1
|
Martins AC, Oliveira-Paula GH, Tinkov AA, Skalny AV, Tizabi Y, Bowman AB, Aschner M. Role of manganese in brain health and disease: Focus on oxidative stress. Free Radic Biol Med 2025; 232:306-318. [PMID: 40086492 PMCID: PMC11985276 DOI: 10.1016/j.freeradbiomed.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Manganese (Mn) is an essential trace element crucial for various physiological processes, but excessive exposure can lead to significant health concerns, particularly neurotoxicity. This review synthesizes current knowledge on Mn-induced oxidative stress and its role in cellular dysfunction and disease. We discuss how Mn promotes toxicity through multiple mechanisms, primarily through reactive oxygen species (ROS) generation, which leads to oxidative stress and disruption of cellular processes. The review examines key pathways affected by Mn toxicity, including mitochondrial dysfunction, endoplasmic reticulum stress, inflammasome activation, and epigenetic modifications. Recent studies have identified promising therapeutic compounds, including both synthetic and natural substances such as probucol, metformin, curcumin, resveratrol, and daidzein, which demonstrate protective effects through various mechanisms, including antioxidant enhancement, mitochondrial function preservation, and epigenetic pathway modulation. Understanding these mechanisms provides new insights into potential therapeutic strategies for Mn-induced disorders. This review also highlights future research directions, emphasizing the need for developing targeted therapies and investigating combination approaches to address multiple aspects of Mn toxicity simultaneously.
Collapse
Affiliation(s)
- Airton C Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Gustavo H Oliveira-Paula
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Alexey A Tinkov
- Institute of Bioelementology, Orenburg State University, Orenburg, 460000, Russia; IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003, Russia
| | - Anatoly V Skalny
- Institute of Bioelementology, Orenburg State University, Orenburg, 460000, Russia; IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003, Russia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington DC, 20059, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
2
|
Fontes A, Jauch AT, Sailer J, Engler J, Azul AM, Zischka H. Metabolic Derangement of Essential Transition Metals and Potential Antioxidant Therapies. Int J Mol Sci 2024; 25:7880. [PMID: 39063122 PMCID: PMC11277342 DOI: 10.3390/ijms25147880] [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: 05/19/2024] [Revised: 07/08/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Essential transition metals have key roles in oxygen transport, neurotransmitter synthesis, nucleic acid repair, cellular structure maintenance and stability, oxidative phosphorylation, and metabolism. The balance between metal deficiency and excess is typically ensured by several extracellular and intracellular mechanisms involved in uptake, distribution, and excretion. However, provoked by either intrinsic or extrinsic factors, excess iron, zinc, copper, or manganese can lead to cellular damage upon chronic or acute exposure, frequently attributed to oxidative stress. Intracellularly, mitochondria are the organelles that require the tightest control concerning reactive oxygen species production, which inevitably leaves them to be one of the most vulnerable targets of metal toxicity. Current therapies to counteract metal overload are focused on chelators, which often cause secondary effects decreasing patients' quality of life. New therapeutic options based on synthetic or natural antioxidants have proven positive effects against metal intoxication. In this review, we briefly address the cellular metabolism of transition metals, consequences of their overload, and current therapies, followed by their potential role in inducing oxidative stress and remedies thereof.
Collapse
Affiliation(s)
- Adriana Fontes
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, D-85764 Neuherberg, Germany;
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Adrian T. Jauch
- School of Medicine and Health, Institute of Toxicology and Environmental Hygiene, Technical University Munich, D-80802 Munich, Germany
| | - Judith Sailer
- School of Medicine and Health, Institute of Toxicology and Environmental Hygiene, Technical University Munich, D-80802 Munich, Germany
| | - Jonas Engler
- School of Medicine and Health, Institute of Toxicology and Environmental Hygiene, Technical University Munich, D-80802 Munich, Germany
| | - Anabela Marisa Azul
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Hans Zischka
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, D-85764 Neuherberg, Germany;
- School of Medicine and Health, Institute of Toxicology and Environmental Hygiene, Technical University Munich, D-80802 Munich, Germany
| |
Collapse
|
3
|
Haberland VMM, Magin S, Iliakis G, Hartwig A. Impact of Manganese and Chromate on Specific DNA Double-Strand Break Repair Pathways. Int J Mol Sci 2023; 24:10392. [PMID: 37373538 DOI: 10.3390/ijms241210392] [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: 04/25/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Manganese is an essential trace element; nevertheless, on conditions of overload, it becomes toxic, with neurotoxicity being the main concern. Chromate is a well-known human carcinogen. The underlying mechanisms seem to be oxidative stress as well as direct DNA damage in the case of chromate, but also interactions with DNA repair systems in both cases. However, the impact of manganese and chromate on DNA double-strand break (DSB) repair pathways is largely unknown. In the present study, we examined the induction of DSB as well as the effect on specific DNA DSB repair mechanisms, namely homologous recombination (HR), non-homologous end joining (NHEJ), single strand annealing (SSA), and microhomology-mediated end joining (MMEJ). We applied DSB repair pathway-specific reporter cell lines, pulsed field gel electrophoresis as well as gene expression analysis, and investigated the binding of specific DNA repair proteins via immunoflourescence. While manganese did not seem to induce DNA DSB and had no impact on NHEJ and MMEJ, HR and SSA were inhibited. In the case of chromate, the induction of DSB was further supported. Regarding DSB repair, no inhibition was seen in the case of NHEJ and SSA, but HR was diminished and MMEJ was activated in a pronounced manner. The results indicate a specific inhibition of error-free HR by manganese and chromate, with a shift towards error-prone DSB repair mechanisms in both cases. These observations suggest the induction of genomic instability and may explain the microsatellite instability involved in chromate-induced carcinogenicity.
Collapse
Affiliation(s)
- Vivien M M Haberland
- Department of Food Chemistry and Toxicology, Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), 76131 Karlsruhe, Germany
| | - Simon Magin
- Institute of Medical Radiation Biology, Medical School, University of Duisburg-Essen, 45122 Essen, Germany
| | - George Iliakis
- Institute of Medical Radiation Biology, Medical School, University of Duisburg-Essen, 45122 Essen, Germany
| | - Andrea Hartwig
- Department of Food Chemistry and Toxicology, Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), 76131 Karlsruhe, Germany
| |
Collapse
|
4
|
Neuron Protection by EDTA May Explain the Successful Outcomes of Toxic Metal Chelation Therapy in Neurodegenerative Diseases. Biomedicines 2022; 10:biomedicines10102476. [PMID: 36289738 PMCID: PMC9599085 DOI: 10.3390/biomedicines10102476] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 11/20/2022] Open
Abstract
Many mechanisms have been related to the etiopathogenesis of neurodegenerative diseases (NDs) such as multiple sclerosis, amyotrophic lateral sclerosis, Parkinson’s disease, and Alzheimer’s disease. In this context, the detrimental role of environmental agents has also been highlighted. Studies focused on the role of toxic metals in the pathogenesis of ND demonstrate the efficacy of treatment with the chelating agent calcium disodium ethylenediaminetetraacetic acid (EDTA) in eliminating toxic metal burden in all ND patients, improving their symptoms. Lead, cadmium, aluminum, nickel, and mercury were the most important toxic metals detected in these patients. Here, I provide an updated review on the damage to neurons promoted by toxic metals and on the impact of EDTA chelation therapy in ND patients, along with the clinical description of a representative case.
Collapse
|
5
|
Werner E, Gokhale A, Ackert M, Xu C, Wen Z, Roberts AM, Roberts BR, Vrailas-Mortimer A, Crocker A, Faundez V. The mitochondrial RNA granule modulates manganese-dependent cell toxicity. Mol Biol Cell 2022; 33:ar108. [PMID: 35921164 PMCID: PMC9635304 DOI: 10.1091/mbc.e22-03-0096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 11/11/2022] Open
Abstract
Prolonged manganese exposure causes manganism, a neurodegenerative movement disorder. The identity of adaptive and nonadaptive cellular processes targeted by manganese remains mostly unexplored. Here we study mechanisms engaged by manganese in genetic cellular models known to increase susceptibility to manganese exposure, the plasma membrane manganese efflux transporter SLC30A10 and the mitochondrial Parkinson's gene PARK2. We found that SLC30A10 and PARK2 mutations as well as manganese exposure compromised the mitochondrial RNA granule composition and function, resulting in disruption of mitochondrial transcript processing. These RNA granule defects led to impaired assembly and function of the mitochondrial respiratory chain. Notably, cells that survived a cytotoxic manganese challenge had impaired RNA granule function, thus suggesting that this granule phenotype was adaptive. CRISPR gene editing of subunits of the mitochondrial RNA granule, FASTKD2 or DHX30, as well as pharmacological inhibition of mitochondrial transcription-translation, were protective rather than deleterious for survival of cells acutely exposed to manganese. Similarly, adult Drosophila mutants with defects in the mitochondrial RNA granule component scully were safeguarded from manganese-induced mortality. We conclude that impairment of the mitochondrial RNA granule function is a protective mechanism for acute manganese toxicity.
