1
|
Xie YH, Song HX, Peng JC, Li SJ, Ou SY, Aschner M, Jiang YM. Treatment of manganese and lead poisoning with sodium para-aminosalicylic acid: A contemporary update. Toxicol Lett 2024; 398:69-81. [PMID: 38909920 DOI: 10.1016/j.toxlet.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/08/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
Sodium para-aminosalicylic acid (PAS-Na) treatment for manganese (Mn) intoxication has shown efficacy in experimental and clinical studies, giving rise to additional studies on its efficacy for lead (Pb) neurotoxicity and its associated mechanisms of neuroprotection. The difference between PAS-Na and other metal complexing agents, such as edetate calcium sodium (CaNa2-EDTA), is firstly that PAS-Na can readily pass through the blood-brain barrier (BBB), and complex and facilitate the excretion of manganese and lead. Secondly, PAS-Na has anti-inflammatory effects. Recent studies have broadened the understanding on the mechanisms associated with efficacy of PAS-Na. The latter has been shown to modulate multifarious manganese- and lead- induced neurotoxicity, via its anti-apoptotic and anti-inflammatory effects, as well as its ability to inhibit pyroptosis, and regulate abnormal autophagic processes. These observations provide novel scientific bases and new concepts for the treatment of lead, mercury, copper, thallium, as well as other toxic encephalopathies, and implicate PAS-Na as a compound with greater prospects for clinical medical application.
Collapse
Affiliation(s)
- Yu-Han Xie
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China
| | - Han-Xiao Song
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China
| | - Jian-Chao Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China
| | - Shao-Jun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China
| | - Shi-Yan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
2
|
Mossine VV, Waters JK, Sun GY, Gu Z, Mawhinney TP. Microglia Signaling Pathway Reporters Unveiled Manganese Activation of the Interferon/STAT1 Pathway and Its Mitigation by Flavonoids. Mol Neurobiol 2023; 60:4679-4692. [PMID: 37140843 PMCID: PMC10293393 DOI: 10.1007/s12035-023-03369-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/25/2023] [Indexed: 05/05/2023]
Abstract
Neuroinflammatory responses to neurotoxic manganese (Mn) in CNS have been associated with the Mn-induced Parkinson-like syndromes. However, the framework of molecular mechanisms contributing to manganism is still unclear. Using an in vitro neuroinflammation model based on the insulated signaling pathway reporter transposon constructs stably transfected into a murine BV-2 microglia line, we tested effects of manganese (II) together with a set of 12 metal salts on the transcriptional activities of the NF-κB, activator protein-1 (AP-1), signal transducer and activator of transcription 1 (STAT1), STAT1/STAT2, STAT3, Nrf2, and metal-responsive transcription factor-1 (MTF-1) via luciferase assay, while concatenated destabilized green fluorescent protein expression provided for simultaneous evaluation of cellular viability. This experiment revealed specific and strong responses to manganese (II) in reporters of the type I and type II interferon-induced signaling pathways, while weaker activation of the NF-κB in the microglia was detected upon treatment of cells with Mn(II) and Ba(II). There was a similarity between Mn(II) and interferon-γ in the temporal STAT1 activation profile and in their antagonism to bacterial LPS. Sixty-four natural and synthetic flavonoids differentially affected both cytotoxicity and the pro-inflammatory activity of Mn (II) in the microglia. Whereas flavan-3-ols, flavanones, flavones, and flavonols were cytoprotective, isoflavones enhanced the cytotoxicity of Mn(II). Furthermore, about half of the tested flavonoids at 10-50 μM could attenuate both basal and 100-200 μM Mn(II)-induced activity at the gamma-interferon activated DNA sequence (GAS) in the cells, suggesting no critical roles for the metal chelation or antioxidant activity in the protective potential of flavonoids against manganese in microglia. In summary, results of the study identified Mn as a specific elicitor of the interferon-dependent pathways that can be mitigated by dietary polyphenols.
Collapse
Affiliation(s)
- Valeri V Mossine
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211, USA.
- Agriculture Experiment Station Chemical Laboratories, University of Missouri, Columbia, MO, 65211, USA.
| | - James K Waters
- Agriculture Experiment Station Chemical Laboratories, University of Missouri, Columbia, MO, 65211, USA
| | - Grace Y Sun
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Thomas P Mawhinney
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211, USA
- Agriculture Experiment Station Chemical Laboratories, University of Missouri, Columbia, MO, 65211, USA
- Department of Child Health, University of Missouri, Columbia, MO, 65211, USA
| |
Collapse
|
3
|
Wolf J, Hoffman L, Southern C. Successful Management of Severe Manganese Toxicosis in Two Dogs. J Am Anim Hosp Assoc 2023; 59:152-157. [PMID: 37167249 DOI: 10.5326/jaaha-ms-7348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2023] [Indexed: 05/13/2023]
Abstract
Manganese is a common component of human joint supplements and may be a source of ingestion and subsequent toxicosis in dogs. Although hepatotoxicity secondary to manganese toxicosis has been reported in dogs before, no descriptions of successful management of manganese toxicosis has been reported in veterinary literature. A 5 yr old spayed female Shetland sheepdog and a 5 yr old female Shetland sheepdog were evaluated following accidental ingestion of a joint supplement. Consultation with a toxicologist revealed concern for manganese toxicosis resulting in hepatic injury. Both dogs developed subsequent acute liver injury, despite decontamination and initial management with N-acetylcysteine and cholestyramine. The patients were managed with calcium ethylenediaminetetraacetic acid, paraaminosalicylic acid, allopurinol, Vitamin E, ginkgo biloba, and S-adenosylmethionine/silybin. Liver values returned to normal in both dogs. Manganese exposure was confirmed with urine manganese analysis in one dog and fecal examination in the other dog. A previous case report detailed the fatal manganese toxicosis in a dog; this case report describes the successful management of severe acute hepatic injury secondary to manganese toxicosis. The combination of medications used above may be used for successful treatment of manganese toxicosis in dogs.
Collapse
Affiliation(s)
- Jacob Wolf
- From the College of Veterinary Medicine, University of Florida, Gainesville, Florida (J.W., C.S.); and the United States Army, San Antonio, Texas (L.H.)
| | - Levi Hoffman
- From the College of Veterinary Medicine, University of Florida, Gainesville, Florida (J.W., C.S.); and the United States Army, San Antonio, Texas (L.H.)
| | - Carl Southern
- From the College of Veterinary Medicine, University of Florida, Gainesville, Florida (J.W., C.S.); and the United States Army, San Antonio, Texas (L.H.)
| |
Collapse
|
4
|
McCabe S, Limesand K, Zhao N. Recent progress toward understanding the role of ZIP14 in regulating systemic manganese homeostasis. Comput Struct Biotechnol J 2023; 21:2332-2338. [PMID: 37020930 PMCID: PMC10070054 DOI: 10.1016/j.csbj.2023.03.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/22/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
ZIP14 is a metal transporter essential for the regulation of body manganese homeostasis. The physiological functions of ZIP14 have been uncovered mainly through two lines of in vivo studies that examined the phenotypes of ZIP14 loss, including studies of humans with ZIP14 mutations and animals with ZIP14 deficiency. This mini review aims at presenting an updated view of the important advances made towards understanding the genetic and pathological mechanisms of brain manganese overload caused by ZIP14 deficiency.
Collapse
|
5
|
Aaseth JO, Nurchi VM. Chelation Combination-A Strategy to Mitigate the Neurotoxicity of Manganese, Iron, and Copper? Biomolecules 2022; 12:1713. [PMID: 36421727 PMCID: PMC9687779 DOI: 10.3390/biom12111713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/13/2022] [Accepted: 11/16/2022] [Indexed: 01/19/2024] Open
Abstract
The chelating thiol dimercaptosuccinate (DMSA) and the traditional agent D-penicillamine (PSH) are effective in enhancing the urinary excretion of copper (Cu) and lead (Pb) in poisoned individuals. However, DMSA, PSH, EDTA (ethylenediamine tetraacetate), and deferoxamine (DFOA) are water-soluble agents with limited access to the central nervous system (CNS). Strategies for mobilization of metals such as manganese (Mn), iron (Fe), and Cu from brain deposits may require the combined use of two agents: one water-soluble agent to remove circulating metal into urine, in addition to an adjuvant shuttler to facilitate the brain-to-blood mobilization. The present review discusses the chemical basis of metal chelation and the ligand exchange of metal ions. To obtain increased excretion of Mn, Cu, and Fe, early experiences showed promising results for CaEDTA, PSH, and DFOA, respectively. Recent experiments have indicated that p-amino salicylate (PAS) plus CaEDTA may be a useful combination to remove Mn from binding sites in CNS, while the deferasirox-DFOA and the tetrathiomolybdate-DMSA combinations may be preferable to promote mobilization of Fe and Cu, respectively, from the CNS. Further research is requested to explore benefits of chelator combinations.
