1
|
Fang S, Clayton PT, Garg D, Yoganathan S, Zaki MS, Helgadottir EA, Palmadottir VK, Landry M, Gospe SM, Mankad K, Bonifati V, Sharma S, Tuschl K. Consensus of Expert Opinion for the Diagnosis and Management of Hypermanganesaemia With Dystonia 1 and 2. J Inherit Metab Dis 2025; 48:e70031. [PMID: 40320765 PMCID: PMC12050909 DOI: 10.1002/jimd.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/31/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025]
Abstract
Hypermanganesaemia with Dystonia 1 and 2 (HMNDYT1 and 2) are inherited, autosomal recessive disorders caused by pathogenic variants in the genes encoding the manganese transporters SLC30A10 and SLC39A14, respectively. Impaired hepatic and enterocytic manganese uptake (SLC39A14) and excretion (SLC30A10) lead to deposition of manganese in the basal ganglia resulting in childhood-onset dystonia-parkinsonism. HMNDYT1 is characterized by additional features due to manganese accumulation in the liver causing cirrhosis, polycythaemia, and depleted iron stores. High blood manganese levels and pathognomonic MRI brain appearances of manganese deposition resulting in T1 hyperintensity of the basal ganglia are diagnostic clues. Treatment is limited to chelation therapy and iron supplementation that can prevent disease progression. Due to their rarity, the awareness of the inherited manganese transporter defects is limited. Here, we provide consensus expert recommendations for the diagnosis and treatment of patients with HMNDYT1 and 2 in order to facilitate early diagnosis and optimize clinical outcome. These recommendations were developed through an evidence and consensus-based process led by a group of 13 international experts across the disciplines of metabolic medicine, neurology, hematology, genetics, and radiology, and address the clinical presentation, diagnostic investigations, principles of treatment, and monitoring of patients with HMNDYT1 and 2.
Collapse
Affiliation(s)
- Sherry Fang
- Department of Metabolic MedicineGreat Ormond Street Hospital for ChildrenLondonUK
| | - Peter T. Clayton
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
| | - Divyani Garg
- Department of NeurologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Sangeetha Yoganathan
- Paediatric Neurology Unit, Department of Neurological SciencesChristian Medical CollegeVelloreIndia
| | - Maha S. Zaki
- Department of Clinical GeneticsHuman Genetics and Genome Research Institute, National Research CentreCairoEgypt
| | | | | | - Maude Landry
- The Moncton Hospital, Horizon Health NetworkMonctonCanada
| | - Sidney M. Gospe
- Department of Neurology and PediatricsUniversity of WashingtonSeattleWashingtonUSA
- Department of PediatricsDuke UniversityDurhamNorth CarolinaUSA
| | - Kshitij Mankad
- Department of RadiologyGreat Ormond Street Hospital for ChildrenLondonUK
| | - Vincenzo Bonifati
- Erasmus MC, University Medical Center RotterdamRotterdamthe Netherlands
| | - Suvasini Sharma
- Department of PediatricsLady Hardinge Medical College and Associated Kalawati Saran Children's HospitalDelhiIndia
| | - Karin Tuschl
- Department of Metabolic MedicineGreat Ormond Street Hospital for ChildrenLondonUK
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
| |
Collapse
|
2
|
Warden AS, Sharma N, Hutchens S, Liu C, Haggerty NR, Gurol KC, Jursa T, Smith DR, Dayne Mayfield R, Mukhopadhyay S. Elevated brain manganese induces motor disease by upregulating the kynurenine pathway of tryptophan metabolism. Proc Natl Acad Sci U S A 2025; 122:e2423628122. [PMID: 40244671 PMCID: PMC12036984 DOI: 10.1073/pnas.2423628122] [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: 11/13/2024] [Accepted: 02/27/2025] [Indexed: 04/18/2025] Open
Abstract
Elevated brain levels of the essential metals manganese (Mn), copper, or iron induce motor disease. However, mechanisms of metal-induced motor disease are unclear and treatments are lacking. Elucidating the mechanisms of Mn-induced motor disease is particularly important because occupational and environmental Mn overexposure is a global public health problem. To address this, here we combined unbiased transcriptomics and metabolomics with functional studies in a mouse model of human environmental Mn exposure. Transcriptomics unexpectedly revealed that Mn exposure up-regulated expression of metabolic pathways in the brain and liver. Notably, genes in the kynurenine pathway of tryptophan metabolism, which produces neuroactive metabolites that impact neurological function, were up-regulated by Mn. Subsequent unbiased metabolomics revealed that Mn treatment altered kynurenine pathway metabolites in the brain and liver. Functional experiments then demonstrated that pharmacological inhibition of the first and rate-limiting step of the kynurenine pathway fully rescued Mn-induced motor deficits. Finally, elevated Mn directly activates hypoxia-inducible factor (HIF) transcription factors, and additional mechanistic assays identified a role for HIF1, but not HIF2, in regulating expression of hepatic kynurenine pathway genes under physiological or Mn exposure conditions, suggesting that Mn-induced HIF1 activation may contribute to the dysregulation of the kynurenine pathway in Mn toxicity. These findings (1) identify the upregulation of the kynurenine pathway by elevated Mn as a fundamental mechanism of Mn-induced motor deficits; (2) provide a pharmacological approach to treat Mn-induced motor disease; and (3) should broadly advance understanding of the general principles underlying neuromotor deficits caused by metal toxicity.
Collapse
Affiliation(s)
- Anna S. Warden
- Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX78712
| | - Nishant Sharma
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Steven Hutchens
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Chunyi Liu
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Noah R. Haggerty
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Kerem C. Gurol
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Thomas Jursa
- Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, CA95064
| | - Donald R. Smith
- Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, CA95064
| | - Roy Dayne Mayfield
- Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX78712
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| |
Collapse
|
3
|
Wei S, Tao HY, Duan Z, Wang Y. Environmental Exposure, Epitranscriptomic Perturbations, and Human Diseases. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:6387-6399. [PMID: 40126397 PMCID: PMC11978485 DOI: 10.1021/acs.est.5c00907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Epitranscriptomics is a rapidly evolving field, and it examines how chemical modifications on RNA regulate gene expression. Increasing lines of evidence support that exposure to various environmental agents can change substantially chemical modifications on RNA, thereby perturbing gene expression and contributing to disease development in humans. However, the molecular mechanisms through which environmental exposure impairs RNA modification-associated proteins ("reader", "writer", and "eraser" or RWE proteins) and alters the landscape of RNA modifications remain poorly understood. Here, we provide our perspectives on the current knowledge about how environmental exposure alters the epitranscriptome, where we focus on dynamic changes in RNA modifications and their regulatory proteins elicited by exposure to environmental agents. We discuss how these epitranscriptomic alterations may contribute to the development of human diseases, especially neurodegeneration and cancer. We also discuss the potential and technical challenges of harnessing RNA modifications as biomarkers for monitoring environmental exposure. Finally, we emphasize the need to integrate multiomics approaches to decipher the complex interplay between environmental exposure and the epitranscriptome and offer a forward-looking viewpoint on future research priorities that may inform public health interventions and environmental regulations.
Collapse
Affiliation(s)
- Songbo Wei
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Huan-Yu Tao
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Zheng Duan
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| |
Collapse
|
4
|
Magro G, Laterza V, Tosto F, Torrente A. Manganese Neurotoxicity: A Comprehensive Review of Pathophysiology and Inherited and Acquired Disorders. J Xenobiot 2025; 15:54. [PMID: 40278159 PMCID: PMC12028444 DOI: 10.3390/jox15020054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/26/2025] Open
Abstract
Manganese (Mn) is an essential trace element and a cofactor for several key enzymes, such as mitochondrial superoxide dismutase. Consequently, it plays an important defense role against reactive oxygen species. Despite this, Mn chronic overexposure can result in a neurological disorder referred to as manganism, which shares some similarities with Parkinson's disease. Mn levels seem regulated by many transporters responsible for its uptake and efflux. These transporters play an established role in many inherited disorders of Mn metabolism and neurotoxicity. Some inherited Mn metabolism disorders, caused by mutations of SLC30A10 and SLC39A14, assume crucial importance since earlier treatment results in a better prognosis. Physicians should be familiar with the clinical presentation of these disorders as the underlying cause of dystonia/parkinsonism and look for other accompanying features, such as liver disease and polycythemia, which are typically associated with SLC30A10 mutations. This review aims to highlight the currently known Mn transporters, Mn-related neurotoxicity, and its consequences, and it provides an overview of inherited and acquired disorders of Mn metabolism. Currently available treatments are also discussed, focusing on the most frequently encountered presentations.
Collapse
Affiliation(s)
- Giuseppe Magro
- Department of Neuroscience, “Giovanni Paolo II” Hospital, Lamezia Terme, 88100 Catanzaro, Italy
| | - Vincenzo Laterza
- Department of Medical and Surgical Sciences, Institute of Neurology, Magna Graecia University, 88100 Catanzaro, Italy
| | - Federico Tosto
- Department of Neuroscience, “Giovanni Paolo II” Hospital, Lamezia Terme, 88100 Catanzaro, Italy
| | - Angelo Torrente
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics (BiND), University of Palermo, 90129 Palermo, Italy;
| |
Collapse
|
5
|
Monsivais H, Dydak U. Subject-Specific Mapping of Excess Manganese Accumulation in the Brain of Welders Using Magnetic Resonance Imaging Relaxometry. TOXICS 2025; 13:157. [PMID: 40137484 PMCID: PMC11945464 DOI: 10.3390/toxics13030157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 03/29/2025]
Abstract
Chronic overexposure to manganese (Mn) can occur in occupational settings, such as welding, leading to increased Mn levels in the brain. Excess brain Mn accumulation may result in neurotoxicity, which is characterized by Parkinsonian-like symptoms including motor and cognitive dysfunctions. In this work, we demonstrate a novel methodology for personalized diagnosis and spatial characterization of abnormal Magnetic Resonance Imaging R1 (R1 = 1/T1) relaxation rates arising from excessive Mn accumulation in welders' brains. Utilizing voxel-wise population-derived norms based on a frequency age-matched non-exposed group (n = 25), we demonstrate the ability to conduct subject-specific assessments and mapping of Mn exposure using MRI relaxometry. Our results show elevated R1 in multiple brain regions in individual welders, but also extreme between-subject variability in Mn accumulation, debasing the concept that high exposures correlate with uniformly high Mn deposition in the brain. Consequently, the presented personalized methodology serves as a counterpart to group-based comparison, which allows for understanding the level of individual exposure and the toxicokinetics of Mn accumulation. This work lays a foundation for improved occupational health assessments and preventive measures against neurotoxic metal exposure.
Collapse
Affiliation(s)
- Humberto Monsivais
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA;
| | - Ulrike Dydak
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA;
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
6
|
Rokad D, Harischandra DS, Samidurai M, Chang YT, Luo J, Lawana V, Sarkar S, Palanisamy BN, Manne S, Kim D, Zenitsky G, Jin H, Anantharam V, Willette A, Kanthasamy A, Kanthasamy AG. Manganese Exposure Enhances the Release of Misfolded α-Synuclein via Exosomes by Impairing Endosomal Trafficking and Protein Degradation Mechanisms. Int J Mol Sci 2024; 25:12207. [PMID: 39596274 PMCID: PMC11594990 DOI: 10.3390/ijms252212207] [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: 08/12/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Excessive exposure to manganese (Mn) increases the risk of chronic neurological diseases, including Parkinson's disease (PD) and other related Parkinsonisms. Aggregated α-synuclein (αSyn), a hallmark of PD, can spread to neighboring cells by exosomal release from neurons. We previously discovered that Mn enhances its spread, triggering neuroinflammatory and neurodegenerative processes. To better understand the Mn-induced release of exosomal αSyn, we examined the effect of Mn on endosomal trafficking and misfolded protein degradation. Exposing MN9D dopaminergic neuronal cells stably expressing human wild-type (WT) αSyn to 300 μM Mn for 24 h significantly suppressed protein and mRNA expression of Rab11a, thereby downregulating endosomal recycling, forcing late endosomes to mature into multivesicular bodies (MVBs). Ectopic expression of WT Rab11a significantly mitigated exosome release, whereas ectopic mutant Rab11a (S25N) increased it. Our in vitro and in vivo studies reveal that Mn exposure upregulated (1) mRNA and protein levels of endosomal Rab27a, which mediates the fusion of MVBs with the plasma membrane; and (2) expression of the autophagosomal markers Beclin-1 and p62, but downregulated the lysosomal marker LAMP2, thereby impairing autophagolysosome formation as confirmed by LysoTracker, cathepsin, and acridine orange assays. Our novel findings demonstrate that Mn promotes the exosomal release of misfolded αSyn by impairing endosomal trafficking and protein degradation.