Collapse
Affiliation(s)
- E. Werner
- Department of Cell Biology, Emory University, Atlanta, GA 30322
| | - A. Gokhale
- Department of Cell Biology, Emory University, Atlanta, GA 30322
| | - M. Ackert
- School of Biological Sciences, Illinois State University, Normal, IL 617901
| | - C. Xu
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322
| | - Z. Wen
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322
| | - A. M. Roberts
- Department of Biochemistry, Emory University, Atlanta, GA 30322
| | - B. R. Roberts
- Department of Biochemistry, Emory University, Atlanta, GA 30322
| | | | - A. Crocker
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753
| | - V. Faundez
- Department of Cell Biology, Emory University, Atlanta, GA 30322
| |
Collapse
|
6
|
Abu-Elfotuh K, Hamdan AME, Abbas AN, Alahmre ATS, Elewa MAF, Masoud RAE, Ali AA, Othman M, Kamal MM, Hassan FAM, Khalil MG, El-Sisi AM, Abdel Hady MMM, Abd-Elhaleim El Azazy MK, Awny MM, Wahid A. Evaluating the neuroprotective activities of vinpocetine, punicalagin, niacin and vitamin E against behavioural and motor disabilities of manganese-induced Parkinson's disease in Sprague Dawley rats. Biomed Pharmacother 2022; 153:113330. [PMID: 35780621 DOI: 10.1016/j.biopha.2022.113330] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 01/22/2023] Open
Abstract
The current study investigated the neuroprotective activity of some drugs and nutriceuticals with antioxidant and anti-inflammatory potential on the pathogenesis of Parkinson's disease (PD). Rats were categorized into seven groups: Rats received tween80 daily for 5 weeks as a control group, MnCl2 (10 mg/kg, i.p) either alone (group II) or in combination with vinpocetine (VIN) (20 mg/kg) (group III), punicalagin (PUN) (30 mg/kg) (group IV), niacin (85 mg/kg) (group V), vitamin E (Vit E) (100 mg/kg) (group VI) or their combination (group VII). Motor activities was examined using open-field and catalepsy. Striatal monamines, acetylcholinesterase, excitatory/inhibitory neurotransmitters, redox status, pro-oxidant content, brain inflammatory, apoptotic and antioxidant biomarkers levels were assessed. Besides, histopathological investigations of different brain regions were determined. Groups (IV -GVII) showed improved motor functions of PD rats. Applied drugs significantly increased the brain levels of monoamines with the strongest effect to PUN. Meanwhile, they significantly decreased levels of acetylcholinesterase with a strongest effect to PUN. Moreover, they exhibited significant neuronal protection and anti-inflammatory abilities through significant reduction of the brain levels of COX2, TNF-α and Il-1β with a strongest effect to the PUN. Interestingly; groups (IV - GVII) showed restored glutamate/GABA balance and exhibited a pronounced decrease in caspase-3 content and GSK-3β protein expression levels. In addition, they significantly increased Bcl2 mRNA expression levels with a strongest effect for PUN. All these findings were further confirmed by the histopathological examinations. As a conclusion, we propose VIN and PUN to mitigate the progression of PD via their antioxidant, anti-inflammatory, anti-apoptotic, neurotrophic and neurogenic activities.
Collapse
Affiliation(s)
- Karema Abu-Elfotuh
- Pharmacology and Toxicology Department (Girls), Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | | | | | | | - Mohammed A F Elewa
- Biochemistry Department, Faculty of Pharmacy, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Rehab Ali Elsayed Masoud
- Forensic Medicine and Clinical Toxicology Department, Faculty of medicine for girls, Al-Azhar University, Cairo, Egypt
| | - Azza A Ali
- Pharmacology and Toxicology Department (Girls), Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mohamed Othman
- Lecturer, Department of anatomy, Faculty of Medicine, King Salman International University, El-Tur Campus, Saini, Egypt
| | - Mona M Kamal
- Pharmacology and Toxicology Department (Girls), Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Fatma Alzahraa M Hassan
- Biochemistry and molecular biology Department, Faculty of Pharmacy, Al-Azhar, University, Cairo, Egypt
| | - Mona G Khalil
- Pharmacology and Toxicology Department, Modern University for Technology and Information, Cairo, Egypt
| | - Ahmed M El-Sisi
- Biochemistry and Molecular Biology Department (boys), Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Nahda University (NUB), Beni-Suef, Egypt
| | - Manal M M Abdel Hady
- Department of Pharmacology, Faculty of Pharmacy, Qantra University, Sinai, Egypt
| | | | - Magdy M Awny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Cairo, Egypt
| | - Ahmed Wahid
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
7
|
Xue J, Li G, Ji X, Liu ZH, Wang HL, Xiao G. Drosophila ZIP13 overexpression or transferrin1 RNAi influences the muscle degeneration of Pink1 RNAi by elevating iron levels in mitochondria. J Neurochem 2022; 160:540-555. [PMID: 35038358 DOI: 10.1111/jnc.15574] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 12/01/2022]
Abstract
Disruption of iron homeostasis in the brain of Parkinson's disease (PD) patients has been reported for many years, but the underlying mechanisms remain unclear. To investigate iron metabolism genes related to PTEN-induced kinase 1 (Pink1) and parkin (E3 ubiquitin ligase), two PD-associated proteins that function to coordinate mitochondrial turnover via induction of selective mitophagy, we conducted a genetic screen in Drosophila and found that altered expression of genes involved in iron metabolism, such as Drosophila ZIP13 (dZIP13) or transferrin1 (Tsf1), significantly influences the disease progression related to Pink1 but not parkin. Several phenotypes of Pink1 mutant and Pink1 RNAi but not parkin mutant were significantly rescued by overexpression (OE) of dZIP13 (dZIP13 OE) or silencing of Tsf1 (Tsf1 RNAi) in the flight muscles. The rescue effects of dZIP13 OE or Tsf1 RNAi were not exerted through mitochondrial disruption or mitophagy, instead, the iron levels in mitochondira were significantly increased, resulting in enhanced activity of enzymes participating in respiration and increased ATP synthesis. Consistently, the rescue effects of dZIP13 OE or Tsf1 RNAi on Pink1 RNAi can be inhibited by decreasing the iron levels in mitochondria through mitoferrin (dmfrn) RNAi. This study suggests that dZIP13, Tsf1 and dmfrn might act independently of parkin in a parallel pathway downstream of Pink1 by modulating respiration and indicates that manipulation of iron levels in mitochondria may provide a novel therapeutic strategy for PD associated with Pink1.
Collapse
Affiliation(s)
- Jinsong Xue
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Guangying Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Xiaowen Ji
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Zhi-Hua Liu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Hui-Li Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Guiran Xiao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| |
Collapse
|
8
|
Critical Involvement of Glial Cells in Manganese Neurotoxicity. BIOMED RESEARCH INTERNATIONAL 2021; 2021:1596185. [PMID: 34660781 PMCID: PMC8514895 DOI: 10.1155/2021/1596185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022]
Abstract
Over the years, most of the research concerning manganese exposure was restricted to the toxicity of neuronal cells. Manganese is an essential trace element that in high doses exerts neurotoxic effects. However, in the last two decades, efforts have shifted toward a more comprehensive approach that takes into account the involvement of glial cells in the development of neurotoxicity as a brain insult. Glial cells provide structural, trophic, and metabolic support to neurons. Nevertheless, these cells play an active role in adult neurogenesis, regulation of synaptogenesis, and synaptic plasticity. Disturbances in glial cell function can lead to neurological disorders, including neurodegenerative diseases. This review highlights the pivotal role that glial cells have in manganese-induced neurotoxicity as well as the most sounding mechanisms involved in the development of this phenomenon.
Collapse
|
9
|
Mitochondrial damage produced by phytotoxic chromenone and chromanone derivatives from endophytic fungus Daldinia eschscholtzii strain GsE13. Appl Microbiol Biotechnol 2021; 105:4225-4239. [PMID: 33970316 DOI: 10.1007/s00253-021-11318-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/16/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022]
Abstract
Bioassay-guided fractionation of the organic extracts of the endophyte Daldinia eschscholtzii strain GsE13 led to the isolation of several phytotoxic compounds, including two chromenone and two chromanone derivatives: 5-hydroxy-8-methoxy-2-methyl-4H-chromen-4-one, 1; 5-hydroxy-2-methyl-4H-chromen-4-one, 2; 5-methoxy-2-methyl-chroman-4-one, 3; and 5-methoxy-2-methyl-chroman-4-ol, 4; as well as other aromatic compounds: 4,8-dihydroxy-1-tetralone, 5; 1,8-dimethoxynaphthalene, 6; and 4,9-dihydroxy-1,2,11,12-tetrahydroperyl-ene-3,10-quinone, 7. Compounds 1, 4, and 7 were isolated for the first time from D. eschscholtzii. The phytotoxicity of all the compounds was determined on germination, root growth, and oxygen uptake in seedlings of a monocotyledonous (Panicum miliaceum) and three dicotyledonous plants (Medicago sativa, Trifolium pratense, and Amaranthus hypochondriacus). In general, root growth was the most affected process in all four weeds, and chromenones 1 and 2 were the most phytotoxic compounds. Phytotoxins 1-4 inhibited basal oxygen consumption rate in isolated mitochondria from M. sativa seedlings and also caused serious damage to their membrane potential (ΔΨm) in percentages greater than 50% at concentrations lower than 2 mM. Based on these results, compounds 1-4 of endophytic origin could be promising for the development of new herbicides potentially useful in agriculture or for the synthesis of promising new molecules. KEY POINTS: • Endophytic fungus Daldinia eschscholtzii produces phytotoxic compounds. • Phytotoxins inhibit basal oxygen consumption rate in isolated M. sativa mitochondria. • Phytotoxins altered the mitochondrial membrane potential.
Collapse
|
10
|
Miah MR, Ijomone OM, Okoh COA, Ijomone OK, Akingbade GT, Ke T, Krum B, da Cunha Martins A, Akinyemi A, Aranoff N, Antunes Soares FA, Bowman AB, Aschner M. The effects of manganese overexposure on brain health. Neurochem Int 2020; 135:104688. [PMID: 31972215 PMCID: PMC7926190 DOI: 10.1016/j.neuint.2020.104688] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 01/12/2020] [Accepted: 01/15/2020] [Indexed: 12/11/2022]
Abstract
Manganese (Mn) is the twelfth most abundant element on the earth and an essential metal to human health. Mn is present at low concentrations in a variety of dietary sources, which provides adequate Mn content to sustain support various physiological processes in the human body. However, with the rise of Mn utility in a variety of industries, there is an increased risk of overexposure to this transition metal, which can have neurotoxic consequences. This risk includes occupational exposure of Mn to workers as well as overall increased Mn pollution affecting the general public. Here, we review exposure due to air pollution and inhalation in industrial settings; we also delve into the toxic effects of manganese on the brain such as oxidative stress, inflammatory response and transporter dysregulation. Additionally, we summarize current understandings underlying the mechanisms of Mn toxicity.
