Warning: mkdir(): Permission denied in /home/virtual/lib/view_data.php on line 81

Warning: fopen(upload/ip_log/ip_log_2024-09.txt): failed to open stream: No such file or directory in /home/virtual/lib/view_data.php on line 83

Warning: fwrite() expects parameter 1 to be resource, boolean given in /home/virtual/lib/view_data.php on line 84
Manganese Neurotoxicity: a Focus on Glutamate Transporters
Skip Navigation
Skip to contents

Ann Occup Environ Med : Annals of Occupational and Environmental Medicine

OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > Ann Occup Environ Med > Volume 25; 2013 > Article
Review Manganese Neurotoxicity: a Focus on Glutamate Transporters
Pratap Karki1, Eunsook Lee1, Michael Aschner2
Annals of Occupational and Environmental Medicine 2013;25:4-4.
DOI: https://doi.org/10.1186/2052-4374-25-4
Published online: May 21, 2013

1Department of Physiology, Meharry Medical College, Nashville, TN, USA

2Department of Pediatrics, Vanderbilt University Medical Center, 2215-B Garland Avenue, 11415 MRB IV, Nashville, TN, 37232-0414, USA

• Received: January 24, 2013   • Accepted: March 1, 2013

Copyright © 2013 Karki et al.; licensee BioMed Central Ltd.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 58 Views
  • 0 Download
  • 55 Web of Science
  • 55 Crossref
prev next
  • Manganese (Mn) is an essential element that is required in trace amount for normal growth, development as well maintenance of proper function and regulation of numerous cellular and biochemical reactions. Yet, excessive Mn brain accumulation upon chronic exposure to occupational or environmental sources of this metal may lead to a neurodegenerative disorder known as manganism, which shares similar symptoms with idiopathic Parkinson’s disease (PD). In recent years, Mn exposure has gained public health interest for two primary reasons: continuous increased usage of Mn in various industries, and experimental findings on its toxicity, linking it to a number of neurological disorders. Since the first report on manganism nearly two centuries ago, there have been substantial advances in the understanding of mechanisms associated with Mn-induced neurotoxicity. This review will briefly highlight various aspects of Mn neurotoxicity with a focus on the role of astrocytic glutamate transporters in triggering its pathophysiology.
Mn is essential but needs to be in the balance
Mn is a ubiquitous element present naturally in the environment, including water and food. In the human body, Mn is found in all tissues where it is required for normal amino acid, lipid, protein and carbohydrate metabolism, and ATP generation. Mn also participates in blood clotting and sugar homeostasis, immune responsiveness, digestion, reproduction, and bone growth [1-3]. It is a critical component of numerous metalloenzymes, including Mn superoxide dismutase, arginase, phosphoenolpyruvate decarboxylase and glutamine synthetase [4-6], to name a few. Despite its requirement in multiple physiological processes, elevated levels of Mn trigger toxicity, particularly within the central nervous system (CNS), causing cognitive, psychiatric and motor abnormalities [7,8]. In humans, Mn deficiency is rare as it is abundant in diets, but in extenuating conditions it may contribute to developmental defects, including malformation of bones, altered macromolecular metabolism, reduced fertility, weakness and enhanced susceptibility to seizures [3,9,10]. Mn deficiency has also been shown to induce skeletal defects and impaired lipid metabolism [11,12].
Sources of human exposure
Mn is naturally abundant in the Earth’s crust (0.1%) and soil (40–900 mg/kg) [13,14]. Mn is found as oxides, carbonates and silicates in minerals. The versatile chemical properties of Mn have expanded its industrial use in glass, ceramics, paint and adhesive industries, as well as in welding. The wide usage of Mn in a range of industries has led to global health concerns. Indeed, occupational exposures to Mn have been documented in multiple industries, including ferroalloy smelting, welding, mining, battery, glass and ceramics [15-19].
The primary source of Mn exposure in the general human population is from diet, which is estimated to contain 0.9-10 mg Mn per day [20]. Rice, grains and nuts are rich sources of Mn with excess of 30 mg/kg, while Mn content in tea is 0.4-1.3 mg/cup. Mn drinking water levels are in the range of 1–100 μg/L [21]. The high Mn content in infant formulas compared to human milk, has recently drawn public attention [22], along with Mn in parenteral nutrition. The latter is considered a risk factor for Mn-induced toxicity since the normal intestinal regulatory mechanisms are bypassed, rendering intravenously delivered Mn 100% bioavailable [23,24]. Mn in fumes, aerosols or suspended particulate matters is deposited in the upper and lower respiratory tract, with subsequent entry into the bloodstream. An Mn-containing gasoline anti-knock additives additive, methylcyclopentadienyl manganese tricarbonyl (MMT), has been introduced in several countries, representing another source of Mn exposure via inhalation [25,26]. Designer drugs, such as methcathinone hydrochloride (ephedrine), where potassium permanganate is used as an oxidant for the synthesis of the illicit drugs [27] has also been shown to cause neurodegenerative sequalae in drug abusers, consistent with PD-like symptomology.
Mn absorption, transport and excretion
Diet represents the major source of Mn in humans. The gastrointestinal tract absorbs 1-5% of ingested Mn; 60-70% of inhaled Mn is exhaled by the lungs [28,29]. The uptake of Mn is tightly regulated and excess Mn is excreted through the bile [30]. Both active transport and passive diffusion mechanisms regulate oral Mn absorption and the process is governed by various factors, including dietary levels of Mn and other minerals as well as age [20,30,31]. Among other metals, iron (Fe) stores are particularly important given the inverse relationship between cellular Fe levels and Mn uptake, as evidenced by increased transport of orally administered Mn in states of Fe deficiency [32,33]. In blood, Mn+2 is predominantly (>99%) in the 2+ oxidation state and mainly bound to β-globulin and albumin. A small fraction of Mn+3 is complexed with transferrin [34,35]. Mn (in the 2+ oxidation state) transport across the blood–brain barrier (BBB) and cell membranes is facilitated by the divalent metal ion transporter 1 (DMT1), N-methyl-d-aspartate (NMDA) receptor channel and Zip8 [36-38], to name a few. Transferrin is the most efficient transport system for Mn in the 3+ oxidation state [39]. Mn is distributed throughout the brain and the highest Mn levels are found in the globus pallidus and other nuclei of the basal ganglia (striatum, substantia nigra) [40,41]. DMT1 and transferrin also regulate Mn uptake both in astrocytes and neurons [42,43]. Generally, the intracellular Mn concentration is higher in tissues rich in mitochondria and pigmentation. The highest Mn levels are noted in bone, liver, pancreas and kidney compared to other tissues [44].
Mn in neurological disorders
Chronic inhalation of air-borne Mn particulates represents the major cause of human neurotoxicity, though there is growing number of reports on Mn toxicity resulting from consumption of Mn-adulterated drinking water [45,46]. Occupational exposures represent the predominant source of excessive Mn exposure [47]. Manganism, first described by Couper in 1837 [48] is a clinical disorder characterized by psychological and neurological abnormalities that shares multiple analogous symptoms with idiopathic PD [Reviewed in [49]]. The early symptoms of manganism include hallucinations, psychoses and various behavioral disturbances soon followed by postural instability, dystonia, bradykinesia and rigidity [50]. Despite their resemblance in clinical features, manganism is clinically distinguishable from PD [51]. Mn-induced neurotoxicity affects mainly the globus pallidus as well as the cortex and hypothalamus [52,53], distinct from the striatal changes associated with PD. Excessive CNS Mn levels may contribute in the pathogenesis of PD, causing loss of dopamine in the striatum, death of non-dopaminergic (DAergic) neurons in the globus pallidus, and damage to glutamatergic and GABAergic projections [54,55]. Mn has also been shown to increase fibril formation by α-synuclein along with its expression and aggregation [53,56,57]. While playing a major role in the etiology of PD [58], the precise role of α-synuclein in Mn-induced neurotoxicity has yet to be determined. A role for Mn has also been advanced in the etiology of Huntington’s disease, amyotrophic lateral sclerosis and Alzheimer’s disease [Reviewed in [49]].
Mechanism of Mn Neurotoxicity

