1
|
Zhang Z, Yang J, Zhou Q, Zhong S, Liu J, Zhang X, Chang X, Wang H. The cGAS-STING-mediated ROS and ferroptosis are involved in manganese neurotoxicity. J Environ Sci (China) 2025; 152:71-86. [PMID: 39617588 DOI: 10.1016/j.jes.2024.05.003] [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: 01/31/2024] [Revised: 04/12/2024] [Accepted: 05/01/2024] [Indexed: 12/12/2024]
Abstract
Manganese (Mn) has been characterized as an environmental pollutant. Excessive releases of Mn due to human activities have increased Mn levels in the environment over the years, posing a threat to human health and the environment. Long-term exposure to high concentrations of Mn can induce neurotoxicity. Therefore, toxicological studies on Mn are of paramount importance. Mn induces oxidative stress through affecting the level of reactive oxygen species (ROS), and the overabundance of ROS further triggers ferroptosis. Additionally, Mn2+ was found to be a novel activator of the cyclic guanosine-adenosine synthase (cGAS)-stimulator of interferon genes (STING) pathway in the innate immune system. Thus, we speculate that Mn exposure may promote ROS production by activating the cGAS-STING pathway, which further induces oxidative stress and ferroptosis, and ultimately triggers Mn neurotoxicity. This review discusses the mechanism between Mn-induced oxidative stress and ferroptosis via activation of the cGAS-STING pathway, which may offer a prospective direction for future in-depth studies on the mechanism of Mn neurotoxicity.
Collapse
Affiliation(s)
- Zhimin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jirui Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qiongli Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Shiyin Zhong
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jingjing Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
2
|
Martins AC, Oliveira-Paula GH, Tinkov AA, Skalny AV, Tizabi Y, Bowman AB, Aschner M. Role of manganese in brain health and disease: Focus on oxidative stress. Free Radic Biol Med 2025; 232:306-318. [PMID: 40086492 PMCID: PMC11985276 DOI: 10.1016/j.freeradbiomed.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Manganese (Mn) is an essential trace element crucial for various physiological processes, but excessive exposure can lead to significant health concerns, particularly neurotoxicity. This review synthesizes current knowledge on Mn-induced oxidative stress and its role in cellular dysfunction and disease. We discuss how Mn promotes toxicity through multiple mechanisms, primarily through reactive oxygen species (ROS) generation, which leads to oxidative stress and disruption of cellular processes. The review examines key pathways affected by Mn toxicity, including mitochondrial dysfunction, endoplasmic reticulum stress, inflammasome activation, and epigenetic modifications. Recent studies have identified promising therapeutic compounds, including both synthetic and natural substances such as probucol, metformin, curcumin, resveratrol, and daidzein, which demonstrate protective effects through various mechanisms, including antioxidant enhancement, mitochondrial function preservation, and epigenetic pathway modulation. Understanding these mechanisms provides new insights into potential therapeutic strategies for Mn-induced disorders. This review also highlights future research directions, emphasizing the need for developing targeted therapies and investigating combination approaches to address multiple aspects of Mn toxicity simultaneously.
Collapse
Affiliation(s)
- Airton C Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Gustavo H Oliveira-Paula
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Alexey A Tinkov
- Institute of Bioelementology, Orenburg State University, Orenburg, 460000, Russia; IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003, Russia
| | - Anatoly V Skalny
- Institute of Bioelementology, Orenburg State University, Orenburg, 460000, Russia; IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003, Russia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington DC, 20059, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
3
|
Li Y, Peng JC, Fang YY, Qin LM, Aschner M, Jiang YM. Effects of Subchronic Manganese and Iron Exposure, Alone or in Combination, on Elemental Distribution in Rats. Biol Trace Elem Res 2025:10.1007/s12011-025-04626-w. [PMID: 40261544 DOI: 10.1007/s12011-025-04626-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
Overexposure to manganese (Mn) or iron (Fe) may lead to neurological damage. The aim of this study was to investigate the effects of subchronic Mn and Fe exposure, alone or in combination, on the distribution of other elements and the relationship between Mn and Fe levels in whole blood and brain. Forty male Sprague-Dawley (SD) rats were divided into control, Mn-exposed, Fe-exposed, and combined Mn-Fe-exposed groups, with 10 rats assigned randomly to each group. The control, Mn-exposed, Fe-exposed group and the combined Mn-Fe-exposed groups were injected intraperitoneally with equal amounts of saline, 5 mg/kg MnCl2, 20 mg/kg FeSO4 or 5 mg/kg MnCl2+20 mg/kg FeSO4 once a day, 5 days a week for 8 weeks. The levels of Mn, Fe and other metallic elements [including barium (Ba), beryllium (Be), strontium (Sr), antimony (Sb), lead (Pb), vanadium (V) and copper (Cu)] in whole blood and brain tissue (including the globus pallidus, hippocampus, striatum and substantia nigra) were determined by inductively coupled plasma-mass spectrometry (ICP-MS). The results of this study show that, in whole blood, Mn levels were increased (p < 0.05) in the Mn-exposed group, Fe levels were decreased in both the Fe-exposed (p < 0.05) and Mn-exposed groups (p < 0.01), and Sb levels were increased in both the Mn-exposed and combined Mn-Fe-exposed groups (p < 0.05). In the substantia nigra, the levels of Be (p < 0.01), Sr (p < 0.05), and Cu (p < 0.001) were increased in the Fe-exposed group; the levels of Cu were also significantly increased in the Mn-exposed group (p < 0.01) and the combined Mn-Fe-exposed group (p < 0.0001); the levels of V were decreased (p < 0.05) in the combined Mn-Fe-exposed group; and the levels of V were decreased in the Fe-exposed group (p < 0.01 ), Mn-exposed group (p < 0.05) and combined Mn-Fe-exposed group (p < 0.001) had decreased Ba levels. In the pallidum, Fe levels were increased in the Mn-Fe co-exposed group (p < 0.0001); Ba (p < 0.01) and Pb (p < 0.05) levels were decreased in the Fe-exposed group; Ba (p < 0.05) levels were decreased in the Mn-exposed group; and Ba levels were increased in the Mn-Fe co-exposed group (p < 0.05). In the hippocampus, Mn (p < 0.01), Cu (p < 0.05), Sb (p < 0.01), and V (p < 0.05) levels were increased in the Fe-exposed group; Mn levels were increased in the Mn-exposed group (p < 0.01) and the combined Mn-Fe-exposed group (p < 0.0001). In the striatum, Be levels were decreased in the Mn-Fe combined exposure group (p < 0.05). Mn and Fe levels in whole blood and brain tissue can reflect the accumulation of Mn and Fe. These measurements can serve as valuable predictive biomarkers for subchronic Mn or Fe exposure and combined Mn-Fe exposure. The interactions between Mn and Fe and the distribution and abnormalities of the essential metal elements in the central nervous system and other organs need to be further investigated.
Collapse
Affiliation(s)
- Yan Li
- Guangxi Zhuang Autonomous Region Institute for the Prevention and Treatment of Occupational Disease, Nanning, 530021, China
| | - Jian-Chao Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd, Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Guangxi Medical University, No. 22, Shuang-yong Rd, Nanning, 530021, Guangxi, China
| | - Yuan-Yuan Fang
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd, Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Guangxi Medical University, No. 22, Shuang-yong Rd, Nanning, 530021, Guangxi, China
| | - Li-Mei Qin
- Guangxi Zhuang Autonomous Region Institute for the Prevention and Treatment of Occupational Disease, Nanning, 530021, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd, Nanning, 530021, Guangxi, China.
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Guangxi Medical University, No. 22, Shuang-yong Rd, Nanning, 530021, Guangxi, China.
| |
Collapse
|
4
|
Chen L, Shen Q, Liu Y, Zhang Y, Sun L, Ma X, Song N, Xie J. Homeostasis and metabolism of iron and other metal ions in neurodegenerative diseases. Signal Transduct Target Ther 2025; 10:31. [PMID: 39894843 PMCID: PMC11788444 DOI: 10.1038/s41392-024-02071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/24/2024] [Accepted: 11/12/2024] [Indexed: 02/04/2025] Open
Abstract
As essential micronutrients, metal ions such as iron, manganese, copper, and zinc, are required for a wide range of physiological processes in the brain. However, an imbalance in metal ions, whether excessive or insufficient, is detrimental and can contribute to neuronal death through oxidative stress, ferroptosis, cuproptosis, cell senescence, or neuroinflammation. These processes have been found to be involved in the pathological mechanisms of neurodegenerative diseases. In this review, the research history and milestone events of studying metal ions, including iron, manganese, copper, and zinc in neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD), will be introduced. Then, the upstream regulators, downstream effector, and crosstalk of mental ions under both physiologic and pathologic conditions will be summarized. Finally, the therapeutic effects of metal ion chelators, such as clioquinol, quercetin, curcumin, coumarin, and their derivatives for the treatment of neurodegenerative diseases will be discussed. Additionally, the promising results and limitations observed in clinical trials of these metal ion chelators will also be addressed. This review will not only provide a comprehensive understanding of the role of metal ions in disease development but also offer perspectives on their modulation for the prevention or treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Leilei Chen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Qingqing Shen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yingjuan Liu
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yunqi Zhang
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Liping Sun
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Xizhen Ma
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Ning Song
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Junxia Xie
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China.
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China.
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China.
| |
Collapse
|
5
|
Khan MA. Targeting Iron Responsive Elements (IREs) of APP mRNA into Novel Therapeutics to Control the Translation of Amyloid-β Precursor Protein in Alzheimer's Disease. Pharmaceuticals (Basel) 2024; 17:1669. [PMID: 39770511 PMCID: PMC11677800 DOI: 10.3390/ph17121669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
The hallmark of Alzheimer's disease (AD) is the buildup of amyloid-β (Aβ), which is produced when the amyloid precursor protein (APP) misfolds and deposits as neurotoxic plaques in the brain. A functional iron responsive element (IRE) RNA stem loop is encoded by the APP 5'-UTR and may be a target for regulating the production of Alzheimer's amyloid precursor protein. Since modifying Aβ protein expression can give anti-amyloid efficacy and protective brain iron balance, targeted regulation of amyloid protein synthesis through modulation of 5'-UTR sequence function is a novel method for the prospective therapy of Alzheimer's disease. Numerous mRNA interference strategies target the 2D RNA structure, even though messenger RNAs like tRNAs and rRNAs can fold into complex, three-dimensional structures, adding even another level of complexity. The IRE family is among the few known 3D mRNA regulatory elements. This review seeks to describe the structural and functional aspects of IREs in transcripts, including that of the amyloid precursor protein, that are relevant to neurodegenerative diseases, including AD. The mRNAs encoding the proteins involved in iron metabolism are controlled by this family of similar base sequences. Like ferritin IRE RNA in their 5'-UTR, iron controls the production of APP in their 5'-UTR. Iron misregulation by iron regulatory proteins (IRPs) can also be investigated and contrasted using measurements of the expression levels of tau production, Aβ, and APP. The development of AD is aided by iron binding to Aβ, which promotes Aβ aggregation. The development of small chemical therapeutics to control IRE-modulated expression of APP is increasingly thought to target messenger RNAs. Thus, IRE-modulated APP expression in AD has important therapeutic implications by targeting mRNA structures.
Collapse
Affiliation(s)
- Mateen A Khan
- Department of Life Science, College of Science and General Studies, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
6
|
Li S, Zhua Y, Liu X. Parkinsonism in liver diseases or dysfunction. Med Clin (Barc) 2024; 163:461-468. [PMID: 38955605 DOI: 10.1016/j.medcli.2024.04.022] [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: 04/11/2024] [Accepted: 04/20/2024] [Indexed: 07/04/2024]
Abstract
Parkinsonism in liver diseases or dysfunction, mainly including neurological manifestations in hereditary liver diseases and neurological complications of advanced liver diseases, occur in isolation or in combination with other movement disorders, and progress along disease course. Prominent akinetic-rigidity syndrome, various onset and progression, poor levodopa response and metabolism abnormalities reflected by serum biomarkers and neuroimaging, make this atypical parkinsonism recognizable and notable in clinical practice. Different susceptibility of brain areas, especially in basal ganglia, to manganese, iron, copper, ammonia overload, together with subsequent oxidative stress, neurotransmitter alterations, disturbed glia-neuron homeostasis and eventually neurotoxicity, contribute to parkinsonism under the circumstances of insufficient liver clearance ability. These mechanisms are interrelated and may interact collectively, adding to the complexity of clinical manifestations and treatment responses. This review summarizes shared clinical features of parkinsonism in liver diseases or dysfunction, depicts their underlying mechanisms and suggests practical flowchart for differential diagnosis.
