1
|
Zheng XW, Fang YY, Lin JJ, Luo JJ, Li SJ, Aschner M, Jiang YM. Signal Transduction Associated with Mn-induced Neurological Dysfunction. Biol Trace Elem Res 2024; 202:4158-4169. [PMID: 38155332 DOI: 10.1007/s12011-023-03999-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
Manganese (Mn) is a heavy metal that occurs widely in nature and has a vital physiological role in growth and development. However, excessive exposure to Mn can cause neurological damage, especially cognitive dysfunction, such as learning disability and memory loss. Numerous studies on the mechanisms of Mn-induced nervous system damage found that this metal targets a variety of metabolic pathways, for example, endoplasmic reticulum stress, apoptosis, neuroinflammation, cellular signaling pathway changes, and neurotransmitter metabolism interference. This article reviews the latest research progress on multiple signaling pathways related to Mn-induced neurological dysfunction.
Collapse
Affiliation(s)
- Xiao-Wei Zheng
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China
| | - Yuan-Yuan Fang
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China
| | - Jun-Jie Lin
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China
| | - Jing-Jing Luo
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China
| | - Shao-Jun Li
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China.
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China.
| | - Michael Aschner
- The Department of Molecular Pharmacology at Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yue-Ming Jiang
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China.
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China.
| |
Collapse
|
2
|
Aschner M, Martins AC, Oliveira-Paula GH, Skalny AV, Zaitseva IP, Bowman AB, Kirichuk AA, Santamaria A, Tizabi Y, Tinkov AA. Manganese in autism spectrum disorder and attention deficit hyperactivity disorder: The state of the art. Curr Res Toxicol 2024; 6:100170. [PMID: 38737010 PMCID: PMC11088232 DOI: 10.1016/j.crtox.2024.100170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/27/2024] [Accepted: 04/23/2024] [Indexed: 05/14/2024] Open
Abstract
The objective of the present narrative review was to synthesize existing clinical and epidemiological findings linking manganese (Mn) exposure biomarkers to autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD), and to discuss key pathophysiological mechanisms of neurodevelopmental disorders that may be affected by this metal. Existing epidemiological data demonstrated both direct and inverse association between Mn body burden and ASD, or lack of any relationship. In contrast, the majority of studies revealed significantly higher Mn levels in subjects with ADHD, as well as direct relationship between Mn body burden with hyperactivity and inattention scores in children, although several studies reported contradictory results. Existing laboratory studies demonstrated that impaired attention and hyperactivity in animals following Mn exposure was associated with dopaminergic dysfunction and neuroinflammation. Despite lack of direct evidence on Mn-induced neurobiological alterations in patients with ASD and ADHD, a plethora of studies demonstrated that neurotoxic effects of Mn overexposure may interfere with key mechanisms of pathogenesis inherent to these neurodevelopmental disorders. Specifically, Mn overload was shown to impair not only dopaminergic neurotransmission, but also affect metabolism of glutamine/glutamate, GABA, serotonin, noradrenaline, thus affecting neuronal signaling. In turn, neurotoxic effects of Mn may be associated with its ability to induce oxidative stress, apoptosis, and neuroinflammation, and/or impair neurogenesis. Nonetheless, additional detailed studies are required to evaluate the association between environmental Mn exposure and/or Mn body burden and neurodevelopmental disorders at a wide range of concentrations to estimate the potential dose-dependent effects, as well as environmental and genetic factors affecting this association.
Collapse
Affiliation(s)
- Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | | | - Anatoly V. Skalny
- Department of Medical Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
| | - Irina P. Zaitseva
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl 150003, Russia
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, USA
| | - Anatoly A. Kirichuk
- Department of Medical Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - Abel Santamaria
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Cuidado de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City 04960, Mexico
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059, USA
| | - Alexey A. Tinkov
- Department of Medical Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl 150003, Russia
| |
Collapse
|
3
|
Sande R, Doshi G, Godad A. Deciphering the role of metal and non-metals in the treatment of epilepsy. Neurochem Int 2023; 167:105536. [PMID: 37178926 DOI: 10.1016/j.neuint.2023.105536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/24/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023]
Abstract
Metals and non-metals have known to play a significant role in various physiological roles in the body including the central nervous system (CNS). The alterations in their concentration in the CNS leads to abnormalities in the normal functions which may lead to various neurological conditions including epilepsy. Manganese is a cofactor required for antioxidant enzymes such as Superoxide dismutase, Glutamine synthetase, etc. The accumulation of iron leads to formation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) which have the potential to cause ferroptosis, one of the reasons for epileptogenesis. Zinc has biphasic response, both neurotoxic and neuroprotective, based on concentration levels in the CNS. Selenium is a main element for selenoproteins which is responsible for the regulation of oxidative state and antioxidant defence mechanism. The reduction in the phosphorous levels in the CNS is widely observed after generalised tonic clonic seizures (GTC), which can be a potential diagnostic biomarker. Copper acts in the CNS in an identical manner, i.e., by blocking both AMPA mediated and GABA mediated neuronal transmission. Magnesium blocks calcium channels in the NMDA receptor and prevents glutamatergic transmission, thus inhibiting excitotoxicity. Lithium acts as a proconvulsive agent and is used in combination with pilocarpine to induce seizures. The identified potential of metals and non-metals in epilepsy can be utilised in order to devise new adjuvant therapies for the management of epilepsy. The article summaries in depth the role of metals and non-metals in the treatment of epilepsy supported with special paragraph on author perspective on to the topic. Furthermore, an update of preclinical and clinical evidences are discussed in the review to give evidence on metal and non-metal based therapies in epilepsy.
Collapse
Affiliation(s)
- Ruksar Sande
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| | - Angel Godad
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India.
