1
|
Zhang Z, Yang J, Zhou Q, Zhong S, Liu J, Zhang X, Chang X, Wang H. The cGAS-STING-mediated ROS and ferroptosis are involved in manganese neurotoxicity. J Environ Sci (China) 2025; 152:71-86. [PMID: 39617588 DOI: 10.1016/j.jes.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/12/2024] [Accepted: 05/01/2024] [Indexed: 12/12/2024]
Abstract
Manganese (Mn) has been characterized as an environmental pollutant. Excessive releases of Mn due to human activities have increased Mn levels in the environment over the years, posing a threat to human health and the environment. Long-term exposure to high concentrations of Mn can induce neurotoxicity. Therefore, toxicological studies on Mn are of paramount importance. Mn induces oxidative stress through affecting the level of reactive oxygen species (ROS), and the overabundance of ROS further triggers ferroptosis. Additionally, Mn2+ was found to be a novel activator of the cyclic guanosine-adenosine synthase (cGAS)-stimulator of interferon genes (STING) pathway in the innate immune system. Thus, we speculate that Mn exposure may promote ROS production by activating the cGAS-STING pathway, which further induces oxidative stress and ferroptosis, and ultimately triggers Mn neurotoxicity. This review discusses the mechanism between Mn-induced oxidative stress and ferroptosis via activation of the cGAS-STING pathway, which may offer a prospective direction for future in-depth studies on the mechanism of Mn neurotoxicity.
Collapse
Affiliation(s)
- Zhimin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jirui Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qiongli Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Shiyin Zhong
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jingjing Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
2
|
Pu B. Association between blood manganese and cardiovascular diseases among U.S. adult population. Sci Rep 2024; 14:31260. [PMID: 39733137 DOI: 10.1038/s41598-024-82673-4] [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: 06/22/2024] [Accepted: 12/09/2024] [Indexed: 12/30/2024] Open
Abstract
Manganese (Mn) is a known toxicant and an essential trace element, and it plays an important role in various mechanisms in relation to cardiovascular health. However, epidemiological studies of the association between blood Mn and cardiovascular diseases (CVD) among U.S. adults are rare. A cross-sectional study of 12,061 participants aged ≥ 20 was conducted using data from the National Health and Nutrition Examination Survey 2011-2018. Logistic regression and restricted cubic spline were used to examine the relationship between blood Mn levels and total CVD risk and specific CVD subtypes. Bayesian kernel-machine regression (BKMR) and weighted quantile sum (WQS) analyses were performed to explore the joint effects of Mn with other metals on CVD. The results showed that individuals with the third quartile group of blood Mn levels had significantly lower risks of CVD, displaying a non-linear U-shaped dose-response relationship. A significant interaction of age on this association was observed. No significant associations were found between Mn levels and specific CVD subtypes. BKMR and WQS analyses showed a positive association between heavy metal mixtures and CVD risks, with no interaction between Mn and other metals. In conclusion, blood Mn levels were significantly associated with CVD risks with a U-shaped relationship in U.S. adults, with possible age-specific differences. Future larger prospective studies are warranted to validate these findings.
Collapse
Affiliation(s)
- Boxuan Pu
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, National Center for Cardiovascular Diseases, Beijing, 102300, China.
| |
Collapse
|
3
|
Urbano T, Vinceti M, Carbone C, Wise LA, Malavolti M, Tondelli M, Bedin R, Vinceti G, Marti A, Chiari A, Zamboni G, Michalke B, Filippini T. Exposure to Cadmium and Other Trace Elements Among Individuals with Mild Cognitive Impairment. TOXICS 2024; 12:933. [PMID: 39771148 PMCID: PMC11679412 DOI: 10.3390/toxics12120933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND A limited number of studies have investigated the role of environmental chemicals in the etiology of mild cognitive impairment (MCI). We performed a cross-sectional study of the association between exposure to selected trace elements and the biomarkers of cognitive decline. METHODS During 2019-2021, we recruited 128 newly diagnosed patients with MCI from two Neurology Clinics in Northern Italy, i.e., Modena and Reggio Emilia. At baseline, we measured serum and cerebrospinal fluid (CSF) concentrations of cadmium, copper, iron, manganese, and zinc using inductively coupled plasma mass spectrometry. With immuno-enzymatic assays, we estimated concentrations of β-amyloid 1-40, β-amyloid 1-42, Total Tau and phosphorylated Tau181 proteins, neurofilament light chain (NfL), and the mini-mental state examination (MMSE) to assess cognitive status. We used spline regression to explore the shape of the association between exposure and each endpoint, adjusted for age at diagnosis, educational attainment, MMSE, and sex. RESULTS In analyses between the serum and CSF concentrations of trace metals, we found monotonic positive correlations between copper and zinc, while an inverse association was observed for cadmium. Serum cadmium concentrations were inversely associated with amyloid ratio and positively associated with Tau proteins. Serum iron concentrations showed the opposite trend, while copper, manganese, and zinc displayed heterogeneous non-linear associations with amyloid ratio and Tau biomarkers. Regarding CSF exposure biomarkers, only cadmium consistently showed an inverse association with amyloid ratio, while iron was positively associated with Tau. Cadmium concentrations in CSF were not appreciably associated with serum NfL levels, while we observed an inverted U-shaped association with CSF NfL, similar to that observed for copper. In CSF, zinc was the only trace element positively associated with NfL at high concentrations. CONCLUSIONS In this cross-sectional study, high serum cadmium concentrations were associated with selected biomarkers of cognitive impairment. Findings for the other trace elements were difficult to interpret, showing complex and inconsistent associations with the neurodegenerative endpoints examined.
Collapse
Affiliation(s)
- Teresa Urbano
- Environmental, Genetics, and Nutritional Epidemiology Research Center (CREAGEN), Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (T.U.); (M.M.); (T.F.)
| | - Marco Vinceti
- Environmental, Genetics, and Nutritional Epidemiology Research Center (CREAGEN), Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (T.U.); (M.M.); (T.F.)
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA;
| | - Chiara Carbone
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (M.T.); (R.B.); (G.Z.)
- Neurology Unit, Baggiovara Hospital, 41126 Modena, Italy; (G.V.); (A.C.)
| | - Lauren A. Wise
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA;
| | - Marcella Malavolti
- Environmental, Genetics, and Nutritional Epidemiology Research Center (CREAGEN), Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (T.U.); (M.M.); (T.F.)
| | - Manuela Tondelli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (M.T.); (R.B.); (G.Z.)
- Neurology Unit, Baggiovara Hospital, 41126 Modena, Italy; (G.V.); (A.C.)
| | - Roberta Bedin
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (M.T.); (R.B.); (G.Z.)
| | - Giulia Vinceti
- Neurology Unit, Baggiovara Hospital, 41126 Modena, Italy; (G.V.); (A.C.)
| | - Alessandro Marti
- Clinical Neuropsychology, Cognitive Disorders and Dyslexia Unit, Neuromotor and Rehabilitation Department, AUSL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Annalisa Chiari
- Neurology Unit, Baggiovara Hospital, 41126 Modena, Italy; (G.V.); (A.C.)
| | - Giovanna Zamboni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (M.T.); (R.B.); (G.Z.)
- Neurology Unit, Baggiovara Hospital, 41126 Modena, Italy; (G.V.); (A.C.)
| | - Bernhard Michalke
- Research Unit Analytical BioGeoChemistry, German Research Center for Environmental Health, Helmholtz Center Munich, 85764 Neuherberg, Germany;
| | - Tommaso Filippini
- Environmental, Genetics, and Nutritional Epidemiology Research Center (CREAGEN), Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (T.U.); (M.M.); (T.F.)
- School of Public Health, University of California Berkeley, Berkeley, CA 94704, USA
| |
Collapse
|
4
|
Ma Y, Chen H, Jiang Y, Wang D, Aschner M, Luo W, Su P. RhoA/ROCK2 signaling pathway regulates Mn-induced alterations in tight junction proteins leading to cognitive dysfunction in mice. Curr Res Toxicol 2024; 8:100207. [PMID: 39834519 PMCID: PMC11745801 DOI: 10.1016/j.crtox.2024.100207] [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: 10/29/2024] [Revised: 11/19/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
Elevated manganese (Mn) exposure has been implicated in a broad spectrum of neurological disorders, including motor dysfunction and cognitive deficits. Previous studies have demonstrated that Mn induces neurotoxicity by disrupting the integrity of the blood-brain barrier (BBB), a critical regulator in maintaining central nervous system homeostasis and a contributing factor in the pathogenesis of numerous neurological disorders. However, the precise molecular mechanisms underlying Mn-induced BBB disruption and its role in facilitating neurotoxicity remain incompletely understood. The primary objectives of this study were to elucidate the mechanisms underlying the relationship between Mn exposure and BBB tight junction proteins (TJPs), and to further investigate potential neuroprotective strategies for mitigating Mn-induced cognitive impairments. In this investigation, we developed Mn exposure models utilizing both murine subjects and cell culture systems to elucidate the mechanisms underlying TJPs involvement and to assess the potential neuroprotective effects of gastrodin (GAS), a bioactive compound extracted from traditional Chinese medicine. Our findings revealed a significant reduction in TJPs expression, both in vivo and in vitro, in Mn-induced BBB disruption. The overexpression of Occludin (OCLN), a crucial component of TJPs, mitigated Mn-induced BBB damage. GAS administration effectively attenuated Mn-induced disruption of the BBB, enhanced the expression of TJPs, and mitigated Mn-induced cognitive dysfunctions, potentially through the modulation of the RhoA/ROCK2 signaling pathway. This research sought to advance our understanding of the molecular pathways involved in Mn-mediated BBB disruption and to identify novel therapeutic approaches for mitigating the deleterious effects of Mn exposure on cognitive function.
