michigan. US Census and neighborhood quality data may be a useful tool to identify population-level lead exposures among US adults. IMPACT Using longitudinal data from a representative cohort of adults in Detroit, Michigan, we demonstrate that Census data regarding housing vacancies and age and neighborhood survey data regarding housing quality are associated with increasing serum lead levels. Previous research has primarily focused on housing quality and lead exposures among children. Here, we demonstrate that area-level metrics of housing quality are associated with lead exposures among adults.
Collapse
Affiliation(s)
- Evans K Lodge
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Chantel L Martin
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for Environmental Health & Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Center for Environmental Health & Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandra J White
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Cavin K Ward-Caviness
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, NC, USA
| | - Sandro Galea
- School of Public Health, Boston University, Boston, MA, USA
| | - Allison E Aiello
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
61
|
Ademiluyi AO, Ogunsuyi OB, Akinduro JO, Aro OP, Oboh G. Evaluating Water bitter leaf ( Struchium sparganophora) and Scent Leaf ( Ocimum gratissimum) extracts as sources of nutraceuticals against manganese-induced toxicity in fruit fly model. Drug Chem Toxicol 2023; 46:236-246. [PMID: 35315297 DOI: 10.1080/01480545.2021.2021928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tropical vegetables remain one of the major sources of functional foods and nutraceuticals, while their constituent phytochemicals, especially alkaloids, have been reported to exhibit neuroprotective properties. Here, the protective effect of alkaloid extracts from Scent leaf (Ocimum gratissimum) and Water bitter leaf (Struchium sparganophora) on manganese (Mn)- induced toxicity in wild type fruit fly (Drosophila melanogaster) model was investigated. Flies were exposed to 30 mM of Mn, the alkaloid extracts (20 and 200 µg/g) and co-treatment of Mn plus extracts, respectively. The survival rate and locomotor performance of the flies were assessed 7 days post-treatment, after which the flies were homogenized and assayed for activities of acetylcholinesterase (AChE), monoamine oxidase (MAO), glutathione-S transferase (GST), catalase, superoxide dismutase SOD), as well as total thiol, reactive oxygen species (ROS) and neural L-DOPA levels. Results showed that the extract significantly reversed Mn-induced reduction in the survival rate and locomotor performance of the flies. Furthermore, both extracts counteracted the Mn-induced elevation in AChE and MAO activities, as well as reduced antioxidant enzyme activities, with a concomitant mitigation of Mn-induced elevated ROS and neural L-DOPA level. The HPLC characterization of the extracts revealed the presence of N-propylamine, Vernomine and Piperidine as predominant in Water bitter leaf extract, while 2, 6-dimethylpyrazine and sesbanimide were found in scent leaf extract. Therefore, the alkaloid extract of these leaves may thus be sources of useful nutraceuticals for the management of pathological conditions associated with manganese toxicity.
Collapse
Affiliation(s)
- Adedayo Oluwaseun Ademiluyi
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| | - Opeyemi Babatunde Ogunsuyi
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology, Akure, Nigeria.,Department of Biomedical Technology, Federal University of Technology, Akure, Nigeria
| | - Josephine Oluwaseun Akinduro
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| | - Olayemi Philemon Aro
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| | - Ganiyu Oboh
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| |
Collapse
|
62
|
Liu C, Ju R. Manganese-induced neuronal apoptosis: new insights into the role of endoplasmic reticulum stress in regulating autophagy-related proteins. Toxicol Sci 2023; 191:193-200. [PMID: 36519822 DOI: 10.1093/toxsci/kfac130] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Manganese (Mn) is an essential trace element that participates in various physiological and pathological processes. However, epidemiological observations indicate that overexposure to Mn is strongly associated with neurodegenerative disorders and has been recognized as a potential risk factor of neuronal apoptosis. Many mechanisms are involved in the pathogenesis of Mn-induced neuronal apoptosis, such as reactive oxygen species generation, neuroinflammation reactions, protein accumulation, endoplasmic reticulum stress (ER stress), and autophagy, all of which collectively accelerate the process of nerve cell damage. As sophisticated cellular processes for maintaining intracellular homeostasis, ER-mediated unfolded protein response and autophagy both play bilateral roles including cell protection and cell injury under pathophysiological conditions, which might interact with each other. Although emerging evidence suggests that ER stress is involved in regulating the compensatory activation of autophagy to promote cell survival, the inherent relationship between ER stress and autophagy on Mn-induced neurotoxicity remains obscure. Here, our review focuses on discussing the existing mechanisms and connections between ER stress, autophagy, and apoptosis, which provide a new perspective on Mn-induced neuronal apoptosis, and the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Chang Liu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu Women's and Children's Central Hospital, Chengdu 611731, China
| | - Rong Ju
- School of Medicine, University of Electronic Science and Technology of China, Chengdu Women's and Children's Central Hospital, Chengdu 611731, China
| |
Collapse
|
63
|
Frydrych A, Krośniak M, Jurowski K. The Role of Chosen Essential Elements (Zn, Cu, Se, Fe, Mn) in Food for Special Medical Purposes (FSMPs) Dedicated to Oncology Patients-Critical Review: State-of-the-Art. Nutrients 2023; 15:1012. [PMID: 36839370 PMCID: PMC9961387 DOI: 10.3390/nu15041012] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/10/2023] [Accepted: 02/12/2023] [Indexed: 02/22/2023] Open
Abstract
The scoping review aimed to characterise the role of selected essential elements (Zn, Cu, Se, Fe, Mn) in food for special medical purposes (FSMPs) aimed at oncology patients. The scope review was conducted using Scopus, Google Scholar, and Web of Science to find published references on this subject. Data from the reviewed literature were related to the physiological functions of the element in the body, and the effects of deficiencies and excesses, referring to the latest ESPEN and EFSA guidelines, among others. Important dietary indices/parameters based on the literature review are provided for each element. On the basis of the literature, data on the level of elements in patients with cancer were collected. The content of these elements in 100 mL of FSMPs was read from the manufacturers' declarations. The literature has been provided on the importance of each element in cancer. Our findings show that the essential elements (Zn, Cu, Se, Fe, and Mn) of FSMPs for cancer patients are not adequately treated. We suggest solutions to ensure the safe use of FSMPs in oncology patients.
Collapse
Affiliation(s)
- Adrian Frydrych
- Laboratory of Innovative Toxicological Research and Analyses, Institute of Medical Studies, Medical College, Rzeszów University, Aleja Majora W. Kopisto 2a, 35-959 Rzeszow, Poland
| | - Mirosław Krośniak
- Department of Food Chemistry and Nutrition, Medical College, Jagiellonian University, Medyczna 9, 30-688 Kraków, Poland
| | - Kamil Jurowski
- Laboratory of Innovative Toxicological Research and Analyses, Institute of Medical Studies, Medical College, Rzeszów University, Aleja Majora W. Kopisto 2a, 35-959 Rzeszow, Poland
- Department of Regulatory and Forensic Toxicology, Institute of Medical Expertises, Aleksandrowska 67/93, 91-205 Łódź, Poland
| |
Collapse
|
64
|
Berger MM, Talwar D, Shenkin A. Pitfalls in the interpretation of blood tests used to assess and monitor micronutrient nutrition status. Nutr Clin Pract 2023; 38:56-69. [PMID: 36335431 DOI: 10.1002/ncp.10924] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/17/2022] [Accepted: 10/02/2022] [Indexed: 11/09/2022] Open
Abstract
Assessment of micronutrient (MN) status is of particular importance in patients who require medical nutrition therapy, especially those requiring parenteral nutrition. Blood testing is generally the only tool available in clinical settings to assess MN status. However, using plasma or serum concentration faces pitfalls mainly because of the impact of inflammation that diverts the MNs from the circulating compartment. This review aims to review the blood tests that are useful and provide information about how to integrate functional markers of status to reach a clinically relevant diagnosis. Most impacted, with a significant and proportional decrease in plasma concentrations, are iron, selenium, zinc, thiamin, folic acid, cobalamin, and vitamins A, C, and D; copper is the only MN for which the plasma concentration increases. Therefore, a surrogate marker of inflammation, C-reactive protein, must always be determined simultaneously. Validated intracellular and functional tests are proposed to improve status assessment. A protocol is suggested for tests required both on commencing and during nutrition support. A timely turnaround of analysis is essential for results to be clinically useful. In some cases, the appropriate provision of MNs should be commenced before results have been obtained to confirm the clinical assessment. Laboratory tests of MN status are an area prone to misuse and misinterpretation. The appropriate use and interpretation of such tests are essential to ensure the correct management of nutrition problems.
Collapse
Affiliation(s)
- Mette M Berger
- Department of Adult Intensive Care, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Dinesh Talwar
- Scottish Trace Element and Micronutrient Diagnostic and Research Laboratory, Department of Clinical Biochemistry, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Alan Shenkin
- Institute of Aging and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
65
|
Pajarillo E, Nyarko-Danquah I, Digman A, Multani HK, Kim S, Gaspard P, Aschner M, Lee E. Mechanisms of manganese-induced neurotoxicity and the pursuit of neurotherapeutic strategies. Front Pharmacol 2022; 13:1011947. [PMID: 36605395 PMCID: PMC9808094 DOI: 10.3389/fphar.2022.1011947] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/01/2022] [Indexed: 01/07/2023] Open
Abstract
Chronic exposure to elevated levels of manganese via occupational or environmental settings causes a neurological disorder known as manganism, resembling the symptoms of Parkinson's disease, such as motor deficits and cognitive impairment. Numerous studies have been conducted to characterize manganese's neurotoxicity mechanisms in search of effective therapeutics, including natural and synthetic compounds to treat manganese toxicity. Several potential molecular targets of manganese toxicity at the epigenetic and transcriptional levels have been identified recently, which may contribute to develop more precise and effective gene therapies. This review updates findings on manganese-induced neurotoxicity mechanisms on intracellular insults such as oxidative stress, inflammation, excitotoxicity, and mitophagy, as well as transcriptional dysregulations involving Yin Yang 1, RE1-silencing transcription factor, transcription factor EB, and nuclear factor erythroid 2-related factor 2 that could be targets of manganese neurotoxicity therapies. This review also features intracellular proteins such as PTEN-inducible kinase 1, parkin, sirtuins, leucine-rich repeat kinase 2, and α-synuclein, which are associated with manganese-induced dysregulation of autophagy/mitophagy. In addition, newer therapeutic approaches to treat manganese's neurotoxicity including natural and synthetic compounds modulating excitotoxicity, autophagy, and mitophagy, were reviewed. Taken together, in-depth mechanistic knowledge accompanied by advances in gene and drug delivery strategies will make significant progress in the development of reliable therapeutic interventions against manganese-induced neurotoxicity.