Collapse
Affiliation(s)
- Jan O. Aaseth
- Department of Research, Innlandet Hospital Trust, P.O. Box 104, N-2381 Brumunddal, Norway
- Faculty of Health and Social Sciences, Inland Norway University of Applied Sciences, P.O. Box 104, N-2418 Elverum, Norway
| | - Valeria M. Nurchi
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy
| |
Collapse
|
6
|
Atteinte neurologique définitive dans un contexte de consommation intraveineuse de méthcathinone (éphédrone). TOXICOLOGIE ANALYTIQUE ET CLINIQUE 2022. [DOI: 10.1016/j.toxac.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
7
|
OUP accepted manuscript. Metallomics 2022; 14:6564191. [DOI: 10.1093/mtomcs/mfac019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/08/2022] [Indexed: 11/13/2022]
|
8
|
Li J, Deng Y, Peng D, Zhao L, Fang Y, Zhu X, Li S, Aschner M, Ou S, Jiang Y. Sodium P-aminosalicylic Acid Attenuates Manganese-Induced Neuroinflammation in BV2 Microglia by Modulating NF-κB Pathway. Biol Trace Elem Res 2021; 199:4688-4699. [PMID: 33447908 DOI: 10.1007/s12011-021-02581-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
Exposure to high levels of manganese (Mn) leads to brain Mn accumulation, and a disease referred to as manganism. Activation of microglia plays an important role in Mn-induced neuroinflammation. Sodium p-aminosalicylic acid (PAS-Na) is a non-steroidal anti-inflammatory drug that inhibits Mn-induced neuroinflammation. The aim of the current study was to explore the role of NF-κB in the protective mechanism of PAS-Na on Mn-induced neuroinflammation in BV2 microglial experimental model. We treated BV2 microglia with 200 μM Mn for 24 h followed by 48 h treatment with graded concentrations of PAS-Na, using an NF-kB inhibitor, JSH-23, as a positive control. MTT results established that 200 and 400 μM PAS-Na treatment increased the Mn-induced cell viability reduction. NF-κB (P65) mRNA expression and the phosphorylation of p65 were increased in Mn-treated BV2 cell, and suppressed by PAS-Na, analogous to the effect of JSH-23 pretreatment. Furthermore, PAS-Na significantly reduced the contents of the inflammatory cytokine TNF-α and IL-1β, both of which were increased by Mn treatment. The current results show that PAS-Na attenuated Mn-induced inflammation by abrogating the activation of the NF-κB signaling pathways and reduced the release of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Junyan Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Yue Deng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Dongjie Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Lin Zhao
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Yuanyuan Fang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Xiaojuan Zhu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Shaojun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Shiyan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Yueming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| |
Collapse
|
9
|
Mishra A, Dahia A, Jaiswal A. Protective effect of Monoisoamyl-2, 3-Dimercaptosuccinic Acid against Manganese-induced Neurotoxicity in rats. Cent Nerv Syst Agents Med Chem 2021; 21:165-171. [PMID: 34433405 DOI: 10.2174/1871524921666210825093134] [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: 01/10/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Apart from being an essential heavy metal, manganese (Mn) serves as an important component of the antioxidant enzyme system in humans. Overexposure to manganese leads to the development of manganism, which is characterized by motor dysfunction along with neurodegeneration. The management of manganism often utilizes chelation therapy. In this regard, Monoisoamyl-2, 3-Dimercaptosuccinic Acid (MiADMSA) has been reported as a novel arsenic chelator, due to the presence of vicinal sulfhydril group. MiADMSA has been reported to reduce the level in divalent ions (like copper) therefore, it may be hypothesized that MiADMSA would be helpful in Mn-induced neurotoxicity. OBJECTIVE This study is envisaged to explore the protective effect of MiADMSA on Mn-induced neurotoxicity. METHOD Mn exposure was carried out by intraperitoneal administration of Mn (as manganese chloride, 10 mg/kg; i.p.). The animals were treated with MiADMSA (50 mg/kg; p.o.) either alone or in combination with Mn. The effect of different treatments on neurobehavioral functions was observed by assessing spontaneous locomotor activity, motor rotarod test, and depression-like behavior in the forced swim test. After behavioral evaluations, all the animals were sacrificed and the brain and liver were isolated for metal estimations. RESULTS Mn exposure leads to loss of motor coordination as observed in spontaneous locomotor activity and rotarod test. However, treatment with MiADMSA significantly improved motor impairments as compared to Mn exposed animals. Accumulation of Mn in the liver and brain has been recorded with Mn exposure; however, MiADMSA treatment significantly reduced the Mn content from the liver and brain. CONCLUSION The outcome of the study suggests that treatment with MiADMSA reversed Mn-induced neurotoxicity by reducing Mn load. Therefore, the use of MiADMSA may be suggested in manganese toxicity, after careful investigation.
Collapse
Affiliation(s)
- Awanish Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli (NIPER-R), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow (UP)- 226002. India
| | - Anjali Dahia
- Department of Biotechnology, Amity University, Jaipur. India
| | - Amit Jaiswal
- Department of Microbiology, Jiwaji University, Gwalior. India
| |
Collapse
|
10
|
Wu Y, Wei G, Zhao N. Restriction of Manganese Intake Prevents the Onset of Brain Manganese Overload in Zip14-/- Mice. Int J Mol Sci 2021; 22:ijms22136773. [PMID: 34202493 PMCID: PMC8268934 DOI: 10.3390/ijms22136773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 02/08/2023] Open
Abstract
As a newly identified manganese transport protein, ZIP14 is highly expressed in the small intestine and liver, which are the two principal organs involved in regulating systemic manganese homeostasis. Loss of ZIP14 function leads to manganese overload in both humans and mice. Excess manganese in the body primarily affects the central nervous system, resulting in irreversible neurological disorders. Therefore, to prevent the onset of brain manganese accumulation becomes critical. In this study, we used Zip14−/− mice as a model for ZIP14 deficiency and discovered that these mice were born without manganese loading in the brain, but started to hyper-accumulate manganese within 3 weeks after birth. We demonstrated that decreasing manganese intake in Zip14−/− mice was effective in preventing manganese overload that typically occurs in these animals. Our results provide important insight into future studies that are targeted to reduce the onset of manganese accumulation associated with ZIP14 dysfunction in humans.
Collapse
|
11
|
Kulshreshtha D, Ganguly J, Jog M. Manganese and Movement Disorders: A Review. J Mov Disord 2021; 14:93-102. [PMID: 33819420 PMCID: PMC8175808 DOI: 10.14802/jmd.20123] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/19/2020] [Accepted: 12/18/2020] [Indexed: 12/21/2022] Open
Abstract
Scientific and technological advances achieved with industrial expansion have led to an ever-increasing demand for heavy metals. This demand has, in turn, led to increased contamination of soil, water and air with these metals. Chronic exposure to metals may be detrimental not only to occupational workers but also to the nonoccupational population exposed to these metals. Manganese (Mn), a commonly used heavy metal, is an essential cofactor for many enzymatic processes that drive biological functions. However, it is also a potential source of neurotoxicity, particularly in the field of movement disorders. The typical manifestation of Mn overexposure is parkinsonism, which may be difficult to differentiate from the more common idiopathic Parkinson's disease. In addition to environmental exposure to Mn, other potential etiologies causing hypermanganesemia include systemic health conditions, total parenteral nutrition and genetic mutations causing Mn dyshomeostasis. In this review, we critically analyze Mn and discuss its sources of exposure, pathophysiology and clinical manifestations. We have highlighted the global public health impact of Mn and emphasize that movement disorder specialists should record a detailed social and occupational history to ensure that a toxic etiology is not misdiagnosed as a neurodegenerative disease. In the absence of a definite therapeutic option, early diagnosis and timely institution of preventive measures are the keys to managing its toxic effects.
Collapse
Affiliation(s)
- Dinkar Kulshreshtha
- Movement Disorder Centre, London Health Sciences Centre, The University of Western Ontario, Ontario, Canada
| | - Jacky Ganguly
- Movement Disorder Centre, London Health Sciences Centre, The University of Western Ontario, Ontario, Canada
| | - Mandar Jog
- Movement Disorder Centre, London Health Sciences Centre, The University of Western Ontario, Ontario, Canada
| |
Collapse
|
12
|
Ullah I, Zhao L, Hai Y, Fahim M, Alwayli D, Wang X, Li H. "Metal elements and pesticides as risk factors for Parkinson's disease - A review". Toxicol Rep 2021; 8:607-616. [PMID: 33816123 PMCID: PMC8010213 DOI: 10.1016/j.toxrep.2021.03.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Essential metals including iron (Fe) and manganese (Mn) with known physiological functions in human body play an important role in cell homeostasis. Excessive exposure to these essential as well as non-essential metals including mercury (Hg) and Aluminum (Al) may contribute to pathological conditions, including PD. Each metal could be toxic through specific pathways. Epidemiological evidences from occupational and ecological studies besides various in vivo and in vitro studies have revealed the possible pathogenic role and neurotoxicity of different metals. Pesticides are substances that aim to mitigate the harm done by pests to plants and crops, and are extensively used to boost agricultural production. This review provides an outline of our current knowledge on the possible association between metals and PD. We have discussed the potential association between these two, furthermore the chemical properties, biological and toxicological aspects as well as possible mechanisms of Fe, Mn, Cu, Zn, Al, Ca, Pb, Hg and Zn in PD pathogenesis. In addition, we review recent evidence on deregulated microRNAs upon pesticide exposure and possible role of deregulated miRNA and pesticides to PD pathogenesis.
Collapse
Affiliation(s)
- Inam Ullah
- School of Life Sciences, Lanzhou University, China
| | - Longhe Zhao
- School of Pharmacy, Lanzhou University, China
| | - Yang Hai
- School of Pharmacy, Lanzhou University, China
| | | | | | - Xin Wang
- School of Pharmacy, Lanzhou University, China
| | - Hongyu Li
- School of Life Sciences, Lanzhou University, China
- School of Pharmacy, Lanzhou University, China
| |
Collapse
|
13
|
Teodorowski O, Adaszek Ł, Erman Or M, Dokuzeylül B, Ercan AM, Tarhan D, Staniec M, Winiarczyk S. Elevated serum manganese concentration in dogs as a possible predisposing factor of cerebral babesiosis in dogs. Acta Vet Hung 2020; 68:354-360. [PMID: 33372913 DOI: 10.1556/004.2020.00053] [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: 06/09/2020] [Accepted: 09/04/2020] [Indexed: 11/19/2022]
Abstract
The aim of this study was to demonstrate a relationship between the occurrence of clinical signs of brain involvement in dogs with babesiosis and the concentration of manganese (Mn) in their serum. The study included seven dogs with early babesiosis (Group 1), seven dogs with cerebral babesiosis (Group 2) and seven healthy dogs (Group 3). Haematological and biochemical blood tests were performed in all dogs, and the results were analysed statistically. The Mann-Whitney rank test was used to demonstrate the differences in Mn concentrations, as well as other haematological and biochemical parameters between groups. In dogs in Group 2 with cerebral babesiosis, as compared to dogs in Groups 1 and 3, a statistically significant increase in serum Mn concentration was shown (P = 0.002 and P = 0.029) that may have been associated with the development of anaemia and/or impairment of liver function. Given the well-established neurotoxic effects of Mn in humans, experimental rodents and primates, additional studies on the role of Mn in the pathogenesis of the cerebral form of canine babesiosis are warranted.