Mitochondrial dysfunction

Mitochondria serve as the primary storage site for intracellular Mn where it is taken up by the calcium uniporter [59]. Mn is also an important cofactor for various mitochondrial enzymes and, thus, the elevation in Mn levels in this organelle can directly interfere with oxidative phosphorylation. Mn inhibits the function of F1-ATPase and the formation of complex I of the electron transport chain, thereby interfering with cellular ATP synthesis [60,61].

Oxidative stress

Elevated intra-mitochondrial Mn levels trigger oxidative stress, generating the excessive reactive oxygen species (ROS), causing mitochondrial dysfunction [61,62]. The transition of Mn+2 to Mn+3 increases its pro-oxidant capacity [63]. Mn-induced oxidative stress leads to the opening of mitochondrial transition pore (MTP), resulting in increased solubility to protons, ions and solutes, loss of the mitochondrial inner membrane potential, impairment of oxidative phosphorylation and ATP synthesis and mitochondrial swelling [64,65]. Furthermore, Mn exposure has also been linked to the activation of signaling pathways involved in response to oxidative stress, including nuclear factor kappa B (NF-kB) and activator protein-1 (AP-1) [66,67].

Apoptosis

Neuronal cell death by apoptosis has been considered to play a major role in neurodegenerative diseases, including PD [68]. Mn has been shown to trigger apoptosis in DAergic neurons in a caspase-3-dependent manner by activation of protein kinase C delta (PKC-δ) [69]. Similarly, Mn has been shown to cause apoptotic cell death in astrocytes by mitochondrial pathways involving cytochrome c release and caspase activation [64,70].

