1
|
Elhassan YH, Alahmadi F, Albadawi EA, Albarakati A, Aljohany AH, Alzaman NS, Albadrani M. The Relationship Between Maternal Exposure to Endocrine-Disrupting Chemicals and the Incidence of Congenital Heart Diseases: A Systematic Review and Meta-Analysis. Metabolites 2024; 14:709. [PMID: 39728490 DOI: 10.3390/metabo14120709] [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: 11/11/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Congenital heart diseases are among the most common birth defects, significantly impacting infant health. Recent evidence suggests that exposure to endocrine-disrupting chemicals may contribute to the incidence of congenital heart diseases. This study systematically reviews and analyzes the association between maternal endocrine-disrupting chemicals exposure and congenital heart diseases. METHODOLOGY This systematic review and meta-analysis followed the Cochrane Handbook and PRISMA guidelines. We included studies assessing the link between maternal exposure to various endocrine-disrupting chemicals and the incidence of congenital heart diseases without restricting the study design or exposure assessment methods. Data were extracted from four databases, including PubMed, Scopus, Web of Science, and Cochrane Library, up to June 2024. Quality assessment of observational studies was conducted using the Newcastle-Ottawa Scale. Statistical analysis was performed using RevMan software version 5.3, presenting results as odds ratios with 95% confidence intervals. RESULTS Fifty-nine studies were included in the meta-analysis. The pooled analysis revealed a significant association between maternal endocrine-disrupting chemical exposure and the incidence of congenital heart diseases when measured using human samples (odds ratio = 1.63, 95% confidence interval [1.35-1.97], p < 0.00001). Notably, exposure to heavy metals, polycyclic aromatic hydrocarbons, and perfluoroalkyl compounds was strongly associated with congenital heart diseases. However, non-sample-based methods showed no significant overall correlation (odds ratio = 1.08, 95% confidence interval [0.93-1.26], p = 0.30), except for housing renovation compounds, which were linked to a higher incidence of congenital heart diseases. CONCLUSIONS Maternal exposure to specific endocrine-disrupting chemicals, particularly heavy metals and polycyclic aromatic hydrocarbons, significantly increases the risk of congenital heart diseases. These findings underscore the need for preventive measures to reduce endocrine-disrupting chemicals exposure during pregnancy and further research to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Yasir Hassan Elhassan
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia
| | - Fahad Alahmadi
- Department of Women and Child Health, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia
| | - Emad Ali Albadawi
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia
| | - Abdullah Albarakati
- Department of Women and Child Health, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia
| | - Azizah Hendi Aljohany
- Department of Women and Child Health, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia
| | | | - Muayad Albadrani
- Department of Family and Community Medicine and Medical Education, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia
| |
Collapse
|
2
|
Dai J, Wang G, Wu C, Pan Z, Li H, Shen L, Wu Y. Exposure to Endocrine-Disrupting Chemicals and Congenital Heart Diseases: The Pooled Results Based on the Current Evidence. Pediatr Cardiol 2024:10.1007/s00246-024-03478-w. [PMID: 38602518 DOI: 10.1007/s00246-024-03478-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 03/19/2024] [Indexed: 04/12/2024]
Abstract
The relationships between maternal exposure to endocrine-disrupting chemicals (EDCs) and congenital heart diseases (CHD) are not elucidated yet. The exposure levels of EDCs are generally estimated based on self-reported questionnaires or occupational exposure evaluations in the literature. Therefore, a study based on epidemiological data from human biospecimens is required to provide stronger evidence between maternal exposure to EDC and CHD. Embase, Pubmed, Scopus, and the Cochrane Library databases were searched for related research which provided risk estimates regarding the relationships between maternal EDC exposure and CHD in human offspring. Baseline characteristics and outcomes of CHD were extracted from each included study. Odds ratios (ORs) with 95% confidence intervals (CIs) were pooled to calculate the overall estimates of CHD. Subgroup and meta-regression analyses were performed to identify the sources of heterogeneity. Bootstrapping techniques were used in analyses where several studies originated from a similar population. A total of seventeen studies were involved in the meta-analyses. Maternal EDC exposure was significantly related to CHD in offspring (OR 2.15; 95%CI 1.64 to 2.83). EDC exposure was significantly associated with septal defects (OR 2.34; 95%CI 1.77 to 3.10), conotruncal defects (OR 2.54; 95%CI 1.89 to 3.43), right ventricular outflow tract obstruction (OR 2.65; 95%CI 1.73 to 4.07), left ventricular outflow tract obstruction (OR 3.58; 95%CI 2.67 to 4.79), anomalous pulmonary venous return (OR 2.31; 95%CI 1.34 to 4.00), and other heart defects (OR 2.49; 95%CI 1.75 to 3.54). In addition, maternal exposure to heavy metals, which included lead (OR 2.19; 95%CI 1.29 to 3.71), cadmium (OR 1.81; 95%CI 1.28 to 2.56), mercury (OR 2.23; 95%CI 1.13 to 4.44), and manganese (OR 2.65; 95%CI 1.48 to 4.74), increased risks for CHD significantly. In conclusion, based on the latest evidence, maternal EDC exposure may increase CHD risks in human offspring, especially in heavy metal exposure conditions.
Collapse
Affiliation(s)
- Jiangtao Dai
- Department of Cardiothoracic Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical DisordersChongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Gang Wang
- Department of Cardiothoracic Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical DisordersChongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Chun Wu
- Department of Cardiothoracic Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical DisordersChongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zhengxia Pan
- Department of Cardiothoracic Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical DisordersChongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Hongbo Li
- Department of Cardiothoracic Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical DisordersChongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lianju Shen
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical DisordersChongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yuhao Wu
- Department of Cardiothoracic Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical DisordersChongqing Key Laboratory of Pediatrics, Chongqing, China.
| |
Collapse
|
3
|
Fan RZ, Sportelli C, Lai Y, Salehe SS, Pinnell JR, Brown HJ, Richardson JR, Luo S, Tieu K. A partial Drp1 knockout improves autophagy flux independent of mitochondrial function. Mol Neurodegener 2024; 19:26. [PMID: 38504290 PMCID: PMC10953112 DOI: 10.1186/s13024-024-00708-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/07/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Dynamin-related protein 1 (Drp1) plays a critical role in mitochondrial dynamics. Partial inhibition of this protein is protective in experimental models of neurological disorders such as Parkinson's disease and Alzheimer's disease. The protective mechanism has been attributed primarily to improved mitochondrial function. However, the observations that Drp1 inhibition reduces protein aggregation in such neurological disorders suggest the involvement of autophagy. To investigate this potential novel protective mechanism of Drp1 inhibition, a model with impaired autophagy without mitochondrial involvement is needed. METHODS We characterized the effects of manganese (Mn), which causes parkinsonian-like symptoms in humans, on autophagy and mitochondria by performing dose-response studies in two cell culture models (stable autophagy HeLa reporter cells and N27 rat immortalized dopamine neuronal cells). Mitochondrial function was assessed using the Seahorse Flux Analyzer. Autophagy flux was monitored by quantifying the number of autophagosomes and autolysosomes, as well as the levels of other autophagy proteins. To strengthen the in vitro data, multiple mouse models (autophagy reporter mice and mutant Drp1+/- mice and their wild-type littermates) were orally treated with a low chronic Mn regimen that was previously reported to increase α-synuclein aggregation and transmission via exosomes. RNAseq, laser captured microdissection, immunofluorescence, immunoblotting, stereological cell counting, and behavioural studies were used. RESULTS IN VITRO: data demonstrate that at low non-toxic concentrations, Mn impaired autophagy flux but not mitochondrial function and morphology. In the mouse midbrain, RNAseq data further confirmed autophagy pathways were dysregulated but not mitochondrial related genes. Additionally, Mn selectively impaired autophagy in the nigral dopamine neurons but not the nearby nigral GABA neurons. In cells with a partial Drp1-knockdown and Drp1+/- mice, Mn induced autophagic impairment was significantly prevented. Consistent with these observations, Mn increased the levels of proteinase-K resistant α-synuclein and Drp1-knockdown protected against this pathology. CONCLUSIONS This study demonstrates that improved autophagy flux is a separate mechanism conferred by Drp1 inhibition independent of its role in mitochondrial fission. Given that impaired autophagy and mitochondrial dysfunction are two prominent features of neurodegenerative diseases, the combined protective mechanisms targeting these two pathways conferred by Drp1 inhibition make this protein an attractive therapeutic target.
Collapse
Affiliation(s)
- Rebecca Z Fan
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Carolina Sportelli
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Yanhao Lai
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Said S Salehe
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Jennifer R Pinnell
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Harry J Brown
- Department of Environmental Health Sciences, Florida International University, Miami, USA
- Biomolecular Sciences Institute, Florida International University, Miami, USA
| | - Jason R Richardson
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Shouqing Luo
- Peninsula Schools of Medicine and Dentistry, Plymouth University, Plymouth, UK
| | - Kim Tieu
- Department of Environmental Health Sciences, Florida International University, Miami, USA.
- Biomolecular Sciences Institute, Florida International University, Miami, USA.
| |
Collapse
|
4
|
Liu Y, Zeng JM, Zhao H, Ao CY, Ao LH, Ban JQ, Li J. Mechanism of KAT2A regulation of H3K36ac in manganese-induced oxidative damage to mitochondria in the nervous system and intervention by curcumin. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 273:116155. [PMID: 38417317 DOI: 10.1016/j.ecoenv.2024.116155] [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: 01/06/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/01/2024]
Abstract
Excessive exposure to manganese in the environment or workplace is strongly linked to neurodegeneration and cognitive impairment, but the precise pathogenic mechanism and preventive measures are still not fully understood. The study aimed to investigate manganese -induced oxidative damage in the nervous system from an epigenetic perspective, focusing on the H3K36ac-dependent antioxidant pathway. Additionally, it sought to examine the potential of curcumin in preventing manganese-induced oxidative damage. Histopathology and transmission electron microscopy revealed that apoptosis and necrosis of neurons and mitochondrial ultrastructure damage were observed in the striatum of manganese-exposed rats. manganese suppressed the expression of mitochondrial antioxidant genes, leading to oxidative damage in the rats' striatum and SH-SY5Y cells. With higher doses of manganese, levels of histone acetyltransferase lysine acetyltransferase 2 A (KAT2A) expression and H3K36ac level decreased. ChIP-qPCR confirmed that H3K36ac enrichment in the promoter regions of antioxidant genes SOD2, PRDX3, and TXN2 was reduced in SH-SY5Y cells after manganese exposure, leading to decreased expression of these genes. Overexpression of KAT2A confirms that it attenuates manganese-induced mitochondrial oxidative damage by regulating H3K36ac levels, which in turn controls the expression of antioxidant genes SOD2, PRDX3, and TXN2 in the manganese-exposed cell model. Furthermore, curcumin might control H3K36ac levels by influencing KAT2A expression, boosting antioxidant genes expression, and reducing manganese-induced mitochondrial oxidative damage. In conclusion, the regulation of mitochondrial oxidative stress by histone acetylation may be an important mechanism of manganese-induced neurotoxicity. This regulation could be achieved by reducing the level of H3K36ac near the promoter region of mitochondrial-associated antioxidant genes via KAT2A. Curcumin mitigates manganese-induced oxidative damage in mitochondria and plays a crucial protective role in manganese-induced oxidative injury in the nervous system.