Collapse
Affiliation(s)
- Mahfuzur R Miah
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Omamuyovwi M Ijomone
- The Neuro-Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology Akure, Ondo, Nigeria
| | - Comfort O A Okoh
- The Neuro-Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology Akure, Ondo, Nigeria
| | - Olayemi K Ijomone
- The Neuro-Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology Akure, Ondo, Nigeria; Department of Anatomy, University of Medical Sciences, Ondo, Nigeria
| | - Grace T Akingbade
- The Neuro-Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology Akure, Ondo, Nigeria
| | - Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bárbara Krum
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | | | - Ayodele Akinyemi
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nicole Aranoff
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Stern College for Women, Yeshiva University, New York, NY, USA
| | - Felix Alexandre Antunes Soares
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
11
|
Li H, Liu X, Wassie M, Chen L. Selenium supplementation alleviates cadmium-induced damages in tall fescue through modulating antioxidant system, photosynthesis efficiency, and gene expression. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:9490-9502. [PMID: 31919821 DOI: 10.1007/s11356-019-06628-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 09/25/2019] [Indexed: 06/10/2023]
Abstract
Selenium (Se) is beneficial for plant growth under different stressful conditions. In this study, we investigated the protective effects of Se supply from Cd-induced damages in tall fescue under Cd stress. Tall fescue seedlings (40 days old) were treated with Cd (30 mg/L, as CdSO4·8/3 H2O) and Se (0.1 mg/L, as Na2SeO3) individually and in combination using 1/2 Hoagland's solution system for 7 days. Various physiological parameters, photosynthetic behaviors, and gene expressions were measured. The results showed that Cd-stressed plants displayed obvious toxicity symptoms such as leaf yellowing, decreasing plant height, and root length. Cd stress significantly increased the malondialdehyde (MDA) content and electrolyte leakage (EL), and remarkably reduced the chlorophyll and soluble protein content, antioxidant enzyme activities, and photosynthetic efficiency. Cd stress significantly inhibited the expression of two photosynthesis-related genes (psbB and psbC), but not psbA. In addition, it significantly inhibited the expression of antioxidant system-related genes such as ChlCu/ZnSOD, CytCu/ZnSOD, GPX, and pAPX, but significantly increased the expression of GR. However, Se improved the overall physiological and photosynthetic behaviors of Cd-stressed plants. Se significantly enhanced the chlorophyll and soluble protein content and CAT and SOD activities, but decreased MDA contents, EL, and Cd content and translocation in tall fescue under Cd stress. Furthermore, under Cd stress, Se increased the expression of psbA, psbB psbC, ChlCu/ZnSOD, CytCu/ZnSOD, GPx, and PAPx. The result suggests that Se alleviated the deleterious effects of Cd and improved Cd resistance in tall fescue through upregulating the antioxidant system, photosynthesis activities, and gene expressions.
Collapse
Affiliation(s)
- Huiying Li
- CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden,The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430074, China
| | - Xiaofei Liu
- Department of Pratacultural Sciences, College of Agronomy, Hunan Agricultural University, Changsha, 410128, China
| | - Misganaw Wassie
- CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden,The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430074, China
- University of Chinese Academy of Sciences China, Chinese Academy of Sciences, Beijing, 100049, China
| | - Liang Chen
- CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden,The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430074, China.
| |
Collapse
|
12
|
L P, D K, J K, Cl H, N L. Integrated approach for data acquisition, visualization and processing of analog polarographic systems for bioenergetics studies. Anal Biochem 2020; 590:113515. [PMID: 31812534 PMCID: PMC6943940 DOI: 10.1016/j.ab.2019.113515] [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: 10/29/2019] [Accepted: 11/25/2019] [Indexed: 11/20/2022]
Abstract
Bioenergetic function is characterized with assays obtained by polarographic systems. Analog systems without data acquisition, visualization, and processing tools are used but require cumbersome operations to derive respiration rate and ADP to oxygen stoichiometry of oxidative phosphorylation (ADP/O ratio). The analog signal of a polarograhic system (YSI-5300) was digitized and a graphical user interface (GUI) was developed in MATLAB to integrate visualization, recording, calibration and processing of bioenergetic data. With the GUI, the signal is continuously visualized during the experiment and respiratory rates and ADP/O ratios can be determined. The integrated system was tested to evaluate bioenergetic function of subpopulations of mitochondria isolated from rat skeletal muscle (n = 10). Signal processing was applied to denoise data recorded at the sampling rate of 1000Hz, and maximize data decimation for computational applications. The error in calculating the bioenergetic outputs using decimated data is negligible when data are denoised. The estimate of respiration rate, ADP/O ratio and RCR obtained with denoised data at sampling rate as low as 5Hz was similar to that obtained with raw data recorded at sampling rate of 1000Hz. In summary, the integrated tools of the GUI overcome the limitations of data processing, accuracy, and utilization of analog polarographic systems.
Collapse
Affiliation(s)
- Potter L
- Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, USA; Biomedical Engineering Institute, Old Dominion University, Norfolk, VA, USA
| | - Krusienski D
- Department of Biomedical Engineering Virginia Commonwealth University, Richmond, VA, USA
| | - Kennedy J
- Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, USA
| | - Hoppel Cl
- Center for Mitochondrial Disease, School of Medicine, Case Western Reserve University, USA; Department of Pharmacology and School of Medicine, Case Western Reserve University, USA; Department of Medicine, School of Medicine, Case Western Reserve University, USA
| | - Lai N
- Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, USA; Biomedical Engineering Institute, Old Dominion University, Norfolk, VA, USA; Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, USA; Department of Mechanical, Chemical and Materials Engineering, University of Cagliari, Italy.
| |
Collapse
|
13
|
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
|
14
|
Okoye CN, MacDonald-Jay N, Kamunde C. Effects of bioenergetics, temperature and cadmium on liver mitochondria reactive oxygen species production and consumption. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 214:105264. [PMID: 31377504 DOI: 10.1016/j.aquatox.2019.105264] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/22/2019] [Accepted: 07/24/2019] [Indexed: 06/10/2023]
Abstract
A by-product of mitochondrial substrate oxidation and electron transfer to generate cellular energy (ATP) is reactive oxygen species (ROS). Superoxide anion radical and hydrogen peroxide (H2O2) are the proximal ROS produced by the mitochondria. Because low levels of ROS serve critical regulatory roles in cell physiology while excessive levels or inappropriately localized ROS result in aberrant physiological states, mitochondrial ROS need to be tightly regulated. While it is known that regulation of mitochondrial ROS involves balancing the rates of production and removal, the effects of stressors on these processes remain largely unknown. To illuminate how stressors modulate mitochondrial ROS homeostasis, we investigated the effects of temperature and cadmium (Cd) on H2O2 emission and consumption in rainbow trout liver mitochondria. We show that H2O2 emission rates increase with temperature and Cd exposure. Energizing mitochondria with malate-glutamate or succinate increased the rate of H2O2 emission; however, Cd exposure imposed different patterns of H2O2 emission depending on the concentration and substrate. Specifically, mitochondria respiring on malate-glutamate exhibited a saturable graded concentration-response curve that plateaued at 5 μM while mitochondria respiring on succinate had a biphasic concentration-response curve characterized by a spike in the emission rate at 1 μM Cd followed by gradual diminution at higher Cd concentrations. To explain the observed substrate- and concentration-dependent effects of Cd, we sequestered specific mitochondrial ROS-emitting sites using blockers of electron transfer and then tested the effect of the metal. The results indicate that the biphasic H2O2 emission response imposed by succinate is due to site IIF but is further modified at sites IQ and IIIQo. Moreover, the saturable graded H2O2 emission response in mitochondria energized with malate-glutamate is consistent with effect of Cd on site IF. Additionally, Cd and temperature acted cooperatively to increase mitochondrial H2O2 emission suggesting that increased toxicity of Cd at high temperature may be due to increased oxidative insult. Surprisingly, despite their clear stimulatory effect on H2O2 emission, Cd, temperature and bioenergetic status did not affect the kinetics of mitochondrial H2O2 consumption; the rate constants and half-lives for all the conditions tested were similar. Overall, our study indicates that the production processes of rainbow trout liver mitochondrial H2O2 metabolism are highly responsive to stressors and bioenergetics while the consumption processes are recalcitrant. The latter denotes the presence of a robust H2O2 scavenging system in liver mitochondria that would maintain H2O2 homeostasis in the face of increased production and reduced scavenging capacity.
Collapse
Affiliation(s)
- Chidozie N Okoye
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE, C1A 4P3, Canada
| | - Nicole MacDonald-Jay
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE, C1A 4P3, Canada
| | - Collins Kamunde
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE, C1A 4P3, Canada.
| |
Collapse
|
15
|
Wan Y, Wang K, Liu Z, Yu Y, Wang Q, Li H. Effect of selenium on the subcellular distribution of cadmium and oxidative stress induced by cadmium in rice (Oryza sativa L.). ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:16220-16228. [PMID: 30972675 DOI: 10.1007/s11356-019-04975-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 03/25/2019] [Indexed: 05/12/2023]
Abstract
Cadmium (Cd) is absorbed readily by rice plants and is transferred to humans when contaminated rice is consumed. Adding selenium (Se) to the plant nutrient solutions reduces the accumulation of Cd in the rice (Oryza sativa L.) seedlings. However, as the relevant underlying mechanism remains unclear, the aim of our study was to improve our understanding of the Se-mediated resistance to Cd stress in rice. We conducted hydroponic experiments to study the effects of selenite or selenate on Cd subcellular distribution and xylem transport in rice seedlings under Cd stress, and we investigated the antioxidative defense responses in the rice plants. We found that the supplementation of both Se forms decreased the Cd accumulations in the roots and shoots of the rice plants. The selenite addition significantly decreased the Cd contents in different subcellular fractions of the rice roots, increased the proportion of Cd distributed to soluble cytosol by 23.41%, and decreased the Cd distribution in the organelle by 28.74% in contrast with the treatment with Cd only. As regards the selenate addition, only the Cd distribution ratio of cytosol was increased by 13.07%. After adding selenite, a decrease of 55.86% in the Cd concentration in xylem sap was observed, whereas little change was found after treatment co-applied with selenate. The hydrogen peroxide (H2O2) and malondialdehyde(MDA) contents in the rice roots were elevated under Cd stress, and the addition of selenite and selenate decreased the H2O2 levels by 77.78% and 59.26%, respectively. Co-exposure to Cd and Se elevated the glutathione (GSH) accumulations in the rice shoots and roots, with the degree of increase being the following: co-applied with selenite > co-applied with selenate > Cd alone treatment. Exposure to Cd increased the catalase (CAT) activity in the roots significantly, whereas it decreased in the shoots. After selenite or selenate supplementation, the CAT activity in the rice roots increased compared with applying only Cd. Compared with the control, the addition of Cd or Se had no significant effect on the activities of peroxidase (POD) or ascorbate peroxidase (APX). Our results showed that Se affected the Cd accumulation in rice seedlings by altering the Cd subcellular distribution and decreasing the ROS induced by Cd stress. Such effects were more significant in the selenite than in the selenate applied treatment.