Collapse
Affiliation(s)
| | - Łukasz Adaszek
- 2Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka str. 30, 20-612 Lublin, Poland
| | - Mehmet Erman Or
- 3Department of Internal Medicine, Faculty of Veterinary Medicine, Istanbul University-Cerrahpasa, Avcilar, Istanbul, Turkey
| | - Banu Dokuzeylül
- 3Department of Internal Medicine, Faculty of Veterinary Medicine, Istanbul University-Cerrahpasa, Avcilar, Istanbul, Turkey
| | - Alev Meltem Ercan
- 4Department of Biophysics, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Fatih, Istanbul, Turkey
| | - Duygu Tarhan
- 4Department of Biophysics, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Fatih, Istanbul, Turkey
| | - Marta Staniec
- 2Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka str. 30, 20-612 Lublin, Poland
| | - Stanisław Winiarczyk
- 2Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka str. 30, 20-612 Lublin, Poland
| |
Collapse
|
14
|
Martins AC, Gubert P, Villas Boas GR, Paes MM, Santamaría A, Lee E, Tinkov AA, Bowman AB, Aschner M. Manganese-induced neurodegenerative diseases and possible therapeutic approaches. Expert Rev Neurother 2020; 20:1109-1121. [PMID: 32799578 PMCID: PMC7657997 DOI: 10.1080/14737175.2020.1807330] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis and prion disease represent important public health concerns. Exposure to high levels of heavy metals such as manganese (Mn) may contribute to their development. AREAS COVERED In this critical review, we address the role of Mn in the etiology of neurodegenerative diseases and discuss emerging treatments of Mn overload, such as chelation therapy. In addition, we discuss natural and synthetic compounds under development as prospective therapeutics. Moreover, bioinformatic approaches to identify new potential targets and therapeutic substances to reverse the neurodegenerative diseases are discussed. EXPERT OPINION Here, the authors highlight the importance of better understanding the molecular mechanisms of toxicity associated with neurodegenerative diseases, and the role of Mn in these diseases. Additional emphasis should be directed to the discovery of new agents to treat Mn-induced diseases, since present day chelator therapies have limited bioavailability. Furthermore, the authors encourage the scientific community to develop research using libraries of compounds to screen those compounds that show efficacy in regulating brain Mn levels. In addition, bioinformatics may provide novel insight for pathways and clinical treatments associated with Mn-induced neurodegeneration, leading to a new direction in Mn toxicological research.
Collapse
Affiliation(s)
- Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Priscila Gubert
- Department of Biochemistry, Laboratory of Immunopathology Keizo Asami, LIKA, Federal, University of Pernambuco, Recife, Brazil
- Postgraduate Program in Pure and Applied Chemistry, Federal University of Western of Bahia, Bahia, Brazil
| | - Gustavo R Villas Boas
- Research Group on Development of Pharmaceutical Products (P&DProFar), Center for Biological and Health Sciences, Federal University of Western Bahia, Barreiras, Bahia, Brazil
| | - Marina Meirelles Paes
- Research Group on Development of Pharmaceutical Products (P&DProFar), Center for Biological and Health Sciences, Federal University of Western Bahia, Barreiras, Bahia, Brazil
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City 14269, Mexico
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32301, USA
| | - Alexey A. Tinkov
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Yaroslavl State University, Yaroslavl, Russia
- Federal Research Centre of Biological Systems and Agro-Technologies of the Russian Academy of Sciences, Orenburg, Russia
| | - Aaron B 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
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
15
|
Horning KJ, Joshi P, Nitin R, Balachandran RC, Yanko FM, Kim K, Christov P, Aschner M, Sulikowski GA, Weaver CD, Bowman AB. Identification of a selective manganese ionophore that enables nonlethal quantification of cellular manganese. J Biol Chem 2020; 295:3875-3890. [PMID: 32047113 PMCID: PMC7086026 DOI: 10.1074/jbc.ra119.009781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/11/2020] [Indexed: 01/14/2023] Open
Abstract
Available assays for measuring cellular manganese (Mn) levels require cell lysis, restricting longitudinal experiments and multiplexed outcome measures. Conducting a screen of small molecules known to alter cellular Mn levels, we report here that one of these chemicals induces rapid Mn efflux. We describe this activity and the development and implementation of an assay centered on this small molecule, named manganese-extracting small molecule (MESM). Using inductively-coupled plasma-MS, we validated that this assay, termed here "manganese-extracting small molecule estimation route" (MESMER), can accurately assess Mn in mammalian cells. Furthermore, we found evidence that MESM acts as a Mn-selective ionophore, and we observed that it has increased rates of Mn membrane transport, reduced cytotoxicity, and increased selectivity for Mn over calcium compared with two established Mn ionophores, calcimycin (A23187) and ionomycin. Finally, we applied MESMER to test whether prior Mn exposures subsequently affect cellular Mn levels. We found that cells receiving continuous, elevated extracellular Mn accumulate less Mn than cells receiving equally-elevated Mn for the first time for 24 h, indicating a compensatory cellular homeostatic response. Use of the MESMER assay versus a comparable detergent lysis-based assay, cellular Fura-2 Mn extraction assay, reduced the number of cells and materials required for performing a similar but cell lethality-based experiment to 25% of the normally required sample size. We conclude that MESMER can accurately quantify cellular Mn levels in two independent cells lines through an ionophore-based mechanism, maintaining cell viability and enabling longitudinal assessment within the same cultures.
Collapse
Affiliation(s)
- Kyle J. Horning
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
| | - Piyush Joshi
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
| | - Rachana Nitin
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
| | | | - Frank M. Yanko
- School of Health Sciences, Purdue University, West Lafayette, Indiana 47907
| | - Kwangho Kim
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232,Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235
| | - Plamen Christov
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Gary A. Sulikowski
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232,Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37212
| | - C. David Weaver
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37212
| | - Aaron B. Bowman
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232,School of Health Sciences, Purdue University, West Lafayette, Indiana 47907, To whom correspondence should be addressed:
Purdue University, 550 Stadium Mall Dr., HAMP 1173A, West Lafayette, IN 47907-2051. E-mail:
| |
Collapse
|
16
|
Iron and other metals in the pathogenesis of Parkinson's disease: Toxic effects and possible detoxification. J Inorg Biochem 2019; 199:110717. [DOI: 10.1016/j.jinorgbio.2019.110717] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/24/2022]
|
17
|
Ammonia-induced mitochondrial impairment is intensified by manganese co-exposure: relevance to the management of subclinical hepatic encephalopathy and cirrhosis-associated brain injury. Clin Exp Hepatol 2019; 5:109-117. [PMID: 31501786 PMCID: PMC6728860 DOI: 10.5114/ceh.2019.85071] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 10/29/2018] [Indexed: 12/30/2022] Open
Abstract
Aim of the study Hepatic encephalopathy (HE) is a neuropsychiatric syndrome ensuing from liver failure. The liver is the major site of ammonia detoxification in the human body. Hence, acute and chronic liver dysfunction can lead to hyperammonemia. Manganese (Mn) is a trace element incorporated in several physiological processes in the human body. Mn is excreted through bile. It has been found that cirrhosis is associated with hyperammonemia as well as body Mn accumulation. The brain is the primary target organ for both ammonia and Mn toxicity. On the other hand, brain mitochondria impairment is involved in the mechanism of Mn and ammonia neurotoxicity. Material and methods The current study was designed to evaluate the effect of Mn and ammonia and their combination on mitochondrial indices of functionality in isolated brain mitochondria. Isolated brain mitochondria were exposed to increasing concentrations of ammonia and Mn alone and/or in combination and several mitochondrial indices were assessed. Results The collapse of mitochondrial membrane potential, increased mitochondrial permeabilization, reactive oxygen species formation, and a significant decrease in mitochondrial dehydrogenase activity and ATP content were evident in Mn-exposed (0.005-1 mM) brain mitochondria. On the other hand, ammonia (0.005-0.5 mM) caused no significant changes in brain mitochondrial function. It was found that co-exposure of the brain mitochondria to Mn and ammonia causes more evident mitochondrial impairment in comparison with Mn and/or ammonia alone. Conclusions These data indicate additive toxicity of ammonia and Mn in isolated brain mitochondria exposed to these neurotoxins.
Collapse
|
18
|
Heidari R. Brain mitochondria as potential therapeutic targets for managing hepatic encephalopathy. Life Sci 2019; 218:65-80. [DOI: 10.1016/j.lfs.2018.12.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/08/2018] [Accepted: 12/16/2018] [Indexed: 02/07/2023]
|
19
|
Atypical Neuropsychiatric Presentation in a Patient Expecting Liver Transplantation. Case Rep Transplant 2018; 2018:4609631. [PMID: 30112246 PMCID: PMC6077324 DOI: 10.1155/2018/4609631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/15/2018] [Accepted: 05/20/2018] [Indexed: 01/17/2023] Open
Abstract
Patients presenting with acute or chronic hepatopathy can develop altered mental status with psychomotor slowing, most commonly indicating encephalopathy. We present the case of a 56-year-old patient who developed subacute atypical neuropsychiatric symptoms including cognitive and behavioural disorganization, manic-like state, and lateralized parkinsonian syndrome. The sequence of events, complete work-up, and detailed neuropsychiatric examination were not compatible with hepatic encephalopathy or delirium; therefore we extended our differential diagnosis and suggested the pathophysiological process described below.