Collapse
Affiliation(s)
- Yan Liu
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang ,Guizhou 561113, China
| | - Jia-Min Zeng
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang ,Guizhou 561113, China
| | - Hua Zhao
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang ,Guizhou 561113, China
| | - Chun-Yan Ao
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang ,Guizhou 561113, China
| | - Li-Hong Ao
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang ,Guizhou 561113, China
| | - Jia-Qi Ban
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang ,Guizhou 561113, China..
| | - Jun Li
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang ,Guizhou 561113, China..
| |
Collapse
|
5
|
Gallo G, Rubattu S, Volpe M. Mitochondrial Dysfunction in Heart Failure: From Pathophysiological Mechanisms to Therapeutic Opportunities. Int J Mol Sci 2024; 25:2667. [PMID: 38473911 DOI: 10.3390/ijms25052667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/17/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
Mitochondrial dysfunction, a feature of heart failure, leads to a progressive decline in bioenergetic reserve capacity, consisting in a shift of energy production from mitochondrial fatty acid oxidation to glycolytic pathways. This adaptive process of cardiomyocytes does not represent an effective strategy to increase the energy supply and to restore the energy homeostasis in heart failure, thus contributing to a vicious circle and to disease progression. The increased oxidative stress causes cardiomyocyte apoptosis, dysregulation of calcium homeostasis, damage of proteins and lipids, leakage of mitochondrial DNA, and inflammatory responses, finally stimulating different signaling pathways which lead to cardiac remodeling and failure. Furthermore, the parallel neurohormonal dysregulation with angiotensin II, endothelin-1, and sympatho-adrenergic overactivation, which occurs in heart failure, stimulates ventricular cardiomyocyte hypertrophy and aggravates the cellular damage. In this review, we will discuss the pathophysiological mechanisms related to mitochondrial dysfunction, which are mainly dependent on increased oxidative stress and perturbation of the dynamics of membrane potential and are associated with heart failure development and progression. We will also provide an overview of the potential implication of mitochondria as an attractive therapeutic target in the management and recovery process in heart failure.
Collapse
Affiliation(s)
- Giovanna Gallo
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035-1039, 00189 Rome, RM, Italy
| | - Speranza Rubattu
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035-1039, 00189 Rome, RM, Italy
- IRCCS Neuromed, 86077 Pozzilli, IS, Italy
| | | |
Collapse
|
6
|
Anbu S, Kenning L, Stasiuk GJ. ATP-responsive Mn(II)-based T1 contrast agent for MRI. Chem Commun (Camb) 2023; 59:13623-13626. [PMID: 37902503 PMCID: PMC10644988 DOI: 10.1039/d3cc03430e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023]
Abstract
A novel diacetylpyridylcarbohydrazide-DAPyCOHz-based manganese(II) chelate with dipicolylamine/zinc(II) (DPA/Zn2+) arms (MnLDPA-Zn2) was developed for adenosine triphosphate (ATP) responsive magnetic resonance imaging (MRI) T1 contrast applications. Compound 2 shows enhanced relaxivity (r1 = 11.52 mM-1 s-1) upon selective ATP binding over other phosphates.
Collapse
Affiliation(s)
- Sellamuthu Anbu
- Departments of Chemistry and Biomedical Sciences, University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
- School of Chemistry, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Lawerence Kenning
- MRI Centre, Royal Infirmary Hospital NHS Trust, Anlaby Road, Hull, HU3 2JZ, UK
| | - Graeme J Stasiuk
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, Fourth Floor Lambeth Wing, St Thomas' Hospital, London, SE1 7EH, UK.
| |
Collapse
|
7
|
Baj J, Flieger W, Barbachowska A, Kowalska B, Flieger M, Forma A, Teresiński G, Portincasa P, Buszewicz G, Radzikowska-Büchner E, Flieger J. Consequences of Disturbing Manganese Homeostasis. Int J Mol Sci 2023; 24:14959. [PMID: 37834407 PMCID: PMC10573482 DOI: 10.3390/ijms241914959] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
Manganese (Mn) is an essential trace element with unique functions in the body; it acts as a cofactor for many enzymes involved in energy metabolism, the endogenous antioxidant enzyme systems, neurotransmitter production, and the regulation of reproductive hormones. However, overexposure to Mn is toxic, particularly to the central nervous system (CNS) due to it causing the progressive destruction of nerve cells. Exposure to manganese is widespread and occurs by inhalation, ingestion, or dermal contact. Associations have been observed between Mn accumulation and neurodegenerative diseases such as manganism, Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. People with genetic diseases associated with a mutation in the gene associated with impaired Mn excretion, kidney disease, iron deficiency, or a vegetarian diet are at particular risk of excessive exposure to Mn. This review has collected data on the current knowledge of the source of Mn exposure, the experimental data supporting the dispersive accumulation of Mn in the brain, the controversies surrounding the reference values of biomarkers related to Mn status in different matrices, and the competitiveness of Mn with other metals, such as iron (Fe), magnesium (Mg), zinc (Zn), copper (Cu), lead (Pb), calcium (Ca). The disturbed homeostasis of Mn in the body has been connected with susceptibility to neurodegenerative diseases, fertility, and infectious diseases. The current evidence on the involvement of Mn in metabolic diseases, such as type 2 diabetes mellitus/insulin resistance, osteoporosis, obesity, atherosclerosis, and non-alcoholic fatty liver disease, was collected and discussed.
Collapse
Affiliation(s)
- Jacek Baj
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Wojciech Flieger
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Aleksandra Barbachowska
- Department of Plastic, Reconstructive and Burn Surgery, Medical University of Lublin, 21-010 Łęczna, Poland;
| | - Beata Kowalska
- Department of Water Supply and Wastewater Disposal, Lublin University of Technology, 20-618 Lublin, Poland;
| | - Michał Flieger
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | - Alicja Forma
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Grzegorz Teresiński
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | - Piero Portincasa
- Clinica Medica A. Murri, Department of Biomedical Sciences & Human Oncology, Medical School, University of Bari, 70124 Bari, Italy;
| | - Grzegorz Buszewicz
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | | | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
8
|
Dorman DC. The Role of Oxidative Stress in Manganese Neurotoxicity: A Literature Review Focused on Contributions Made by Professor Michael Aschner. Biomolecules 2023; 13:1176. [PMID: 37627240 PMCID: PMC10452838 DOI: 10.3390/biom13081176] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
This literature review focuses on the evidence implicating oxidative stress in the pathogenesis of manganese neurotoxicity. This review is not intended to be a systematic review of the relevant toxicologic literature. Instead, in keeping with the spirit of this special journal issue, this review highlights contributions made by Professor Michael Aschner's laboratory in this field of study. Over the past two decades, his laboratory has made significant contributions to our scientific understanding of cellular responses that occur both in vitro and in vivo following manganese exposure. These studies have identified molecular targets of manganese toxicity and their respective roles in mitochondrial dysfunction, inflammation, and cytotoxicity. Other studies have focused on the critical role astrocytes play in manganese neurotoxicity. Recent studies from his laboratory have used C. elegans to discover new facets of manganese-induced neurotoxicity. Collectively, his body of work has dramatically advanced the field and presents broader implications beyond metal toxicology.
Collapse
Affiliation(s)
- David C Dorman
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, 1052 William Moore Dr, Raleigh, NC 27606, USA
| |
Collapse
|
9
|
Fan RZ, Sportelli C, Lai Y, Salehe S, Pinnell JR, Richardson JR, Luo S, Tieu K. A partial Drp1 knockout improves autophagy flux independent of mitochondrial function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547095. [PMID: 37425803 PMCID: PMC10327068 DOI: 10.1101/2023.06.29.547095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Dynamin-related protein 1 (Drp1) is typically known for its role in mitochondrial fission. A partial inhibition of this protein has been reported to be protective in experimental models of neurodegenerative diseases. The protective mechanism has been attributed primarily to improved mitochondrial function. Herein, we provide evidence showing that a partial Drp1-knockout improves autophagy flux independent of mitochondria. First, we characterized in cell and animal models that at low non-toxic concentrations, manganese (Mn), which causes parkinsonian-like symptoms in humans, impaired autophagy flux but not mitochondrial function and morphology. Furthermore, nigral dopaminergic neurons were more sensitive than their neighbouring GABAergic counterparts. Second, in cells with a partial Drp1-knockdown and Drp1 +/- mice, autophagy impairment induced by Mn was significantly attenuated. This study demonstrates that autophagy is a more vulnerable target than mitochondria to Mn toxicity. Furthermore, improving autophagy flux is a separate mechanism conferred by Drp1 inhibition independent of mitochondrial fission.
Collapse
|
10
|
Huang Y, Ruan Y, Ma Y, Chen D, Zhang T, Fan S, Lin W, Huang Y, Lu H, Xu JF, Pi J, Zheng B. Immunomodulatory activity of manganese dioxide nanoparticles: Promising for novel vaccines and immunotherapeutics. Front Immunol 2023; 14:1128840. [PMID: 36926351 PMCID: PMC10011163 DOI: 10.3389/fimmu.2023.1128840] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/09/2023] [Indexed: 03/08/2023] Open
Abstract
Manganese (Mn), a nutrient inorganic trace element, is necessary for a variety of physiological processes of animal body due to their important roles in oxidative regulation effects and other aspects of activities. Moreover, manganese ion (Mn2+) has widely reported to be crucial for the regulations of different immunological responses, thus showing promising application as potential adjuvants and immunotherapeutics. Taking the advantages of Mn-based biological and immunological activities, Manganese dioxide nanoparticles (MnO2 NPs) are a new type of inorganic nanomaterials with numerous advantages, including simple preparation, low cost, environmental friendliness, low toxicity, biodegradable metabolism and high bioavailability. MnO2 NPs, as a kind of drug carrier, have also shown the ability to catalyze hydrogen peroxide (H2O2) to produce oxygen (O2) under acidic conditions, which can enhance the efficacy of radiotherapy, chemotherapy and other therapeutics for tumor treatment by remodeling the tumor microenvironment. More importantly, MnO2 NPs also play important roles in immune regulations both in innate and adaptive immunity. In this review, we summarize the biological activities of Manganese, followed by the introduction for the biological and medical functions and mechanisms of MnO2 NPs. What's more, we emphatically discussed the immunological regulation effects and mechanisms of MnO2 NPs, as well as their potentials to serve as adjuvants and immunomodulators, which might benefit the development of novel vaccines and immunotherapies for more effective disease control.
Collapse
Affiliation(s)
- Yuhe Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yongdui Ruan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Yuhe Ma
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Dongsheng Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Tangxin Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.,Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Shuhao Fan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Wensen Lin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yifan Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Hongmei Lu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Biying Zheng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| |
Collapse
|
11
|
Chen P, Cheng H, Zheng F, Li S, Bornhorst J, Yang B, Lee KH, Ke T, Li Y, Schwerdtle T, Yang X, Bowman AB, Aschner M. BTBD9 attenuates manganese-induced oxidative stress and neurotoxicity by regulating insulin growth factor signaling pathway. Hum Mol Genet 2022; 31:2207-2222. [PMID: 35134179 PMCID: PMC9262395 DOI: 10.1093/hmg/ddac025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/29/2021] [Accepted: 01/21/2022] [Indexed: 02/05/2023] Open
Abstract
Manganese (Mn) is an essential mineral, but excess exposure can cause dopaminergic neurotoxicity. Restless legs syndrome (RLS) is a common neurological disorder, but the etiology and pathology remain largely unknown. The purpose of this study was to identify the role of Mn in the regulation of an RLS genetic risk factor BTBD9, characterize the function of BTBD9 in Mn-induced oxidative stress and dopaminergic neuronal dysfunction. We found that human subjects with high blood Mn levels were associated with decreased BTBD9 mRNA levels, when compared with subjects with low blood Mn levels. In A549 cells, Mn exposure decreased BTBD9 protein levels. In Caenorhabditis elegans, loss of hpo-9 (BTBD9 homolog) resulted in more susceptibility to Mn-induced oxidative stress and mitochondrial dysfunction, as well as decreased dopamine levels and alternations of dopaminergic neuronal morphology and behavior. Overexpression of hpo-9 in mutant animals restored these defects and the protection was eliminated by mutation of the forkhead box O (FOXO). In addition, expression of hpo-9 upregulated FOXO protein levels and decreased protein kinase B levels. These results suggest that elevated Mn exposure might be an environmental risk factor for RLS. Furthermore, BTBD9 functions to alleviate Mn-induced oxidative stress and neurotoxicity via regulation of insulin/insulin-like growth factor signaling pathway.