Collapse
Affiliation(s)
- Yanan Wan
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, Key Laboratory of Plant-Soil Interactions of the Ministry of Education, College of Resources and Environmental Sciences, China Agricultural University, Beijing, 100193, People's Republic of China
- Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
| | - Kang Wang
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, Key Laboratory of Plant-Soil Interactions of the Ministry of Education, College of Resources and Environmental Sciences, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Zhe Liu
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, Key Laboratory of Plant-Soil Interactions of the Ministry of Education, College of Resources and Environmental Sciences, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Yao Yu
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, Key Laboratory of Plant-Soil Interactions of the Ministry of Education, College of Resources and Environmental Sciences, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Qi Wang
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, Key Laboratory of Plant-Soil Interactions of the Ministry of Education, College of Resources and Environmental Sciences, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Huafen Li
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, Key Laboratory of Plant-Soil Interactions of the Ministry of Education, College of Resources and Environmental Sciences, China Agricultural University, Beijing, 100193, People's Republic of China.
| |
Collapse
|
16
|
Kanthasamy A, Jin H, Charli A, Vellareddy A, Kanthasamy A. Environmental neurotoxicant-induced dopaminergic neurodegeneration: a potential link to impaired neuroinflammatory mechanisms. Pharmacol Ther 2019; 197:61-82. [PMID: 30677475 PMCID: PMC6520143 DOI: 10.1016/j.pharmthera.2019.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
With the increased incidence of neurodegenerative diseases worldwide, Parkinson's disease (PD) represents the second-most common neurodegenerative disease. PD is a progressive multisystem neurodegenerative disorder characterized by a marked loss of nigrostriatal dopaminergic neurons and the formation of Lewy pathology in diverse brain regions. Although the mechanisms underlying dopaminergic neurodegeneration remain poorly characterized, data from animal models and postmortem studies have revealed that heightened inflammatory responses mediated via microglial and astroglial activation and the resultant release of proinflammatory factors may act as silent drivers of neurodegeneration. In recent years, numerous studies have demonstrated a positive association between the exposure to environmental neurotoxicants and the etiology of PD. Although it is unclear whether neuroinflammation drives pesticide-induced neurodegeneration, emerging evidence suggests that the failure to dampen neuroinflammatory mechanisms may account for the increased vulnerability to pesticide neurotoxicity. Furthermore, recent studies provide additional evidence that shifts the focus from a neuron-centric view to glial-associated neurodegeneration following pesticide exposure. In this review, we propose to summarize briefly the possible factors that regulate neuroinflammatory processes during environmental neurotoxicant exposure with a focus on the potential roles of mitochondria-driven redox mechanisms. In this context, a critical discussion of the data obtained from experimental research and possible epidemiological studies is included. Finally, we hope to provide insights on the pivotal role of exosome-mediated intercellular transmission of aggregated proteins in microglial activation response and the resultant dopaminergic neurodegeneration after exposure to pesticides. Collectively, an improved understanding of glia-mediated neuroinflammatory signaling might provide novel insights into the mechanisms that contribute to neurodegeneration induced by environmental neurotoxicant exposure.
Collapse
Affiliation(s)
- Arthi Kanthasamy
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA.
| | - Huajun Jin
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Adhithiya Charli
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Anantharam Vellareddy
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Anumantha Kanthasamy
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
17
|
Fernandes J, Chandler JD, Liu KH, Uppal K, Hao L, Hu X, Go YM, Jones DP. Metabolomic Responses to Manganese Dose in SH-SY5Y Human Neuroblastoma Cells. Toxicol Sci 2019; 169:84-94. [PMID: 30715528 PMCID: PMC6484887 DOI: 10.1093/toxsci/kfz028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Manganese (Mn)-associated neurotoxicity has been well recognized. However, Mn is also an essential nutrient to maintain physiological function. Our previous study of human neuroblastoma SH-SY5Y cells showed that Mn treatment comparable to physiological and toxicological concentrations in human brain resulted in different mitochondrial responses, yet cellular metabolic responses associated with such different outcomes remain uncharacterized. Herein, SH-SY5Y cells were examined for metabolic responses discriminated by physiological and toxicological levels of Mn using high-resolution metabolomics (HRM). Before performing HRM, we examined Mn dose (from 0 to100 μM) and time effects on cell death. Although we did not observe any immediate cell death after 5 h exposure to any of the Mn concentrations assessed (0-100 μM), cell loss was present after a 24-h recovery period in cultures treated with Mn ≥ 50 μM. Exposure to Mn for 5 h resulted in a wide range of changes in cellular metabolism including amino acids (AA), neurotransmitters, energy, and fatty acids metabolism. Adaptive responses at 10 μM showed increases in neuroprotective AA metabolites (creatine, phosphocreatine, phosphoserine). A 5-h exposure to 100 µM Mn, a time before any cell death occurred, resulted in decreases in energy and fatty acid metabolites (hexose-1,6 bisphosphate, acyl carnitines). The results show that adjustments in AA metabolism occur in response to Mn that does not cause cell death while disruption in energy and fatty acid metabolism occur in response to Mn that results in subsequent cell death. The present study establishes utility for metabolomics analyses to discriminate adaptive and toxic molecular responses in a human in vitro cellular model that could be exploited in evaluation of Mn toxicity.
Collapse
Affiliation(s)
- Jolyn Fernandes
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Joshua D Chandler
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Ken H Liu
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Karan Uppal
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Li Hao
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Xin Hu
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Young-Mi Go
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
18
|
Role of NADPH oxidase in cooperative reactive oxygen species generation in dopaminergic neurons induced by combined treatment with dieldrin and lindane. Toxicol Lett 2018; 299:47-55. [DOI: 10.1016/j.toxlet.2018.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/31/2018] [Accepted: 09/16/2018] [Indexed: 11/19/2022]
|
19
|
Yu Y, Yuan S, Zhuang J, Wan Y, Wang Q, Zhang J, Li H. Effect of selenium on the uptake kinetics and accumulation of and oxidative stress induced by cadmium in Brassica chinensis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 162:571-580. [PMID: 30031318 DOI: 10.1016/j.ecoenv.2018.07.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/15/2018] [Accepted: 07/10/2018] [Indexed: 06/08/2023]
Abstract
Pak choi can readily accumulate cadmium (Cd) into its edible parts; this can pose a threat to human health. Although not essential for higher plants, selenium (Se) can be favorable for plant growth and antioxidative defense under heavy metal stress conditions. A pak choi hydroponic experiment was conducted to investigate the effect of two forms of Se on the Cd uptake kinetics and accumulation and oxidative stress. The results showed that selenite and selenate remarkably enhanced Cd uptake kinetics in pak choi. The maximum Cd uptake rate increased by more than 100% after treatment with 5 µM of selenite and selenate, and it further increased after treatment with 20 µM of both Se forms. The effects of Se on Cd content depended on the Se form, exposure time, and Cd dosage. Selenite reduced the Cd content in shoots by 41% after 3 days of treatment with 10 µM Cd, whereas selenate increased this rate by 89%. Both forms of Se decreased Cd content in the shoots by 40% after 7 days of treatment with 10 µM Cd, but they increased the Cd content by approximately 30% after treatment with 50 µM Cd. Se enhanced Cd-induced oxidative stress in pak choi. Malondialdehyde (MDA) generation was promoted by more than 33% by selenite and selenate treatments in combination with 10 µM Cd, and it was further enhanced by 106% and 185% at 50 µM Cd, respectively. Selenite also increased the H2O2 content at both Cd doses, but selenate did not have significant effects on H2O2 production. The effects of Se on antioxidative enzyme activity also depended on the dose of Cd. Selenite and selenate inhibited catalase activity by 11% and 29%, respectively, at 10 µM Cd, and by 13% and 42%, respectively, at 50 µM Cd. Moreover, both forms of Se increased superoxide dismutase activity after treatment with 10 µM Cd but inhibited its activity at 50 µM Cd. Therefore, Se exhibits dual effects on Cd accumulation and oxidative stress in pak choi and might cause further stress when combined with higher doses of Cd.