Collapse
|
20
|
SLC39A14 deficiency alters manganese homeostasis and excretion resulting in brain manganese accumulation and motor deficits in mice. Proc Natl Acad Sci U S A 2018; 115:E1769-E1778. [PMID: 29437953 DOI: 10.1073/pnas.1720739115] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Solute carrier family 39, member 14 (SLC39A14) is a transmembrane transporter that can mediate the cellular uptake of zinc, iron, and manganese (Mn). Studies of Slc39a14 knockout (Slc39a14-/-) mice have documented that SLC39A14 is required for systemic growth, hepatic zinc uptake during inflammation, and iron loading of the liver in iron overload. The normal physiological roles of SLC39A14, however, remain incompletely characterized. Here, we report that Slc39a14-/- mice spontaneously display dramatic alterations in tissue Mn concentrations, suggesting that Mn is a main physiological substrate for SLC39A14. Specifically, Slc39a14-/- mice have abnormally low Mn levels in the liver coupled with markedly elevated Mn concentrations in blood and most other organs, especially the brain and bone. Radiotracer studies using 54Mn reveal that Slc39a14-/- mice have impaired Mn uptake by the liver and pancreas and reduced gastrointestinal Mn excretion. In the brain of Slc39a14-/- mice, Mn accumulated in the pons and basal ganglia, including the globus pallidus, a region susceptible to Mn-related neurotoxicity. Brain Mn accumulation in Slc39a14-/- mice was associated with locomotor impairments, as assessed by various behavioral tests. Although a low-Mn diet started at weaning was able to reverse brain Mn accumulation in Slc39a14-/- mice, it did not correct their motor deficits. We conclude that SLC39A14 is essential for efficient Mn uptake by the liver and pancreas, and its deficiency results in impaired Mn excretion and accumulation of the metal in other tissues. The inability of Mn depletion to correct the motor deficits in Slc39a14-/- mice suggests that the motor impairments represent lasting effects of early-life Mn exposure.
Collapse
|
21
|
Zebrafish slc30a10 deficiency revealed a novel compensatory mechanism of Atp2c1 in maintaining manganese homeostasis. PLoS Genet 2017; 13:e1006892. [PMID: 28692648 PMCID: PMC5524415 DOI: 10.1371/journal.pgen.1006892] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 07/24/2017] [Accepted: 06/23/2017] [Indexed: 12/28/2022] Open
Abstract
Recent studies found that mutations in the human SLC30A10 gene, which encodes a manganese (Mn) efflux transporter, are associated with hypermanganesemia with dystonia, polycythemia, and cirrhosis (HMDPC). However, the relationship between Mn metabolism and HMDPC is poorly understood, and no specific treatments are available for this disorder. Here, we generated two zebrafish slc30a10 mutant lines using the CRISPR/Cas9 system. Compared to wild-type animals, mutant adult animals developed significantly higher systemic Mn levels, and Mn accumulated in the brain and liver of mutant embryos in response to exogenous Mn. Interestingly, slc30a10 mutants developed neurological deficits in adulthood, as well as environmental Mn-induced manganism in the embryonic stage; moreover, mutant animals had impaired dopaminergic and GABAergic signaling. Finally, mutant animals developed steatosis, liver fibrosis, and polycythemia accompanied by increased epo expression. This phenotype was rescued partially by EDTA- CaNa2 chelation therapy and iron supplementation. Interestingly, prior to the onset of slc30a10 expression, expressing ATP2C1 (ATPase secretory pathway Ca2+ transporting 1) protected mutant embryos from Mn exposure, suggesting a compensatory role for Atp2c1 in the absence of Slc30a10. Notably, expressing either wild-type or mutant forms of SLC30A10 was sufficient to inhibit the effect of ATP2C1 in response to Mn challenge in both zebrafish embryos and HeLa cells. These findings suggest that either activating ATP2C1 or restoring the Mn-induced trafficking of ATP2C1 can reduce Mn accumulation, providing a possible target for treating HMDPC. Impaired function of the manganese transporter SLC30A10 has been implicated in HMDPC (hypermanganesemia with dystonia, polycythemia, and cirrhosis), an early-onset metabolic disorder clinically characterized by increased systemic Mn levels, neurological impairment, polycythemia, and hepatic injury. No specific treatment is currently available for HMDPC. Moreover, the mechanisms that underlie Mn metabolism are poorly understood, thereby hindering the development of effective treatments. To investigate the physiological processes underlying Mn metabolism and to develop new disease models of HMDPC, we generated two zebrafish slc30a10 mutant lines using the CRISPR/Cas9 system and found that these mutants develop clinical deficits typically associated with HMDPC. Furthermore, we identified a putative compensatory role for ATP2C1 in the absence of SLC30A10 with respect to modulating Mn metabolism. These findings provide a valuable tool for investigating the role of manganese dysregulation in neurological degenerative diseases and which can be used to develop new pharmacological approaches for managing Mn accumulation.
Collapse
|
22
|
Calap-Quintana P, González-Fernández J, Sebastiá-Ortega N, Llorens JV, Moltó MD. Drosophila melanogaster Models of Metal-Related Human Diseases and Metal Toxicity. Int J Mol Sci 2017; 18:E1456. [PMID: 28684721 PMCID: PMC5535947 DOI: 10.3390/ijms18071456] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 06/27/2017] [Accepted: 06/30/2017] [Indexed: 12/21/2022] Open
Abstract
Iron, copper and zinc are transition metals essential for life because they are required in a multitude of biological processes. Organisms have evolved to acquire metals from nutrition and to maintain adequate levels of each metal to avoid damaging effects associated with its deficiency, excess or misplacement. Interestingly, the main components of metal homeostatic pathways are conserved, with many orthologues of the human metal-related genes having been identified and characterized in Drosophila melanogaster. Drosophila has gained appreciation as a useful model for studying human diseases, including those caused by mutations in pathways controlling cellular metal homeostasis. Flies have many advantages in the laboratory, such as a short life cycle, easy handling and inexpensive maintenance. Furthermore, they can be raised in a large number. In addition, flies are greatly appreciated because they offer a considerable number of genetic tools to address some of the unresolved questions concerning disease pathology, which in turn could contribute to our understanding of the metal metabolism and homeostasis. This review recapitulates the metabolism of the principal transition metals, namely iron, zinc and copper, in Drosophila and the utility of this organism as an experimental model to explore the role of metal dyshomeostasis in different human diseases. Finally, a summary of the contribution of Drosophila as a model for testing metal toxicity is provided.
Collapse
Affiliation(s)
- Pablo Calap-Quintana
- Department of Genetics, University of Valencia, Campus of Burjassot, 46100 Valencia, Spain.
| | - Javier González-Fernández
- Department of Genetics, University of Valencia, Campus of Burjassot, 46100 Valencia, Spain.
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain.
| | - Noelia Sebastiá-Ortega
- Department of Genetics, University of Valencia, Campus of Burjassot, 46100 Valencia, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Spain.
| | - José Vicente Llorens
- Department of Genetics, University of Valencia, Campus of Burjassot, 46100 Valencia, Spain.
| | - María Dolores Moltó
- Department of Genetics, University of Valencia, Campus of Burjassot, 46100 Valencia, Spain.
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Spain.
| |
Collapse
|
23
|
Levin OS, Chimagomedova AS, Skripkina NA, Lyashenko EA, Babkina OV. Nonmotor Symptoms in Vascular and Other Secondary Parkinsonism. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 134:1303-1334. [PMID: 28805574 DOI: 10.1016/bs.irn.2017.05.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Vascular parkinsonism (VP) is a relatively frequent variant of secondary parkinsonism caused by ischemic or hemorrhagic lesions of basal ganglia, midbrain, or their links with frontal cortex. According to different investigations, various forms of cerebrovascular disease cause 1%-15% of parkinsonism cases. Nonmotor symptoms are frequently found in VP and may negatively influence on quality of life. However, nonmotor symptoms such as hallucinations, orthostatic hypotension, REM-sleep behavior disorder, and anosmia are rarely revealed in VP, which may be noted to another diagnosis or mixed pathology. Clinical value of nonmotor symptoms in normal pressure hydrocephalus, toxic, and drug-induced parkinsonism is also discussed.
Collapse
Affiliation(s)
- Oleg S Levin
- Russian Medical Academy of Professional Continuous Education, Centre of Extrapyramidal Disorders, Moscow, Russia.
| | - Achcha Sh Chimagomedova
- Russian Medical Academy of Professional Continuous Education, Centre of Extrapyramidal Disorders, Moscow, Russia
| | - Natalia A Skripkina
- Russian Medical Academy of Professional Continuous Education, Centre of Extrapyramidal Disorders, Moscow, Russia
| | - Elena A Lyashenko
- Russian Medical Academy of Professional Continuous Education, Centre of Extrapyramidal Disorders, Moscow, Russia
| | - Olga V Babkina
- Russian Medical Academy of Professional Continuous Education, Centre of Extrapyramidal Disorders, Moscow, Russia
| |
Collapse
|
24
|
Mackenzie Ross S. A reply to Pigatto, Ronchi & Guzzi's commentary. Cortex 2017; 94:202-203. [PMID: 28545676 DOI: 10.1016/j.cortex.2017.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 04/28/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Sarah Mackenzie Ross
- Research Department of Clinical, Educational and Health Psychology, University College London, UK
| |
Collapse
|
25
|
Pigatto PD, Ronchi A, Guzzi G. Neurocognitive disorders and chronic manganese exposure. Cortex 2017; 94:200-201. [PMID: 28372792 DOI: 10.1016/j.cortex.2017.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 10/10/2016] [Accepted: 03/08/2017] [Indexed: 11/16/2022]
Affiliation(s)
- Paolo D Pigatto
- Department of Biomedical, Surgical and Dental Sciences, Unit of Dermatology, IRCCS Galeazzi Hospital, University of Milan, Milan, Italy
| | - Anna Ronchi
- Pavia Poison Control Center and National Toxicology Information Centre, Toxicology Unit, IRCCS Maugeri Foundation and University of Pavia, Italy
| | - Gianpaolo Guzzi
- Italian Association for Metals and Biocompatibility Research - A.I.R.M.E.B., Milan, Italy.
