1
|
Ludwig HD, Carpenter JM, Filipov NM. Behavioral Alterations in Mice Exposed to Manganese via Drinking Water: Effects of Sex and a Lipopolysaccharide Challenge. J Appl Toxicol 2024. [PMID: 39647842 DOI: 10.1002/jat.4739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/12/2024] [Accepted: 11/21/2024] [Indexed: 12/10/2024]
Abstract
Manganese (Mn) is an essential and important metal; however, overexposures lead to adverse neurological outcomes. Nonoccupational Mn overexposure occurs primarily through consumption of Mn-contaminated drinking water (DW). Sex differences in terms of nervous and immune systems' responsiveness to excessive Mn in the DW are understudied. Thus, this study investigated behavioral and sex differences in response to Mn DW treatment (0.4 g Mn/L for up to 8 weeks) and a lipopolysaccharide (LPS) challenge of adult C57BL/6 mice with GFP-tagged monocytes/microglia. After 6 weeks, in motor function tests, Mn exposure resulted in decreased activity and gait deficits. In two different mood tests (open field test [OFT]/elevated zero maze), Mn-exposed mice exhibited decreased fear/anxiety-like behavior. Two weeks after behavioral assessment, when mice were challenged with LPS, circulating inflammatory cytokines, and acute phase proteins increased in both sexes. After 8 weeks of Mn exposure, liver and brain Mn levels were increased, but Mn alone did not affect circulating cytokines in either sex. Notably, Mn-exposed/LPS-challenged males had potentiated plasma cytokine output, whereas the reverse was seen in females. Males, but not females, continued to exhibit increased fearlessness (i.e., increased OFT center time), even when challenged with LPS. Overall, our results show that Mn DW exposure increases brain Mn levels and it leads to behavioral alterations in both sexes. However, males might be more susceptible to the effect of Mn on mood, and this effect is recalcitrant to an inflammagen challenge. Mn augmented post-LPS cytokine production only in males, further indicating that important Mn effects are sex-biased.
Collapse
Affiliation(s)
- Helaina D Ludwig
- Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, USA
| | - Jessica M Carpenter
- Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia, USA
- Neuroscience PhD Program, University of Georgia, Athens, Georgia, USA
| | - Nikolay M Filipov
- Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, USA
- Neuroscience PhD Program, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
2
|
Mohamed Ibrahim N, Lin CH. Early Onset Parkinsonism: Differential diagnosis and what not to miss. Parkinsonism Relat Disord 2024; 129:107100. [PMID: 39183141 DOI: 10.1016/j.parkreldis.2024.107100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/07/2024] [Accepted: 08/10/2024] [Indexed: 08/27/2024]
Abstract
Early Onset Parkinsonism (EOP) refers to parkinsonism occurring before the age of 50 years. The causes are diverse and include secondary and genetic causes. Secondary causes related to medications, inflammatory and infective disorders are mostly treatable and well recognized as they usually present with a relatively more rapid clinical course compared to idiopathic Parkinson's disease. Genetic causes of EOP are more challenging to diagnose especially as more of the non-PARK genes are recognized to present with typical and atypical parkinsonism. Some of the genetic disorders such as Spinocerebellar ataxia 2 (SCA2) and Spinocerebellar ataxia 3 (SCA3) may present with levodopa-responsive parkinsonism, indistinguishable from idiopathic Parkinson's disease. Additionally, some of the genetic disorders, including Wilson's disease and cerebrotendinous xanthomatosis (CTX), are potentially treatable and should not be missed. Due to the advent of next generating sequencing techniques, genetic analyses facilitate early identification and proper treatment of diverse causes of EOP. In this review, we outline the clinical approach of EOP highlighting the key clinical features of some of the non-PARK genetic causes of EOP and related investigations, which could assist in clinical diagnosis. This review also encompass genetic diagnostic approaches, emphasizing the significance of pretest counseling and the principles of bioinformatics analysis strategies.