Collapse
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Ivan Nyarko-Danquah
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Alexis Digman
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Harpreet Kaur Multani
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL, United States
| | - Sanghoon Kim
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Patric Gaspard
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY, United States
| | - Eunsook Lee
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| |
Collapse
|
66
|
Zhang X, Liu J, Wang H. The cGAS-STING-autophagy pathway: Novel perspectives in neurotoxicity induced by manganese exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 315:120412. [PMID: 36240967 DOI: 10.1016/j.envpol.2022.120412] [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: 07/26/2022] [Revised: 08/28/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
Chronic high-level heavy metal exposure increases the risk of developing different neurodegenerative diseases. Chronic excessive manganese (Mn) exposure is known to lead to neurodegenerative diseases. In addition, some evidence suggests that autophagy dysfunction plays an important role in the pathogenesis of various neurodegenerative diseases. Over the past decade, the DNA-sensing receptor cyclic GMP-AMP synthase (cGAS) and its downstream signal-efficient interferon gene stimulator (STING), as well as the molecular composition and regulatory mechanisms of this pathway have been well understood. The cGAS-STING pathway has emerged as a crucial mechanism to induce effective innate immune responses by inducing type I interferons in mammalian cells. Moreover, recent studies have found that Mn2+ is the second activator of the cGAS-STING pathway besides dsDNA, and inducing autophagy is a primitive function for the activation of the cGAS-STING pathway. However, overactivation of the immune response can lead to tissue damage. This review discusses the mechanism of neurotoxicity induced by Mn exposure from the cGAS-STING-autophagy pathway. Future work exploiting the cGAS-STING-autophagy pathway may provide a novel perspective for manganese neurotoxicity.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Jingjing Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
67
|
Association between Heavy Metal Exposure and Parkinson's Disease: A Review of the Mechanisms Related to Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11122467. [PMID: 36552676 PMCID: PMC9774122 DOI: 10.3390/antiox11122467] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Parkinson's disease (PD) is a gradually progressing neurodegenerative condition that is marked by a loss of motor coordination along with non-motor features. Although the precise cause of PD has not been determined, the disease condition is mostly associated with the exposure to environmental toxins, such as metals, and their abnormal accumulation in the brain. Heavy metals, such as iron (Fe), mercury (Hg), manganese (Mn), copper (Cu), and lead (Pb), have been linked to PD and contribute to its progression. In addition, the interactions among the components of a metal mixture may result in synergistic toxicity. Numerous epidemiological studies have demonstrated a connection between PD and either single or mixed exposure to these heavy metals, which increase the prevalence of PD. Chronic exposure to heavy metals is related to the activation of proinflammatory cytokines resulting in neuronal loss through neuroinflammation. Similarly, metals disrupt redox homeostasis while inducing free radical production and decreasing antioxidant levels in the substantia nigra. Furthermore, these metals alter molecular processes and result in oxidative stress, DNA damage, mitochondrial dysfunction, and apoptosis, which can potentially trigger dopaminergic neurodegenerative disorders. This review focuses on the roles of Hg, Pb, Mn, Cu, and Fe in the development and progression of PD. Moreover, it explores the plausible roles of heavy metals in neurodegenerative mechanisms that facilitate the development of PD. A better understanding of the mechanisms underlying metal toxicities will enable the establishment of novel therapeutic approaches to prevent or cure PD.
Collapse
|
68
|
Adamson A, Buck SA, Freyberg Z, De Miranda BR. Sex Differences in Dopaminergic Vulnerability to Environmental Toxicants - Implications for Parkinson's Disease. Curr Environ Health Rep 2022; 9:563-573. [PMID: 36201109 PMCID: PMC10201647 DOI: 10.1007/s40572-022-00380-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW Sex dimorphism in Parkinson's disease (PD) is an ostensible feature of the neurological disorder, particularly as men are 1.5-2 times more likely to develop PD than women. Clinical features of the disease, such as presentation at onset, most prevalent symptoms, and response to treatment, are also affected by sex. Despite these well-known sex differences in PD risk and phenotype, the mechanisms that impart sex dimorphisms in PD remain poorly understood. RECENT FINDINGS As PD incidence is influenced by environmental factors, an intriguing pattern has recently emerged in research studies suggesting a male-specific vulnerability to dopaminergic neurodegeneration caused by neurotoxicant exposure, with relative protection in females. These new experimental data have uncovered potential mechanisms that provide clues to the source of sex differences in dopaminergic neurodegeneration and other PD pathology such as alpha-synuclein toxicity. In this review, we discuss the emerging evidence of increased male sensitivity to neurodegeneration from environmental exposures. We examine mechanisms underlying dopaminergic neurodegeneration and PD-related pathologies with evidence supporting the roles of estrogen, SRY expression, the vesicular glutamate transporter VGLUT2, and the microbiome as prospective catalysts for male vulnerability. We also highlight the importance of including sex as a biological variable, particularly when evaluating dopaminergic neurotoxicity in the context of PD.
Collapse
Affiliation(s)
- Ashley Adamson
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1719 6th Ave South, CIRC 560, Birmingham, AL, 35294, USA
| | - Silas A Buck
- Center for Neuroscience, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary Freyberg
- Center for Neuroscience, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Briana R De Miranda
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1719 6th Ave South, CIRC 560, Birmingham, AL, 35294, USA.
| |
Collapse
|
69
|
Daksh S, Kaul A, Deep S, Datta A. Current advancement in the development of manganese complexes as magnetic resonance imaging probes. J Inorg Biochem 2022; 237:112018. [PMID: 36244313 DOI: 10.1016/j.jinorgbio.2022.112018] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/14/2022] [Accepted: 09/28/2022] [Indexed: 01/18/2023]
Abstract
Emerging non-invasive molecular imaging modalities can detect a pathophysiological state at the molecular level before any anatomic changes are observed. Magnetic resonance imaging (MRI) is preferred over other nuclear imaging techniques owing to its radiation-free approach. Conventionally, most MRI contrast agents employed predominantly involve lanthanide metal: Gadolinium (Gd) until the discovery of associated severe nephrogenic toxicity issues. This limitation led a way to the development of manganese-based contrast agents which offer similar positive contrast enhancement capability. A vast quantity of experimental data has been accumulated over the last decade to define the physicochemical characteristics of manganese chelates with various ligand scaffolds. One can now observe how the ligand configurations, rigidity, and donor-acceptor characteristics impact the stability of the complex. This review covers the current trends in the development of manganese-based MRI contrast agents, the mechanisms they are based on and design considerations for newer manganese-based contrast agents with higher diagnostic strength along with better safety profiles.
Collapse
Affiliation(s)
- Shivani Daksh
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization, Brig S. K. Mazumdar Marg, Delhi 110054, India; Department of Chemistry, Indian Institute of Technology, Hauz-Khas, New Delhi 110016, India
| | - Ankur Kaul
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization, Brig S. K. Mazumdar Marg, Delhi 110054, India
| | - Shashank Deep
- Department of Chemistry, Indian Institute of Technology, Hauz-Khas, New Delhi 110016, India.
| | - Anupama Datta
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization, Brig S. K. Mazumdar Marg, Delhi 110054, India.
| |
Collapse
|
70
|
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
|
71
|
Diessl J, Berndtsson J, Broeskamp F, Habernig L, Kohler V, Vazquez-Calvo C, Nandy A, Peselj C, Drobysheva S, Pelosi L, Vögtle FN, Pierrel F, Ott M, Büttner S. Manganese-driven CoQ deficiency. Nat Commun 2022; 13:6061. [PMID: 36229432 PMCID: PMC9563070 DOI: 10.1038/s41467-022-33641-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 09/26/2022] [Indexed: 12/24/2022] Open
Abstract
Overexposure to manganese disrupts cellular energy metabolism across species, but the molecular mechanism underlying manganese toxicity remains enigmatic. Here, we report that excess cellular manganese selectively disrupts coenzyme Q (CoQ) biosynthesis, resulting in failure of mitochondrial bioenergetics. While respiratory chain complexes remain intact, the lack of CoQ as lipophilic electron carrier precludes oxidative phosphorylation and leads to premature cell and organismal death. At a molecular level, manganese overload causes mismetallation and proteolytic degradation of Coq7, a diiron hydroxylase that catalyzes the penultimate step in CoQ biosynthesis. Coq7 overexpression or supplementation with a CoQ headgroup analog that bypasses Coq7 function fully corrects electron transport, thus restoring respiration and viability. We uncover a unique sensitivity of a diiron enzyme to mismetallation and define the molecular mechanism for manganese-induced bioenergetic failure that is conserved across species.