| |
Collapse
|
26
|
Selikhova M, Tripoliti E, Fedoryshyn L, Matvienko Y, Stanetska H, Boychuk M, Komnatska I, Lees A, Sanotsky Y. Analysis of a distinct speech disorder seen in chronic manganese toxicity following Ephedrone abuse. Clin Neurol Neurosurg 2016; 147:71-7. [DOI: 10.1016/j.clineuro.2016.05.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 03/11/2016] [Accepted: 05/29/2016] [Indexed: 11/17/2022]
|
27
|
Yuan ZX, Chen HB, Li SJ, Huang XW, Mo YH, Luo YN, He SN, Deng XF, Lu GD, Jiang YM. The influence of manganese treatment on the distribution of metal elements in rats and the protection by sodium para-amino salicylic acid. J Trace Elem Med Biol 2016; 36:84-9. [PMID: 27259357 DOI: 10.1016/j.jtemb.2016.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 03/11/2016] [Accepted: 04/08/2016] [Indexed: 12/15/2022]
Abstract
Manganese (Mn) overexposure induced neurological damages, which could be potentially protected by sodium para-aminosalicylic acid (PAS-Na). In this study, we systematically detected the changes of divalent metal elements in most of the organs and analyzed the distribution of the metals in Mn-exposed rats and the protection by PAS-Na. Sprague Dawley (SD) rats received intraperitoneal injections of 15mg/kg MnCl2·4H2O (5d/week for 3 weeks), followed by subcutaneous (back) injections of PAS-Na (100 and 200mg/kg, everyday for 5 weeks). The concentrations of Mn and other metal elements [Iron (Fe), Copper (Cu), Zinc (Zn), Magnesium (Mg), Calcium (Ca)] in major organs (liver, spleen, kidney, thighbone and iliac bone, cerebral cortex, hippocampus and testes) and blood by Inductively Coupled Plasma-Atomic Emission Spectrometry (ICP-AES). The results showed that Mn overexposure significantly increased Mn in most organs, Fe and Zn in liver, Fe and Mg in blood; however decreased Fe, Cu, Zn, Mg and Ca in cortex, Cu and Zn in kidney, Cu and Mg in iliac bone, and Zn in blood. In contrast, PAS-Na treatment restored most changes particularly in cortex. In conclusion, excessive Mn exposure disturbed the balance of other metal elements but PAS-Na post-treatments could restore these alterations.
Collapse
Affiliation(s)
- Zong-Xiang Yuan
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Hai-Bin Chen
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China; Center for Disease Control and Prevention, Nanning, PR China
| | - Shao-Jun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Xiao-Wei Huang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Yu-Huan Mo
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Yi-Ni Luo
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Sheng-Nan He
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Xiang-Fa Deng
- Department of Human Anatomy, School of Basic Medicine, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Guo-Dong Lu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China; The Key Laboratory of High-Incidence Diseases Prevention and Control, Guangxi Medical University, Nanning, PR China.
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China.
| |
Collapse
|
28
|
Manganese-Induced Parkinsonism and Parkinson's Disease: Shared and Distinguishable Features. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:7519-40. [PMID: 26154659 PMCID: PMC4515672 DOI: 10.3390/ijerph120707519] [Citation(s) in RCA: 226] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/12/2014] [Accepted: 01/06/2015] [Indexed: 11/30/2022]
Abstract
Manganese (Mn) is an essential trace element necessary for physiological processes that support development, growth and neuronal function. Secondary to elevated exposure or decreased excretion, Mn accumulates in the basal ganglia region of the brain and may cause a parkinsonian-like syndrome, referred to as manganism. The present review discusses the advances made in understanding the essentiality and neurotoxicity of Mn. We review occupational Mn-induced parkinsonism and the dynamic modes of Mn transport in biological systems, as well as the detection and pharmacokinetic modeling of Mn trafficking. In addition, we review some of the shared similarities, pathologic and clinical distinctions between Mn-induced parkinsonism and Parkinson’s disease. Where possible, we review the influence of Mn toxicity on dopamine, gamma aminobutyric acid (GABA), and glutamate neurotransmitter levels and function. We conclude with a survey of the preventive and treatment strategies for manganism and idiopathic Parkinson’s disease (PD).
Collapse
|
29
|
Chen P, Chakraborty S, Mukhopadhyay S, Lee E, Paoliello MMB, Bowman AB, Aschner M. Manganese homeostasis in the nervous system. J Neurochem 2015; 134:601-10. [PMID: 25982296 DOI: 10.1111/jnc.13170] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 04/30/2015] [Accepted: 05/04/2015] [Indexed: 12/31/2022]
Abstract
Manganese (Mn) is an essential heavy metal that is naturally found in the environment. Daily intake through dietary sources provides the necessary amount required for several key physiological processes, including antioxidant defense, energy metabolism, immune function and others. However, overexposure from environmental sources can result in a condition known as manganism that features symptomatology similar to Parkinson's disease (PD). This disorder presents with debilitating motor and cognitive deficits that arise from a neurodegenerative process. In order to maintain a balance between its essentiality and neurotoxicity, several mechanisms exist to properly buffer cellular Mn levels. These include transporters involved in Mn uptake, and newly discovered Mn efflux mechanisms. This review will focus on current studies related to mechanisms underlying Mn import and export, primarily the Mn transporters, and their function and roles in Mn-induced neurotoxicity. Though and essential metal, overexposure to manganese may result in neurodegenerative disease analogous to Parkinson's disease. Manganese homeostasis is tightly regulated by transporters, including transmembrane importers (divalent metal transporter 1, transferrin and its receptor, zinc transporters ZIP8 and Zip14, dopamine transporter, calcium channels, choline transporters and citrate transporters) and exporters (ferroportin and SLC30A10), as well as the intracellular trafficking proteins (SPCA1 and ATP12A2). A manganese-specific sensor, GPP130, has been identified, which affords means for monitoring intracellular levels of this metal.
Collapse
Affiliation(s)
- Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sudipta Chakraborty
- Neuroscience Graduate Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology & Toxicology, College of Pharmacy; Institute for Cellular & Molecular Biology; and Institute for Neuroscience, The University of Texas at Austin, Austin, Texas, USA
| | - Eunsook Lee
- Department of Physiology, Meharry Medical College, Nashville, Tennessee, USA
| | - Monica M B Paoliello
- Graduate Program in Public Health, Department of Pathology, Clinical and Toxicological Analysis, Center of Health Science, State University of Londrina, Parana, Brazil
| | - Aaron B Bowman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA.,Neuroscience Graduate Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
30
|
Neth K, Lucio M, Walker A, Kanawati B, Zorn J, Schmitt-Kopplin P, Michalke B. Diverse Serum Manganese Species Affect Brain Metabolites Depending on Exposure Conditions. Chem Res Toxicol 2015; 28:1434-42. [PMID: 26024413 DOI: 10.1021/acs.chemrestox.5b00104] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Occupational and environmental exposure to increased concentrations of manganese (Mn) can lead to an accumulation of this element in the brain. The consequence is an irreversible damage of dopaminergic neurons leading to a disease called manganism with a clinical presentation similar to the one observed in Parkinson's disease. Human as well as animal studies indicate that Mn is mainly bound to low molecular mass (LMM) compounds such as Mn-citrate when crossing neural barriers. The shift toward LMM compounds might already take place in serum due to elevated Mn concentrations in the body. In this study, we investigated Mn-species pattern in serum in two different animal models by size exclusion chromatography-inductively coupled plasma mass spectrometry (SEC-ICP-MS). A subchronic feeding of rats with elevated levels of Mn led to an increase in LMM compounds, mainly Mn-citrate and Mn bound to amino acids. In addition, a single i.v. injection of Mn showed an increase in Mn-transferrin and Mn bound to amino acids 1 h after injection, while species values were more or less rebalanced 4 days after the injection. Results from Mn-speciation were correlated to the brain metabolome determined by means of electrospray ionization ion cyclotron resonance Fourier transform mass spectrometry (ESI-ICR/FT-MS). The powerful combination of Mn-speciation in serum with metabolomics of the brain underlined the need for Mn-speciation in exposure scenarios instead of the determination of whole Mn concentrations in blood. The progress of Mn-induced neuronal injury might therefore be assessed on the basis of known serum Mn-species.
Collapse
Affiliation(s)
| | | | | | | | | | - Philippe Schmitt-Kopplin
- §Technische Universität München, Chair of Analytical Food Chemistry, Alte Akademie 10, D-85354 Freising, Germany
| | | |
Collapse
|
31
|
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
|
32
|
Dimovasili C, Aschner M, Plaitakis A, Zaganas I. Differential interaction of hGDH1 and hGDH2 with manganese: Implications for metabolism and toxicity. Neurochem Int 2015; 88:60-5. [PMID: 25837286 DOI: 10.1016/j.neuint.2015.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 03/13/2015] [Indexed: 10/23/2022]
Abstract
Manganese (Mn) is an essential trace element that serves as co-factor for many important mammalian enzymes. In humans, the importance of this cation is highlighted by the fact that low levels of Mn cause developmental and metabolic abnormalities and, on the other hand, chronic exposure to excessive amounts of Mn is characterized by neurotoxicity, possibly mediated by perturbation of astrocytic mitochondrial energy metabolism. Here we sought to study the effect of Mn on the two human glutamate dehydrogenases (hGDH1 and hGDH2, respectively), key mitochondrial enzymes involved in numerous cellular processes, including mitochondrial metabolism, glutamate homeostasis and neurotransmission, and cell signaling. Our studies showed that, compared to magnesium (Mg) and calcium (Ca), Mn exerted a significant inhibitory effect on both human isoenzymes with hGDH2 being more sensitive than hGDH1, especially under conditions of low ADP levels. Specifically, in the presence of 0.25 mM ADP, the Mn IC50 was 1.14 ± 0.02 mM and 1.54 ± 0.08 mM for hGDH2 and for hGDH1, respectively (p = 0.0001). Increasing Mn levels potentiated this differential effect, with 3 mM Mn inhibiting hGDH2 by 96.5% and hGDH1 by 70.2%. At 1 mM ADP, the Mn IC50 was 1.84 ± 0.02 mM and 2.04 ± 0.07 mM (p = 0.01) for hGDH2 and hGDH1, respectively, with 3 mM Mn inhibiting hGDH2 by 93.6% and hGDH1 by 70.9%. These results were due to the sigmoidal inhibitory curve of Mn that was more pronounced for hGDH2 than for hGDH1. Indeed, at 0.25 mM, the Hill coefficient value was higher for hGDH2 (3.42 ± 0.20) than for hGDH1 (1.94 ± 0.25; p = 0.0002) indicating that interaction of Mn with hGDH2 was substantially more co-operative than for hGDH1. These findings, showing an enhanced sensitivity of the hGDH2 isoenzyme to Mn, especially at low ADP levels, might be of pathophysiological relevance under conditions of Mn neurotoxicity.