Collapse
Affiliation(s)
- Sichen Li
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxia Zhua
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xi Liu
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
7
|
Cahill CM, Sarang SS, Bakshi R, Xia N, Lahiri DK, Rogers JT. Neuroprotective Strategies and Cell-Based Biomarkers for Manganese-Induced Toxicity in Human Neuroblastoma (SH-SY5Y) Cells. Biomolecules 2024; 14:647. [PMID: 38927051 PMCID: PMC11201412 DOI: 10.3390/biom14060647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/24/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Manganese (Mn) is an essential heavy metal in the human body, while excess Mn leads to neurotoxicity, as observed in this study, where 100 µM of Mn was administered to the human neuroblastoma (SH-SY5Y) cell model of dopaminergic neurons in neurodegenerative diseases. We quantitated pathway and gene changes in homeostatic cell-based adaptations to Mn exposure. Utilizing the Gene Expression Omnibus, we accessed the GSE70845 dataset as a microarray of SH-SY5Y cells published by Gandhi et al. (2018) and applied statistical significance cutoffs at p < 0.05. We report 74 pathway and 10 gene changes with statistical significance. ReactomeGSA analyses demonstrated upregulation of histones (5 out of 10 induced genes) and histone deacetylases as a neuroprotective response to remodel/mitigate Mn-induced DNA/chromatin damage. Neurodegenerative-associated pathway changes occurred. NF-κB signaled protective responses via Sirtuin-1 to reduce neuroinflammation. Critically, Mn activated three pathways implicating deficits in purine metabolism. Therefore, we validated that urate, a purine and antioxidant, mitigated Mn-losses of viability in SH-SY5Y cells. We discuss Mn as a hypoxia mimetic and trans-activator of HIF-1α, the central trans-activator of vascular hypoxic mitochondrial dysfunction. Mn induced a 3-fold increase in mRNA levels for antioxidant metallothionein-III, which was induced 100-fold by hypoxia mimetics deferoxamine and zinc.
Collapse
Affiliation(s)
- Catherine M. Cahill
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| | - Sanjan S. Sarang
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| | - Rachit Bakshi
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| | - Ning Xia
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| | - Debomoy K. Lahiri
- Department of Psychiatry and Medical & Molecular Genetics, Indiana Alzheimer’s Disease Research Center, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Jack T. Rogers
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| |
Collapse
|
8
|
Aschner M, Martins AC, Oliveira-Paula GH, Skalny AV, Zaitseva IP, Bowman AB, Kirichuk AA, Santamaria A, Tizabi Y, Tinkov AA. Manganese in autism spectrum disorder and attention deficit hyperactivity disorder: The state of the art. Curr Res Toxicol 2024; 6:100170. [PMID: 38737010 PMCID: PMC11088232 DOI: 10.1016/j.crtox.2024.100170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/27/2024] [Accepted: 04/23/2024] [Indexed: 05/14/2024] Open
Abstract
The objective of the present narrative review was to synthesize existing clinical and epidemiological findings linking manganese (Mn) exposure biomarkers to autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD), and to discuss key pathophysiological mechanisms of neurodevelopmental disorders that may be affected by this metal. Existing epidemiological data demonstrated both direct and inverse association between Mn body burden and ASD, or lack of any relationship. In contrast, the majority of studies revealed significantly higher Mn levels in subjects with ADHD, as well as direct relationship between Mn body burden with hyperactivity and inattention scores in children, although several studies reported contradictory results. Existing laboratory studies demonstrated that impaired attention and hyperactivity in animals following Mn exposure was associated with dopaminergic dysfunction and neuroinflammation. Despite lack of direct evidence on Mn-induced neurobiological alterations in patients with ASD and ADHD, a plethora of studies demonstrated that neurotoxic effects of Mn overexposure may interfere with key mechanisms of pathogenesis inherent to these neurodevelopmental disorders. Specifically, Mn overload was shown to impair not only dopaminergic neurotransmission, but also affect metabolism of glutamine/glutamate, GABA, serotonin, noradrenaline, thus affecting neuronal signaling. In turn, neurotoxic effects of Mn may be associated with its ability to induce oxidative stress, apoptosis, and neuroinflammation, and/or impair neurogenesis. Nonetheless, additional detailed studies are required to evaluate the association between environmental Mn exposure and/or Mn body burden and neurodevelopmental disorders at a wide range of concentrations to estimate the potential dose-dependent effects, as well as environmental and genetic factors affecting this association.
Collapse
Affiliation(s)
- Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | | | - Anatoly V. Skalny
- Department of Medical Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
| | - Irina P. Zaitseva
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl 150003, Russia
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, USA
| | - Anatoly A. Kirichuk
- Department of Medical Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - Abel Santamaria
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Cuidado de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City 04960, Mexico
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059, USA
| | - Alexey A. Tinkov
- Department of Medical Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl 150003, Russia
| |
Collapse
|
9
|
Ivantsik O, John A, Kydonopoulou K, Mitropoulos K, Gerou S, Ali BR, Patrinos GP. Novel Pathogenic Variants Leading to Sporadic Amyotrophic Lateral Sclerosis in Greek Patients. Genes (Basel) 2024; 15:309. [PMID: 38540370 PMCID: PMC10970271 DOI: 10.3390/genes15030309] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 06/14/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive disease that affects motor neurons, leading to paralysis and death usually 3-5 years after the onset of symptoms. The investigation of both sporadic and familial ALS highlighted four main genes that contribute to the pathogenesis of the disease: SOD1, FUS, TARDBP and C9orf72. This study aims to provide a comprehensive investigation of genetic variants found in SOD1, FUS and TARDBP genes in Greek sporadic ALS (sALS) cases. Our sequencing analysis of the coding regions of the abovementioned genes that include the majority of the variants that lead to ALS in 32 sALS patients and 3 healthy relatives revealed 6 variants in SOD1, 19 variants in FUS and 37 variants in TARDBP, of which the SOD1 p.D90A and the FUS c.*356G>A (rs886051940) variants have been previously associated with ALS, while two novel nonsense pathogenic variants were also identified, namely FUS p.R241* and TDP-43 p.Y214*. Our study contributes to the worldwide effort toward clarifying the genetic basis of sALS to better understand the disease's molecular pathology.
Collapse
Affiliation(s)
- Ouliana Ivantsik
- Laboratory of Pharmacogenomics and Individualized Therapy, Division of Pharmacology and Biosciences, Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Rion, Greece
| | - Anne John
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 15551, United Arab Emirates
| | | | - Konstantinos Mitropoulos
- Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Spyridon Gerou
- ANALYSI Biomedical Laboratories S.A., 54623 Thessaloniki, Greece
| | - Bassam R. Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 15551, United Arab Emirates
- ASPIRE Abu Dhabi Precision Medicine Ρesearch Institute, Al-Ain P.O. Box 15551, United Arab Emirates
| | - George P. Patrinos
- Laboratory of Pharmacogenomics and Individualized Therapy, Division of Pharmacology and Biosciences, Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Rion, Greece
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 15551, United Arab Emirates
- ASPIRE Abu Dhabi Precision Medicine Ρesearch Institute, Al-Ain P.O. Box 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 15551, United Arab Emirates
- Clinical Bioinformatics Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
10
|
Schmued L, Maloney B, Schmued C, Lahiri DK. Treatment with 1, 10 Phenanthroline-5-Amine Reduced Amyloid Burden in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2024; 97:239-247. [PMID: 38073385 PMCID: PMC10789349 DOI: 10.3233/jad-221285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent age-related dementia, and, despite numerous attempts to halt or reverse its devastating progression, no effective therapeutics have yet been confirmed clinically. However, one class of agents that has shown promise is certain metal chelators. OBJECTIVE For the novel assessment of the effect of oral administration of 1,10-phenanthroline-5-amine (PAA) on the severity of amyloid plaque load, we used a transgenic (Tg) mouse model with inserted human autosomally dominant (familial) AD genes: amyloid-β protein precursor (AβPP) and tau. METHODS AβPP/Tau transgenic mice that model AD were allotted into one of two groups. The control group received no treatment while the experimental group received PAA in their drinking water starting at 4 months of age. All animals were sacrificed at 1 year of age and their brains were stained with two different markers of amyloid plaques, Amylo-Glo+ and HQ-O. RESULTS The control animals exhibited numerous dense core plaques throughout the neo- and allo- cortical brain regions. The experimental group treated with PAA, however, showed 62% of the amyloid plaque burden seen in the control group. CONCLUSIONS Oral daily dosing with PAA will significantly reduce the amyloid plaque burden in transgenic mice that model AD. The underlying mechanism for this protection is not fully known; however, one proposed mechanism involves inhibiting the "metal-seeding" of Aβ.
Collapse
Affiliation(s)
| | - Bryan Maloney
- Department of Psychiatry, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Debomoy K. Lahiri
- Department of Psychiatry, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Departments of Medical & Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
11
|
Kawahara M, Kato-Negishi M, Tanaka KI. Dietary Trace Elements and the Pathogenesis of Neurodegenerative Diseases. Nutrients 2023; 15:2067. [PMID: 37432185 PMCID: PMC10180548 DOI: 10.3390/nu15092067] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 07/12/2023] Open
Abstract
Trace elements such as iron (Fe), zinc (Zn), copper (Cu), and manganese (Mn) are absorbed from food via the gastrointestinal tract, transported into the brain, and play central roles in normal brain functions. An excess of these trace elements often produces reactive oxygen species and damages the brain. Moreover, increasing evidence suggests that the dyshomeostasis of these metals is involved in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease, prion diseases, and Lewy body diseases. The disease-related amyloidogenic proteins can regulate metal homeostasis at the synapses, and thus loss of the protective functions of these amyloidogenic proteins causes neurodegeneration. Meanwhile, metal-induced conformational changes of the amyloidogenic proteins contribute to enhancing their neurotoxicity. Moreover, excess Zn and Cu play central roles in the pathogenesis of vascular-type senile dementia. Here, we present an overview of the intake, absorption, and transport of four essential elements (Fe, Zn, Cu, Mn) and one non-essential element (aluminum: Al) in food and their connections with the pathogenesis of neurodegenerative diseases based on metal-protein, and metal-metal cross-talk.
Collapse
Affiliation(s)
- Masahiro Kawahara
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| | - Midori Kato-Negishi
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| | - Ken-Ichiro Tanaka
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| |
Collapse
|
12
|
Nakagawa Y, Yamada S. The Relationships Among Metal Homeostasis, Mitochondria, and Locus Coeruleus in Psychiatric and Neurodegenerative Disorders: Potential Pathogenetic Mechanism and Therapeutic Implications. Cell Mol Neurobiol 2023; 43:963-989. [PMID: 35635600 PMCID: PMC11414457 DOI: 10.1007/s10571-022-01234-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 05/15/2022] [Indexed: 11/03/2022]
Abstract
While alterations in the locus coeruleus-noradrenergic system are present during early stages of neuropsychiatric disorders, it is unclear what causes these changes and how they contribute to other pathologies in these conditions. Data suggest that the onset of major depressive disorder and schizophrenia is associated with metal dyshomeostasis that causes glial cell mitochondrial dysfunction and hyperactivation in the locus coeruleus. The effect of the overactive locus coeruleus on the hippocampus, amygdala, thalamus, and prefrontal cortex can be responsible for some of the psychiatric symptoms. Although locus coeruleus overactivation may diminish over time, neuroinflammation-induced alterations are presumably ongoing due to continued metal dyshomeostasis and mitochondrial dysfunction. In early Alzheimer's and Parkinson's diseases, metal dyshomeostasis and mitochondrial dysfunction likely induce locus coeruleus hyperactivation, pathological tau or α-synuclein formation, and neurodegeneration, while reduction of glymphatic and cerebrospinal fluid flow might be responsible for β-amyloid aggregation in the olfactory regions before the onset of dementia. It is possible that the overactive noradrenergic system stimulates the apoptosis signaling pathway and pathogenic protein formation, leading to further pathological changes which can occur in the presence or absence of locus coeruleus hypoactivation. Data are presented in this review indicating that although locus coeruleus hyperactivation is involved in pathological changes at prodromal and early stages of these neuropsychiatric disorders, metal dyshomeostasis and mitochondrial dysfunction are critical factors in maintaining ongoing neuropathology throughout the course of these conditions. The proposed mechanistic model includes multiple pharmacological sites that may be targeted for the treatment of neuropsychiatric disorders commonly.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan.
| | - Shizuo Yamada
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| |
Collapse
|
13
|
Bazi Alahri M, Jibril Ibrahim A, Barani M, Arkaban H, Shadman SM, Salarpour S, Zarrintaj P, Jaberi J, Turki Jalil A. Management of Brain Cancer and Neurodegenerative Disorders with Polymer-Based Nanoparticles as a Biocompatible Platform. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020841. [PMID: 36677899 PMCID: PMC9864049 DOI: 10.3390/molecules28020841] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/27/2022] [Accepted: 01/10/2023] [Indexed: 01/19/2023]
Abstract
The blood-brain barrier (BBB) serves as a protective barrier for the central nervous system (CNS) against drugs that enter the bloodstream. The BBB is a key clinical barrier in the treatment of CNS illnesses because it restricts drug entry into the brain. To bypass this barrier and release relevant drugs into the brain matrix, nanotechnology-based delivery systems have been developed. Given the unstable nature of NPs, an appropriate amount of a biocompatible polymer coating on NPs is thought to have a key role in reducing cellular cytotoxicity while also boosting stability. Human serum albumin (HSA), poly (lactic-co-glycolic acid) (PLGA), Polylactide (PLA), poly (alkyl cyanoacrylate) (PACA), gelatin, and chitosan are only a few of the significant polymers mentioned. In this review article, we categorized polymer-coated nanoparticles from basic to complex drug delivery systems and discussed their application as novel drug carriers to the brain.