Collapse
Affiliation(s)
- Dharmin Rokad
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Dilshan S. Harischandra
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Manikandan Samidurai
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| | - Yuan-Teng Chang
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| | - Jie Luo
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Vivek Lawana
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Souvarish Sarkar
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Bharathi N. Palanisamy
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Sireesha Manne
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Dongsuk Kim
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Gary Zenitsky
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| | - Huajun Jin
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| | - Vellareddy Anantharam
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| | - Auriel Willette
- Department of Neurology, Rutgers University, New Brunswick, NJ 07101, USA;
| | - Arthi Kanthasamy
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| | - Anumantha G. Kanthasamy
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| |
Collapse
|
7
|
Fan RZ, Sportelli C, Lai Y, Salehe SS, Pinnell JR, Brown HJ, Richardson JR, Luo S, Tieu K. A partial Drp1 knockout improves autophagy flux independent of mitochondrial function. Mol Neurodegener 2024; 19:26. [PMID: 38504290 PMCID: PMC10953112 DOI: 10.1186/s13024-024-00708-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/07/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Dynamin-related protein 1 (Drp1) plays a critical role in mitochondrial dynamics. Partial inhibition of this protein is protective in experimental models of neurological disorders such as Parkinson's disease and Alzheimer's disease. The protective mechanism has been attributed primarily to improved mitochondrial function. However, the observations that Drp1 inhibition reduces protein aggregation in such neurological disorders suggest the involvement of autophagy. To investigate this potential novel protective mechanism of Drp1 inhibition, a model with impaired autophagy without mitochondrial involvement is needed. METHODS We characterized the effects of manganese (Mn), which causes parkinsonian-like symptoms in humans, on autophagy and mitochondria by performing dose-response studies in two cell culture models (stable autophagy HeLa reporter cells and N27 rat immortalized dopamine neuronal cells). Mitochondrial function was assessed using the Seahorse Flux Analyzer. Autophagy flux was monitored by quantifying the number of autophagosomes and autolysosomes, as well as the levels of other autophagy proteins. To strengthen the in vitro data, multiple mouse models (autophagy reporter mice and mutant Drp1+/- mice and their wild-type littermates) were orally treated with a low chronic Mn regimen that was previously reported to increase α-synuclein aggregation and transmission via exosomes. RNAseq, laser captured microdissection, immunofluorescence, immunoblotting, stereological cell counting, and behavioural studies were used. RESULTS IN VITRO: data demonstrate that at low non-toxic concentrations, Mn impaired autophagy flux but not mitochondrial function and morphology. In the mouse midbrain, RNAseq data further confirmed autophagy pathways were dysregulated but not mitochondrial related genes. Additionally, Mn selectively impaired autophagy in the nigral dopamine neurons but not the nearby nigral GABA neurons. In cells with a partial Drp1-knockdown and Drp1+/- mice, Mn induced autophagic impairment was significantly prevented. Consistent with these observations, Mn increased the levels of proteinase-K resistant α-synuclein and Drp1-knockdown protected against this pathology. CONCLUSIONS This study demonstrates that improved autophagy flux is a separate mechanism conferred by Drp1 inhibition independent of its role in mitochondrial fission. Given that impaired autophagy and mitochondrial dysfunction are two prominent features of neurodegenerative diseases, the combined protective mechanisms targeting these two pathways conferred by Drp1 inhibition make this protein an attractive therapeutic target.
Collapse
Affiliation(s)
- Rebecca Z Fan
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Carolina Sportelli
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Yanhao Lai
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Said S Salehe
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Jennifer R Pinnell
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Harry J Brown
- Department of Environmental Health Sciences, Florida International University, Miami, USA
- Biomolecular Sciences Institute, Florida International University, Miami, USA
| | - Jason R Richardson
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Shouqing Luo
- Peninsula Schools of Medicine and Dentistry, Plymouth University, Plymouth, UK
| | - Kim Tieu
- Department of Environmental Health Sciences, Florida International University, Miami, USA.
- Biomolecular Sciences Institute, Florida International University, Miami, USA.
| |
Collapse
|
8
|
Monsivais H, Yeh CL, Edmondson A, Harold R, Snyder S, Wells EM, Schmidt-Wilcke T, Foti D, Zauber SE, Dydak U. Whole-brain mapping of increased manganese levels in welders and its association with exposure and motor function. Neuroimage 2024; 288:120523. [PMID: 38278427 PMCID: PMC11124758 DOI: 10.1016/j.neuroimage.2024.120523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/29/2023] [Accepted: 01/23/2024] [Indexed: 01/28/2024] Open
Abstract
Although manganese (Mn) is a trace metal essential for humans, chronic exposure to Mn can cause accumulation of this metal ion in the brain leading to an increased risk of neurological and neurobehavioral health effects. This is a concern for welders exposed to Mn through welding fumes. While brain Mn accumulation in occupational settings has mostly been reported in the basal ganglia, several imaging studies also revealed elevated Mn in other brain areas. Since Mn functions as a magnetic resonance imaging (MRI) T1 contrast agent, we developed a whole-brain MRI approach to map in vivo Mn deposition differences in the brains of non-exposed factory controls and exposed welders. This is a cross-sectional analysis of 23 non-exposed factory controls and 36 exposed full-time welders from the same truck manufacturer. We collected high-resolution 3D MRIs of brain anatomy and R1 relaxation maps to identify regional differences using voxel-based quantification (VBQ) and statistical parametric mapping. Furthermore, we investigated the associations between excess Mn deposition and neuropsychological and motor test performance. Our results indicate that: (1) Using whole-brain MRI relaxometry methods we can generate excess Mn deposition maps in vivo, (2) excess Mn accumulation due to occupational exposure occurs beyond the basal ganglia in cortical areas associated with motor and cognitive functions, (3) Mn likely diffuses along white matter tracts in the brain, and (4) Mn deposition in specific brain regions is associated with exposure (cerebellum and frontal cortex) and motor metrics (cerebellum and hippocampus).
Collapse
Affiliation(s)
| | - Chien-Lin Yeh
- School of Health Sciences, Purdue University, West Lafayette, IN, USA; Takeda Pharmaceutical Company Ltd, Cambridge, MA, USA
| | - Alex Edmondson
- Cincinnati Children's Hospital Medical Center, Imaging Research Center, Cincinnati, OH, USA; University of Cincinnati College of Medicine, Department of Environmental and Public Health Sciences, Cincinnati, OH USA
| | - Roslyn Harold
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, USA
| | - Sandy Snyder
- School of Health Sciences, Purdue University, West Lafayette, IN, USA; Department of Speech, Language and Hearing Sciences, Purdue University, West Lafayette, IN, USA
| | - Ellen M Wells
- School of Health Sciences, Purdue University, West Lafayette, IN, USA; Department of Public Health, Purdue University, West Lafayette, IN, USA
| | - Tobias Schmidt-Wilcke
- Department of Neurology, St. Mauritius Therapieklinik, Meerbusch, Germany; Institute of Clinical Neuroscience and Medical Psychology, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Dan Foti
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, USA
| | - S Elizabeth Zauber
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
| | - Ulrike Dydak
- School of Health Sciences, Purdue University, West Lafayette, IN, USA; Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
9
|
Mattison DR, Momoli F, Alyanak C, Aschner M, Baker M, Cashman N, Dydak U, Farhat N, Guilarte TR, Karyakina N, Ramoju S, Shilnikova N, Taba P, Krewski D. Diagnosis of manganism and manganese neurotoxicity: A workshop report. MEDICINE INTERNATIONAL 2024; 4:11. [PMID: 38410758 PMCID: PMC10895461 DOI: 10.3892/mi.2024.135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 01/19/2024] [Indexed: 02/28/2024]
Abstract
With declining exposures to manganese (Mn) in occupational settings, there is a need for more sensitive exposure assessments and clinical diagnostic criteria for manganism and Mn neurotoxicity. To address this issue, a workshop was held on November 12-13, 2020, with international experts on Mn toxicity. The workshop discussions focused on the history of the diagnostic criteria for manganism, including those developed by the Institut de Recherche Robert-Sauvé en Santé et en Sécurité du Travail (IRSST) in Quebec in 2005 and criteria developed by the Chinese government in 2002 and updated in 2006; the utility of biomarkers of exposure; recent developments in magnetic resonance imaging (MRI) for assessing Mn accumulation in the brain and diagnosing manganism; and potential future applications of metabolomics. The suggestions of the participants for updating manganism diagnostic criteria included the consideration of: i) A history of previous occupational and environmental exposure to Mn; ii) relevant clinical symptoms such as dystonia; iii) MRI imaging to document Mn accumulation in the neural tissues, including the basal ganglia; and iv) criteria for the differential diagnosis of manganism and other neurological conditions. Important research gaps include the characterization of Mn exposure and other co-exposures, exploration of the roles of different brain regions with MRI, understanding the complexity of metal ion transporters involved in Mn homeostasis, and a need for information on other neurotransmitter systems and brain regions underlying the pathophysiology of manganism.
Collapse
Affiliation(s)
- Donald R. Mattison
- Risk Sciences International, Ottawa, ON K1P 5J6, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
- Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Franco Momoli
- Risk Sciences International, Ottawa, ON K1P 5J6, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
| | - Cemil Alyanak
- Risk Sciences International, Ottawa, ON K1P 5J6, Canada
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Marissa Baker
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Neil Cashman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- ProMIS Neurosciences, Inc., Toronto, ON M4S 3E2, Canada
| | - Ulrike Dydak
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Nawal Farhat
- Risk Sciences International, Ottawa, ON K1P 5J6, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
| | - Tomás R. Guilarte
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | | | - Siva Ramoju
- Risk Sciences International, Ottawa, ON K1P 5J6, Canada
| | - Natalia Shilnikova
- Risk Sciences International, Ottawa, ON K1P 5J6, Canada
- R. Samuel McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
| | - Pille Taba
- Department of Neurology and Neurosurgery, Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia
- Neurology Clinic, Tartu University Hospital, 50406 Tartu, Estonia
| | - Daniel Krewski
- Risk Sciences International, Ottawa, ON K1P 5J6, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
- R. Samuel McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
| |
Collapse
|
10
|
Lee EY, Kim J, Prado-Rico JM, Du G, Lewis MM, Kong L, Yanosky JD, Eslinger P, Kim BG, Hong YS, Mailman RB, Huang X. Effects of mixed metal exposures on MRI diffusion features in the medial temporal lobe. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.07.18.23292828. [PMID: 37503124 PMCID: PMC10371112 DOI: 10.1101/2023.07.18.23292828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Background Environmental exposure to metal mixtures is common and may be associated with increased risk for neurodegenerative disorders including Alzheimer's disease. Objective This study examined associations of mixed metal exposures with medial temporal lobe (MTL) MRI structural metrics and neuropsychological performance. Methods Metal exposure history, whole blood metal, and neuropsychological tests were obtained from subjects with/without a history of mixed metal exposure from welding fumes (42 exposed subjects; 31 controls). MTL structures (hippocampus, entorhinal and parahippocampal cortices) were assessed by morphologic (volume, cortical thickness) and diffusion tensor imaging [mean (MD), axial (AD), radial diffusivity (RD), and fractional anisotropy (FA)] metrics. In exposed subjects, correlation, multiple linear, Bayesian kernel machine regression, and mediation analyses were employed to examine effects of single- or mixed-metal predictor(s) and their interactions on MTL structural and neuropsychological metrics; and on the path from metal exposure to neuropsychological consequences. Results Compared to controls, exposed subjects had higher blood Cu, Fe, K, Mn, Pb, Se, and Zn levels (p's<0.026) and poorer performance in processing/psychomotor speed, executive, and visuospatial domains (p's<0.046). Exposed subjects displayed higher MD, AD, and RD in all MTL ROIs (p's<0.040) and lower FA in entorhinal and parahippocampal cortices (p's<0.033), but not morphological differences. Long-term mixed-metal exposure history indirectly predicted lower processing speed performance via lower parahippocampal FA (p=0.023). Higher whole blood Mn and Cu predicted higher entorhinal diffusivity (p's<0.043) and lower Delayed Story Recall performance (p=0.007) without overall metal mixture or interaction effects. Discussion Mixed metal exposure predicted MTL structural and neuropsychological features that are similar to Alzheimer's disease at-risk populations. These data warrant follow-up as they may illuminate the path for environmental exposure to Alzheimer's disease-related health outcomes.