| |
Collapse
|
4
|
Chin-Chan M, Montes S, Blanco-Álvarez VM, Aguirre-Alarcón HA, Hernández-Rodríguez I, Bautista E. Relevance of biometals during neuronal differentiation and myelination: in vitro and in vivo studies. Biometals 2022; 35:395-427. [DOI: 10.1007/s10534-022-00380-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 02/27/2022] [Indexed: 12/20/2022]
|
5
|
Szabó Z, Péter M, Héja L, Kardos J. Dual Role for Astroglial Copper-Assisted Polyamine Metabolism during Intense Network Activity. Biomolecules 2021; 11:604. [PMID: 33921742 PMCID: PMC8073386 DOI: 10.3390/biom11040604] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 12/29/2022] Open
Abstract
Astrocytes serve essential roles in human brain function and diseases. Growing evidence indicates that astrocytes are central players of the feedback modulation of excitatory Glu signalling during epileptiform activity via Glu-GABA exchange. The underlying mechanism results in the increase of tonic inhibition by reverse operation of the astroglial GABA transporter, induced by Glu-Na+ symport. GABA, released from astrocytes, is synthesized from the polyamine (PA) putrescine and this process involves copper amino oxidase. Through this pathway, putrescine can be considered as an important source of inhibitory signaling that counterbalances epileptic discharges. Putrescine, however, is also a precursor for spermine that is known to enhance gap junction channel communication and, consequently, supports long-range Ca2+ signaling and contributes to spreading of excitatory activity through the astrocytic syncytium. Recently, we presented the possibility of neuron-glia redox coupling through copper (Cu+/Cu2+) signaling and oxidative putrescine catabolism. In the current work, we explore whether the Cu+/Cu2+ homeostasis is involved in astrocytic control on neuronal excitability by regulating PA catabolism. We provide supporting experimental data underlying this hypothesis. We show that the blockade of copper transporter (CTR1) by AgNO3 (3.6 µM) prevents GABA transporter-mediated tonic inhibitory currents, indicating causal relationship between copper (Cu+/Cu2+) uptake and the catabolism of putrescine to GABA in astrocytes. In addition, we show that MnCl2 (20 μM), an inhibitor of the divalent metal transporter DMT1, also prevents the astrocytic Glu-GABA exchange. Furthermore, we observed that facilitation of copper uptake by added CuCl2 (2 µM) boosts tonic inhibitory currents. These findings corroborate the hypothesis that modulation of neuron-glia coupling by copper uptake drives putrescine → GABA transformation, which leads to subsequent Glu-GABA exchange and tonic inhibition. Findings may in turn highlight the potential role of copper signaling in fine-tuning the activity of the tripartite synapse.
Collapse
Affiliation(s)
- Zsolt Szabó
- Functional Pharmacology Research Group, Research Centre for Natural Sciences, Institute of Organic Chemistry, H-1117 Budapest, Hungary; (Z.S.); (M.P.); (J.K.)
| | - Márton Péter
- Functional Pharmacology Research Group, Research Centre for Natural Sciences, Institute of Organic Chemistry, H-1117 Budapest, Hungary; (Z.S.); (M.P.); (J.K.)
- Hevesy György Ph.D. School of Chemistry, ELTE Eötvös Loránd University, H-1117 Budapest, Hungary
| | - László Héja
- Functional Pharmacology Research Group, Research Centre for Natural Sciences, Institute of Organic Chemistry, H-1117 Budapest, Hungary; (Z.S.); (M.P.); (J.K.)
| | - Julianna Kardos
- Functional Pharmacology Research Group, Research Centre for Natural Sciences, Institute of Organic Chemistry, H-1117 Budapest, Hungary; (Z.S.); (M.P.); (J.K.)
| |
Collapse
|
6
|
Totten MS, Pierce DM, Erikson KM. Diet-Induced Obesity Disrupts Trace Element Homeostasis and Gene Expression in the Olfactory Bulb. Nutrients 2020; 12:E3909. [PMID: 33371327 PMCID: PMC7767377 DOI: 10.3390/nu12123909] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to determine the impact of diet-induced obesity (DIO) on trace element homeostasis and gene expression in the olfactory bulb and to identify potential interaction effects between diet, sex, and strain. Our study is based on evidence that obesity and olfactory bulb impairments are linked to neurodegenerative processes. Briefly, C57BL/6J (B6J) and DBA/2J (D2J) male and female mice were fed either a low-fat diet or a high-fat diet for 16 weeks. Brain tissue was then evaluated for iron, manganese, copper, and zinc concentrations and mRNA gene expression. There was a statistically significant diet-by-sex interaction for iron and a three-way interaction between diet, sex, and strain for zinc in the olfactory bulb. Obese male B6J mice had a striking 75% increase in iron and a 50% increase in manganese compared with the control. There was an increase in zinc due to DIO in B6J males and D2J females, but a decrease in zinc in B6J females and D2J males. Obese male D2J mice had significantly upregulated mRNA gene expression for divalent metal transporter 1, alpha-synuclein, amyloid precursor protein, dopamine receptor D2, and tyrosine hydroxylase. B6J females with DIO had significantly upregulated brain-derived neurotrophic factor expression. Our results demonstrate that DIO has the potential to disrupt trace element homeostasis and mRNA gene expression in the olfactory bulb, with effects that depend on sex and genetics. We found that DIO led to alterations in iron and manganese predominantly in male B6J mice, and gene expression dysregulation mainly in male D2J mice. These results have important implications for health outcomes related to obesity with possible connections to neurodegenerative disease.
Collapse
Affiliation(s)
- Melissa S. Totten
- Department of Nutrition, School of Health and Human Sciences, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (D.M.P.); (K.M.E.)
| | | | | |
Collapse
|
7
|
Kazimierska K, Biel W, Witkowicz R. Mineral Composition of Cereal and Cereal-Free Dry Dog Foods versus Nutritional Guidelines. Molecules 2020; 25:E5173. [PMID: 33172044 PMCID: PMC7664208 DOI: 10.3390/molecules25215173] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 12/18/2022] Open
Abstract
The aims of the present work are to estimate the nutritional value and to evaluate and compare the levels of macroelements (Ca, P, K, Na, Mg), microelements (Fe, Zn, Mn, Cu), heavy metals (Co, Cd, Pb, Mo, Cr, Ni), and their ratios in extruded complete foods for adult dogs, their compatibility with nutritional guidelines, as well as food profile similarity. Basic composition was determined according to Association of Official Analytical Chemists (AOAC). Analyses for elements were performed using an atomic absorption spectrometer. All the evaluated dry dog foods met the minimum recommended levels for protein and fat. Eighteen tested dog foods (60%) did not meet at least one recommendation of nutritional guidelines. Four dog foods exceeded the legal limit of Fe and five foods exceeded the legal limit of Zn; in one of them, Zn level was almost twice higher. Dog foods with insect protein exceeded the legal limit for Mn content. Eight dog foods had an inappropriate Ca:P ratio. Heavy metals were below detection limit in all analyzed dog foods. The results seem to show the need for regular feed analyses of the elemental composition in raw materials before introducing supplementation and for the monitoring of the mineral composition of finished pet food.