Collapse
Affiliation(s)
- Yan Ma
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Chang Le Xi Road, Xi’an,Shaanxi 710032, China
| | - Honggang Chen
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Chang Le Xi Road, Xi’an,Shaanxi 710032, China
| | - Yuxin Jiang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Chang Le Xi Road, Xi’an,Shaanxi 710032, China
| | - Diya Wang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Chang Le Xi Road, Xi’an,Shaanxi 710032, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, United States of America
| | - Wenjing Luo
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Chang Le Xi Road, Xi’an,Shaanxi 710032, China
| | - Peng Su
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Chang Le Xi Road, Xi’an,Shaanxi 710032, China
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
5
|
Yu L, Zhang H, Liu J, Cao S, Li S, Li F, Xia W, Xu S, Li Y. Thyroid-stimulating hormone (TSH) mediates the associations between maternal metals and neurodevelopment in children: A prospective cohort study. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125150. [PMID: 39427953 DOI: 10.1016/j.envpol.2024.125150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/22/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
Insufficient research has focused on the effects of metal mixtures on children's neurodevelopment and TSH's potential mediating effect. Plasma concentrations of ten metals were measured among 2887 pregnant women in a persistent Chinese birth cohort. At age two, children's neurodevelopment was assessed using mental development indexes (MDIs) and psychomotor development indexes (PDIs), defining neurodevelopmental delay as MDI≤ 79 (cognitive delay) or PDI≤ 79 (motor delay). The associations between single and mixed metals with neurodevelopment delay risk were examined using generalized linear regression complemented by weighted quantile sum (WQS) regression. To investigate the mediated effects of infant Thyroid-Stimulating Hormone (TSH) on metal-associated neurodevelopment delay risk, mediation analyses were conducted. According to the single-metal model, V, Mn, and Pb levels are positively associated with neurodevelopment delay. The WQS model found consistent associations (Odds Ratio [OR] 1.55, 95% Confidence Interval [CI] 1.23 to 1.95), highlighting V, Mn, and Pb as the main causes of cognitive delay. Further mediation analysis revealed that the association between metals (mainly V, Mn, and Pb) and neurodevelopment delay risk is mediated by TSH, with proportions ranging from 3.18 to 10.14% (all P < 0.05). Our findings highlighted prenatal exposure to metals was associated with higher risks of neurodevelopmental delay, with TSH possibly mediating this effect.
Collapse
Affiliation(s)
- Ling Yu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | | | - Jiangtao Liu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuting Cao
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shulan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fasheng Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Wei Xia
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shunqing Xu
- School of Environmental Science and Engineering, Hainan University, Haikou, Hainan, China.
| | - Yuanyuan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
6
|
Ren J, Wu W, Li J, Hu Q, Zhang M, Wang J, Li X, Li Y, Huang B. Association of metalloestrogens exposure with depression in women across reproductive lifespan. Front Psychiatry 2024; 15:1486402. [PMID: 39691784 PMCID: PMC11649658 DOI: 10.3389/fpsyt.2024.1486402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/15/2024] [Indexed: 12/19/2024] Open
Abstract
Background Exposure to metal could impact women's depression risk. However, the connection and mechanisms between metalloestrogens exposure and depression are still not fully understood. We aim to explore the associations between metalloestrogens and the risk of depression in women across reproductive lifespan. Methods Using data from NHANES 2011-2018, we employed logistic regression and baknernel machine regression (BKMR) to study links between metalloestrogen exposure and depression in US women. We analyzed how contraceptive use affects this relationship. Results The study involved 3,374 adult women, with 345 of them experiencing depression. Our research revealed that certain metalloestrogens like Ba, Ca, Pb, Sb, and Sn were linked to higher depression risk in women, while Hg was associated with lower depression risk in older women. For women aged 18-44, a blend of metalloestrogens showed a significant positive correlation with depression risk, and the likelihood of depression in later years notably rose when the metal mixture concentration reached or exceeded the 60th percentile. Oral contraceptives would have an effect on the impact of metalloestrogen mixture exposure on depression in women during the reproductive stage. Conclusions Our study indicates a significant link between metalloestrogen exposure and a higher risk of depression in adult women in the United States. This finding can aid in identifying the connection and enhancing women's mental well-being.
Collapse
Affiliation(s)
- Junjie Ren
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, Anhui, China
| | - Wanxin Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Jia Li
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Qifang Hu
- Shenzhen Hospital of Southern Medical University, Shenzhen Clinical Medical School, Shenzhen, China
| | - Mi Zhang
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, Anhui, China
| | - Jing Wang
- Department of Materials Science and Engineering, Shanghai University of Engineering Science, Shanghai, China
| | - Xiaoming Li
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, Anhui, China
| | - Yanwen Li
- Department of Clinical Laboratory, Clinical Laboratory Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Binbin Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
7
|
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
|
8
|
Bai YZ, Zhang SQ. Selenium intake is an effective strategy for the improvement of cognitive decline in low cognition older Americans. Int J Food Sci Nutr 2024; 75:687-694. [PMID: 39034505 DOI: 10.1080/09637486.2024.2380758] [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: 12/13/2023] [Revised: 06/11/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
Age-related cognitive decline is a prominent concern in older adults and selenium (Se) deficiency has been found to be associated with cognitive deficits. For the first time, the present study explored the association between Se intake and cognitive performance in older people with/without cognitive impairment using the data from the National Health and Nutrition Examination Survey 2011-2014. Weighted linear regression models were conducted to evaluate the association between dietary Se/total Se intakes and cognitive assessments. A total of 2387 participants were included. The significant positive association between dietary Se/total Se intakes and total scores of cognitive functioning tests existed only in the older people with low cognitive performance (p < 0.001), not in those with normal cognitive performance. In conclusion, Se intake was beneficial for cognitive decline only in the low cognition older people but failed in normal cognition older people.
Collapse
Affiliation(s)
- Ya-Zhi Bai
- Department of Nutrition and Metabolism, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shuang-Qing Zhang
- Department of Nutrition and Metabolism, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
9
|
Okeowo OM, Anadu VE, Ijomone OK, Aschner M, Ijomone OM. Combined Restraint Stress and Metal Exposure Paradigms in Rats: Unravelling Behavioural and Neurochemical Perturbations. Mol Neurobiol 2024:10.1007/s12035-024-04570-1. [PMID: 39443350 DOI: 10.1007/s12035-024-04570-1] [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: 06/09/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Accumulation of heavy metals (Mn and Ni) and prolonged exposure to stress are associated with adverse health outcomes. Various studies have shown the impacts of stress and metal exposures on brain function. However, no study has examined the effects of co-exposure to stress, Mn, and Ni on the brain. This study addresses this gap by evaluating oxidative and glial responses, apoptotic activity, as well as cognitive processes in a rat model. Adult Wistar rats were exposed to vehicle (control), restraint stress, 25 mg/kg of manganese (Mn) or nickel (Ni), or combined restraint stress plus Mn or Ni. Following treatment, rats were subjected to several behavioural paradigms to assess cognitive function. Enzyme activity, as well as ATPase levels, were evaluated. Thereafter, an immunohistochemical procedure was utilised to evaluate neurochemical markers of glial function, myelination, oxidative stress, and apoptosis in the hippocampus, prefrontal cortex (PFC), and striatum. Results showed that stress and metal exposure increased oxidative stress markers and reduced antioxidant levels. Further, combined stress and metal exposure reduced various forms of learning and memory ability in rats. In addition, there were alterations in Iba1 activity and Nrf2 levels, reduced Olig2 and myelin basic protein (MBP) levels, and increased caspase-3 expression. These neurotoxic outcomes were mostly exacerbated by co-exposure to stress and metals. Overall, our findings establish that stress and metal exposures impaired cognitive performance, induced oxidative stress and apoptosis, and led to demyelination effects which were worsened by combined stress and metal exposure.
Collapse
Affiliation(s)
- Oritoke M Okeowo
- Department of Physiology, School of Basic Medical Sciences, Federal University of Technology, Akure, Nigeria
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Nigeria
| | - Victor E Anadu
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Nigeria
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Medical Sciences, Ondo, Nigeria
| | - Olayemi K Ijomone
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Nigeria
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Medical Sciences, Ondo, Nigeria
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Omamuyovwi M Ijomone
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Nigeria.
- Department of Human Anatomy, School of Basic Medical Sciences, Federal University of Technology, Akure, Nigeria.
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Medical Sciences, Ondo, Nigeria.
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
10
|
Mehrdad SA, Cucchiarini A, Mergny JL, Kazemi Noureini S. Heavy metal ions interactions with G-quadruplex-prone DNA sequences. Biochimie 2024; 225:146-155. [PMID: 38821199 DOI: 10.1016/j.biochi.2024.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
The industrial world exposes living organisms to a variety of metal pollutants. Here we investigated whether such elements affect G-rich sequences susceptible to fold into G-quadruplex (GQ) structures. Thermal stability and conformation of these oligoncleotides was studied at various molar ratios of a variety of heavy metal salts using thermal FRET, transition-FRET (t-FRET) and circular dichroism. Metal ions affected the thermal stability of the GQs to different extents; some metals had no effect on Tm while other metals caused small to moderate changes in Tm at 1:1 or 1:10 molar ratio. While most of the metals had no major effect, Al3+, Cd2+, Pb2+, Hg2+ and Zn2+ altered the thermal stability and structural features of the GQs. Some metals such as Pb2+ and Hg2+ exhibit differential interactions with telomere, c-myc and c-kit GQs. Overall, toxic heavy metals affect G-quadruplex stability in a sequence and topology dependent manner. This study provides new insight into how heavy metal exposure may affect gene expression and cellular responses.