Collapse
Affiliation(s)
- Eun-Young Lee
- Department of Health Care and Science, Dong-A University, Busan, South-Korea
| | - Juhee Kim
- Department of Health Care and Science, Dong-A University, Busan, South-Korea
| | - Janina Manzieri Prado-Rico
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Guangwei Du
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Mechelle M. Lewis
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Lan Kong
- Department of Public Health Sciences, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Jeff D. Yanosky
- Department of Public Health Sciences, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Paul Eslinger
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Byoung-Gwon Kim
- Department of Preventive Medicine, College of Medicine, Dong-A University, Busan, South Korea
| | - Young-Seoub Hong
- Department of Preventive Medicine, College of Medicine, Dong-A University, Busan, South Korea
| | - Richard B. Mailman
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Xuemei Huang
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Radiology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Neurosurgery, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Kinesiology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| |
Collapse
|
11
|
Ghosn ZA, Sparks KM, Spaulding JL, Vutukuri S, Ahmed MJJ, VanBerkum MFA. Divalent metal content in diet affects severity of manganese toxicity in Drosophila. Biol Open 2024; 13:bio060204. [PMID: 38117005 PMCID: PMC10810561 DOI: 10.1242/bio.060204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023] Open
Abstract
Dysregulation of manganese (Mn) homeostasis is a contributing factor in many neuro-degenerative diseases. Adult Drosophila are sensitive to excessive levels of dietary Mn, dying relatively early, and exhibiting biochemical and mobility changes reminiscent of Parkinsonian conditions. To further study Mn homeostasis in Drosophila, we sought to test lower levels of dietary Mn (5 mM) and noted a striking difference in Canton-S adult survivorship on different food. On a cornmeal diet, Mn-treated flies live only about half as long as untreated siblings. Yet, with the same Mn concentration in a molasses diet, adults survive about 80% as long as untreated siblings, and adults raised on a sucrose-yeast diet are completely insensitive to this low dose of dietary Mn. By manipulating metal ion content in the cornmeal diet, and measuring the metal content in each diet, we traced the difference in lifespan to the levels of calcium and magnesium in the food, suggesting that these ions are involved in Mn uptake and/or use. Based on these findings, it is recommended that the total dietary load of metal ions be considered when assessing Mn toxicity.
Collapse
Affiliation(s)
- Zahraa A. Ghosn
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Kailynn M. Sparks
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Jacob L. Spaulding
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Sanjana Vutukuri
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Mirza J. J. Ahmed
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Mark F. A. VanBerkum
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
12
|
Vaidya B, Padhy DS, Joshi HC, Sharma SS, Singh JN. Ion Channels and Metal Ions in Parkinson's Disease: Historical Perspective to the Current Scenario. Methods Mol Biol 2024; 2761:529-557. [PMID: 38427260 DOI: 10.1007/978-1-0716-3662-6_36] [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] [Indexed: 03/02/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative condition linked to the deterioration of motor and cognitive performance. It produces degeneration of the dopaminergic neurons along the nigrostriatal pathway in the central nervous system (CNS), which leads to symptoms such as bradykinesias, tremors, rigidity, and postural instability. There are several medications currently approved for the therapy of PD, but a permanent cure for it remains elusive. With the aging population set to increase, a number of PD cases are expected to shoot up in the coming times. Hence, there is a need to look for new molecular targets that could be investigated both preclinically and clinically for PD treatment. Among these, several ion channels and metal ions are being studied for their effects on PD pathology and the functioning of dopaminergic neurons. Ion channels such as N-methyl-D-aspartate (NMDA), γ-aminobutyric acid A (GABAA), voltage-gated calcium channels, potassium channels, HCN channels, Hv1 proton channels, and voltage-gated sodium channels and metal ions such as mercury, zinc, copper, iron, manganese, calcium, and lead showed prominent involvement in PD. Pharmacological agents have been used to target these ion channels and metal ions to prevent or treat PD. Hence, in the present review, we summarize the pathophysiological events linked to PD with an emphasis on the role of ions and ion channels in PD pathology, and pharmacological agents targeting these ion channels have also been listed.
Collapse
Affiliation(s)
- Bhupesh Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India
| | - Dibya S Padhy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India
| | - Hem C Joshi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India
| | - Shyam S Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India.
| | - Jitendra Narain Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India.
| |
Collapse
|
13
|
Gionco JT, Bernstein AI. Emerging Role of Environmental Epitranscriptomics and RNA Modifications in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:643-656. [PMID: 38578904 PMCID: PMC11191529 DOI: 10.3233/jpd-230457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/10/2024] [Indexed: 04/07/2024]
Abstract
Environmental risk factors and gene-environment interactions play a critical role in Parkinson's disease (PD). However, the relatively large contribution of environmental risk factors in the overwhelming majority of PD cases has been widely neglected in the field. A "PD prevention agenda" proposed in this journal laid out a set of research priorities focused on preventing PD through modification of environmental risk factors. This agenda includes a call for preclinical studies to employ new high-throughput methods for analyzing transcriptomics and epigenomics to provide a deeper understanding of the effects of exposures linked to PD. Here, we focus on epitranscriptomics as a novel area of research with the potential to add to our understanding of the interplay between genes and environmental exposures in PD. Both epigenetics and epitranscriptomics have been recognized as potential mediators of the complex relationship between genes, environment, and disease. Multiple studies have identified epigenetic alterations, such as DNA methylation, associated with PD and PD-related exposures in human studies and preclinical models. In addition, recent technological advancements have made it possible to study epitranscriptomic RNA modifications, such as RNA N6-methyladenosine (m6A), and a handful of recent studies have begun to explore epitranscriptomics in PD-relevant exposure models. Continued exploration of epitranscriptomic mechanisms in environmentally relevant PD models offers the opportunity to identify biomarkers, pre-degenerative changes that precede symptom onset, and potential mitigation strategies for disease prevention and treatment.
Collapse
Affiliation(s)
- John T. Gionco
- Graduate Program in Cell and Developmental Biology, Rutgers University, Piscataway, NJ, USA
| | - Alison I. Bernstein
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
14
|
Key J, Gispert S, Koepf G, Steinhoff-Wagner J, Reichlmeir M, Auburger G. Translation Fidelity and Respiration Deficits in CLPP-Deficient Tissues: Mechanistic Insights from Mitochondrial Complexome Profiling. Int J Mol Sci 2023; 24:17503. [PMID: 38139332 PMCID: PMC10743472 DOI: 10.3390/ijms242417503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
The mitochondrial matrix peptidase CLPP is crucial during cell stress. Its loss causes Perrault syndrome type 3 (PRLTS3) with infertility, neurodegeneration, and a growth deficit. Its target proteins are disaggregated by CLPX, which also regulates heme biosynthesis via unfolding ALAS enzymes, providing access for pyridoxal-5'-phosphate (PLP). Despite efforts in diverse organisms with multiple techniques, CLPXP substrates remain controversial. Here, avoiding recombinant overexpression, we employed complexomics in mitochondria from three mouse tissues to identify endogenous targets. A CLPP absence caused the accumulation and dispersion of CLPX-VWA8 as AAA+ unfoldases, and of PLPBP. Similar changes and CLPX-VWA8 co-migration were evident for mitoribosomal central protuberance clusters, translation factors like GFM1-HARS2, the RNA granule components LRPPRC-SLIRP, and enzymes OAT-ALDH18A1. Mitochondrially translated proteins in testes showed reductions to <30% for MTCO1-3, the mis-assembly of the complex IV supercomplex, and accumulated metal-binding assembly factors COX15-SFXN4. Indeed, heavy metal levels were increased for iron, molybdenum, cobalt, and manganese. RT-qPCR showed compensatory downregulation only for Clpx mRNA; most accumulated proteins appeared transcriptionally upregulated. Immunoblots validated VWA8, MRPL38, MRPL18, GFM1, and OAT accumulation. Co-immunoprecipitation confirmed CLPX binding to MRPL38, GFM1, and OAT, so excess CLPX and PLP may affect their activity. Our data mechanistically elucidate the mitochondrial translation fidelity deficits which underlie progressive hearing impairment in PRLTS3.
Collapse
Affiliation(s)
- Jana Key
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (S.G.); (M.R.); (G.A.)
| | - Suzana Gispert
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (S.G.); (M.R.); (G.A.)
| | - Gabriele Koepf
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (S.G.); (M.R.); (G.A.)
| | - Julia Steinhoff-Wagner
- TUM School of Life Sciences, Animal Nutrition and Metabolism, Technical University of Munich, Liesel-Beckmann-Str. 2, 85354 Freising-Weihenstephan, Germany;
| | - Marina Reichlmeir
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (S.G.); (M.R.); (G.A.)
| | - Georg Auburger
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (S.G.); (M.R.); (G.A.)
| |
Collapse
|
15
|
Turck D, Bohn T, Castenmiller J, de Henauw S, Hirsch‐Ernst K, Knutsen HK, Maciuk A, Mangelsdorf I, McArdle HJ, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Bornhorst J, Cubadda F, Dopter A, FitzGerald R, de Sesmaisons Lecarré A, das Neves Ferreira P, Fabiani L, Horvath Z, Matijević L, Naska A. Scientific opinion on the tolerable upper intake level for manganese. EFSA J 2023; 21:e8413. [PMID: 38075631 PMCID: PMC10704406 DOI: 10.2903/j.efsa.2023.8413] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024] Open
Abstract
Following a request from the European Commission (EC), the EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) was asked to deliver a scientific opinion on the tolerable upper intake level (UL) for manganese. Systematic reviews of the literature of human and animal data were conducted to assess evidence regarding excess manganese intake (including authorised manganese salts) and the priority adverse health effect, i.e. manganese-induced neurotoxicity. Available human and animal studies support neurotoxicity as a critical effect, however, data are not sufficient and suitable to characterise a dose-response relationship and identify a reference point for manganese-induced neurotoxicity. In the absence of adequate data to establish an UL, estimated background dietary intakes (i.e. manganese intakes from natural dietary sources only) observed among high consumers (95th percentile) were used to provide an indication of the highest level of intake where there is reasonable confidence on the absence of adverse effects. A safe level of intake of 8 mg/day was established for adults ≥ 18 years (including pregnant and lactating women) and ranged between 2 and 7 mg/day for other population groups. The application of the safe level of intake is more limited than an UL because the intake level at which the risk of adverse effects starts to increase is not defined.
Collapse
|
16
|
Baj J, Flieger W, Barbachowska A, Kowalska B, Flieger M, Forma A, Teresiński G, Portincasa P, Buszewicz G, Radzikowska-Büchner E, Flieger J. Consequences of Disturbing Manganese Homeostasis. Int J Mol Sci 2023; 24:14959. [PMID: 37834407 PMCID: PMC10573482 DOI: 10.3390/ijms241914959] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
Manganese (Mn) is an essential trace element with unique functions in the body; it acts as a cofactor for many enzymes involved in energy metabolism, the endogenous antioxidant enzyme systems, neurotransmitter production, and the regulation of reproductive hormones. However, overexposure to Mn is toxic, particularly to the central nervous system (CNS) due to it causing the progressive destruction of nerve cells. Exposure to manganese is widespread and occurs by inhalation, ingestion, or dermal contact. Associations have been observed between Mn accumulation and neurodegenerative diseases such as manganism, Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. People with genetic diseases associated with a mutation in the gene associated with impaired Mn excretion, kidney disease, iron deficiency, or a vegetarian diet are at particular risk of excessive exposure to Mn. This review has collected data on the current knowledge of the source of Mn exposure, the experimental data supporting the dispersive accumulation of Mn in the brain, the controversies surrounding the reference values of biomarkers related to Mn status in different matrices, and the competitiveness of Mn with other metals, such as iron (Fe), magnesium (Mg), zinc (Zn), copper (Cu), lead (Pb), calcium (Ca). The disturbed homeostasis of Mn in the body has been connected with susceptibility to neurodegenerative diseases, fertility, and infectious diseases. The current evidence on the involvement of Mn in metabolic diseases, such as type 2 diabetes mellitus/insulin resistance, osteoporosis, obesity, atherosclerosis, and non-alcoholic fatty liver disease, was collected and discussed.