Collapse
Affiliation(s)
- Katarzyna Kazimierska
- Department of Monogastric Animal Sciences, Division of Animal Nutrition and Food, West Pomeranian University of Technology in Szczecin, 29 Klemensa Janickiego, 71270 Szczecin, Poland;
| | - Wioletta Biel
- Department of Monogastric Animal Sciences, Division of Animal Nutrition and Food, West Pomeranian University of Technology in Szczecin, 29 Klemensa Janickiego, 71270 Szczecin, Poland;
| | - Robert Witkowicz
- Department of Agroecology and Crop Production, University of Agriculture in Krakow, 21 Mickiewicza, 31120 Krakow, Poland;
| |
Collapse
|
8
|
Kissani N, Naji Y, Mebrouk Y, Chraa M, Ghanima A. Parkinsonism and chronic manganese exposure: Pilot study with clinical, environmental and experimental evidence. Clin Park Relat Disord 2020; 3:100057. [PMID: 34316640 PMCID: PMC8298767 DOI: 10.1016/j.prdoa.2020.100057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 04/21/2020] [Accepted: 05/01/2020] [Indexed: 11/17/2022] Open
Abstract
Parkinsonism related to chronic Manganese exposure is notably due to focal lesions of the basal ganglia. Our study focused on epidemiological, clinical, toxicological and experimental aspects of Manganese-induced Parkinsonism in south of Morocco. It is a prospective study concerning the workers and the residents bordering on the 2 mines in the south of Morocco. The results of the study concerned 120 cases divided into 4 groups of patients: G1: 30 cases exposed to different incriminated toxic products, which present Parkinsonian signs, G2: 30 cases healthy and exposed, G3: 30 cases affected with Idiopathic Parkinson's disease, and G4: 30 cases healthy and unexposed (controls). The results from the first mine show that 5.7% of the sample developed Manganese-Induced Parkinsonism and this percentage is slightly higher (4.5%) than the second mine site. Chemical and biological analysis revealed high levels of Manganese. The majority of patients did not improve the clinical signs under L-dopa treatment. The authors underline the gravity of Manganese-induced Parkinsonism and propose a listing of the various exposures as well as a cartography of the regions of risk in Morocco. The objective of this study was to update the data about Manganese-Induced Parkinsonism in the South of Morocco. Multidisciplinary study putting together clinicians, biologists, biochemists, and pathologists to map accurately areas of exposure to heavy metals. Specific considerations should be taken concerning miners by improving work conditions and obliging the use of protective equipment. General approach on primary prevention for the population living near the mines in the way to decrease the number of cases.
Collapse
Affiliation(s)
- Najib Kissani
- Clinical and Experimental Neuroscience Research Laboratory, Faculty of Medicine, Cadi Ayyad University, Marrakech, Morocco
- Neurology department, University Teaching Hospital Mohammed VI, Marrakesh, Morocco
- Corresponding author at: Neurology Department, University Hospital Mohamed VI, Marrakech, Morocco.
| | - Yahya Naji
- Clinical and Experimental Neuroscience Research Laboratory, Faculty of Medicine, Cadi Ayyad University, Marrakech, Morocco
| | - Yassine Mebrouk
- Neurology Department, University Hospital Mohammed VI, Oujda, Morocco
| | - Mohamed Chraa
- Clinical and Experimental Neuroscience Research Laboratory, Faculty of Medicine, Cadi Ayyad University, Marrakech, Morocco
| | - Abderrazzak Ghanima
- Laboratory of Bio-Organic and Macromolecular Chemistry, Department of Biology, Faculty of Sciences and Techniques, Marrakech, Morocco
| |
Collapse
|
9
|
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
|
10
|
Ommati MM, Heidari R, Ghanbarinejad V, Abdoli N, Niknahad H. Taurine Treatment Provides Neuroprotection in a Mouse Model of Manganism. Biol Trace Elem Res 2019; 190:384-395. [PMID: 30357569 DOI: 10.1007/s12011-018-1552-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022]
Abstract
Manganese (Mn) is a trace element involved in many physiological processes. However, excessive Mn exposure leads to neurological complications. Although no precise mechanism(s) has been found for Mn-induced neurotoxicity, oxidative stress and mitochondrial injury seem to play a relevant role in this complication. On the other hand, there is no protective strategy against Mn neurotoxicity so far. Taurine is an amino acid with significant neuroprotective properties. The current study was designed to evaluate the effect of taurine supplementation and its potential mechanism(s) of action in a mouse model of manganism. Animals were treated with Mn (100 mg/kg, s.c) alone and/or in combination with taurine (50, 100, and 500 mg/kg, i.p, for eight consecutive days). Severe locomotor dysfunction along with a significant elevation in brain tissue biomarkers of oxidative stress was evident in Mn-exposed mice. On the other hand, it was revealed that mitochondrial indices of functionality were hampered in Mn-treated animals. Taurine supplementation (50, 100, and 500 mg/kg, i.p) alleviated Mn-induced locomotor deficit. Moreover, this amino acid mitigated oxidative stress biomarkers and preserved brain tissue mitochondrial indices of functionality. These data introduce taurine as a potential neuroprotective agent against Mn neurotoxicity. Antioxidative and mitochondria protecting effects of taurine might play a fundamental role in its neuroprotective properties against Mn toxicity.
Collapse
Affiliation(s)
- Mohammad Mehdi Ommati
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P. O. Box 158371345, Roknabad, Karafarin St., Shiraz, Fars, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P. O. Box 158371345, Roknabad, Karafarin St., Shiraz, Fars, Iran.
| | - Vahid Ghanbarinejad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P. O. Box 158371345, Roknabad, Karafarin St., Shiraz, Fars, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Abdoli
- Iran Food and Drug Administration (IFDA), Ministry of Health, Tehran, Iran
| | - Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P. O. Box 158371345, Roknabad, Karafarin St., Shiraz, Fars, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
11
|
Lasley SM. The Use of Intracerebral Microdialysis to Elucidate Environmentally Induced Neurotoxic Mechanisms. CURRENT PROTOCOLS IN TOXICOLOGY 2019; 80:e72. [PMID: 30939232 PMCID: PMC6615941 DOI: 10.1002/cptx.72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The technique of microdialysis permits the assessment of neurotransmitter activity and the monitoring of other cellular entities in tissue extracellular fluid. The method is widely used for quantifying biogenic amine and amino acid transmitters, peptides, administered drugs, and other molecules in response to various experimental treatments. This article provides an overview of the manner in which the methodology of intracerebral microdialysis is utilized in the field of neurotoxicology to elucidate the actions of environmental agents. The technique is employed in a variety of creative ways to address specific experimental goals involving myriad toxicants. With appropriate consideration of method parameters, investigators have also been able to address mechanistic issues in their studies. These investigations consist of sampling of neurotransmitters in extracellular fluid after various protocols of environmental metal exposure as well as assessments of blood-brain barrier permeability, the detection of reactive oxygen species, and description of the toxicodynamics of environmental agents. The purpose of this examination is not to review the investigational findings, per se, but to highlight the various approaches utilized with this methodology and the experimental questions that have been addressed. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Stephen M Lasley
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, Illinois
| |
Collapse
|
12
|
Wang G, Liu Q, Guo L, Zeng H, Ding C, Zhang W, Xu D, Wang X, Qiu J, Dong Q, Fan Z, Zhang Q, Pan J. Gut Microbiota and Relevant Metabolites Analysis in Alcohol Dependent Mice. Front Microbiol 2018; 9:1874. [PMID: 30158912 PMCID: PMC6104187 DOI: 10.3389/fmicb.2018.01874] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/25/2018] [Indexed: 12/21/2022] Open
Abstract
Alcohol abuse is a major public health crisis. Relative evidences supported that the gut microbiota (GM) played an important role in central nervous system (CNS) function, and the composition of them had changed after alcohol drinking. We sought to explore the changes of GM in alcohol dependence. In our study, the GM of mice with alcohol administration was detected through analyzed 16S rRNA gene sequencing and the fecal metabolites were analyzed by LC-MS. The microbial diversity was significantly higher in the alcohol administration group, the abundance of phylum Firmicutes and its class Clostridiales were elevated, meanwhile the abundance of Lachnospiraceae, Alistipes, and Odoribacter showed significant differences among the three groups. Based on LC-MS results, bile acid, secondary bile acid, serotonin and taurine level had varying degrees of changes in alcohol model. From paraffin sections, tissue damage was observed in liver and colon. These findings provide direct evidence that alcohol intake affects the composition of GM, enable a better understanding of the function of GM in the microbiota-gut-brain (MGB) axis, and give a new thought for alcohol addiction treatment.