Collapse
Affiliation(s)
- Seyyed-Ali Mehrdad
- Department of Biology, Faculty of Basic Sciences, Hakim Sabzevari University, Sabzevar, Iran
| | - Anne Cucchiarini
- Laboratoire d'Optique et Biosciences (LOB), Ecole Polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, 91120, Palaiseau, France
| | - Jean-Louis Mergny
- Laboratoire d'Optique et Biosciences (LOB), Ecole Polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, 91120, Palaiseau, France
| | - Sakineh Kazemi Noureini
- Department of Biology, Faculty of Basic Sciences, Hakim Sabzevari University, Sabzevar, Iran.
| |
Collapse
|
11
|
Wei R, Wei P, Yuan H, Yi X, Aschner M, Jiang YM, Li SJ. Inflammation in Metal-Induced Neurological Disorders and Neurodegenerative Diseases. Biol Trace Elem Res 2024; 202:4459-4481. [PMID: 38206494 DOI: 10.1007/s12011-023-04041-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024]
Abstract
Essential metals play critical roles in maintaining human health as they participate in various physiological activities. Nonetheless, both excessive accumulation and deficiency of these metals may result in neurotoxicity secondary to neuroinflammation and the activation of microglia and astrocytes. Activation of these cells can promote the release of pro-inflammatory cytokines. It is well known that neuroinflammation plays a critical role in metal-induced neurotoxicity as well as the development of neurological disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Initially seen as a defense mechanism, persistent inflammatory responses are now considered harmful. Astrocytes and microglia are key regulators of neuroinflammation in the central nervous system, and their excessive activation may induce sustained neuroinflammation. Therefore, in this review, we aim to emphasize the important role and molecular mechanisms underlying metal-induced neurotoxicity. Our objective is to raise the awareness on metal-induced neuroinflammation in neurological disorders. However, it is not only just neuroinflammation that different metals could induce; they can also cause harm to the nervous system through oxidative stress, apoptosis, and autophagy, to name a few. The primary pathophysiological mechanism by which these metals induce neurological disorders remains to be determined. In addition, given the various pathways through which individuals are exposed to metals, it is necessary to also consider the effects of co-exposure to multiple metals on neurological disorders.
Collapse
Affiliation(s)
- Ruokun Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Peiqi Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Haiyan Yuan
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Xiang Yi
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, 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, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| | - Shao-Jun Li
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| |
Collapse
|
12
|
McCabe SM, Zhao N. Expression of Manganese Transporters ZIP8, ZIP14, and ZnT10 in Brain Barrier Tissues. Int J Mol Sci 2024; 25:10342. [PMID: 39408669 PMCID: PMC11476488 DOI: 10.3390/ijms251910342] [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] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/15/2024] [Accepted: 09/22/2024] [Indexed: 10/20/2024] Open
Abstract
Manganese (Mn) is an essential trace mineral for brain function, but excessive accumulation can cause irreversible nervous system damage, highlighting the need for proper Mn balance. ZIP14, ZnT10, and ZIP8 are key transporters involved in maintaining Mn homeostasis, particularly in the absorption and excretion of Mn in the intestine and liver. However, their roles in the brain are less understood. The blood-cerebrospinal fluid barrier and the blood-brain barrier, formed by the choroid plexus and brain blood vessels, respectively, are critical for brain protection and brain metal homeostasis. This study identified ZIP14 on the choroid plexus epithelium, and ZIP8 and ZnT10 in brain microvascular tissue. We show that despite significant Mn accumulation in the CSF of Znt10 knockout mice, ZIP14 expression levels in the blood-cerebrospinal fluid barrier remain unchanged, indicating that ZIP14 does not have a compensatory mechanism for regulating Mn uptake in the brain in vivo. Additionally, Mn still enters the CSF without ZIP14 when systemic levels rise. This indicates that alternative transport mechanisms or compensatory pathways ensure Mn balance in the CSF, shedding light on potential strategies for managing Mn-related disorders.
Collapse
Affiliation(s)
| | - Ningning Zhao
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ 85721, USA;
| |
Collapse
|
13
|
Qiu Y, Liu Y, Gan M, Wang W, Jiang T, Jiang Y, Lv H, Lu Q, Qin R, Tao S, Huang L, Xu X, Liu C, Dou Y, Ke K, Sun T, Jiang Y, Xu B, Jin G, Ma H, Shen H, Hu Z, Lin Y, Du J. Association of prenatal multiple metal exposures with child neurodevelopment at 3 years of age: A prospective birth cohort study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 942:173812. [PMID: 38857795 DOI: 10.1016/j.scitotenv.2024.173812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/13/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Prenatal exposures to toxic metals and trace elements have been linked to childhood neurodevelopment. However, existing evidence remains inconclusive, and further research is needed to investigate the mixture effects of multiple metal exposures on childhood neurodevelopment. We aimed to examine the associations between prenatal exposure to specific metals and metal mixtures and neurodevelopment in children. In this prospective cohort study, we used the multivariable linear regressions and the robust modified Poisson regressions to explore the associations of prenatal exposure to 25 specific metals with neurodevelopment among children at 3 years of age in 854 mother-child pairs from the Jiangsu Birth Cohort (JBC) Study. The Bayesian kernel machine regression (BKMR) was employed to assess the joint effects of multiple metals on neurodevelopment. Prenatal manganese (Mn) exposure was negatively associated with the risk of non-optimal cognition development of children, while vanadium (V), copper (Cu), zinc (Zn), antimony (Sb), cerium (Ce) and uranium (U) exposures were positively associated with the risk of non-optimal gross motor development. BKMR identified an interaction effect between Sb and Ce on non-optimal gross motor development. Additionally, an element risk score (ERS), representing the mixture effect of multiple metal exposures including V, Cu, Zn, Sb, Ce and U was constructed based on weights from a Poisson regression model. Children with ERS in the highest tertile had higher probability of non-optimal gross motor development (RR = 2.37, 95 % CI: 1.15, 4.86) versus those at the lowest tertile. Notably, Sb [conditional-posterior inclusion probabilities (cPIP) = 0.511] and U (cPIP = 0.386) mainly contributed to the increased risk of non-optimal gross motor development. The findings highlight the importance of paying attention to the joint effects of multiple metals on children's neurodevelopment. The ERS score may serve as an indicator of comprehensive metal exposure risk for children's neurodevelopment.
Collapse
Affiliation(s)
- Yun Qiu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, Jiangsu, China
| | - Yuxin Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Ming Gan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Weiting Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Tao Jiang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Yangqian Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Hong Lv
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, Jiangsu, China
| | - Qun Lu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Rui Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Shiyao Tao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Lei Huang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Xin Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Cong Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Yuanyan Dou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Kang Ke
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Tianyu Sun
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Yue Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Bo Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Guangfu Jin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, Jiangsu, China
| | - Hongxia Ma
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, Jiangsu, China
| | - Hongbing Shen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, Jiangsu, China
| | - Yuan Lin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, Jiangsu, China; Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| | - Jiangbo Du
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, Jiangsu, China.
| |
Collapse
|
14
|
Wei S, Ma X, Liang G, He J, Wang J, Chen H, Lu W, Qin H, Zou Y. The role of circHmbox1(3,4) in ferroptosis-mediated cognitive impairments induced by manganese. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135212. [PMID: 39024764 DOI: 10.1016/j.jhazmat.2024.135212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/07/2024] [Accepted: 07/13/2024] [Indexed: 07/20/2024]
Abstract
Excessive environmental exposure to manganese (Mn) has been linked to cognitive impairments, circular RNAs (circRNAs) have been recognized for their roles in epigenetic regulation in various biological processes, including neurological pathogenesis. Previous studies found that ferroptosis, an iron ion-dependent programmed cell death, may be involved in cognitive impairments. However, specific mechanisms underlying the relationship among circRNA, ferroptosis, and neurotoxicity of Mn are not well-understood. In the current study, RNA sequencing was performed to profile RNA expression in Neuro-2a (N2a) cells that were treated with 300 μM Mn. The potential molecular mechanisms of circHmbox1(3,4) in Mn-induced cognitive impairments were investigated via various experiments, such as Western blot and intracerebroventricular injection in mice. We observed a significant decrease in the expression of circHmbox1(3,4) both in vitro and in vivo following Mn treatment. The results of Y maze test and Morris water maze test demonstrated an improvement in learning and memory abilities following circHmbox1(3,4) overexpression in Mn treated mice. Mn treatment may reduce circHmbox1(3,4) biogenesis through lowered expression of E2F1/QKI. Inhibiting circHmbox1(3,4) expression led to GPX4 protein degradation through protein ligation and ubiquitination. Overall, the current study showed that Mn exposure-induced cognitive dysfunction may be mediated through ferroptosis regulated by circHmbox1(3,4).