Collapse
Affiliation(s)
- Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Hong Cheng
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 53021, China
| | - Fuli Zheng
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Shaojun Li
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 53021, China
| | - Julia Bornhorst
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal 42119, Germany
| | - Bobo Yang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kun He Lee
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yunhui Li
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Key Laboratory of Environmental Medicine Engineering Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210000, China
| | - Tanja Schwerdtle
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Nuthetal 14558, Germany
- TraceAge—DFG Research Group on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena 14558, Germany
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 53021, China
- Department of Public Health, School of Medicine, Guangxi University of Science and Technology, Liuzhou 545026, China
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
12
|
Olude MA, Mouihate A, Mustapha OA, Farina C, Quintana FJ, Olopade JO. Astrocytes and Microglia in Stress-Induced Neuroinflammation: The African Perspective. Front Immunol 2022; 13:795089. [PMID: 35707531 PMCID: PMC9190229 DOI: 10.3389/fimmu.2022.795089] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Africa is laden with a youthful population, vast mineral resources and rich fauna. However, decades of unfortunate historical, sociocultural and leadership challenges make the continent a hotspot for poverty, indoor and outdoor pollutants with attendant stress factors such as violence, malnutrition, infectious outbreaks and psychological perturbations. The burden of these stressors initiate neuroinflammatory responses but the pattern and mechanisms of glial activation in these scenarios are yet to be properly elucidated. Africa is therefore most vulnerable to neurological stressors when placed against a backdrop of demographics that favor explosive childbearing, a vast population of unemployed youths making up a projected 42% of global youth population by 2030, repressive sociocultural policies towards women, poor access to healthcare, malnutrition, rapid urbanization, climate change and pollution. Early life stress, whether physical or psychological, induces neuroinflammatory response in developing nervous system and consequently leads to the emergence of mental health problems during adulthood. Brain inflammatory response is driven largely by inflammatory mediators released by glial cells; namely astrocytes and microglia. These inflammatory mediators alter the developmental trajectory of fetal and neonatal brain and results in long-lasting maladaptive behaviors and cognitive deficits. This review seeks to highlight the patterns and mechanisms of stressors such as poverty, developmental stress, environmental pollutions as well as malnutrition stress on astrocytes and microglia in neuroinflammation within the African context.
Collapse
Affiliation(s)
- Matthew Ayokunle Olude
- Vertebrate Morphology, Environmental Toxicology and Neuroscience Unit, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
- *Correspondence: Matthew Ayokunle Olude,
| | - Abdeslam Mouihate
- Department of Physiology, Faculty of Medicine, Health Sciences Centre, Kuwait University, Kuwait City, Kuwait
| | - Oluwaseun Ahmed Mustapha
- Vertebrate Morphology, Environmental Toxicology and Neuroscience Unit, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
| | - Cinthia Farina
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) San Raffaele Scientific Institute, Institute of Experimental Neurology (INSPE) and Division of Neuroscience, Milan, Italy
| | - Francisco Javier Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - James Olukayode Olopade
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
13
|
Multi-Elemental Analysis of Human Optic Chiasm-A New Perspective to Reveal the Pathomechanism of Nerve Fibers' Degeneration. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19074420. [PMID: 35410100 PMCID: PMC8998695 DOI: 10.3390/ijerph19074420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/04/2023]
Abstract
The effect of metals on the functioning of the human eye is multifactorial and includes enzyme activity modulation, trace metal metabolic pathways changes, and cytotoxic activity. Functional dysfunctions appear mostly as a result of the accumulation of toxic xenobiotic metals or disturbances of micronutrients’ homeostasis. So far, the affinity of selected metals to eye tissues, i.e., the cornea, choroid, lens, and anterior chamber fluid, has been most studied. However, it is known that many eye symptoms are related to damage to the optic nerve. In order to fill this gap, the aim of the study is to perform a multi-element analysis of tissue collected postmortem from optic chiasm and optic nerves. A total of 178 samples from 107 subjects were tested. The concentrations of 51 elements were quantified by inductively coupled plasma mass spectrometry (ICP-MS) after the wet-mineralization step. In terms of elemental composition, the optic chiasm is dominated by two trace elements, i.e., iron (Fe) and zinc (Zn), besides macro-elements Ca, K, Na, P, and Mg. The subjects formed a homogeneous cluster (over 70% subjects) with the highest accumulation of aluminum (Al). The remaining two departing clusters were characterized by an increased content of most of the elements, including toxic elements such as bismuth (Bi), uranium (U), lead (Pb), chromium (Cr), and cadmium (Cd). Changes in elemental composition with age were analyzed statistically for the selected groups, i.e., females, males, and subjects with alcohol use disorder (AUD) and without AUD. A tendency of women to lose Se, Cu, Zn, Fe with age was observed, and a disturbed Ca/Mg, Na/K ratio in subjects with AUD. Although the observed trends were not statistically significant, they shed new light on the risks and possible pathologies associated with metal neurotoxicity in the visual tract.
Collapse
|
14
|
Wang M, Tian Y, Yu P, Li N, Deng Y, Li L, Kang H, Chen D, Wang H, Liu Z, Liang J. Association between congenital heart defects and maternal manganese and iron concentrations: a case-control study in China. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:26950-26959. [PMID: 34865185 PMCID: PMC8989826 DOI: 10.1007/s11356-021-17054-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/11/2021] [Indexed: 05/10/2023]
Abstract
To investigate the correlation between maternal manganese and iron concentrations and the risk of CHD among their infant. A multi-center hospital-based case control study was conducted in China. There were 322 cases and 333 controls have been selected from pregnant women who received prenatal examinations. Correlations between CHDs and maternal manganese and iron concentrations were estimated by conditional logistic regression. Moreover, the interaction between manganese and iron on CHDs was analyzed. Compared with the controls, mothers whose hair manganese concentration was 3.01 μg/g or more were more likely to have a child with CHD than those with a lower concentration. The adjusted OR was 2.68 (95%CI = 1.44-4.99). The results suggested that mothers whose iron content was 52.95 μg/g or more had a significantly higher risk of having a child with CHD (aOR = 2.87, 95%CI = 1.54-5.37). No interaction between maternal manganese and iron concentrations was observed in the multiplicative or additive model. The concurrently existing high concentration of manganese and iron may bring higher risk of CHD (OR = 7.02). Women with excessive manganese concentrations have a significantly increased risk of having offspring with CHDs. The high maternal iron status also correlates with CHDs. The concurrently existing high concentration of manganese and iron may bring higher risk of CHD.
Collapse
Affiliation(s)
- Meixian Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yan Tian
- Liupanshui Maternal and Child Health Care Hospital, Liupanshui Children's Hospital, Liupanshui, Guizhou, China
| | - Ping Yu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Nana Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ying Deng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lu Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hong Kang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dapeng Chen
- Chenghua District Maternal and Child Health Hospital of Chengdu, Chengdu, Sichuan, China
| | - Hui Wang
- Mianyang Maternal and Child Health Care Hospital, Mianyang, Sichuan, China
| | - Zhen Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.
- National Office for Maternal and Child Health Surveillance of China, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Juan Liang
- National Office for Maternal and Child Health Surveillance of China, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Monteith AJ, Miller JM, Beavers WN, Juttukonda LJ, Skaar EP. Increased Dietary Manganese Impairs Neutrophil Extracellular Trap Formation Rendering Neutrophils Ineffective at Combating Staphylococcus aureus. Infect Immun 2022; 90:e0068521. [PMID: 35191757 PMCID: PMC8929375 DOI: 10.1128/iai.00685-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 11/20/2022] Open
Abstract
Dietary metals can modify the risk to infection. Previously, we demonstrated that heightened dietary manganese (Mn) during systemic Staphylococcus aureus infection increases S. aureus virulence. However, immune cells also operate in these same environments and the effect of dietary Mn on neutrophil function in vivo has not been assessed. This study reveals that increased concentrations of Mn impairs mitochondrial respiration and superoxide production in neutrophils responding to S. aureus. As a result, high Mn accelerates primary degranulation, while impairing suicidal neutrophil extracellular trap (NET) formation, which decreases bactericidal activity. In vivo, elevated dietary Mn accumulated extracellularly in the heart, indicating that excess Mn may be more bioavailable in the heart. Coinciding with this phenotype, neutrophil function in the heart was most impacted by a high Mn diet, as neutrophils produced lower levels of mitochondrial superoxide and underwent less suicidal NET formation. Consistent with an ineffective neutrophil response when mice are on a high Mn diet, S. aureus burdens were increased in the heart and mice were more susceptible to systemic infection. Therefore, elevated dietary Mn not only affects S. aureus but also renders neutrophils less capable of restricting staphylococcal infection.
Collapse
Affiliation(s)
- Andrew J. Monteith
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeanette M. Miller
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - William N. Beavers
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Lillian J. Juttukonda
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, & Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
16
|
Zheng R, Guo J, Cai X, Bin L, Lu C, Singh A, Trivedi M, Kumar A, Liu J. Manganese complexes and manganese-based metal-organic frameworks as contrast agents in MRI and chemotherapeutics agents: Applications and prospects. Colloids Surf B Biointerfaces 2022; 213:112432. [PMID: 35259704 DOI: 10.1016/j.colsurfb.2022.112432] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/17/2022] [Accepted: 02/27/2022] [Indexed: 12/20/2022]
Abstract
Manganese-based Metal-organic Frameworks (Mn-MOFs) represents a unique sub-class of MOFs with low toxicity, oxidative ability, and biocompatibility, which plays vital role in the application of this class of MOFs in medical field. Mn-MOFs show great potential in biomedical applications, and has been extensively studied as compared to other MOFs in transition metal series. They are important in medical applications because Mn(II) possess large electron spin number and longer electron relaxation time. They display fast water exchange rate and could be employed as a potential MRI contrast agent because of their strong targeting ability. Manganese complexes with different ligands also display prospective applications in area such as carrier for drug targeting in anti-tumor and antimicrobial therapy. In the review presented herewith, the application of Mn-based complexes and Mn-MOFs have been emphasized in the area such as imaging viz. MRI, multimodal imaging, antitumor activities such as chemodynamic therapy, photodynamic therapy, sonodynamic therapy and antimicrobial applications. Also, how rational designing and syntheses of targeted Mn-based complexes and Mn-MOFs can engender desired applications.