Collapse
Affiliation(s)
- Jutta Diessl
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden
| | - Jens Berndtsson
- Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden
| | - Filomena Broeskamp
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden
| | - Lukas Habernig
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden
| | - Verena Kohler
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden
| | - Carmela Vazquez-Calvo
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden
- Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden
| | - Arpita Nandy
- Institute of Biochemistry and Molecular Biology, ZBMZ, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Carlotta Peselj
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden
| | - Sofia Drobysheva
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden
| | - Ludovic Pelosi
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, 38000, Grenoble, France
| | - F-Nora Vögtle
- Institute of Biochemistry and Molecular Biology, ZBMZ, University of Freiburg, 79104, Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, 69120, Heidelberg, Germany
- Network Aging Research, Heidelberg University, 69120, Heidelberg, Germany
| | - Fabien Pierrel
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, 38000, Grenoble, France
| | - Martin Ott
- Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Sabrina Büttner
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden.
| |
Collapse
|
72
|
Soto-Verdugo J, Siva-Parra J, Hernández-Kelly LC, Ortega A. Acute Manganese Exposure Modifies the Translation Machinery via PI3K/Akt Signaling in Glial Cells. ASN Neuro 2022; 14:17590914221131452. [PMID: 36203371 PMCID: PMC9551334 DOI: 10.1177/17590914221131452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SUMMARY STATEMENT We demonstrate herein that short-term exposure of radial glia cells to Manganese, a neurotoxic metal, induces an effect on protein synthesis, altering the protein repertoire of these cells.
Collapse
Affiliation(s)
| | | | | | - Arturo Ortega
- Arturo Ortega, Departamento de Toxicología,
Centro de Investigación y de Estudios Avanzados del Instituto Politécnico
Nacional, México City, México, 07360.
| |
Collapse
|
73
|
Afsheen N, Rafique S, Rafeeq H, Irshad K, Hussain A, Huma Z, Kumar V, Bilal M, Aleya L, Iqbal HMN. Neurotoxic effects of environmental contaminants-measurements, mechanistic insight, and environmental relevance. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:70808-70821. [PMID: 36059010 DOI: 10.1007/s11356-022-22779-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 08/24/2022] [Indexed: 06/15/2023]
Abstract
Pollution is a significant and growing concern for any population regardless of age because these environmental contaminants exhibit different neurodegenerative effects on persons of different ages. These environmental contaminants are the products of human welfare projects like industry, automobile exhaust, clinical and research laboratory extrudes, and agricultural chemicals. These contaminants are found in various forms in environmental matrices like nanoparticles, particulate matter, lipophilic vaporized toxicants, and ultrafine particulate matter. Because of their small size, they can easily cross blood-brain barriers or use different cellular mechanisms for assistance. Other than this, these contaminants cause an innate immune response in different cells of the central nervous system and cause neurotoxicity. Considering the above critiques and current needs, this review summarizes different protective strategies based on bioactive compounds present in plants. Various bioactive compounds from medicinal plants with neuroprotective capacities are discussed with relevant examples. Many in vitro studies on clinical trials have shown promising outcomes using plant-based bioactive compounds against neurological disorders.
Collapse
Affiliation(s)
- Nadia Afsheen
- Department of Biochemistry, Riphah International University, Faisalabad, 38000, Pakistan
| | - Sadia Rafique
- Department of Pharmacy, Riphah International University, Faisalabad, 38000, Pakistan
| | - Hamza Rafeeq
- Department of Biochemistry, Riphah International University, Faisalabad, 38000, Pakistan
| | - Kanwal Irshad
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, 38000, Pakistan
| | - Asim Hussain
- Department of Biochemistry, University of Agriculture, Faisalabad, 38000, Pakistan
| | - Zille Huma
- Department of Chemistry, Riphah International University, Faisalabad, 38000, Pakistan
| | - Vineet Kumar
- Department of Basic and Applied Sciences, School of Engineering and Sciences, GD Goenka University, Sohna Road, Gurugram, Haryana, 122103, India
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon, France
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, 64849, Monterrey, Mexico.
| |
Collapse
|
74
|
Lodge EK, Guseh NS, Martin CL, Fry RC, White AJ, Ward-Caviness CK, Galea S, Aiello AE. The effect of residential proximity to brownfields, highways, and heavy traffic on serum metal levels in the Detroit Neighborhood Health Study. ENVIRONMENTAL ADVANCES 2022; 9:100278. [PMID: 36034484 PMCID: PMC9401556 DOI: 10.1016/j.envadv.2022.100278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Research in environmental sciences has demonstrated that land in close proximity to brownfields and heavily trafficked highways is contaminated with toxic metals. Despite this, little is known about the influence of brownfields and highways on metal levels in residents living nearby. We used data from 774 participants in the Detroit Neighborhood Health Study to estimate the effect of residential proximity to brownfields, highways, and present-day traffic on serum levels of lead, mercury, manganese, and copper using generalized estimating equations. We found that a 1 standard deviation increase in residential brownfield density within 200m was associated with increased serum lead levels (β: 0.04, 95% CI: -0.01, 0.09). The same modeled increase in a subset of historic industrial-use brownfields was associated with elevated serum mercury (β: 0.06, 95% CI: 0.03, 0.09). Increased highway and traffic density was positively associated with serum manganese (β: 0.02, 95% CI: 0.01, 0.04). Highway and traffic density was also positively associated with serum lead (β: 0.02, 95% CI: 0.01, 0.03) after restricting the analysis to participants who did not move during the study follow-up period. These findings draw attention to the importance of remediating polluted post-industrial sites in heavily populated areas, particularly as residents continue to move into densely populated cities around the globe.
Collapse
Affiliation(s)
- Evans K. Lodge
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nahnsan S. Guseh
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, NC, USA
| | - Chantel L. Martin
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for Environmental Health & Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandra J. White
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Cavin K. Ward-Caviness
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, NC, USA
| | - Sandro Galea
- School of Public Health, Boston University, Boston, MA, USA
| | - Allison E. Aiello
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
75
|
Jewell S, Herath AM, Gordon R. Inflammasome Activation in Parkinson’s Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:S113-S128. [PMID: 35848038 PMCID: PMC9535572 DOI: 10.3233/jpd-223338] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Chronic sterile inflammation and persistent immune activation is a prominent pathological feature of Parkinson’s disease (PD). Inflammasomes are multi-protein intracellular signaling complexes which orchestrate inflammatory responses in immune cells to a diverse range of pathogens and host-derived signals. Widespread inflammasome activation is evident in PD patients at the sites of dopaminergic degeneration as well as in blood samples and mucosal biopsies. Inflammasome activation in the nigrostriatal system is also a common pathological feature in both neurotoxicant and α-synuclein models of PD where dopaminergic degeneration occurs through distinct mechanisms. The NLRP3 (NLR Family Pyrin Domain Containing 3) inflammasome has been shown to be the primary driver of inflammatory neurotoxicity in PD and other neurodegenerative diseases. Chronic NLRP3 inflammasome activation is triggered by pathogenic misfolded α-synuclein aggregates which accumulate and spread over the disease course in PD. Converging lines of evidence suggest that blocking inflammasome activation could be a promising therapeutic strategy for disease modification, with both NLRP3 knockout mice and CNS-permeable pharmacological inhibitors providing robust neuroprotection in multiple PD models. This review summarizes the current evidence and knowledge gaps around inflammasome activation in PD, the pathological mechanisms by which persistent inflammasome activation can drive dopaminergic degeneration and the therapeutic opportunities for disease modification using NLRP3 inhibitors.
Collapse
Affiliation(s)
- Shannon Jewell
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Ashane M. Herath
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Richard Gordon
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
76
|
Lodge EK, Dhingra R, Martin CL, Fry RC, White AJ, Ward-Caviness CK, Wani AH, Uddin M, Wildman DE, Galea S, Aiello AE. Serum lead, mercury, manganese, and copper and DNA methylation age among adults in Detroit, Michigan. ENVIRONMENTAL EPIGENETICS 2022; 8:dvac018. [PMID: 36330039 PMCID: PMC9620967 DOI: 10.1093/eep/dvac018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/03/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
Although the effects of lead, mercury, manganese, and copper on individual disease processes are well understood, estimating the health effects of long-term exposure to these metals at the low concentrations often observed in the general population is difficult. In addition, the health effects of joint exposure to multiple metals are difficult to estimate. Biological aging refers to the integrative progression of multiple physiologic and molecular changes that make individuals more at risk of disease. Biomarkers of biological aging may be useful to estimate the population-level effects of metal exposure prior to the development of disease in the population. We used data from 290 participants in the Detroit Neighborhood Health Study to estimate the effect of serum lead, mercury, manganese, and copper on three DNA methylation-based biomarkers of biological aging (Horvath Age, PhenoAge, and GrimAge). We used mixed models and Bayesian kernel machine regression and controlled for participant sex, race, ethnicity, cigarette use, income, educational attainment, and block group poverty. We observed consistently positive estimates of the effects between lead and GrimAge acceleration and mercury and PhenoAge acceleration. In contrast, we observed consistently negative associations between manganese and PhenoAge acceleration and mercury and Horvath Age acceleration. We also observed curvilinear relationships between copper and both PhenoAge and GrimAge acceleration. Increasing total exposure to the observed mixture of metals was associated with increased PhenoAge and GrimAge acceleration and decreased Horvath Age acceleration. These findings indicate that an increase in serum lead or mercury from the 25th to 75th percentile is associated with a ∼0.25-year increase in two epigenetic markers of all-cause mortality in a population of adults in Detroit, Michigan. While few of the findings were statistically significant, their consistency and novelty warrant interest.