Collapse
Affiliation(s)
- Shengtao Wei
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Xiaoli Ma
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Guiqiang Liang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Jiacheng He
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Jian Wang
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China; Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Hao Chen
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China; Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Wenmin Lu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Huiyan Qin
- Guangxi Zhuang Autonomous Region Center for Disease Control and Prevention, Nanning 530028, Guangxi, China
| | - Yunfeng Zou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China; Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Nanning 530021, Guangxi, China.
| |
Collapse
|
15
|
Wen Y, Fu Z, Li J, Liu M, Wang X, Chen J, Chen Y, Wang H, Wen S, Zhang K, Deng Y. Targeting m 6A mRNA demethylase FTO alleviates manganese-induced cognitive memory deficits in mice. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:134969. [PMID: 38908185 DOI: 10.1016/j.jhazmat.2024.134969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Manganese (Mn) induced learning and memory deficits through mechanisms that are not fully understood. In this study, we discovered that the demethylase FTO was significantly downregulated in hippocampal neurons in an experimental a mouse model of Mn exposure. This decreased expression of FTO was associated with Mn-induced learning and memory impairments, as well as the dysfunction in synaptic plasticity and damage to regional neurons. The overexpression of FTO, or its positive modulation with agonists, provides protection against neurological damage and cognitive impairments. Mechanistically, FTO interacts synergistically with the reader YTHDF3 to facilitate the degradation of GRIN1 and GRIN3B through the m6A modification pathway. Additionally, Mn decreases the phosphorylation of SOX2, which specifically impairs the transcriptional regulation of FTO activity. Additionally, we found that the natural compounds artemisinin and apigenin that can bind molecularly with SOX2 and reduce Mn-induced cognitive dysfunction in mice. Our findings suggest that the SOX2-FTO-Grins axis represents a viable target for addressing Mn-induced neurotoxicity and cognitive impairments.
Collapse
Affiliation(s)
- Yi Wen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Zhushan Fu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Jiashuo Li
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China; Department of Occupational and Environmental Health, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Mingyue Liu
- Department of Developmental Cell Biology, School of Life Sciences, China Medical University, Shenyang, China; Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Xinmiao Wang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Jingqi Chen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Yue Chen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Haocheng Wang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Sihang Wen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Ke Zhang
- Department of Developmental Cell Biology, School of Life Sciences, China Medical University, Shenyang, China; Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China; Institute of Health Professions Education Assessment and Reform, China Medical University, Shenyang, China.
| |
Collapse
|
16
|
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] [Grants] [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
|
17
|
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
|
18
|
Dai G, Sun H, Lan Y, Jiang J, Fang B. The association of manganese levels with red cell distribution width: A population-based study. PLoS One 2024; 19:e0292569. [PMID: 39146304 PMCID: PMC11326586 DOI: 10.1371/journal.pone.0292569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 05/08/2024] [Indexed: 08/17/2024] Open
Abstract
OBJECTIVES Experimental and acute exposure studies imply that manganese affects red blood cell production. Nevertheless, the association between environmental exposure and red blood cell distribution width (RDW) has yet to be explored. This research sought to assess the correlation between blood manganese levels and RDW within the general population of the United States. MATERIALS AND METHODS Employing weighted multiple linear regression models, data from the 2011-2018 National Health and Nutrition Examination Survey (NHANES) were utilized to assess the correlation between manganese levels in the blood and RDW. Restricted cubic spline plots and two-piecewise linear regression models were also employed. RESULT The analysis included a total of 15882 participants in which we determined an independent positive relationship between blood manganese levels and RDW among participants(β = 0.079, P<0.001). Moreover, we identified a J-shaped association between blood manganese levels and RDW in total participants (inflection point for blood manganese: 7.32 ug/L) and distinct subgroups following adjusted covariates. Women exhibited a more pronounced association, even after controlling for adjusted covariates. CONCLUSIONS We determined a J-shaped relationship between blood manganese levels and RDW with an inflection point at 7.32 ug/L for blood manganese. Nevertheless, fundamental research and large sample prospective studies are needed to determine the extent to which blood manganese levels correlate with RDW.
Collapse
Affiliation(s)
- Guanmian Dai
- Department of Hematology, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Huanhuan Sun
- Department of Traditional Chinese Medicine, FuYang Women and Children's Hospital, Fuyang, Anhui, China
| | - Yanli Lan
- Department of Oncology, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Jinhong Jiang
- Department of Hematology, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Bingmu Fang
- Department of Hematology, Lishui People's Hospital, Lishui, Zhejiang, China
| |
Collapse
|
19
|
Gu Q, Liu J, Zhang X, Huang A, Yu X, Wu K, Huang Y. Association between heavy metals exposure and risk of attention deficit hyperactivity disorder (ADHD) in children: a systematic review and meta-analysis. Eur Child Adolesc Psychiatry 2024:10.1007/s00787-024-02546-z. [PMID: 39126497 DOI: 10.1007/s00787-024-02546-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024]
Abstract
Heavy metals can negatively affect children's neurodevelopment, yet the relationship between heavy metals exposure and attention deficit hyperactivity disorder (ADHD) in children remains unclear. We aimed to examine associations between exposure to five common heavy metals (lead, arsenic, mercury, cadmium, and manganese) with neurodevelopmental toxicity and the risk of ADHD in children. Online databases of PubMed, Web of Science, and Embase were searched before February 29, 2024. A total of 31 studies involving 25,258 children were included in the final analysis. Our findings revealed that lead exposure was positively associated with ADHD risk in children (OR = 1.95, 95% CI: 1.57-2.41) overall, while the associations varied among different WHO regions, with the strongest in the Americas. Sensitivity analyses revealed significant associations between arsenic (OR = 1.53, 95% CI: 1.01-2.32) and manganese (OR = 1.79, 95% CI: 1.28-2.49) exposure and ADHD risk after omitting one study. Arsenic exposure was positively associated with ADHD risk in studies conducted in the Americas and adjusted for environmental smoke exposure. Positive associations between manganese exposure and ADHD risk were also found in several subgroup analyses. No significant associations were found for mercury and cadmium exposure. Dose-response meta-analysis suggested that children with higher blood lead levels exhibited a higher probability of ADHD diagnosis. Lead exposure consistently increases the risk of ADHD in children, while arsenic and manganese exposure may be associated with ADHD under different occasions. More research is required to understand heavy metals' impact on ADHD across varying exposure levels, particularly in less contaminated regions.
Collapse
Affiliation(s)
- Qianfei Gu
- School of Public Health, Shantou University, Shantou, Guangdong Province, China
- Mental Health Center of Shantou University, Shantou, Guangdong Province, China
| | - Jiayu Liu
- Mental Health Center of Shantou University, Shantou, Guangdong Province, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong Province, China
| | - Xuanzhi Zhang
- Mental Health Center of Shantou University, Shantou, Guangdong Province, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong Province, China
| | - Anyan Huang
- Shantou Maternity & Child Healthcare Hospital, Shantou, Guangdong Province, China
| | - Xinle Yu
- Mental Health Center of Shantou University, Shantou, Guangdong Province, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong Province, China
| | - Kusheng Wu
- Department of Preventive Medicine, Shantou University Medical College, Shantou, Guangdong Province, China.
| | - Yanhong Huang
- Mental Health Center of Shantou University, Shantou, Guangdong Province, China.
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong Province, China.
| |
Collapse
|
20
|
Xia Z, Tang B, Li X, Li X, Jia Y, Jiang J, Chen J, Song J, Liu S, Min J, Wang F. A Novel Role for the Longevity-Associated Protein SLC39A11 as a Manganese Transporter. RESEARCH (WASHINGTON, D.C.) 2024; 7:0440. [PMID: 39114488 PMCID: PMC11304475 DOI: 10.34133/research.0440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024]
Abstract
The identification of aging- and longevity-associated genes is important for promoting healthy aging. By analyzing a large cohort of Chinese centenarians, we previously found that single-nucleotide polymorphisms (SNPs) in the SLC39A11 gene (also known as ZIP11) are associated with longevity in males. However, the function of the SLC39A11 protein remains unclear. Here, we found that SLC39A11 expression is significantly reduced in patients with Hutchinson-Gilford progeria syndrome (HGPS). In addition, we found that zebrafish with a mutation in slc39a11 that significantly reduces its expression have an accelerated aging phenotype, including a shortened average lifespan, muscle atrophy and reduced swimming, impaired muscle regeneration, gut damage, and abnormal morphology in the reproductive system. Interestingly, these signs of premature aging were more pronounced in male zebrafish than in females. RNA-sequencing analysis revealed that cellular senescence may serve as a potential mechanism for driving this slc39a11 deficiency-induced phenotype in mutant zebrafish. Moreover, immunofluorescence showed significantly increased DNA damage and reactive oxygen species signaling in slc39a11 mutant zebrafish. Using inductively coupled plasma mass spectrometry (ICP-MS), we found that manganese significantly accumulates in slc39a11 mutant zebrafish, as well as in the serum of both global Slc39a11 knockout and hepatocyte-specific Slc39a11 knockout mice, suggesting that this metal transporter regulates systemic manganese levels. Finally, using cultured human fibroblasts, we found that both knocking down SLC39A11 and exposure to high extracellular manganese increased cellular senescence. These findings provide compelling evidence that SLC39A11 serves to protect against the aging process, at least in part by regulating cellular manganese homeostasis.