Collapse
Affiliation(s)
- Christina Dimovasili
- Neurology Laboratory, School of Health Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Andreas Plaitakis
- Neurology Laboratory, School of Health Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Ioannis Zaganas
- Neurology Laboratory, School of Health Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece.
| |
Collapse
|
33
|
Sikk K, Taba P. Methcathinone "Kitchen Chemistry" and Permanent Neurological Damage. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 120:257-71. [PMID: 26070761 DOI: 10.1016/bs.irn.2015.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Methcathinone abuse is a significant cause of parkinsonism among young patients in the Eastern European countries. The drug is synthesized from over-the-counter cold remedies containing ephedrine or pseudoephedrine. The final mixture contains a high concentration of manganese if potassium permanganate is used as the oxidant agent. Though manganese is an essential trace element and its homeostasis is well maintained, exposure to a high level of manganese is neurotoxic. The use of manganese-contaminated methcathinone may cause permanent neurological damage and severe disability. Drug users develop a distinctive extrapyramidal syndrome that resembles classic manganese intoxication. Methcathinone could have additive neurotoxic effect to the progression of parkinsonism. The most prevalent symptoms are symmetrical bradykinesia, dystonias, and early postural, gait, and speech impairment. After cessation of exposure, the syndrome is generally irreversible and can even progress.
Collapse
Affiliation(s)
- Katrin Sikk
- Department of Neurology, North-Estonian Regional Hospital, Tallinn, Estonia
| | - Pille Taba
- Department of Neurology and Neurosurgery, University of Tartu, Tartu, Estonia.
| |
Collapse
|
34
|
Dusek P, Roos PM, Litwin T, Schneider SA, Flaten TP, Aaseth J. The neurotoxicity of iron, copper and manganese in Parkinson's and Wilson's diseases. J Trace Elem Med Biol 2015; 31:193-203. [PMID: 24954801 DOI: 10.1016/j.jtemb.2014.05.007] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 05/05/2014] [Accepted: 05/22/2014] [Indexed: 12/14/2022]
Abstract
Impaired cellular homeostasis of metals, particularly of Cu, Fe and Mn may trigger neurodegeneration through various mechanisms, notably induction of oxidative stress, promotion of α-synuclein aggregation and fibril formation, activation of microglial cells leading to inflammation and impaired production of metalloproteins. In this article we review available studies concerning Fe, Cu and Mn in Parkinson's disease and Wilson's disease. In Parkinson's disease local dysregulation of iron metabolism in the substantia nigra (SN) seems to be related to neurodegeneration with an increase in SN iron concentration, accompanied by decreased SN Cu and ceruloplasmin concentrations and increased free Cu concentrations and decreased ferroxidase activity in the cerebrospinal fluid. Available data in Wilson's disease suggest that substantial increases in CNS Cu concentrations persist for a long time during chelating treatment and that local accumulation of Fe in certain brain nuclei may occur during the course of the disease. Consequences for chelating treatment strategies are discussed.
Collapse
Affiliation(s)
- Petr Dusek
- Department of Neurology and Center of Clinical Neuroscience, Charles University in Prague, 1st Faculty of Medicine and General University Hospital in Prague, Czech Republic; Institute of Neuroradiology, University Medicine Göttingen, Göttingen, Germany.
| | - Per M Roos
- Department of Neurology, Division of Clinical Neurophysiology, Oslo University Hospital, Oslo, Norway; Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tomasz Litwin
- 2nd Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland
| | | | - Trond Peder Flaten
- Department of Chemistry, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jan Aaseth
- Department of Medicine, Innlandet Hospital Trust, Kongsvinger Hospital Division, Kongsvinger, Norway
| |
Collapse
|
35
|
Hong L, Xu C, O'Neal S, Bi HC, Huang M, Zheng W, Zeng S. Roles of P-glycoprotein and multidrug resistance protein in transporting para-aminosalicylic acid and its N-acetylated metabolite in mice brain. Acta Pharmacol Sin 2014; 35:1577-85. [PMID: 25418377 PMCID: PMC4261121 DOI: 10.1038/aps.2014.103] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 09/02/2014] [Indexed: 12/27/2022] Open
Abstract
AIM Para-aminosalicylic acid (PAS) is effective in the treatment of manganism-induced neurotoxicity (manganism). In this study we investigated the roles of P-glycoprotein (MDR1a) and multidrug resistance protein (MRP) in transporting PAS and its N-acetylated metabolite AcPAS through blood-brain barrier. METHODS MDR1a-null or wild-type mice were intravenously injected with PAS (200 mg/kg). Thirty minutes after the injection, blood samples and brains were collected, and the concentrations of PAS and AcPAS in brain capillaries and parenchyma were measured using HPLC. Both MDCK-MDR1 and MDCK-MRP1 cells that overexpressed P-gp and MRP1, respectively, were used in two-chamber Transwell transport studies in vitro. RESULTS After injection of PAS, the brain concentration of PAS was substantially higher in MDR1a-null mice than in wild-type mice, but the brain concentration of AcPAS had no significant difference between MDR1a-null mice and wild-type mice. Concomitant injection of PAS with the MRP-specific inhibitor MK-571 (50 mg/kg) further increased the brain concentration of PAS in MDR1a-null mice, and increased the brain concentration of AcPAS in both MDR1a-null mice and wild-type mice. Two-chamber Transwell studies with MDCK-MDR1 cells demonstrated that PAS was not only a substrate but also a competitive inhibitor of P-gp, while AcPAS was not a substrate of P-gp. Two-chamber Transwell studies with the MDCK-MRP1 cells showed that MRP1 had the ability to transport both PAS and AcPAS across the BBB. CONCLUSION P-gp plays a major role in the efflux of PAS from brain parenchyma into blood in mice, while MRP1 is involved in both PAS and AcPAS transport in the brain.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/deficiency
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Acetylation
- Aminosalicylic Acid/administration & dosage
- Aminosalicylic Acid/blood
- Aminosalicylic Acid/pharmacokinetics
- Aminosalicylic Acids/blood
- Aminosalicylic Acids/pharmacokinetics
- Animals
- Biotransformation
- Blood-Brain Barrier/metabolism
- Brain/drug effects
- Brain/metabolism
- Capillary Permeability
- Dogs
- Injections, Intravenous
- Madin Darby Canine Kidney Cells
- Male
- Membrane Transport Modulators/pharmacology
- Mice, Knockout
- Multidrug Resistance-Associated Proteins/antagonists & inhibitors
- Multidrug Resistance-Associated Proteins/genetics
- Multidrug Resistance-Associated Proteins/metabolism
- Tissue Distribution
- Transfection
Collapse
Affiliation(s)
- Lan Hong
- College of Pharmaceutical Sciences, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou 310058, China
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Cong Xu
- College of Pharmaceutical Sciences, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou 310058, China
| | - Stefanie O'Neal
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Hui-chang Bi
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Min Huang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Su Zeng
- College of Pharmaceutical Sciences, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
36
|
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: 5.3] [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
|
37
|
Brazier MW, Wedd AG, Collins SJ. Antioxidant and Metal Chelation-Based Therapies in the Treatment of Prion Disease. Antioxidants (Basel) 2014; 3:288-308. [PMID: 26784872 PMCID: PMC4665489 DOI: 10.3390/antiox3020288] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 02/13/2014] [Accepted: 02/28/2014] [Indexed: 12/31/2022] Open
Abstract
Many neurodegenerative disorders involve the accumulation of multimeric assemblies and amyloid derived from misfolded conformers of constitutively expressed proteins. In addition, the brains of patients and experimental animals afflicted with prion disease display evidence of heightened oxidative stress and damage, as well as disturbances to transition metal homeostasis. Utilising a variety of disease model paradigms, many laboratories have demonstrated that copper can act as a cofactor in the antioxidant activity displayed by the prion protein while manganese has been implicated in the generation and stabilisation of disease-associated conformers. This and other evidence has led several groups to test dietary and chelation therapy-based regimens to manipulate brain metal concentrations in attempts to influence the progression of prion disease in experimental mice. Results have been inconsistent. This review examines published data on transition metal dyshomeostasis, free radical generation and subsequent oxidative damage in the pathogenesis of prion disease. It also comments on the efficacy of trialed therapeutics chosen to combat such deleterious changes.
Collapse
Affiliation(s)
- Marcus W Brazier
- Department of Pathology, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Anthony G Wedd
- The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia.
- School of Chemistry, The University of Melbourne, Victoria 3010, Australia.
| | - Steven J Collins
- Department of Pathology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
38
|
Kilpatrick S, Jacinto A, Foale RD, Tappin SW, Burton C, Frowde PE, Elwood CM, Powell R, Duncan A, Mellanby RJ, Gow AG. Whole blood manganese concentrations in dogs with primary hepatitis. J Small Anim Pract 2014; 55:241-6. [PMID: 24593275 DOI: 10.1111/jsap.12196] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2014] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Increased whole blood manganese concentrations have been reported in humans with primary liver disease. Due to the neurotoxic effects of manganese, altered manganese homeostasis has been linked to the development of hepatic encephalopathy. Whole blood manganese concentrations are increased in cases of canine congenital portosystemic shunts, but it remains unclear whether dogs with primary hepatopathies also have altered manganese homeostasis. METHODS Whole blood manganese concentrations were measured by graphite furnace atomic absorption spectrometry in 21 dogs with primary hepatitis, 65 dogs with a congenital portosystemic shunt, 31 dogs with non-hepatic illnesses and 18 healthy dogs. RESULTS The whole blood manganese concentrations were significantly different between dogs with primary hepatitis, dogs with non-hepatic illnesses and healthy dogs (P=0·002). Dogs with primary hepatitis had significantly increased whole blood manganese concentrations compared with healthy dogs (P<0·05) and dogs with non-hepatic illnesses (P<0·01). Dogs with primary hepatitis had significantly lower whole blood manganese concentration compared with dogs with congenital portosystemic shunts (P=0·0005). CLINICAL SIGNIFICANCE Dogs with primary hepatopathies have increased concentrations of whole blood manganese although these concentrations are not as high as those in dogs with congenital portosystemic shunts. The role of altered manganese homeostasis in canine hepatic encephalopathy is worthy of further study.