Collapse
Affiliation(s)
- Yao Yu
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, Key Laboratory of Plant-Soil Interactions of the Ministry of Education, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, PR China
| | - Sili Yuan
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, Key Laboratory of Plant-Soil Interactions of the Ministry of Education, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, PR China
| | - Jian Zhuang
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, Key Laboratory of Plant-Soil Interactions of the Ministry of Education, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, PR China
| | - Yanan Wan
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, Key Laboratory of Plant-Soil Interactions of the Ministry of Education, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, PR China
| | - Qi Wang
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, Key Laboratory of Plant-Soil Interactions of the Ministry of Education, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, PR China
| | - Jingsuo Zhang
- Beijing Municipal Station of Agro-environmental Monitoring, Beijing 100029, PR China
| | - Huafen Li
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, Key Laboratory of Plant-Soil Interactions of the Ministry of Education, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, PR China.
| |
Collapse
|
20
|
Porte Alcon S, Gorojod RM, Kotler ML. Regulated Necrosis Orchestrates Microglial Cell Death in Manganese-Induced Toxicity. Neuroscience 2018; 393:206-225. [PMID: 30316909 DOI: 10.1016/j.neuroscience.2018.10.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/01/2018] [Accepted: 10/04/2018] [Indexed: 12/17/2022]
Abstract
Microglia, the brain resident immune cells, play prominent roles in immune surveillance, tissue repair and neural regeneration. Despite these pro-survival actions, the relevance of these cells in the progression of several neuropathologies has been established. In the context of manganese (Mn) overexposure, it has been proposed that microglial activation contributes to enhance the neurotoxicity. However, the occurrence of a direct cytotoxic effect of Mn on microglial cells remains controversial. In the present work, we investigated the potential vulnerability of immortalized mouse microglial cells (BV-2) toward Mn2+, focusing on the signaling pathways involved in cell death. Evidence obtained showed that Mn2+ induces a decrease in cell viability which is associated with reactive oxygen species (ROS) generation. In this report we demonstrated, for the first time, that Mn2+ triggers regulated necrosis (RN) in BV-2 cells involving two central mechanisms: parthanatos and lysosomal disruption. The occurrence of parthanatos is supported by several cellular and molecular events: (i) DNA damage; (ii) AIF translocation from mitochondria to the nucleus; (iii) mitochondrial membrane permeabilization; and (iv) PARP1-dependent cell death. On the other hand, Mn2+ induces lysosomal membrane permeabilization (LMP) and cathepsin D (CatD) release into the cytosol supporting the lysosomal disruption. Pre-incubation with CatB and D inhibitors partially prevented the Mn2+-induced cell viability decrease. Altogether these events point to lysosomes as players in the execution of RN. In summary, our results suggest that microglial cells could be direct targets of Mn2+ damage. In this scenario, Mn2+ triggers cell death involving RN pathways.
Collapse
Affiliation(s)
- Soledad Porte Alcon
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina, Buenos Aires, Argentina.
| | - Roxana Mayra Gorojod
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina, Buenos Aires, Argentina.
| | - Mónica Lidia Kotler
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina, Buenos Aires, Argentina.
| |
Collapse
|
21
|
Pardo-Peña K, Lorea-Hernández JJ, Camacho-Hernández NP, Ordaz B, Villasana-Salazar B, Morales-Villagrán A, Peña-Ortega F. Hydrogen peroxide extracellular concentration in the ventrolateral medulla and its increase in response to hypoxia in vitro: Possible role of microglia. Brain Res 2018; 1692:87-99. [DOI: 10.1016/j.brainres.2018.04.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 03/31/2018] [Accepted: 04/25/2018] [Indexed: 12/12/2022]
|
22
|
Garza-Lombó C, Posadas Y, Quintanar L, Gonsebatt ME, Franco R. Neurotoxicity Linked to Dysfunctional Metal Ion Homeostasis and Xenobiotic Metal Exposure: Redox Signaling and Oxidative Stress. Antioxid Redox Signal 2018; 28:1669-1703. [PMID: 29402131 PMCID: PMC5962337 DOI: 10.1089/ars.2017.7272] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE Essential metals such as copper, iron, manganese, and zinc play a role as cofactors in the activity of a wide range of processes involved in cellular homeostasis and survival, as well as during organ and tissue development. Throughout our life span, humans are also exposed to xenobiotic metals from natural and anthropogenic sources, including aluminum, arsenic, cadmium, lead, and mercury. It is well recognized that alterations in the homeostasis of essential metals and an increased environmental/occupational exposure to xenobiotic metals are linked to several neurological disorders, including neurodegeneration and neurodevelopmental alterations. Recent Advances: The redox activity of essential metals is key for neuronal homeostasis and brain function. Alterations in redox homeostasis and signaling are central to the pathological consequences of dysfunctional metal ion homeostasis and increased exposure to xenobiotic metals. Both redox-active and redox-inactive metals trigger oxidative stress and damage in the central nervous system, and the exact mechanisms involved are starting to become delineated. CRITICAL ISSUES In this review, we aim to appraise the role of essential metals in determining the redox balance in the brain and the mechanisms by which alterations in the homeostasis of essential metals and exposure to xenobiotic metals disturb the cellular redox balance and signaling. We focus on recent literature regarding their transport, metabolism, and mechanisms of toxicity in neural systems. FUTURE DIRECTIONS Delineating the specific mechanisms by which metals alter redox homeostasis is key to understand the pathological processes that convey chronic neuronal dysfunction in neurodegenerative and neurodevelopmental disorders. Antioxid. Redox Signal. 28, 1669-1703.
Collapse
Affiliation(s)
- Carla Garza-Lombó
- 1 Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln , Lincoln, Nebraska.,2 Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas , Universidad Nacional Autónoma de México, Mexico City, México
| | - Yanahi Posadas
- 3 Departamentos de Farmacología y de, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México .,4 Departamentos de Química, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México
| | - Liliana Quintanar
- 4 Departamentos de Química, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México
| | - María E Gonsebatt
- 2 Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas , Universidad Nacional Autónoma de México, Mexico City, México
| | - Rodrigo Franco
- 1 Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln , Lincoln, Nebraska
| |
Collapse
|
23
|
Song D, Ma J, Chen L, Guo C, Zhang Y, Chen T, Zhang S, Zhu Z, Tian L, Niu P. FOXO3 promoted mitophagy via nuclear retention induced by manganese chloride in SH-SY5Y cells. Metallomics 2018; 9:1251-1259. [PMID: 28661534 DOI: 10.1039/c7mt00085e] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES To evaluate the role of FOXO3 during the process of mitophagy induced by manganese chloride (MnCl2), mitochondrial dysfunction and mitophagy were detected before and after FOXO3 was knocked down in SH-SY5Y cells. METHOD Transmission electron microscopy (TEM), flow cytometry, confocal microscopy and a western blot were used to detect mitochondrial ultrastructure and autophagy, Ca2+ levels, mitochondrial reactive oxygen species (ROS) and the mitochondrial membrane potential (MMP), autophagosomes and mitophagy marker proteins (p62, LC3-II/LC3-I, Beclin-1, PINK1 and P-parkin), respectively. RESULTS After SH-SY5Y cells were exposed to MnCl2, the levels of cytoplasmic Ca2+ and mitochondrial ROS increased but the mitochondrial MMP decreased significantly compared to the control in a dose- and time-dependent manner (p < 0.05), which indicated that MnCl2 can lead to mitochondrial dysfunction. Under TEM, mitophagy and autolysosomes were observed. The WB results also showed that mitophagy marker proteins including LC3-II/LC3-I, Beclin-1, PINK1 and P-parkin except for p62 increased in a dose- and time-dependent manner, accompanied by FOXO3 nuclear retention, which indicated that MnCl2 can lead to mitophagy and FOXO3 nuclear translocation may be involved in this process. After FOXO3 was knocked down, the inverse results of mitophagy and the levels of mitochondrial ROS decreasing were observed, which showed that FOXO3 silencing could inhibit mitophagy and mitochondrial dysfunction induced by MnCl2. CONCLUSIONS Our results indicated that Mn could induce mitophagy by enhancing FOXO3 nuclear retention, which might promote mitophagy induced by MnCl2.
Collapse
Affiliation(s)
- Dongmei Song
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, No. 10 Xitoutiao Road, You'anmenwai Street, Fengtai District, Beijing, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Protective effect of vinpocetine against neurotoxicity of manganese in adult male rats. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:729-742. [PMID: 29671021 DOI: 10.1007/s00210-018-1498-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/05/2018] [Indexed: 01/10/2023]
Abstract
Manganese (Mn) is required for many essential biological processes as well as in the development and functioning of the brain. Extensive accumulation of Mn in the brain may cause central nervous system dysfunction known as manganism, a motor disorder associated with cognitive and neuropsychiatric deficits similar to parkinsonism. Vinpocetine, a synthetic derivative of the alkaloid vincamine, is used to improve the cognitive function in cerebrovascular diseases. It possesses antioxidant and antiinflammatory properties. The present work was designed to explore the potential neuroprotective mechanisms exerted by vinpocetine in the Mn-induced neurotoxicity in rats. Rats were allocated into four groups. First group was given saline. The other three groups were given MnCl2; two of them were treated with either L-dopa, the gold standard antiparkinsonian drug, or vinpocetine. Rats receiving MnCl2 exhibited lengthened catalepsy duration in the grid and bar tests, motor impairment in the open-field test and short-term memory deficit in the Y-maze test. Additionally, histological examination revealed structural alterations and degeneration in different brain regions. Besides, striatal monoamines and mitochondrial complex I contents were declined, apoptotic biomarker caspase-3 expression and acetylcholinesterase activity were elevated. Moreover, oxidative stress and inflammation were detected in the striata. L-dopa or vinpocetine exerted protective effects against MnCl2-induced neurotoxicity. It could be hypothesized that modulation of monoamines, upregulation of mitochondrial complex I, antioxidant, antiinflammatory, and antiapoptotic activities are significant mechanisms underlying the neuroprotective effect of vinpocetine in the Mn-induced neurotoxicity model in rats.
Collapse
|
25
|
Critical Role of Zinc as Either an Antioxidant or a Prooxidant in Cellular Systems. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9156285. [PMID: 29743987 PMCID: PMC5884210 DOI: 10.1155/2018/9156285] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/09/2018] [Accepted: 01/16/2018] [Indexed: 01/11/2023]
Abstract
Zinc is recognized as an essential trace metal required for human health; its deficiency is strongly associated with neuronal and immune system defects. Although zinc is a redox-inert metal, it functions as an antioxidant through the catalytic action of copper/zinc-superoxide dismutase, stabilization of membrane structure, protection of the protein sulfhydryl groups, and upregulation of the expression of metallothionein, which possesses a metal-binding capacity and also exhibits antioxidant functions. In addition, zinc suppresses anti-inflammatory responses that would otherwise augment oxidative stress. The actions of zinc are not straightforward owing to its numerous roles in biological systems. It has been shown that zinc deficiency and zinc excess cause cellular oxidative stress. To gain insights into the dual action of zinc, as either an antioxidant or a prooxidant, and the conditions under which each role is performed, the oxidative stresses that occur in zinc deficiency and zinc overload in conjunction with the intracellular regulation of free zinc are summarized. Additionally, the regulatory role of zinc in mitochondrial homeostasis and its impact on oxidative stress are briefly addressed.