Collapse
Affiliation(s)
- Zhidan Xia
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Biyao Tang
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
- School of Public Health, Basic Medical Sciences, The First Affiliated Hospital, Hengyang Medical School,
University of South China, Hengyang, China
- School of Public Health, School of Basic Medical Sciences, The First Affiliated Hospital,
Xinxiang Medical University, Xinxiang, China
| | - Xiaopeng Li
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Xinran Li
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Yangfan Jia
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Jianwei Jiang
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Jingyao Chen
- The Core Facilities,
Zhejiang University School of Medicine, Hangzhou, China
| | - Jingshu Song
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Siyi Liu
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Junxia Min
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Fudi Wang
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
- School of Public Health, Basic Medical Sciences, The First Affiliated Hospital, Hengyang Medical School,
University of South China, Hengyang, China
- School of Public Health, School of Basic Medical Sciences, The First Affiliated Hospital,
Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
21
|
Snell-Rood EC, Kjaer SJ, Marek-Spartz M, Devitz AC, Jansa SA. Pronounced declines in heavy metal burdens of Minnesotan mammals over the last century. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:52473-52484. [PMID: 39150665 PMCID: PMC11374866 DOI: 10.1007/s11356-024-34667-y] [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] [Received: 02/21/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
Humans have drastically altered the ecology of heavy metals, which can have negative effects on animal development and neural functioning. Many species have shown the ability to adapt to anthropogenic increases in metal pollution, but such evolutionary responses will depend on the extent of metal variation over space and time. For terrestrial vertebrates, it is unclear how metal exposure has changed over time: some studies suggest metal content peaked with the enactment of policies controlling lead emissions, while other studies suggest metal levels peaked at least a century earlier. We used 162 specimens of four mammal species (a mouse, shrew, bat, and squirrel) to ask how metal content of the fur and skin has changed over a 90-year time period, and impacts on individual performance (body size and cranial capacity). Using ICP-MS, we show that for lead, cadmium, copper, and chromium, there were significant declines in metal content in mammal tissue over the 90-year time period, with lead levels five times lower now than in the early 1900s. Importantly, metal content began to drop well before the pollution regulation of the 1970s. Effects of time greatly outweighed any effects of an individual living near a human population center. Surprisingly, there were no effects of body metal content on body size, and only manganese was negatively related to relative cranial capacity. Taken together, these results suggest that present day populations of mammals are experiencing levels of heavy metal exposure that are less stressful than they were 100 years ago. In addition, temporal decreases in metal loads likely partly reflect global patterns of pollution decline that affect atmospheric metal deposition rather than local point sources of exposure.
Collapse
Affiliation(s)
- Emilie C Snell-Rood
- Department Ecology, Evolution and Behavior, University of Minnesota, Twin Cities, 1479 Gortner Ave, Gortner 140, St Paul, MN, 55108, USA.
| | - Savannah J Kjaer
- Department Ecology, Evolution and Behavior, University of Minnesota, Twin Cities, 1479 Gortner Ave, Gortner 140, St Paul, MN, 55108, USA
| | - Mary Marek-Spartz
- Department Ecology, Evolution and Behavior, University of Minnesota, Twin Cities, 1479 Gortner Ave, Gortner 140, St Paul, MN, 55108, USA
| | - Amy-Charlotte Devitz
- Department Ecology, Evolution and Behavior, University of Minnesota, Twin Cities, 1479 Gortner Ave, Gortner 140, St Paul, MN, 55108, USA
| | - Sharon A Jansa
- Department Ecology, Evolution and Behavior, University of Minnesota, Twin Cities, 1479 Gortner Ave, Gortner 140, St Paul, MN, 55108, USA
| |
Collapse
|
22
|
Friedman A, Schildroth S, Fruh V, Krengel MH, Tripodis Y, Placidi D, White RF, Lucchini RG, Smith DR, Wright RO, Horton MK, Claus Henn B. Sex-specific associations of a ferroalloy metal mixture with motor function in Italian adolescents. Environ Epidemiol 2024; 8:e321. [PMID: 39022189 PMCID: PMC11254121 DOI: 10.1097/ee9.0000000000000321] [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: 11/28/2023] [Accepted: 06/11/2024] [Indexed: 07/20/2024] Open
Abstract
Background Motor function is critical for children's health, yet remains an understudied neurodevelopmental domain. Exposure to metals has been linked with motor function, but no study has examined the joint effects of metal mixtures. Methods We evaluated cross-sectional associations between a metal mixture and motor function among 569 adolescents (10-14 years old) living near the ferroalloy industry. Concentrations of blood lead, hair manganese, hair copper, and hair chromium were quantified using inductively coupled plasma mass spectrometry. Neuropsychologists administered multiple fine motor function assessments: pursuit aiming, finger tapping, visual reaction time (VRT), and subtests from the Luria Nebraska battery. We estimated associations between motor function and the metal mixture using quantile-based g-computation and multivariable linear regression, adjusting for child age, sex, and socioeconomic status. We explored sex-specific associations in stratified models. Results Associations between the metal mixture and motor function were mostly null but were modified by sex. We observed a beneficial association among females: a quartile increase in all metals in the mixture was associated with a 2.6% faster average response time on the VRT (95% confidence interval [CI] = -4.7%, -0.5%), driven by Cu and Cr. In contrast, this association was adverse among males (ß = 1.5% slower response time [95% CI = -0.7%, 3.9%]), driven by Cu and Mn. Conclusions Results suggest that males may be more susceptible to the adverse effects of metal exposure on motor function during adolescence than females. Future studies, particularly prospective study designs, are warranted to further understand the associations of metal mixtures with motor function.
Collapse
Affiliation(s)
- Alexa Friedman
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts
| | - Samantha Schildroth
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts
| | - Victoria Fruh
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts
| | - Maxine H. Krengel
- Department of Neurology, Boston University Medical School, Boston, Massachusetts
| | - Yorghos Tripodis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Donatella Placidi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Roberta F. White
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts
- Department of Neurology, Boston University Medical School, Boston, Massachusetts
| | - Roberto G. Lucchini
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
- Department of Environmental Health Sciences, School of Public Health, Florida International University, Miami, Florida
| | - Donald R. Smith
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, California
| | - Robert O. Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Megan K. Horton
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Birgit Claus Henn
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts
| |
Collapse
|
23
|
Zhou X, Azimi M, Handin N, Riselli A, Vora B, Chun E, Yee SW, Artursson P, Giacomini KM. Proteomic Profiling Reveals Age-Related Changes in Transporter Proteins in the Human Blood-Brain Barrier. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.604313. [PMID: 39091855 PMCID: PMC11291171 DOI: 10.1101/2024.07.26.604313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The Blood-Brain Barrier (BBB) is a crucial, selective barrier that regulates the entry of molecules including nutrients, environmental toxins, and therapeutic medications into the brain. This function relies heavily on brain endothelial cell proteins, particularly transporters and tight junction proteins. The BBB continues to develop postnatally, adapting its selective barrier function across different developmental phases, and alters with aging and disease. Here we present a global proteomics analysis focused on the ontogeny and aging of proteins in human brain microvessels (BMVs), predominantly composed of brain endothelial cells. Our proteomic profiling quantified 6,223 proteins and revealed possible age-related alteration in BBB permeability due to basement membrane component changes through the early developmental stage and age-dependent changes in transporter expression. Notable changes in expression levels were observed with development and age in nutrient transporters and transporters that play critical roles in drug disposition. This research 1) provides important information on the mechanisms that drive changes in the metabolic content of the brain with age and 2) enables the creation of physiologically based pharmacokinetic models for CNS drug distribution across different life stages.
Collapse
Affiliation(s)
- Xujia Zhou
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Mina Azimi
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Niklas Handin
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Andrew Riselli
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Bianca Vora
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Eden Chun
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| |
Collapse
|
24
|
Ouisselsat M, El Maouaki A, Maidoumi S, François Y, Pineau A, Sedki A. Assessment of Essential and Toxic Element Levels in the Toenails of Children with Autism Spectrum Disorder. Biol Trace Elem Res 2024:10.1007/s12011-024-04319-w. [PMID: 39042314 DOI: 10.1007/s12011-024-04319-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024]
Abstract
Autism spectrum disorder (ASD) has become a global public health concern, impacting the quality of life. The question of gene-environment interaction in the emergence of ASD remains a subject of ongoing debate, and exploring its pathophysiology is thoroughly related to metals as a risk factor. Therefore, this study aims to assess the levels of toxic (Al, Cd, Hg, and Pb) and essential (Cr, Mn, Fe, Ni, Cu, Zn, and Se) elements in toenail samples collected in children with ASD and neurotypical children, by ICP-MS. Parallelly, we will discuss the use of toenails as an exposure indicator. The study involved 208 children aged 3 to 14 from Marrakech, Morocco. One hundred two were diagnosed with ASD and 106 were neurotypical children. Significant statistical differences in the concentration of Cr, Mn, and Fe were documented between the two groups. Higher levels of Pb in toenails compared to reference values have been reported. No association was established between concentrations of elements and age. Spearman correlation coefficients revealed a significantly different pattern of mutual dependence for toxic and essential elements between the two groups. The strongest positive correlations were found in the neurotypical group (Fe-Mn (ρ = 0.750), and Se-Zn (ρ = 0.800)). These results provide additional, although inconclusive, evidence on the probable role of element disturbance in the pathogenesis of ASD. Further studies should be performed to explore other nutritional, cultural, sociodemographic, environmental, and methodological factors that may impact the levels of these elements in the nails and their possible correlation with the incidence of ASD.