Collapse
Affiliation(s)
- Norlinah Mohamed Ibrahim
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Chin Hsien Lin
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan; College of Medicine, National Taiwan University, Taipei, Taiwan; Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
3
|
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
|
4
|
Lewis F, Shoieb D, Azmoun S, Colicino E, Jin Y, Chi J, Gu H, Placidi D, Padovani A, Pilotto A, Pepe F, Turla M, Crippa P, Wang X, Lucchini RG. Metabolomic and Lipidomic Analysis of Manganese-Associated Parkinsonism: a Case-Control Study in Brescia, Italy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.04.24313002. [PMID: 39281765 PMCID: PMC11398432 DOI: 10.1101/2024.09.04.24313002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Background and Objectives Excessive Manganese (Mn) exposure is neurotoxic and can cause Mn-Induced Parkinsonism (MnIP), marked by cognitive and motor dysfunction. Although metabolomic and lipidomic research in Parkinsonism (PD) patients exists, it remains limited. This study hypothesizes distinct metabolomic and lipidomic profiles based on exposure status, disease diagnosis, and their interaction. Methods We used a case-control design with a 2×2 factorial framework to investigate the metabolomic and lipidomic alterations associated with Mn exposure and their link to PD. The study population of 97 individuals was divided into four groups: non-exposed controls (n=23), exposed controls (n=25), non-exposed with PD (n=26) and exposed with PD (n=23). Cases, defined by at least two cardinal PD features (excluding vascular, iatrogenic, and traumatic origins), were recruited from movement disorder clinics in four hospitals in Brescia, Northern Italy. Controls, free from neurological or psychiatric conditions, were selected from the same hospitals. Exposed subjects resided in metallurgic regions (Val Camonica and Bagnolo Mella) for at least 8 continuous years, while non-exposed subjects lived in low-exposure areas around Lake Garda and Brescia city. We conducted untargeted analyses of metabolites and lipids in whole blood samples using ultra-high-performance liquid chromatography (UHPLC) and mass spectrometry (MS), followed by statistical analyses including Principal Component Analysis (PCA), Partial Least Squares-Discriminant Analysis (PLS-DA), and Two-Way Analysis of Covariance (ANCOVA). Results Metabolomic analysis revealed modulation of alanine, aspartate, and glutamate metabolism (Impact=0.05, p=0.001) associated with disease effect; butanoate metabolism (Impact=0.03, p=0.004) with the exposure effect; and vitamin B6 metabolism (Impact=0.08, p=0.03) with the interaction effect. Differential relative abundances in 3-sulfoxy-L-Tyrosine (β=1.12, FDR p<0.001), glycocholic acid (β=0.48, FDR p=0.03), and palmitelaidic acid (β=0.30, FDR p<0.001) were linked to disease, exposure, and interaction effects, respectively. In the lipidome, ferroptosis (Pathway Lipids=11, FDR p=0.03) associated with the disease effect and sphingolipid signaling (Pathway Lipids=9, FDR p=0.04) associated with the interaction effect were significantly altered. Lipid classes triacylglycerols, ceramides, and phosphatidylethanolamines showed differential relative abundances associated with disease, exposure, and interaction effects, respectively. Discussion These findings suggest that PD and Mn exposure induce unique metabolomic and lipidomic changes, potentially serving as biomarkers for MnIP and warranting further study.