Collapse
Affiliation(s)
- Rouqiao Zheng
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Junru Guo
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Xinyi Cai
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Lianjie Bin
- Department of General Surgery, Dongguan People's Hospital, Wanjiang District, Dongguan 523000, China.
| | - Chengyu Lu
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Amita Singh
- Department of Chemistry, Dr. Ram Manohar Lohiya Awadh University, Ayodhya, India
| | - Manoj Trivedi
- Department of Chemistry, Sri Venkateswara College, University of Delhi, New Delhi 110021, India
| | - Abhinav Kumar
- Department of Chemistry, Faculty of Science, University of Lucknow, Lucknow 226007, India.
| | - Jianqiang Liu
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
17
|
Liu HS, Zhou MY, Zhang X, Li YL, Kong JW, Gao X, Ge DY, Liu JJ, Ma PG, Peng GY, Liao Y. Sagittaria sagittifolia polysaccharide protects against six-heavy-metal-induced hepatic injury associated with the activation of Nrf2 pathway to regulate oxidative stress and apoptosis. J Inorg Biochem 2022; 232:111810. [DOI: 10.1016/j.jinorgbio.2022.111810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 12/12/2022]
|
18
|
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
|
19
|
Garg V, Suzuki J, Paranjpe I, Unsulangi T, Boyman L, Milescu LS, Lederer WJ, Kirichok Y. The mechanism of MICU-dependent gating of the mitochondrial Ca 2+uniporter. eLife 2021; 10:e69312. [PMID: 34463251 PMCID: PMC8437439 DOI: 10.7554/elife.69312] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 08/09/2021] [Indexed: 12/11/2022] Open
Abstract
Ca2+ entry into mitochondria is through the mitochondrial calcium uniporter complex (MCUcx), a Ca2+-selective channel composed of five subunit types. Two MCUcx subunits (MCU and EMRE) span the inner mitochondrial membrane, while three Ca2+-regulatory subunits (MICU1, MICU2, and MICU3) reside in the intermembrane space. Here, we provide rigorous analysis of Ca2+ and Na+ fluxes via MCUcx in intact isolated mitochondria to understand the function of MICU subunits. We also perform direct patch clamp recordings of macroscopic and single MCUcx currents to gain further mechanistic insights. This comprehensive analysis shows that the MCUcx pore, composed of the EMRE and MCU subunits, is not occluded nor plugged by MICUs during the absence or presence of extramitochondrial Ca2+ as has been widely reported. Instead, MICUs potentiate activity of MCUcx as extramitochondrial Ca2+ is elevated. MICUs achieve this by modifying the gating properties of MCUcx allowing it to spend more time in the open state.
Collapse
Affiliation(s)
- Vivek Garg
- Department of Physiology, University of California San FranciscoSan FranciscoUnited States
- Department of Physiology, University of MarylandBaltimoreUnited States
| | - Junji Suzuki
- Department of Physiology, University of California San FranciscoSan FranciscoUnited States
| | - Ishan Paranjpe
- Department of Physiology, University of California San FranciscoSan FranciscoUnited States
| | - Tiffany Unsulangi
- Department of Physiology, University of California San FranciscoSan FranciscoUnited States
| | - Liron Boyman
- Department of Physiology, University of MarylandBaltimoreUnited States
| | - Lorin S Milescu
- Department of Biology, University of MarylandCollege ParkUnited States
| | | | - Yuriy Kirichok
- Department of Physiology, University of California San FranciscoSan FranciscoUnited States
| |
Collapse
|
20
|
Oxidative Stress as a Common Key Event in Developmental Neurotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6685204. [PMID: 34336113 PMCID: PMC8315852 DOI: 10.1155/2021/6685204] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/29/2021] [Accepted: 07/06/2021] [Indexed: 12/20/2022]
Abstract
The developing brain is extremely sensitive to many chemicals. Perinatal exposure to neurotoxicants has been implicated in several neurodevelopmental disorders, including autism spectrum disorder, attention-deficit hyperactive disorder, and schizophrenia. Studies of the molecular and cellular events related to developmental neurotoxicity have identified a number of “adverse outcome pathways,” many of which share oxidative stress as a key event. Oxidative stress occurs when the balance between the production of free oxygen radicals and the activity of the cellular antioxidant system is dysregulated. In this review, we describe some of the developmental neurotoxins that target the antioxidant system and the mechanisms by which they elicit stress, including oxidative phosphorylation in mitochondria and plasma membrane redox system in rodent models. We also discuss future directions for identifying adverse outcome pathways related to oxidative stress and developmental neurotoxicity, with the goal of improving our ability to quickly and accurately screen chemicals for their potential developmental neurotoxicity.
Collapse
|
21
|
Mostafa HES, Alaa El-Din EA, El-Shafei DA, Abouhashem NS, Abouhashem AA. Protective roles of thymoquinone and vildagliptin in manganese-induced nephrotoxicity in adult albino rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:31174-31184. [PMID: 33595798 DOI: 10.1007/s11356-021-12997-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 02/11/2021] [Indexed: 06/12/2023]
Abstract
Despite being important in the body's mechanisms, excessive accumulation of manganese (Mn) can induce severe toxicity in vital organs of the body. Thymoquinone (TQ) is extracted from Nigella sativa seeds which recently gained popularity as dietary supplements and plant-based antioxidants. Vildagliptin (VLD) is a dipeptidyl peptidase IV (DPPIV) inhibitor, approved as anti-hyperglycemic agents with cardioprotective and renoprotective effects. The present study aimed to investigate the nephrotoxicity of Mn and the potential protective effects of thymoquinone and vildagliptin. Sixty-four adult male albino rats were equally divided into 8 groups: group I (control, received no medication), group II (vehicle, received normal saline), group III (TQ, 50 mg/kg/day), group IV (VLD, 10 mg/kg/day), group V (MnCl2, 50 mg/kg/day), group VI (Mn+TQ), group VII (Mn+VLD), and group VIII (Mn+TQ+VLD). Groups VI, VII, and VIII, received the same previously mentioned doses. All drugs were orally gavaged for 12 weeks. Manganese administration resulted in an elevation in the levels of serum and tissues Mn, blood glucose, serum urea, creatinine, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and reduction in insulin, kidney superoxide dismutase (SOD), glutathione (GSH), and interleukin-10. Histopathological structural renal damage was detected associated with strong positive immunoexpression of caspase-3. On the other hand, individual or combined TQ and VLD administration with Mn significantly decreased the serum and tissue levels of Mn, declined the blood glucose, inflammatory markers, oxidative stress markers, ameliorated the histopathological effects, and down-regulated the immunoexpression of caspase-3. In conclusion, TQ and VLD co-administration elicited protective effects against Mn-induced nephrotoxicity.
Collapse
Affiliation(s)
- Heba El-Sayed Mostafa
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Eman Ahmed Alaa El-Din
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt.
| | - Dalia Abdallah El-Shafei
- Department of Community, Environmental & Occupational Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Nehal S Abouhashem
- Department of Pathology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Aisha Abdallah Abouhashem
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
22
|
Wei X, Cai M, Jin L. The Function of the Metals in Regulating Epigenetics During Parkinson's Disease. Front Genet 2021; 11:616083. [PMID: 33603768 PMCID: PMC7884633 DOI: 10.3389/fgene.2020.616083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/30/2020] [Indexed: 11/13/2022] Open
Abstract
Parkinson's means Parkinson's disease, a chronic degenerative disease of central nervous system. The main area which is affected by this disease is motor system. Since it firstly founded by James Parkinson in his 1817 publication, nowadays, people still have lots of questions about this disease. This review mainly summarizes the epigenetics of Parkinson's. DNA methylation is one of the epigenetic mechanisms of Parkinson's. During the development of disease, global hypomethylation, and hypermethylation happen in different areas of patients. Another epigenetic mechanism is histone modification. People believe that some metals can induce Parkinson's disease by modulating epigenetic mechanisms. This review summarizes the relationships between different metals and Parkinson's disease. However, the specific roles of most metals in epigenetics are still unknown, which need further research.
Collapse
Affiliation(s)
- Xiangzhen Wei
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Menghua Cai
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Lifang Jin
- Department of Biological Sciences, Shaoxing University, Shaoxing, China
| |
Collapse
|
23
|
Illarionova NB, Morozova KN, Petrovskii DV, Sharapova MB, Romashchenko AV, Troitskii SY, Kiseleva E, Moshkin YM, Moshkin MP. 'Trojan-Horse' stress-granule formation mediated by manganese oxide nanoparticles. Nanotoxicology 2020; 14:1432-1444. [PMID: 33320703 DOI: 10.1080/17435390.2020.1856433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Exposure to nanomaterials is considered as one of the risk factors for neurodegenerative pathology. In vitro inorganic nanoparticles (NPs) absorb intrinsically disordered proteins, many of which are the constituents of stress-granules (SGs). SGs normally form in response to cellular stress and, here, we addressed whether selected inorganic NPs could trigger SGs formation in cells. To this end, we have tested a series of inorganic NPs for their ability to induce SGs formation in human glioblastoma and fibroblast cell lines. Among tested NPs, only Mn3O4 NPs triggered SGs formation in cell-type-specific and metabolic-dependent manner. In human glioblastoma U87 MG cell line, Mn3O4 NPs entered cells within minutes and resided inside intracellular vesicles for at least 48 h. Mn3O4 NPs induced a strong reduction in oxidative phosphorylation rate, but not glycolysis. We showed that Mn3O4 NPs slowly dissolve producing a local net of Mn2+ cations, which are known to inhibit oxidative phosphorylation. Indeed, direct incubation of cells with equimolar amounts of Mn2+ cations triggered SGs formation and reduced cellular respiration rate. However, while SGs formed in response to Mn3O4 NPs persisted for hours, SGs formation by Mn2+ peaked and dropped within minutes. Finally, Mn3O4 NPs mediated SGs formation via the phosphorylation of eIF2α. Thus, we conclude that exposure of U87 MG cells to Mn3O4 NPs caused a 'Trojan-horse' prolonged SGs response.
Collapse
Affiliation(s)
| | - Ksenia N Morozova
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Dmitry V Petrovskii
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | | | | | - Sergey Y Troitskii
- Department of Heterogeneous Catalysis, Boreskov Institute of Catalysis, Siberian Branch of RAS, Novosibirsk, Russia
| | - Elena Kiseleva
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Yuri M Moshkin
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Mikhail P Moshkin
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia.,National Research Tomsk State University, Tomsk, Russia
| |
Collapse
|
24
|
Iqubal A, Ahmed M, Ahmad S, Sahoo CR, Iqubal MK, Haque SE. Environmental neurotoxic pollutants: review. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:41175-41198. [PMID: 32820440 DOI: 10.1007/s11356-020-10539-z] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 08/16/2020] [Indexed: 05/23/2023]
Abstract
Environmental pollutants are recognized as one of the major concerns for public health and responsible for various forms of neurological disorders. Some of the common sources of environmental pollutants related to neurotoxic manifestations are industrial waste, pesticides, automobile exhaust, laboratory waste, and burning of terrestrial waste. Among various environmental pollutants, particulate matter, ultrafine particulate matter, nanoparticles, and lipophilic vaporized toxicant (acrolein) easily cross the blood-brain barrier, activate innate immune responses in the astrocytes, microglia, and neurons, and exert neurotoxicity. Growing shreds of evidence from human epidemiological studies have correlated the environmental pollutants with neuroinflammation, oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction, myelin sheath disruption, and alterations in the blood-brain barrier anatomy leading to cognitive dysfunction and poor quality of life. These environmental pollutants also considerably cause developmental neurotoxicity, exhibit teratogenic effect and mental growth retardance, and reduce IQ level. Until now, the exact mechanism of pollutant-induced neurotoxicity is not known, but studies have shown interference of pollutants with the endogenous antioxidant defense system, inflammatory pathway (Nrf2/NF-kB, MAPKs/PI3K, and Akt/GSK3β), modulation of neurotransmitters, and reduction in long-term potentiation. In the current review, various sources of pollutants and exposure to the human population, developmental neurotoxicity, and molecular mechanism of different pollutants involved in the pathogenesis of different neurological disorders have been discussed.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Musheer Ahmed
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Shahnawaz Ahmad
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Chita Ranjan Sahoo
- Central Research Laboratory, Institute of Medical Sciences & Sum Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
25
|
Hernández RB, Carrascal M, Abian J, Michalke B, Farina M, Gonzalez YR, Iyirhiaro GO, Moteshareie H, Burnside D, Golshani A, Suñol C. Manganese-induced neurotoxicity in cerebellar granule neurons due to perturbation of cell network pathways with potential implications for neurodegenerative disorders. Metallomics 2020; 12:1656-1678. [PMID: 33206086 DOI: 10.1039/d0mt00085j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Manganese (Mn) is essential for living organisms, playing an important role in nervous system function. Nevertheless, chronic and/or acute exposure to this metal, especially during early life stages, can lead to neurotoxicity and dementia by unclear mechanisms. Thus, based on previous works of our group with yeast and zebrafish, we hypothesized that the mechanisms mediating manganese-induced neurotoxicity can be associated with the alteration of protein metabolism. These mechanisms may also depend on the chemical speciation of manganese. Therefore, the current study aimed at investigating the mechanisms mediating the toxic effects of manganese in primary cultures of cerebellar granule neurons (CGNs). By exposing cultured CGNs to different chemical species of manganese ([[2-[(dithiocarboxy)amino]ethyl]carbamodithioato]](2-)-kS,kS']manganese, named maneb (MB), and [[1,2-ethanediylbis[carbamodithioato]](2-)]manganese mixture with [[1,2-ethanediylbis[carbamodithioato]](2-)]zinc, named mancozeb (MZ), and manganese chloride (MnCl2)), and using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, we observed that both MB and MZ induced similar cytotoxicity (LC50∼ 7-9 μM), which was higher than that of MnCl2 (LC50∼ 27 μM). Subsequently, we applied systems biology approaches, including metallomics, proteomics, gene expression and bioinformatics, and revealed that independent of chemical speciation, for non-cytotoxic concentrations (0.3-3 μM), Mn-induced neurotoxicity in CGNs is associated with metal dyshomeostasis and impaired protein metabolism. In this way, we verified that MB induced more post-translational alterations than MnCl2, which can be a plausible explanation for cytotoxic differences between both chemical species. The metabolism of proteins is one of the most energy consuming cellular processes and its impairment appears to be a key event of some cellular stress processes reported separately in other studies such as cell cycle arrest, energy impairment, cell signaling, excitotoxicity, immune response, potential protein accumulation and apoptosis. Interestingly, we verified that Mn-induced neurotoxicity shares pathways associated with the development of Alzheimer's disease, Amyotrophic Lateral Sclerosis, Huntington's disease, and Parkinson's disease. This has been observed in baker's yeast and zebrafish suggesting that the mode of action of Mn may be evolutionarily conserved.