Collapse
Affiliation(s)
- Evans K Lodge
- *Correspondence address. Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC 27599, USA. Tel: +574-339-0253; Fax: +919-966-2089; E-mail:
| | - Radhika Dhingra
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 135 Dauer Dr, Chapel Hill, NC 27599, USA
- Institute for Environmental Health Solutions, University of North Carolina at Chapel Hill, 135 Dauer Dr, Chapel Hill, NC 27599, USA
| | - Chantel L Martin
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 135 Dauer Dr, Chapel Hill, NC 27599, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, 123 W Franklin St, Chapel Hill, NC 27516, USA
- Center for Environmental Health & Susceptibility, University of North Carolina at Chapel Hill, 135 Dauer Dr, Chapel Hill, NC 27599, USA
| | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 135 Dauer Dr, Chapel Hill, NC 27599, USA
- Center for Environmental Health & Susceptibility, University of North Carolina at Chapel Hill, 135 Dauer Dr, Chapel Hill, NC 27599, USA
| | - Alexandra J White
- Epidemiology Branch, National Institute of Environmental Health Sciences, A323 David P Rall Building, Research Triangle Park, NC 27709, USA
| | - Cavin K Ward-Caviness
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, 104 Mason Farm Rd, Chapel Hill, NC 27514, USA
| | - Agaz H Wani
- Genomics Program, College of Public Health, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Monica Uddin
- Genomics Program, College of Public Health, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Derek E Wildman
- Genomics Program, College of Public Health, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Sandro Galea
- School of Public Health, Boston University, 715 Albany St, Boston, MA 02118, USA
| | - Allison E Aiello
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 135 Dauer Dr, Chapel Hill, NC 27599, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, 123 W Franklin St, Chapel Hill, NC 27516, USA
| |
Collapse
|
77
|
Huang M, Bargues-Carot A, Riaz Z, Wickham H, Zenitsky G, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Impact of Environmental Risk Factors on Mitochondrial Dysfunction, Neuroinflammation, Protein Misfolding, and Oxidative Stress in the Etiopathogenesis of Parkinson's Disease. Int J Mol Sci 2022; 23:10808. [PMID: 36142718 PMCID: PMC9505762 DOI: 10.3390/ijms231810808] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
As a prevalent progressive neurodegenerative disorder, Parkinson's disease (PD) is characterized by the neuropathological hallmark of the loss of nigrostriatal dopaminergic (DAergic) innervation and the appearance of Lewy bodies with aggregated α-synuclein. Although several familial forms of PD have been reported to be associated with several gene variants, most cases in nature are sporadic, triggered by a complex interplay of genetic and environmental risk factors. Numerous epidemiological studies during the past two decades have shown positive associations between PD and several environmental factors, including exposure to neurotoxic pesticides/herbicides and heavy metals as well as traumatic brain injury. Other environmental factors that have been implicated as potential risk factors for PD include industrial chemicals, wood pulp mills, farming, well-water consumption, and rural residence. In this review, we summarize the environmental toxicology of PD with the focus on the elaboration of chemical toxicity and the underlying pathogenic mechanisms associated with exposure to several neurotoxic chemicals, specifically 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), rotenone, paraquat (PQ), dichloro-diphenyl-trichloroethane (DDT), dieldrin, manganese (Mn), and vanadium (V). Our overview of the current findings from cellular, animal, and human studies of PD provides information for possible intervention strategies aimed at halting the initiation and exacerbation of environmentally linked PD.
Collapse
Affiliation(s)
- Minhong Huang
- Department of Biomedical Sciences, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Alejandra Bargues-Carot
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Zainab Riaz
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Hannah Wickham
- Department of Biomedical Sciences, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Gary Zenitsky
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Huajun Jin
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Vellareddy Anantharam
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Arthi Kanthasamy
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Anumantha G. Kanthasamy
- Department of Biomedical Sciences, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| |
Collapse
|
78
|
Al-Ghraiybah NF, Wang J, Alkhalifa AE, Roberts AB, Raj R, Yang E, Kaddoumi A. Glial Cell-Mediated Neuroinflammation in Alzheimer's Disease. Int J Mol Sci 2022; 23:10572. [PMID: 36142483 PMCID: PMC9502483 DOI: 10.3390/ijms231810572] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder; it is the most common cause of dementia and has no treatment. It is characterized by two pathological hallmarks, the extracellular deposits of amyloid beta (Aβ) and the intraneuronal deposits of Neurofibrillary tangles (NFTs). Yet, those two hallmarks do not explain the full pathology seen with AD, suggesting the involvement of other mechanisms. Neuroinflammation could offer another explanation for the progression of the disease. This review provides an overview of recent advances on the role of the immune cells' microglia and astrocytes in neuroinflammation. In AD, microglia and astrocytes become reactive by several mechanisms leading to the release of proinflammatory cytokines that cause further neuronal damage. We then provide updates on neuroinflammation diagnostic markers and investigational therapeutics currently in clinical trials to target neuroinflammation.
Collapse
Affiliation(s)
- Nour F. Al-Ghraiybah
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Junwei Wang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Amer E. Alkhalifa
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Andrew B. Roberts
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Ruchika Raj
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Euitaek Yang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| |
Collapse
|
79
|
Alhasan KA, Alshuaibi W, Hamad MH, Salim S, Jamjoom DZ, Alhashim AH, AlGhamdi MA, Kentab AY, Bashiri FA. Hypermanganesemia with Dystonia Type 2: A Potentially Treatable Neurodegenerative Disorder: A Case Series in a Tertiary University Hospital. CHILDREN 2022; 9:children9091335. [PMID: 36138644 PMCID: PMC9497897 DOI: 10.3390/children9091335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 11/16/2022]
Abstract
Importance: Hypermanganesemia with dystonia type 2 is a rare autosomal recessive neurodegenerative disorder characterized by the loss of previously acquired milestones, dystonia, parkinsonian features, a high serum manganese level, and characteristic neuroimaging findings such as bilateral and symmetrically increased T1 and decreased T2/fluid-attenuated inversion recovery signal intensity in the basal ganglia. This condition is secondary to a mutation in the SLC39A14 gene. Objective: To present a series of three cases of hypermanganesemia with dystonia type 2, which was genetically confirmed secondary to a mutation in the SLC39A14 gene, and to describe the treatment and clinical course in these cases. Design: A retrospective case series. Setting: University, Tertiary hospital. Participants: Three unrelated pediatric patients with hypermanganesemia with dystonia type 2, genetically confirmed to be secondary to a mutation in the SLC39A14 gene. Exposures: Chelation therapy using calcium disodium edetate. Main outcome(s) and measure(s): The response to chelation therapy based on clinical improvements in motor and cognition developments. Results: All three patients were started on chelation therapy using calcium disodium edetate, and two of them showed an improvement in their clinical course. The chelation therapy could alter the course of the disease and prevent deterioration in the clinical setting. Conclusions and Relevance: Early diagnosis and intervention with chelating agents, such as calcium disodium edetate, will help change the outcome in patients with hypermanganesemia with dystonia type 2. This finding highlights the importance of early diagnosis and treatment in improving the outcomes of patients with treatable neurodegenerative disorders.
Collapse
Affiliation(s)
- Khalid A. Alhasan
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
- Division of Pediatric Nephrology, Department of Pediatrics, King Saud University Medical City, Riyadh 11461, Saudi Arabia
| | - Walaa Alshuaibi
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
- Division of Medical Genetics, Department of Pediatrics, King Saud University Medical City, Riyadh 11461, Saudi Arabia
| | - Muddathir H. Hamad
- Division of Pediatric Neurology, Department of Pediatrics, King Saud University Medical City, Riyadh 11461, Saudi Arabia
| | - Suha Salim
- Division of Pediatric Nephrology, Department of Pediatrics, King Saud University Medical City, Riyadh 11461, Saudi Arabia
| | - Dima Z. Jamjoom
- Department of Radiology and Medical Imaging, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Aqeela H. Alhashim
- Pediatric Neurology Department, King Fahad Medical City, Riyadh 11525, Saudi Arabia
| | - Malak Ali AlGhamdi
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
- Division of Medical Genetics, Department of Pediatrics, King Saud University Medical City, Riyadh 11461, Saudi Arabia
| | - Amal Y. Kentab
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
- Division of Pediatric Neurology, Department of Pediatrics, King Saud University Medical City, Riyadh 11461, Saudi Arabia
| | - Fahad A. Bashiri
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
- Division of Pediatric Neurology, Department of Pediatrics, King Saud University Medical City, Riyadh 11461, Saudi Arabia
- Correspondence: ; Tel.: +966-118066331
| |
Collapse
|
80
|
Liu K, Liu Z, Liu Z, Ma Z, Deng Y, Liu W, Xu B. Manganese induces S-nitrosylation of PINK1 leading to nerve cell damage by repressing PINK1/Parkin-mediated mitophagy. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 834:155358. [PMID: 35460769 DOI: 10.1016/j.scitotenv.2022.155358] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
Chronic exposure to excess manganese (Mn) causes neurotoxicity, which is characterized by Parkinson-like symptoms and referred to as manganism. In the last few decades, mitochondrial damage and subsequent energy failure have been reported to be important mechanisms of Mn toxicity, yet how Mn causes mitochondrial damage remains largely unknown. Here, we demonstrated that Mn induced S-nitrosation of phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1), a master regulator in the mitophagy pathway, results in dysregulation of mitophagy and nerve cell injury in the rat striatum. We cultured primary neurons and used 1400 W, a potent and selective inducible nitric oxide synthase (iNOS) inhibitor, as an intervention to verify the precise mechanism of Mn-induced dysregulation of mitophagy. We demonstrated that Mn-induced S-nitrosylation of PINK1 decreased the phosphorylated level of parkin RBR E3 ubiquitin-protein ligase (Parkin), as well as the translocation of Parkin to damaged mitochondria, which led to the accumulation of damaged mitochondria and mitochondrial-mediated apoptosis. Our findings indicated the unusual connection between nitrative stress and mitochondrial dysfunction in Mn-induced neurotoxicity. These data highlight the role of S-nitrosation of PINK1 in Mn-induced dysregulation of mitophagy and provide a reliable target for the development of specific drugs and the early treatment of manganism, which has important theoretical and practical significance.