Collapse
Affiliation(s)
- Freeman Lewis
- Environmental Health Sciences, Florida International University, 11200 SW 8th St, Miami, 33199, Florida, USA
| | - Daniel Shoieb
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Viale Europa 11, Brescia, 25123, Italy
| | - Somaiyeh Azmoun
- Environmental Health Sciences, Florida International University, 11200 SW 8th St, Miami, 33199, Florida, USA
| | - Elena Colicino
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, 17 E 102nd St, New York, 10029, New York, USA
| | - Yan Jin
- Environmental Health Sciences, Florida International University, 11200 SW 8th St, Miami, 33199, Florida, USA
| | - Jinhua Chi
- Environmental Health Sciences, Florida International University, 11200 SW 8th St, Miami, 33199, Florida, USA
| | - Haiwei Gu
- Environmental Health Sciences, Florida International University, 11200 SW 8th St, Miami, 33199, Florida, USA
| | - Donatella Placidi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Viale Europa 11, Brescia, 25123, Italy
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, Brescia, 25123, Italy and Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy
| | - Andrea Pilotto
- Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, Brescia, 25123, Italy and Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy
| | - Fulvio Pepe
- Clinic of Neurology, Poliambulanza Foundation, Brescia, Italy
| | - Marinella Turla
- Clinic of Neurology, Esine Hospital of Valcamonica, Brescia, Italy
| | | | - Xuexia Wang
- Department of Biostatistics, Florida International University, 11200 SW 8th St, Miami, 33199, Florida, USA
| | - Roberto G Lucchini
- Environmental Health Sciences, Florida International University, 11200 SW 8th St, Miami, 33199, Florida, USA
- Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, Via Universitá, 4, Modena, 610101, Italy
| |
Collapse
|
5
|
Pajarillo E, Kim S, Digman A, Ajayi I, Nyarko-Danquah I, Son DS, Aschner M, Lee E. Dopaminergic REST/NRSF is protective against manganese-induced neurotoxicity in mice. J Biol Chem 2024; 300:107707. [PMID: 39178947 PMCID: PMC11421342 DOI: 10.1016/j.jbc.2024.107707] [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: 03/05/2024] [Revised: 07/23/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
Chronic exposure to elevated levels of manganese (Mn) may cause a neurological disorder referred to as manganism. The transcription factor REST is dysregulated in several neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease. REST upregulated tyrosine hydroxylase and induced protection against Mn toxicity in neuronal cultures. In the present study, we investigated if dopaminergic REST plays a critical role in protecting against Mn-induced toxicity in vivo using dopaminergic REST conditional knockout (REST-cKO) mice and REST loxP mice as wild-type (WT) controls. Restoration of REST in the substantia nigra (SN) with neuronal REST AAV vector infusion was performed to further support the role of REST in Mn toxicity. Mice were exposed to Mn (330 μg, intranasal, daily for 3 weeks), followed by behavioral tests and molecular biology experiments. Results showed that Mn decreased REST mRNA/protein levels in the SN-containing midbrain, as well as locomotor activity and motor coordination in WT mice, which were further decreased in REST-cKO mice. Mn-induced mitochondrial insults, such as impairment of fission/fusion and mitophagy, apoptosis, and oxidative stress, in the midbrain of WT mice were more pronounced in REST-cKO mice. However, REST restoration in the SN of REST-cKO mice attenuated Mn-induced neurotoxicity. REST's molecular target for its protection is unclear, but REST attenuated Mn-induced mitochondrial dysregulation, indicating that it is a primary intracellular target for both Mn and REST. These novel findings suggest that dopaminergic REST in the nigrostriatal pathway is critical in protecting against Mn toxicity, underscoring REST as a potential therapeutic target for treating manganism.
Collapse
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Sanghoon Kim
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Alexis Digman
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Itunu Ajayi
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Ivan Nyarko-Danquah
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Deok-Soo Son
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, New York, USA
| | - Eunsook Lee
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA.
| |
Collapse
|
6
|
Zheng XW, Fang YY, Lin JJ, Luo JJ, Li SJ, Aschner M, Jiang YM. Signal Transduction Associated with Mn-induced Neurological Dysfunction. Biol Trace Elem Res 2024; 202:4158-4169. [PMID: 38155332 DOI: 10.1007/s12011-023-03999-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
Manganese (Mn) is a heavy metal that occurs widely in nature and has a vital physiological role in growth and development. However, excessive exposure to Mn can cause neurological damage, especially cognitive dysfunction, such as learning disability and memory loss. Numerous studies on the mechanisms of Mn-induced nervous system damage found that this metal targets a variety of metabolic pathways, for example, endoplasmic reticulum stress, apoptosis, neuroinflammation, cellular signaling pathway changes, and neurotransmitter metabolism interference. This article reviews the latest research progress on multiple signaling pathways related to Mn-induced neurological dysfunction.
Collapse
Affiliation(s)
- Xiao-Wei Zheng
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China
| | - Yuan-Yuan Fang
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China
| | - Jun-Jie Lin
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China
| | - Jing-Jing Luo
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China
| | - Shao-Jun Li
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China.
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China.
| | - Michael Aschner
- The Department of Molecular Pharmacology at Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yue-Ming Jiang
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-Yong Rd., Nanning, 530021, Guangxi, China.