Collapse
Affiliation(s)
- Mariam Ouisselsat
- Laboratory of Water, Biodiversity and Climate Change, Department of Biology, Faculty of Sciences - Semlalia, Cadi Ayyad University, Bd Moulay Abdellah BP, 2390-40001, Marrakech, Morocco.
| | - Amal El Maouaki
- Laboratory of Clinical, Experimental and Environmental Neurosciences, Faculty of Medicine and Pharmacy, Cadi Ayyad University, 274, 40000, Marrakech, Morocco
| | - Sana Maidoumi
- Laboratory of Water, Biodiversity and Climate Change, Department of Biology, Faculty of Sciences - Semlalia, Cadi Ayyad University, Bd Moulay Abdellah BP, 2390-40001, Marrakech, Morocco
- Higher Institute of Nursing Professions and Health Techniques, Rue Abdelouahab Derraq, 40000, Marrakech, Morocco
| | - Yannick François
- Mineral Element Dosing Centre, UFR of Pharmaceutical and Biological Sciences, University of Nantes, 9, Rue Bias, 44035 - 44000, Nantes Cedex 1, France
| | - Alain Pineau
- Mineral Element Dosing Centre, UFR of Pharmaceutical and Biological Sciences, University of Nantes, 9, Rue Bias, 44035 - 44000, Nantes Cedex 1, France
| | - Azeddine Sedki
- Laboratory of Water, Biodiversity and Climate Change, Department of Biology, Faculty of Sciences - Semlalia, Cadi Ayyad University, Bd Moulay Abdellah BP, 2390-40001, Marrakech, Morocco
| |
Collapse
|
25
|
Almatrafi AM, Alayoubi AM, Alluqmani M, Hashmi JA, Basit S. Exome Sequence Analysis to Characterize Undiagnosed Family Segregating Motor Impairment and Dystonia. J Clin Med 2024; 13:4252. [PMID: 39064292 PMCID: PMC11278008 DOI: 10.3390/jcm13144252] [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: 07/01/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Background: Hypermanganesemia with dystonia 1 (HMNDYT1) is a rare genetic disorder characterized by elevated blood manganese levels. This condition is associated with polycythemia, motor neurodegeneration with extrapyramidal features, and hepatic dysfunction, which can progress to cirrhosis in some patients. Materials and Methods: In this study, a consanguineous Saudi family with two affected individuals exhibiting symptoms of severe motor impairment, spastic paraparesis, postural instability, and dystonia was studied. Clinical and radiographic evaluations were conducted on the affected individuals. Whole exome sequencing (WES) was performed to diagnose the disease and to determine the causative variant underlying the phenotype. Moreover, Sanger sequencing was used for validation and segregation analysis of the identified variant. Bioinformatics tools were utilized to predict the pathogenicity of candidate variants based on ACMG criteria. Results: Exome sequencing detected a recurrent homozygous missense variant (c.266T>C; p.L89P) in exon 1 of the SLC30A10 gene. Sanger sequencing was employed to validate the segregation of the discovered variant in all available family members. Bioinformatics tools predicted that the variant is potentially pathogenic. Moreover, conservation analysis showed that the variant is highly conserved in vertebrates. Conclusions: This study shows that exome sequencing is instrumental in diagnosing undiagnosed neurodevelopmental disorders. Moreover, this study expands the mutation spectrum of SLC30A10 in distinct populations.
Collapse
Affiliation(s)
- Ahmad M. Almatrafi
- Department of Biology, College of Science, Taibah University, Medina 42353, Saudi Arabia
- Center for Genetics and Inherited Diseases, Taibah University, Madinah 42353, Saudi Arabia;
| | - Abdulfatah M. Alayoubi
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia;
| | - Majed Alluqmani
- Department of Neurology, College of Medicine, Taibah University, Medina 42353, Saudi Arabia;
| | - Jamil A. Hashmi
- Center for Genetics and Inherited Diseases, Taibah University, Madinah 42353, Saudi Arabia;
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia;
| | - Sulman Basit
- Center for Genetics and Inherited Diseases, Taibah University, Madinah 42353, Saudi Arabia;
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia;
| |
Collapse
|
26
|
Stojsavljević A, Jagodić J, Perović T, Manojlović D, Pavlović S. Changes of Target Essential Trace Elements in Multiple Sclerosis: A Systematic Review and Meta-Analysis. Biomedicines 2024; 12:1589. [PMID: 39062163 PMCID: PMC11274787 DOI: 10.3390/biomedicines12071589] [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: 05/28/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
(1) Background: Multiple sclerosis (MS) is a chronic, complex, and demyelinating disease closely associated with altered levels of trace elements. Although the first studies into the role of trace elements in MS were published in the 1970s, for five decades it has remained unknown whether trace elements can be part of this heterogeneous neurological disease. (2) Materials and methods: To drive toward at a potential solution, we conducted a systematic review and meta-analysis to elucidate whether there were differences in circulating levels of neurologically important essential trace elements (Zn, Fe, Co, Cu, Mn, and Se) between MS cases and controls. (3) Results: This study revealed significantly lower serum/plasma Zn and Fe levels and higher Cu levels in MS-affected individuals compared to controls. At the same time, no significant differences were found between the MS cases and controls regarding their serum/plasma levels of Co, Mn, or Se. Thus, the loss of Fe and Zn should be considered in supplementation/nutrition strategies for MS patients. On the other hand, since high serum Cu levels indicate a burden on the bloodstreams of MS patients, Cu should be excluded from mineral supplement strategies. Furthermore, all three trace elements (Fe, Zn, and Cu) should be considered from an etiological point of view, and, most importantly, their levels in the bloodstreams of MS patients should be monitored. (4) Conclusions: This study highlights the way for personalized and targeted strategies in the management of MS.
Collapse
Affiliation(s)
- Aleksandar Stojsavljević
- Innovation Center, Faculty of Chemistry, University of Belgrade, Studentski Trg 12-16, 11000 Belgrade, Serbia
| | - Jovana Jagodić
- Faculty of Chemistry, University of Belgrade, 11000 Belgrade, Serbia; (J.J.); (D.M.)
| | - Tatjana Perović
- Psychiatric Hospital, University Medical Center Zvezdara, 11000 Belgrade, Serbia;
- Serbian RE&CBT Centre, 11000 Belgrade, Serbia
| | - Dragan Manojlović
- Faculty of Chemistry, University of Belgrade, 11000 Belgrade, Serbia; (J.J.); (D.M.)
| | - Slađan Pavlović
- Institute for Biological Research “Siniša Stanković”-National Institute of the Republic of Serbia, University of Belgrade, 11108 Belgrade, Serbia;
| |
Collapse
|
27
|
Chen N, Zhou H, He B, Peng S, Ding F, Liu QH, Ma Z, Liu W, Xu B. Melatonin promotes cell cycle progression of neural stem cells subjected to manganese via Nurr1. ENVIRONMENTAL TOXICOLOGY 2024; 39:3883-3896. [PMID: 38563506 DOI: 10.1002/tox.24258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/04/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
Excessive exposure to manganese (Mn) through drinking water and food during pregnancy significantly heightens the likelihood of neurodevelopmental damage in offspring. Multiple studies have indicated that melatonin (Mel) may help to relieve neurodevelopmental disorders caused by Mn, but potential mechanisms underlying this effect require further exploration. Here, we utilized primary neural stem cells (NSCs) as a model to elucidate the molecular mechanism underlying the protective function of Mel on Mn-induced cell proliferation dysfunction and cycle arrest. Our results showed that Mn disrupted the cell cycle in NSCs by suppressing positive regulatory proteins (CDK2, Cyclin A, Cyclin D1, and E2F1) and enhancing negative ones (p27KIP1 and p57KIP2), leading to cell proliferation dysfunction. Mel inhibited the Mn-dependent changes to these proteins and the cell cycle through nuclear receptor-related protein 1 (Nurr1), thus alleviating the proliferation dysfunction. Knockdown of Nurr1 using lentivirus-expressed shRNA in NSCs resulted in a diminished protective effect of Mel. We concluded that Mel mitigated Mn-induced proliferation dysfunction and cycle arrest in NSCs through Nurr1.
Collapse
Affiliation(s)
- Nan Chen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Han Zhou
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Bin He
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Sen Peng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Feng Ding
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Qi-Hao Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Zhuo Ma
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
- Key laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, PR China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
- Key laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, PR China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
- Key laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, PR China
| |
Collapse
|
28
|
Totten MS, Howell JM, Tomberlin JA, Erikson KM. Relationship Between a High-Fat Diet, Reduced Mobility, and Trace Element Overload in the Olfactory Bulbs of C57BL/6J and DBA/2J Mice. Biol Trace Elem Res 2024; 202:3215-3224. [PMID: 37864044 DOI: 10.1007/s12011-023-03911-w] [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: 07/08/2023] [Accepted: 10/06/2023] [Indexed: 10/22/2023]
Abstract
The dysregulation of trace elements in the brain, which can be caused by genetic or environmental factors, has been associated with disease and compromised mobility. Research regarding trace elements and motor function has focused mainly on the basal ganglia, but few studies have examined the olfactory bulb in this context. Diets high in fat have been shown to have consequences of dysregulated iron and manganese in the brain and disrupted motor activity. The aim of our study was to examine the relationship between mobility and trace element disruption in the olfactory bulb in male and female C57BL/6J and DBA/2J mice fed a high-fat diet. Mobility was significantly reduced in male C57BL/6Js, but the correlation between iron and manganese in the olfactory bulb with velocity, distance travelled, and habituation was not statistically significant. However, there appears to be an overall pattern of a high-fat diet having a statistically significant impact individually on elevated iron and manganese in the olfactory bulb, reduced velocity, reduced distance travelled, and reduced habituation mainly in the male C57BL/6J strain. We found similar trends within the scientific literature to suggest that dysregulated trace element status in the olfactory bulb may be related to motor function in both humans and animals and that males may be more susceptible to the negative outcomes. Our findings contribute new information regarding the impact of diet on the brain, behavior, and potential connection between trace element dysregulation in the olfactory bulb with mobility.