Collapse
Affiliation(s)
- Raúl Bonne Hernández
- Laboratory of Bioinorganic and Environmental Toxicology - LABITA, Department of Exact and Earth Sciences, Federal University of São Paulo, Rua Prof. Artur Riedel, 275, CEP 09972-270, Diadema, SP, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Synthesis, structural characterization, DFT, kinetics and mechanism of oxidation of bromothymol blue: application to textile industrial wastewater treatment. CHEMICAL PAPERS 2020. [DOI: 10.1007/s11696-020-01299-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
27
|
Kumar V, Santhosh Kumar TR, Kartha CC. Mitochondrial membrane transporters and metabolic switch in heart failure. Heart Fail Rev 2020; 24:255-267. [PMID: 30535838 DOI: 10.1007/s10741-018-9756-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mitochondrial dysfunction is widely recognized as a major factor for the progression of cardiac failure. Mitochondrial uptake of metabolic substrates and their utilization for ATP synthesis, electron transport chain activity, reactive oxygen species levels, ion homeostasis, mitochondrial biogenesis, and dynamics as well as levels of reactive oxygen species in the mitochondria are key factors which regulate mitochondrial function in the normal heart. Alterations in these functions contribute to adverse outcomes in heart failure. Iron imbalance and oxidative stress are also major factors for the evolution of cardiac hypertrophy, heart failure, and aging-associated pathological changes in the heart. Mitochondrial ATP-binding cassette (ABC) transporters have a key role in regulating iron metabolism and maintenance of redox status in cells. Deficiency of mitochondrial ABC transporters is associated with an impaired mitochondrial electron transport chain complex activity, iron overload, and increased levels of reactive oxygen species, all of which can result in mitochondrial dysfunction. In this review, we discuss the role of mitochondrial ABC transporters in mitochondrial metabolism and metabolic switch, alterations in the functioning of ABC transporters in heart failure, and mitochondrial ABC transporters as possible targets for therapeutic intervention in cardiac failure.
Collapse
Affiliation(s)
- Vikas Kumar
- Cardiovascular Diseases and Diabetes Biology group, Rajiv Gandhi Centre for Biotechnology (RGCB), Poojappura, Thycaud Post, Trivandrum, Kerala, 695014, India.,Graduate Studies, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - T R Santhosh Kumar
- Cardiovascular Diseases and Diabetes Biology group, Rajiv Gandhi Centre for Biotechnology (RGCB), Poojappura, Thycaud Post, Trivandrum, Kerala, 695014, India.,Graduate Studies, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India.,Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Trivandrum, Kerala, India
| | - C C Kartha
- Cardiovascular Diseases and Diabetes Biology group, Rajiv Gandhi Centre for Biotechnology (RGCB), Poojappura, Thycaud Post, Trivandrum, Kerala, 695014, India.
| |
Collapse
|
28
|
Martins AC, Morcillo P, Ijomone OM, Venkataramani V, Harrison FE, Lee E, Bowman AB, Aschner M. New Insights on the Role of Manganese in Alzheimer's Disease and Parkinson's Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E3546. [PMID: 31546716 PMCID: PMC6801377 DOI: 10.3390/ijerph16193546] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
Manganese (Mn) is an essential trace element that is naturally found in the environment and is necessary as a cofactor for many enzymes and is important in several physiological processes that support development, growth, and neuronal function. However, overexposure to Mn may induce neurotoxicity and may contribute to the development of Alzheimer's disease (AD) and Parkinson's disease (PD). The present review aims to provide new insights into the involvement of Mn in the etiology of AD and PD. Here, we discuss the critical role of Mn in the etiology of these disorders and provide a summary of the proposed mechanisms underlying Mn-induced neurodegeneration. In addition, we review some new therapy options for AD and PD related to Mn overload.
Collapse
Affiliation(s)
- Airton Cunha Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA (P.M.)
| | - Patricia Morcillo
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA (P.M.)
| | - Omamuyovwi Meashack Ijomone
- Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology Akure, Akure 340252, Nigeria;
| | - Vivek Venkataramani
- Department of Hematology and Medical Oncology and Institute of Pathology, University Medical Center Göttingen (UMG), 37075 Göttingen, Germany;
| | - Fiona Edith Harrison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32301, USA;
| | - Aaron Blaine Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, USA;
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA (P.M.)
| |
Collapse
|
29
|
Kupsco A, Sanchez-Guerra M, Amarasiriwardena C, Brennan KJM, Estrada-Gutierrez G, Svensson K, Schnaas L, Pantic I, Téllez-Rojo MM, Baccarelli AA, Wright RO. Prenatal manganese and cord blood mitochondrial DNA copy number: Effect modification by maternal anemic status. ENVIRONMENT INTERNATIONAL 2019; 126:484-493. [PMID: 30849576 PMCID: PMC6471611 DOI: 10.1016/j.envint.2019.02.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 05/06/2023]
Abstract
INTRODUCTION Manganese (Mn) is an essential nutrient but also a toxicant at high exposures, when it can induce oxidative stress (OS). Mn uptake is inversely correlated with iron status, therefore anemic individuals may be more susceptible to Mn overload induced-OS, which can manifest as changes in mitochondrial DNA copy number (mtDNA CN). Our objectives were to: 1) determine stage-specific associations of prenatal Mn exposure with cord blood MtDNA CN; and 2) investigate effect modification by maternal anemia, ferritin, and mean corpuscular volume (MCV). MATERIALS AND METHODS We measured whole blood Mn, hemoglobin, serum ferritin, and MCV in the 2nd and 3rd trimester, in maternal blood at birth, and in cord blood from a prospective birth cohort in Mexico City, Mexico (n = 485). We then extracted DNA from cord blood leukocytes to determine mtDNA CN. We used robust regression to measure associations between Mn and mtDNA CN at each trimester and at birth. Anemia (hemoglobin ≤11 g/dL), iron deficiency (ferritin ≤15 ng/mL) and MCV (stratified at median), were examined as effect modifiers. RESULTS Mn levels increased throughout pregnancy, and Mn was inversely correlated with ferritin. We observed a positive association between Mn in the 3rd trimester and Mn in cord blood and mtDNA CN (β = 0.04-0.05; 95% CI = 0.01, 0.08). Anemia significantly modified the association between mtDNA CN and Mn in the 2nd trimester. We found a positive association between 2nd trimester Mn and mtDNA CN in mothers with normal hemoglobin, and a negative association in those with low hemoglobin. (βhigh = 0.06; 95% CI = 0.01, 0.11; p = 0.01 and βlow = -0.06; 95% CI = 0.03, -0.13; p = 0.06). No associations were detected between anemia, iron deficiency and MCV and mtDNA CN. CONCLUSIONS Maternal blood Mn in the 3rd trimester and in cord blood was positively associated with mtDNA CN, suggesting that higher late pregnancy prenatal Mn exposures can impact newborn mitochondria by promoting OS. Furthermore, 2nd trimester Mn was positively associated with mtDNA in non-anemic mother-child pairs but inversely associated in anemic individuals, indicating potential interactions between Mn and chronic anemia.
Collapse
Affiliation(s)
- Allison Kupsco
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA.
| | | | - Chitra Amarasiriwardena
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kasey J M Brennan
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA
| | | | - Katherine Svensson
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Ivan Pantic
- National Institute of Perinatology, Mexico City, Mexico
| | - Martha María Téllez-Rojo
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
30
|
Chen P, Totten M, Zhang Z, Bucinca H, Erikson K, Santamaría A, Bowma AB, Aschner M. Iron and manganese-related CNS toxicity: mechanisms, diagnosis and treatment. Expert Rev Neurother 2019; 19:243-260. [PMID: 30759034 PMCID: PMC6422746 DOI: 10.1080/14737175.2019.1581608] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 02/08/2019] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Iron (Fe) and manganese (Mn) are essential nutrients for humans. They act as cofactors for a variety of enzymes. In the central nervous system (CNS), these two metals are involved in diverse neurological activities. Dyshomeostasis may interfere with the critical enzymatic activities, hence altering the neurophysiological status and resulting in neurological diseases. Areas covered: In this review, the authors cover the molecular mechanisms of Fe/Mn-induced toxicity and neurological diseases, as well as the diagnosis and potential treatment. Given that both Fe and Mn are abundant in the earth crust, nutritional deficiency is rare. In this review the authors focus on the neurological disorders associated with Mn and Fe overload. Expert commentary: Oxidative stress and mitochondrial dysfunction are the primary molecular mechanism that mediates Fe/Mn-induced neurotoxicity. Although increased Fe or Mn concentrations have been found in brain of patients, it remains controversial whether the elevated metal amounts are the primary cause or secondary consequence of neurological diseases. Currently, treatments are far from satisfactory, although chelation therapy can significantly decrease brain Fe and Mn levels. Studies to determine the primary cause and establish the molecular mechanism of toxicity may help to adapt more comprehensive and satisfactory treatments in the future.