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning, 530021, China.
| |
Collapse
|
7
|
Kang J, Wu X, Li Y, Zhao S, Wang S, Yu D. Association between inflammatory bowel disease and osteoporosis in European and East Asian populations: exploring causality, mediation by nutritional status, and shared genetic architecture. Front Immunol 2024; 15:1425610. [PMID: 39136019 PMCID: PMC11317921 DOI: 10.3389/fimmu.2024.1425610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/24/2024] [Indexed: 08/15/2024] Open
Abstract
Background While previous research has established an association between inflammatory bowel disease (IBD) and osteoporosis (OP), the nature of this association in different populations remains unclear. Objective Our study used linkage disequilibrium scores(LDSC) regression analysis and Mendelian randomization(MR) to assess the genetic correlation and causal relationship between IBD and OP in European and East Asian populations. Methods We performed separate genetic correlation and causal analyses for IBD and OP in European and East Asian populations, used the product of coefficients method to estimate the mediating effect of nutritional status on the causal relationship, and used multi-trait analysis to explore the biological mechanisms underlying the IBD-nutrition-OP causal pathway. Results Our analysis revealed a significant genetic correlation and causal relationship between IBD and OP in the European population. Conversely, no such correlation or causal relationship was observed in the East Asian population. Mediation analysis revealed a significant mediating effect of nutritional status on the causal pathway between IBD and OP in the European population. Multi-trait analysis of the IBD-nutrition-OP causal pathway identified MFAP2, ATP13A2, SERPINA1, FTO and VCAN as deleterious variants. Conclusion Our findings establish a genetic correlation and causal relationship between IBD and OP in the European population, with nutritional status playing a crucial mediating role.
Collapse
Affiliation(s)
- Jian Kang
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xize Wu
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yue Li
- Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Shuangli Zhao
- Orthopedics and Traumatology, The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Shixuan Wang
- Orthopedics and Traumatology, The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Dongdong Yu
- Orthopedics and Traumatology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
8
|
Reis J, Tranchant C, Lucchini RG, Spencer PS. Concerns with the new biological research criteria for synucleinopathy. Lancet Neurol 2024; 23:663. [PMID: 38876740 PMCID: PMC11344483 DOI: 10.1016/s1474-4422(24)00215-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/09/2024] [Indexed: 06/16/2024]
Affiliation(s)
- Jacques Reis
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France; Association RISE, Oberhausbergen 67205, France.
| | - Christine Tranchant
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France
| | - Roberto G Lucchini
- Environmental Health Sciences, Florida International University, Miami, FL, USA; University of Modena and Reggio Emilia, Modena, Italy
| | - Peter S Spencer
- Department of Neurology, School of Medicine, Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
9
|
Ngwa HA, Bargues-Carot A, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Manganese and Vanadium Co-Exposure Induces Severe Neurotoxicity in the Olfactory System: Relevance to Metal-Induced Parkinsonism. Int J Mol Sci 2024; 25:5285. [PMID: 38791326 PMCID: PMC11121436 DOI: 10.3390/ijms25105285] [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: 03/28/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic environmental exposure to toxic heavy metals, which often occurs as a mixture through occupational and industrial sources, has been implicated in various neurological disorders, including Parkinsonism. Vanadium pentoxide (V2O5) typically presents along with manganese (Mn), especially in welding rods and high-capacity batteries, including electric vehicle batteries; however, the neurotoxic effects of vanadium (V) and Mn co-exposure are largely unknown. In this study, we investigated the neurotoxic impact of MnCl2, V2O5, and MnCl2-V2O5 co-exposure in an animal model. C57BL/6 mice were intranasally administered either de-ionized water (vehicle), MnCl2 (252 µg) alone, V2O5 (182 µg) alone, or a mixture of MnCl2 (252 µg) and V2O5 (182 µg) three times a week for up to one month. Following exposure, we performed behavioral, neurochemical, and histological studies. Our results revealed dramatic decreases in olfactory bulb (OB) weight and levels of tyrosine hydroxylase, dopamine, and 3,4-dihydroxyphenylacetic acid in the treatment groups compared to the control group, with the Mn/V co-treatment group producing the most significant changes. Interestingly, increased levels of α-synuclein expression were observed in the substantia nigra (SN) of treated animals. Additionally, treatment groups exhibited locomotor deficits and olfactory dysfunction, with the co-treatment group producing the most severe deficits. The treatment groups exhibited increased levels of the oxidative stress marker 4-hydroxynonenal in the striatum and SN, as well as the upregulation of the pro-apoptotic protein PKCδ and accumulation of glomerular astroglia in the OB. The co-exposure of animals to Mn/V resulted in higher levels of these metals compared to other treatment groups. Taken together, our results suggest that co-exposure to Mn/V can adversely affect the olfactory and nigral systems. These results highlight the possible role of environmental metal mixtures in the etiology of Parkinsonism.