Collapse
Affiliation(s)
- Mehdi Bazi Alahri
- Department of Clinical Psychology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1971653313, Iran
| | - Alhawarin Jibril Ibrahim
- Department of Chemistry, Faculty of Science, Al-Hussein Bin Talal University, Ma’an 71111, Jordan
| | - Mahmood Barani
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman 7616913555, Iran
- Correspondence:
| | - Hassan Arkaban
- Department of Chemistry, University of Isfahan, Isfahan 8174673441, Iran
| | | | - Soodeh Salarpour
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616913555, Iran
| | - Payam Zarrintaj
- School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK 74078, USA
| | - Javad Jaberi
- Department of Chemistry, University of Isfahan, Isfahan 8174673441, Iran
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla 51001, Iraq
| |
Collapse
|
14
|
Rogers JT, Cahill CM. Iron Responsiveness to Lysosomal Disruption: A Novel Pathway to Alzheimer's Disease. J Alzheimers Dis 2023; 96:41-45. [PMID: 37781810 DOI: 10.3233/jad-230953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Familial Alzheimer's disease (fAD) mutations in the amyloid-β protein precursor (AβPP) enhance brain AβPP C-Terminal Fragment (CTF) levels to inhibit lysosomal v-ATPase. Consequent disrupted acidification of the endolysosomal pathway may trigger brain iron deficiencies and mitochondrial dysfunction. The iron responsive element (IRE) in the 5'Untranslated-region of AβPP mRNA should be factored into this cycle where reduced bioavailable Fe-II would decrease IRE-dependent AβPP translation and levels of APP-CTFβ in a cycle to adaptively restore iron homeostasis while increases of transferrin-receptors is evident. In healthy younger individuals, Fe-dependent translational modulation of AβPP is part of the neuroprotective function of sAβPPα with its role in iron transport.
Collapse
Affiliation(s)
- Jack T Rogers
- Neurochemistry Laboratory, Massachusetts General Hospital (East), and Harvard Medical School, Charlestown, MA, USA
| | - Catherine M Cahill
- Neurochemistry Laboratory, Massachusetts General Hospital (East), and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
15
|
Ferroptosis as a mechanism of non-ferrous metal toxicity. Arch Toxicol 2022; 96:2391-2417. [PMID: 35727353 DOI: 10.1007/s00204-022-03317-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/11/2022] [Indexed: 11/02/2022]
Abstract
Ferroptosis is a recently discovered form of regulated cell death, implicated in multiple pathologies. Given that the toxicity elicited by some metals is linked to alterations in iron metabolism and induction of oxidative stress and lipid peroxidation, ferroptosis might be involved in such toxicity. Although direct evidence is insufficient, certain pioneering studies have demonstrated a crosstalk between metal toxicity and ferroptosis. Specifically, the mechanisms underlying metal-induced ferroptosis include induction of ferritinophagy, increased DMT-1 and TfR cellular iron uptake, mitochondrial dysfunction and mitochondrial reactive oxygen species (mitoROS) generation, inhibition of Xc-system and glutathione peroxidase 4 (GPX4) activity, altogether resulting in oxidative stress and lipid peroxidation. In addition, there is direct evidence of the role of ferroptosis in the toxicity of arsenic, cadmium, zinc, manganese, copper, and aluminum exposure. In contrast, findings on the impact of cobalt and nickel on ferroptosis are scant and nearly lacking altogether for mercury and especially lead. Other gaps in the field include limited studies on the role of metal speciation in ferroptosis and the critical cellular targets. Although further detailed studies are required, it seems reasonable to propose even at this early stage that ferroptosis may play a significant role in metal toxicity, and its modulation may be considered as a potential therapeutic tool for the amelioration of metal toxicity.
Collapse
|
16
|
Michalke B. Review about Powerful Combinations of Advanced and Hyphenated Sample Introduction Techniques with Inductively Coupled Plasma-Mass Spectrometry (ICP-MS) for Elucidating Trace Element Species in Pathologic Conditions on a Molecular Level. Int J Mol Sci 2022; 23:ijms23116109. [PMID: 35682788 PMCID: PMC9181184 DOI: 10.3390/ijms23116109] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/23/2022] [Accepted: 05/27/2022] [Indexed: 02/05/2023] Open
Abstract
Element analysis in clinical or biological samples is important due to the essential role in clinical diagnostics, drug development, and drug-effect monitoring. Particularly, the specific forms of element binding, actual redox state, or their spatial distribution in tissue or in single cells are of interest in medical research. This review summarized exciting combinations of sophisticated sample delivery systems hyphenated to inductively coupled plasma-mass spectrometry (ICP-MS), enabling a broadening of information beyond the well-established outstanding detection capability. Deeper insights into pathological disease processes or intracellular distribution of active substances were provided, enabling a better understanding of biological processes and their dynamics. Examples were presented from spatial elemental mapping in tissue, cells, or spheroids, also considering elemental tagging. The use of natural or artificial tags for drug monitoring was shown. In the context of oxidative stress and ferroptosis iron, redox speciation gained importance. Quantification methods for Fe2+, Fe3+, and ferritin-bound iron were introduced. In Wilson’s disease, free and exchangeable copper play decisive roles; the respective paragraph provided information about hyphenated Cu speciation techniques, which provide their fast and reliable quantification. Finally, single cell ICP-MS provides highly valuable information on cell-to-cell variance, insights into uptake of metal-containing drugs, and their accumulation and release on the single-cell level.
Collapse
Affiliation(s)
- Bernhard Michalke
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München-German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| |
Collapse
|
17
|
Gonzalez-Alvarez MA, Hernandez-Bonilla D, Plascencia-Alvarez NI, Riojas-Rodriguez H, Rosselli D. Environmental and occupational exposure to metals (manganese, mercury, iron) and Parkinson's disease in low and middle-income countries: a narrative review. REVIEWS ON ENVIRONMENTAL HEALTH 2022; 37:1-11. [PMID: 33768768 DOI: 10.1515/reveh-2020-0140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/08/2021] [Indexed: 06/12/2023]
Abstract
OBJECTIVES We designed and conducted a narrative review consistent with the PRISMA guidelines (PROSPERO registration number: CRD42018099498) to evaluate the association between environmental metals (manganese, mercury, iron) and Parkinson's Disease (PD) in low and middle-income countries (LMIC). METHODS Data sources: A total of 19 databases were screened, and 2,048 references were gathered. Study selection: Randomized controlled trials, cluster trials, cohort studies, case-control studies, nested case-control studies, ecological studies, cross-sectional studies, case series, and case reports carried out in human adults of LMIC, in which the association between at least one of these three metals and the primary outcome were reported. Data extraction: We extracted qualitative and quantitative data. The primary outcome was PD cases, defined by clinical criteria. A qualitative analysis was conducted. RESULTS Fourteen observational studies fulfilled the selection criteria. Considerable variation was observed between these studies' methodologies for the measurement of metal exposure and outcome assessment. A fraction of studies suggested an association between the exposure and primary outcome; nevertheless, these findings should be weighted and appraised on the studies' design and its implementation limitations, flaws, and implications. CONCLUSIONS Further research is required to confirm a potential risk of metal exposure and its relationship to PD. To our awareness, this is the first attempt to evaluate the association between environmental and occupational exposure to metals and PD in LMIC settings using the social determinants of health as a framework.
Collapse
Affiliation(s)
| | - David Hernandez-Bonilla
- Environmental Health Department, National Institute of Public Health, Ciudad de Mexico, CDMX, Mexico
- Environmental Health Department, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | | | - Horacio Riojas-Rodriguez
- Environmental Health Department, National Institute of Public Health, Ciudad de Mexico, CDMX, Mexico
- Environmental Health Department, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Diego Rosselli
- Clinical Epidemiology and Biostatistics Department, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
18
|
Differences and Interactions in Placental Manganese and Iron Transfer across an In Vitro Model of Human Villous Trophoblasts. Int J Mol Sci 2022; 23:ijms23063296. [PMID: 35328723 PMCID: PMC8951728 DOI: 10.3390/ijms23063296] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/03/2022] [Accepted: 03/14/2022] [Indexed: 12/10/2022] Open
Abstract
Manganese (Mn) as well as iron (Fe) are essential trace elements (TE) important for the maintenance of physiological functions including fetal development. However, in the case of Mn, evidence suggests that excess levels of intrauterine Mn are associated with adverse pregnancy outcomes. Although Mn is known to cross the placenta, the fundamentals of Mn transfer kinetics and mechanisms are largely unknown. Moreover, exposure to combinations of TEs should be considered in mechanistic transfer studies, in particular for TEs expected to share similar transfer pathways. Here, we performed a mechanistic in vitro study on the placental transfer of Mn across a BeWo b30 trophoblast layer. Our data revealed distinct differences in the placental transfer of Mn and Fe. While placental permeability to Fe showed a clear inverse dose-dependency, Mn transfer was largely independent of the applied doses. Concurrent exposure of Mn and Fe revealed transfer interactions of Fe and Mn, indicating that they share common transfer mechanisms. In general, mRNA and protein expression of discussed transporters like DMT1, TfR, or FPN were only marginally altered in BeWo cells despite the different exposure scenarios highlighting that Mn transfer across the trophoblast layer likely involves a combination of active and passive transport processes.
Collapse
|
19
|
Han J, Fan Y, Wu P, Huang Z, Li X, Zhao L, Ji Y, Zhu M. Parkinson's Disease Dementia: Synergistic Effects of Alpha-Synuclein, Tau, Beta-Amyloid, and Iron. Front Aging Neurosci 2021; 13:743754. [PMID: 34707492 PMCID: PMC8542689 DOI: 10.3389/fnagi.2021.743754] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/21/2021] [Indexed: 12/31/2022] Open
Abstract
Parkinson’s disease dementia (PDD) is a common complication of Parkinson’s disease that seriously affects patients’ health and quality of life. At present, the process and pathological mechanisms of PDD remain controversial, which hinders the development of treatments. An increasing number of clinical studies have shown that alpha-synuclein (α-syn), tau, beta-amyloid (Aβ), and iron are closely associated with PDD severity. Thus, we inferred the vicious cycle that causes oxidative stress (OS), due to the synergistic effects of α-syn, tau, Aβ, and, iron, and which plays a pivotal role in the mechanism underlying PDD. First, iron-mediated reactive oxygen species (ROS) production can lead to neuronal protein accumulation (e.g., α-syn andAβ) and cytotoxicity. In addition, regulation of post-translational modification of α-syn by iron affects the aggregation or oligomer formation of α-syn. Iron promotes tau aggregation and neurofibrillary tangles (NFTs) formation. High levels of iron, α-syn, Aβ, tau, and NFTs can cause severe OS and neuroinflammation, which lead to cell death. Then, the increasing formation of α-syn, Aβ, and NFTs further increase iron levels, which promotes the spread of α-syn and Aβ in the central and peripheral nervous systems. Finally, iron-induced neurotoxicity promotes the activation of glycogen synthase kinase 3β (GSK3β) related pathways in the synaptic terminals, which in turn play an important role in the pathological synergistic effects of α-syn, tau and Aβ. Thus, as the central factor regulating this vicious cycle, GSK3β is a potential target for the prevention and treatment of PDD; this is worthy of future study.