Collapse
Affiliation(s)
- Melissa S Totten
- Department of Chemistry and Physics, Salem College, Winston-Salem, NC, USA.
| | - Jenna M Howell
- Department of Chemistry and Physics, Salem College, Winston-Salem, NC, USA
| | | | - Keith M Erikson
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC, USA
| |
Collapse
|
29
|
Wang T, Xie TN, Shi JH, Zhang WJ. Dietary fructose regulates hepatic manganese homeostasis in female mice. Heliyon 2024; 10:e33278. [PMID: 39022091 PMCID: PMC11253509 DOI: 10.1016/j.heliyon.2024.e33278] [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: 03/06/2024] [Revised: 05/31/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
Arginase, an enzyme dependent on manganese (Mn), plays a crucial role in the production of urea and processing of ammonia in the liver. Previous studies have shown that overconsumption of fructose disrupts Mn homeostasis in the liver of male mice. However, the potential sex-specific differences in the impact of fructose on hepatic Mn homeostasis remain uncertain. In this study, we provide evidence that heightened fructose intake disrupts liver Mn homeostasis in female mice. Elevated fructose exposure led to a reduction in liver Mn levels, resulting in decreased arginase and manganese superoxide dismutase (Mn-SOD) activity in the liver of female mice. The underlying mechanism involves the upregulation of carbohydrate-responsive element binding protein (ChREBP) expression and the Mn exporting gene Slc30a10 in the liver in response to fructose consumption. In summary, our findings support the involvement of fructose in liver Mn metabolism via the ChREBP/Slc30a10 pathway in female mice, and indicate that there is no disparity in the impact of fructose on hepatic Mn homeostasis between sexes.
Collapse
Affiliation(s)
- Ting Wang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Tie-Ning Xie
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Jian-Hui Shi
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Weiping J. Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| |
Collapse
|
30
|
Sarawi WS, Attia HA, Alomar HA, Alhaidar R, Rihan E, Aldurgham N, Ali RA. The protective role of sesame oil against Parkinson's-like disease induced by manganese in rats. Behav Brain Res 2024; 465:114969. [PMID: 38548024 DOI: 10.1016/j.bbr.2024.114969] [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: 12/07/2023] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/14/2024]
Abstract
Chronic exposure to manganese (Mn) results in motor dysfunction, biochemical and pathological alterations in the brain. Oxidative stress, inflammation, and dysfunction of dopaminergic and GABAergic systems stimulate activating transcription factor-6 (ATF-6) and protein kinase RNA-like ER kinase (PERK) leading to apoptosis. This study aimed to investigate the protective effect of sesame oil (SO) against Mn-induced neurotoxicity. Rats received 25 mg/kg MnCl2 and were concomitantly treated with 2.5, 5, or 8 ml/kg of SO for 5 weeks. Mn-induced motor dysfunction was indicated by significant decreases in the time taken by rats to fall during the rotarod test and in the number of movements observed during the open field test. Also, Mn resulted in neuronal degeneration as observed by histological staining. The striatal levels of lipid peroxides and reduced glutathione (oxidative stress markers), interleukin-6 and tumor necrosis factor-α (inflammatory markers) were significantly elevated. Mn significantly reduced the levels of dopamine and Bcl-2, while GABA, PERK, ATF-6, Bax, and caspase-3 were increased. Interestingly, all SO doses, especially at 8 ml/kg, significantly improved locomotor activity, biochemical deviations and reduced neuronal degeneration. In conclusion, SO may provide potential therapeutic benefits in enhancing motor performance and promoting neuronal survival in individuals highly exposed to Mn.
Collapse
Affiliation(s)
- Wedad S Sarawi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia.
| | - Hala A Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
| | - Hatun A Alomar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
| | - Rawan Alhaidar
- College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
| | - Esraa Rihan
- College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
| | - Nora Aldurgham
- College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
| | - Rehab A Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
| |
Collapse
|
31
|
Zhang PP, Ding GC, Tao CY, Zhang L, Wang YX, Yuan QY, Zhang SM, Wang LP. Levels of trace metals and their impact on oocyte: A review. Taiwan J Obstet Gynecol 2024; 63:307-311. [PMID: 38802192 DOI: 10.1016/j.tjog.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2024] [Indexed: 05/29/2024] Open
Abstract
Trace metals play a vital role in a variety of biological processes, but excessive amounts can be toxic and are receiving increasing attention. Trace metals in the environment are released from natural sources, such as rock weathering, volcanic eruptions, and other human activities, such as industrial emissions, mineral extraction, and vehicle exhaust. Lifestyle, dietary habits and environmental quality are the main sources of human exposure to trace metals, which play an important role in inducing human reproductive infertility. The purpose of this review is to summarize the distribution of various trace metals in oocyte and to identify the trace metals that may cause oocyte used in the design and execution of toxicological studies.
Collapse
Affiliation(s)
- Ping-Ping Zhang
- Yangzhou Maternity and Child Health Care Hospital, Yangzhou, Jiangsu, China
| | - Gui-Chun Ding
- Yangzhou Maternity and Child Health Care Hospital, Yangzhou, Jiangsu, China
| | - Chen-Yue Tao
- School of Nursing·School of Public Health, Yangzhou University, Yangzhou, Jiangsu, China
| | - Lei Zhang
- Yangzhou Maternity and Child Health Care Hospital, Yangzhou, Jiangsu, China
| | - Yi-Xiong Wang
- Yangzhou Maternity and Child Health Care Hospital, Yangzhou, Jiangsu, China
| | | | - Sheng-Min Zhang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China.
| | - Li-Ping Wang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
32
|
McBride DE, Bhattacharya A, Sucharew H, Brunst KJ, Barnas M, Cox C, Altman L, Hilbert TJ, Burkle J, Westneat S, Martin KV, Parsons PJ, Praamsma ML, Palmer CD, Kannan K, Smith DR, Wright R, Amarasiriwardena C, Dietrich KN, Cecil KM, Haynes EN. Child and Adolescent Manganese Biomarkers and Adolescent Postural Balance in Marietta CARES Cohort Participants. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:57010. [PMID: 38780454 PMCID: PMC11114102 DOI: 10.1289/ehp13381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 03/04/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Manganese (Mn) plays a significant role in both human health and global industries. Epidemiological studies of exposed populations demonstrate a dose-dependent association between Mn and neuromotor effects ranging from subclinical effects to a clinically defined syndrome. However, little is known about the relationship between early life Mn biomarkers and adolescent postural balance. OBJECTIVES This study investigated the associations between childhood and adolescent Mn biomarkers and adolescent postural balance in participants from the longitudinal Marietta Communities Actively Researching Exposures Study (CARES) cohort. METHODS Participants were recruited into CARES when they were 7-9 y old, and reenrolled at 13-18 years of age. At both time points, participants provided samples of blood, hair, and toenails that were analyzed for blood Mn and lead (Pb), serum cotinine, hair Mn, and toenail Mn. In adolescence, participants completed a postural balance assessment. Greater sway indicates postural instability (harmful effect), whereas lesser sway indicates postural stability (beneficial effect). Multivariable linear regression models were conducted to investigate the associations between childhood and adolescent Mn biomarkers and adolescent postural balance adjusted for age, sex, height-weight ratio, parent/caregiver intelligence quotient, socioeconomic status, blood Pb, and serum cotinine. RESULTS CARES participants who completed the adolescent postural balance assessment (n = 123 ) were 98% White and 54% female and had a mean age of 16 y (range: 13-18 y). In both childhood and adolescence, higher Mn biomarker concentrations were significantly associated with greater adolescent sway measures. Supplemental analyses revealed sex-specific associations; higher childhood Mn biomarker concentrations were significantly associated with greater sway in females compared with males. DISCUSSION This study found childhood and adolescent Mn biomarkers were associated with subclinical neuromotor effects in adolescence. This study demonstrates postural balance as a sensitive measure to assess the association between Mn biomarkers and neuromotor function. https://doi.org/10.1289/EHP13381.
Collapse
Affiliation(s)
- Danielle E. McBride
- Department of Epidemiology and Environmental Health, College of Public Health, University of Kentucky, Lexington, Kentucky, USA
| | - Amit Bhattacharya
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Heidi Sucharew
- Department of Emergency Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kelly J. Brunst
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Mary Barnas
- Department of Psychology, Marietta College, Marietta, Ohio, USA
| | - Cyndy Cox
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lorenna Altman
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Timothy J. Hilbert
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jeff Burkle
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Susan Westneat
- Department of Epidemiology and Environmental Health, College of Public Health, University of Kentucky, Lexington, Kentucky, USA
| | - Kaitlin Vollet Martin
- Department of Epidemiology and Environmental Health, College of Public Health, University of Kentucky, Lexington, Kentucky, USA
| | - Patrick J. Parsons
- Division of Environmental Health Sciences, Wadsworth Center, New York State Department of Health, Albany, New York, USA
- Department of Environmental Health Sciences, School of Public Health, University at Albany, State University of New York, Rensselaer, New York, USA
| | - Meredith L. Praamsma
- Division of Environmental Health Sciences, Wadsworth Center, New York State Department of Health, Albany, New York, USA
- Department of Environmental Health Sciences, School of Public Health, University at Albany, State University of New York, Rensselaer, New York, USA
| | - Christopher D. Palmer
- Division of Environmental Health Sciences, Wadsworth Center, New York State Department of Health, Albany, New York, USA
- Department of Environmental Health Sciences, School of Public Health, University at Albany, State University of New York, Rensselaer, New York, USA
| | - Kurunthachalam Kannan
- Division of Environmental Health Sciences, Wadsworth Center, New York State Department of Health, Albany, New York, USA
- Department of Environmental Health Sciences, School of Public Health, University at Albany, State University of New York, Rensselaer, New York, USA
| | - Donald R. Smith
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, California, USA
| | - Robert Wright
- Environmental Medicine and Public Health, Mount Sinai School of Medicine, New York, New York, USA
| | - Chitra Amarasiriwardena
- Environmental Medicine and Public Health, Mount Sinai School of Medicine, New York, New York, USA
| | - Kim N. Dietrich
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kim M. Cecil
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
- Department of Radiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Erin N. Haynes
- Department of Epidemiology and Environmental Health, College of Public Health, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
33
|
Alba-González A, Dragomir EI, Haghdousti G, Yáñez J, Dadswell C, González-Méndez R, Wilson SW, Tuschl K, Folgueira M. Manganese Overexposure Alters Neurogranin Expression and Causes Behavioral Deficits in Larval Zebrafish. Int J Mol Sci 2024; 25:4933. [PMID: 38732149 PMCID: PMC11084468 DOI: 10.3390/ijms25094933] [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: 03/14/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Manganese (Mn), a cofactor for various enzyme classes, is an essential trace metal for all organisms. However, overexposure to Mn causes neurotoxicity. Here, we evaluated the effects of exposure to Mn chloride (MnCl2) on viability, morphology, synapse function (based on neurogranin expression) and behavior of zebrafish larvae. MnCl2 exposure from 2.5 h post fertilization led to reduced survival (60%) at 5 days post fertilization. Phenotypical changes affected body length, eye and olfactory organ size, and visual background adaptation. This was accompanied by a decrease in both the fluorescence intensity of neurogranin immunostaining and expression levels of the neurogranin-encoding genes nrgna and nrgnb, suggesting the presence of synaptic alterations. Furthermore, overexposure to MnCl2 resulted in larvae exhibiting postural defects, reduction in motor activity and impaired preference for light environments. Following the removal of MnCl2 from the fish water, zebrafish larvae recovered their pigmentation pattern and normalized their locomotor behavior, indicating that some aspects of Mn neurotoxicity are reversible. In summary, our results demonstrate that Mn overexposure leads to pronounced morphological alterations, changes in neurogranin expression and behavioral impairments in zebrafish larvae.