Collapse
Affiliation(s)
- Guanhao Wang
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Qing Liu
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Liang Guo
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Haijuan Zeng
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Chengchao Ding
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Wentong Zhang
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Dongpo Xu
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Xiang Wang
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jingxuan Qiu
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Qingli Dong
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Ziquan Fan
- Thermo Fisher Scientific, Shanghai, China
| | - Qi Zhang
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jing Pan
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
13
|
Ben-Shahar Y. The Impact of Environmental Mn Exposure on Insect Biology. Front Genet 2018; 9:70. [PMID: 29545824 PMCID: PMC5837978 DOI: 10.3389/fgene.2018.00070] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/16/2018] [Indexed: 01/18/2023] Open
Abstract
Manganese (Mn) is an essential trace element that acts as a metal co-factor in diverse biochemical and cellular functions. However, chronic environmental exposure to high levels of Mn is a well-established risk factor for the etiology of severe, atypical parkinsonian syndrome (manganism) via its accumulation in the basal ganglia, pallidum, and striatum brain regions, which is often associated with abnormal dopamine, GABA, and glutamate neural signaling. Recent studies have indicated that chronic Mn exposure at levels that are below the risk for manganism can still cause behavioral, cognitive, and motor dysfunctions via poorly understood mechanisms at the molecular and cellular levels. Furthermore, in spite of significant advances in understanding Mn-induced behavioral and neuronal pathologies, available data are primarily for human and rodents. In contrast, the possible impact of environmental Mn exposure on brain functions and behavior of other animal species, especially insects and other invertebrates, remains mostly unknown both in the laboratory and natural habitats. Yet, the effects of environmental exposure to metals such as Mn on insect development, physiology, and behavior could also have major indirect impacts on human health via the long-term disruptions of food webs, as well as direct impact on the economy because of the important role insects play in crop pollination. Indeed, laboratory and field studies indicate that chronic exposures to metals such as Mn, even at levels that are below what is currently considered toxic, affect the dopaminergic signaling pathway in the insect brain, and have a major impact on the behavior of insects, including foraging activity of important pollinators such as the honey bee. Together, these studies highlight the need for a better understanding of the neuronal, molecular, and genetic processes that underlie the toxicity of Mn and other metal pollutants in diverse animal species, including insects.
Collapse
Affiliation(s)
- Yehuda Ben-Shahar
- Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
14
|
Marreilha dos Santos AP, Andrade V, Aschner M. Neuroprotective and Therapeutic Strategies for Manganese-Induced Neurotoxicity. CLINICAL PHARMACOLOGY AND TRANSLATIONAL MEDICINE 2017; 1:54-62. [PMID: 30854510 PMCID: PMC6402347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Manganese (Mn) is an essential element required for growth, development and general maintenance of health. However, chronic or high occupational and environmental exposure to excessive levels of Mn has long been known to lead to a progressive neurological disorder similar to Parkinsonism. Manganism patients display a variety of symptoms, including mental, cognitive and behavioural impediments, as well as motor dysfunctions that are associated with basal ganglia dysfunction. Taking into account the pharmacokinetics and Mn-related toxicity mechanisms, several neuroprotective compounds and therapeutic approaches have been investigated to assess their efficacy in mitigating its neurotoxicity. Here, we will briefly address some of the toxic mechanisms of Mn, followed by neuroprotective strategies and therapeutic approaches aiming to reduce or treat Mn induced neurotoxicity. Natural and synthetic antioxidants, anti-inflammatory compounds, ATP/ADP ratio protectors and glutamate protectors have been introduced in view of decreasing Mn-induced neurotoxicity. In addition, the efficacy and mechanisms of several therapeutic interventions such as levodopa, ethylene-diamine-tetraacetic acid (EDTA) and para-aminosalicylic acid (PAS), aimed at ameliorating Mn neurotoxic symptoms in humans, will be reviewed.
Collapse
Affiliation(s)
- AP Marreilha dos Santos
- Institute of Medicine Research (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon. Portugal
| | - V Andrade
- Institute of Medicine Research (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon. Portugal
| | - M Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, USA
| |
Collapse
|
15
|
Bouabid S, Tinakoua A, Lakhdar-Ghazal N, Benazzouz A. Manganese neurotoxicity: behavioral disorders associated with dysfunctions in the basal ganglia and neurochemical transmission. J Neurochem 2015; 136:677-691. [PMID: 26608821 DOI: 10.1111/jnc.13442] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 10/24/2015] [Accepted: 11/10/2015] [Indexed: 11/30/2022]
Abstract
Manganese (Mn) is an essential element required for many physiological functions. While it is essential at physiological levels, excessive accumulation of Mn in the brain causes severe dysfunctions in the central nervous system known as manganism. Manganism is an extrapyramidal disorder characterized by motor disturbances associated with neuropsychiatric and cognitive disabilities similar to Parkinsonism. As the primary brain regions targeted by Mn are the basal ganglia, known to be involved in the pathophysiology of extrapyramidal disorders, this review will examine the impact of Mn exposure on the basal ganglia circuitry and neurotransmitters in relation to motor and non-motor disorders. The collected data from recent available studies in humans and experimental animal models provide new information about the mechanisms by which Mn affects behavior, neurotransmitters, and basal ganglia function observed in manganism. The effects of the alterations of metals on basal ganglia and neurochemical functioning are critical to develop effective modalities not only for the treatment of vulnerable populations (e.g., Mn-exposed workers) but also for understanding the etiology of neurodegenerative diseases where brain metal imbalances are involved, such as Parkinson's disease. We examine the impact of manganese (Mn) exposure on the basal ganglia circuitry and neurotransmitters in relation with motor and non-motor disorders. The collected data from available studies show that when accumulated in the globus pallidus, Mn influences the subthalamic (STN) and substantia nigra (SN) neurons, which are at the origin of changes in the thalamus and the cortex.