Collapse
|
26
|
Langley MR, Ghaisas S, Ay M, Luo J, Palanisamy BN, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Manganese exposure exacerbates progressive motor deficits and neurodegeneration in the MitoPark mouse model of Parkinson's disease: Relevance to gene and environment interactions in metal neurotoxicity. Neurotoxicology 2018; 64:240-255. [PMID: 28595911 PMCID: PMC5736468 DOI: 10.1016/j.neuro.2017.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/02/2017] [Accepted: 06/02/2017] [Indexed: 10/19/2022]
Abstract
Parkinson's disease (PD) is now recognized as a neurodegenerative condition caused by a complex interplay of genetic and environmental influences. Chronic manganese (Mn) exposure has been implicated in the development of PD. Since mitochondrial dysfunction is associated with PD pathology as well as Mn neurotoxicity, we investigated whether Mn exposure augments mitochondrial dysfunction and neurodegeneration in the nigrostriatal dopaminergic system using a newly available mitochondrially defective transgenic mouse model of PD, the MitoPark mouse. This unique PD model recapitulates key features of the disease including progressive neurobehavioral changes and neuronal degeneration. We exposed MitoPark mice to a low dose of Mn (10mg/kg, p.o.) daily for 4 weeks starting at age 8 wks and then determined the behavioral, neurochemical and histological changes. Mn exposure accelerated the rate of progression of motor deficits in MitoPark mice when compared to the untreated MitoPark group. Mn also worsened olfactory function in this model. Most importantly, Mn exposure intensified the depletion of striatal dopamine and nigral TH neuronal loss in MitoPark mice. The neurodegenerative changes were accompanied by enhanced oxidative damage in the striatum and substantia nigra (SN) of MitoPark mice treated with Mn. Furthermore, Mn-treated MitoPark mice had significantly more oligomeric protein and IBA-1-immunoreactive microglia cells, suggesting Mn augments neuroinflammatory processes in the nigrostriatal pathway. To further confirm the direct effect of Mn on impaired mitochondrial function, we also generated a mitochondrially defective dopaminergic cell model by knocking out the TFAM transcription factor by using a CRISPR-Cas9 gene-editing method. Seahorse mitochondrial bioenergetic analysis revealed that Mn decreases mitochondrial basal and ATP-linked respiration in the TFAM KO cells. Collectively, our results reveal that Mn can augment mitochondrial dysfunction to exacerbate nigrostriatal neurodegeneration and PD-related behavioral symptoms. Our study also demonstrates that the MitoPark mouse is an excellent model to study the gene-environment interactions associated with mitochondrial defects in the nigral dopaminergic system as well as to evaluate the contribution of potential environmental toxicant interactions in a slowly progressive model of Parkinsonism.
Collapse
Affiliation(s)
- Monica R Langley
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Shivani Ghaisas
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Muhammet Ay
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Jie Luo
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Bharathi N Palanisamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Huajun Jin
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Vellareddy Anantharam
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Arthi Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Anumantha G Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
27
|
Xu J, Church SJ, Patassini S, Begley P, Waldvogel HJ, Curtis MA, Faull RLM, Unwin RD, Cooper GJS. Evidence for widespread, severe brain copper deficiency in Alzheimer's dementia. Metallomics 2017; 9:1106-1119. [PMID: 28654115 DOI: 10.1039/c7mt00074j] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Datasets comprising simultaneous measurements of many essential metals in Alzheimer's disease (AD) brain are sparse, and available studies are not entirely in agreement. To further elucidate this matter, we employed inductively-coupled-plasma mass spectrometry to measure post-mortem levels of 8 essential metals and selenium, in 7 brain regions from 9 cases with AD (neuropathological severity Braak IV-VI), and 13 controls who had normal ante-mortem mental function and no evidence of brain disease. Of the regions studied, three undergo severe neuronal damage in AD (hippocampus, entorhinal cortex and middle-temporal gyrus); three are less-severely affected (sensory cortex, motor cortex and cingulate gyrus); and one (cerebellum) is relatively spared. Metal concentrations in the controls differed among brain regions, and AD-associated perturbations in most metals occurred in only a few: regions more severely affected by neurodegeneration generally showed alterations in more metals, and cerebellum displayed a distinctive pattern. By contrast, copper levels were substantively decreased in all AD-brain regions, to 52.8-70.2% of corresponding control values, consistent with pan-cerebral copper deficiency. This copper deficiency could be pathogenic in AD, since levels are lowered to values approximating those in Menkes' disease, an X-linked recessive disorder where brain-copper deficiency is the accepted cause of severe brain damage. Our study reinforces others reporting deficient brain copper in AD, and indicates that interventions aimed at safely and effectively elevating brain copper could provide a new experimental-therapeutic approach.
Collapse
Affiliation(s)
- Jingshu Xu
- School of Biological Sciences, Faculty of Science, and the Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand. and Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester M13 9WL, UK and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Stephanie J Church
- Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester M13 9WL, UK and Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, and Manchester Academic Health Science Centre, Manchester M13 9NT, UK
| | - Stefano Patassini
- School of Biological Sciences, Faculty of Science, and the Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand. and Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester M13 9WL, UK and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Paul Begley
- Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester M13 9WL, UK and Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, and Manchester Academic Health Science Centre, Manchester M13 9NT, UK
| | - Henry J Waldvogel
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard D Unwin
- Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester M13 9WL, UK and Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, and Manchester Academic Health Science Centre, Manchester M13 9NT, UK
| | - Garth J S Cooper
- School of Biological Sciences, Faculty of Science, and the Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand. and Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester M13 9WL, UK and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand and Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, and Manchester Academic Health Science Centre, Manchester M13 9NT, UK
| |
Collapse
|
28
|
Gugnani KS, Vu N, Rondón-Ortiz AN, Böhlke M, Maher TJ, Pino-Figueroa AJ. Neuroprotective activity of macamides on manganese-induced mitochondrial disruption in U-87 MG glioblastoma cells. Toxicol Appl Pharmacol 2017; 340:67-76. [PMID: 29288688 DOI: 10.1016/j.taap.2017.12.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/09/2017] [Accepted: 12/23/2017] [Indexed: 02/07/2023]
Abstract
Macamides are a distinct class of secondary metabolites, benzylamides of long chain fatty acids, which were isolated from the Peruvian plant Lepidium meyenii (Maca). As structural analogues of the endocannabinoid anandamide (AEA), they have demonstrated neuroprotective effects in vitro and in vivo. The purpose of this study was to demonstrate the neuroprotective activity of the macamides: N-(3-methoxybenzyl)oleamide (MAC 18:1), N-(3-methoxybenzyl)linoleamide (MAC 18:2) and N-(3-methoxybenzyl)linolenamide (MAC 18:3) in a neurotoxic environment caused by exposure of U-87 MG glioblastoma cells to manganese chloride (MnCl2). The neuroprotective effects of these macamides were reversed by the CB1 antagonist AM251. The mechanism by which manganese (Mn) induces cell damage was investigated by studying its effects on mitochondria. Reactive oxygen species (ROS) increase intracellular calcium and enhance the opening of mitochondrial permeability transition pores (MPTP), which leads to decreased mitochondrial membrane potential (MMP), to disruption of mitochondria and to neuron death in neurodegenerative disorders. In this study, MnCl2 at 50μM was responsible for mitochondrial disruption, which was attenuated by all three of the macamides tested. Human peroxisome proliferator-activated receptor gamma (PPARγ) has been proposed to be a cannabinoid target, and PPARγ has also been demonstrated to mediate some of the longer-term vascular effects of the plant cannabinoid, ∆9-tetrahydrocannabinol. PPARγ activation was observed in response to exposures of cells to MAC 18:2 and MAC 18:3. These findings suggest that macamides achieve their neuroprotective effects by binding to CB1 receptors to protect against Mn-induced toxicity in U-87 MG glioblastoma cells. Additionally these macamides, in a manner similar to the analogous endocannabinoid AEA, interact with other targets such as PPARγ to regulate metabolism and energy homeostasis, cell differentiation and inflammation.
Collapse
Affiliation(s)
- Kuljeet S Gugnani
- Department of Pharmaceutical Sciences, MCPHS University, 179 Longwood Avenue, Boston, MA, USA
| | - Nguyen Vu
- School of Pharmacy, MCPHS University, 179 Longwood Avenue, Boston, MA, USA
| | | | - Mark Böhlke
- Department of Pharmaceutical Sciences, MCPHS University, 179 Longwood Avenue, Boston, MA, USA
| | - Timothy J Maher
- Department of Pharmaceutical Sciences, MCPHS University, 179 Longwood Avenue, Boston, MA, USA
| | | |
Collapse
|
29
|
Nam E, Han J, Suh JM, Yi Y, Lim MH. Link of impaired metal ion homeostasis to mitochondrial dysfunction in neurons. Curr Opin Chem Biol 2017; 43:8-14. [PMID: 29100100 DOI: 10.1016/j.cbpa.2017.09.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/03/2017] [Accepted: 09/12/2017] [Indexed: 02/07/2023]
Abstract
Manganese, iron, copper, and zinc are observed to play essential roles in mitochondria. The overload and depletion of metal ions in mitochondria under pathological conditions, however, could disturb mitochondrial compartments and functions leading to cell death. In this review, we mainly summarize how impaired metal ion homeostasis affects mitochondrial systems, such as membrane potentials, the tricarboxylic acid cycle, oxidative phosphorylation, and glutathione metabolism. In addition, based on current findings, we briefly describe a recent understanding of the relationship among metal ion dysregulation, mitochondrial dysfunction, and the pathogeneses of neurodegenerative diseases.