Collapse
Affiliation(s)
- Kuan Liu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, People's Republic of China
| | - Zhiqi Liu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, People's Republic of China
| | - Zhuofan Liu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, People's Republic of China
| | - Zhuo Ma
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 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 Province 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 Province 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 Province 110122, People's Republic of China.
| |
Collapse
|
81
|
Liu X, Yao C, Tang Y, Liu X, Duan C, Wang C, Han F, Xiang Y, Wu L, Li Y, Ji A, Cai T. Role of p53 methylation in manganese-induced cyclooxygenase-2 expression in BV2 microglial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 241:113824. [PMID: 36068751 DOI: 10.1016/j.ecoenv.2022.113824] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/14/2022] [Accepted: 06/27/2022] [Indexed: 05/21/2023]
Abstract
Manganese (Mn) is an essential cofactor for many enzymes and plays an important role in normal growth and development. However, excess exposure to manganese (Mn) may be an important environmental factor leading to neurodegeneration. The overexpression of microglial cyclooxygenase-2 (COX-2) plays a key role in neuroinflammation in neurodegenerative diseases. The existing data suggest that Mn can induce neuroinflammation by up-regulating COX-2 expression. However, the mechanisms involved in Mn-induced microglial COX-2 up-regulation remain to be determined. The aim of this study was to investigate the role of p53 in Mn-induced COX-2 expression in microglial cells. The results showed that Mn exposure induced the up-regulation of COX-2 and inhibited the expression of p53 in BV2 microglial cells. The addition of p53 activator and the over-expression of p53 blocked the expression of COX-2 and prostaglandin E2 (PGE2), a COX-2 downstream effector, induced by Mn. Further, Mn increased the methylation of p53 DNA in microglia, while the addition of demethylation reagent 5-Aza-dC enhanced the expression of p53 but decreased the expression of COX-2. These results suggested that Mn may inhibit p53 expression through induction of DNA methylation, which can further induce the expression of COX-2 in microglial cells.
Collapse
Affiliation(s)
- Xiaoling Liu
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Chunyan Yao
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yan Tang
- Experimental Teaching Center, School of Public Health, Southwest Medical University, Luzhou, China
| | - Xiaoyan Liu
- The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Chenggang Duan
- Department of Pathophysiology, Southwest Medical University, Luzhou, China
| | - Chunmei Wang
- Experimental Teaching Center, School of Public Health, Southwest Medical University, Luzhou, China
| | - Fei Han
- Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ying Xiang
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Long Wu
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yafei Li
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ailing Ji
- Department of Preventive Medicine & Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing, China.
| | - Tongjian Cai
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
82
|
Nicastro R, Gaillard H, Zarzuela L, Péli-Gulli MP, Fernández-García E, Tomé M, García-Rodríguez N, Durán RV, De Virgilio C, Wellinger RE. Manganese is a physiologically relevant TORC1 activator in yeast and mammals. eLife 2022; 11:80497. [PMID: 35904415 PMCID: PMC9337852 DOI: 10.7554/elife.80497] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/18/2022] [Indexed: 12/09/2022] Open
Abstract
The essential biometal manganese (Mn) serves as a cofactor for several enzymes that are crucial for the prevention of human diseases. Whether intracellular Mn levels may be sensed and modulate intracellular signaling events has so far remained largely unexplored. The highly conserved target of rapamycin complex 1 (TORC1, mTORC1 in mammals) protein kinase requires divalent metal cofactors such as magnesium (Mg2+) to phosphorylate effectors as part of a homeostatic process that coordinates cell growth and metabolism with nutrient and/or growth factor availability. Here, our genetic approaches reveal that TORC1 activity is stimulated in vivo by elevated cytoplasmic Mn levels, which can be induced by loss of the Golgi-resident Mn2+ transporter Pmr1 and which depend on the natural resistance-associated macrophage protein (NRAMP) metal ion transporters Smf1 and Smf2. Accordingly, genetic interventions that increase cytoplasmic Mn2+ levels antagonize the effects of rapamycin in triggering autophagy, mitophagy, and Rtg1-Rtg3-dependent mitochondrion-to-nucleus retrograde signaling. Surprisingly, our in vitro protein kinase assays uncovered that Mn2+ activates TORC1 substantially better than Mg2+, which is primarily due to its ability to lower the Km for ATP, thereby allowing more efficient ATP coordination in the catalytic cleft of TORC1. These findings, therefore, provide both a mechanism to explain our genetic observations in yeast and a rationale for how fluctuations in trace amounts of Mn can become physiologically relevant. Supporting this notion, TORC1 is also wired to feedback control mechanisms that impinge on Smf1 and Smf2. Finally, we also show that Mn2+-mediated control of TORC1 is evolutionarily conserved in mammals, which may prove relevant for our understanding of the role of Mn in human diseases.
Collapse
Affiliation(s)
- Raffaele Nicastro
- University of Fribourg, Department of Biology, Fribourg, Switzerland
| | - Hélène Gaillard
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Seville, Spain.,Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| | - Laura Zarzuela
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Seville, Spain
| | | | - Elisabet Fernández-García
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Seville, Spain.,Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| | - Mercedes Tomé
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Seville, Spain
| | - Néstor García-Rodríguez
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Seville, Spain.,Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| | - Raúl V Durán
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Seville, Spain
| | | | - Ralf Erik Wellinger
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Seville, Spain.,Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
83
|
Shan S, Zhang Y, Zhao H, Zeng T, Zhao X. Polystyrene nanoplastics penetrate across the blood-brain barrier and induce activation of microglia in the brain of mice. CHEMOSPHERE 2022; 298:134261. [PMID: 35302003 DOI: 10.1016/j.chemosphere.2022.134261] [Citation(s) in RCA: 150] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/21/2022] [Accepted: 03/06/2022] [Indexed: 06/14/2023]
Abstract
Microplastics (MPs) have been well demonstrated as potential threats to the ecosystem, whereas the neurotoxicity of MPs in mammals remains to be elucidated. The current study was designed to investigate whether 50 nm polystyrene nanoplastics (PS-NPs) could pass through the blood-brain barrier (BBB), and to elucidate the underlying mechanisms and the following neurotoxic manifestation. In vivo study showed that PS-NPs (0.5-50 mg/kg. bw PS-NPs for 7 days) significantly induced the increase of permeability of BBB, and dose-dependently accumulated in the brain of mice. In addition, PS-NPs were found to be present in microglia, and induced microglia activation and neuron damage in the mouse brain. In vitro studies using the immortalized human cerebral microvascular endothelial cell (hCMEC/D3), the most commonly used cell model for BBB-related studies, revealed that PS-NPs could be internalized into cells, and caused reactive oxygen species (ROS) production, nuclear factor kappa-B (NF-κB) activation, tumor necrosis factors α (TNF-α) secretion, and necroptosis of hCMEC/D3 cells. Furthermore, PS-NPs exposure led to disturbance of the tight junction (TJ) formed by hCMEC/D3, as demonstrated by the decline of transendothelial electrical resistance (TEER) and decreased expression of occludin. Lastly, PS-NPs exposure resulted in the activation of murine microglia BV2 cells, and the cell medium of PS-NPs-exposed BV2 induced obvious damage to murine neuron HT-22 cells. Collectively, these results suggest that PS-NPs could pass through BBB and induce neurotoxicity in mammals probably by inducing activation of microglia.
Collapse
Affiliation(s)
- Shan Shan
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yifan Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huiwen Zhao
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Xiulan Zhao
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
84
|
Ferreira SA, Loreto JS, Dos Santos MM, Barbosa NV. Environmentally relevant manganese concentrations evoke anxiety phenotypes in adult zebrafish. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 93:103870. [PMID: 35523392 DOI: 10.1016/j.etap.2022.103870] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/23/2022] [Accepted: 04/26/2022] [Indexed: 06/14/2023]
Abstract
Manganese (Mn) is an essential metal for living organisms. However, the excess of Mn can be toxic, especially for the central nervous system. Herein, we used adult zebrafish as model organism to investigate the relationship of an environmentally relevant Mn exposure with the onset of neurobehavioral disturbances and brain biochemical alterations. Fish were exposed to MnCl2 at 0.5, 2.0, 7.5 and 15.0 mg/L for 96 h, and after submitted to trials for examining exploratory, locomotor and anxiety-related behaviors. The neurobehavioral parameters were followed by the analyses of cell viability, Mn accumulation and acetylcholinesterase activity in the brain, and whole-body cortisol levels. By Novel tank, Light dark and Social preference test, we found that the exposure to Mn, along with locomotor deficits induced anxiety-like phenotypes in zebrafish. Most of these behavioral changes were evoked by the highest concentrations, which also caused cell viability loss, higher accumulation of Mn and increased AChE activity in the brain, and an increase in the whole-body cortisol content. Our findings demonstrated that zebrafish are quite sensitive to levels of Mn found in the environment, and that the magnitude of the neurotoxic effects may be associated with the levels of manganese accumulated in the brain. Interestingly, we showed that Mn exposure in addition to motor deficits may also cause psychiatric abnormalities, namely anxiety.
Collapse
Affiliation(s)
- Sabrina Antunes Ferreira
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, Santa Maria, RS 97105-900, Brazil
| | - Julia Sepel Loreto
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, Santa Maria, RS 97105-900, Brazil
| | - Matheus Mülling Dos Santos
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, Santa Maria, RS 97105-900, Brazil
| | - Nilda Vargas Barbosa
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, Santa Maria, RS 97105-900, Brazil.
| |
Collapse
|
85
|
Kim H, Harrison FE, Aschner M, Bowman AB. Exposing the role of metals in neurological disorders: a focus on manganese. Trends Mol Med 2022; 28:555-568. [PMID: 35610122 PMCID: PMC9233117 DOI: 10.1016/j.molmed.2022.04.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/14/2022] [Accepted: 04/25/2022] [Indexed: 02/06/2023]
Abstract
Metals are ubiquitous chemical entities involved in a myriad of biological processes. Despite their integral role in sustaining life, overexposure can lead to deleterious neurological outcomes posing a public health concern. Excess exposure to metals has been associated with aberrant neurodevelopmental and neurodegenerative diseases and prominently contributes to environmental risk for neurological disorders. Here, we use manganese (Mn) to exemplify the gap in our understanding of the mechanisms behind acute metal toxicity and their relationship to chronic toxicity and disease. This challenge frustrates understanding of how individual exposure histories translate into preventing and treating brain diseases from childhood through old age. We discuss ways to enhance the predictive value of preclinical models and define mechanisms of chronic, persistent, and latent neurotoxicity.