Collapse
Affiliation(s)
- Safa Bouabid
- University de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,Université Mohammed V, Faculté des Sciences, Equipe Rythmes Biologiques, Neurosciences et Environnement, Rabat, Morocco
| | - Anass Tinakoua
- University de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,Université Mohammed V, Faculté des Sciences, Equipe Rythmes Biologiques, Neurosciences et Environnement, Rabat, Morocco
| | - Nouria Lakhdar-Ghazal
- Université Mohammed V, Faculté des Sciences, Equipe Rythmes Biologiques, Neurosciences et Environnement, Rabat, Morocco
| | - Abdelhamid Benazzouz
- University de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| |
Collapse
|
16
|
Abstract
The understanding of manganese (Mn) biology, in particular its cellular regulation and role in neurological disease, is an area of expanding interest. Mn is an essential micronutrient that is required for the activity of a diverse set of enzymatic proteins (e.g., arginase and glutamine synthase). Although necessary for life, Mn is toxic in excess. Thus, maintaining appropriate levels of intracellular Mn is critical. Unlike other essential metals, cell-level homeostatic mechanisms of Mn have not been identified. In this review, we discuss common forms of Mn exposure, absorption, and transport via regulated uptake/exchange at the gut and blood-brain barrier and via biliary excretion. We present the current understanding of cellular uptake and efflux as well as subcellular storage and transport of Mn. In addition, we highlight the Mn-dependent and Mn-responsive pathways implicated in the growing evidence of its role in Parkinson's disease and Huntington's disease. We conclude with suggestions for future focuses of Mn health-related research.
Collapse
Affiliation(s)
- Kyle J Horning
- Department of Neurology, Vanderbilt University, Nashville, Tennessee 37232; , ,
| | | | | | | | | |
Collapse
|
17
|
Lu CL, Tang S, Meng ZJ, He YY, Song LY, Liu YP, Ma N, Li XY, Guo SC. Taurine improves the spatial learning and memory ability impaired by sub-chronic manganese exposure. J Biomed Sci 2014; 21:51. [PMID: 24885898 PMCID: PMC4045917 DOI: 10.1186/1423-0127-21-51] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 05/18/2014] [Indexed: 11/29/2022] Open
Abstract
Background Excessive manganese exposure induced cognitive deficit. Several lines of evidence have demonstrated that taurine improves cognitive impairment induced by numerous neurotoxins. However, the role of taurine on manganese-induced damages in learning and memory is still elusive. This goal of this study was to investigate the beneficial effect of taurine on learning and memory capacity impairment by manganese exposure in an animal model. Results The escape latency in the Morris Water Maze test was significantly longer in the rats injected with manganese than that in the rats received both taurine and manganese. Similarly, the probe trial showed that the annulus crossings were significantly greater in the taurine plus manganese treated rats than those in the manganese-treated rats. However, the blood level of manganese was not altered by the taurine treatment. Interestingly, the exposure of manganese led to a significant increase in the acetylcholinesterase activity and an evidently decrease in the choline acetyltransferase activity, which were partially restored by the addition of taurine. Additionally, we identified 9 differentially expressed proteins between the rat hippocampus treated by manganese and the control or the manganese plus taurine in the proteomic analysis using the 2-dimensional gel electrophoresis followed by the tandem mass spectrometry (MS/MS). Most of these proteins play a role in energy metabolism, oxidative stress, inflammation, and neuron synapse. Conclusions In summary, taurine restores the activity of AChE and ChAT, which are critical for the regulation of acetylcholine. We have identified seven differentially expressed proteins specifically induced by manganese and two proteins induced by taurine from the rat hippocampus. Our results support that taurine improves the impaired learning and memory ability caused by excessive exposure of manganese.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xi-Yi Li
- Department of Food and Nutrition, School of Public Health, Guangxi Medical University, 22 Shuangyong Road, 530021 Nanning, Guangxi, P,R, China.
| | | |
Collapse
|
18
|
Fordahl SC, Erikson KM. Manganese accumulation in membrane fractions of primary astrocytes is associated with decreased γ-aminobutyric acid (GABA) uptake, and is exacerbated by oleic acid and palmitate. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 37:1148-1156. [PMID: 24814258 DOI: 10.1016/j.etap.2014.03.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 03/20/2014] [Accepted: 03/23/2014] [Indexed: 06/03/2023]
Abstract
Manganese (Mn) exposure interferes with GABA uptake; however, the effects of Mn on GABA transport proteins (GATs) have not been identified. We sought to characterize how Mn impairs GAT function in primary rat astrocytes. Astrocytes exposed to Mn (500 μM) had significantly reduced (3)H-GABA uptake despite no change in membrane or cytosolic GAT3 protein levels. Co-treatment with 100 μM oleic or palmitic acids (both known to be elevated in Mn neurotoxicity), exacerbated the Mn-induced decline in (3)H-GABA uptake. Mn accumulation in the membrane fraction of astrocytes was enhanced with fatty acid administration, and was negatively correlated with (3)H-GABA uptake. Furthermore, control cells exposed to Mn only during the experimental uptake had significantly reduced (3)H-GABA uptake, and the addition of GABA (50 μM) blunted cytosolic Mn accumulation. These data indicate that reduced GAT function in astrocytes is influenced by Mn and fatty acids accumulating at or interacting with the plasma membrane.
Collapse
Affiliation(s)
- Steve C Fordahl
- Department of Nutrition, University of North Carolina at Greensboro, 318 Stone Building, P.O. Box 26170, Greensboro, NC 27402-6170, United States.
| | - Keith M Erikson
- Department of Nutrition, University of North Carolina at Greensboro, 318 Stone Building, P.O. Box 26170, Greensboro, NC 27402-6170, United States.