Collapse
Affiliation(s)
- Jiajun Han
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yaohua Fan
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Peipei Wu
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zifeng Huang
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xinrong Li
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lijun Zhao
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yichun Ji
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Meiling Zhu
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
20
|
Hin N, Newman M, Pederson S, Lardelli M. Iron Responsive Element-Mediated Responses to Iron Dyshomeostasis in Alzheimer's Disease. J Alzheimers Dis 2021; 84:1597-1630. [PMID: 34719489 DOI: 10.3233/jad-210200] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Iron trafficking and accumulation is associated with Alzheimer's disease (AD) pathogenesis. However, the role of iron dyshomeostasis in early disease stages is uncertain. Currently, gene expression changes indicative of iron dyshomeostasis are not well characterized, making it difficult to explore these in existing datasets. OBJECTIVE To identify sets of genes predicted to contain iron responsive elements (IREs) and use these to explore possible iron dyshomeostasis-associated gene expression responses in AD. METHODS Comprehensive sets of genes containing predicted IRE or IRE-like motifs in their 3' or 5' untranslated regions (UTRs) were identified in human, mouse, and zebrafish reference transcriptomes. Further analyses focusing on these genes were applied to a range of cultured cell, human, mouse, and zebrafish gene expression datasets. RESULTS IRE gene sets are sufficiently sensitive to distinguish not only between iron overload and deficiency in cultured cells, but also between AD and other pathological brain conditions. Notably, changes in IRE transcript abundance are among the earliest observable changes in zebrafish familial AD (fAD)-like brains, preceding other AD-typical pathologies such as inflammatory changes. Unexpectedly, while some IREs in the 3' untranslated regions of transcripts show significantly increased stability under iron deficiency in line with current assumptions, many such transcripts instead display decreased stability, indicating that this is not a generalizable paradigm. CONCLUSION Our results reveal IRE gene expression changes as early markers of the pathogenic process in fAD and are consistent with iron dyshomeostasis as an important driver of this disease. Our work demonstrates how differences in the stability of IRE-containing transcripts can be used to explore and compare iron dyshomeostasis-associated gene expression responses across different species, tissues, and conditions.
Collapse
Affiliation(s)
- Nhi Hin
- South Australian Genomics Centre, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA, Australia.,Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, The University of Adelaide, North Terrace, Adelaide, SA, Australia
| | - Morgan Newman
- Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, The University of Adelaide, North Terrace, Adelaide, SA, Australia
| | - Stephen Pederson
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Michael Lardelli
- Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, The University of Adelaide, North Terrace, Adelaide, SA, Australia
| |
Collapse
|
21
|
Zhang J, Yang Y, Yang X, Qin J, Wei X, Peng Y, Li Z, Zhang L, Zhang Z, Zou Y. Influence of manganese exposure on cognitive function, plasma APP and Aβ levels in older men. J Trace Elem Med Biol 2021; 67:126788. [PMID: 34015662 DOI: 10.1016/j.jtemb.2021.126788] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/20/2021] [Accepted: 05/10/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Elevated manganese (Mn) exposure impairs cognition in adults and children, but the association between Mn and cognitive function in elderly people is unclear. Previous studies have linked Mn neurotoxicity in AD to Aβ-dependent mechanisms. However, the association between Mn and plasma APP and Aβ in the general elderly population remains unknown. This study aimed to investigate the association between Mn exposure and cognitive function, plasma APP and plasma Aβ in older adults. METHODS Cognitive abilities in 375 men aged 60 and older in Guangxi, China were assessed using the Mini-Mental State Examination (MMSE) and cognitive impairment were identified using education-stratified cut-off points of MMSE scores. Urinary Mn levels and plasma APP, and Aβ levels were measured using ICP-MS and ELISA, respectively. RESULTS A total of 109 (29.07 %) older men were identified as having cognitive impairment. The median urinary Mn level was 0.22 μg/g creatinine. Urinary Mn levels were negatively correlated with MMSE scores (β = -1.35, 95 % CI: -2.65 to -0.06; p = 0.041). In addition, higher concentrations of urinary manganese were associated with a greater risk of cognitive impairment (OR = 2.03, 95 % CI: 1.14-3.59; comparing the highest and lowest manganese; p = 0.025). Moreover, plasma APP levels were inversely associated with urinary Mn levels (r = -0.123, p = 0.020), and positively associated with MMSE scores (r = 0.158, p = 0.002). Surprisingly, no correlations were observed between plasma Aβ42, Aβ40, Aβ40/Aβ42, or Aβ42/Aβ40 and urinary Mn levels and MMSE scores. CONCLUSION These results suggested that Mn exposure is negatively associated with older men's cognition and plasma APP levels, but not plasma Aβ levels.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yiping Yang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jian Qin
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiao Wei
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yang Peng
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zhiying Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Li'e Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zhiyong Zhang
- Department of Environmental Health and Occupational Medicine, School of Public Health, Guilin Medical University, Guilin, 541100, Guangxi, China.
| | - Yunfeng Zou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
22
|
Yang Y, Zhang J, Yang X, Li Z, Wang J, Lu C, Nan A, Zou Y. Dysregulated APP expression and α-secretase processing of APP is involved in manganese-induced cognitive impairment. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 220:112365. [PMID: 34058678 DOI: 10.1016/j.ecoenv.2021.112365] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 06/12/2023]
Abstract
Excessive exposure to manganese (Mn) can cause cognitive impairment, a common feature of Alzheimer's disease (AD), but the mechanisms remain unclear. Amyloid precursor protein (APP) is key to AD pathogenesis, and whether APP and its secretase processing are involved in Mn-induced cognitive impairment remains unknown. In the present study, we established a model of Mn-induced neurotoxicity in vivo (male C57BL/6, 0-100 mg/kg Mn, 90 days, gastric gavage) and in vitro (Neuro-2a (N2a) cells, 0-800 μM Mn for 24 h; APP overexpression and APP shRNA N2a cells, 0 and 800 μM Mn for 24 h). We found impaired cognition of Mn-treated mice. Both in vivo and in vitro results consistently showed that Mn exposure inhibited the expression of APP, α-secretase, soluble APP alpha protein (sAPPα), and synapse proteins as well as the activity of α-secretase. However, Mn exposure showed no effect on the protein levels of β-secretase, Aβ40, and Aβ42 or the activity of β-secretase. Collectively, these findings demonstrate key roles of APP and its α-secretase processing in the regulation of Mn-induced cognitive impairment, which may act as a target for ameliorating Mn-induced neurotoxicity.
Collapse
Affiliation(s)
- Yiping Yang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Jie Zhang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Zhiying Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Jian Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Cailing Lu
- Department of Nutrition and Food Hygiene, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Aruo Nan
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China.
| | - Yunfeng Zou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China.
| |
Collapse
|
23
|
Michalke B, Berthele A, Venkataramani V. Simultaneous Quantification and Speciation of Trace Metals in Paired Serum and CSF Samples by Size Exclusion Chromatography-Inductively Coupled Plasma-Dynamic Reaction Cell-Mass Spectrometry (SEC-DRC-ICP-MS). Int J Mol Sci 2021; 22:8892. [PMID: 34445607 PMCID: PMC8396360 DOI: 10.3390/ijms22168892] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/12/2021] [Accepted: 08/15/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Transition metals play a crucial role in brain metabolism: since they exist in different oxidation states they are involved in ROS generation, but they are also co-factors of enzymes in cellular energy metabolism or oxidative defense. METHODS Paired serum and cerebrospinal fluid (CSF) samples were analyzed for iron, zinc, copper and manganese as well as for speciation using SEC-ICP-DRC-MS. Brain extracts from Mn-exposed rats were additionally analyzed with SEC-ICP-DRC-MS. RESULTS The concentration patterns of transition metal size fractions were correlated between serum and CSF: Total element concentrations were significantly lower in CSF. Fe-ferritin was decreased in CSF whereas a LMW Fe fraction was relatively increased. The 400-600 kDa Zn fraction and the Cu-ceruloplasmin fraction were decreased in CSF, by contrast the 40-80 kDa fraction, containing Cu- and Zn-albumin, relatively increased. For manganese, the α-2-macroglobulin fraction showed significantly lower concentration in CSF, whereas the citrate Mn fraction was enriched. Results from the rat brain extracts supported the findings from human paired serum and CSF samples. CONCLUSIONS Transition metals are strictly controlled at neural barriers (NB) of neurologic healthy patients. High molecular weight species are down-concentrated along NB, however, the Mn-citrate fraction seems to be less controlled, which may be problematic under environmental load.
Collapse
Affiliation(s)
- Bernhard Michalke
- Research Unit Analytical BioGeoChemistry, Helmholtz Center Munich—German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Achim Berthele
- Department of Neurology, School of Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany;
| | - Vivek Venkataramani
- Department of Medicine II, Hematology/Oncology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany;
- Institute of Pathology, University Medical Center Göttingen (UMG), 37075 Göttingen, Germany
| |
Collapse
|
24
|
Violi F, Solovyev N, Vinceti M, Mandrioli J, Lucio M, Michalke B. The study of levels from redox-active elements in cerebrospinal fluid of amyotrophic lateral sclerosis patients carrying disease-related gene mutations shows potential copper dyshomeostasis. Metallomics 2021; 12:668-681. [PMID: 32373852 DOI: 10.1039/d0mt00051e] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Amyotrophic lateral sclerosis is a progressive neurodegenerative disease characterized by a loss of function of motor neurons. The etiology of this disorder is still largely unknown. Gene-environment interaction arises as a possible key factor in the development of amyotrophic lateral sclerosis. We assessed the levels of trace metals, copper (Cu), iron (Fe), and manganese (Mn), of 9 amyotrophic lateral sclerosis cases and 40 controls by measuring their content in cerebrospinal fluid. The following trace element species were quantified using ion chromatography-inductively coupled plasma mass spectrometry: univalent copper (Cu-I), divalent Cu (Cu-II), divalent Fe (Fe-II), trivalent Fe (Fe-III), divalent Mn (Mn-II), trivalent Mn (Mn-III), and also unidentified Mn species (Mn-unknown) were present in some samples. When computing the relative risks for amyotrophic lateral sclerosis through an unconditional logistic regression model, we observed a weak and imprecise positive association for iron (Fe III, adjusted odds ratio 1.48, 95% CI 0.46-4.76) and manganese (total-Mn and Mn-II; adjusted odds ratio 1.11, 95% CI 0.74-1.67, and 1.13, 95% CI 0.79-1.61, respectively). Increased risk for copper was found both in the crude analysis (odds ratio 1.14, 95% CI 0.99-1.31) and in multivariable analysis after adjusting for sex, age, and year of storage (1.09, 95% CI 0.90-1.32). Our results suggest a possible positive association between Cu and genetic amyotrophic lateral sclerosis, while they give little indication of involvement of Fe and Mn in disease, though some correlations found also for these elements deserve further investigation.
Collapse
Affiliation(s)
- Federica Violi
- CREAGEN Research Center of Environmental, Genetic and Nutritional Epidemiology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | |
Collapse
|
25
|
Martins AC, Ruella Oliveira S, Barbosa F, Tinkov AA, V A, Santamaría A, Lee E, Bowman AB, Aschner M. Evaluating the risk of manganese-induced neurotoxicity of parenteral nutrition: review of the current literature. Expert Opin Drug Metab Toxicol 2021; 17:581-593. [PMID: 33620266 PMCID: PMC8122055 DOI: 10.1080/17425255.2021.1894123] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Several diseases and clinical conditions can affect enteral nutrition and adequate gastrointestinal uptake. In this respect, parenteral nutrition (PN) is necessary for the provision of deficient trace elements. However, some essential elements, such as manganese (Mn) may be toxic to children and adults when parenterally administered in excess, leading to toxic, especially neurotoxic effects. AREAS COVERED Here, we briefly provide an overview on Mn, addressing its sources of exposure, the role of Mn in the etiology of neurodegenerative diseases, and focusing on potential mechanisms associated with Mn-induced neurotoxicity. In addition, we discuss the potential consequences of overexposure to Mn inherent to PN. EXPERT OPINION In this critical review, we suggest that additional research is required to safely set Mn levels in PN, and that eliminating Mn as an additive should be considered by physicians and nutritionists on a case by case basis in the meantime to avoid the greater risk of neurotoxicity by its presence. There is a need to better define clinical biomarkers for Mn toxicity by PN, as well as identify new effective agents to treat Mn-neurotoxicity. Moreover, we highlight the importance of the development of new guidelines and practice safeguards to protect patients from excessive Mn exposure and neurotoxicity upon PN administration.
Collapse
Affiliation(s)
- Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Silvana Ruella Oliveira
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, Brazil
| | - Fernando Barbosa
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, Brazil
| | - Alexey A. Tinkov
- Yaroslavl State University, Yaroslavl, Russia
- IM Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anatoly V
- IM Sechenov First Moscow State Medical University, Moscow, Russia
- Federal Scientific Center of Biological Systems and Agrotechnologies of the Russian Academy of Sciences, Orenburg, Russia
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- IM Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
26
|
Tukker AM, Royal CD, Bowman AB, McAllister KA. The Impact of Environmental Factors on Monogenic Mendelian Diseases. Toxicol Sci 2021; 181:3-12. [PMID: 33677604 PMCID: PMC8599782 DOI: 10.1093/toxsci/kfab022] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Environmental factors and gene-environment interactions modify the variable expressivity, progression, severity, and onset of some classic (monogenic) Mendelian-inherited genetic diseases. Cystic fibrosis, Huntington disease, Parkinson's disease, and sickle cell disease are examples of well-known Mendelian disorders that are influenced by exogenous exposures. Environmental factors may act by direct or indirect mechanisms to modify disease severity, timing, and presentation, including through epigenomic influences, protein misfolding, miRNA alterations, transporter activity, and mitochondrial effects. Because pathological features of early-onset Mendelian diseases can mimic later onset complex diseases, we propose that studies of environmental exposure vulnerabilities using monogenic model systems of rare Mendelian diseases have high potential to provide insight into complex disease phenotypes arising from multi-genetic/multi-toxicant interactions. Mendelian disorders can be modeled by homologous mutations in animal model systems with strong recapitulation of human disease etiology and natural history, providing an important advantage for study of these diseases. Monogenic high penetrant mutations are ideal for toxicant challenge studies with a wide variety of environmental stressors, because background genetic variability may be less able to alter the relatively strong phenotype driving disease-causing mutations. These models promote mechanistic understandings of gene-environment interactions and biological pathways relevant to both Mendelian and related sporadic complex disease outcomes by creating a sensitized background for relevant environmental risk factors. Additionally, rare disease communities are motivated research participants, creating the potential of strong research allies among rare Mendelian disease advocacy groups and disease registries and providing a variety of translational opportunities that are under-utilized in genetic or environmental health science.