Collapse
Affiliation(s)
- Jacek Baj
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Wojciech Flieger
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Aleksandra Barbachowska
- Department of Plastic, Reconstructive and Burn Surgery, Medical University of Lublin, 21-010 Łęczna, Poland;
| | - Beata Kowalska
- Department of Water Supply and Wastewater Disposal, Lublin University of Technology, 20-618 Lublin, Poland;
| | - Michał Flieger
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | - Alicja Forma
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Grzegorz Teresiński
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | - Piero Portincasa
- Clinica Medica A. Murri, Department of Biomedical Sciences & Human Oncology, Medical School, University of Bari, 70124 Bari, Italy;
| | - Grzegorz Buszewicz
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | | | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
17
|
Liu Q, Jenkitkasemwong S, Prami TA, McCabe SM, Zhao N, Hojyo S, Fukada T, Knutson MD. Metal-ion transporter SLC39A8 is required for brain manganese uptake and accumulation. J Biol Chem 2023; 299:105078. [PMID: 37482277 PMCID: PMC10457451 DOI: 10.1016/j.jbc.2023.105078] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/25/2023] Open
Abstract
Manganese (Mn) is an essential nutrient, but is toxic in excess. Whole-body Mn levels are regulated in part by the metal-ion influx transporter SLC39A8, which plays an essential role in the liver by reclaiming Mn from bile. Physiological roles of SLC39A8 in Mn homeostasis in other tissues, however, remain largely unknown. To screen for extrahepatic requirements for SLC39A8 in tissue Mn homeostasis, we crossed Slc39a8-inducible global-KO (Slc39a8 iKO) mice with Slc39a14 KO mice, which display markedly elevated blood and tissue Mn levels. Tissues were then analyzed by inductively coupled plasma-mass spectrometry to determine levels of Mn. Although Slc39a14 KO; Slc39a8 iKO mice exhibited systemic hypermanganesemia and increased Mn loading in the bone and kidney due to Slc39a14 deficiency, we show Mn loading was markedly decreased in the brains of these animals, suggesting a role for SLC39A8 in brain Mn accumulation. Levels of other divalent metals in the brain were unaffected, indicating a specific effect of SLC39A8 on Mn. In vivo radiotracer studies using 54Mn in Slc39a8 iKO mice revealed that SLC39A8 is required for Mn uptake by the brain, but not most other tissues. Furthermore, decreased 54Mn uptake in the brains of Slc39a8 iKO mice was associated with efficient inactivation of Slc39a8 in isolated brain microvessels but not in isolated choroid plexus, suggesting SLC39A8 mediates brain Mn uptake via the blood-brain barrier. These findings establish SLC39A8 as a candidate therapeutic target for mitigating Mn uptake and accumulation in the brain, the primary organ of Mn toxicity.
Collapse
Affiliation(s)
- Qingli Liu
- Food Science and Human Nutrition Department, University of Florida, Gainesville, Florida, USA
| | - Supak Jenkitkasemwong
- Food Science and Human Nutrition Department, University of Florida, Gainesville, Florida, USA
| | - Tamanna Afrin Prami
- Food Science and Human Nutrition Department, University of Florida, Gainesville, Florida, USA
| | - Shannon Morgan McCabe
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, Arizona, USA
| | - Ningning Zhao
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, Arizona, USA
| | - Shintaro Hojyo
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Toshiyuki Fukada
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Mitchell D Knutson
- Food Science and Human Nutrition Department, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
18
|
Lucchini R, Tieu K. Manganese-Induced Parkinsonism: Evidence from Epidemiological and Experimental Studies. Biomolecules 2023; 13:1190. [PMID: 37627255 PMCID: PMC10452806 DOI: 10.3390/biom13081190] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Manganese (Mn) exposure has evolved from acute, high-level exposure causing manganism to low, chronic lifetime exposure. In this latter scenario, the target areas extend beyond the globus pallidus (as seen with manganism) to the entire basal ganglia, including the substantia nigra pars compacta. This change of exposure paradigm has prompted numerous epidemiological investigations of the occurrence of Parkinson's disease (PD), or parkinsonism, due to the long-term impact of Mn. In parallel, experimental research has focused on the underlying pathogenic mechanisms of Mn and its interactions with genetic susceptibility. In this review, we provide evidence from both types of studies, with the aim to link the epidemiological data with the potential mechanistic interpretation.
Collapse
Affiliation(s)
- Roberto Lucchini
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Kim Tieu
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
19
|
Fan RZ, Sportelli C, Lai Y, Salehe S, Pinnell JR, Richardson JR, Luo S, Tieu K. A partial Drp1 knockout improves autophagy flux independent of mitochondrial function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547095. [PMID: 37425803 PMCID: PMC10327068 DOI: 10.1101/2023.06.29.547095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Dynamin-related protein 1 (Drp1) is typically known for its role in mitochondrial fission. A partial inhibition of this protein has been reported to be protective in experimental models of neurodegenerative diseases. The protective mechanism has been attributed primarily to improved mitochondrial function. Herein, we provide evidence showing that a partial Drp1-knockout improves autophagy flux independent of mitochondria. First, we characterized in cell and animal models that at low non-toxic concentrations, manganese (Mn), which causes parkinsonian-like symptoms in humans, impaired autophagy flux but not mitochondrial function and morphology. Furthermore, nigral dopaminergic neurons were more sensitive than their neighbouring GABAergic counterparts. Second, in cells with a partial Drp1-knockdown and Drp1 +/- mice, autophagy impairment induced by Mn was significantly attenuated. This study demonstrates that autophagy is a more vulnerable target than mitochondria to Mn toxicity. Furthermore, improving autophagy flux is a separate mechanism conferred by Drp1 inhibition independent of mitochondrial fission.
Collapse
|
20
|
Lee EY, Kim J, Prado-Rico JM, Du G, Lewis MM, Kong L, Kim BG, Hong YS, Yanosky JD, Mailman RB, Huang X. Higher hippocampal diffusivity values in welders are associated with greater R2* in the red nucleus and lower psychomotor performance. Neurotoxicology 2023; 96:53-68. [PMID: 36966945 PMCID: PMC10445214 DOI: 10.1016/j.neuro.2023.03.005] [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: 11/03/2022] [Revised: 02/17/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023]
Abstract
INTRODUCTION Chronic excessive welding exposure may be related to higher metal accumulation and structural differences in different subcortical structures. We examined how welding affected brain structures and their associations with metal exposure and neurobehavioral consequences. METHODS Study includes 42 welders and 31 controls without a welding history. Welding-related structural differences were assessed by volume and diffusion tensor imaging (DTI) metrics in basal ganglia, red nucleus (RN), and hippocampus. Metal exposure was estimated by both exposure questionnaires and whole blood metal levels. Brain metal accumulations were estimated by R1 (for Mn) and R2* (for Fe). Neurobehavioral status was assessed by standard neuropsychological tests. RESULTS Compared to controls, welders displayed higher hippocampal mean (MD), axial (AD), and radial diffusivity (RD) (p's < 0.036), but similar DTI or volume in other ROIs (p's > 0.117). Welders had higher blood metal levels (p's < 0.004), higher caudate and RN R2* (p's < 0.014), and lower performance on processing/psychomotor speed, executive function, and visuospatial processing tasks (p's < 0.046). Higher caudate and RN R2* were associated with higher blood Fe and Pb (p's < 0.043), respectively. RN R2* was a significant predictor of all hippocampal diffusivity metrics (p's < 0.006). Higher hippocampal MD and RD values were associated with lower Trail Making Test-A scores (p's < 0.025). A mediation analysis of both groups revealed blood Pb indirectly affected hippocampal diffusivity via RN R2* (p's < 0.041). DISCUSSION Welding-related higher hippocampal diffusivity metrics may be associated with higher RN R2* and lower psychomotor speed performance. Future studies are warranted to test the role of Pb exposure in these findings.
Collapse
Affiliation(s)
- Eun-Young Lee
- Department of Health Care and Science, Dong-A University, Busan, South Korea.
| | - Juhee Kim
- Department of Health Care and Science, Dong-A University, Busan, South Korea
| | - Janina Manzieri Prado-Rico
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Guangwei Du
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Mechelle M Lewis
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA 17033, USA; Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Lan Kong
- Department of Public Health Sciences, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Byoung-Gwon Kim
- Department of Preventive Medicine, College of Medicine, Dong-A University, Busan, South Korea
| | - Young-Seoub Hong
- Department of Preventive Medicine, College of Medicine, Dong-A University, Busan, South Korea
| | - Jeff D Yanosky
- Department of Public Health Sciences, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Richard B Mailman
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA 17033, USA; Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Xuemei Huang
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA 17033, USA; Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA 17033, USA; Department of Radiology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA 17033, USA; Department of Neurosurgery, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA 17033, USA; Department of Kinesiology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA 17033, USA.
| |
Collapse
|
21
|
Taylor CA, Grant SM, Jursa T, Melkote A, Fulthorpe R, Aschner M, Smith DR, Gonzales RA, Mukhopadhyay S. SLC30A10 manganese transporter in the brain protects against deficits in motor function and dopaminergic neurotransmission under physiological conditions. Metallomics 2023; 15:mfad021. [PMID: 36990693 PMCID: PMC10103839 DOI: 10.1093/mtomcs/mfad021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/28/2023] [Indexed: 03/31/2023]
Abstract
Loss-of-function mutations in SLC30A10 induce hereditary manganese (Mn)-induced neuromotor disease in humans. We previously identified SLC30A10 to be a critical Mn efflux transporter that controls physiological brain Mn levels by mediating hepatic and intestinal Mn excretion in adolescence/adulthood. Our studies also revealed that in adulthood, SLC30A10 in the brain regulates brain Mn levels when Mn excretion capacity is overwhelmed (e.g. after Mn exposure). But, the functional role of brain SLC30A10 under physiological conditions is unknown. We hypothesized that, under physiological conditions, brain SLC30A10 may modulate brain Mn levels and Mn neurotoxicity in early postnatal life because body Mn excretion capacity is reduced in this developmental stage. We discovered that Mn levels of pan-neuronal/glial Slc30a10 knockout mice were elevated in specific brain regions (thalamus) during specific stages of early postnatal development (postnatal day 21), but not in adulthood. Furthermore, adolescent or adult pan-neuronal/glial Slc30a10 knockouts exhibited neuromotor deficits. The neuromotor dysfunction of adult pan-neuronal/glial Slc30a10 knockouts was associated with a profound reduction in evoked striatal dopamine release without dopaminergic neurodegeneration or changes in striatal tissue dopamine levels. Put together, our results identify a critical physiological function of brain SLC30A10-SLC30A10 in the brain regulates Mn levels in specific brain regions and periods of early postnatal life, which protects against lasting deficits in neuromotor function and dopaminergic neurotransmission. These findings further suggest that a deficit in dopamine release may be a likely cause of early-life Mn-induced motor disease.
Collapse
Affiliation(s)
- Cherish A Taylor
- Division of Pharmacology & Toxicology, College of Pharmacy; and Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Stephanie M Grant
- Division of Pharmacology & Toxicology, College of Pharmacy; and Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Thomas Jursa
- Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, CA 95064, USA
| | - Ashvini Melkote
- Division of Pharmacology & Toxicology, College of Pharmacy; and Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Rebecca Fulthorpe
- Division of Pharmacology & Toxicology, College of Pharmacy; and Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx NY 10461, USA
| | - Donald R Smith
- Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, CA 95064, USA
| | - Rueben A Gonzales
- Division of Pharmacology & Toxicology, College of Pharmacy; and Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology & Toxicology, College of Pharmacy; and Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
22
|
Gurol KC, Li D, Broberg K, Mukhopadhyay S. Manganese efflux transporter SLC30A10 missense polymorphism T95I associated with liver injury retains manganese efflux activity. Am J Physiol Gastrointest Liver Physiol 2023; 324:G78-G88. [PMID: 36414535 PMCID: PMC9829465 DOI: 10.1152/ajpgi.00213.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/18/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022]
Abstract
The activity of the manganese (Mn) efflux transporter SLC30A10 in the liver and intestines is critical for Mn excretion and preventing Mn toxicity. Homozygous loss-of-function mutations in SLC30A10 are a well-established cause of hereditary Mn toxicity. But, the relationship between more common SLC30A10 polymorphisms, Mn homeostasis, and disease is only recently emerging. In 2021, the first coding SNP in SLC30A10 (T95I) was associated with liver disease raising the hypothesis that the T95I substitution may induce disease by inhibiting the Mn efflux function of SLC30A10. Here, we test this hypothesis using structural, viability, and metal quantification approaches. Analyses of a predicted structure of SLC30A10 revealed that the side chain of T95 pointed away from the putative Mn-binding cavity, raising doubts about the impact of the T95I substitution on SLC30A10 function. In HeLa or HepG2 cells, overexpression of SLC30A10-WT or T95I resulted in comparable reductions of intracellular Mn levels and protection against Mn-induced cell death. Furthermore, ΔSLC30A10 HepG2 cells, generated using CRISPR/Cas9, exhibited elevated Mn levels and heightened sensitivity to Mn-induced cell death, and these phenotypic changes were similarly rescued by expression of SLC30A10-WT or T95I. Finally, turnover rates of SLC30A10-WT or T95I were also comparable. In summary, our results indicate that the Mn transport activity of SLC30A10-T95I is essentially comparable to the WT protein. Our findings imply that SLC30A10-T95I either has a complex association with liver injury that extends beyond the simple reduction in SLC30A10 activity or alternatively the T95I mutation lacks a causal role in liver disease.NEW & NOTEWORTHY This study demonstrates that the T95I polymorphism in the manganese transporter SLC30A10, which has been associated with liver disease in human GWAS studies, does not impact transporter function in cell culture. These findings raise doubts about the causal relationship of the T95I polymorphism with human disease and highlight the importance of validating GWAS findings using mechanistic approaches.