Collapse
Affiliation(s)
- Eunju Nam
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jiyeon Han
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jong-Min Suh
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Yelim Yi
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| |
Collapse
|
30
|
Tikamdas R, Singhal S, Zhang P, Smith JA, Krause EG, Stevens SM, Song S, Liu B. Ischemia-responsive protein 94 is a key mediator of ischemic neuronal injury-induced microglial activation. J Neurochem 2017. [PMID: 28640931 DOI: 10.1111/jnc.14111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Neuroinflammation, especially activation of microglia, the key immune cells in the brain, has been proposed to contribute to the pathogenesis of ischemic stroke. However, the dynamics and the potential mediators of microglial activation following ischemic neuronal injury are not well understood. In this study, using oxygen/glucose deprivation and reoxygenation with neuronal and microglial cell cultures as an in vitro model of ischemic neuronal injury, we set out to identify neuronal factors released from injured neurons that are capable of inducing microglial activation. Conditioned media (CM) from hippocampal and cortical neurons exposed to oxygen/glucose deprivation and reoxygenation induced significant activation of microglial cells as well as primary microglia, evidenced by up-regulation of inducible nitric oxide synthase, increased production of nitrite and reactive oxygen species, and increased expression of microglial markers. Mechanistically, neuronal ischemia-responsive protein 94 (Irp94) was a key contributor to microglial activation since significant increase in Irp94 was detected in the neuronal CM following ischemic insult and immunodepletion of Irp94 rendered ischemic neuronal CM ineffective in inducing microglial activation. Ischemic insult-augmented oxidative stress was a major facilitator of neuronal Irp94 release, and pharmacological inhibition of NADPH oxidase significantly reduced the ischemic injury-induced neuronal reactive oxygen species production and Irp94 release. Taken together, these results indicate that neuronal Irp94 may play a pivotal role in the propagation of ischemic neuronal damage. Continued studies may help identify Irp94 and/or related proteins as potential therapeutic targets and/or diagnostic/prognostic biomarkers for managing ischemia-associated brain disorders.
Collapse
Affiliation(s)
- Rajiv Tikamdas
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Sarthak Singhal
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Ping Zhang
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Justin A Smith
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Eric G Krause
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Stanley M Stevens
- Department of Cell Biology, Microbiology and Molecular Biology, College of Arts and Sciences, University of South Florida, Tampa, Florida, USA
| | - Sihong Song
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Bin Liu
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
31
|
Park E, Chun HS. Protective Effects of Curcumin on Manganese-Induced BV-2 Microglial Cell Death. Biol Pharm Bull 2017; 40:1275-1281. [PMID: 28529240 DOI: 10.1248/bpb.b17-00160] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Curcumin, a bioactive component in tumeric, has been shown to exert antioxidant, anti-inflammatory, anticarcinogenic, hepatoprotective, and neuroprotective effects, but the effects of curcumin against manganese (Mn)-mediated neurotoxicity have not been studied. This study examined the protective effects of curcumin on Mn-induced cytotoxicity in BV-2 microglial cells. Curcumin (0.1-10 µM) dose-dependently prevented Mn (250 µM)-induced cell death. Mn-induced mitochondria-related apoptotic characteristics, such as caspase-3 and -9 activation, cytochrome c release, Bax increase, and Bcl-2 decrease, were significantly suppressed by curcumin. In addition, curcumin significantly increased intracellular glutathione (GSH) and moderately potentiated superoxide dismutase (SOD), both which were diminished by Mn treatment. Curcumin pretreatment effectively suppressed Mn-induced upregulation of malondialdehyde (MDA), total reactive oxygen species (ROS). Moreover, curcumin markedly inhibited the Mn-induced mitochondrial membrane potential (MMP) loss. Furthermore, curcumin was able to induce heme oxygenase (HO)-1 expression. Curcumin-mediated inhibition of ROS, down-regulation of caspases, restoration of MMP, and recovery of cell viability were partially reversed by HO-1 inhibitor (SnPP). These results suggest the first evidence that curcumin can prevent Mn-induced microglial cell death through the induction of HO-1 and regulation of oxidative stress, mitochondrial dysfunction, and apoptotic events.
Collapse
Affiliation(s)
- Euteum Park
- Department of Biomedical Science, Chosun University
| | | |
Collapse
|
32
|
Smith MR, Fernandes J, Go YM, Jones DP. Redox dynamics of manganese as a mitochondrial life-death switch. Biochem Biophys Res Commun 2017; 482:388-398. [PMID: 28212723 PMCID: PMC5382988 DOI: 10.1016/j.bbrc.2016.10.126] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/25/2016] [Accepted: 10/27/2016] [Indexed: 12/16/2022]
Abstract
Sten Orrenius, M.D., Ph.D., pioneered many areas of cellular and molecular toxicology and made seminal contributions to our knowledge of oxidative stress and glutathione (GSH) metabolism, organellar functions and Ca+2-dependent mechanisms of cell death, and mechanisms of apoptosis. On the occasion of his 80th birthday, we summarize current knowledge on redox biology of manganese (Mn) and its role in mechanisms of cell death. Mn is found in all organisms and has critical roles in cell survival and death mechanisms by regulating Mn-containing enzymes such as manganese superoxide dismutase (SOD2) or affecting expression and activity of caspases. Occupational exposures to Mn cause "manganism", a Parkinson's disease-like condition of neurotoxicity, and experimental studies show that Mn exposure leads to accumulation of Mn in the brain, especially in mitochondria, and neuronal cell death occurs with features of an apoptotic mechanism. Interesting questions are why a ubiquitous metal that is essential for mitochondrial function would accumulate to excessive levels, cause increased H2O2 production and lead to cell death. Is this due to the interactions of Mn with other essential metals, such as iron, or with toxic metals, such as cadmium? Why is the Mn loading in the human brain so variable, and why is there such a narrow window between dietary adequacy and toxicity? Are non-neuronal tissues similarly vulnerable to insufficiency and excess, yet not characterized? We conclude that Mn is an important component of the redox interface between an organism and its environment and warrants detailed studies to understand the role of Mn as a mitochondrial life-death switch.
Collapse
Affiliation(s)
- Matthew Ryan Smith
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Jolyn Fernandes
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Young-Mi Go
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
33
|
Fernandes J, Hao L, Bijli KM, Chandler JD, Orr M, Hu X, Jones DP, Go YM. From the Cover: Manganese Stimulates Mitochondrial H2O2 Production in SH-SY5Y Human Neuroblastoma Cells Over Physiologic as well as Toxicologic Range. Toxicol Sci 2016; 155:213-223. [PMID: 27701121 DOI: 10.1093/toxsci/kfw196] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Manganese (Mn) is an abundant redox-active metal with well-characterized mitochondrial accumulation and neurotoxicity due to excessive exposures. Mn is also an essential co-factor for the mitochondrial antioxidant protein, superoxide dismutase-2 (SOD2), and the range for adequate intake established by the Institute of Medicine Food and Nutrition Board is 20% of the interim guidance value for toxicity by the Agency for Toxic Substances and Disease Registry, leaving little margin for safety. To study toxic mechanisms over this critical dose range, we treated human neuroblastoma SH-SY5Y cells with a series of MnCl2 concentrations (from 0 to 100 μM) and measured cellular content to compare to human brain Mn content. Concentrations ≤10 μM gave cellular concentrations comparable to literature values for normal human brain, whereas concentrations ≥50 μM resulted in values comparable to brains from individuals with toxic Mn exposures. Cellular oxygen consumption rate increased as a function of Mn up to 10 μM and decreased with Mn dose ≥50 μM. Over this range, Mn had no effect on superoxide production as measured by aconitase activity or MitoSOX but increased H2O2 production as measured by MitoPY1. Consistent with increased production of H2O2, SOD2 activity, and steady-state oxidation of total thiol increased with increasing Mn. These findings have important implications for Mn toxicity by re-directing attention from superoxide anion radical to H2O2-dependent mechanisms and to investigation over the entire physiologic range to toxicologic range. Additionally, the results show that controlled Mn exposure provides a useful cell manipulation for toxicological studies of mitochondrial H2O2 signaling.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Young-Mi Go
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
34
|
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
|
35
|
da Rosa MS, Seminotti B, Ribeiro CAJ, Parmeggiani B, Grings M, Wajner M, Leipnitz G. 3-Hydroxy-3-methylglutaric and 3-methylglutaric acids impair redox status and energy production and transfer in rat heart: relevance for the pathophysiology of cardiac dysfunction in 3-hydroxy-3-methylglutaryl-coenzyme A lyase deficiency. Free Radic Res 2016; 50:997-1010. [PMID: 27430492 DOI: 10.1080/10715762.2016.1214952] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
3-Hydroxy-3-methylglutaryl-coenzyme A lyase (HL) deficiency is characterized by tissue accumulation of 3-hydroxy-3-methylglutaric (HMG), and 3-methylglutaric (MGA) acids. Affected patients present cardiomyopathy, whose pathomechanisms are not yet established. We investigated the effects of HMG and MGA on energy and redox homeostasis in rat heart using in vivo and in vitro models. In vivo experiments showed that intraperitoneal administration of HMG and MGA decreased the activities of the respiratory chain complex II and creatine kinase (CK), whereas HMG also decreased the activity of complex II-III. Furthermore, HMG and MGA injection increased reactive species production and carbonyl formation, and decreased glutathione concentrations. Regarding the enzymatic antioxidant defenses, HMG and MGA increased glutathione peroxidase (GPx) and glutathione reductase (GR) activities, while only MGA diminished the activities of superoxide dismutase (SOD) and catalase, as well as the protein content of SOD1. Pre-treatment with melatonin (MEL) prevented MGA-induced decrease of CK activity and SOD1 levels. In vitro results demonstrated that HMG and MGA increased reactive species formation, induced lipid peroxidation and decreased glutathione. We also verified that reactive species overproduction and glutathione decrease provoked by HMG and MGA were abrogated by MEL and lipoic acid (LA), while only MEL prevented HMG- and MGA-induced lipoperoxidation. Allopurinol (ALP) also prevented reactive species overproduction caused by both metabolites. Our data provide solid evidence that bioenergetics dysfunction and oxidative stress are induced by HMG and MGA in heart, which may explain the cardiac dysfunction observed in HL deficiency, and also suggest that antioxidant supplementation could be considered as adjuvant therapy for affected patients.