Collapse
Affiliation(s)
- Hyunjin Kim
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Fiona E Harrison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
86
|
Xu Y, Peng T, Xiang Y, Liao G, Zou F, Meng X. Neurotoxicity and gene expression alterations in zebrafish larvae in response to manganese exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 825:153778. [PMID: 35150691 DOI: 10.1016/j.scitotenv.2022.153778] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/03/2022] [Accepted: 02/06/2022] [Indexed: 06/14/2023]
Abstract
Manganese (Mn) is an essential trace element, but excessive exposure can damage mental, cognitive, and motor functions. Although many studies have reported the toxicity of Mn, the underlying mechanism remains unclear. Here, wild-type and/or Tg(NBT:DsRed) zebrafish embryos/larvae were exposed to different dosages of Mn to determine the effects on mortality, malformation, and hatching rates. A video tracking system was used to analyze the locomotor activities of zebrafish larvae. The terminal deoxynucleotidyl transferase dUTP nick end labeling assay and acridine orange staining were performed to monitor cell apoptosis, while dopamine transporter and tyrosine hydroxylase (TH) expression were detected by immunohistochemical staining. Meanwhile, transcriptome sequencing of the head tissues of zebrafish larvae was performed to search for molecular targets of Mn neurotoxicity. The results showed that Mn exposure increased the mortality and malformation rates of zebrafish larvae, and significantly reduced swim distance and velocity. In addition, the proportion of apoptotic dopaminergic neurons increased, while TH expression significantly decreased. The results of transcriptome sequencing showed that a large number of differentially expressed genes associated with apoptosis and DNA damage repair were upregulated, consistent with the above results. Meanwhile, Western blot analysis showed that higher exposure level of Mn could induce activation of MAPK pathway. These data demonstrate that Mn exposure can damage dopaminergic neurons and cause apoptosis, which has detrimental effects on the motor abilities of zebrafish larvae.
Collapse
Affiliation(s)
- Yongjie Xu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Tao Peng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Yang Xiang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Gengze Liao
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
87
|
Berger MM, Shenkin A, Schweinlin A, Amrein K, Augsburger M, Biesalski HK, Bischoff SC, Casaer MP, Gundogan K, Lepp HL, de Man AME, Muscogiuri G, Pietka M, Pironi L, Rezzi S, Cuerda C. ESPEN micronutrient guideline. Clin Nutr 2022; 41:1357-1424. [PMID: 35365361 DOI: 10.1016/j.clnu.2022.02.015] [Citation(s) in RCA: 230] [Impact Index Per Article: 76.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Trace elements and vitamins, named together micronutrients (MNs), are essential for human metabolism. Recent research has shown the importance of MNs in common pathologies, with significant deficiencies impacting the outcome. OBJECTIVE This guideline aims to provide information for daily clinical nutrition practice regarding assessment of MN status, monitoring, and prescription. It proposes a consensus terminology, since many words are used imprecisely, resulting in confusion. This is particularly true for the words "deficiency", "repletion", "complement", and "supplement". METHODS The expert group attempted to apply the 2015 standard operating procedures (SOP) for ESPEN which focuses on disease. However, this approach could not be applied due to the multiple diseases requiring clinical nutrition resulting in one text for each MN, rather than for diseases. An extensive search of the literature was conducted in the databases Medline, PubMed, Cochrane, Google Scholar, and CINAHL. The search focused on physiological data, historical evidence (published before PubMed release in 1996), and observational and/or randomized trials. For each MN, the main functions, optimal analytical methods, impact of inflammation, potential toxicity, and provision during enteral or parenteral nutrition were addressed. The SOP wording was applied for strength of recommendations. RESULTS There was a limited number of interventional trials, preventing meta-analysis and leading to a low level of evidence. The recommendations underwent a consensus process, which resulted in a percentage of agreement (%): strong consensus required of >90% of votes. Altogether the guideline proposes sets of recommendations for 26 MNs, resulting in 170 single recommendations. Critical MNs were identified with deficiencies being present in numerous acute and chronic diseases. Monitoring and management strategies are proposed. CONCLUSION This guideline should enable addressing suboptimal and deficient status of a bundle of MNs in at-risk diseases. In particular, it offers practical advice on MN provision and monitoring during nutritional support.
Collapse
Affiliation(s)
- Mette M Berger
- Department of Adult Intensive Care, Lausanne University Hospital (CHUV), Lausanne, Switzerland.
| | - Alan Shenkin
- Institute of Aging and Chronic Disease, University of Liverpool, Liverpool, UK.
| | - Anna Schweinlin
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany.
| | - Karin Amrein
- Medical University of Graz, Department of Internal Medicine, Division of Endocrinology and Diabetology, Austria.
| | - Marc Augsburger
- University Centre of Legal Medicine Lausanne-Geneva, Lausanne University Hospital and University of Lausanne, Geneva University Hospital and University of Geneva, Lausanne-Geneva, Switzerland.
| | | | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany.
| | - Michael P Casaer
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Intensive Care Medicine, Leuven, Belgium.
| | - Kursat Gundogan
- Division of Intensive Care Medicine, Department of Internal Medicine, Erciyes University School of Medicine, Kayseri, Turkey.
| | | | - Angélique M E de Man
- Department of Intensive Care Medicine, Research VUmc Intensive Care (REVIVE), Amsterdam Cardiovascular Science (ACS), Amsterdam Infection and Immunity Institute (AI&II), Amsterdam Medical Data Science (AMDS), Amsterdam UMC, Location VUmc, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands.
| | - Giovanna Muscogiuri
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy; United Nations Educational, Scientific and Cultural Organization (UNESCO) Chair for Health Education and Sustainable Development, Federico II, University, Naples, Italy.
| | - Magdalena Pietka
- Pharmacy Department, Stanley Dudrick's Memorial Hospital, Skawina, Poland.
| | - Loris Pironi
- Alma Mater Studiorum - University of Bologna, Department of Medical and Surgical Sciences, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, Centre for Chronic Intestinal Failure - Clinical Nutrition and Metabolism Unit, Italy.
| | - Serge Rezzi
- Swiss Nutrition and Health Foundation (SNHf), Epalinges, Switzerland.
| | - Cristina Cuerda
- Departamento de Medicina, Universidad Complutense de Madrid, Nutrition Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| |
Collapse
|
88
|
Long noncoding RNA Sh2d3c promotes manganese-induced neuronal apoptosis through the mmu-miR-675-5p/Chmp4b/Bax axis. Toxicol Lett 2022; 365:24-35. [DOI: 10.1016/j.toxlet.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/30/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022]
|
89
|
Tuschl K, White RJ, Trivedi C, Valdivia LE, Niklaus S, Bianco IH, Dadswell C, González-Méndez R, Sealy IM, Neuhauss SCF, Houart C, Rihel J, Wilson SW, Busch-Nentwich EM. Loss of slc39a14 causes simultaneous manganese hypersensitivity and deficiency in zebrafish. Dis Model Mech 2022; 15:dmm044594. [PMID: 35514229 PMCID: PMC9227717 DOI: 10.1242/dmm.044594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/25/2022] [Indexed: 12/15/2022] Open
Abstract
Manganese neurotoxicity is a hallmark of hypermanganesemia with dystonia 2, an inherited manganese transporter defect caused by mutations in SLC39A14. To identify novel potential targets of manganese neurotoxicity, we performed transcriptome analysis of slc39a14-/- mutant zebrafish that were exposed to MnCl2. Differentially expressed genes mapped to the central nervous system and eye, and pathway analysis suggested that Ca2+ dyshomeostasis and activation of the unfolded protein response are key features of manganese neurotoxicity. Consistent with this interpretation, MnCl2 exposure led to decreased whole-animal Ca2+ levels, locomotor defects and changes in neuronal activity within the telencephalon and optic tectum. In accordance with reduced tectal activity, slc39a14-/- zebrafish showed changes in visual phototransduction gene expression, absence of visual background adaptation and a diminished optokinetic reflex. Finally, numerous differentially expressed genes in mutant larvae normalised upon MnCl2 treatment indicating that, in addition to neurotoxicity, manganese deficiency is present either subcellularly or in specific cells or tissues. Overall, we assembled a comprehensive set of genes that mediate manganese-systemic responses and found a highly correlated and modulated network associated with Ca2+ dyshomeostasis and cellular stress. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Karin Tuschl
- UCL GOS Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
- Department of Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, IoPPN, Kings College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Richard J. White
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London E1 4NS, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Chintan Trivedi
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Leonardo E. Valdivia
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Camino La Pirámide 5750, Huechuraba 8580745, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Camino La Pirámide 5750, Huechuraba 8580745, Chile
| | - Stephanie Niklaus
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Isaac H. Bianco
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Chris Dadswell
- School of Life Sciences, University of Sussex, Brighton BN1 9QJ, UK
| | | | - Ian M. Sealy
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London E1 4NS, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Stephan C. F. Neuhauss
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Corinne Houart
- Department of Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, IoPPN, Kings College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Stephen W. Wilson
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Elisabeth M. Busch-Nentwich
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London E1 4NS, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| |
Collapse
|
90
|
Durin Z, Houdou M, Morelle W, Barré L, Layotte A, Legrand D, Ouzzine M, Foulquier F. Differential Effects of D-Galactose Supplementation on Golgi Glycosylation Defects in TMEM165 Deficiency. Front Cell Dev Biol 2022; 10:903953. [PMID: 35693943 PMCID: PMC9178294 DOI: 10.3389/fcell.2022.903953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Glycosylation is a ubiquitous and universal cellular process in all domains of life. In eukaryotes, many glycosylation pathways occur simultaneously onto proteins and lipids for generating a complex diversity of glycan structures. In humans, severe genetic diseases called Congenital Disorders of Glycosylation (CDG), resulting from glycosylation defects, demonstrate the functional relevance of these processes. No real cure exists so far, but oral administration of specific monosaccharides to bypass the metabolic defects has been used in few CDG, then constituting the simplest and safest treatments. Oral D-Galactose (Gal) therapy was seen as a promising tailored treatment for specific CDG and peculiarly for TMEM165-CDG patients. TMEM165 deficiency not only affects the N-glycosylation process but all the other Golgi-related glycosylation types, then contributing to the singularity of this defect. Our previous results established a link between TMEM165 deficiency and altered Golgi manganese (Mn2+) homeostasis. Besides the fascinating power of MnCl2 supplementation to rescue N-glycosylation in TMEM165-deficient cells, D-Gal supplementation has also been shown to be promising in suppressing the observed N-glycosylation defects. Its effect on the other Golgi glycosylation types, most especially O-glycosylation and glycosaminoglycan (GAG) synthesis, was however unknown. In the present study, we demonstrate the differential impact of D-Gal or MnCl2 supplementation effects on the Golgi glycosylation defects caused by TMEM165 deficiency. Whereas MnCl2 supplementation unambiguously fully rescues the N- and O-linked as well as GAG glycosylations in TMEM165-deficient cells, D-Gal supplementation only rescues the N-linked glycosylation, without any effects on the other Golgi-related glycosylation types. According to these results, we would recommend the use of MnCl2 for TMEM165-CDG therapy.