Collapse
Affiliation(s)
- Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Melissa Totten
- Department of Nutrition, University of North Carolina Greensboro, Greensboro, NC, USA
| | - Ziyan Zhang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hana Bucinca
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Keith Erikson
- Department of Nutrition, University of North Carolina Greensboro, Greensboro, NC, USA
| | - Abel Santamaría
- Laboratory of Excitatory Amino Acids, National Institute of Neurology and Neurosurgery, Mexico, Mexico City, Mexico
| | - Aaron B. Bowma
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
31
|
Kamemura N. Methylcyclopentadienyl manganese tricarbonyl increases cell vulnerability to oxidative stress on rat thymocytes. Drug Chem Toxicol 2018; 42:140-146. [PMID: 29359594 DOI: 10.1080/01480545.2018.1424180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Methylcyclopentadienyl manganese tricarbonyl (MMT) is used as a gasoline antiknock additive. However, the toxic effect of MMT is currently not well understood. In this study, we investigated the toxic effect of MMT on rat thymocytes using a flow cytometer and fluorescent probes. MMT at 100-300 µM significantly increased the population of cells exhibiting propidium fluorescence, i.e., the population of dead cells. The intensity of BES-So-AM fluorescence significantly increased when using 100 µM MMT. In addition, the intensity of oxonol fluorescence in rat thymocytes increased with the treatment with MMT in a concentration-dependent manner (10-100 µM). The toxic effect of MMT was inhibited by quercetin, antioxidant flavonoid. Moreover, co-treatment with 30-100 µM MMT and 100 µM H2O2 increased the cell lethality further. These results indicate that MMT increases cell vulnerability to oxidative stress on rat thymocytes. This study provides insight into the toxic effect of MMT on the immune system.
Collapse
Affiliation(s)
- Norio Kamemura
- a Division of Bioscience and Bioindustry , Tokushima University , Tokushima , Japan
| |
Collapse
|
32
|
Helley MP, Pinnell J, Sportelli C, Tieu K. Mitochondria: A Common Target for Genetic Mutations and Environmental Toxicants in Parkinson's Disease. Front Genet 2017; 8:177. [PMID: 29204154 PMCID: PMC5698285 DOI: 10.3389/fgene.2017.00177] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/01/2017] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is a devastating neurological movement disorder. Since its first discovery 200 years ago, genetic and environmental factors have been identified to play a role in PD development and progression. Although genetic studies have been the predominant driving force in PD research over the last few decades, currently only a small fraction of PD cases can be directly linked to monogenic mutations. The remaining cases have been attributed to other risk associated genes, environmental exposures and gene-environment interactions, making PD a multifactorial disorder with a complex etiology. However, enormous efforts from global research have yielded significant insights into pathogenic mechanisms and potential therapeutic targets for PD. This review will highlight mitochondrial dysfunction as a common pathway involved in both genetic mutations and environmental toxicants linked to PD.
Collapse
Affiliation(s)
- Martin P. Helley
- Department of Environmental Health Sciences, Florida International University, Miami, FL, United States
| | - Jennifer Pinnell
- Department of Environmental Health Sciences, Florida International University, Miami, FL, United States
- Peninsula Schools of Medicine and Dentistry, Plymouth University, Plymouth, United Kingdom
| | - Carolina Sportelli
- Department of Environmental Health Sciences, Florida International University, Miami, FL, United States
- Peninsula Schools of Medicine and Dentistry, Plymouth University, Plymouth, United Kingdom
| | - Kim Tieu
- Department of Environmental Health Sciences, Florida International University, Miami, FL, United States
| |
Collapse
|
33
|
Neely MD, Davison CA, Aschner M, Bowman AB. From the Cover: Manganese and Rotenone-Induced Oxidative Stress Signatures Differ in iPSC-Derived Human Dopamine Neurons. Toxicol Sci 2017; 159:366-379. [PMID: 28962525 PMCID: PMC5837701 DOI: 10.1093/toxsci/kfx145] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) is the result of complex interactions between genetic and environmental factors. Two chemically distinct environmental stressors relevant to PD are the metal manganese and the pesticide rotenone. Both are thought to exert neurotoxicity at least in part via oxidative stress resulting from impaired mitochondrial activity. Identifying shared mechanism of action may reveal clues towards an understanding of the mechanisms underlying PD pathogenesis. Here we compare the effects of manganese and rotenone in human-induced pluripotent stem cells-derived postmitotic mesencephalic dopamine neurons by assessing several different oxidative stress endpoints. Manganese, but not rotenone caused a concentration and time-dependent increase in intracellular reactive oxygen/nitrogen species measured by quantifying the fluorescence of oxidized chloromethyl 2',7'-dichlorodihydrofluorescein diacetate (DCF) assay. In contrast, rotenone but not manganese caused an increase in cellular isoprostane levels, an indicator of lipid peroxidation. Manganese and rotenone both caused an initial decrease in cellular reduced glutathione; however, glutathione levels remained low in neurons treated with rotenone for 24 h but recovered in manganese-exposed cells. Neurite length, a sensitive indicator of overall neuronal health was adversely affected by rotenone, but not manganese. Thus, our observations suggest that the cellular oxidative stress evoked by these 2 agents is distinct yielding unique oxidative stress signatures across outcome measures. The protective effect of rasagiline, a compound used in the clinic for PD, had negligible impact on any of oxidative stress outcome measures except a subtle significant decrease in manganese-dependent production of reactive oxygen/nitrogen species detected by the DCF assay.
Collapse
Affiliation(s)
- M. Diana Neely
- Department of Pediatrics
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Carrie Ann Davison
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Aaron B. Bowman
- Department of Pediatrics
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232
| |
Collapse
|
34
|
Kaur G, Kumar V, Arora A, Tomar A, Ashish, Sur R, Dutta D. Affected energy metabolism under manganese stress governs cellular toxicity. Sci Rep 2017; 7:11645. [PMID: 28928443 PMCID: PMC5605510 DOI: 10.1038/s41598-017-12004-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 08/31/2017] [Indexed: 12/03/2022] Open
Abstract
Excessive manganese exposure is toxic, but a comprehensive biochemical picture of this assault is poorly understood. Whether oxidative stress or reduced energy metabolism under manganese exposure causes toxicity is still a debate. To address this, we chose ΔmntPEscherichia coli, a highly manganese-sensitive strain, in this study. Combining microarray, proteomics, and biochemical analyses, we show that the chronic manganese exposure rewires diverse regulatory and metabolic pathways. Manganese stress affects protein and other macromolecular stability, and envelope biogenesis. Most importantly, manganese exposure disrupts both iron-sulfur cluster and heme-enzyme biogenesis by depleting cellular iron level. Therefore, the compromised function of the iron-dependent enzymes in the tricarboxylic acid cycle, and electron transport chain impede ATP synthesis, leading to severe energy deficiency. Manganese stress also evokes reactive oxygen species, inducing oxidative stress. However, suppressing oxidative stress does not improve oxidative phosphorylation and cell growth. On the contrary, iron supplementation resumed cell growth stimulating oxidative phosphorylation. Therefore, we hypothesize that affected energy metabolism is the primal cause of manganese toxicity.
Collapse
Affiliation(s)
- Gursharan Kaur
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India.,Department of Biophysics, Molecular Biology & Bioinformatics, Calcutta University, Kolkata, India
| | - Vineet Kumar
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
| | - Amit Arora
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
| | - Ajay Tomar
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
| | - Ashish
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
| | - Runa Sur
- Department of Biophysics, Molecular Biology & Bioinformatics, Calcutta University, Kolkata, India
| | - Dipak Dutta
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India.
| |
Collapse
|
35
|
Pfalzer AC, Bowman AB. Relationships Between Essential Manganese Biology and Manganese Toxicity in Neurological Disease. Curr Environ Health Rep 2017; 4:223-228. [PMID: 28417441 PMCID: PMC5515274 DOI: 10.1007/s40572-017-0136-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Manganese (Mn) is critical for neurodevelopment but also has been implicated in the pathophysiology of several neurological diseases. We discuss how Mn requirements intersect with Mn biology and toxicity, and how these requirements may be altered in neurological disease. Furthermore, we discuss the emerging evidence that the level of Mn associated with optimal overall efficiency for Mn biology does not necessarily coincide with optimal cognitive outcomes. RECENT FINDINGS Studies have linked Mn exposures with urea cycle metabolism and autophagy, with evidence that exposures typically neurotoxic may be able to correct deficiencies in these processes at least short term. The line between Mn-dependent biology and toxicity is thus blurred. Further, new work suggests that Mn exposures correlating to optimal cognitive scores in children are associated with cognitive decline in adults. This review explores relationships between Mn-dependent neurobiology and Mn-dependent neurotoxicity. We propose the hypothesis that Mn levels/exposures that are toxic to some biological processes are beneficial for other biological processes and influenced by developmental stage and disease state.
Collapse
Affiliation(s)
- Anna C Pfalzer
- Departments of Pediatrics, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Aaron B Bowman
- Departments of Pediatrics, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA.
- Department of Neurology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA.
- Department of Biochemistry, Vanderbilt Brain Institute, Kennedy Center for Research and Human Development, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
36
|
Marreilha dos Santos AP, Andrade V, Aschner M. Neuroprotective and Therapeutic Strategies for Manganese-Induced Neurotoxicity. CLINICAL PHARMACOLOGY AND TRANSLATIONAL MEDICINE 2017; 1:54-62. [PMID: 30854510 PMCID: PMC6402347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Manganese (Mn) is an essential element required for growth, development and general maintenance of health. However, chronic or high occupational and environmental exposure to excessive levels of Mn has long been known to lead to a progressive neurological disorder similar to Parkinsonism. Manganism patients display a variety of symptoms, including mental, cognitive and behavioural impediments, as well as motor dysfunctions that are associated with basal ganglia dysfunction. Taking into account the pharmacokinetics and Mn-related toxicity mechanisms, several neuroprotective compounds and therapeutic approaches have been investigated to assess their efficacy in mitigating its neurotoxicity. Here, we will briefly address some of the toxic mechanisms of Mn, followed by neuroprotective strategies and therapeutic approaches aiming to reduce or treat Mn induced neurotoxicity. Natural and synthetic antioxidants, anti-inflammatory compounds, ATP/ADP ratio protectors and glutamate protectors have been introduced in view of decreasing Mn-induced neurotoxicity. In addition, the efficacy and mechanisms of several therapeutic interventions such as levodopa, ethylene-diamine-tetraacetic acid (EDTA) and para-aminosalicylic acid (PAS), aimed at ameliorating Mn neurotoxic symptoms in humans, will be reviewed.
Collapse
Affiliation(s)
- AP Marreilha dos Santos
- Institute of Medicine Research (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon. Portugal
| | - V Andrade
- Institute of Medicine Research (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon. Portugal
| | - M Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, USA
| |
Collapse
|
37
|
Nielsen BS, Larsen EH, Ladefoged O, Lam HR. Subchronic, Low-Level Intraperitoneal Injections of Manganese (IV) Oxide and Manganese (II) Chloride Affect Rat Brain Neurochemistry. Int J Toxicol 2017; 36:239-251. [PMID: 28460583 DOI: 10.1177/1091581817704378] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Manganese (Mn) is neurotoxic and can induce manganism, a Parkinson-like disease categorized as being a serious central nervous system irreversible neurodegenerative disease. An increased risk of developing symptoms of Parkinson disease has been linked to work-related exposure, for example, for workers in agriculture, horticulture, and people living near areas with frequent use of Mn-containing pesticides. In this study, the focus was placed on neurochemical effects of Mn. Rats were dosed intraperitoneally with 0.9% NaCl (control), 1.22 mg Mn (as MnO2)/kg bodyweight (bw)/day, or 2.5 mg Mn (as MnCl2)/kg bw/day for 7 d/wk for 8 or 12 weeks. This dosing regimen adds relevant new knowledge about Mn neurotoxicity as a consequence of low-dose subchronic Mn dosing. Manganese concentrations increased in the striatum, the rest of the brain, and in plasma, and regional brain neurotransmitter concentrations, including noradrenaline, dopamine (DA), 5-hydroxytrytamine, glutamate, taurine, and γ-amino butyric acid, and the activity of acetylcholinesterase changed. Importantly, a target parameter for Parkinson disease and manganism, the striatal DA concentration, was reduced after 12 weeks of dosing with MnCl2. Plasma prolactin concentration was not significantly affected due to a potentially reduced dopaminergic inhibition of the prolactin release from the anterior hypophysis. No effects on the striatal α-synuclein and synaptophysin protein levels were detected.