Inflammation

Although the oxidative stress induced by mitochondrial dysfunction is regarded as the major pathological mechanism of Mn neurotoxicity, recent studies also suggest a proinflammatory role for Mn, which involves the activation of glial cells characterized by the release of non-neuronal derived ROS, such as nitric oxide (NO), cytokines, prostaglandins and H2O2[71-73]. Mn potentiates the release of several cytokines, including TNF-α, IL-6, IL-1β from the activated glial cells, thereby activating various transcription factors including NF-kB, AP-1 and kinases including ERK, JNK, AKT, and PKC-α [Reviewed in [74]].
Astrocytes are the site of early dysfunction and damage in Mn neurotoxicity. Astrocytes are the most abundant CNS cells (~50% by volume), and they perform numerous essential functions for normal neuronal activity, such as glutamate uptake, glutamine release, K+ and H+ buffering and volume regulation [36,75,76]. Astrocytes accumulate up to 50-fold higher Mn concentrations compared to neurons, thus serving as the main homeostatic and storage site for this metal [75,77]. The intracellular concentration of Mn in astrocytes is ~50-75 μM where it is an essential cofactor for the astrocyte-specific enzyme glutamine synthetase, which catalyzes the conversion of glutamate to glutamine [78,79]. The excessive accumulation of Mn in astrocytes mediates neurotoxicity primarily by oxidative stress and impairment of glutamate transporters [80,81]. Mn toxicity has been shown to cause astrocytic alterations in primate models, and exposure of pathophysiologically relevant Mn concentrations in astrocytes in vitro causes time-and concentration-dependent cell swelling secondary to oxidative stress [82,83]. One of the proposed mechanisms of Mn-induced neurotoxicity in astrocytes is alteration in glutamate homeostasis due to impairment of glutamate transporters [84]. Mn has also been shown to downregulate the expression and function of glutamine transporters, resulting in reduced glutamine uptake [85]. The impairment of glutamate/glutamine transporters results in increased extracellular glutamate levels that elicit excitatory neurotoxicity. In support of this mechanism, we and others have shown that estrogen and selective estrogen receptor modulators protect astrocytes from Mn-induced neurotoxicity by upregulating the expression and function of glutamate transporters [86-88]. Moreover, Mn also activates selective cellular signaling pathways that mediate alterations in glutamate-glutamine homeostasis. The decrease in glutamine uptake after the activation of PKC-δ by Mn represents a typical example of the involvement of signaling pathways in Mn-induced neurotoxicity [89].
Although it is widely accepted that Mn impairs the expression and function of the two main glutamate transporters (GLAST and GLT-1), its mechanism of action at the transcriptional levels remains unknown. The increased production of ROS and TNF-α by Mn is thought to be the principal cause that leads to impairment in glutamate transporter function. ROS inhibit astrocyte glutamate uptake, and TNF-α decreases GLAST and GLT-1 protein and mRNA levels [90-92]. Oxidative stress also plays an important role in the regulation of glutamate transporter function since the activity of glutamate transporters is regulated by the redox state of reactive cysteine residues, with a dramatic decrease in activity once the reduced cysteine is oxidized [93]. Furthermore, glutamate uptake by the recombinant glutamate transporters EAAT1, EAAT2 and EAAT3 was found to be inhibited by peroxynitrite and H2O2 and restored upon treatment with the reducing agent, dithithreotol, suggesting a role for oxidative stress in the regulation of glutamate transporters activity [94]. TNF-α is a key neuroinflammatory mediator of neurotoxicity and neurodegeneration, and Mn increases the levels of this cytokine [95]. Several studies also corroborate the reduction in the expression and activity of glutamate transporters by TNF-α, highlighting its role as a negative regulator of the transporter [90,92]. NF-kB and MAPK signaling pathways mediate TNF-α-induced reduction in GLT-1 expression, since the inhibition of these pathways restores the decrease in TNF-α induced GLT-1 expression and function [92].
Chronic excessive exposures to Mn represent a global health concern as growing evidences suggests that Mn accumulation in the brain may be a predisposing factor for several neurodegenerative diseases. Studies over the past several decades have provided invaluable insights into the cause, effects and mechanisms of Mn-induced neurotoxicity. The recent findings on the involvement of glutamate transporters and cellular signaling pathways in Mn-induced neurotoxicity provide not only new insights into the molecular mechanisms of Mn-induced neurotoxicity, but also provide new therapeutic targets in the development of novel drugs to attenuate the symptoms associated with manganism, PD and other related neurodegenerative disorders.
The authors declare that there is no any competing interest.
PK drafted the initial manuscript, EL and MA corrected, edited and finalized it. All the authors read and approve the final version of the manuscript.
The studies in our laboratories are supported by NIH grants R01 ES 10563, P30 ES 00267 and SC1 GM089630.
  • Aschner M, Erikson KM, Dorman DC: Crit Rev Toxicol. 2005;25: 1–32. 10.1080/10408440590905920. 15742901.
  • Erikson KM, Thompson K, Aschner J, Aschner M: Pharmacol Ther. 2007;25: 369–377. 10.1016/j.pharmthera.2006.09.002. 17084903.
  • Aschner JL, Aschner M: Mol Aspects Med. 2005;25: 353–362. 10.1016/j.mam.2005.07.003. 16099026.