| |
Collapse
|
19
|
Kinawy AA, Ezzat AR, Al-Suwaigh BR. Inhalation of air polluted with gasoline vapours alters the levels of amino acid neurotransmitters in the cerebral cortex, hippocampus, and hypothalamus of the rat. ACTA ACUST UNITED AC 2014; 66:219-24. [PMID: 24690269 DOI: 10.1016/j.etp.2014.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 01/27/2014] [Accepted: 02/07/2014] [Indexed: 01/25/2023]
Abstract
BACKGROUND This study was designed to investigate the impact of exposure to the vapours of two kinds of gasoline, a widely used fuel for the internal combustion engines on the levels of the amino acid neurotransmitters of the rat brain. Recent studies provide strong evidence for a causative role for traffic-related air pollution on morbidity outcomes as well as premature death (Health Effects Institute, 2009; Levy et al., 2010; von Stackelberg et al., 2013). Exposure to the vapours of gasoline or its constituents may be accidental, occupational by workers at fuel stations and factories, or through abuse as a mean of mood alteration (Fortenberry, 1985; Mc Garvey et al., 1999). Two kinds of gasoline that are common in Egypt have been used in this study. The first contains octane enhancers in the form of lead derivatives (leaded gasoline; G1) and the other contains methyl-tertiary butyl ether (MTBE) as the octane enhancer (unleaded gasoline; G2). The levels of the major excitatory (aspartic acid and glutamic acid) and the inhibitory (GABA and glycine) amino acid neurotransmitters were determined in the cerebral cortex, hippocampus, and hypothalamus. RESULTS The current study revealed that the acute inhalation of air polluted with the two types of gasoline vapours (1/2 LC50 for 30 min) induced elevation in the levels of aspartic and glutamic acids along with a decrease in glycine and GABA in most studied brain areas. Chronic inhalation of both types of gasoline (a single daily 30-min session of 1/5 LC50 for 60 days) caused a significant increase in the aspartic and glutamic acid concentrations of the hippocampus without affecting the levels of GABA or glycine. CONCLUSION Acute and chronic inhalation of either one of G1 and G2 vapours induced a disturbance and fluctuation in the levels of the free amino acids that act as excitatory and inhibitory neurotransmitters in the brain areas under investigation. These neurotransmitters are fundamental for the communicative functioning of the neurons and such effects may have a profound impact on the cognitive and sensorimotor functions of the brain resulting in serious psychological and physiological disorders..
Collapse
Affiliation(s)
- Amal A Kinawy
- Biology Department, College of Science, Dammam University, Postal Code 31113, P.O. Box 838, Dammam, Saudi Arabia.
| | - Ahmed R Ezzat
- Zoology Department, Faculty of Science, Ain Shams University, Postal Code 11566, Cairo, Egypt.
| | - Badryah R Al-Suwaigh
- Biology Department, College of Science, Dammam University, Postal Code 31113, P.O. Box 838, Dammam, Saudi Arabia.
| |
Collapse
|
20
|
Thalamic GABA predicts fine motor performance in manganese-exposed smelter workers. PLoS One 2014; 9:e88220. [PMID: 24505436 PMCID: PMC3913772 DOI: 10.1371/journal.pone.0088220] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 01/05/2014] [Indexed: 02/02/2023] Open
Abstract
Overexposure to manganese (Mn) may lead to parkinsonian symptoms including motor deficits. The main inhibitory neurotransmitter gamma-aminobutyric acid (GABA) is known to play a pivotal role in the regulation and performance of movement. Therefore this study was aimed at testing the hypothesis that an alteration of GABA following Mn exposure may be associated with fine motor performance in occupationally exposed workers and may underlie the mechanism of Mn-induced motor deficits. A cohort of nine Mn-exposed male smelter workers from an Mn-iron alloy factory and 23 gender- and age-matched controls were recruited and underwent neurological exams, magnetic resonance spectroscopy (MRS) measurements, and Purdue pegboard motor testing. Short-echo-time MRS was used to measure N-Acetyl-aspartate (NAA) and myo-inositol (mI). GABA was detected with a MEGA-PRESS J-editing MRS sequence. The mean thalamic GABA level was significantly increased in smelter workers compared to controls (p = 0.009). Multiple linear regression analysis reveals (1) a significant association between the increase in GABA level and the duration of exposure (R2 = 0.660, p = 0.039), and (2) significant inverse associations between GABA levels and all Purdue pegboard test scores (for summation of all scores R2 = 0.902, p = 0.001) in the smelter workers. In addition, levels of mI were reduced significantly in the thalamus and PCC of smelter workers compared to controls (p = 0.030 and p = 0.009, respectively). In conclusion, our results show clear associations between thalamic GABA levels and fine motor performance. Thus in Mn-exposed subjects, increased thalamic GABA levels may serve as a biomarker for subtle deficits in motor control and may become valuable for early diagnosis of Mn poisoning.
Collapse
|
21
|
Vorhees CV, Graham DL, Amos-Kroohs RM, Braun AA, Grace CE, Schaefer TL, Skelton MR, Erikson KM, Aschner M, Williams MT. Effects of developmental manganese, stress, and the combination of both on monoamines, growth, and corticosterone. Toxicol Rep 2014; 1:1046-1061. [PMID: 25574457 PMCID: PMC4285371 DOI: 10.1016/j.toxrep.2014.10.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Developmental exposure to manganese (Mn) or stress can each be detrimental to brain development. Here, Sprague-Dawley rats were exposed to two housing conditions and Mn from postnatal day (P)4–28. Within each litter two males and two females were assigned to the following groups: 0 (vehicle), 50, or 100 mg/kg Mn by gavage every other day. Half the litters were reared in cages with standard bedding and half with no bedding. One pair/group in each litter had an acute shallow water stressor before tissue collection (i.e., standing in shallow water). Separate litters were assessed at P11, 19, or 29. Mn-treated rats raised in standard cages showed no change in baseline corticosterone but following acute stress increased more than controls on P19; no Mn effects were seen on P11 or P29. Mn increased neostriatal dopamine in females at P19 and norepinephrine at P11 and P29. Mn increased hippocampal dopamine at P11 and P29 and 5-HT at P29 regardless of housing or sex. Mn had no effect on hypothalamic dopamine, but increased norepinephrine in males at P29 and 5-HT in males at all ages irrespective of rearing condition. Barren reared rats showed no or opposite effects of Mn, i.e., barren rearing + Mn attenuated corticosterone increases to acute stress. Barren rearing also altered the Mn-induced changes in dopamine and norepinephrine in the neostriatum, but not in the hippocampus. Barren rearing caused a Mn-associated increase in hypothalamic dopamine at P19 and P29 not seen in standard reared Mn-treated groups. Developmental Mn alters monoamines and corticosterone as a function of age, stress (acute and chronic), and sex.