Collapse
Affiliation(s)
- Kerem C Gurol
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| | - Danyang Li
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| | - Karin Broberg
- Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
23
|
Muacevic A, Adler JR, Kassiri N. An Overview of the Relationship Between Occupational Manganese Exposure and Parkinsonism. Cureus 2022; 14:e32161. [PMID: 36601184 PMCID: PMC9807224 DOI: 10.7759/cureus.32161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2022] [Indexed: 12/05/2022] Open
Abstract
Manganese (Mn) is an essential element used in many industries, such as welding, foundries, the production of metal alloys, especially stainless steel, and the production of dry batteries, pesticides, paints, and explosives. Individuals are exposed to Mn through inhalation of fumes, dermal absorption, and ingestion. This metal is an essential trace element required for normal growth, development, and cellular homeostasis. It has also toxic effects on the central nervous system and can cause Parkinsonism symptoms in exposed patients. Studies on human and animal models reveal that neurons of the globus pallidus, the cerebellum, pons, red nucleus, the thalamus, cortex, and the anterior horn of the spinal cord could be affected by Mn toxicity. Although the diagnosis of manganese-induced Parkinsonism is primarily clinical, there are some supporting features on brain MRI images that may be helpful to objectively distinguish it. This study was designed to review the ways of exposure to Mn, clinical symptoms in case of exposure, and discover the relationship between exposure to Mn and Parkinsonism in the working population.
Collapse
|
24
|
Rodichkin AN, Guilarte TR. Hereditary Disorders of Manganese Metabolism: Pathophysiology of Childhood-Onset Dystonia-Parkinsonism in SLC39A14 Mutation Carriers and Genetic Animal Models. Int J Mol Sci 2022; 23:12833. [PMID: 36361624 PMCID: PMC9653914 DOI: 10.3390/ijms232112833] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 07/30/2023] Open
Abstract
Over the last decade, several clinical reports have outlined cases of childhood-onset manganese (Mn)-induced dystonia-parkinsonism, resulting from loss-of-function mutations in the Mn influx transporter gene SLC39A14. These clinical cases have provided a wealth of knowledge on Mn toxicity and homeostasis. However, our current understanding of the underlying neuropathophysiology is severely lacking. The recent availability of Slc39a14 knockout (KO) murine and zebrafish animal models provide a powerful platform to investigate the neurological effects of elevated blood and brain Mn concentrations in vivo. As such, the objective of this review was to organize and summarize the current clinical literature and studies utilizing Slc39a14-KO animal models and assess the validity of the animal models based on the clinical presentation of the disease in human mutation carriers.
Collapse
|
25
|
Chhetri G, Ke Y, Wang P, Usman M, Li Y, Sapp E, Wang J, Ghosh A, Islam MA, Wang X, Boudi A, DiFiglia M, Li X. Impaired XK recycling for importing manganese underlies striatal vulnerability in Huntington's disease. J Cell Biol 2022; 221:213461. [PMID: 36099524 PMCID: PMC9475296 DOI: 10.1083/jcb.202112073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/15/2022] [Accepted: 07/29/2022] [Indexed: 02/08/2023] Open
Abstract
Mutant huntingtin, which causes Huntington's disease (HD), is ubiquitously expressed but induces preferential loss of striatal neurons by unclear mechanisms. Rab11 dysfunction mediates homeostatic disturbance of HD neurons. Here, we report that Rab11 dysfunction also underscores the striatal vulnerability in HD. We profiled the proteome of Rab11-positive endosomes of HD-vulnerable striatal cells to look for protein(s) linking Rab11 dysfunction to striatal vulnerability in HD and found XK, which triggers the selective death of striatal neurons in McLeod syndrome. XK was trafficked together with Rab11 and was diminished on the surface of immortalized HD striatal cells and striatal neurons in HD mouse brains. We found that XK participated in transporting manganese, an essential trace metal depleted in HD brains. Introducing dominantly active Rab11 into HD striatal cells improved XK dynamics and increased manganese accumulation in an XK-dependent manner. Our study suggests that impaired Rab11-based recycling of XK onto cell surfaces for importing manganese is a driver of striatal dysfunction in Huntington's disease.
Collapse
Affiliation(s)
- Gaurav Chhetri
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yuting Ke
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | - Ping Wang
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA.,Department of Neurology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji'nan, China
| | - Muhammad Usman
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Ellen Sapp
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | - Jing Wang
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, China
| | - Arabinda Ghosh
- Department of Botany, Microbiology Division, Gauhati University, Guwahati, Assam, India
| | - Md Ariful Islam
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaolong Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Adel Boudi
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | - Marian DiFiglia
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | - Xueyi Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| |
Collapse
|
26
|
Rodichkin AN, Edler MK, McGlothan JL, Guilarte TR. Pathophysiological studies of aging Slc39a14 knockout mice to assess the progression of manganese-induced dystonia-parkinsonism. Neurotoxicology 2022; 93:92-102. [PMID: 36152728 DOI: 10.1016/j.neuro.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Over the last decade, several clinical reports have outlined cases of early-onset manganese (Mn)-induced dystonia-parkinsonism, resulting from loss of function mutations of the Mn transporter gene SLC39A14. Previously, we have performed characterization of the behavioral, neurochemical, and neuropathological changes in 60-day old (PN60) Slc39a14-knockout (KO) murine model of the human disease. Here, we extend our studies to aging Slc39a14-KO mice to assess the progression of the disease. Our results indicate that 365-day old (PN365) Slc39a14-KO mice present with markedly elevated blood and brain Mn levels, similar to those found in the PN60 mice and representative of the human cases of the disease. Furthermore, aging Slc39a14-KO mice consistently manifest a hypoactive and dystonic behavioral deficits, similar to the PN60 animals, suggesting that the behavioral changes are established early in life without further age-associated deterioration. Neurochemical, neuropathological, and functional assessment of the dopaminergic system of the basal ganglia revealed absence of neurodegenerative changes of dopamine (DA) neurons in the substantia nigra pars compacta (SNc), with no changes in DA or metabolite concentrations in the striatum of Slc39a14-KO mice relative to wildtype (WT). Similar to the PN60 animals, aging Slc39a14-KO mice expressed a marked inhibition of potassium-stimulated DA release in the striatum. Together our findings indicate that the pathophysiological changes observed in the basal ganglia of aging Slc39a14-KO animals are similar to those at PN60 and aging does not have a significant effect on these parameters.
Collapse
Affiliation(s)
- Alexander N Rodichkin
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States.
| | - Melissa K Edler
- Department of Anthropology and Brain Health Research Institute, Kent State University, Kent, OH 44242, United States.
| | - Jennifer L McGlothan
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States.
| | - Tomás R Guilarte
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States.
| |
Collapse
|
27
|
Zhu QF, Lu LL, Fang YY, Wu J, Huang ZY, Zheng XW, Song HX, Aschner M, Song C, Jiang YM. Methylcyclopentadienyl Manganese Tricarbonyl Alter Behavior and Cause Ultrastructural Changes in the Substantia Nigra of Rats: Comparison with Inorganic Manganese Chloride. Neurochem Res 2022; 47:2198-2210. [PMID: 35513760 DOI: 10.1007/s11064-022-03606-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/23/2022] [Accepted: 04/10/2022] [Indexed: 10/18/2022]
Abstract
The antiknock additive methylcyclopentadienyl manganese tricarbonyl (MMT) is an organic manganese(Mn) compound. Mn neurotoxicity caused by occupational Mn exposure (mostly inorganic MnCl2) is associated with motor and cognitive disturbances, referred to as Manganism. However, the impact of environmentally relevant Mn exposure on MMT-induced Manganism is poorly understood. In this investigation, we studied the effects of MMT on motor function and brain structure, and compared its effects with those of inorganic MnCl2. After adaptive feeding for 7 days, male and female Sprague-Dawley (SD) rats in the MMT-treated groups and positive control group were treated for 8 weeks with MMT (1, 2 and 4 mg/kg/i.g.) or MnCl2·4H2O (200 mg/kg/i.g.). Mn content in blood, liver, spleen and distinct brain regions was determined by inductively coupled plasma-mass spectrometer (ICP-MS). We found that MMT and MnCl2 exposure led to slower body-weight-gain in female rats, impaired motor and balance function and spatial learning and memory both in male and female rats. HE staining showed that MMT and MnCl2 led to altered structure of the substantia nigra pars compacta (SNpc), and Nissl staining corroborated MMT's propensity to damage the SNpc both in male and female rat. In addition, Immunostaining of the SNpc showed decreased TH-positive neurons in MMT- and MnCl2-treated rats, concomitant with Iba1 activation in microglia. Moreover, no statistically significant difference was noted between the rats in the H-MMT and MnCl2 groups. In summary, these findings suggest that MMT and MnCl2 exposure cause ultrastructural changes in the SNpc neurons culminating in altered motor behavior and cognition, suggesting that altered SNpc structure and function may underline the motor and cognitive deficits inherent to Manganism, and accounting for MMT and MnCl2's manifestations of atypical parkinsonism.
Collapse
Affiliation(s)
- Qi-Feng Zhu
- Department of Neurology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530011, China
- Department of Centre for Translational Medical Research in Integrative Chinese and Western Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, No. 10, Hua-dong Rd., Nanning, 530011, Guangxi, China
| | - Li-Li Lu
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Yuan-Yuan Fang
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Jie Wu
- Department of Cardiology, Institute of Cardiovascular Diseases, the First Institute of Clinical Medicine Guangxi Medical University, Nanning, 530021, China
| | - Zhao-Ying Huang
- Department of Neurology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530011, China
| | - Xiao-Wei Zheng
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Han-Xiao Song
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ce Song
- Department of Centre for Translational Medical Research in Integrative Chinese and Western Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, No. 10, Hua-dong Rd., Nanning, 530011, Guangxi, China.
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| |
Collapse
|
28
|
Kim H, Harrison FE, Aschner M, Bowman AB. Exposing the role of metals in neurological disorders: a focus on manganese. Trends Mol Med 2022; 28:555-568. [PMID: 35610122 PMCID: PMC9233117 DOI: 10.1016/j.molmed.2022.04.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/14/2022] [Accepted: 04/25/2022] [Indexed: 02/06/2023]
Abstract
Metals are ubiquitous chemical entities involved in a myriad of biological processes. Despite their integral role in sustaining life, overexposure can lead to deleterious neurological outcomes posing a public health concern. Excess exposure to metals has been associated with aberrant neurodevelopmental and neurodegenerative diseases and prominently contributes to environmental risk for neurological disorders. Here, we use manganese (Mn) to exemplify the gap in our understanding of the mechanisms behind acute metal toxicity and their relationship to chronic toxicity and disease. This challenge frustrates understanding of how individual exposure histories translate into preventing and treating brain diseases from childhood through old age. We discuss ways to enhance the predictive value of preclinical models and define mechanisms of chronic, persistent, and latent neurotoxicity.
Collapse
Affiliation(s)
- Hyunjin Kim
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Fiona E Harrison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
29
|
Freydenzon A, Nabais MF, Lin T, Williams KL, Wallace L, Henders AK, Blair IP, Wray NR, Pamphlett R, McRae AF. Association between DNA methylation variability and self-reported exposure to heavy metals. Sci Rep 2022; 12:10582. [PMID: 35732753 PMCID: PMC9217962 DOI: 10.1038/s41598-022-13892-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 05/30/2022] [Indexed: 11/30/2022] Open
Abstract
Individuals encounter varying environmental exposures throughout their lifetimes. Some exposures such as smoking are readily observed and have high personal recall; others are more indirect or sporadic and might only be inferred from long occupational histories or lifestyles. We evaluated the utility of using lifetime-long self-reported exposures for identifying differential methylation in an amyotrophic lateral sclerosis cases-control cohort of 855 individuals. Individuals submitted paper-based surveys on exposure and occupational histories as well as whole blood samples. Genome-wide DNA methylation levels were quantified using the Illumina Infinium Human Methylation450 array. We analyzed 15 environmental exposures using the OSCA software linear and MOA models, where we regressed exposures individually by methylation adjusted for batch effects and disease status as well as predicted scores for age, sex, cell count, and smoking status. We also regressed on the first principal components on clustered environmental exposures to detect DNA methylation changes associated with a more generalised definition of environmental exposure. Five DNA methylation probes across three environmental exposures (cadmium, mercury and metalwork) were significantly associated using the MOA models and seven through the linear models, with one additionally across a principal component representing chemical exposures. Methylome-wide significance for four of these markers was driven by extreme hyper/hypo-methylation in small numbers of individuals. The results indicate the potential for using self-reported exposure histories in detecting DNA methylation changes in response to the environment, but also highlight the confounded nature of environmental exposure in cohort studies.