Collapse
Affiliation(s)
- S Kilpatrick
- Division of Veterinary Clinical Sciences, The Roslin Institute, Royal (Dick) School of Veterinary Studies, Hospital for Small Animals, The University of Edinburgh, Roslin, Midlothian EH25 9RG
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Mot AI, Wedd AG, Sinclair L, Brown DR, Collins SJ, Brazier MW. Metal attenuating therapies in neurodegenerative disease. Expert Rev Neurother 2014; 11:1717-45. [DOI: 10.1586/ern.11.170] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
40
|
Stepens A, Groma V, Skuja S, Platkājis A, Aldiņš P, Ekšteina I, Mārtiņsone I, Bricis R, Donaghy M. The outcome of the movement disorder in methcathinone abusers: clinical, MRI and manganesemia changes, and neuropathology. Eur J Neurol 2013; 21:199-205. [PMID: 23678867 DOI: 10.1111/ene.12185] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 03/25/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE There is limited knowledge regarding the long-term outcome of the methcathinone/manganese-induced movement disorder. Our purpose was to define prognosis in intravenous methcathinone abusers affected by this distinctive disorder attributed to manganese (Mn) toxicity. Also, neuropathology from a globus pallidus region biopsy from a former user is reported. METHODS Eighteen methcathinone abusers were categorized as active (five), discontinued (four) or former (nine) users. They were reassessed after a median of 32.5 months (range 3.4-59.6) clinically, on rating scales, and with MRI and blood Mn levels. The biopsy was examined ultrastructurally. RESULTS Overall the group showed a slight tendency to deterioration at follow-up on clinical assessment of motor functioning, especially the active users. No significant change occurred on parkinsonian rating scale reassessment. Significant reduction in Mn levels occurred in former users, and decreased T1-weighted hyperintensity on basal ganglia MRI occurred in 3 of 4 former and 2 of 3 discontinued users, despite lack of clinical improvement. The biopsy consisted of white matter showing decompacted myelin sheaths and frequent abnormalities of mitochondria. CONCLUSIONS No improvement in this Mn-induced movement disorder occurs after cessation of methcathinone abuse despite improvement of Mn blood levels and/or MRI abnormalities. Ultrastructural abnormalities in a former user confirm structural damage to white matter is associated with the disorder. Methcathinone/Mn toxicity is an important, disabling and permanent medical sequel of intravenous drug abuse in the former Soviet Union.
Collapse
Affiliation(s)
- A Stepens
- Laboratory for Research in Rehabilitation, Riga Stradins University, Riga, Latvia
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Santos D, Batoreu MC, Aschner M, Marreilha dos Santos A. Comparison between 5-aminosalicylic acid (5-ASA) and para-aminosalicylic acid (4-PAS) as potential protectors against Mn-induced neurotoxicity. Biol Trace Elem Res 2013; 152:113-6. [PMID: 23315311 PMCID: PMC3594465 DOI: 10.1007/s12011-012-9597-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 12/29/2012] [Indexed: 01/12/2023]
Abstract
Manganese (Mn) is an essential metal for biological systems; however, occupational or clinical exposure to high levels of Mn can produce a neurological disorder called manganism. Oxidative stress and neuroinflammation play major roles in the Mn-induced neurodegeneration leading to dysfunction of the basal ganglia. We investigated the toxic effects of MnCl2 in an immortalized rat brain endothelial cell line (RBE4) and the protective effects of the radical scavenging aminosalicylic acids, 5-aminosalicylic acid (5-ASA) and 4-aminosalicylic acid (4-PAS). Mn cytotoxicity was determined with 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) reduction and lactate dehydrogenase (LDH) activity. A significant decrease in MTT reduction concomitant with increased LDH release was noted in RBE4 cells exposed for 24 h to MnCl2 (600 and 800 μM; p < 0.0001). Our results establish that compared to 4-PAS, 5-ASA has greater efficacy in protecting RBE4 cells from Mn-induced neurotoxicity after preexposure to MnCl2 800 μM (p < 0.0001).
Collapse
Affiliation(s)
- Dinamene Santos
- I-Med. UL, Department of Toxicology and Food Sciences, Faculty of Pharmacy, University of Lisbon, Lisbon. Portugal
| | - M Camila Batoreu
- I-Med. UL, Department of Toxicology and Food Sciences, Faculty of Pharmacy, University of Lisbon, Lisbon. Portugal
| | - Michael Aschner
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN37232, USA
| | - A.P. Marreilha dos Santos
- I-Med. UL, Department of Toxicology and Food Sciences, Faculty of Pharmacy, University of Lisbon, Lisbon. Portugal
- Corresponding author: , Phone- 351217946400, Fax- 351217946470
| |
Collapse
|
42
|
Sikk K, Haldre S, Aquilonius SM, Asser A, Paris M, Roose Ä, Petterson J, Eriksson SL, Bergquist J, Taba P. Manganese-induced parkinsonism in methcathinone abusers: bio-markers of exposure and follow-up. Eur J Neurol 2013; 20:915-20. [PMID: 23347399 DOI: 10.1111/ene.12088] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 11/29/2012] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND PURPOSE Methcathinone abuse is a new cause of manganism. The psychostimulant is prepared from pseudoephedrine using potassium permanganate as an oxidant. We describe the clinical, biological, neuroimaging characteristics and follow-up results in a large Estonian cohort of intravenous methcathinone users. METHODS During 2006-2012 we studied 38 methcathinone abusers with a mean age of 33 years. Subjects were rated by the Unified Parkinson's Disease Rating Scale (UPDRS), Hoehn and Yahr (HY), and Schwab and England (SE) rating scales. Twenty-four cases were reassessed 9-70 (20 ± 15) months after the initial evaluation. Manganese (Mn) in plasma and hair was analysed by inductively coupled plasma-atom emission spectrometry. Magnetic resonance imaging (MRI) was performed in 11, and single-photon emission computed tomography (SPECT) with iodobenzamide (IBZM) in eight subjects. RESULTS The average total UPDRS score was 43 ± 21. The most severely affected domains in UPDRS Part III were speech and postural stability, the least affected domain was resting tremor. At follow-up there was worsening of HY and SE rating scales. Subjects had a higher mean level of Mn in hair (2.9 ± 3.8 ppm) than controls (0.82 ± 1.02 ppm), P = 0.02. Plasma Mn concentrations were higher (11.5 ± 6.2 ppb) in active than in former users (5.6 ± 1.8 ppb), P = 0.006. Active methcathinone users had increased MRI T1-signal intensity in the globus pallidus, substantia nigra and periaquaductal gray matter. IBZM-SPECT showed normal symmetric tracer uptake in striatum. CONCLUSION Methcathinone abusers develop a distinctive hypokinetic syndrome. Though the biomarkers of Mn exposure are characteristic only of recent abuse, the syndrome is not reversible.
Collapse
Affiliation(s)
- K Sikk
- Department of Neurology and Neurosurgery, University of Tartu, Tartu, Estonia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
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
|
44
|
Robison G, Zakharova T, Fu S, Jiang W, Fulper R, Barrea R, Marcus MA, Zheng W, Pushkar Y. X-ray fluorescence imaging: a new tool for studying manganese neurotoxicity. PLoS One 2012. [PMID: 23185282 PMCID: PMC3501493 DOI: 10.1371/journal.pone.0048899] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The neurotoxic effect of manganese (Mn) establishes itself in a condition known as manganism or Mn induced parkinsonism. While this condition was first diagnosed about 170 years ago, the mechanism of the neurotoxic action of Mn remains unknown. Moreover, the possibility that Mn exposure combined with other genetic and environmental factors can contribute to the development of Parkinson's disease has been discussed in the literature and several epidemiological studies have demonstrated a correlation between Mn exposure and an elevated risk of Parkinson's disease. Here, we introduce X-ray fluorescence imaging as a new quantitative tool for analysis of the Mn distribution in the brain with high spatial resolution. The animal model employed mimics deficits observed in affected human subjects. The obtained maps of Mn distribution in the brain demonstrate the highest Mn content in the globus pallidus, the thalamus, and the substantia nigra pars compacta. To test the hypothesis that Mn transport into/distribution within brain cells mimics that of other biologically relevant metal ions, such as iron, copper, or zinc, their distributions were compared. It was demonstrated that the Mn distribution does not follow the distributions of any of these metals in the brain. The majority of Mn in the brain was shown to occur in the mobile state, confirming the relevance of the chelation therapy currently used to treat Mn intoxication. In cells with accumulated Mn, it can cause neurotoxic action by affecting the mitochondrial respiratory chain. This can result in increased susceptibility of the neurons of the globus pallidus, thalamus, and substantia nigra pars compacta to various environmental or genetic insults. The obtained data is the first demonstration of Mn accumulation in the substantia nigra pars compacta, and thus, can represent a link between Mn exposure and its potential effects for development of Parkinson's disease.