Collapse
Affiliation(s)
- Charles V. Vorhees
- Department of Pediatrics, Division of Neurology, Cincinnati Children's Research Foundation and University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Corresponding authors. Tel.: +513 636 8624 MTW; 513 636 8622 CVV; fax: +513 636 3912
| | - Devon L. Graham
- Department of Pediatrics, Division of Neurology, Cincinnati Children's Research Foundation and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Robyn M. Amos-Kroohs
- Department of Pediatrics, Division of Neurology, Cincinnati Children's Research Foundation and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Amanda A. Braun
- Department of Pediatrics, Division of Neurology, Cincinnati Children's Research Foundation and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Curtis E. Grace
- Department of Pediatrics, Division of Neurology, Cincinnati Children's Research Foundation and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tori L. Schaefer
- Department of Pediatrics, Division of Neurology, Cincinnati Children's Research Foundation and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Matthew R. Skelton
- Department of Pediatrics, Division of Neurology, Cincinnati Children's Research Foundation and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Keith M. Erikson
- Department of Nutrition, University of North Carolina, Greensboro, NC, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael T. Williams
- Department of Pediatrics, Division of Neurology, Cincinnati Children's Research Foundation and University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Corresponding authors. Tel.: +513 636 8624 MTW; 513 636 8622 CVV; fax: +513 636 3912
| |
Collapse
|
22
|
Martinez-Finley EJ, Gavin CE, Aschner M, Gunter TE. Manganese neurotoxicity and the role of reactive oxygen species. Free Radic Biol Med 2013; 62:65-75. [PMID: 23395780 PMCID: PMC3713115 DOI: 10.1016/j.freeradbiomed.2013.01.032] [Citation(s) in RCA: 210] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 01/25/2013] [Accepted: 01/28/2013] [Indexed: 12/21/2022]
Abstract
Manganese (Mn) is an essential dietary nutrient, but an excess or accumulation can be toxic. Disease states, such as manganism, are associated with overexposure or accumulation of Mn and are due to the production of reactive oxygen species, free radicals, and toxic metabolites; alteration of mitochondrial function and ATP production; and depletion of cellular antioxidant defense mechanisms. This review focuses on all of the preceding mechanisms and the scientific studies that support them as well as providing an overview of the absorption, distribution, and excretion of Mn and the stability and transport of Mn compounds in the body.
Collapse
Affiliation(s)
- Ebany J Martinez-Finley
- Division of Clinical Pharmacology and Pediatric Toxicology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37240, USA; Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | | | - Michael Aschner
- Division of Clinical Pharmacology and Pediatric Toxicology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37240, USA; Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN 37240, USA; Center for Molecular Neuroscience, Vanderbilt University Medical Center, Nashville, TN 37240, USA; The Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, TN 37240, USA.
| | - Thomas E Gunter
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| |
Collapse
|
23
|
Abstract
Manganese (Mn) is an essential trace metal that is pivotal for normal cell function and metabolism. Its homeostasis is tightly regulated; however, the mechanisms of Mn homeostasis are poorly characterized. While a number of proteins such as the divalent metal transporter 1, the transferrin/transferrin receptor complex, the ZIP family metal transporters ZIP-8 and ZIP-14, the secretory pathway calcium ATPases SPCA1 and SPCA2, ATP13A2, and ferroportin have been suggested to play a role in Mn transport, the degree that each of them contributes to Mn homeostasis has still to be determined. The recent discovery of SLC30A10 as a crucial Mn transporter in humans has shed further light on our understanding of Mn transport across the cell. Although essential, Mn is toxic at high concentrations. Mn neurotoxicity has been attributed to impaired dopaminergic (DAergic), glutamatergic and GABAergic transmission, mitochondrial dysfunction, oxidative stress, and neuroinflammation. As a result of preferential accumulation of Mn in the DAergic cells of the basal ganglia, particularly the globus pallidus, Mn toxicity causes extrapyramidal motor dysfunction. Firstly described as "manganism" in miners during the nineteenth century, this movement disorder resembles Parkinson's disease characterized by hypokinesia and postural instability. To date, a variety of acquired causes of brain Mn accumulation can be distinguished from an autosomal recessively inherited disorder of Mn metabolism caused by mutations in the SLC30A10 gene. Both, acquired and inherited hypermanganesemia, lead to Mn deposition in the basal ganglia associated with pathognomonic magnetic resonance imaging appearances of hyperintense basal ganglia on T1-weighted images. Current treatment strategies for Mn toxicity combine chelation therapy to reduce the body Mn load and iron (Fe) supplementation to reduce Mn binding to proteins that interact with both Mn and Fe. This chapter summarizes our current understanding of Mn homeostasis and the mechanisms of Mn toxicity and highlights the clinical disorders associated with Mn neurotoxicity.
Collapse
Affiliation(s)
- Karin Tuschl
- Clinical and Molecular Genetics Unit, UCL Institute of Child Health, London, United Kingdom.
| | | | | |
Collapse
|
24
|
Ripps H, Shen W. Review: taurine: a "very essential" amino acid. Mol Vis 2012; 18:2673-86. [PMID: 23170060 PMCID: PMC3501277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 11/08/2012] [Indexed: 11/22/2022] Open
Abstract
Taurine is an organic osmolyte involved in cell volume regulation, and provides a substrate for the formation of bile salts. It plays a role in the modulation of intracellular free calcium concentration, and although it is one of the few amino acids not incorporated into proteins, taurine is one of the most abundant amino acids in the brain, retina, muscle tissue, and organs throughout the body. Taurine serves a wide variety of functions in the central nervous system, from development to cytoprotection, and taurine deficiency is associated with cardiomyopathy, renal dysfunction, developmental abnormalities, and severe damage to retinal neurons. All ocular tissues contain taurine, and quantitative analysis of ocular tissue extracts of the rat eye revealed that taurine was the most abundant amino acid in the retina, vitreous, lens, cornea, iris, and ciliary body. In the retina, taurine is critical for photoreceptor development and acts as a cytoprotectant against stress-related neuronal damage and other pathological conditions. Despite its many functional properties, however, the cellular and biochemical mechanisms mediating the actions of taurine are not fully known. Nevertheless, considering its broad distribution, its many cytoprotective attributes, and its functional significance in cell development, nutrition, and survival, taurine is undoubtedly one of the most essential substances in the body. Interestingly, taurine satisfies many of the criteria considered essential for inclusion in the inventory of neurotransmitters, but evidence of a taurine-specific receptor has yet to be identified in the vertebrate nervous system. In this report, we present a broad overview of the functional properties of taurine, some of the consequences of taurine deficiency, and the results of studies in animal models suggesting that taurine may play a therapeutic role in the management of epilepsy and diabetes.