Collapse
Affiliation(s)
- Anke M Tukker
- School of Health Sciences, Purdue University, West Lafayette, Indiana 47907-2051
| | - Charmaine D Royal
- Departments of African and African American Studies, Biology, Global Health, and Family Medicine and Community Health and Center on Genomics, Race, Identity, Difference, Duke University, Durham, North Carolina 27708
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, Indiana 47907-2051
| | - Kimberly A McAllister
- Genes Environment and Health Branch, Division of Extramural Research and Training, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| |
Collapse
|
27
|
Cheng J, Zhu Y, Xing X, Xiao J, Chen H, Zhang H, Wang D, Zhang Y, Zhang G, Wu Z, Liu Y. Manganese-deposited iron oxide promotes tumor-responsive ferroptosis that synergizes the apoptosis of cisplatin. Theranostics 2021; 11:5418-5429. [PMID: 33859755 PMCID: PMC8039957 DOI: 10.7150/thno.53346] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/26/2021] [Indexed: 01/09/2023] Open
Abstract
Background: Ferroptosis is a form of iron-dependent programmed cell death that differs from apoptosis with regards to both mechanism and cell morphology. Therefore, ferroptotic-based cancer therapy has shown significant potential to overcome the weaknesses of conventional therapeutics mediated by apoptosis pathways. Effective ferroptosis can be induced by the intracellular Fenton reaction that is dependent on the adequate supply of iron ions and H2O2 in cells. However, these are often insufficient due to intrinsic cellular regulation. Methods: In this study, we designed a cisplatin prodrug-loaded manganese-deposited iron oxide nanoplatform (Pt-FMO) to trigger intracellular cascade reactions that lead to generation of reactive oxygen species (ROS) to enhance ferroptotic effect. The Pt-FMO causes the tumor microenvironment responsive to release manganese, iron ions and Pt-drugs. As manganese is an element that is able to catalyze the Fenton reaction more effectively than iron, coupled with the Pt-drugs that can promote generation of H2O2 in cells, the Pt-FMO is expected to significantly strengthen catalysis of the Fenton reaction, which favors the ferroptotic effect. Moreover, the Pt-drugs will eventually function as cisplatin. Thus, Pt-FMO is an ideal candidate for tumor ferroptotic combined with apoptotic treatment. Results:In vivo results demonstrated that, at a dosage of only 8.89% Pt content, Pt-FMO is able to achieve a similar treatment effect as cisplatin. Hence, Pt-FMO exhibited significantly lower systemic toxicity compared to cisplatin. Additionally, Pt-FMO exhibits effective T2 -weighted MRI enhancement for tumor imaging. Conclusion: The Pt-FMO nanoplatform is designed to introduce mutual beneficial cascade reactions for promoting ferroptosis and apoptosis in combination with tumor MRI. The Pt-FMO system, which causes ferroptosis combined with apoptosis, can efficiently induce tumor cell death.
Collapse
Affiliation(s)
- Junjie Cheng
- Department of Pharmacy, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine; Department of Chemistry, University of Science and Technology of China, Hefei 230001, China
| | - Yang Zhu
- Department of Pharmacy, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine; Department of Chemistry, University of Science and Technology of China, Hefei 230001, China
| | - Xin Xing
- Department of Stomatology, the First Affiliated Hospital of Wannan Medical College, Wuhu 241000, China
| | - Jianmin Xiao
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Hui Chen
- Department of Dental Implant Center, Stomatologic Hospital & College, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Hongwei Zhang
- Department of Pharmacy, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine; Department of Chemistry, University of Science and Technology of China, Hefei 230001, China
| | - Dan Wang
- Department of Dental Implant Center, Stomatologic Hospital & College, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Yuanyuan Zhang
- School of Life Science, Anhui Medical University, Hefei 230032, China
| | - Guilong Zhang
- School of Pharmacy, the Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai 264003, China
| | - Zhengyan Wu
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Yangzhong Liu
- Department of Pharmacy, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine; Department of Chemistry, University of Science and Technology of China, Hefei 230001, China
| |
Collapse
|
28
|
Kawahara M, Kato-Negishi M, Tanaka KI. Neurometals in the Pathogenesis of Prion Diseases. Int J Mol Sci 2021; 22:ijms22031267. [PMID: 33525334 PMCID: PMC7866166 DOI: 10.3390/ijms22031267] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 12/29/2022] Open
Abstract
Prion diseases are progressive and transmissive neurodegenerative diseases. The conformational conversion of normal cellular prion protein (PrPC) into abnormal pathogenic prion protein (PrPSc) is critical for its infection and pathogenesis. PrPC possesses the ability to bind to various neurometals, including copper, zinc, iron, and manganese. Moreover, increasing evidence suggests that PrPC plays essential roles in the maintenance of homeostasis of these neurometals in the synapse. In addition, trace metals are critical determinants of the conformational change and toxicity of PrPC. Here, we review our studies and other new findings that inform the current understanding of the links between trace elements and physiological functions of PrPC and the neurotoxicity of PrPSc.
Collapse
|
29
|
Bagheri S, Saboury AA. What role do metals play in Alzheimer's disease? JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2021. [DOI: 10.1007/s13738-021-02181-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
30
|
Ebrahim AM, Idris AM, Alnajjar AO, Michalke B. Cr and Mn total, accessible species, and protein-fraction contents in plants used for traditional anti-diabetes treatment. J Trace Elem Med Biol 2020; 62:126645. [PMID: 32937239 DOI: 10.1016/j.jtemb.2020.126645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/18/2020] [Accepted: 08/31/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Our survey has found eleven plants that have being consumed for traditional treatment of diabetes mellitus, particularly in Saudi Arabia and generally in many countries across the world. The literature reported about trace elements such as Cr and Mn positively affecting diabetes mellitus. The aim of this work is to determine the total, accessible element species, and protein-fraction contents of Cr and Mn in the edible parts of those plants. METHODS The total contents of Mn were determined by inductively coupled plasma-optical emission spectrometry (ICP-OES), while the total contents of Cr was determined by ICP-sector field (sf)-mass spectrometry (MS) due to lower concentration. The protein-fraction contents were determined in accessible element species by size-exclusion chromatography (SEC)-ICP-MS. RESULTS AND DISCUSSION The separation was successfully carried out to quantify Cr and Mn bound up to 11 protein fractions. The examined plants recorded wide ranges of total contents (Cr 44.7-1880.0 μg/kg and Mn 3.7-59.0 mg/kg) and accessible element species contents (Cr 0.93-29.40 μg/kg and Mn 0.82-35.85 mg/kg). Also wide ranges of percentages of accessible element species contents to total contents of Cr (0.65-4.21%) and Mn (5.43-68.42%) were obtained. The hazardous indices of both trace elements Cr and Mn for all examined plants consumed by both children and adults were all < 1, indicating no probability of health risk to occur. Moreover, Cr as carcinogen element reported no probability of cancer risk to occur from the consumption of all examined plants. Irrespective of plant species, Cr was quantified in all SEC fractions (mainly protein fractions), with the exception of 1.9-3.7 kDa, while Mn was quantified in all SEC fractions with the exception of 100-120 and 1.3-3.7 kDa. Nevertheless, the majority of accessible Cr species contents bound to the 10-14 and 0.05-0.40 kDa fractions, while that of Mn bound to 0.05-0.40 kDa fraction. To gather, the benefits of specific plant species in terms of accessible Cr and Mn species contents, in addition to accessible Zn species contents reported in our previous study, Haloxylon Salicornicum, Olea Europaea Momordica and Charantia are recommended to be consumed for traditionally controlling T2DM.
Collapse
Affiliation(s)
- Ammar M Ebrahim
- Consultants unit, Research and Consulting Institute, King Faisal University, P.O Box 400, Al-Ahsa 31982, Saudi Arabia; Sudan Atomic Energy Commission, Khartoum, Sudan
| | - Abubakr M Idris
- Department of Chemistry, College of Science, King Khalid University, Abha 61321, Saudi Arabia; Research Center for Advance Materials Science (RCAMS), King Khalid University, Abha 1413, P.O. Box 9004, Saudi Arabia.
| | - Ahmed O Alnajjar
- Department of Chemistry, College of Science, King Faisal University, P.O Box 400, Al-Ahsa 31982, Saudi Arabia
| | - Bernhard Michalke
- Research Unit Analytical BioGeoChemistry, Helmholtz Center, Munich, German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
31
|
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: 7.0] [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
|
32
|
Cahill CM, Aleyadeh R, Gao J, Wang C, Rogers JT. Alpha-Synuclein in Alcohol Use Disorder, Connections with Parkinson's Disease and Potential Therapeutic Role of 5' Untranslated Region-Directed Small Molecules. Biomolecules 2020; 10:E1465. [PMID: 33096655 PMCID: PMC7589448 DOI: 10.3390/biom10101465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/28/2020] [Accepted: 10/09/2020] [Indexed: 12/23/2022] Open
Abstract
Alpha-synuclein (α-Syn) is a 140-amino acid (aa) protein encoded by the Synuclein alpha SNCA gene. It is the synaptic protein associated with Parkinson's disease (PD) and is the most highly expressed protein in the Lewy bodies associated with PD and other alpha synucleopathies, including Lewy body dementia (LBD) and multiple system atrophy (MSA). Iron deposits are present in the core of Lewy bodies, and there are reports suggesting that divalent metal ions including Cu2+ and Fe2+ enhance the aggregation of α-Syn. Differential expression of α-Syn is associated with alcohol use disorder (AUD), and specific genetic variants contribute to the risk for alcoholism, including alcohol craving. Spliced variants of α-Syn, leading to the expression of several shorter forms which are more prone to aggregation, are associated with both PD and AUD, and common transcript variants may be able to predict at-risk populations for some movement disorders or subtypes of PD, including secondary Parkinsonism. Both PD and AUD are associated with liver and brain iron dyshomeostasis. Research over the past decade has shown that α-Syn has iron import functions with an ability to oxidize the Fe3+ form of iron to Fe2+ to facilitate its entry into cells. Our prior research has identified an iron-responsive element (IRE) in the 5' untranslated region (5'UTR) of α-Syn mRNA, and we have used the α-Syn 5'UTR to screen for small molecules that modulate its expression in the H4 neuronal cell line. These screens have led us to identify several interesting small molecules capable of both decreasing and increasing α-Syn expression and that may have the potential, together with the recently described mesenchymal stem cell therapies, to normalize α-Syn expression in different regions of the alcoholic and PD brain.
Collapse
Affiliation(s)
- Catherine M. Cahill
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA;
| | | | - Jin Gao
- Department of Clinical Psychology, Qilu Hospital of Shandong University, Qingdao 266011, China;
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA;
| | - Jack T. Rogers
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA;
| |
Collapse
|
33
|
Sun Y, He Y, Yang L, Liang D, Shi W, Zhu X, Jiang Y, Ou C. Manganese induced nervous injury by α-synuclein accumulation via ATP-sensitive K(+) channels and GABA receptors. Toxicol Lett 2020; 332:164-170. [PMID: 32659473 DOI: 10.1016/j.toxlet.2020.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/27/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023]
Abstract
Manganese (Mn) is an environmental pollutant having a toxic effect on Parkinson's disease, with significant damage seen in the neurons of basal ganglia. Hence, Mn pollution is a public health concern. A Sprague-Dawley rat model was used to determine the damage to basal nuclei, and the effect of Mn intake was detected using the Morris water maze test and transmission electron microscopy. The SH-SY5Y cell line was exposed to Mn, and downstream signaling was assessed to determine the mechanism of toxicity. Mn exposure injured neurons, repressing GABAAR receptors and inducing GABABR receptors. The synergistic effect of the GABABR receptor and Kir6.1-SUR1 or Kir6.2-SUR1 was found to be one of the potential factors for the secretion of α-synuclein. The accumulation of α-synuclein regulated downstream factors calmodulin (CAM) cAMP response element-binding protein (CREB), thereby impairing learning and memory. Other genes downstream of CREB, rather than the feedback regulation of CREB, and brain-derived neurotrophic factor might also be involved.