  • Baly DL: J Nutr. 1985;25: 327.
  • Bentle LA, Lardy HA: J Biol Chem. 1976;25: 2916–2921. 1270433.
  • Stallings WC, Metzger AL, Pattridge KA, Fee JA, Ludwig ML: Free Radic Res Commun. 1991;25(Pt 1):259–268. 2071033.
  • Dobson AW, Erikson KM, Aschner M: Ann N Y Acad Sci. 2004;25: 115–128. 10.1196/annals.1306.009. 15105259.
  • Pal PK, Samii A, Calne DB: Neurotoxicology. 1999;25: 227–238. 10385886.
  • Aschner M, Shanker G, Erikson K, Yang J, Mutkus LA: Neurotoxicology. 2002;25: 165–168. 10.1016/S0161-813X(02)00056-6. 12224757.
  • Keen CL, Ensunsa JL, Watson MH, Baly DL, Donovan SM, Monaco MH, Clegg MS: Neurotoxicology. 1999;25: 213–223. 10385885.
  • Hurley LS: Physiol Rev. 1981;25: 249–295. 7012858.
  • Klimis-Tavantzis DJ, Leach RM, Kris-Etherton PM: J Nutr. 1983;25: 328–336. 6822906.
  • Burton NC, Guilarte TR: Environ Health Perspect. 2009;25: 325–332. 19337503.
  • Cooper WC: J Toxicol Environ Health. 1984;25: 23–46. 10.1080/15287398409530561. 6389893.
  • Bader M, Dietz MC, Ihrig A, Triebig G: Int Arch Occup Environ Health. 1999;25: 521–527. 10.1007/s004200050410. 10592004.
  • Bast-Pettersen R, Ellingsen DG, Hetland SM, Thomassen Y: Int Arch Occup Environ Health. 2004;25: 277–287. 10.1007/s00420-003-0491-0. 15024571.
  • Bowler RM, Nakagawa S, Drezgic M, Roels HA, Park RM, Diamond E, Mergler D, Bouchard M, Bowler RP, Koller W: Neurotoxicology. 2007;25: 298–311. 10.1016/j.neuro.2006.11.001. 17169432.
  • Montes S, Riojas-Rodriguez H, Sabido-Pedraza E, Rios C: Environ Res. 2008;25: 89–95. 10.1016/j.envres.2007.08.008. 17915211.
  • Srivastava AK, Gupta BN, Mathur N, Murty RC, Garg N, Chandra SV: Vet Hum Toxicol. 1991;25: 280–282. 1858313.
  • Finley JW, Davis CD: BioFactors (Oxford, England). 1999;25: 15–24. 10.1002/biof.5520100102.
  • ATSDR. Toxicological Profile for Manganese. 2000, Atlanta. GA: Agency for Toxic Substances and Disease Registry.
  • Krachler M, Rossipal E: Ann Nutr Metab. 2000;25: 68–74. 10.1159/000012823. 10970995.
  • Bertinet DB, Tinivella M, Balzola FA, de Francesco A, Davini O, Rizzo L, Massarenti P, Leonardi MA, Balzola F: JPEN. Journal of parenteral and enteral nutrition. 2000;25: 223–227. 10.1177/0148607100024004223. 10885716.
  • Hardy G: Gastroenterology. 2009;25: S29–S35. 10.1053/j.gastro.2009.08.011. 19874947.
  • Davis JM: Environ Health Perspect. 1998;25(Suppl 1):191–201. 10.1289/ehp.98106s1191. 9539013.
  • Kaiser J: Science (New York, N.Y.). 2003;25: 926–928. 10.1126/science.300.5621.926.
  • Sikk K, Taba P, Haldre S, Bergquist J, Nyholm D, Askmark H, Danfors T, Sorensen J, Thurfjell L, Raininko R, Eriksson R, Flink R, Farnstrand C, Aquilonius SM: Acta Neurol Scand. 2010;25: 237–243. 10.1111/j.1600-0404.2009.01189.x. 20028341.
  • Davidsson L, Cederblad A, Hagebo E, Lonnerdal B, Sandstrom B: J Nutr. 1998;25: 1517–1521.
  • Mena I: Ann Clin Lab Sci. 1974;25: 487–491. 4429343.
  • Davis CD, Zech L, Greger JL: Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine (New York, N.Y.) 1993;25:103–108.Article
  • Dorman DC, Struve MF, James RA, McManus BE, Marshall MW, Wong BA: Toxicological sciences : an official journal of the Society of Toxicology. 2001;25: 242–251. 10.1093/toxsci/60.2.242. 11248135.
  • Davis CD, Malecki EA, Greger JL: Am J Clin Nutr. 1992;25: 926–932. 1415012.
  • Gunshin H, Mackenzie B, Berger UV, Gunshin Y, Romero MF, Boron WF, Nussberger S, Gollan JL, Hediger MA: Nature. 1997;25: 482–488. 10.1038/41343. 9242408.
  • Aisen P, Aasa R, Redfield AG: J Biol Chem. 1969;25: 4628–4633. 4309148.
  • Critchfield JW, Keen CL: Metabolism: clinical and experimental. 1992;25: 1087–1092. 10.1016/0026-0495(92)90290-Q. 1406294.
  • Aschner M, Gannon M: Brain Res Bull. 1994;25: 345–349. 10.1016/0361-9230(94)90204-6. 8293318.
  • Au C, Benedetto A, Aschner M: Neurotoxicology. 2008;25: 569–576. 10.1016/j.neuro.2008.04.022. 18565586.
  • Itoh K, Sakata M, Watanabe M, Aikawa Y, Fujii H: Neuroscience. 2008;25: 732–740. 10.1016/j.neuroscience.2008.03.080. 18495352.
  • Aschner M, Aschner JL: Brain Res Bull. 1990;25: 857–860. 10.1016/0361-9230(90)90152-P. 2372703.
  • Dorman DC, Struve MF, Marshall MW, Parkinson CU, James RA, Wong BA: Toxicological sciences : an official journal of the Society of Toxicology. 2006;25: 201–210. 10.1093/toxsci/kfj206. 16624849.
  • Guilarte TR, McGlothan JL, Degaonkar M, Chen MK, Barker PB, Syversen T, Schneider JS: Toxicological sciences : an official journal of the Society of Toxicology. 2006;25: 351–358. 10.1093/toxsci/kfl106. 16968886.
  • Erikson KM, Aschner M: Neurotoxicology. 2006;25: 125–130. 10.1016/j.neuro.2005.07.003. 16140386.
  • Suarez N, Eriksson H: J Neurochem. 1993;25: 127–131. 10.1111/j.1471-4159.1993.tb03546.x. 8515258.
  • Rehnberg GL, Hein JF, Carter SD, Laskey JW: J Toxicol Environ Health. 1980;25: 217–226. 10.1080/15287398009529844. 7381972.
  • Mena I, Marin O, Fuenzalida S, Cotzias GC: Neurology. 1967;25: 128–136. 10.1212/WNL.17.2.128. 6066873.
  • Roels HA, Bowler RM, Kim Y, Claus Henn B, Mergler D, Hoet P, Gocheva VV, Bellinger DC, Wright RO, Harris MG, Chang Y, Bouchard MF, Riojas-Rodriguez H, Menezes-Filho JA, Tellez-Rojo MM: Neurotoxicology. 2012;25: 872–880. 10.1016/j.neuro.2012.03.009. 22498092.
  • Mergler D, Huel G, Bowler R, Iregren A, Belanger S, Baldwin M, Tardif R, Smargiassi A, Martin L: Environ Res. 1994;25: 151–180. 10.1006/enrs.1994.1013. 8306949.
  • Couper J: Br Ann Med Pharmacol. 1837;25: 41–42.
  • Bowman AB, Kwakye GF, Hernandez EH, Aschner M: Journal of trace elements in medicine and biology : organ of the Society for Minerals and Trace Elements (GMS). 2011;25: 191–203. 10.1016/j.jtemb.2011.08.144. 21963226.