Collapse
Affiliation(s)
| | - Erik H Larsen
- 2 Division of Food Production, National Food Institute, Søborg, Denmark
| | - Ole Ladefoged
- 3 Division of Toxicology and Risk Assessment, National Food Institute, Søborg, Denmark
| | - Henrik R Lam
- 1 Environment and Toxicology, DHI, Hørsholm, Denmark
| |
Collapse
|
38
|
Smith MR, Fernandes J, Go YM, Jones DP. Redox dynamics of manganese as a mitochondrial life-death switch. Biochem Biophys Res Commun 2017; 482:388-398. [PMID: 28212723 PMCID: PMC5382988 DOI: 10.1016/j.bbrc.2016.10.126] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/25/2016] [Accepted: 10/27/2016] [Indexed: 12/16/2022]
Abstract
Sten Orrenius, M.D., Ph.D., pioneered many areas of cellular and molecular toxicology and made seminal contributions to our knowledge of oxidative stress and glutathione (GSH) metabolism, organellar functions and Ca+2-dependent mechanisms of cell death, and mechanisms of apoptosis. On the occasion of his 80th birthday, we summarize current knowledge on redox biology of manganese (Mn) and its role in mechanisms of cell death. Mn is found in all organisms and has critical roles in cell survival and death mechanisms by regulating Mn-containing enzymes such as manganese superoxide dismutase (SOD2) or affecting expression and activity of caspases. Occupational exposures to Mn cause "manganism", a Parkinson's disease-like condition of neurotoxicity, and experimental studies show that Mn exposure leads to accumulation of Mn in the brain, especially in mitochondria, and neuronal cell death occurs with features of an apoptotic mechanism. Interesting questions are why a ubiquitous metal that is essential for mitochondrial function would accumulate to excessive levels, cause increased H2O2 production and lead to cell death. Is this due to the interactions of Mn with other essential metals, such as iron, or with toxic metals, such as cadmium? Why is the Mn loading in the human brain so variable, and why is there such a narrow window between dietary adequacy and toxicity? Are non-neuronal tissues similarly vulnerable to insufficiency and excess, yet not characterized? We conclude that Mn is an important component of the redox interface between an organism and its environment and warrants detailed studies to understand the role of Mn as a mitochondrial life-death switch.
Collapse
Affiliation(s)
- Matthew Ryan Smith
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Jolyn Fernandes
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Young-Mi Go
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
39
|
Bonke E, Siebels I, Zwicker K, Dröse S. Manganese ions enhance mitochondrial H 2O 2 emission from Krebs cycle oxidoreductases by inducing permeability transition. Free Radic Biol Med 2016; 99:43-53. [PMID: 27474449 DOI: 10.1016/j.freeradbiomed.2016.07.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/21/2016] [Accepted: 07/25/2016] [Indexed: 11/24/2022]
Abstract
Manganese-induced toxicity has been linked to mitochondrial dysfunction and an increased generation of reactive oxygen species (ROS). We could recently show in mechanistic studies that Mn2+ ions induce hydrogen peroxide (H2O2) production from the ubiquinone binding site of mitochondrial complex II (IIQ) and generally enhance H2O2 formation by accelerating the rate of superoxide dismutation. The present study with intact mitochondria reveals that manganese additionally enhances H2O2 emission by inducing mitochondrial permeability transition (mPT). In mitochondria fed by NADH-generating substrates, the combination of Mn2+ and different respiratory chain inhibitors led to a dynamically increasing H2O2emission which was sensitive to the mPT inhibitor cyclosporine A (CsA) as well as Ru-360, an inhibitor of the mitochondrial calcium uniporter (MCU). Under these conditions, flavin-containing enzymes of the mitochondrial matrix, e.g. the mitochondrial 2-oxoglutaratedehydrogenase (OGDH), were major sources of ROS. With succinate as substrate, Mn2+ stimulated ROS production mainly at complex II, whereby the applied succinate concentration had a marked effect on the tendency for mPT. Also Ca2+ increased the rate of H2O2 emission by mPT, while no direct effect on ROS-production of complex II was observed. The present study reveals a complex scenario through which manganese affects mitochondrial H2O2 emission: stimulating its production from distinct sites (e.g. site IIQ), accelerating superoxide dismutation and enhancing the emission via mPT which also leads to the loss of soluble components of the mitochondrial antioxidant systems and favors the ROS production from flavin-containing oxidoreductases of the Krebs cycle.
Collapse
Affiliation(s)
- Erik Bonke
- Department of Anesthesiology, Intensive-Care Medicine and Pain Therapy, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | - Ilka Siebels
- Department of Anesthesiology, Intensive-Care Medicine and Pain Therapy, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | - Klaus Zwicker
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Stefan Dröse
- Department of Anesthesiology, Intensive-Care Medicine and Pain Therapy, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
40
|
Abstract
Metals play important roles in the human body, maintaining cell structure and regulating gene expression, neurotransmission, and antioxidant response, to name a few. However, excessive metal accumulation in the nervous system may be toxic, inducing oxidative stress, disrupting mitochondrial function, and impairing the activity of numerous enzymes. Damage caused by metal accumulation may result in permanent injuries, including severe neurological disorders. Epidemiological and clinical studies have shown a strong correlation between aberrant metal exposure and a number of neurological diseases, including Alzheimer’s disease, amyotrophic lateral sclerosis, autism spectrum disorders, Guillain–Barré disease, Gulf War syndrome, Huntington’s disease, multiple sclerosis, Parkinson’s disease, and Wilson’s disease. Here, we briefly survey the literature relating to the role of metals in neurodegeneration.
Collapse
Affiliation(s)
- Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York, USA
| | - Mahfuzur Rahman Miah
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
41
|
Abstract
SIGNIFICANCE Mitochondria are structurally and biochemically diverse, even within a single type of cell. Protein complexes localized to the inner mitochondrial membrane synthesize ATP by coupling electron transport and oxidative phosphorylation. The organelles produce reactive oxygen species (ROS) from mitochondrial oxygen and ROS can, in turn, alter the function and expression of proteins used for aerobic respiration by post-translational and transcriptional regulation. RECENT ADVANCES New interest is emerging not only into the roles of mitochondria in disease development and progression but also as a target for environmental toxicants. CRITICAL ISSUES Dysregulation of respiration has been linked to cell death and is a major contributor to acute neuronal trauma, peripheral diseases, as well as chronic neurodegenerative diseases, such as Parkinson's disease and Alzheimer's disease. FUTURE DIRECTIONS Here, we discuss the mechanisms underlying the sensitivity of the mitochondrial respiratory complexes to redox modulation, as well as examine the effects of environmental contaminants that have well-characterized mitochondrial toxicity. The contaminants discussed in this review are some of the most prevalent and potent environmental contaminants that have been linked to neurological dysfunction, altered cellular respiration, and oxidation.
Collapse
Affiliation(s)
- Samuel W Caito
- Department of Molecular Pharmacology, Albert Einstein College of Medicine , Bronx, New York
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine , Bronx, New York
| |
Collapse
|
42
|
Bonke E, Zwicker K, Dröse S. Manganese ions induce H2O2 generation at the ubiquinone binding site of mitochondrial complex II. Arch Biochem Biophys 2015; 580:75-83. [DOI: 10.1016/j.abb.2015.06.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 06/18/2015] [Accepted: 06/19/2015] [Indexed: 12/28/2022]
|
43
|
Chen P, Bowman AB, Mukhopadhyay S, Aschner M. SLC30A10: A novel manganese transporter. WORM 2015; 4:e1042648. [PMID: 26430566 DOI: 10.1080/21624054.2015.1042648] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/31/2015] [Accepted: 04/10/2015] [Indexed: 10/23/2022]
Abstract
Homozygous mutations in SLC30A10 cause familial parkinsonism associated with manganese (Mn) retention. We recently identified SLC30A10 to be a cell surface-localized Mn efflux transporter and demonstrated that parkinsonism-causing mutations block its intracellular trafficking and efflux function. In C. elegans, SLC30A10 over-expression protected against Mn-induced lethality and dopaminergic neurotoxicity, consistent with results in mammalian systems. Here, we present new data about SLC30A10 function in C. elegans. SLC30A10 expression did not protect worms against ZnSO4toxicity, suggesting that SLC30A10 does not mediate Zn export in C. elegans. Furthermore, while a blast search identified 5 potential SLC30A10 homologs in worms (cdf-1, cdf-2, ttm-1 and toc-1; sequence identity <35%), knock-down of these genes showed a tendency of increased survival after Mn exposure (although only ttm-1 was statistically significant), suggesting that the worm homologs may function differently.
Collapse
Affiliation(s)
- Pan Chen
- Department of Molecular Pharmacology; Albert Einstein College of Medicine ; Bronx, NY USA
| | - Aaron B Bowman
- Department of Neurology; Vanderbilt University Medical Center ; Nashville, TN USA
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology & Toxicology; Institute for Cellular and Molecular Biology and Institute for Neuroscience; College of Pharmacy; The University of Texas at Austin ; Austin, TX USA
| | - Michael Aschner
- Department of Molecular Pharmacology; Albert Einstein College of Medicine ; Bronx, NY USA
| |
Collapse
|
44
|
Dimovasili C, Aschner M, Plaitakis A, Zaganas I. Differential interaction of hGDH1 and hGDH2 with manganese: Implications for metabolism and toxicity. Neurochem Int 2015; 88:60-5. [PMID: 25837286 DOI: 10.1016/j.neuint.2015.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 03/13/2015] [Indexed: 10/23/2022]
Abstract
Manganese (Mn) is an essential trace element that serves as co-factor for many important mammalian enzymes. In humans, the importance of this cation is highlighted by the fact that low levels of Mn cause developmental and metabolic abnormalities and, on the other hand, chronic exposure to excessive amounts of Mn is characterized by neurotoxicity, possibly mediated by perturbation of astrocytic mitochondrial energy metabolism. Here we sought to study the effect of Mn on the two human glutamate dehydrogenases (hGDH1 and hGDH2, respectively), key mitochondrial enzymes involved in numerous cellular processes, including mitochondrial metabolism, glutamate homeostasis and neurotransmission, and cell signaling. Our studies showed that, compared to magnesium (Mg) and calcium (Ca), Mn exerted a significant inhibitory effect on both human isoenzymes with hGDH2 being more sensitive than hGDH1, especially under conditions of low ADP levels. Specifically, in the presence of 0.25 mM ADP, the Mn IC50 was 1.14 ± 0.02 mM and 1.54 ± 0.08 mM for hGDH2 and for hGDH1, respectively (p = 0.0001). Increasing Mn levels potentiated this differential effect, with 3 mM Mn inhibiting hGDH2 by 96.5% and hGDH1 by 70.2%. At 1 mM ADP, the Mn IC50 was 1.84 ± 0.02 mM and 2.04 ± 0.07 mM (p = 0.01) for hGDH2 and hGDH1, respectively, with 3 mM Mn inhibiting hGDH2 by 93.6% and hGDH1 by 70.9%. These results were due to the sigmoidal inhibitory curve of Mn that was more pronounced for hGDH2 than for hGDH1. Indeed, at 0.25 mM, the Hill coefficient value was higher for hGDH2 (3.42 ± 0.20) than for hGDH1 (1.94 ± 0.25; p = 0.0002) indicating that interaction of Mn with hGDH2 was substantially more co-operative than for hGDH1. These findings, showing an enhanced sensitivity of the hGDH2 isoenzyme to Mn, especially at low ADP levels, might be of pathophysiological relevance under conditions of Mn neurotoxicity.