Collapse
Affiliation(s)
- Hilary Afeseh Ngwa
- Iowa Center for Advanced Neurotoxicity, Department of Biomedical Sciences, Iowa State University, Ames, IA 50010, USA
| | - Alejandra Bargues-Carot
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (A.B.-C.); (H.J.); (V.A.)
| | - Huajun Jin
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (A.B.-C.); (H.J.); (V.A.)
| | - Vellareddy Anantharam
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (A.B.-C.); (H.J.); (V.A.)
| | - Arthi Kanthasamy
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (A.B.-C.); (H.J.); (V.A.)
| | - Anumantha G. Kanthasamy
- Iowa Center for Advanced Neurotoxicity, Department of Biomedical Sciences, Iowa State University, Ames, IA 50010, USA
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (A.B.-C.); (H.J.); (V.A.)
| |
Collapse
|
10
|
Jules Christophe MK, Marlène YT, Valery Jean François N, Merlin NN, Inocent G, Mathieu N. Assessment of cooking methods and freezing on the nutritional value and health risks of heavy metals in four fish species consumed in Douala, Cameroon. Heliyon 2024; 10:e28316. [PMID: 38576585 PMCID: PMC10990866 DOI: 10.1016/j.heliyon.2024.e28316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 04/06/2024] Open
Abstract
The effects of smoking, boiling and freezing on the nutritional value and health risks of heavy metals in four fish species consumed in Douala was investigated. Fish samples from Cyprinus carpio, Arius parkii, Ethmalosa fimbriata and Polydactilis quadrifilis were collected at the Douala Fishing seaport, carried to the laboratory, washed with distilled water and processed. Proximate composition, mineral content, heavy metals and lipid quality were analyzed using AOAC standard methods. Estimated Daily Intake (EDI), Targeted Hazard Quotient (THQ), Hazard Index (HI) and Carcinogenic Risk (CR) were used to estimate the human health risk. Results showed that smoking and boiling increased significantly (P < 0.05) protein and ash levels. Lipid were reduced significantly (P < 0.05) with boiling and freezing compared to raw and smoked sample. Smoking increased significantly (P < 0.05) cadmium, lead, mercury and arsenic contents compared to boiling and freezing. EDI values of cadmium in all species of fish smoked were not acceptable for human consumption. THQ values of mercury in raw, smoked, boiled and frozen were not acceptable for human consumption. HI suggested a non potential carcinogenic effect for all fish while CR for cadmium and arsenic suggested a carcinogenic health risk for Arius parkii (smoked and boiled). All treatment decreased significantly (P < 0.05) iodine value and increased acid, peroxide, anisidine values, thiobarbituric acid reactive substances and total oxidation index compared to raw fish. Boiling was the best cooking method compared to smoking.
Collapse
Affiliation(s)
| | - Youogo Tegueu Marlène
- Laboratory of Biochemistry, Faculty of Science, University of Douala, Po Box 24157, Douala, Cameroon
| | | | - Nchoutpouen Ngafon Merlin
- Laboratory of Biochemistry, Faculty of Science, University of Douala, Po Box 24157, Douala, Cameroon
| | - Gouado Inocent
- Laboratory of Biochemistry, Faculty of Science, University of Douala, Po Box 24157, Douala, Cameroon
| | - Ndomou Mathieu
- Laboratory of Biochemistry, Faculty of Science, University of Douala, Po Box 24157, Douala, Cameroon
| |
Collapse
|
11
|
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
|