  • Olanow CW: Ann N Y Acad Sci. 2004;25: 209–223. 10.1196/annals.1306.018. 15105268.
  • Calne DB, Chu NS, Huang CC, Lu CS, Olanow W: Neurology. 1994;25: 1583–1586. 10.1212/WNL.44.9.1583. 7936278.
  • Yamada M, Ohno S, Okayasu I, Okeda R, Hatakeyama S, Watanabe H, Ushio K, Tsukagoshi H: Acta Neuropathol. 1986;25: 273–278. 10.1007/BF00686083. 3766127.
  • Verina T, Schneider JS, Guilarte TR: Toxicol Lett. 2012.
  • Guilarte TR: Environ Health Perspect. 2010;25: 1071–80. 10.1289/ehp.0901748. 20403794.
  • Sikk K, Haldre S, Aquilonius SM, Taba P: Parkinson's disease. 2011;25: 865319. 23671637.
  • Li Y, Sun L, Cai T, Zhang Y, Lv S, Wang Y, Ye L: Brain Res Bull. 2010;25: 428–33. 10.1016/j.brainresbull.2009.11.007. 19932157.
  • Uversky VN, Li J, Fink AL: J Biol Chem. 2001;25: 44284–96. 10.1074/jbc.M105343200. 11553618.
  • Eller M, Williams DR: Clinical chemistry and laboratory medicine: CCLM / FESCC. 2011;25: 403–8. 21342025.
  • Gavin CE, Gunter KK, Gunter TE: Neurotoxicology. 1999;25: 445–53. 10385903.
  • Chen JY, Tsao GC, Zhao Q, Zheng W: Toxicol Appl Pharmacol. 2001;25: 160–8. 10.1006/taap.2001.9245. 11543648.
  • Gavin CE, Gunter KK, Gunter TE: Toxicol Appl Pharmacol. 1992;25: 1–5. 10.1016/0041-008X(92)90360-5. 1631887.
  • Milatovic D, Yin Z, Gupta RC, Sidoryk M, Albrecht J, Aschner JL, Aschner M: Toxicological sciences : an official journal of the Society of Toxicology. 2007;25: 198–205. 10.1093/toxsci/kfm095. 17468184.
  • Reaney SH, Smith DR: Toxicol Appl Pharmacol. 2005;25: 271–81. 10.1016/j.taap.2004.10.013. 15922012.
  • Yin Z, Aschner JL, dos Santos AP, Aschner M: Brain Res. 2008;25: 1–11. 18313649.
  • Zoratti M, Szabo I: Biochim Biophys Acta. 1995;25: 139–76. 10.1016/0304-4157(95)00003-A. 7640294.
  • Ramesh GT, Ghosh D, Gunasekar PG: Toxicol Lett. 2002;25: 151–8. 10.1016/S0378-4274(02)00332-6. 12425965.
  • Wise K, Manna S, Barr J, Gunasekar P, Ramesh G: Toxicol Lett. 2004;25: 237–44. 10.1016/j.toxlet.2003.11.007. 15104115.
  • Dauer W, Przedborski S: Neuron. 2003;25: 889–909. 10.1016/S0896-6273(03)00568-3. 12971891.
  • Latchoumycandane C, Anantharam V, Kitazawa M, Yang Y, Kanthasamy A, Kanthasamy AG: J Pharmacol Exp Ther. 2005;25: 46–55. 15608081.
  • Gonzalez LE, Juknat AA, Venosa AJ, Verrengia N, Kotler ML: Neurochem Int. 2008;25: 408–15. 10.1016/j.neuint.2008.09.008. 18930091.
  • Liu X, Sullivan KA, Madl JE, Legare M, Tjalkens RB: Toxicological sciences: an official journal of the Society of Toxicology. 2006;25: 521–31. 10.1093/toxsci/kfj150. 16551646.
  • Zhang P, Lokuta KM, Turner DE, Liu B: J Neurochem. 2010;25: 434–43. 10.1111/j.1471-4159.2009.06477.x. 19895668.
  • Zhang P, Wong TA, Lokuta KM, Turner DE, Vujisic K, Liu B: Exp Neurol. 2009;25: 219–30. 10.1016/j.expneurol.2009.02.013. 19268665.
  • Filipov NM, Dodd CA: Journal of applied toxicology: JAT. 2012;25: 310–7. 10.1002/jat.1762. 22120544.
  • Aschner M, Guilarte TR, Schneider JS, Zheng W: Toxicol Appl Pharmacol. 2007;25: 131–47. 10.1016/j.taap.2007.03.001. 17466353.
  • Chen MK, Lee JS, McGlothan JL, Furukawa E, Adams RJ, Alexander M, Wong DF, Guilarte TR: Neurotoxicology. 2006;25: 229–36. 10.1016/j.neuro.2005.10.008. 16325911.
  • Aschner M, Erikson KM, Herrero Hernandez E, Tjalkens R: Neuromolecular Med. 2009;25: 252–66. 10.1007/s12017-009-8083-0. 19657747.
  • Gorovits R, Avidan N, Avisar N, Shaked I, Vardimon L: Proc Natl Acad Sci USA. 1997;25: 7024–9. 10.1073/pnas.94.13.7024. 9192685.
  • Tholey G, Ledig M, Mandel P, Sargentini L, Frivold AH, Leroy M, Grippo AA, Wedler FC: Neurochem Res. 1988;25: 45–50. 10.1007/BF00971853. 2897087.
  • Brouillet EP, Shinobu L, McGarvey U, Hochberg F, Beal MF: Exp Neurol. 1993;25: 89–94. 10.1006/exnr.1993.1042. 8477830.
  • Desole MS, Sciola L, Delogu MR, Sircana S, Migheli R, Miele E: Neurochem Int. 1997;25: 169–76. 10.1016/S0197-0186(96)00146-5. 9220449.
  • Olanow CW, Good PF, Shinotoh H, Hewitt KA, Vingerhoets F, Snow BJ, Beal MF, Calne DB, Perl DP: Neurology. 1996;25: 492–8. 10.1212/WNL.46.2.492. 8614520.
  • Rama Rao KV, Reddy PV, Hazell AS, Norenberg MD: Neurotoxicology. 2007;25: 807–12. 10.1016/j.neuro.2007.03.001. 17408748.
  • Erikson K, Aschner M: Neurotoxicology. 2002;25: 595–602. 10.1016/S0161-813X(02)00012-8. 12428731.
  • Sidoryk-Wegrzynowicz M, Lee E, Albrecht J, Aschner M: J Neurochem. 2009;25: 822–30. 10.1111/j.1471-4159.2009.06172.x. 19457077.
  • Deng Y, Xu Z, Xu B, Xu D, Tian Y, Feng W: Biol Trace Elem Res. 2012;25: 242–9. 10.1007/s12011-012-9365-1. 22391793.
  • Lee ES, Sidoryk M, Jiang H, Yin Z, Aschner M: J Neurochem. 2009;25: 530–44. 10.1111/j.1471-4159.2009.06105.x. 19453300.
  • Lee ES, Yin Z, Milatovic D, Jiang H, Aschner M: Toxicological sciences: an official journal of the Society of Toxicology. 2009;25: 156–67. 10.1093/toxsci/kfp081. 19383943.
  • Sidoryk-Wegrzynowicz M, Lee ES, Ni M, Aschner M: Glia. 2010;25: 1905–12. 10.1002/glia.21060. 20737472.
  • Sitcheran R, Gupta P, Fisher PB, Baldwin AS: EMBO J. 2005;25: 510–20. 10.1038/sj.emboj.7600555. 15660126.
  • Sorg O, Horn TF, Yu N, Gruol DL, Bloom FE: Molecular medicine (Cambridge, Mass.). 1997;25: 431–40.
  • Su ZZ, Leszczyniecka M, Kang DC, Sarkar D, Chao W, Volsky DJ, Fisher PB: Proc Natl Acad Sci USA. 2003;25: 1955–60. 10.1073/pnas.0136555100. 12578975.
  • Trotti D, Danbolt NC, Volterra A: Trends Pharmacol Sci. 1998;25: 328–34. 10.1016/S0165-6147(98)01230-9. 9745361.
  • Miralles VJ, Martinez-Lopez I, Zaragoza R, Borras E, Garcia C, Pallardo FV, Vina JR: Brain Res. 2001;25: 21–9. 10.1016/S0006-8993(01)03124-9. 11730698.
  • Crittenden PL, Filipov NM: Toxicology in vitro : an international journal published in association with BIBRA. 2008;25: 18–27. 10.1016/j.tiv.2007.07.004. 17845838.