Collapse
Affiliation(s)
- Anna Freydenzon
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Marta F Nabais
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.,University of Exeter Medical School, Exeter, EX2 5DW, Devon, UK
| | - Tian Lin
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Kelly L Williams
- Centre for Motor Neuron Disease Research, Macquarie University, Exeter, NSW, 2109, Australia
| | - Leanne Wallace
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Anjali K Henders
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Ian P Blair
- Centre for Motor Neuron Disease Research, Macquarie University, Exeter, NSW, 2109, Australia
| | - Naomi R Wray
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Roger Pamphlett
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Allan F McRae
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
30
|
Tuschl K, White RJ, Trivedi C, Valdivia LE, Niklaus S, Bianco IH, Dadswell C, González-Méndez R, Sealy IM, Neuhauss SCF, Houart C, Rihel J, Wilson SW, Busch-Nentwich EM. Loss of slc39a14 causes simultaneous manganese hypersensitivity and deficiency in zebrafish. Dis Model Mech 2022; 15:dmm044594. [PMID: 35514229 PMCID: PMC9227717 DOI: 10.1242/dmm.044594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/25/2022] [Indexed: 12/15/2022] Open
Abstract
Manganese neurotoxicity is a hallmark of hypermanganesemia with dystonia 2, an inherited manganese transporter defect caused by mutations in SLC39A14. To identify novel potential targets of manganese neurotoxicity, we performed transcriptome analysis of slc39a14-/- mutant zebrafish that were exposed to MnCl2. Differentially expressed genes mapped to the central nervous system and eye, and pathway analysis suggested that Ca2+ dyshomeostasis and activation of the unfolded protein response are key features of manganese neurotoxicity. Consistent with this interpretation, MnCl2 exposure led to decreased whole-animal Ca2+ levels, locomotor defects and changes in neuronal activity within the telencephalon and optic tectum. In accordance with reduced tectal activity, slc39a14-/- zebrafish showed changes in visual phototransduction gene expression, absence of visual background adaptation and a diminished optokinetic reflex. Finally, numerous differentially expressed genes in mutant larvae normalised upon MnCl2 treatment indicating that, in addition to neurotoxicity, manganese deficiency is present either subcellularly or in specific cells or tissues. Overall, we assembled a comprehensive set of genes that mediate manganese-systemic responses and found a highly correlated and modulated network associated with Ca2+ dyshomeostasis and cellular stress. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Karin Tuschl
- UCL GOS Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
- Department of Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, IoPPN, Kings College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Richard J. White
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London E1 4NS, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Chintan Trivedi
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Leonardo E. Valdivia
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Camino La Pirámide 5750, Huechuraba 8580745, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Camino La Pirámide 5750, Huechuraba 8580745, Chile
| | - Stephanie Niklaus
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Isaac H. Bianco
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Chris Dadswell
- School of Life Sciences, University of Sussex, Brighton BN1 9QJ, UK
| | | | - Ian M. Sealy
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London E1 4NS, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Stephan C. F. Neuhauss
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Corinne Houart
- Department of Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, IoPPN, Kings College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Stephen W. Wilson
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Elisabeth M. Busch-Nentwich
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London E1 4NS, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| |
Collapse
|
31
|
Prado-Rico JM, Lee EY, Wang EW, Yanosky JD, Kong L, Chen H, Navas-Acien A, Du G, Lewis MM, Mailman RB, Huang X. Higher R2* in the Red Nucleus Is Associated With Lead Exposure in an Asymptomatic Welder Cohort. Toxicol Sci 2022; 187:345-354. [PMID: 35357496 PMCID: PMC9154244 DOI: 10.1093/toxsci/kfac035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Lead is a nonessential metal and may be a coexposure in welding fumes. Preclinical data indicate lead may affect iron regulation. The current study investigated blood lead concentrations and their association with brain iron accumulation in workers with chronic welding fume exposure, with a focus on iron-rich subcortical regions of the cerebellum and basal ganglia. Occupational exposure, whole blood metal, and brain MRI data were obtained from 29 controls and 42 welders. R2* (1/T2*) and R1 (T1 relaxation rate) values were used to estimate brain iron and manganese content, respectively. Blood metals and brain R2* (in the red nucleus [RN], dentate nucleus, caudate, putamen, globus pallidus, and substantia nigra) were compared between groups. Associations between brain R2* values and exposure metrics were tested within each group, and analyses were adjusted for potential confounders. Welders had significantly higher levels of whole blood lead, manganese, iron, and copper. Welders also had higher R2* RN (p = .002), but not R1. A 2nd-order polynomial modeled the association between R2* RN and a long-term welding exposure metric. In welders, but not controls, R2* RN was associated positively with whole blood lead (r = 0.54, p = .003), and negatively with whole blood manganese (r = -0.43, p = .02). Higher blood Pb and lower blood Mn independently accounted for variance in high RN R2*. Together, these data suggest that higher RN R2* values may mark lead exposure in welders. Because lead is a known neurotoxicant, additional studies are warranted to confirm this finding, and ascertain its scientific and public/occupational health implications.
Collapse
Affiliation(s)
- Janina Manzieri Prado-Rico
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
| | - Eun-Young Lee
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
| | - Ernest W Wang
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
| | - Jeff D Yanosky
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
| | - Lan Kong
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
| | - Hairong Chen
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York City, New York, USA
| | - Guangwei Du
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
| | - Mechelle M Lewis
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
- Department of Pharmacology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
| | - Richard B Mailman
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
- Department of Pharmacology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
| | - Xuemei Huang
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
- Department of Pharmacology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
- Department of Radiology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
| |
Collapse
|
32
|
Schmidt CW. Hemispheres of Influence: Bridging the Disconnect between Environmental Neurotoxicology and Clinical Practice. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:52001. [PMID: 35543742 PMCID: PMC9093734 DOI: 10.1289/ehp9013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 02/23/2021] [Indexed: 06/14/2023]
|
33
|
Manganese Intoxication Recovery and the Expression Changes of Park2/Parkin in Rats. Neurochem Res 2021; 47:897-906. [PMID: 34839452 DOI: 10.1007/s11064-021-03493-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/18/2021] [Accepted: 11/17/2021] [Indexed: 10/19/2022]
Abstract
Occupational overexposure to manganese (Mn) produces Parkinson's disease-like manganism. Acute Mn intoxication in rats causes dopaminergic neuron loss, impairment of motor activity and reduction of the expression of Park2/Parkin. The expression of Park2/Parkin is also reduced. Whether these changes are reversible after cessation of Mn exposure is unknown, and is the goal of this investigation. Adult male rats were injected with Mn2+ at doses 1 mg/kg and 5 mg/kg in the form of MnCl2·4H2O, every other day for one-month to produce acute Mn neurotoxicity. For a half of rats Mn exposure was suspended for recovery for up to 5 months. Mn neurotoxicity was evaluated by the accumulation of Mn in blood and brain, behavioral activities, dopaminergic neuron loss, and the expression of Park2/Parkin in the blood cells and brain. Dose-dependent Mn neurotoxicity in rats was evidenced by Mn accumulation, rotarod impairments, reduction of tyrosine hydroxylase (TH)-positive neurons in the substantia nigra, decreased level of Park2 mRNA in the blood and brain, and decreased Parkin protein in the brain. After cessation of Mn exposure, the amount of Park2 mRNA in the blood started to increase one month after the recovery. After 5-month of recovery, blood and brain Mn returned to normal, rotarod activity recovered, the reduction of TH-positive dopaminergic neurons ameliorated, and the level of Park2 mRNA in the blood and Park2/Parkin in the midbrain and striatum were returned to the normal. Mn neurotoxicity in rats is reversible after cessation of Mn exposure. The level of Park2 mRNA in the blood could be used as a novel biomarker for Mn exposure and recovery.
Collapse
|
34
|
Budinger D, Barral S, Soo AKS, Kurian MA. The role of manganese dysregulation in neurological disease: emerging evidence. Lancet Neurol 2021; 20:956-968. [PMID: 34687639 DOI: 10.1016/s1474-4422(21)00238-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022]
Abstract
Manganese is an essential trace metal. The dysregulation of manganese seen in a broad spectrum of neurological disorders reflects its importance in brain development and key neurophysiological processes. Historically, the observation of acquired manganism in miners and people who misuse drugs provided early evidence of brain toxicity related to manganese exposure. The identification of inherited manganese transportopathies, which cause neurodevelopmental and neurodegenerative syndromes, further corroborates the neurotoxic potential of this element. Moreover, manganese dyshomoeostasis is also implicated in Parkinson's disease and other neurodegenerative conditions, such as Alzheimer's disease and Huntington's disease. Ongoing and future research will facilitate the development of better targeted therapeutical strategies than are currently available for manganese-associated neurological disorders.
Collapse
Affiliation(s)
- Dimitri Budinger
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | - Serena Barral
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | - Audrey K S Soo
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London, London, UK; Department of Neurology, Great Ormond Street Hospital, London, UK
| | - Manju A Kurian
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London, London, UK; Department of Neurology, Great Ormond Street Hospital, London, UK.
| |
Collapse
|
35
|
Gibson R, Dalvi SP, Dalvi PS. DJ-1 and Parkinson's disease. BRAIN DISORDERS 2021. [DOI: 10.1016/j.dscb.2021.100020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
36
|
Sharma A, Feng L, Muresanu DF, Sahib S, Tian ZR, Lafuente JV, Buzoianu AD, Castellani RJ, Nozari A, Wiklund L, Sharma HS. Manganese nanoparticles induce blood-brain barrier disruption, cerebral blood flow reduction, edema formation and brain pathology associated with cognitive and motor dysfunctions. PROGRESS IN BRAIN RESEARCH 2021; 265:385-406. [PMID: 34560926 DOI: 10.1016/bs.pbr.2021.06.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Nanoparticles affect blood-brain barrier (BBB) and brain edema formation resulting in sensory-motor dysfunction. Exposure of Mn nanoparticles from industrial sources in humans could target basal ganglia resulting in Parkinson's disease. In present investigation, Mn exposure on brain pathology in a rat model was examined. Rats received Mn nanoparticles (30-40nm size) in a dose of 10 or 20mg/kg, i.p. once daily for 7 days and behavioral dysfunctions on Rota Rod performance, inclined plane angle and grid-walking tests as well as gait performances were examined. In addition, BBB breakdown to Evans blue and radioiodine, brain edema formation and neural injuries were also evaluated. Mn nanoparticles treated rats exhibited cognitive and motor dysfunction on the 8th day. At this time, BBB disruption, reduction in cerebral blood flow (CBF), brain edema formation and brain pathology were most marked in the sensory-motor cortex, hippocampus, caudate putamen, cerebellum and thalamus followed by hypothalamus, pons, medulla and spinal cord. In these brain areas, neuronal injuries using Nissl staining was clearly seen. These effects of Mn nanoparticle are dose dependent. These results are the first to demonstrate that Mn nanoparticles induce selective brain pathology resulting in cognitive and motor dysfunction, not reported earlier.
Collapse
Affiliation(s)
- Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
37
|
Behavioral and neurochemical studies of inherited manganese-induced dystonia-parkinsonism in Slc39a14-knockout mice. Neurobiol Dis 2021; 158:105467. [PMID: 34358615 DOI: 10.1016/j.nbd.2021.105467] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/14/2021] [Accepted: 08/02/2021] [Indexed: 11/21/2022] Open
Abstract
Inherited autosomal recessive mutations of the manganese (Mn) transporter gene SLC39A14 in humans, results in elevated blood and brain Mn concentrations and childhood-onset dystonia-parkinsonism. The pathophysiology of this disease is unknown, but the nigrostriatal dopaminergic system of the basal ganglia has been implicated. Here, we describe pathophysiological studies in Slc39a14-knockout (KO) mice as a preclinical model of dystonia-parkinsonism in SLC39A14 mutation carriers. Blood and brain metal concentrations in Slc39a14-KO mice exhibited a pattern similar to the human disease with highly elevated Mn concentrations. We observed an early-onset backward-walking behavior at postnatal day (PN) 21 which was also noted in PN60 Slc39a14-KO mice as well as dystonia-like movements. Locomotor activity and motor coordination were also impaired in Slc39a14-KO relative to wildtype (WT) mice. From a neurochemical perspective, striatal dopamine (DA) and metabolite concentrations and their ratio in Slc39a14-KO mice did not differ from WT. Striatal tyrosine hydroxylase (TH) immunohistochemistry did not change in Slc39a14-KO mice relative to WT. Unbiased stereological cell quantification of TH-positive and Nissl-stained estimated neuron number, neuron density, and soma volume in the substantia nigra pars compacta (SNc) was the same in Slc39a14-KO mice as in WT. However, we measured a marked inhibition (85-90%) of potassium-stimulated DA release in the striatum of Slc39a14-KO mice relative to WT. Our findings indicate that the dystonia-parkinsonism observed in this genetic animal model of the human disease is associated with a dysfunctional but structurally intact nigrostriatal dopaminergic system. The presynaptic deficit in DA release is unlikely to explain the totality of the behavioral phenotype and points to the involvement of other neuronal systems and brain regions in the pathophysiology of the disease.