Collapse
Affiliation(s)
- Christina Dimovasili
- Neurology Laboratory, School of Health Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Andreas Plaitakis
- Neurology Laboratory, School of Health Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Ioannis Zaganas
- Neurology Laboratory, School of Health Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece.
| |
Collapse
|
45
|
Villalobos V, Hernández-Fonseca JP, Bonilla E, Medina-Leendertz S, Mora M, Mosquera J. Ultrastructural Changes of Caudate Nucleus in Mice Chronically Treated with Manganese. Ultrastruct Pathol 2015; 39:217-25. [PMID: 25569534 DOI: 10.3109/01913123.2014.991885] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Manganese (Mn) is able to cross the blood-brain barrier and induces functional and structural alterations during the intoxication by this metal. Therefore, the effects of chronic administration of Mn in the caudate nucleus of mice were evaluated by electron microscopy. Male albino mice were injected intraperitoneally with MnCl2 (5 mg/kg/d) 5 d per week during 9 weeks. The control group received only 0.9% of NaCl solution. The caudate nuclei were extracted and subsequently processed to be observed on a conventional transmission electron microscope at 2, 4, 6, and 9 weeks after treatment. A high percentage of vacuolated and swollen mitochondria were found throughout all the analyzed periods. Myelin disarrangement and ultrastructural alterations related to edema were observed increased in Mn-treated mice at week 9. Granular degeneration of myelin at week 9 accompanied with deposition of electron dense granules in the neuropil was also observed. Edema in neuropil and glial cells was detected from week 2 to week 9 accompanied by swollen mitochondria. Neuronal bodies, synaptic terminals, and perivascular cells were found swollen. Decreased electron density in postsynaptic areas and decreased and dispersed synaptic vesicles in presynaptic areas were noted in Mn-treated animals. Some neurons from Mn-treated mice showed cisternae dilation of the Golgi apparatus. These results suggest that Mn-treatment produces structural alterations in the caudate nucleus that could be responsible for some of the neurotoxic effects of this metal.
Collapse
Affiliation(s)
- Virginia Villalobos
- Departamento de Biología, Facultad Experimental de Ciencias, Universidad del Zulia , Maracaibo , Venezuela
| | | | | | | | | | | |
Collapse
|
46
|
Chen P, Chakraborty S, Peres TV, Bowman AB, Aschner M. Manganese-induced Neurotoxicity: From C. elegans to Humans. Toxicol Res (Camb) 2014; 4:191-202. [PMID: 25893090 DOI: 10.1039/c4tx00127c] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Manganese (Mn) is one of the most abundant metals on the earth. It is required for normal cellular activities, but overexposure leads to toxicity. Neurons are more susceptible to Mn-induced toxicity than other cells, and accumulation of Mn in the brain results in Manganism that presents with Parkinson's disease (PD)-like symptoms. In the last decade, a number of Mn transporters have been identified, which improves our understanding of Mn transport in and out of cells. However, the mechanism of Mn-induced neurotoxicity is only partially uncovered, with further research needed to explore the whole picture of Mn-induced toxicity. In this review, we will address recent progress in Mn-induced neurotoxicity from C. elegans to humans, and explore future directions that will help understand the mechanisms of its neurotoxicity.
Collapse
Affiliation(s)
- Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sudipta Chakraborty
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tanara V Peres
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA ; Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Aaron B Bowman
- Department of Neurology, Vanderbilt University Medical Center, Nashville TN, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
47
|
Chtourou Y, Garoui EM, Boudawara T, Zeghal N. Protective role of silymarin against manganese-induced nephrotoxicity and oxidative stress in rat. ENVIRONMENTAL TOXICOLOGY 2014; 29:1147-1154. [PMID: 23339144 DOI: 10.1002/tox.21845] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 12/15/2012] [Accepted: 12/25/2012] [Indexed: 06/01/2023]
Abstract
Metal toxicity may occur after exposure from many sources. Oxidative stress is thought to be involved in manganese-induced toxicity and leads to various health disorders. Silymarin (SIL), a natural flavonoid, has been reported to have many benefits and medicinal properties. The aim of this study was to assess the toxicity of manganese (Mn) on oxidative stress and DNA damage in the kidney of rats and its alleviation by SIL. Manganese was given orally in drinking water (20 mg MnCl2 /mL) with or without SIL administration (100 mg /kg intraperitoneally) for 30 days. Our data showed that SIL significantly prevented Mn induced nephrotoxicity, indicated by both diagnostic indicators of kidney injury like plasma urea, uric acid and creatinine and urinary electrolyte levels and by histopathological analysis. Moreover, Mn-induced profound elevation of the production of reactive oxygen species (ROS) and altered the levels of oxidative stress related biomarkers in kidney tissue. This is evidenced by the increase of lipid peroxidation, protein carbonylation, DNA fragmentation and urinary hydrogen peroxide, while, the activities of enzymatic antioxidant and glutathione level were decreased. Treatment with SIL reduced the alterations in the renal and urine markers, decreasing lipid peroxidation markers, increasing the antioxidant cascade and decreasing the Mn-induced damage. All these changes were supported by histopathological observations. These findings suggested that the inhibition of Mn-induced damage by SIL was due at least in part to its antioxidant activity and its capacity to modulate the oxidative damage.
Collapse
Affiliation(s)
- Yassine Chtourou
- Department of Life Sciences, Animal Physiology Laboratory, UR/08-73, University of Sfax, Faculty of Sciences, BP 1171, 3000 Sfax, Tunisia
| | | | | | | |
Collapse
|
48
|
Korotkov S, Konovalova S, Emelyanova L, Brailovskaya I. Y3+, La3+, and some bivalent metals inhibited the opening of the Tl+-induced permeability transition pore in Ca2+-loaded rat liver mitochondria. J Inorg Biochem 2014; 141:1-9. [PMID: 25172992 DOI: 10.1016/j.jinorgbio.2014.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 08/05/2014] [Accepted: 08/07/2014] [Indexed: 11/18/2022]
Abstract
We showed earlier that diminution of 2,4-dinitrophenol (DNP)-stimulated respiration and increase of both mitochondrial swelling and electrochemical potential (ΔΨmito) dissipation in medium containing TlNO3 and KNO3 were caused by opening of Tl(+)-induced mitochondrial permeability transition pore (MPTP) in the inner membrane of Ca(2+)-loaded rat liver mitochondria. The MPTP opening was studied in the presence of bivalent metal ions (Sr(2+), Ba(2+), Mn(2+), Co(2+) and Ni(2+)), trivalent metal ions (Y(3+) and La(3+)), and ruthenium red. We found that these metal ions (except Ba(2+) and Co(2+)) as well as ruthenium red inhibited to the MPTP opening that manifested in preventing both diminution of the DNP-stimulated respiration and increase of the swelling and of the ΔΨmito dissipation in medium containing TlNO3, KNO3, and Ca(2+). Inhibition of the MPTP opening by Sr(2+) and Mn(2+) is suggested because of their interaction with high affinity Ca(2+) sites, facing the matrix side and participating in the MPTP opening. The inhibitory effects of metal ions (Y(3+), La(3+), and Ni(2+)), and ruthenium red are accordingly discussed in regard to competitive and noncompetitive inhibition of the mitochondrial Ca(2+)-uniporter. High concentrations (50μM) of Y(3+) and La(3+) favored of MPTP opening in the inner membrane of rat liver mitochondria in Ca(2+) free medium containing TlNO3. The latter MPTP opening was markedly eliminated by MPTP inhibitors (cyclosporine A and ADP).
Collapse
Affiliation(s)
- Sergey Korotkov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, Thorez pr. 44, 194223 St. Petersburg, Russian Federation.
| | - Svetlana Konovalova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, Thorez pr. 44, 194223 St. Petersburg, Russian Federation
| | - Larisa Emelyanova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, Thorez pr. 44, 194223 St. Petersburg, Russian Federation
| | - Irina Brailovskaya
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, Thorez pr. 44, 194223 St. Petersburg, Russian Federation
| |
Collapse
|
49
|
Liu Y, Wang D. Administration of chromium(III) and manganese(II) as a potential protective approach against daunorubicin-induced cardiotoxicity: in vitro and in vivo experimental evidence. Biol Trace Elem Res 2013; 156:253-61. [PMID: 24189981 DOI: 10.1007/s12011-013-9851-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 10/22/2013] [Indexed: 10/26/2022]
Abstract
Daunorubicin (DNR) is a widely used antitumor drug, but its application is limited because of its cardiotoxic side effects. The present study was designed to investigate the interaction between DNR and cardiac myosin (CM) in the presence of chromium(III) (Cr(3+)) and manganese(II) (Mn(2+)) using fluorescence spectrometry under simulative physiological conditions with the aim of exploring the influence of metal ion on DNR-CM complex and finding out an aggressive approach to abrogate of DNR-induced cardiotoxicity. In detail, the quenching and binding constant of ternary system, including metal ion, DNR, and CM, were measured and compared with the DNR-CM. The data from in vitro experiments indicate that the presence of Cr(3+) or Mn(2+) distinctly decreased the binding force between DNR and CM, and alleviated the cardiac toxicity caused by DNR. In addition, the variations in mice body weight and myocardial enzyme level were examined by in vivo experiments. Animals receiving Cr(3+) or Mn(2+) supplementation of DNR showed preservation of the normal pattern of the heart, especially 2.0 mg Cr(3+)/kg body wt or 50.0 mg Mn(2+)/kg body wt exhibited an obviously protective effect accompanied with body weight raise when compared with the mice treated with DNR alone, decreased the ratio of heart to body weight (BW) and the ratio of left ventricular mass to BW to the normal levels, and inhibited the leak of myocardial enzyme caused by DNR. As a result, this study suggests that pretreatment of lower dose of Cr(3+) (2 mg/kg wt) and moderate dose of Mn(2+) (50 mg/kg wt) might be useful and play an important role in ameliorating the cardiotoxicity of DNR treatment in cancer patients.
Collapse
Affiliation(s)
- Yang Liu
- School of Life Science, Wuchang University of Technology, Wuhan, Hubei Province, 430223, People's Republic of China,
| | | |
Collapse
|
50
|
Martinez-Finley EJ, Gavin CE, Aschner M, Gunter TE. Manganese neurotoxicity and the role of reactive oxygen species. Free Radic Biol Med 2013; 62:65-75. [PMID: 23395780 PMCID: PMC3713115 DOI: 10.1016/j.freeradbiomed.2013.01.032] [Citation(s) in RCA: 210] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 01/25/2013] [Accepted: 01/28/2013] [Indexed: 12/21/2022]
Abstract
Manganese (Mn) is an essential dietary nutrient, but an excess or accumulation can be toxic. Disease states, such as manganism, are associated with overexposure or accumulation of Mn and are due to the production of reactive oxygen species, free radicals, and toxic metabolites; alteration of mitochondrial function and ATP production; and depletion of cellular antioxidant defense mechanisms. This review focuses on all of the preceding mechanisms and the scientific studies that support them as well as providing an overview of the absorption, distribution, and excretion of Mn and the stability and transport of Mn compounds in the body.
Collapse
Affiliation(s)
- Ebany J Martinez-Finley
- Division of Clinical Pharmacology and Pediatric Toxicology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37240, USA; Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | | | - Michael Aschner
- Division of Clinical Pharmacology and Pediatric Toxicology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37240, USA; Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN 37240, USA; Center for Molecular Neuroscience, Vanderbilt University Medical Center, Nashville, TN 37240, USA; The Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, TN 37240, USA.
| | - Thomas E Gunter
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| |
Collapse
|