Collapse
Affiliation(s)
- Yi Sun
- Department of Toxicology, School of Public Health, Guilin Medical University, Guilin 541004, China
| | - Yonghua He
- Department of Toxicology, School of Public Health, Guilin Medical University, Guilin 541004, China
| | - Lin Yang
- Department of Toxicology, School of Public Health, Guilin Medical University, Guilin 541004, China
| | - Dan Liang
- Department of Toxicology, School of Public Health, Guilin Medical University, Guilin 541004, China
| | - Wenxiang Shi
- Department of Toxicology, School of Public Health, Guilin Medical University, Guilin 541004, China
| | - Xiaonian Zhu
- Department of Toxicology, School of Public Health, Guilin Medical University, Guilin 541004, China
| | - Yueming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Naning 530021, China
| | - Chaoyan Ou
- Department of Toxicology, School of Public Health, Guilin Medical University, Guilin 541004, China.
| |
Collapse
|
34
|
Liu W, Xin Y, Li Q, Shang Y, Ping Z, Min J, Cahill CM, Rogers JT, Wang F. Biomarkers of environmental manganese exposure and associations with childhood neurodevelopment: a systematic review and meta-analysis. Environ Health 2020; 19:104. [PMID: 33008482 PMCID: PMC7531154 DOI: 10.1186/s12940-020-00659-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/22/2020] [Indexed: 05/17/2023]
Abstract
BACKGROUND Although prior studies showed a correlation between environmental manganese (Mn) exposure and neurodevelopmental disorders in children, the results have been inconclusive. There has yet been no consistent biomarker of environmental Mn exposure. Here, we summarized studies that investigated associations between manganese in biomarkers and childhood neurodevelopment and suggest a reliable biomarker. METHODS We searched PubMed and Web of Science for potentially relevant articles published until December 31th 2019 in English. We also conducted a meta-analysis to quantify the effects of manganese exposure on Intelligence Quotient (IQ) and the correlations of manganese in different indicators. RESULTS Of 1754 citations identified, 55 studies with 13,388 subjects were included. Evidence from cohort studies found that higher manganese exposure had a negative effect on neurodevelopment, mostly influencing cognitive and motor skills in children under 6 years of age, as indicated by various metrics. Results from cross-sectional studies revealed that elevated Mn in hair (H-Mn) and drinking water (W-Mn), but not blood (B-Mn) or teeth (T-Mn), were associated with poorer cognitive and behavioral performance in children aged 6-18 years old. Of these cross-sectional studies, most papers reported that the mean of H-Mn was more than 0.55 μg/g. The meta-analysis concerning H-Mn suggested that a 10-fold increase in hair manganese was associated with a decrease of 2.51 points (95% confidence interval (CI), - 4.58, - 0.45) in Full Scale IQ, while the meta-analysis of B-Mn and W-Mn generated no such significant effects. The pooled correlation analysis revealed that H-Mn showed a more consistent correlation with W-Mn than B-Mn. Results regarding sex differences of manganese associations were inconsistent, although the preliminary meta-analysis found that higher W-Mn was associated with better Performance IQ only in boys, at a relatively low water manganese concentrations (most below 50 μg/L). CONCLUSIONS Higher manganese exposure is adversely associated with childhood neurodevelopment. Hair is the most reliable indicator of manganese exposure for children at 6-18 years of age. Analysis of the publications demonstrated sex differences in neurodevelopment upon manganese exposure, although a clear pattern has not yet been elucidated for this facet of our study.
Collapse
Affiliation(s)
- Weiwei Liu
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yongjuan Xin
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Qianwen Li
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yanna Shang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Zhiguang Ping
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Junxia Min
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Catherine M. Cahill
- Neurochemistry Laboratory, Department of Psychiatry-Neuroscience, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA USA
| | - Jack T. Rogers
- Neurochemistry Laboratory, Department of Psychiatry-Neuroscience, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA USA
| | - Fudi Wang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou, China
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
35
|
Wu B, Cai H, Tang S, Xu Y, Shi Q, Wei L, Meng L, Zhang N, Wang X, Xiao D, Zou Y, Yang X, Li X, Lu C. Methionine-Mediated Protein Phosphatase 2A Catalytic Subunit (PP2Ac) Methylation Ameliorates the Tauopathy Induced by Manganese in Cell and Animal Models. Neurotherapeutics 2020; 17:1878-1896. [PMID: 32959271 PMCID: PMC7851222 DOI: 10.1007/s13311-020-00930-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2020] [Indexed: 01/10/2023] Open
Abstract
The molecular mechanism of Alzheimer-like cognitive impairment induced by manganese (Mn) exposure has not yet been fully clarified, and there are currently no effective interventions to treat neurodegenerative lesions related to manganism. Protein phosphatase 2 A (PP2A) is a major tau phosphatase and was recently identified as a potential therapeutic target molecule for neurodegenerative diseases; its activity is directed by the methylation status of the catalytic C subunit. Methionine is an essential amino acid, and its downstream metabolite S-adenosylmethionine (SAM) participates in transmethylation pathways as a methyl donor. In this study, the neurotoxic mechanism of Mn and the protective effect of methionine were evaluated in Mn-exposed cell and rat models. We show that Mn-induced neurotoxicity is characterized by PP2Ac demethylation accompanied by abnormally decreased LCMT-1 and increased PME-1, which are associated with tau hyperphosphorylation and spatial learning and memory deficits, and that the poor availability of SAM in the hippocampus is likely to determine the loss of PP2Ac methylation. Importantly, maintenance of local SAM levels through continuous supplementation with exogenous methionine, or through specific inhibition of PP2Ac demethylation by ABL127 administration in vitro, can effectively prevent tau hyperphosphorylation to reduce cellular oxidative stress, apoptosis, damage to cell viability, and rat memory deficits in cell or animal Mn exposure models. In conclusion, our data suggest that SAM and PP2Ac methylation may be novel targets for the treatment of Mn poisoning and neurotoxic mechanism-related tauopathies.
Collapse
Affiliation(s)
- Bin Wu
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Haiqing Cai
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Shen Tang
- School of Basic Medical Sciences, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Yilu Xu
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Qianqian Shi
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Lancheng Wei
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Ling Meng
- School of Basic Medical Sciences, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Ning Zhang
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Xinhang Wang
- School of Basic Medical Sciences, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Deqiang Xiao
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Yunfeng Zou
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Xiaobo Yang
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Xiyi Li
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| | - Cailing Lu
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| |
Collapse
|
36
|
Kuang Y, Zheng X, Zhang L, Ai X, Venkataramani V, Kilic E, Hermann DM, Majid A, Bähr M, Doeppner TR. Adipose-derived mesenchymal stem cells reduce autophagy in stroke mice by extracellular vesicle transfer of miR-25. J Extracell Vesicles 2020; 10:e12024. [PMID: 33304476 PMCID: PMC7710129 DOI: 10.1002/jev2.12024] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/25/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022] Open
Abstract
Grafted mesenchymal stem cells (MSCs) yield neuroprotection in preclinical stroke models by secreting extracellular vesicles (EVs). The neuroprotective cargo of EVs, however, has not yet been identified. To investigate such cargo and its underlying mechanism, primary neurons were exposed to oxygen-glucose-deprivation (OGD) and cocultured with adipose-derived MSCs (ADMSCs) or ADMSC-secreted EVs. Under such conditions, both ADMSCs and ADMSC-secreted EVs significantly reduced neuronal death. Screening for signalling cascades being involved in the interaction between ADMSCs and neurons revealed a decreased autophagic flux as well as a declined p53-BNIP3 activity in neurons receiving either treatment paradigm. However, the aforementioned effects were reversed when ADMSCs were pretreated with the inhibitor of exosomal secretion GW4869 or when Hrs was knocked down. In light of miR-25-3p being the most highly expressed miRNA in ADMSC-EVs interacting with the p53 pathway, further in vitro work focused on this pathway. Indeed, a miR-25-3p oligonucleotide mimic reduced cell death, whereas the anti-oligonucleotide increased autophagic flux and cell death by modulating p53-BNIP3 signalling in primary neurons exposed to OGD. Likewise, native ADMSC-EVs but not EVs obtained from ADMSCs pretreated with the anti-miR-25-3p oligonucleotide (ADMSC-EVsanti-miR-25-3p) confirmed the aforementioned in vitro observations in C57BL/6 mice exposed to cerebral ischemia. The infarct size was reduced, and neurological recovery was increased in mice treated with native ADMSC-EVs when compared to ADMSC-EVsanti-miR-25-3p. ADMSCs induce neuroprotection by improved autophagic flux through secreted EVs containing miR-25-3p. Hence, our work uncovers a novel key factor in naturally secreted ADMSC-EVs for the regulation of autophagy and induction of neuroprotection in a preclinical stroke model.
Collapse
Affiliation(s)
- Yaoyun Kuang
- University Medical Center GoettingenDepartment of NeurologyGoettingenGermany
| | - Xuan Zheng
- University Medical Center GoettingenDepartment of NeurologyGoettingenGermany
| | - Lin Zhang
- University Medical Center GoettingenDepartment of NeurologyGoettingenGermany
| | - Xiaoyu Ai
- University Medical Center GoettingenDepartment of NeurologyGoettingenGermany
| | - Vivek Venkataramani
- University Medical Center GoettingenInstitute for PathologyGoettingenGermany
| | - Ertugrul Kilic
- Istanbul Medipol UniversityRegenerative and Restorative Medical Research CenterIstanbulTurkey
| | - Dirk M. Hermann
- Department of NeurologyUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Arshad Majid
- Sheffield Institute for Translational NeuroscienceUniversity of SheffieldSheffieldUK
| | - Mathias Bähr
- University Medical Center GoettingenDepartment of NeurologyGoettingenGermany
| | - Thorsten R. Doeppner
- University Medical Center GoettingenDepartment of NeurologyGoettingenGermany
- Istanbul Medipol UniversityRegenerative and Restorative Medical Research CenterIstanbulTurkey
| |
Collapse
|
37
|
Brosseron F, Kleemann K, Kolbe CC, Santarelli F, Castro-Gomez S, Tacik P, Latz E, Jessen F, Heneka MT. Interrelations of Alzheimer´s disease candidate biomarkers neurogranin, fatty acid-binding protein 3 and ferritin to neurodegeneration and neuroinflammation. J Neurochem 2020; 157:2210-2224. [PMID: 32894885 DOI: 10.1111/jnc.15175] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 01/01/2023]
Abstract
There is growing evidence that promising biomarkers of inflammation in Alzheimer´s disease (AD) and other neurodegenerative diseases correlate strongest to levels of tau or neurofilament, indicating an inflammatory response to neuronal damage or death. To test this hypothesis, we investigated three AD candidate markers (ferritin, fatty acid binding protein 3 (FABP-3), and neurogranin) in interrelation to established AD and inflammatory protein markers. We further aimed to determine if such interrelations would be evident in pathological subjects only or also under non-pathological circumstances. Cerebrospinal fluid levels of the three proteins were quantified in samples from the University Clinic of Bonn (UKB) Department of Neurodegenerative Diseases & Geriatric Psychiatry, Germany. Data were analyzed based on clinical or biomarker-defined stratification of subjects with adjustment for covariates age, sex, and APOE status. Levels of ferritin, FABP-3 and neurogranin were elevated in subjects with pathological levels of t-tau independent of beta-amyloid status. The three markers correlated with each other, tau isoforms, age, and those inflammatory markers previously described as related to neurodegeneration, predominantly sTREM2, macrophage migration inhibitory factor, soluble vascular endothelial growth factor receptor, soluble vascular cell adhesion molecule 1 (sVCAM-1), and C1q. These interrelations existed in subjects with pathological and sub-pathological tau levels, in particular for FABP-3 and neurogranin. Relations to ferritin were independent of absolute levels of tau, too, but showed differing trajectories between pathological and non-pathological subjects. A specific set of inflammatory markers is highly related to markers of neuronal damage such as tau, neurogranin, or FABP-3. These proteins could be used as readouts of the inflammatory response during the neurodegeneration phase of AD.