Collapse
|
38
|
Chopra D, Sharma S, Sharma N, Nehru B. N-Acetylcysteine Ameliorates Neurotoxic Effects of Manganese Intoxication in Rats: A Biochemical and Behavioral Study. Neurochem Res 2021; 46:1953-1969. [PMID: 33950473 DOI: 10.1007/s11064-021-03312-2] [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: 09/24/2020] [Revised: 03/16/2021] [Accepted: 03/24/2021] [Indexed: 11/26/2022]
Abstract
Clinical and experimental evidences reveal that excess exposure to manganese is neurotoxic and leads to cellular damage. However, the mechanism underlying manganese neurotoxicity remains poorly understood but oxidative stress has been implicated to be one of the key pathophysiological features related to it. The present study investigates the effects associated with manganese induced toxicity in rats and further to combat these alterations with a well-known antioxidant N-acetylcysteine which is being used in mitigating the damage by its radical scavenging activity. The study was designed to note the sequential changes along with the motor and memory dysfunction associated with biochemical and histo-pathological alterations following exposure and treatment for 2 weeks. The results so obtained showed decrease in the body weights, behavioral deficits with increased stress markers and also neuronal degeneration in histo-pathological examination after manganese intoxication in rats. To overcome the neurotoxic effects of manganese, N-acetylcysteine was used in the current study due to its pleiotropic potential in several pathological ailments. Taken together, N-acetylcysteine helped in ameliorating manganese induced neurotoxic effects by diminishing the behavioral deficits, normalizing acetylcholinesterase activity, and augmentation of redox status.
Collapse
Affiliation(s)
- Devika Chopra
- Department of Biophysics, Basic Medical Sciences Block II, Panjab University, Chandigarh, 160014, India
| | - Sheetal Sharma
- Department of Biophysics, Basic Medical Sciences Block II, Panjab University, Chandigarh, 160014, India
| | - Neha Sharma
- Department of Biophysics, Basic Medical Sciences Block II, Panjab University, Chandigarh, 160014, India
| | - Bimla Nehru
- Department of Biophysics, Basic Medical Sciences Block II, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
39
|
Tarnacka B, Jopowicz A, Maślińska M. Copper, Iron, and Manganese Toxicity in Neuropsychiatric Conditions. Int J Mol Sci 2021; 22:ijms22157820. [PMID: 34360586 PMCID: PMC8346158 DOI: 10.3390/ijms22157820] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 12/18/2022] Open
Abstract
Copper, manganese, and iron are vital elements required for the appropriate development and the general preservation of good health. Additionally, these essential metals play key roles in ensuring proper brain development and function. They also play vital roles in the central nervous system as significant cofactors for several enzymes, including the antioxidant enzyme superoxide dismutase (SOD) and other enzymes that take part in the creation and breakdown of neurotransmitters in the brain. An imbalance in the levels of these metals weakens the structural, regulatory, and catalytic roles of different enzymes, proteins, receptors, and transporters and is known to provoke the development of various neurological conditions through different mechanisms, such as via induction of oxidative stress, increased α-synuclein aggregation and fibril formation, and stimulation of microglial cells, thus resulting in inflammation and reduced production of metalloproteins. In the present review, the authors focus on neurological disorders with psychiatric signs associated with copper, iron, and manganese excess and the diagnosis and potential treatment of such disorders. In our review, we described diseases related to these metals, such as aceruloplasminaemia, neuroferritinopathy, pantothenate kinase-associated neurodegeneration (PKAN) and other very rare classical NBIA forms, manganism, attention-deficit/hyperactivity disorder (ADHD), ephedrone encephalopathy, HMNDYT1-SLC30A10 deficiency (HMNDYT1), HMNDYT2-SLC39A14 deficiency, CDG2N-SLC39A8 deficiency, hepatic encephalopathy, prion disease and “prion-like disease”, amyotrophic lateral sclerosis, Huntington’s disease, Friedreich’s ataxia, and depression.
Collapse
Affiliation(s)
- Beata Tarnacka
- Department of Rehabilitation Medicine, Faculty of Medicine, Warsaw Medical University, Spartańska 1, 02-637 Warsaw, Poland
- Correspondence: ; Tel.: +48-603944804
| | - Anna Jopowicz
- Department of Rehabilitation, Eleonora Reicher National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, 02-637 Warsaw, Poland;
| | - Maria Maślińska
- Department of Early Arthritis, Eleonora Reicher National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, 02-637 Warsaw, Poland;
| |
Collapse
|
40
|
Schächtle MA, Rosshart SP. The Microbiota-Gut-Brain Axis in Health and Disease and Its Implications for Translational Research. Front Cell Neurosci 2021; 15:698172. [PMID: 34335190 PMCID: PMC8321234 DOI: 10.3389/fncel.2021.698172] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/16/2021] [Indexed: 12/22/2022] Open
Abstract
Over the past decades, microbiome research has evolved rapidly and became a hot topic in basic, preclinical and clinical research, for the pharmaceutical industry and for the general public. With the help of new high-throughput sequencing technologies tremendous progress has been made in the characterization of host-microbiota interactions identifying the microbiome as a major factor shaping mammalian physiology. This development also led to the discovery of the gut-brain axis as the crucial connection between gut microbiota and the nervous system. Consequently, a rapidly growing body of evidence emerged suggesting that the commensal gut microbiota plays a vital role in brain physiology. Moreover, it became evident that the communication along this microbiota-gut-brain axis is bidirectional and primarily mediated by biologically active microbial molecules and metabolites. Further, intestinal dysbiosis leading to changes in the bidirectional relationship between gut microbiota and the nervous system was linked to the pathogenesis of several psychiatric and neurological disorders. Here, we discuss the impact of the gut microbiota on the brain in health and disease, specifically as regards to neuronal homeostasis, development and normal aging as well as their role in neurological diseases of the highest socioeconomic burden such as Alzheimer's disease and stroke. Subsequently, we utilize Alzheimer's disease and stroke to examine the translational research value of current mouse models in the spotlight of microbiome research. Finally, we propose future strategies on how we could conduct translational microbiome research in the field of neuroscience that may lead to the identification of novel treatments for human diseases.
Collapse
Affiliation(s)
- Melanie Anna Schächtle
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stephan Patrick Rosshart
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
41
|
Kaviani S, Shahab S, Sheikhi M, Khaleghian M, Al Saud S. Characterization of the binding affinity between some anti-Parkinson agents and Mn2+, Fe3+ and Zn2+ metal ions: A DFT insight. INORG CHEM COMMUN 2021. [DOI: 10.1016/j.inoche.2021.108582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
42
|
Kulkarni N, Gadde R, Gugnani KS, Vu N, Yoo C, Zaveri R, Betharia S. Neuroprotective effects of disubstituted dithiolethione ACDT against manganese-induced toxicity in SH-SY5Y cells. Neurochem Int 2021; 147:105052. [PMID: 33905764 DOI: 10.1016/j.neuint.2021.105052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/05/2021] [Accepted: 04/21/2021] [Indexed: 11/17/2022]
Abstract
Dithiolethiones are lipophilic, organosulfur compounds that activate the Nrf2 transcription factor causing an upregulation of various phase II antioxidant enzymes. A disubstituted dithiolethione 5-amino-3-thioxo-3H-(1,2) dithiole-4-carboxylic acid ethyl ester (ACDT) retains the functional pharmacophore while also containing modifiable functional groups. Neuroprotection against autoimmune encephalomyelitis in vivo and 6-hydroxy dopamine (a model for Parkinson's disease) in vitro have been previously reported with ACDT. Manganese (Mn) is a metal essential for metabolic processes at low concentrations. Overexposure and accumulation of Mn leads to a neurological condition called manganism which shares pathophysiological sequelae with parkinsonism. Here we hypothesized ACDT to be protective against manganese-induced cytotoxicity. SH-SY5Y human neuroblastoma cells exposed to 300 μM MnCl2 displayed approximately 50% cell death, and a 24-h pretreatment with 75 μM ACDT significantly reversed this cytotoxicity. ACDT pretreatment was also found to increase total GSH levels (2.18-fold) and the protein levels of NADPH:quinone oxidoreductase-1 (NQO1) enzyme (6.33-fold), indicating an overall increase in the cells' antioxidant defense stores. A corresponding 2.32-fold reduction in the level of Mn-induced reactive oxygen species was also observed in cells pretreated with ACDT. While no changes were observed in the protein levels of apoptotic markers Bax and Bcl-2, pretreatment with 75 μM ACDT led to a 2.09-fold downregulation of ZIP14 import transporter, indicating a potential reduction in the cellular uptake of Mn as an additional neuroprotective mechanism. These effects did not extend to other transporters like the divalent metal transporter 1 (DMT1) or ferroportin. Collectively, ACDT showed substantial neuroprotection against Mn-induced cytotoxicity, opening a path for dithiolethiones as a potential novel therapeutic option against heavy metal neurotoxicity.
Collapse
Affiliation(s)
- Neha Kulkarni
- Department of Pharmaceutical Sciences, MCPHS University, School of Pharmacy, 179 Longwood Avenue, Boston, MA, 02115, USA.
| | - Rajitha Gadde
- Department of Pharmaceutical Sciences, MCPHS University, School of Pharmacy, 179 Longwood Avenue, Boston, MA, 02115, USA
| | - Kuljeet S Gugnani
- Department of Pharmaceutical Sciences, MCPHS University, School of Pharmacy, 179 Longwood Avenue, Boston, MA, 02115, USA
| | - Nguyen Vu
- Department of Pharmaceutical Sciences, MCPHS University, School of Pharmacy, 179 Longwood Avenue, Boston, MA, 02115, USA
| | - Claude Yoo
- Department of Pharmaceutical Sciences, MCPHS University, School of Pharmacy, 179 Longwood Avenue, Boston, MA, 02115, USA
| | - Rohan Zaveri
- Department of Pharmaceutical Sciences, MCPHS University, School of Pharmacy, 179 Longwood Avenue, Boston, MA, 02115, USA
| | - Swati Betharia
- Department of Pharmaceutical Sciences, MCPHS University, School of Pharmacy, 179 Longwood Avenue, Boston, MA, 02115, USA
| |
Collapse
|
43
|
Lee EY, Flynn MR, Du G, Lewis MM, Goldenberg M, Kong L, Mailman RB, Hong YS, Huang X. Nigral MRI features of asymptomatic welders. Parkinsonism Relat Disord 2021; 85:37-43. [PMID: 33691274 DOI: 10.1016/j.parkreldis.2021.02.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 02/05/2021] [Accepted: 02/14/2021] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Manganese (Mn)-induced parkinsonism involves motor symptoms similar to those observed in Parkinson's disease (PD). Previous literature suggests that chronic Mn- exposure may increase PD risk, although Mn-induced clinical syndromes are considered atypical for PD. This study investigated whether asymptomatic welders display differences in the substantia nigra (SN), the key pathological locus of PD. METHOD Brain MRI data and occupational exposure history were obtained in welders (N = 43) and matched controls (N = 31). Diffusion tensor imaging fractional anisotropy (FA; estimate of microstructural integrity) and R2* (estimate of iron and other PD-related brain differences) values in the SN pars compacta (SNc), SN reticulata (SNr), and globus pallidus (GP) were compared between the two groups. The MRI markers of the SN and GP within welders were related to exposure estimates. RESULTS Compared to controls, welders who had chronic, but low-level, Mn-exposure had similar FA and R2* values in both SN regions (p's > 0.082), but significantly lower FA (p = 0.0013), although not R2* (p = 0.553), in the GP. In welders, FA values in the SN and GP showed a second-order polynomial relationship with cumulative lifetime welding exposure (p's < 0.03). CONCLUSION Neurotoxic processes associated with Mn-exposure may be different from those in PD when the exposure-level is relatively low. Greater welding duration and level, however, were associated with FA differences in the GP and SN, indicating that welding exposures above a certain level may induce neurotoxicity in the SN, a finding that should be explored further in future studies.