Collapse
Affiliation(s)
- Mateus Struecker da Rosa
- a Programa de Pós-Graduação em Ciências Biológicas: Bioquímica , Instituto de Ciências Básicas da Saúde , UFRGS , Porto Alegre, RS , Brazil
| | - Bianca Seminotti
- a Programa de Pós-Graduação em Ciências Biológicas: Bioquímica , Instituto de Ciências Básicas da Saúde , UFRGS , Porto Alegre, RS , Brazil
| | - César Augusto João Ribeiro
- b Centro de Ciências Naturais e Humanas , Universidade Federal do ABC , São Bernardo do Campo , SP , Brazil
| | - Belisa Parmeggiani
- a Programa de Pós-Graduação em Ciências Biológicas: Bioquímica , Instituto de Ciências Básicas da Saúde , UFRGS , Porto Alegre, RS , Brazil
| | - Mateus Grings
- a Programa de Pós-Graduação em Ciências Biológicas: Bioquímica , Instituto de Ciências Básicas da Saúde , UFRGS , Porto Alegre, RS , Brazil
| | - Moacir Wajner
- a Programa de Pós-Graduação em Ciências Biológicas: Bioquímica , Instituto de Ciências Básicas da Saúde , UFRGS , Porto Alegre, RS , Brazil ;,c Departamento de Bioquímica , Instituto de Ciências Básicas da Saúde , UFRGS , Porto Alegre, RS , Brazil ;,d Serviço de Genética Médica , Hospital de Clínicas de Porto Alegre , Porto Alegre , RS , Brazil
| | - Guilhian Leipnitz
- a Programa de Pós-Graduação em Ciências Biológicas: Bioquímica , Instituto de Ciências Básicas da Saúde , UFRGS , Porto Alegre, RS , Brazil ;,c Departamento de Bioquímica , Instituto de Ciências Básicas da Saúde , UFRGS , Porto Alegre, RS , Brazil
| |
Collapse
|
36
|
Coherent and Contradictory Facts, Feats and Fictions Associated with Metal Accumulation in Parkinson's Disease: Epicenter or Outcome, Yet a Demigod Question. Mol Neurobiol 2016; 54:4738-4755. [PMID: 27480264 DOI: 10.1007/s12035-016-0016-y] [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] [Received: 03/14/2016] [Accepted: 07/12/2016] [Indexed: 01/30/2023]
Abstract
Unwarranted exposure due to liberal use of metals for maintaining the lavish life and to achieve the food demand for escalating population along with an incredible boost in the average human life span owing to orchestrated progress in rejuvenation therapy have gradually increased the occurrence of Parkinson's disease (PD). Etiology is albeit elusive; association of PD with metal accumulation has never been overlooked due to noteworthy similitude between metal-exposure symptoms and a few cardinal features of disease. Even though metals are entailed in the vital functions, a hysterical shift, primarily augmentation, escorts the stern nigrostriatal dopaminergic neurodegeneration. An increase in the passage of metals through the blood brain barrier and impaired metabolic activity and elimination system could lead to metal accumulation in the brain, which eventually makes dopaminergic neurons quite susceptible. In the present article, an update on implication of metal accumulation in PD/Parkinsonism has been provided. Moreover, encouraging and paradoxical facts and fictions associated with metal accumulation in PD/Parkinsonism have also been compiled. Systematic literature survey of PD is performed to describe updated information if metal accumulation is an epicenter or merely an outcome. Finally, a perspective on the association of metal accumulation with pesticide-induced Parkinsonism has been explained to unveil the likely impact of the former in the latter.
Collapse
|
37
|
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: 27] [Impact Index Per Article: 2.7] [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]
|
38
|
Mokgobu MI, Cholo MC, Anderson R, Steel HC, Motheo MP, Hlatshwayo TN, Tintinger GR, Theron AJ. Oxidative induction of pro-inflammatory cytokine formation by human monocyte-derived macrophages following exposure to manganese in vitro. J Immunotoxicol 2014; 12:98-103. [PMID: 24806275 DOI: 10.3109/1547691x.2014.902877] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Manganese (as Mn(2+)), a superoxide dismutase mimetic, catalyzes the formation of the relatively stable membrane-permeable reactive oxygen species (ROS) hydrogen peroxide (H2O2), a mediator of intracellular redox signaling in immune and inflammatory cells. The goal of this study was to investigate the potential for Mn(2+), via its pro-oxidative properties, to activate production of pro-inflammatory cytokines/chemokines IL-1β, IL-6, IL-8, IFNγ, TNFα, and G-CSF by human monocyte-derived macrophages in vitro. For these studies, the cells were isolated from peripheral blood mononuclear leukocytes and matured to generate a population of large CD14/CD16 co-expressing cells. The monocyte-derived macrophages were then exposed to bacterial lipopolysaccharide (LPS, 1 μg/ml) or MnCl2 (25-100 μM)-alone or in combination-for 24 h at 37 °C, after which cell-free supernatants were analyzed using a multiplex cytokine assay procedure. Exposure of the cells to LPS caused modest statistically insignificant increases in cytokine production; MnCl2 caused dose-related increases in production of all six cytokines (achieving statistical significance of p < 0.0171- < 0.0005 for IL-1β, IL-6, IL-8, IFNγ, and TNFα). In the case of LPS and MnCl2 combinations, the observed increases in production of IL-1β, IL-6, IL-8, IFNγ, and G-CSF were greater than those seen with cells exposed to the individual agents. The Mn(2+)-mediated induction of cytokine production was associated with increased production of H2O2 and completely attenuated by inclusion of the H2O2-scavenger dithiothreitol, and partially by inhibitors of NF-κB and p38MAP kinase. The findings from the studies here help to further characterize the pro-inflammatory mechanisms that may underpin clinical disorders associated with excess exposure to Mn(2+), particularly those disorders seen in the central nervous and respiratory systems.
Collapse
Affiliation(s)
- Matlou I Mokgobu
- Medical Research Council Unit for Inflammation and Immunity, Department of Immunology, Faculty of Health Sciences, University of Pretoria , Pretoria , South Africa
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Do metals that translocate to the brain exacerbate traumatic brain injury? Med Hypotheses 2014; 82:558-62. [DOI: 10.1016/j.mehy.2014.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 01/22/2014] [Accepted: 02/08/2014] [Indexed: 11/19/2022]
|
40
|
Upregulation of mitochondrial protease HtrA2/Omi contributes to manganese-induced neuronal apoptosis in rat brain striatum. Neuroscience 2014; 268:169-79. [DOI: 10.1016/j.neuroscience.2014.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 03/04/2014] [Accepted: 03/04/2014] [Indexed: 11/22/2022]
|
41
|
Bornhorst J, Chakraborty S, Meyer S, Lohren H, Brinkhaus SG, Knight AL, Caldwell KA, Caldwell GA, Karst U, Schwerdtle T, Bowman A, Aschner M. The effects of pdr1, djr1.1 and pink1 loss in manganese-induced toxicity and the role of α-synuclein in C. elegans. Metallomics 2014; 6:476-90. [PMID: 24452053 DOI: 10.1039/c3mt00325f] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative brain disorder characterized by selective dopaminergic (DAergic) cell loss that results in overt motor and cognitive deficits. Current treatment options exist to combat PD symptomatology, but are unable to directly target its pathogenesis due to a lack of knowledge concerning its etiology. Several genes have been linked to PD, including three genes associated with an early-onset familial form: parkin, pink1 and dj1. All three genes are implicated in regulating oxidative stress pathways. Another hallmark of PD pathophysiology is Lewy body deposition, associated with the gain-of-function genetic risk factor α-synuclein. The function of α-synuclein is poorly understood, as it shows both neurotoxic and neuroprotective activities in PD. Using the genetically tractable invertebrate Caenorhabditis elegans (C. elegans) model system, the neurotoxic or neuroprotective role of α-synuclein upon acute Mn exposure in the background of mutated pdr1, pink1 or djr1.1 was examined. The pdr1 and djr1.1 mutants showed enhanced Mn accumulation and oxidative stress that was reduced by α-synuclein. Moreover, DAergic neurodegeneration, while unchanged with Mn exposure, returned to wild-type (WT) levels for pdr1, but not djr1.1 mutants expressing α-synuclein. Taken together, this study uncovers a novel, neuroprotective role for WT human α-synuclein in attenuating Mn-induced toxicity in the background of PD-associated genes, and further supports the role of extracellular dopamine in exacerbating Mn neurotoxicity.
Collapse
Affiliation(s)
- Julia Bornhorst
- Institute of Food Chemistry, University of Münster, Münster, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
In vitro manganese exposure disrupts MAPK signaling pathways in striatal and hippocampal slices from immature rats. BIOMED RESEARCH INTERNATIONAL 2013; 2013:769295. [PMID: 24324973 PMCID: PMC3845707 DOI: 10.1155/2013/769295] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 10/07/2013] [Indexed: 12/17/2022]
Abstract
The molecular mechanisms mediating manganese (Mn)-induced neurotoxicity, particularly in the immature central nervous system, have yet to be completely understood. In this study, we investigated whether mitogen-activated protein kinases (MAPKs) and tyrosine hydroxylase (TH) could represent potential targets of Mn in striatal and hippocampal slices obtained from immature rats (14 days old). The aim of this study was to evaluate if the MAPK pathways are modulated after subtoxic Mn exposure, which do not significantly affect cell viability. The concentrations of manganese chloride (MnCl₂; 10-1,000 μM) caused no change in cell viability in slices exposed for 3 or 6 hours. However, Mn exposure significantly increased extracellular signal-regulated kinase (ERK) 1/2, as well as c-Jun N-terminal kinase (JNK) 1/2/3 phosphorylation at both 3 and 6 hours incubations, in both brain structures. Furthermore, Mn exposure did not change the total content or phosphorylation of TH at the serine 40 site in striatal slices. Thus, Mn at concentrations that do not disrupt cell viability causes activation of MAPKs (ERK1/2 and JNK1/2/3) in immature hippocampal and striatal slices. These findings suggest that altered intracellular MAPKs signaling pathways may represent an early event concerning the effects of Mn in the immature brain.
Collapse
|