Collapse
Affiliation(s)
- Frederic Brosseron
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases & Geropsychiatry/Neurology, University of Bonn Medical Center, Bonn, Germany
| | | | | | - Francesco Santarelli
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases & Geropsychiatry/Neurology, University of Bonn Medical Center, Bonn, Germany
| | - Sergio Castro-Gomez
- Department of Neurodegenerative Diseases & Geropsychiatry/Neurology, University of Bonn Medical Center, Bonn, Germany
| | - Pawel Tacik
- Department of Neurodegenerative Diseases & Geropsychiatry/Neurology, University of Bonn Medical Center, Bonn, Germany
| | - Eicke Latz
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Institute of Innate Immune, University of Bonn Medical Center, Bonn, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases & Geropsychiatry/Neurology, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
38
|
Hernández RB, Carrascal M, Abian J, Michalke B, Farina M, Gonzalez YR, Iyirhiaro GO, Moteshareie H, Burnside D, Golshani A, Suñol C. Manganese-induced neurotoxicity in cerebellar granule neurons due to perturbation of cell network pathways with potential implications for neurodegenerative disorders. Metallomics 2020; 12:1656-1678. [PMID: 33206086 DOI: 10.1039/d0mt00085j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Manganese (Mn) is essential for living organisms, playing an important role in nervous system function. Nevertheless, chronic and/or acute exposure to this metal, especially during early life stages, can lead to neurotoxicity and dementia by unclear mechanisms. Thus, based on previous works of our group with yeast and zebrafish, we hypothesized that the mechanisms mediating manganese-induced neurotoxicity can be associated with the alteration of protein metabolism. These mechanisms may also depend on the chemical speciation of manganese. Therefore, the current study aimed at investigating the mechanisms mediating the toxic effects of manganese in primary cultures of cerebellar granule neurons (CGNs). By exposing cultured CGNs to different chemical species of manganese ([[2-[(dithiocarboxy)amino]ethyl]carbamodithioato]](2-)-kS,kS']manganese, named maneb (MB), and [[1,2-ethanediylbis[carbamodithioato]](2-)]manganese mixture with [[1,2-ethanediylbis[carbamodithioato]](2-)]zinc, named mancozeb (MZ), and manganese chloride (MnCl2)), and using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, we observed that both MB and MZ induced similar cytotoxicity (LC50∼ 7-9 μM), which was higher than that of MnCl2 (LC50∼ 27 μM). Subsequently, we applied systems biology approaches, including metallomics, proteomics, gene expression and bioinformatics, and revealed that independent of chemical speciation, for non-cytotoxic concentrations (0.3-3 μM), Mn-induced neurotoxicity in CGNs is associated with metal dyshomeostasis and impaired protein metabolism. In this way, we verified that MB induced more post-translational alterations than MnCl2, which can be a plausible explanation for cytotoxic differences between both chemical species. The metabolism of proteins is one of the most energy consuming cellular processes and its impairment appears to be a key event of some cellular stress processes reported separately in other studies such as cell cycle arrest, energy impairment, cell signaling, excitotoxicity, immune response, potential protein accumulation and apoptosis. Interestingly, we verified that Mn-induced neurotoxicity shares pathways associated with the development of Alzheimer's disease, Amyotrophic Lateral Sclerosis, Huntington's disease, and Parkinson's disease. This has been observed in baker's yeast and zebrafish suggesting that the mode of action of Mn may be evolutionarily conserved.
Collapse
Affiliation(s)
- Raúl Bonne Hernández
- Laboratory of Bioinorganic and Environmental Toxicology - LABITA, Department of Exact and Earth Sciences, Federal University of São Paulo, Rua Prof. Artur Riedel, 275, CEP 09972-270, Diadema, SP, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ivleva I, Pestereva N, Zubov A, Karpenko M. Intranasal exposure of manganese induces neuroinflammation and disrupts dopamine metabolism in the striatum and hippocampus. Neurosci Lett 2020; 738:135344. [PMID: 32889006 DOI: 10.1016/j.neulet.2020.135344] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/18/2020] [Accepted: 08/27/2020] [Indexed: 01/26/2023]
Abstract
Prolonged exposure to manganese (Mn) may lead to toxic effects on the central nervous system (CNS). The mechanisms underlying neuronal death from exposure to Mn are not well understood but undoubtedly involve inflammatory processes. The aim of this study was to explore the effects of long-lasting intranasal Mn exposure in rats focusing on inflammatory processes and catecholamine (dopamine, norepinephrine) levels in the striatum and hippocampus. It was found that intranasal administration by instillation of MnCl2 solution once a day for 90 days leads to impaired movement and gait. We also observed that Mn concentration increased in the hippocampus (by 30 %) and in the striatum (by 220 %), dopamine (24 %) and DOPAC (35 %) were reduced in the striatum, and dopamine (190 %) and DOPAC (220 %) levels increased with simultaneously norepinephrine reduction (30 %) in the hippocampus. Observation of cytokine mRNA revealed increased expression of both assayed cytokines (IL-1β and TNF-α) in the hippocampus. There was a 3-fold increase in the expression of IBA-1 mRNA, 2-fold increase in NFκB mRNA, and dramatic reduction in IkB mRNA in the striatum. Taken together, intranasal exposure to a high dose of MnCl2 induces neuroinflammation and neurotransmission disturbance, but the effects are specific for each studied brain region.
Collapse
Affiliation(s)
- Irina Ivleva
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia.
| | - Nina Pestereva
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia
| | - Alexander Zubov
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia
| | - Marina Karpenko
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia; Institute of Physics, Nanotechnology and Telecommunications, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| |
Collapse
|
40
|
Iron-responsive-like elements and neurodegenerative ferroptosis. ACTA ACUST UNITED AC 2020; 27:395-413. [PMID: 32817306 PMCID: PMC7433652 DOI: 10.1101/lm.052282.120] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/26/2022]
Abstract
A set of common-acting iron-responsive 5′untranslated region (5′UTR) motifs can fold into RNA stem loops that appear significant to the biology of cognitive declines of Parkinson's disease dementia (PDD), Lewy body dementia (LDD), and Alzheimer's disease (AD). Neurodegenerative diseases exhibit perturbations of iron homeostasis in defined brain subregions over characteristic time intervals of progression. While misfolding of Aβ from the amyloid-precursor-protein (APP), alpha-synuclein, prion protein (PrP) each cause neuropathic protein inclusions in the brain subregions, iron-responsive-like element (IRE-like) RNA stem–loops reside in their transcripts. APP and αsyn have a role in iron transport while gene duplications elevate the expression of their products to cause rare familial cases of AD and PDD. Of note, IRE-like sequences are responsive to excesses of brain iron in a potential feedback loop to accelerate neuronal ferroptosis and cognitive declines as well as amyloidosis. This pathogenic feedback is consistent with the translational control of the iron storage protein ferritin. We discuss how the IRE-like RNA motifs in the 5′UTRs of APP, alpha-synuclein and PrP mRNAs represent uniquely folded drug targets for therapies to prevent perturbed iron homeostasis that accelerates AD, PD, PD dementia (PDD) and Lewy body dementia, thus preventing cognitive deficits. Inhibition of alpha-synuclein translation is an option to block manganese toxicity associated with early childhood cognitive problems and manganism while Pb toxicity is epigenetically associated with attention deficit and later-stage AD. Pathologies of heavy metal toxicity centered on an embargo of iron export may be treated with activators of APP and ferritin and inhibitors of alpha-synuclein translation.
Collapse
|
41
|
Miah MR, Ijomone OM, Okoh COA, Ijomone OK, Akingbade GT, Ke T, Krum B, da Cunha Martins A, Akinyemi A, Aranoff N, Antunes Soares FA, Bowman AB, Aschner M. The effects of manganese overexposure on brain health. Neurochem Int 2020; 135:104688. [PMID: 31972215 PMCID: PMC7926190 DOI: 10.1016/j.neuint.2020.104688] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 01/12/2020] [Accepted: 01/15/2020] [Indexed: 12/11/2022]
Abstract
Manganese (Mn) is the twelfth most abundant element on the earth and an essential metal to human health. Mn is present at low concentrations in a variety of dietary sources, which provides adequate Mn content to sustain support various physiological processes in the human body. However, with the rise of Mn utility in a variety of industries, there is an increased risk of overexposure to this transition metal, which can have neurotoxic consequences. This risk includes occupational exposure of Mn to workers as well as overall increased Mn pollution affecting the general public. Here, we review exposure due to air pollution and inhalation in industrial settings; we also delve into the toxic effects of manganese on the brain such as oxidative stress, inflammatory response and transporter dysregulation. Additionally, we summarize current understandings underlying the mechanisms of Mn toxicity.
Collapse
Affiliation(s)
- Mahfuzur R Miah
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Omamuyovwi M Ijomone
- The Neuro-Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology Akure, Ondo, Nigeria
| | - Comfort O A Okoh
- The Neuro-Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology Akure, Ondo, Nigeria
| | - Olayemi K Ijomone
- The Neuro-Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology Akure, Ondo, Nigeria; Department of Anatomy, University of Medical Sciences, Ondo, Nigeria
| | - Grace T Akingbade
- The Neuro-Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology Akure, Ondo, Nigeria
| | - Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bárbara Krum
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | | | - Ayodele Akinyemi
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nicole Aranoff
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Stern College for Women, Yeshiva University, New York, NY, USA
| | - Felix Alexandre Antunes Soares
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
42
|
Kawahara M, Kato-Negishi M, Tanaka KI. Amyloids: Regulators of Metal Homeostasis in the Synapse. Molecules 2020; 25:molecules25061441. [PMID: 32210005 PMCID: PMC7145306 DOI: 10.3390/molecules25061441] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/17/2020] [Accepted: 03/20/2020] [Indexed: 12/11/2022] Open
Abstract
Conformational changes in amyloidogenic proteins, such as β-amyloid protein, prion proteins, and α-synuclein, play a critical role in the pathogenesis of numerous neurodegenerative diseases, including Alzheimer’s disease, prion disease, and Lewy body disease. The disease-associated proteins possess several common characteristics, including the ability to form amyloid oligomers with β-pleated sheet structure, as well as cytotoxicity, although they differ in amino acid sequence. Interestingly, these amyloidogenic proteins all possess the ability to bind trace metals, can regulate metal homeostasis, and are co-localized at the synapse, where metals are abundantly present. In this review, we discuss the physiological roles of these amyloidogenic proteins in metal homeostasis, and we propose hypothetical models of their pathogenetic role in the neurodegenerative process as the loss of normal metal regulatory functions of amyloidogenic proteins. Notably, these amyloidogenic proteins have the capacity to form Ca2+-permeable pores in membranes, suggestive of a toxic gain of function. Therefore, we focus on their potential role in the disruption of Ca2+ homeostasis in amyloid-associated neurodegenerative diseases.
Collapse
|
43
|
Interactions between iron and manganese in neurotoxicity. Arch Toxicol 2020; 94:725-734. [PMID: 32180038 DOI: 10.1007/s00204-020-02652-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/27/2020] [Indexed: 12/31/2022]
Abstract
The essential and naturally occurring transition metal manganese (Mn) is present in the soil, water, air, and various foods. Manganese can accumulate in the brain if the Mn intake or exposure is excessive and this can result in neurotoxic effects. Manganese is important for the proper activation of different metabolic and antioxidant enzymes. There are numerous Mn importers and exporters. However, the exact transport mechanism for Mn is not fully understood. On the other hand, iron (Fe) is another well-known essential metal, which has redox activity in addition to chemical characteristics resembling those of Mn. Existing data show that interactions occur between Fe and Mn due to certain similarities regarding their mechanisms of the absorption and the transport. It has been disclosed that Mn-specific transporters, together with Fe transporters, regulate the Mn distribution in the brain and other peripheral tissues. In PC12 cells, a significant increase of transferrin receptor (TfR) mRNA expression was linked to Mn exposure and accompanied by elevated Fe uptake. In both humans and animals, there is a strong relationship between Fe and Mn metabolism. In the present review, special attention is paid to the interaction between Mn and Fe. In particular, Fe and Mn distribution, as well as the potential molecular mechanisms of Mn-induced neurotoxicity in cases of Fe deficiency, are discussed.
Collapse
|
44
|
Bakulski KM, Seo YA, Hickman RC, Brandt D, Vadari HS, Hu H, KyunPark S. Heavy Metals Exposure and Alzheimer's Disease and Related Dementias. J Alzheimers Dis 2020; 76:1215-1242. [PMID: 32651318 PMCID: PMC7454042 DOI: 10.3233/jad-200282] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease and related dementias lack effective treatment or cures and are major public health challenges. Risk for Alzheimer's disease and related dementias is partially attributable to environmental factors. The heavy metals lead, cadmium, and manganese are widespread and persistent in our environments. Once persons are exposed to these metals, they are adept at entering cells and reaching the brain. Lead and cadmium are associated with numerous health outcomes even at low levels of exposure. Although manganese is an essential metal, deficiency or environmental exposure or high levels of the metal can be toxic. In cell and animal model systems, lead, cadmium, and manganese are well documented neurotoxicants that contribute to canonical Alzheimer's disease pathologies. Adult human epidemiologic studies have consistently shown lead, cadmium, and manganese are associated with impaired cognitive function and cognitive decline. No longitudinal human epidemiology study has assessed lead or manganese exposure on Alzheimer's disease specifically though two studies have reported a link between cadmium and Alzheimer's disease mortality. More longitudinal epidemiologic studies with high-quality time course exposure data and incident cases of Alzheimer's disease and related dementias are warranted to confirm and estimate the proportion of risk attributable to these exposures. Given the widespread and global exposure to lead, cadmium, and manganese, even small increases in the risks of Alzheimer's disease and related dementias would have a major population impact on the burden on disease. This article reviews the experimental and epidemiologic literature of the associations between lead, cadmium, and manganese on Alzheimer's disease and related dementias and makes recommendations of critical areas of future investment.