Figure & Data

REFERENCES

    Citations

    Citations to this article as recorded by  
    • Sex-specific associations of a ferroalloy metal mixture with motor function in Italian adolescents
      Alexa Friedman, Samantha Schildroth, Victoria Fruh, Maxine H. Krengel, Yorghos Tripodis, Donatella Placidi, Roberta F. White, Roberto G. Lucchini, Donald R. Smith, Robert O. Wright, Megan K. Horton, Birgit Claus Henn
      Environmental Epidemiology.2024; 8(4): e321.     CrossRef
    • Astrocyte dysregulation as an epileptogenic factor: a systematic review
      Komang Trisna Sumadewi, Bryan Gervais de Liyis, Ni Made Linawati, I Putu Eka Widyadharma, I Nyoman Mantik Astawa
      The Egyptian Journal of Neurology, Psychiatry and Neurosurgery.2024;[Epub]     CrossRef
    • Lactational Exposure of Human Infants to Metal (loid)s: A Comparison of Industrial and Urban Inhabitants in North of the Persian Gulf
      Leila Amiri, Atefeh Khalili Doroodzani, Afshin Ostovar, Sina Dobaradaran, Azam Mohammadi, Iraj Nabipour, Alireza Raeisi, Hasan Malekizadeh, Akram Farhadi, Reza Saeedi, Sima Afrashteh, Shahrokh Nazmara, Mozhgan Keshtkar
      Biological Trace Element Research.2024; 202(5): 1829.     CrossRef
    • Pathogenesis and management of multiple sclerosis revisited
      Abhi Shah, Viraj Panchal, Kashyap Patel, Zainab Alimohamed, Nirja Kaka, Yashendra Sethi, Neil Patel
      Disease-a-Month.2023; 69(9): 101497.     CrossRef
    • Structures and coordination chemistry of transporters involved in manganese and iron homeostasis
      Shamayeeta Ray, Rachelle Gaudet
      Biochemical Society Transactions.2023; 51(3): 897.     CrossRef
    • The role of excitatory amino acid transporter 2 (EAAT2) in epilepsy and other neurological disorders
      Sahar Alijanpour, Mohammad Miryounesi, Soudeh Ghafouri-Fard
      Metabolic Brain Disease.2023; 38(1): 1.     CrossRef
    • Transcranial direct current stimulation ameliorates motor and cognitive functions by regulating neuronal excitotoxicity in experimental Parkinson’s disease model
      Güven AKÇAY, Serhan TAMERER
      Cukurova Medical Journal.2023; 48(3): 919.     CrossRef
    • Effects of Chronic Hydrocodone Exposure and Ceftriaxone on the Expression of Astrocytic Glutamate Transporters in Mesocorticolimbic Brain Regions of C57/BL Mice
      Woonyen Wong, Youssef Sari
      Toxics.2023; 11(10): 870.     CrossRef
    • Upregulation of glutamate transporter 1 by mTOR/Akt pathway in astrocyte culture during oxygen–glucose deprivation and reoxygenation
      Mi Li, Jingmei Yu, Huan Deng, Shansha Xie, Qiuling Li, Yuping Zhao, Shubin Yin, Yi-Fei Ji
      Experimental Brain Research.2023; 241(1): 201.     CrossRef
    • Munc18-1 Contributes to Hippocampal Injury in Septic Rats Through Regulation of Syntanxin1A and Synaptophysin and Glutamate Levels
      Fajuan Tang, Lin Chen, Hu Gao, Yupeng Lei, Linli Pan, Dongqiong Xiao, Xihong Li
      Neurochemical Research.2023; 48(3): 791.     CrossRef
    • Comparative analysis of astrocytes in the prefrontal cortex of primates: Insights into the evolution of human brain energetics
      Emily L. Munger, Melissa K. Edler, William D. Hopkins, Patrick R. Hof, Chet C. Sherwood, Mary Ann Raghanti
      Journal of Comparative Neurology.2022; 530(18): 3106.     CrossRef
    • Ultrasmall Fe@Fe3O4 nanoparticles as T1–T2 dual-mode MRI contrast agents for targeted tumor imaging
      Donglin Liu, Jiaojiao Li, Chengbin Wang, Lu An, Jiaomin Lin, Qiwei Tian, Shiping Yang
      Nanomedicine: Nanotechnology, Biology and Medicine.2021; 32: 102335.     CrossRef
    • Effects of Pentoxifylline in a Rat Model of Manganism: Evaluation of the Possible Toxicity
      Romina Tanideh, Omid Farshad, Akram Jamshidzadeh, Aida Iraji, Murat Senturk
      Journal of Chemistry.2021; 2021: 1.     CrossRef
    • Exercise increases striatal Glu reuptake and improves motor dysfunction in 6-OHDA-induced Parkinson’s disease rats
      Yan Feng, Shifang Zhou, Jian Sun
      Experimental Brain Research.2021; 239(11): 3277.     CrossRef
    • Single step nanospray drying preparation technique of gabapentin-loaded nanoparticles-mediated brain delivery for effective treatment of PTZ-induced seizures
      Alaa H. Salama, Abeer A.A. Salama, Mona Elhabak
      International Journal of Pharmaceutics.2021; 602: 120604.     CrossRef
    • Micellar buccal film for safe and effective control of seizures: Preparation, in vitro characterization, ex vivo permeation studies and in vivo assessment
      Alaa H. Salama, Mona Basha, Abeer A.A. Salama
      European Journal of Pharmaceutical Sciences.2021; 166: 105978.     CrossRef
    • Magnetic Nanoparticles in Cancer Therapy and Diagnosis
      Ali Farzin, Seyed Alireza Etesami, Jacob Quint, Adnan Memic, Ali Tamayol
      Advanced Healthcare Materials.2020;[Epub]     CrossRef
    • Associations between manganese exposure and multiple immunological parameters in manganese-exposed workers healthy cohort
      Xiang Chen, Zhenfang Liu, Xiaoting Ge, Xiaoyu Luo, Sifang Huang, Yanting Zhou, Defu Li, Hong Cheng, Longman Li, Lulu Huang, Qingzhi Hou, Gaohui Zan, Yanli Tan, Chaoqun Liu, Yunfeng Zou, Xiaobo Yang
      Journal of Trace Elements in Medicine and Biology.2020; 59: 126454.     CrossRef
    • Glutamate Transporters (EAAT-1–3) as a Factor in the Pathogenesis and a Potential Therapeutic Target in Epilepsy