Collapse
Affiliation(s)
- Zoé Durin
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Marine Houdou
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Willy Morelle
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Lydia Barré
- Faculty of Medicine, UMR7365 CNRS-University of Lorraine, Biopôle, Nancy, France
| | - Aurore Layotte
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Dominique Legrand
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Mohamed Ouzzine
- Faculty of Medicine, UMR7365 CNRS-University of Lorraine, Biopôle, Nancy, France
| | - François Foulquier
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
- *Correspondence: François Foulquier,
| |
Collapse
|
91
|
Nabi M, Tabassum N. Role of Environmental Toxicants on Neurodegenerative Disorders. FRONTIERS IN TOXICOLOGY 2022; 4:837579. [PMID: 35647576 PMCID: PMC9131020 DOI: 10.3389/ftox.2022.837579] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/22/2022] [Indexed: 12/22/2022] Open
Abstract
Neurodegeneration leads to the loss of structural and functioning components of neurons over time. Various studies have related neurodegeneration to a number of degenerative disorders. Neurological repercussions of neurodegeneration can have severe impacts on the physical and mental health of patients. In the recent past, various neurodegenerative ailments such as Alzheimer’s and Parkinson’s illnesses have received global consideration owing to their global occurrence. Environmental attributes have been regarded as the main contributors to neural dysfunction-related disorders. The majority of neurological diseases are mainly related to prenatal and postnatal exposure to industrially produced environmental toxins. Some neurotoxic metals, like lead (Pb), aluminium (Al), Mercury (Hg), manganese (Mn), cadmium (Cd), and arsenic (As), and also pesticides and metal-based nanoparticles, have been implicated in Parkinson’s and Alzheimer’s disease. The contaminants are known for their ability to produce senile or amyloid plaques and neurofibrillary tangles (NFTs), which are the key features of these neurological dysfunctions. Besides, solvent exposure is also a significant contributor to neurological diseases. This study recapitulates the role of environmental neurotoxins on neurodegeneration with special emphasis on major neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease.
Collapse
Affiliation(s)
- Masarat Nabi
- Department of Environmental Science, University of Kashmir, Srinagar, India
- *Correspondence: Masarat Nabi, , orcid.org/0000-0003-1677-6498; Nahida Tabassum,
| | - Nahida Tabassum
- Department of Pharmaceutical Sciences, University of Kashmir, Srinagar, India
- *Correspondence: Masarat Nabi, , orcid.org/0000-0003-1677-6498; Nahida Tabassum,
| |
Collapse
|
92
|
Wu Z, Heineman WR, Haynes EN, Papautsky I. Electrochemical Determination of Manganese in Whole Blood with Indium Tin Oxide Electrode. JOURNAL OF THE ELECTROCHEMICAL SOCIETY 2022; 169:057508. [PMID: 35755409 PMCID: PMC9229665 DOI: 10.1149/1945-7111/ac6a19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
In this work, we demonstrate accurate and precise measurement of manganese (Mn) concentration in human whole blood with indium tin oxide (ITO) electrode using square wave stripping voltammetry. While an essential trace metal for human health, elevated levels of Mn due to environmental or occupational exposure have been associated with severe neuromotor dysfunction characterized by parkinsonism and cognitive dysfunction making the monitoring of Mn in whole blood necessary. Pediatric populations are particularly susceptible to Mn given their developing brain and potential long-term impacts on neurodevelopment. The current gold standard for whole blood Mn measurements is by ICP-MS, which is costly and time consuming. The electrochemical detection with ITO working electrode in this work showed a limit of detection of 0.5 μg l-1 and a linear range of 5 to 500 μg l-1, which encompasses the physiological Mn levels in human whole blood (5-18 μg l-1). Our results of Mn measurement in whole blood show an average precision of 96.5% and an average accuracy of 90.3% compared to ICP-MS for both the normal range (5-18 μg l-1) and the elevated levels (>36 μg l-1) that require medical intervention. These results demonstrate the feasibility of Mn measurements in human blood with electrochemical sensors.
Collapse
Affiliation(s)
- Zhizhen Wu
- Department of Biomedical Engineering, University of Illinois Chicago, Illinois 60607, USA
| | - William R Heineman
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, USA
| | - Erin N Haynes
- Department of Epidemiology and Preventive Medicine and Environmental Health, University of Kentucky, Kentucky 40536, USA
| | - Ian Papautsky
- Department of Biomedical Engineering, University of Illinois Chicago, Illinois 60607, USA
| |
Collapse
|
93
|
Jensen N, Terrell R, Ramoju S, Shilnikova N, Farhat N, Karyakina N, Cline BH, Momoli F, Mattison D, Krewski D. Magnetic resonance imaging T1 indices of the brain as biomarkers of inhaled manganese exposure. Crit Rev Toxicol 2022; 52:358-370. [PMID: 36412542 DOI: 10.1080/10408444.2022.2128719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Excessive exposure to manganese (Mn) is linked to its accumulation in the brain and adverse neurological effects. Paramagnetic properties of Mn allow the use of magnetic resonance imaging (MRI) techniques to identify it in biological tissues. A critical review was conducted to evaluate whether MRI techniques could be used as a diagnostic tool to detect brain Mn accumulation as a quantitative biomarker of inhaled exposure. A comprehensive search was conducted in MEDLINE, EMBASE, and PubMed to identify potentially relevant studies published prior to 9 May 2022. Two reviewers independently screened identified references using a two-stage process. Of the 6452 unique references identified, 36 articles were retained for data abstraction. Eligible studies used T1-weighted MRI techniques and reported direct or indirect T1 measures to characterize Mn accumulation in the brain. Findings demonstrate that, in subjects exposed to high levels of Mn, deposition in the brain is widespread, accumulating both within and outside the basal ganglia. Available evidence indicates that T1 MRI techniques can be used to distinguish Mn-exposed individuals from unexposed. Additionally, T1 MRI may be useful for semi-quantitative evaluation of inhaled Mn exposure, particularly when interpreted along with other exposure indices. T1 MRI measures appear to have a nonlinear relationship to Mn exposure duration, with R1 signal only increasing after critical thresholds. The strength of the association varied depending on the regions of interest imaged and the method of exposure measurement. Overall, available evidence suggests potential for future clinical and risk assessment applications of MRI as a diagnostic tool.