Collapse
Affiliation(s)
- Gregory Robison
- Department of Physics, Purdue University, West Lafayette, Indiana, United States of America
| | - Taisiya Zakharova
- Department of Physics, Purdue University, West Lafayette, Indiana, United States of America
| | - Sherleen Fu
- School of Health Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Wendy Jiang
- School of Health Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Rachael Fulper
- Department of Physics, Purdue University, West Lafayette, Indiana, United States of America
| | - Raul Barrea
- Advanced Photon Source, Argonne National Laboratory, Argonne, Illinois, United States of America
| | - Matthew A. Marcus
- Advanced Light Source, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Yulia Pushkar
- Department of Physics, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail:
| |
Collapse
|
45
|
Stamelou M, Tuschl K, Chong WK, Burroughs AK, Mills PB, Bhatia KP, Clayton PT. Dystonia with brain manganese accumulation resulting from SLC30A10 mutations: a new treatable disorder. Mov Disord 2012; 27:1317-22. [PMID: 22926781 PMCID: PMC3664426 DOI: 10.1002/mds.25138] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 05/22/2012] [Accepted: 07/03/2012] [Indexed: 11/12/2022] Open
Abstract
Background The first gene causing early-onset generalized dystonia with brain manganese accumulation has recently been identified. Mutations in the SLC30A10 gene, encoding a manganese transporter, cause a syndrome of hepatic cirrhosis, dystonia, polycythemia, and hypermanganesemia. Methods We present 10-year longitudinal clinical features, MRI data, and treatment response to chelation therapy of the originally described patient with a proven homozygous mutation in SLC30A10. Results The patient presented with early-onset generalized dystonia and mild hyperbilirubinemia accompanied by elevated whole-blood manganese levels. T1-sequences in MRI showed hyperintensities in the basal ganglia and cerebellum, characteristic of manganese deposition. Treatment with intravenous disodium calcium edetate led to clinical improvement and reduction of hyperintensities in brain imaging. Conclusions We wish to highlight this rare disorder, which, together with Wilson's disease, is the only potentially treatable inherited metal storage disorder to date, that otherwise can be fatal as a result of complications of cirrhosis. © 2012 Movement Disorder Society
Collapse
Affiliation(s)
- Maria Stamelou
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
46
|
Charradi K, Elkahoui S, Karkouch I, Limam F, Hassine FB, Aouani E. Grape seed and skin extract prevents high-fat diet-induced brain lipotoxicity in rat. Neurochem Res 2012; 37:2004-13. [PMID: 22684284 DOI: 10.1007/s11064-012-0821-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 05/26/2012] [Accepted: 05/28/2012] [Indexed: 11/30/2022]
Abstract
Obesity is related to an elevated risk of dementia and the physiologic mechanisms whereby fat adversely affects the brain are poorly understood. The present investigation analyzed the effect of a high fat diet (HFD) on brain steatosis and oxidative stress and the intracellular mediators involved in signal transduction, as well as the protection offered by grape seed and skin extract (GSSE). HFD induced ectopic deposition of cholesterol and phospholipid but not triglyceride. Moreover brain lipotoxicity is linked to an oxidative stress characterized by increased lipoperoxidation and carbonylation, inhibition of glutathione peroxidase and superoxide dismutase activities, depletion of manganese and a concomitant increase in ionizable calcium and acetylcholinesterase activity. Importantly GSSE alleviated all the deleterious effects of HFD treatment. Altogether our data indicated that HFD could find some potential application in the treatment of manganism and that GSSE should be used as a safe anti-lipotoxic agent in the prevention and treatment of fat-induced brain injury.
Collapse
Affiliation(s)
- Kamel Charradi
- Laboratoire des Substances Bioactives, Centre de Biotechnologie de Borj Cedria, BP-901, 2050 Hammam-Lif, Tunisia
| | | | | | | | | | | |
Collapse
|
47
|
Tuschl K, Clayton P, Gospe S, Gulab S, Ibrahim S, Singhi P, Aulakh R, Ribeiro R, Barsottini O, Zaki M, Del Rosario M, Dyack S, Price V, Rideout A, Gordon K, Wevers R, “Kling” Chong W, Mills P. Syndrome of hepatic cirrhosis, dystonia, polycythemia, and hypermanganesemia caused by mutations in SLC30A10, a manganese transporter in man. Am J Hum Genet 2012; 90:457-66. [PMID: 22341972 DOI: 10.1016/j.ajhg.2012.01.018] [Citation(s) in RCA: 232] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 01/01/2012] [Accepted: 01/25/2012] [Indexed: 01/06/2023] Open
Abstract
Environmental manganese (Mn) toxicity causes an extrapyramidal, parkinsonian-type movement disorder with characteristic magnetic resonance images of Mn accumulation in the basal ganglia. We have recently reported a suspected autosomal recessively inherited syndrome of hepatic cirrhosis, dystonia, polycythemia, and hypermanganesemia in cases without environmental Mn exposure. Whole-genome mapping of two consanguineous families identified SLC30A10 as the affected gene in this inherited type of hypermanganesemia. This gene was subsequently sequenced in eight families, and homozygous sequence changes were identified in all affected individuals. The function of the wild-type protein and the effect of sequence changes were studied in the manganese-sensitive yeast strain Δpmr1. Expressing human wild-type SLC30A10 in the Δpmr1 yeast strain rescued growth in high Mn conditions, confirming its role in Mn transport. The presence of missense (c.266T>C [p.Leu89Pro]) and nonsense (c.585del [p.Thr196Profs(∗)17]) mutations in SLC30A10 failed to restore Mn resistance. Previously, SLC30A10 had been presumed to be a zinc transporter. However, this work has confirmed that SLC30A10 functions as a Mn transporter in humans that, when defective, causes Mn accumulation in liver and brain. This is an important step toward understanding Mn transport and its role in neurodegenerative processes.
Collapse
|
48
|
Abstract
Manganese-induced parkinsonism has been recognized since 1837. It has been reported primarily in miners, grinders, and smelters since that time. More recently, isolated case reports involving welders have appeared in the medical literature. Manganism can be distinguished from other forms of parkinsonism by clinical presentation with support from laboratory and radiologic findings. The controversy regarding the risk of parkinsonism in welders is reviewed.
Collapse
Affiliation(s)
- Brent Furbee
- Indiana Poison Center, Indiana University School of Medicine, B408 Methodist Hospital, Indianapolis, IN 46206, USA.
| |
Collapse
|
49
|
Sikk K, Haldre S, Aquilonius SM, Taba P. Manganese-Induced Parkinsonism due to Ephedrone Abuse. PARKINSON'S DISEASE 2011; 2011:865319. [PMID: 21403909 PMCID: PMC3043321 DOI: 10.4061/2011/865319] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2010] [Accepted: 01/09/2011] [Indexed: 11/20/2022]
Abstract
During recent years, a syndrome of hypokinesia, dysarthria, dystonia, and postural impairment, related to intravenous use of a "designer" psychostimulant derived from pseudoephedrine using potassium permanganate as the oxidant, has been observed in drug addicts in several countries in Eastern Europe with some cases also in Western countries. A levodopa unresponsive Parkinsonian syndrome occurs within a few months of abusing the homemade drug mixture containing ephedrone (methcathinone) and manganese. The development of this neurological syndrome has been attributed to toxic effects of manganese, but the role of the psychostimulant ephedrone is unclear. This paper describes the clinical syndrome, results of neuroimaging, and therapeutic attempts.
Collapse
Affiliation(s)
- Katrin Sikk
- Department of Neurology and Neurosurgery, University of Tartu, L. Puusepa 8, 51014 Tartu, Estonia
| | - Sulev Haldre
- Department of Neurology and Neurosurgery, University of Tartu, L. Puusepa 8, 51014 Tartu, Estonia
| | - Sten-Magnus Aquilonius
- Department of Neuroscience, Neurology, Uppsala University Hospital, 75185 Uppsala, Sweden
| | - Pille Taba
- Department of Neurology and Neurosurgery, University of Tartu, L. Puusepa 8, 51014 Tartu, Estonia
| |
Collapse
|
50
|
Hobson A, Seixas N, Sterling D, Racette BA. Estimation of particulate mass and manganese exposure levels among welders. THE ANNALS OF OCCUPATIONAL HYGIENE 2011; 55:113-25. [PMID: 20870928 PMCID: PMC3020674 DOI: 10.1093/annhyg/meq069] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 08/03/2010] [Indexed: 11/14/2022]
Abstract
BACKGROUND Welders are frequently exposed to Manganese (Mn), which may increase the risk of neurological impairment. Historical exposure estimates for welding-exposed workers are needed for epidemiological studies evaluating the relationship between welding and neurological or other health outcomes. The objective of this study was to develop and validate a multivariate model to estimate quantitative levels of welding fume exposures based on welding particulate mass and Mn concentrations reported in the published literature. METHODS Articles that described welding particulate and Mn exposures during field welding activities were identified through a comprehensive literature search. Summary measures of exposure and related determinants such as year of sampling, welding process performed, type of ventilation used, degree of enclosure, base metal, and location of sampling filter were extracted from each article. The natural log of the reported arithmetic mean exposure level was used as the dependent variable in model building, while the independent variables included the exposure determinants. Cross-validation was performed to aid in model selection and to evaluate the generalizability of the models. RESULTS A total of 33 particulate and 27 Mn means were included in the regression analysis. The final model explained 76% of the variability in the mean exposures and included welding process and degree of enclosure as predictors. There was very little change in the explained variability and root mean squared error between the final model and its cross-validation model indicating the final model is robust given the available data. CONCLUSIONS This model may be improved with more detailed exposure determinants; however, the relatively large amount of variance explained by the final model along with the positive generalizability results of the cross-validation increases the confidence that the estimates derived from this model can be used for estimating welder exposures in absence of individual measurement data.
Collapse
Affiliation(s)
- Angela Hobson
- Department of Neurology, School of Medicine, Washington University, St Louis, MO 63116, USA
| | - Noah Seixas
- Department of Environmental and Occupational Health Sciences, School of Public Health and Community Medicine, University of Washington, Seattle, WA 98105, USA
| | - David Sterling
- Department of Environmental and Occupational Health, School of Public Health, University of North Texas, Fort Worth, TX 76107, USA
| | - Brad A. Racette
- Department of Neurology, School of Medicine, Washington University, St Louis, MO 63116, USA
| |
Collapse
|