      I. V. Smolensky, S. V. Ovsepian, A. V. Zaitsev
      Neuroscience and Behavioral Physiology.2020; 50(6): 777.     CrossRef
    • Sodium Para-aminosalicylic Acid Reverses Changes of Glutamate Turnover in Manganese-Exposed Rats
      Zhao-Cong Li, Fang Wang, Shao-Jun Li, Lin Zhao, Jun-Yan Li, Yue Deng, Xiao-Juan Zhu, Yu-Wen Zhang, Dong-Jie Peng, Yue-Ming Jiang
      Biological Trace Element Research.2020; 197(2): 544.     CrossRef
    • 6-OHDA ile Oluşturulan Parkinson Hastalığı Modelinde Astrogliozis ve Glutamat Taşıyıcı Protein GLT1 Ekspresyonu
      Zehra MİNBAY, Bülent GÖREN, Özhan EYİGÖR
      Uludağ Üniversitesi Tıp Fakültesi Dergisi.2020; 46(3): 385.     CrossRef
    • Dysregulation of TFEB contributes to manganese-induced autophagic failure and mitochondrial dysfunction in astrocytes
      Ziyan Zhang, Jingqi Yan, Aaron B. Bowman, Miles R. Bryan, Rajat Singh, Michael Aschner
      Autophagy.2020; 16(8): 1506.     CrossRef
    • Neurobiology, Functions, and Relevance of Excitatory Amino Acid Transporters (EAATs) to Treatment of Refractory Epilepsy
      Aleksey V. Zaitsev, Ilya V. Smolensky, Pascal Jorratt, Saak V. Ovsepian
      CNS Drugs.2020; 34(11): 1089.     CrossRef
    • Dietary Intake and Urinary Excretion of Manganese in Korean Healthy Adults
      Mi-Kyeong Choi, Yun Jung Bae
      Biological Trace Element Research.2020; 196(2): 384.     CrossRef
    • Ceftriaxone Treatment Affects EAAT2 Expression and Glutamatergic Neurotransmission and Exerts a Weak Anticonvulsant Effect in Young Rats
      Aleksey V. Zaitsev, Sergey L. Malkin, Tatyana Y. Postnikova, Ilya V. Smolensky, Olga E. Zubareva, Irina V. Romanova, Maria V. Zakharova, Vladimir B. Karyakin, Vladimir Zavyalov
      International Journal of Molecular Sciences.2019; 20(23): 5852.     CrossRef
    • Brain mitochondria as potential therapeutic targets for managing hepatic encephalopathy
      Reza Heidari
      Life Sciences.2019; 218: 65.     CrossRef
    • Role of Astrocytes in Manganese Neurotoxicity Revisited
      Tao Ke, Marta Sidoryk-Wegrzynowicz, Edward Pajarillo, Asha Rizor, Félix Alexandre Antunes Soares, Eunsook Lee, Michael Aschner
      Neurochemical Research.2019; 44(11): 2449.     CrossRef
    • The role of zinc, copper, manganese and iron in neurodegenerative diseases
      Leda Mezzaroba, Daniela Frizon Alfieri, Andrea Name Colado Simão, Edna Maria Vissoci Reiche
      NeuroToxicology.2019; 74: 230.     CrossRef
    • The role of astrocytic glutamate transporters GLT-1 and GLAST in neurological disorders: Potential targets for neurotherapeutics
      Edward Pajarillo, Asha Rizor, Jayden Lee, Michael Aschner, Eunsook Lee
      Neuropharmacology.2019; 161: 107559.     CrossRef
    • Association of circulating manganese levels with Parkinson’s disease: A meta-analysis
      Ke Du, Ming-Yan Liu, Yan-Zhu Pan, Xin Zhong, Min-Jie Wei
      Neuroscience Letters.2018; 665: 92.     CrossRef
    • Synthesis and characterization of [Mn(phen)(H 2 O) 4 ]·SO 4 ·2H 2 O
      Ibanphylla Syiemlieh, Arvind Kumar, Sunshine D. Kurbah, Ram A. Lal
      Journal of Molecular Structure.2018; 1166: 102.     CrossRef
    • Manganese exposure induces neuroinflammation by impairing mitochondrial dynamics in astrocytes
      Souvarish Sarkar, Emir Malovic, Dilshan S. Harischandra, Hilary A. Ngwa, Anamitra Ghosh, Colleen Hogan, Dharmin Rokad, Gary Zenitsky, Huajun Jin, Vellareddy Anantharam, Anumantha G. Kanthasamy, Arthi Kanthasamy
      NeuroToxicology.2018; 64: 204.     CrossRef
    • The Impact of Environmental Mn Exposure on Insect Biology
      Yehuda Ben-Shahar
      Frontiers in Genetics.2018;[Epub]     CrossRef
    • Assessment of gender and age effects on serum and hair trace element levels in children with autism spectrum disorder
      Anatoly V. Skalny, Natalia V. Simashkova, Anastasia A. Skalnaya, Tatiana P. Klyushnik, Geir Bjørklund, Margarita G. Skalnaya, Alexey A. Tinkov
      Metabolic Brain Disease.2017; 32(5): 1675.     CrossRef
    • Cellular Fates of Manganese(II) Pentaazamacrocyclic Superoxide Dismutase (SOD) Mimetics: Fluorescently Labeled MnSOD Mimetics, X-ray Absorption Spectroscopy, and X-ray Fluorescence Microscopy Studies
      Claire M. Weekley, Isabell Kenkel, Rainer Lippert, Shengwei Wei, Dominik Lieb, Tiffanny Cranwell, Jason L. Wedding, Annika S. Zillmann, Robin Rohr, Milos R. Filipovic, Ivana Ivanović-Burmazović, Hugh H. Harris
      Inorganic Chemistry.2017; 56(11): 6076.     CrossRef
    • Health status of male steel workers at an electric arc furnace (EAF) in Trentino, Italy
      Roberto Cappelletti, Marcello Ceppi, Justina Claudatus, Valerio Gennaro
      Journal of Occupational Medicine and Toxicology.2016;[Epub]     CrossRef
    • Biochemical and ecophysiological responses to manganese stress by ectomycorrhizal fungus Pisolithus tinctorius and in association with Eucalyptus grandis
      Gabriela C. Canton, Amanda A. Bertolazi, Antônio J. D. Cogo, Frederico Jacob Eutrópio, Juliana Melo, Sávio Bastos de Souza, Cesar A. Krohling, Eliemar Campostrini, Ary Gomes da Silva, Arnoldo R. Façanha, Nuno Sepúlveda, Cristina Cruz, Alessandro C. Ramos
      Mycorrhiza.2016; 26(5): 475.     CrossRef
    • Null allele mutants of trt-1 , the catalytic subunit of telomerase in Caenorhabditis elegans , are less sensitive to Mn-induced toxicity and DAergic degeneration
      Omamuyovwi M. Ijomone, Mahfuzur R. Miah, Tanara V. Peres, Polycarp U. Nwoha, Michael Aschner