Collapse
Affiliation(s)
- Eun-Young Lee
- Department of Health Care and Science, Dong-A University, Busan, South Korea.
| | - Michael R Flynn
- Department of Kinesiology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Environmental Sciences, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Guangwei Du
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Mechelle M Lewis
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Pharmacology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Michael Goldenberg
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Lan Kong
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Richard B Mailman
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Pharmacology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Young-Seoub Hong
- Department of Preventive Medicine, Dong-A University College of Medicine, Busan, South Korea.
| | - Xuemei Huang
- Department of Neurology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Pharmacology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Radiology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Neurosurgery, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Kinesiology, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, 17033, USA; Department of Environmental Sciences, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
44
|
Imataka G, Yoshihara S. A Case Report of Non-Herpetic Limbic Encephalitis with Psychological Symptoms and Parkinsonism. IRANIAN JOURNAL OF PUBLIC HEALTH 2021; 50:203-204. [PMID: 34178781 PMCID: PMC8213637 DOI: 10.18502/ijph.v50i1.5089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- George Imataka
- Department of Pediatrics, Dokkyo Medical University, Tochigi, Japan
| | | |
Collapse
|
45
|
Zhu X, Yang L, He Y, Sun Y, Shi W, Ou C. Liver Function of Male Rats Exposed to Manganese at Different Time Points. Biol Trace Elem Res 2020; 198:224-230. [PMID: 32100273 DOI: 10.1007/s12011-020-02067-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 01/30/2020] [Indexed: 01/15/2023]
Abstract
As an essential trace element in the human body, manganese (Mn) is involved in many important biochemical reactions. However, excessive exposure to manganese can cause multiple systematic damages to the body. This study aims to investigate the effects of manganese exposure on serum hepatic enzymes in male rats at different time points. After adaptive feeding for 7 days, male Sprague-Dawley (SD) rats were injected intraperitoneally with 30 mg/kg MnCl2·4H2O once a day for 21 days at zeitgeber time point 2 (ZT2), ZT8, ZT14, and ZT20, respectively. We found that short-term repeated exposure to manganese caused slower body weight gain and increased relative liver and spleen weight index in male rats at different time points. Moreover, serum total bile acid (TBA) increased while aspartate aminotransferase (AST) decreased at ZT2, ZT8, and ZT20. Cholinesterase (ChE) decreased at ZT2 and ZT20, lactic dehydrogenase (LDH) decreased at ZT2, ZT14, and ZT20, and acid phosphatase (ACP) decreased at ZT2 and ZT14. Alkaline phosphatase (ALP) decreased at ZT2, ZT14, and ZT20, but increased at ZT8. Alanine amino transferase (ALT) decreased at ZT2 and ZT20, but increased at ZT8. There was a negative correlation between relative liver weight index with AST, ACP, ALP, and LDH, while a positive correlation with TBA. However, relative spleen weight index had a positive correlation with relative liver weight index and TBA, while a negative correlation with ALT, AST, ACP, ALP, LDH, and ChE. Our study shows that the injury of liver function is caused by short-term repeated manganese exposure at different time points. The time effect should be considered in manganese toxicity evaluation.
Collapse
Affiliation(s)
- Xiaonian Zhu
- Department of Health Toxicology, School of Public Health, Guilin Medical University, 109 Huancheng North Road 2, Guilin, Guangxi, 541004, People's Republic of China
| | - Lin Yang
- Department of Health Toxicology, School of Public Health, Guilin Medical University, 109 Huancheng North Road 2, Guilin, Guangxi, 541004, People's Republic of China
| | - Yonghua He
- Department of Health Toxicology, School of Public Health, Guilin Medical University, 109 Huancheng North Road 2, Guilin, Guangxi, 541004, People's Republic of China
| | - Yi Sun
- Department of Health Toxicology, School of Public Health, Guilin Medical University, 109 Huancheng North Road 2, Guilin, Guangxi, 541004, People's Republic of China
| | - Wenxiang Shi
- Department of Health Toxicology, School of Public Health, Guilin Medical University, 109 Huancheng North Road 2, Guilin, Guangxi, 541004, People's Republic of China
| | - Chaoyan Ou
- Department of Health Toxicology, School of Public Health, Guilin Medical University, 109 Huancheng North Road 2, Guilin, Guangxi, 541004, People's Republic of China.
| |
Collapse
|
46
|
Martin KV, Edmondson D, Cecil KM, Bezi C, Vance ML, McBride D, Haynes EN. Manganese Exposure and Neurologic Outcomes in Adult Populations. Neurol Clin 2020; 38:913-936. [PMID: 33040869 PMCID: PMC8978550 DOI: 10.1016/j.ncl.2020.07.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A review of published articles examining the effects of manganese exposure to workers and community residents shows adverse neurologic outcomes. Innovative biomarkers, including those from neuroimaging, were incorporated into many of these studies to assess both manganese exposure and neurologic outcomes. A variety of health effects were evaluated, including cognitive and motor impairments. Studies of community participants residing near manganese point sources show variability in outcomes, reflecting the complexities of exposure measurement, individual absorption, and assessment of neurologic effects. The aging population provides insight into the impacts of chronic exposure in younger populations.
Collapse
Affiliation(s)
- Kaitlin V Martin
- Department of Epidemiology, College of Public Health, University of Kentucky, 111 Washington Avenue Room 212C, Lexington, KY 40536, USA.
| | - David Edmondson
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Imaging Research Center, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 5033, Cincinnati, OH 45229, USA
| | - Kim M Cecil
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Imaging Research Center, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 5033, Cincinnati, OH 45229, USA; Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Cassandra Bezi
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7017, Cincinnati, OH 45229, USA
| | - Miriam Leahshea Vance
- Department of Epidemiology, College of Public Health, University of Kentucky, 111 Washington Avenue, Lexington, KY 40536, USA
| | - Dani McBride
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Erin N Haynes
- Department of Epidemiology, College of Public Health, University of Kentucky, 111 Washington Avenue Room 212G, Lexington, KY 40536, USA
| |
Collapse
|
47
|
Pingale T, Gupta GL. Classic and evolving animal models in Parkinson's disease. Pharmacol Biochem Behav 2020; 199:173060. [PMID: 33091373 DOI: 10.1016/j.pbb.2020.173060] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with motor and non-motor symptoms. PD is characterized by the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and deficiency of dopamine in the striatal region. The primary objective in PD research is to understand the pathogenesis, targets, and development of therapeutic interventions to control the progress of the disease. The anatomical and physiological resemblances between humans and animals gathered the researcher's attention towards the use of animals in PD research. Due to varying age of onset, symptoms, and progression rate, PD becomes heterogeneous which demands the variety of animal models to study diverse features of the disease. Parkinson is a multifactorial disorder, selection of models become important as not a single model shows all the biochemical features of the disease. Currently, conventional pharmacological, neurotoxin-induced, genetically modified and cellular models are available for PD research, but none of them recapitulate all the biochemical characteristics of the disease. In this review, we included the updated knowledge on the main features of currently available in vivo and in vitro models as well as their strengths and weaknesses.
Collapse
Affiliation(s)
- Tanvi Pingale
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, India
| | - Girdhari Lal Gupta
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, India; School of Pharmacy & Technology Management, SVKM'S NMIMS, Shirpur, Maharashtra, India.
| |
Collapse
|
48
|
Edmondson DA, Yeh CL, Hélie S, Dydak U. Whole-brain R1 predicts manganese exposure and biological effects in welders. Arch Toxicol 2020; 94:3409-3420. [PMID: 32875357 DOI: 10.1007/s00204-020-02839-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 07/09/2020] [Indexed: 12/25/2022]
Abstract
Manganese (Mn) is a neurotoxicant that, due to its paramagnetic property, also functions as a magnetic resonance imaging (MRI) T1 contrast agent. Previous studies in Mn toxicity have shown that Mn accumulates in the brain, which may lead to parkinsonian symptoms. In this article, we trained support vector machines (SVM) using whole-brain R1 (R1 = 1/T1) maps from 57 welders and 32 controls to classify subjects based on their air Mn concentration ([Mn]Air), Mn brain accumulation (ExMnBrain), gross motor dysfunction (UPDRS), thalamic GABA concentration (GABAThal), and total years welding. R1 was highly predictive of [Mn]Air above a threshold of 0.20 mg/m3 with an accuracy of 88.8% and recall of 88.9%. R1 was also predictive of subjects with GABAThal having less than or equal to 2.6 mM with an accuracy of 82% and recall of 78.9%. Finally, we used an SVM to predict age as a method of verifying that the results could be attributed to Mn exposure. We found that R1 was predictive of age below 48 years of age with accuracies ranging between 75 and 82% with recall between 94.7% and 76.9% but was not predictive above 48 years of age. Together, this suggests that lower levels of exposure (< 0.20 mg/m3 and < 18 years of welding on the job) do not produce discernable signatures, whereas higher air exposures and subjects with more total years welding produce signatures in the brain that are readily identifiable using SVM.
Collapse
Affiliation(s)
- David A Edmondson
- School of Health Sciences, Purdue University, 550 Stadium Dr., Hampton Hall of Civil Engineering, West Lafayette, IN, 47907, USA.,Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.,Imaging Research Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Chien-Lin Yeh
- School of Health Sciences, Purdue University, 550 Stadium Dr., Hampton Hall of Civil Engineering, West Lafayette, IN, 47907, USA.,Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sébastien Hélie
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, USA
| | - Ulrike Dydak
- School of Health Sciences, Purdue University, 550 Stadium Dr., Hampton Hall of Civil Engineering, West Lafayette, IN, 47907, USA. .,Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
49
|
Freeman DM, O'Neal R, Zhang Q, Bouwer EJ, Wang Z. Manganese-induced Parkinsonism in mice is reduced using a novel contaminated water sediment exposure model. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 78:103399. [PMID: 32380377 DOI: 10.1016/j.etap.2020.103399] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 04/15/2020] [Accepted: 04/18/2020] [Indexed: 06/11/2023]
Abstract
Heavy metals enter the aquatic environment and accumulate within water sediments, but these metal-sediment interactions remain to be explored within toxicity studies. We developed an exposure model in mice that encapsulates the aquatic microenvironment of metals before exposure. Male and female C57/BL6 mice were exposed via their drinking water to manganese contaminated sediment (Sed_Mn) or to manganese without sediment interaction (Mn) for six weeks. Sediment interaction did not alter weekly manganese ingestion from water in males or females. We analyzed motor impairment, a common feature in manganese-induced Parkinsonism, using the beam traversal, cylinder, and accelerating rotarod tests. Sed_Mn mice performed better overall compared to Mn mice and males were more sensitive to manganese than females in both Sed_Mn and Mn treatment groups. Our study indicates that metal-sediment interactions may alter metal toxicity in mammals and introduces a new exposure model to test the toxicity of metal contaminants of drinking water.
Collapse
Affiliation(s)
- Dana M Freeman
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Rachel O'Neal
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Qiang Zhang
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Edward J Bouwer
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zhibin Wang
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
50
|
Wang H, Yang F, Xin R, Cui D, He J, Zhang S, Sun Y. The gut microbiota attenuate neuroinflammation in manganese exposure by inhibiting cerebral NLRP3 inflammasome. Biomed Pharmacother 2020; 129:110449. [PMID: 32768944 DOI: 10.1016/j.biopha.2020.110449] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/12/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023] Open
Abstract
Manganese (Mn) exposure has been reported to cause neurodegenerative disorders. β-Amyloid (Aβ) induced Tau pathology in an NLRP3-dependent manner is at the heart of Alzheimer's and Parkinson's diseases. The gut microbiota plays a crucial role in the bidirectional gut-brain axis that integrates the gut and central nervous system (CNS) activities. In this study, we found that Mn exposure increases Aβ1-40 and Tau production in brain, and causes hippocampal degeneration and necrosis. Meanwhile, Mn exposure can stimulate neurotoxicity by increasing inflammation either in peripheral blood and CNS. Importantly, we found that transplantation of gut microbiota from normal rats into Mn exposure rats reduced Aβ and Tau expression, and the cerebral expression of NLRP3 was downregulated, and the expression of neuroinflammatory factors was also downregulated. Therefore, improving the composition of gut microbiota in Mn exposure rats can attenuate neuroinflammation, which is considered as a novel therapeutic strategy for Mn exposure by remodelling the gut microbiota.
Collapse
Affiliation(s)
- Hui Wang
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Feng Yang
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Ruihua Xin
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Dongan Cui
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Jiongjie He
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Shidong Zhang
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
| | - Yan Sun
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
| |
Collapse
|