Collapse
Affiliation(s)
- Kelly M. Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Young Ah Seo
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Ruby C. Hickman
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Daniel Brandt
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Harita S. Vadari
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Howard Hu
- School of Public Health, University of Washington, Seattle, WA, USA
| | - Sung KyunPark
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
45
|
Nakagawa Y, Yamada S. Metal homeostasis disturbances in neurodegenerative disorders, with special emphasis on Creutzfeldt-Jakob disease - Potential pathogenetic mechanism and therapeutic implications. Pharmacol Ther 2019; 207:107455. [PMID: 31863817 DOI: 10.1016/j.pharmthera.2019.107455] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022]
Abstract
Creutzfeldt-Jakob disease (CJD) is characterized by a rapidly progressive dementia often accompanied by myoclonus and other signs of brain dysfunction, relying on the conversion of the normal cellular form of the prion protein (PrPC) to a misfolded form (PrPSc). The neuropathological changes include spongiform degeneration, neuronal loss, astrogliosis, and deposition of PrPSc. It is still unclear how these pathological changes correlate with the development of CJD symptoms because few patients survive beyond 2 years after diagnosis. Inasmuch as the symptoms of CJD overlap some of those observed in Alzheimer's, Parkinson's, and Huntington's diseases, there may be some underlying pathologic mechanisms associated with CJD that may contribute to the symptoms of non-prion neurodegenerative diseases as well. Data suggest that imbalance of metals, including copper, zinc, iron, and manganese, induces abnormalities in processing and degradation of prion proteins that are accompanied by self-propagation of PrPSc. These events appear to be responsible for glutamatergic synaptic dysfunctions, neuronal death, and PrPSc aggregation. Given that the prodromal symptoms of CJD such as sleep disturbances and mood disorders are associated with brain stem and limbic system dysfunction, the pathological changes may initially occur in these brain regions, then spread throughout the entire brain. Alterations in cerebrospinal fluid homeostasis, which may be linked to imbalance of these metals, seem to be more important than neuroinflammation in causing the cell death. It is proposed that metal dyshomeostasis could be responsible for the initiation and progression of the pathological changes associated with symptoms of CJD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Shizuo Yamada
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
46
|
Yang Y, Wei F, Wang J, Chen R, Zhang J, Li D, Gan D, Yang X, Zou Y. Manganese modifies Neurotrophin-3 (NT3) and its tropomyosin receptor kinase C (TrkC) in the cortex: Implications for manganese-induced neurotoxicity. Food Chem Toxicol 2019; 135:110925. [PMID: 31676349 DOI: 10.1016/j.fct.2019.110925] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 10/07/2019] [Accepted: 10/24/2019] [Indexed: 12/26/2022]
Abstract
Manganese (Mn), an essential micronutrient, has the potential to induce apoptosis. The NT3/TrkC ligand/receptor pair known as part of the classic neurotrophic theory plays a critical role in neuronal survival. However, whether the NT3/TrkC-mediated signaling pathways are involved in Mn-induced apoptosis of cortical neurons remains unknown. The present study was designed to investigate the interactions between NT3/TrkC-mediated signaling pathways and Mn-induced apoptosis in cortical neurons. This study showed that subacute Mn exposure significantly increased the levels of pro-apoptotic Bax while decreasing the levels of anti-apoptotic Bcl 2 in the cortex compared with the corresponding control. Markedly reduced NT3 and TrkC levels along with decreased Ras/MAPK and PI3/Akt signaling in the cortex were observed following subacute Mn exposure. We further found increased levels of Bax, cleaved caspase-3, and the total apoptosis rate, and decreased levels of Bcl 2, NT3, TrkC, and Ras/MAPK and PI3/Akt signaling in Mn-treated primary cortical neurons. Pretreatment with hNT3 or Z-VAD-FAM ameliorated Mn-induced apoptosis by increasing the levels of NT3 and TrkC and its Ras/MAPK and PI3/Akt signaling pathways. Taken together, our findings clearly indicate that NT3/TrkC and mediated Ras/MAPK and PI3/Akt signaling pathways play a crucial role in Mn-induced neurotoxicity.
Collapse
Affiliation(s)
- Yiping Yang
- 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
| | - Fu Wei
- Center for Reproductive Medicine and Genetics, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Jian Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Rui Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Jie Zhang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Danni Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Dong Gan
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, China.
| | - Yunfeng Zou
- 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
|
47
|
Liu C, Yan DY, Wang C, Ma Z, Deng Y, Liu W, Xu B. Manganese activates autophagy to alleviate endoplasmic reticulum stress-induced apoptosis via PERK pathway. J Cell Mol Med 2019; 24:328-341. [PMID: 31639278 PMCID: PMC6933331 DOI: 10.1111/jcmm.14732] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/22/2019] [Accepted: 09/27/2019] [Indexed: 12/17/2022] Open
Abstract
Overexposure to manganese (Mn) is neurotoxic. Our previous research has demonstrated that the interaction of endoplasmic reticulum (ER) stress and autophagy participates in the early stage of Mn‐mediated neurotoxicity in mouse. However, the mechanisms of ER stress signalling pathways in the initiation of autophagy remain confused. In the current study, we first validated that ER stress–mediated cell apoptosis is accompanied by autophagy in SH‐SY5Y cells. Then, we found that inhibiting ER stress with 4‐phenylbutyrate (4‐PBA) decreased ER stress–related protein expression and reduced cell apoptosis, whereas blocking autophagy with 3‐methyladenine (3‐MA) increased cell apoptosis. These data indicate that protective autophagy was activated to alleviate ER stress–mediated apoptosis. Knockdown of the protein kinase RNA‐like ER kinase (PERK) gene inhibited Mn‐induced autophagy and weakened the interaction between ATF4 and the LC3 promoter. Our results reveal a novel molecular mechanism in which ER stress may regulate autophagy via the PERK/eIF2α/ATF4 signalling pathway. Additionally, Mn may activate protective autophagy to alleviate ER stress–mediated apoptosis via the PERK/eIF2α/ATF4 signalling pathway in SH‐SY5Y cells.
Collapse
Affiliation(s)
- Chang Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Dong-Ying Yan
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Can Wang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Zhuo Ma
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| |
Collapse
|
48
|
Martins AC, Morcillo P, Ijomone OM, Venkataramani V, Harrison FE, Lee E, Bowman AB, Aschner M. New Insights on the Role of Manganese in Alzheimer's Disease and Parkinson's Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E3546. [PMID: 31546716 PMCID: PMC6801377 DOI: 10.3390/ijerph16193546] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
Manganese (Mn) is an essential trace element that is naturally found in the environment and is necessary as a cofactor for many enzymes and is important in several physiological processes that support development, growth, and neuronal function. However, overexposure to Mn may induce neurotoxicity and may contribute to the development of Alzheimer's disease (AD) and Parkinson's disease (PD). The present review aims to provide new insights into the involvement of Mn in the etiology of AD and PD. Here, we discuss the critical role of Mn in the etiology of these disorders and provide a summary of the proposed mechanisms underlying Mn-induced neurodegeneration. In addition, we review some new therapy options for AD and PD related to Mn overload.
Collapse
Affiliation(s)
- Airton Cunha Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA (P.M.)
| | - Patricia Morcillo
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA (P.M.)
| | - Omamuyovwi Meashack Ijomone
- Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology Akure, Akure 340252, Nigeria;
| | - Vivek Venkataramani
- Department of Hematology and Medical Oncology and Institute of Pathology, University Medical Center Göttingen (UMG), 37075 Göttingen, Germany;
| | - Fiona Edith Harrison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32301, USA;
| | - Aaron Blaine Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, USA;
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA (P.M.)
| |
Collapse
|
49
|
Peng DJ, Zhang YW, Li ZC, Li SJ, Cai M, Qin WX, Ou SY, Huang XW, Yuan ZX, Jiang YM. Preventive impacts of PAS-Na on the slow growth and activated inflammatory responses in Mn-exposed rats. J Trace Elem Med Biol 2019; 54:134-141. [PMID: 31109603 DOI: 10.1016/j.jtemb.2019.04.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/26/2019] [Accepted: 04/23/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Sodium para-aminosalicylic acid (PAS-Na), an anti-tuberculosis drug, has been demonstrated its function in facilitating the Mn elimination in manganism patients and Mn-exposed models in vivo and improving the symptoms of Mn poisoning. But whether it can improve the growth retardation and inflammatory responses induced by Mn have not been reported. OBJECTIVES This study was designed to investigate the preventive effects of PAS-Na on the development of retardation and inflammatory responses in Mn-exposed rats. METHODS Male Sprague Dawley (SD) rats (8 weeks old, weighing 180 ± 20 g) were randomly divided into normal control group and Mn-exposed group in the 4 weeks experiment observation and normal control group, Mn-exposed group, PAS-Na preventive group and PAS-Na control group in the 8 weeks experiment observation. The Mn-exposed group received an intraperitoneal injection (i.p.) of 15 mg/kg MnCl2 and the normal control group i.p. physiological Saline in the same volume once a day for 4 or 8 weeks, 5 days per week. The PAS-Na preventive group i.p. 15 mg/kg MnCl2 along with back subcutaneous (s.c.) injection of 240 mg/kg PAS-Na once a day for 8 weeks, 5 days per week. PAS-Na control group received s.c. injection of 240 mg/kg PAS-Na along with i.p. injection of saline once daily. The body weight was determined once a week until the end of the experiment. The manganese contents in the blood were detected by graphite furnace atomic absorption spectrometry. The inflammatory factor levels (TNF-α, IL-1β, IL-6, and PGE2) in the blood were detected by using enzyme-linked immunosorbent assay (Elisa) and each organ taking from rats were weighed and recorded. RESULTS Mn exposure significantly suppressed the growth in rats and increased heart, liver, spleen and kidney coefficients as compared with the control group. The whole blood Mn level and serum levels of IL-1β, IL-6, PGE2, and TNF-α in sub-chronic Mn-exposure group were markedly higher than those in the control group. However, preventive treatment with PAS-Na obviously reduced the whole blood Mn level, the spleen and liver coefficients of the Mn-exposed rats. And serum levels of IL-1β and TNF-α were significantly reduced by 33.9% and 14.7% respectively in PAS-Na prevention group. CONCLUSIONS PAS-Na could improve the growth retardation and alleviate inflammatory responses in Mn-exposed rats.
Collapse
Affiliation(s)
- Dong-Jie Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Yu-Wen Zhang
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Zhao-Cong Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Shao-Jun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Mei Cai
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China.
| | - Wen-Xia Qin
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Department of Child Health, Guilin Women and Children Hospital, Guilin, China.
| | - Shi-Yan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Xiao-Wei Huang
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Zong-Xiang Yuan
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, China.
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, 530021, Nanning, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| |
Collapse
|
50
|
Clinical utility of serum hepcidin and iron profile measurements in Alzheimer's disease. J Neurol Sci 2019; 403:85-91. [PMID: 31233974 DOI: 10.1016/j.jns.2019.06.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/06/2019] [Accepted: 06/09/2019] [Indexed: 01/13/2023]
Abstract
OBJECTIVES There are no generally accepted serum biomarkers for Alzheimer's disease (AD). We investigated the clinical usefulness of measuring the serum hepcidin levels and iron profile in patients with AD. MATERIALS & METHODS The iron profile and hepcidin levels were measured in patients with AD (N = 70), minimal cognitive impairment (MCI, N = 39), and vascular dementia (VD, N = 25) and normal controls (N = 124). General cognitive tests were performed, and the relationships between cognition and hepcidin levels or the iron profile were assessed. RESULTS Patients with AD had higher hepcidin values than those with MCI and VD and normal controls (median value: 39.00 vs. 30.81, 32.52, and 5.51 ng/ml, respectively, P < 0.001), and these differences were found in both men and women. The total iron-binding capacity was significantly lower in the AD group than in any other groups (308.0 vs. 332.0, 329.0, and 330.5 μg/dl, respectively, P = 0.018), and serum iron levels were lower in the AD group than controls (79.1 vs. 107.2 μg/dl, P = 0.007). Hepcidin levels were statistically significantly correlated with the clinical dementia rating (CDR, P = 0.040) with a Pearson's correlation coefficient of 0.253, and the patients with AD with a CDR value >1 had significantly higher hepcidin values than those with a CDR value of 1 (65.26 vs. 23.49 ng/ml, P = 0.020). CONCLUSION The measurement of serum hepcidin levels and the iron profile in patients with early manifestations of cognitive functional loss might aid in the diagnosis of AD and the assessment of disease severity when combined with other diagnostic parameters.
Collapse
|