      NeuroToxicology.2016; 57: 54.     CrossRef
    • Iron Oxide Based Nanoparticles for Multimodal Imaging and Magnetoresponsive Therapy
      Nohyun Lee, Dongwon Yoo, Daishun Ling, Mi Hyeon Cho, Taeghwan Hyeon, Jinwoo Cheon
      Chemical Reviews.2015; 115(19): 10637.     CrossRef
    • Changes in Brain Metallome/Metabolome Pattern due to a Single i.v. Injection of Manganese in Rats
      Katharina Neth, Marianna Lucio, Alesia Walker, Julia Zorn, Philippe Schmitt-Kopplin, Bernhard Michalke, Fanis Missirlis
      PLOS ONE.2015; 10(9): e0138270.     CrossRef
    • Manganese-induced neurotoxicity: from C. elegans to humans
      Pan Chen, Sudipta Chakraborty, Tanara V. Peres, Aaron B. Bowman, Michael Aschner
      Toxicology Research.2015; 4(2): 191.     CrossRef
    • Complex liaisons moving forward the Parkinson’s disease? An appraisal
      Graziella Rubino
      Basal Ganglia.2015; 5(4): 77.     CrossRef
    • α-Synuclein Protects Against Manganese Neurotoxic Insult During the Early Stages of Exposure in a Dopaminergic Cell Model of Parkinson’s Disease
      Dilshan S. Harischandra, Huajun Jin, Vellareddy Anantharam, Arthi Kanthasamy, Anumantha G. Kanthasamy
      Toxicological Sciences.2015; 143(2): 454.     CrossRef
    • Effects of developmental exposure to manganese and/or low iron diet: Changes to metal transporters, sucrose preference, elevated zero-maze, open-field, and locomotion in response to fenfluramine, amphetamine, and MK-801
      Robyn M. Amos-Kroohs, Colin P. Bloor, Momina A. Qureshi, Charles V. Vorhees, Michael T. Williams
      Toxicology Reports.2015; 2: 1046.     CrossRef
    • Is it possible to enhance the organism’s resistance to toxic effects of metallic nanoparticles?
      Boris A. Katsnelson, Larisa I. Privalova, Marina P. Sutunkova, Ilzira A. Minigalieva, Vladimir B. Gurvich, Vladimir Y. Shur, Oleg H. Makeyev, Irina E. Valamina, Ekaterina V. Grigoryeva
      Toxicology.2015; 337: 79.     CrossRef
    • Silymarin as a Natural Antioxidant: An Overview of the Current Evidence and Perspectives
      Peter Surai
      Antioxidants.2015; 4(1): 204.     CrossRef
    • Attenuation of Combined Nickel(II) Oxide and Manganese(II, III) Oxide Nanoparticles’ Adverse Effects with a Complex of Bioprotectors
      Ilzira Minigalieva, Boris Katsnelson, Larisa Privalova, Marina Sutunkova, Vladimir Gurvich, Vladimir Shur, Ekaterina Shishkina, Irene Valamina, Oleg Makeyev, Vladimir Panov, Anatoly Varaksin, Ekaterina Grigoryeva, Ekaterina Meshtcheryakova
      International Journal of Molecular Sciences.2015; 16(9): 22555.     CrossRef
    • Blood Metal Concentrations of Manganese, Lead, and Cadmium in Relation to Serum Ferritin Levels in Ohio Residents
      Yangho Kim, Danelle T. Lobdell, Chris W. Wright, Vihra V. Gocheva, Edward Hudgens, Rosemarie M. Bowler
      Biological Trace Element Research.2015; 165(1): 1.     CrossRef
    • Neutron Activation Analysis of Scalp Hair from ALS Patients and Residents in the Kii Peninsula, Japan
      Tameko Kihira, Iori Sakurai, Sohei Yoshida, Ikuro Wakayama, Koichi Takamiya, Ryo Okumura, Yuhto Iinuma, Keiko Iwai, Yoshinori Kajimoto, Yasuhiro Hiwatani, Junko Kohmoto, Kazushi Okamoto, Yasumasa Kokubo, Shigeki Kuzuhara
      Biological Trace Element Research.2015; 164(1): 36.     CrossRef
    • Correlation between the biochemical pathways altered by mutated parkinson-related genes and chronic exposure to manganese
      Jerome A. Roth
      NeuroToxicology.2014; 44: 314.     CrossRef
    • Iron Deficiency is Not Associated with Increased Blood Cadmium in Infants
      Jung-Hun Park, Sangkyu Park, Yangho Kim
      Annals of Occupational and Environmental Medicine.2014;[Epub]     CrossRef
    • Manganese leads to an increase in markers of oxidative stress as well as to a shift in the ratio of Fe(ii)/(iii) in rat brain tissue
      Katharina Fernsebner, Julia Zorn, Basem Kanawati, Alesia Walker, Bernhard Michalke
      Metallomics.2014; 6(4): 921.     CrossRef
    • Iron deficiency increases blood concentrations of neurotoxic metals in children
      Yangho Kim, Sangkyu Park
      Korean Journal of Pediatrics.2014; 57(8): 345.     CrossRef
    • Sex-specific Profiles of Blood Metal Levels Associated with Metal–Iron Interactions
      Byung-Kook Lee, Yangho Kim
      Safety and Health at Work.2014; 5(3): 113.     CrossRef
    • Blood Manganese Concentration is Elevated in Infants with Iron Deficiency
      Sangkyu Park, Chang-Sun Sim, Heun Lee, Yangho Kim
      Biological Trace Element Research.2013; 155(2): 184.     CrossRef

    • PubReader PubReader
    • ePub LinkePub Link
    • Cite
      CITE
      export Copy Download
      Close
      Download Citation
      Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

      Format:
      • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
      • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
      Include:
      • Citation for the content below
      Manganese Neurotoxicity: a Focus on Glutamate Transporters
      Ann Occup Environ Med. 2013;25:4-4.   Published online May 21, 2013
      Close
    • XML DownloadXML Download
    Manganese Neurotoxicity: a Focus on Glutamate Transporters
    Manganese Neurotoxicity: a Focus on Glutamate Transporters

    Ann Occup Environ Med : Annals of Occupational and Environmental Medicine
    Close layer
    TOP