Collapse
Affiliation(s)
- Harris Ripps
- Departments of Ophthalmology and Visual Science, Anatomy and Cell Biology, Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| | | |
Collapse
|
25
|
Abstract
Manganese (Mn(2+))-enhanced magnetic resonance imaging studies of the neuronal pathways of the hypothalamus showed that information about the regulation of food intake and energy balance circulate through specific hypothalamic nuclei. The dehydration-induced anorexia (DIA) model demonstrated to be appropriate for studying the hypothalamus with Mn(2+)-enhanced magnetic resonance imaging. Manganese is involved in the normal functioning of a variety of physiological processes and is associated with enzymes contributing to neurotransmitter synthesis and metabolism. It also induces psychiatric and motor disturbances. The molecular mechanisms by which Mn(2+) produces alterations of the hypothalamic physiological processes are not well understood. (1)H-magnetic resonance spectroscopy measurements of the rodent hypothalamus are challenging due to the distant location of the hypothalamus resulting in limited measurement sensitivity. The present study proposed to investigate the effects of Mn(2+) on the neurochemical profile of the hypothalamus in normal, DIA, and overnight fasted female rats at 14.1 T. Results provide evidence that γ-aminobutyric acid has an essential role in the maintenance of energy homeostasis in the hypothalamus but is not condition specific. On the contrary, glutamine, glutamate, and taurine appear to respond more accurately to Mn(2+) exposure. An increase in glutamine levels could also be a characteristic response of the hypothalamus to DIA.
Collapse
|
26
|
Fordahl S, Cooney P, Qiu Y, Xie G, Jia W, Erikson KM. Waterborne manganese exposure alters plasma, brain, and liver metabolites accompanied by changes in stereotypic behaviors. Neurotoxicol Teratol 2011; 34:27-36. [PMID: 22056924 DOI: 10.1016/j.ntt.2011.10.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 10/11/2011] [Accepted: 10/12/2011] [Indexed: 12/12/2022]
Abstract
Overexposure to waterborne manganese (Mn) is linked with cognitive impairment in children and neurochemical abnormalities in other experimental models. In order to characterize the threshold between Mn-exposure and altered neurochemistry, it is important to identify biomarkers that positively correspond with brain Mn-accumulation. The objective of this study was to identify Mn-induced alterations in plasma, liver, and brain metabolites using liquid/gas chromatography-time of flight-mass spectrometry metabolomic analyses; and to monitor corresponding Mn-induced behavior changes. Weanling Sprague-Dawley rats had access to deionized drinking water either Mn-free or containing 1g Mn/L for 6 weeks. Behaviors were monitored during the sixth week for a continuous 24h period while in a home cage environment using video surveillance. Mn-exposure significantly increased liver, plasma, and brain Mn concentrations compared to control, specifically targeting the globus pallidus (GP). Mn significantly altered 98 metabolites in the brain, liver, and plasma; notably shifting cholesterol and fatty acid metabolism in the brain (increased oleic and palmitic acid; 12.57 and 15.48 fold change (FC), respectively), and liver (increased oleic acid, 14.51 FC; decreased hydroxybutyric acid, -14.29 FC). Additionally, Mn-altered plasma metabolites homogentisic acid, chenodeoxycholic acid, and aspartic acid correlated significantly with GP and striatal Mn. Total distance traveled was significantly increased and positively correlated with Mn-exposure, while nocturnal stereotypic and exploratory behaviors were reduced with Mn-exposure and performed largely during the light cycle compared to unexposed rats. These data provide putative biomarkers for Mn-neurotoxicity and suggest that Mn disrupts the circadian cycle in rats.
Collapse
Affiliation(s)
- Steve Fordahl
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC 27402-6170, USA
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Glutamine (Gln) is found abundantly in the central nervous system (CNS) where it participates in a variety of metabolic pathways. Its major role in the brain is that of a precursor of the neurotransmitter amino acids: the excitatory amino acids, glutamate (Glu) and aspartate (Asp), and the inhibitory amino acid, γ-amino butyric acid (GABA). The precursor-product relationship between Gln and Glu/GABA in the brain relates to the intercellular compartmentalization of the Gln/Glu(GABA) cycle (GGC). Gln is synthesized from Glu and ammonia in astrocytes, in a reaction catalyzed by Gln synthetase (GS), which, in the CNS, is almost exclusively located in astrocytes (Martinez-Hernandez et al., 1977). Newly synthesized Gln is transferred to neurons and hydrolyzed by phosphate-activated glutaminase (PAG) to give rise to Glu, a portion of which may be decarboxylated to GABA or transaminated to Asp. There is a rich body of evidence which indicates that a significant proportion of the Glu, Asp and GABA derived from Gln feed the synaptic, neurotransmitter pools of the amino acids. Depolarization-induced-, calcium- and PAG activity-dependent releases of Gln-derived Glu, GABA and Asp have been observed in CNS preparations in vitro and in the brain in situ. Immunocytochemical studies in brain slices have documented Gln transfer from astrocytes to neurons as well as the location of Gln-derived Glu, GABA and Asp in the synaptic terminals. Patch-clamp studies in brain slices and astrocyte/neuron co-cultures have provided functional evidence that uninterrupted Gln synthesis in astrocytes and its transport to neurons, as mediated by specific carriers, promotes glutamatergic and GABA-ergic transmission. Gln entry into the neuronal compartment is facilitated by its abundance in the extracellular spaces relative to other amino acids. Gln also appears to affect neurotransmission directly by interacting with the NMDA class of Glu receptors. Transmission may also be modulated by alterations in cell membrane polarity related to the electrogenic nature of Gln transport or to uncoupled ion conductances in the neuronal or glial cell membranes elicited by Gln transporters. In addition, Gln appears to modulate the synthesis of the gaseous messenger, nitric oxide (NO), by controlling the supply to the cells of its precursor, arginine. Disturbances of Gln metabolism and/or transport contribute to changes in Glu-ergic or GABA-ergic transmission associated with different pathological conditions of the brain, which are best recognized in epilepsy, hepatic encephalopathy and manganese encephalopathy.
Collapse
|
28
|
Cannon JR, Greenamyre JT. The role of environmental exposures in neurodegeneration and neurodegenerative diseases. Toxicol Sci 2011; 124:225-50. [PMID: 21914720 DOI: 10.1093/toxsci/kfr239] [Citation(s) in RCA: 268] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neurodegeneration describes the loss of neuronal structure and function. Numerous neurodegenerative diseases are associated with neurodegeneration. Many are rare and stem from purely genetic causes. However, the prevalence of major neurodegenerative diseases is increasing with improvements in treating major diseases such as cancers and cardiovascular diseases, resulting in an aging population. The neurological consequences of neurodegeneration in patients can have devastating effects on mental and physical functioning. The causes of most cases of prevalent neurodegenerative diseases are unknown. The role of neurotoxicant exposures in neurodegenerative disease has long been suspected, with much effort devoted to identifying causative agents. However, causative factors for a significant number of cases have yet to be identified. In this review, the role of environmental neurotoxicant exposures on neurodegeneration in selected major neurodegenerative diseases is discussed. Alzheimer's disease, Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis were chosen because of available data on environmental influences. The special sensitivity the nervous system exhibits to toxicant exposure and unifying mechanisms of neurodegeneration are explored.
Collapse
Affiliation(s)
- Jason R Cannon
- Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | | |
Collapse
|