Collapse
Affiliation(s)
- N Jensen
- Risk Sciences International, Ottawa, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | - R Terrell
- Risk Sciences International, Ottawa, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | - S Ramoju
- Risk Sciences International, Ottawa, Canada
| | - N Shilnikova
- Risk Sciences International, Ottawa, Canada.,McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Canada
| | - N Farhat
- Risk Sciences International, Ottawa, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada.,School of Mathematics and Statistics, Carleton University, Ottawa, Canada
| | - N Karyakina
- Risk Sciences International, Ottawa, Canada.,McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Canada
| | - B H Cline
- International Manganese Institute, Paris, France
| | - F Momoli
- Risk Sciences International, Ottawa, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | - D Mattison
- Risk Sciences International, Ottawa, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada.,Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - D Krewski
- Risk Sciences International, Ottawa, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada.,McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Canada.,School of Mathematics and Statistics, Carleton University, Ottawa, Canada
| |
Collapse
|
94
|
Mitra S, Chakraborty AJ, Tareq AM, Emran TB, Nainu F, Khusro A, Idris AM, Khandaker MU, Osman H, Alhumaydhi FA, Simal-Gandara J. Impact of heavy metals on the environment and human health: Novel therapeutic insights to counter the toxicity. JOURNAL OF KING SAUD UNIVERSITY - SCIENCE 2022; 34:101865. [DOI: 10.1016/j.jksus.2022.101865] [Citation(s) in RCA: 326] [Impact Index Per Article: 108.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
95
|
DHASARATHAN S, SHUNMUGAPERUMAL S, SELVARAJ P K. Exploration of Role of Concentration on Sensing Activities using Novel unsymmetrical Schiff bases. JOURNAL OF THE TURKISH CHEMICAL SOCIETY, SECTION A: CHEMISTRY 2022. [DOI: 10.18596/jotcsa.1008926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
96
|
LA-ICP-MS bioimaging demonstrated disturbance of metal ions in the brain of Parkinson's disease model mouse undergoing manganese-enhanced MRI. Anal Bioanal Chem 2022; 414:5561-5571. [PMID: 35275218 DOI: 10.1007/s00216-022-03994-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/10/2022] [Accepted: 02/25/2022] [Indexed: 11/01/2022]
Abstract
Manganese-enhanced MRI (MEMRI) is a powerful tool to study neuronal activity and microarchitecture in vivo. Yet the influence of exogenous manganese on the brain of the Parkinson's disease (PD) model mouse is poorly understood. Laser ablation connected to inductively coupled plasma mass spectrometry (LA-ICP-MS) imaging for tissue section is an ideal tool to simultaneously analyze the metabolism of endogenous metal ions. In this study, DJ-1 knockout PD model mice were subjected to an MnCl2 saline treatment and the distribution of Mn and several other endogenous metal ions in brain regions was assessed by MEMRI and LA-ICP-MS imaging. The results demonstrated that Mn mainly deposited in subcortical regions, such as ventricles, hippocampus (HC), medial preoptic nucleus (MPO), lateral septal nucleus (LS), and ventromedial hypothalamic nucleus (VMH). The enhanced signal-to-noise ratio (S/N) determined by MEMRI for Mn is closely related to the signal in LA-ICP-MS imaging. Significantly, the treatment of MnCl2 disturbs the homeostasis of iron, zinc, copper, and calcium in the DJ-1 mouse, which could result in more severe symptoms of PD. Therefore, the application of MEMRI in the study of neurological disease must be made with caution.
Collapse
|
97
|
Manganese Exposure and Metabolic Syndrome: A Systematic Review and Meta-Analysis. Nutrients 2022; 14:nu14040825. [PMID: 35215474 PMCID: PMC8876230 DOI: 10.3390/nu14040825] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 12/11/2022] Open
Abstract
Manganese (Mn) is an essential element acting as a co-factor of superoxide dismutase, and it is potentially beneficial for cardiometabolic health by reducing oxidative stress. Although some studies have examined the relationship between Mn and metabolic syndrome (MetS), no systematic review and meta-analysis has been presented to summarize the evidence. Therefore, the present review examined the association between dietary and environmental Mn exposure, and MetS risk. A total of nine cross-sectional studies and three case-control studies were included, which assessed Mn from diet, serum, urine, and whole blood. The association of the highest Mn level from diet (three studies, odds ratio (OR): 0.83, 95% confidence interval (C.I.) = 0.57, 1.21), serum (two studies, OR: 0.87, 95% C.I. = 0.66, 1.14), urine (two studies, OR: 0.84, 95% C.I. = 0.59, 1.19), and whole blood (two studies, OR: 0.92, 95% C.I. = 0.53, 1.60) were insignificant, but some included studies have suggested a non-linear relationship of urinary and blood Mn with MetS, and higher dietary Mn may associate with a lower MetS risk in some of the included studies. While more evidence from prospective cohorts is needed, future studies should use novel statistical approaches to evaluate relative contribution of Mn on MetS risk along with other inter-related exposures.
Collapse
|
98
|
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: 7] [Impact Index Per Article: 2.3] [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
|
99
|
Błażewicz A, Grywalska E, Macek P, Mertowska P, Mertowski S, Wojnicka J, Durante N, Makarewicz A. Research into the Association of Cadmium and Manganese Excretion with Thyroid Function and Behavioral Areas in Adolescents with Autism Spectrum Disorders. J Clin Med 2022; 11:jcm11030579. [PMID: 35160030 PMCID: PMC8837100 DOI: 10.3390/jcm11030579] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/10/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023] Open
Abstract
Thyroid dysfunction and toxic metal exposure have been linked to the increased risk of autism spectrum disorders (ASD); however, the relationship between those factors remains unclear. We aimed to evaluate the relationship between the serum level of hormones, namely thyroid-stimulating hormone (TSH), free triiodothyronine (fT3), free thyroxine (fT4), and urinary cadmium (U-Cd) and urinary manganese (U-Mn), in patients with ASD. The study group consisted of 129 adolescents with ASD, and the control group consisted of 86 healthy persons. Ion chromatography with spectrophotometric detection (IC-UV/ViS) was used to quantitatively determine Cd and Mn in all 24-h urine samples. These results indicate that severity of certain symptoms in autism is associated with thyroid function. Correlation analysis between Childhood Autism Rating Scale (CARS) results and the content of both U-Mn and U-Cd as well as fT3, fT4 and TSH values in ASD patients showed significantly positive correlation of CARS7 (visual reaction) with fT3 and fT4 and a negative correlation with TSH for the whole study group. In the group of adolescents over 14 years of age, it was also observed that CARS10 (anxiety reaction) negatively correlates with serum TSH levels, and among younger individuals, CARS9 (near receptor responsiveness, taste, smell) positively correlates with TSH.
Collapse
Affiliation(s)
- Anna Błażewicz
- Department of Pathobiochemistry and Interdisciplinary Applications of Ion Chromatography, Medical University of Lublin, 1 Chodźki Street, 20-093 Lublin, Poland; (J.W.); (N.D.)
- Correspondence: ; Tel.: +48-81448-7300
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodźki Street, 20-093 Lublin, Poland; (E.G.); (P.M.); (S.M.)
| | - Paweł Macek
- Department of Oncology, Institute of Health Sciences, Collegium Medicum, Jan Kochanowski University, 25-713 Kielce, Poland;
- Department of Epidemiology and Cancer Control, Holycross Cancer Centre, 25-734 Kielce, Poland
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodźki Street, 20-093 Lublin, Poland; (E.G.); (P.M.); (S.M.)
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodźki Street, 20-093 Lublin, Poland; (E.G.); (P.M.); (S.M.)
| | - Julia Wojnicka
- Department of Pathobiochemistry and Interdisciplinary Applications of Ion Chromatography, Medical University of Lublin, 1 Chodźki Street, 20-093 Lublin, Poland; (J.W.); (N.D.)
| | - Nicolo Durante
- Department of Pathobiochemistry and Interdisciplinary Applications of Ion Chromatography, Medical University of Lublin, 1 Chodźki Street, 20-093 Lublin, Poland; (J.W.); (N.D.)
| | - Agata Makarewicz
- Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, 20-439 Lublin, Poland;
| |
Collapse
|
100
|
Forero-Rodríguez LJ, Josephs-Spaulding J, Flor S, Pinzón A, Kaleta C. Parkinson's Disease and the Metal-Microbiome-Gut-Brain Axis: A Systems Toxicology Approach. Antioxidants (Basel) 2021; 11:71. [PMID: 35052575 PMCID: PMC8773335 DOI: 10.3390/antiox11010071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/02/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson's Disease (PD) is a neurodegenerative disease, leading to motor and non-motor complications. Autonomic alterations, including gastrointestinal symptoms, precede motor defects and act as early warning signs. Chronic exposure to dietary, environmental heavy metals impacts the gastrointestinal system and host-associated microbiome, eventually affecting the central nervous system. The correlation between dysbiosis and PD suggests a functional and bidirectional communication between the gut and the brain. The bioaccumulation of metals promotes stress mechanisms by increasing reactive oxygen species, likely altering the bidirectional gut-brain link. To better understand the differing molecular mechanisms underlying PD, integrative modeling approaches are necessary to connect multifactorial perturbations in this heterogeneous disorder. By exploring the effects of gut microbiota modulation on dietary heavy metal exposure in relation to PD onset, the modification of the host-associated microbiome to mitigate neurological stress may be a future treatment option against neurodegeneration through bioremediation. The progressive movement towards a systems toxicology framework for precision medicine can uncover molecular mechanisms underlying PD onset such as metal regulation and microbial community interactions by developing predictive models to better understand PD etiology to identify options for novel treatments and beyond. Several methodologies recently addressed the complexity of this interaction from different perspectives; however, to date, a comprehensive review of these approaches is still lacking. Therefore, our main aim through this manuscript is to fill this gap in the scientific literature by reviewing recently published papers to address the surrounding questions regarding the underlying molecular mechanisms between metals, microbiota, and the gut-brain-axis, as well as the regulation of this system to prevent neurodegeneration.
Collapse
Affiliation(s)
- Lady Johanna Forero-Rodríguez
- Research Group Bioinformatics and Systems Biology, Instituto de Genetica, Universidad Nacional de Colombia, Bogotá 111321, Colombia; (L.J.F.-R.); (A.P.)
- Research Group Medical Systems Biology, Christian-Albrechts-Universität Kiel, Brunswiker Straße 10, 24105 Kiel, Germany; (S.F.); (C.K.)
| | - Jonathan Josephs-Spaulding
- Research Group Medical Systems Biology, Christian-Albrechts-Universität Kiel, Brunswiker Straße 10, 24105 Kiel, Germany; (S.F.); (C.K.)
| | - Stefano Flor
- Research Group Medical Systems Biology, Christian-Albrechts-Universität Kiel, Brunswiker Straße 10, 24105 Kiel, Germany; (S.F.); (C.K.)
| | - Andrés Pinzón
- Research Group Bioinformatics and Systems Biology, Instituto de Genetica, Universidad Nacional de Colombia, Bogotá 111321, Colombia; (L.J.F.-R.); (A.P.)
| | - Christoph Kaleta
- Research Group Medical Systems Biology, Christian-Albrechts-Universität Kiel, Brunswiker Straße 10, 24105 Kiel, Germany; (S.F.); (C.K.)
| |
Collapse
|