Collapse
Affiliation(s)
- Anabel Alba-González
- Department of Biology, Faculty of Sciences, University of A Coruña, 15008 A Coruña, Spain; (A.A.-G.); (J.Y.)
- Centro Interdisciplinar de Química y Biología, (CICA), University of A Coruña, 15071 A Coruña, Spain
| | - Elena I. Dragomir
- Department of Cell and Developmental, University College London, London, WC1E 6BT, UK; (E.I.D.); (G.H.); (S.W.W.)
| | - Golsana Haghdousti
- Department of Cell and Developmental, University College London, London, WC1E 6BT, UK; (E.I.D.); (G.H.); (S.W.W.)
| | - Julián Yáñez
- Department of Biology, Faculty of Sciences, University of A Coruña, 15008 A Coruña, Spain; (A.A.-G.); (J.Y.)
- Centro Interdisciplinar de Química y Biología, (CICA), University of A Coruña, 15071 A Coruña, Spain
| | - Chris Dadswell
- School of Life Sciences, University of Sussex, Brighton, BN1 9QJ, UK; (C.D.); (R.G.-M.)
| | - Ramón González-Méndez
- School of Life Sciences, University of Sussex, Brighton, BN1 9QJ, UK; (C.D.); (R.G.-M.)
| | - Stephen W. Wilson
- Department of Cell and Developmental, University College London, London, WC1E 6BT, UK; (E.I.D.); (G.H.); (S.W.W.)
| | - Karin Tuschl
- UCL GOSH Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Mónica Folgueira
- Department of Biology, Faculty of Sciences, University of A Coruña, 15008 A Coruña, Spain; (A.A.-G.); (J.Y.)
- Centro Interdisciplinar de Química y Biología, (CICA), University of A Coruña, 15071 A Coruña, Spain
| |
Collapse
|
34
|
Jensen GC, Janis MK, Nguyen HN, David OW, Zastrow ML. Fluorescent Protein-Based Sensors for Detecting Essential Metal Ions across the Tree of Life. ACS Sens 2024; 9:1622-1643. [PMID: 38587931 PMCID: PMC11073808 DOI: 10.1021/acssensors.3c02695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Genetically encoded fluorescent metal ion sensors are powerful tools for elucidating metal dynamics in living systems. Over the last 25 years since the first examples of genetically encoded fluorescent protein-based calcium indicators, this toolbox of probes has expanded to include other essential and non-essential metal ions. Collectively, these tools have illuminated fundamental aspects of metal homeostasis and trafficking that are crucial to fields ranging from neurobiology to human nutrition. Despite these advances, much of the application of metal ion sensors remains limited to mammalian cells and tissues and a limited number of essential metals. Applications beyond mammalian systems and in vivo applications in living organisms have primarily used genetically encoded calcium ion sensors. The aim of this Perspective is to provide, with the support of historical and recent literature, an updated and critical view of the design and use of fluorescent protein-based sensors for detecting essential metal ions in various organisms. We highlight the historical progress and achievements with calcium sensors and discuss more recent advances and opportunities for the detection of other essential metal ions. We also discuss outstanding challenges in the field and directions for future studies, including detecting a wider variety of metal ions, developing and implementing a broader spectral range of sensors for multiplexing experiments, and applying sensors to a wider range of single- and multi-species biological systems.
Collapse
Affiliation(s)
- Gary C Jensen
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Makena K Janis
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Hazel N Nguyen
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Ogonna W David
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Melissa L Zastrow
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
35
|
Moksnes MR, Hansen AF, Wolford BN, Thomas LF, Rasheed H, Simić A, Bhatta L, Brantsæter AL, Surakka I, Zhou W, Magnus P, Njølstad PR, Andreassen OA, Syversen T, Zheng J, Fritsche LG, Evans DM, Warrington NM, Nøst TH, Åsvold BO, Flaten TP, Willer CJ, Hveem K, Brumpton BM. A genome-wide association study provides insights into the genetic etiology of 57 essential and non-essential trace elements in humans. Commun Biol 2024; 7:432. [PMID: 38594418 PMCID: PMC11004147 DOI: 10.1038/s42003-024-06101-z] [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: 05/09/2023] [Accepted: 03/22/2024] [Indexed: 04/11/2024] Open
Abstract
Trace elements are important for human health but may exert toxic or adverse effects. Mechanisms of uptake, distribution, metabolism, and excretion are partly under genetic control but have not yet been extensively mapped. Here we report a comprehensive multi-element genome-wide association study of 57 essential and non-essential trace elements. We perform genome-wide association meta-analyses of 14 trace elements in up to 6564 Scandinavian whole blood samples, and genome-wide association studies of 43 trace elements in up to 2819 samples measured only in the Trøndelag Health Study (HUNT). We identify 11 novel genetic loci associated with blood concentrations of arsenic, cadmium, manganese, selenium, and zinc in genome-wide association meta-analyses. In HUNT, several genome-wide significant loci are also indicated for other trace elements. Using two-sample Mendelian randomization, we find several indications of weak to moderate effects on health outcomes, the most precise being a weak harmful effect of increased zinc on prostate cancer. However, independent validation is needed. Our current understanding of trace element-associated genetic variants may help establish consequences of trace elements on human health.
Collapse
Affiliation(s)
- Marta R Moksnes
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway.
| | - Ailin F Hansen
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Brooke N Wolford
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Laurent F Thomas
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- BioCore-Bioinformatics Core Facility, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Humaira Rasheed
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Division of Medicine and Laboratory Sciences, University of Oslo, Oslo, Norway
| | - Anica Simić
- Department of Chemistry, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Laxmi Bhatta
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Anne Lise Brantsæter
- Department of Food Safety, Division of Climate and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Ida Surakka
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Pål R Njølstad
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
| | - Ole A Andreassen
- NORMENT Centre, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Tore Syversen
- Department of Neuroscience, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Bristol, UK
| | - Lars G Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - David M Evans
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
| | - Nicole M Warrington
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
| | - Therese H Nøst
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Bjørn Olav Åsvold
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Levanger, Norway
- Department of Endocrinology, Clinic of Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Trond Peder Flaten
- Department of Chemistry, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Cristen J Willer
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Kristian Hveem
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Levanger, Norway
| | - Ben M Brumpton
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway.
- HUNT Research Centre, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Levanger, Norway.
- Clinic of Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway.
| |
Collapse
|
36
|
Rosa MJ, Foppa Pedretti N, Goldson B, Mathews N, Merced-Nieves F, Xhani N, Bosquet Enlow M, Gershon R, Ho E, Huddleston K, Wright RO, Wright RJ, Colicino E. Integrating Data Across Multiple Sites in the Northeastern United States to Examine Associations Between a Prenatal Metal Mixture and Child Cognition. Am J Epidemiol 2024; 193:606-616. [PMID: 37981721 PMCID: PMC11484645 DOI: 10.1093/aje/kwad233] [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: 04/20/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023] Open
Abstract
We applied a novel hierarchical Bayesian weighted quantile sum (HBWQS) regression to combine data across 3 study sites to examine associations between prenatal exposure to metals and cognitive functioning in childhood. Data from 326 mother-child dyads enrolled in an ongoing cohort study, the Programming of Intergenerational Stress Mechanisms (PRISM) Study, based in New York, New York (recruitment in 2013-2020) and Boston, Massachusetts (recruitment 2011-2013), and the First Thousand Days of Life (FTDL) cohort study (recruitment 2012-2019), based in northern Virginia, were used. Arsenic, cadmium, manganese, lead, and antimony were measured in urine collected during pregnancy. Cognitive functioning was assessed in children aged 3-11 years using the National Institutes of Health Toolbox Cognition Battery. The HBWQS regression showed a negative association between the urinary metal mixture and the Cognition Early Childhood Composite Score in the PRISM New York City (β = -3.67, 95% credible interval (CrI): -7.61, -0.01) and FTDL (β = -3.76, 95% CrI: -7.66, -0.24) samples, with a similar trend in the PRISM Boston sample (β = -3.24, 95% CrI: -6.77, 0.144). We did not detect these associations in traditionally pooled models. HBWQS regression allowed us to account for site heterogeneity and detect associations between prenatal metal-mixture exposure and cognitive outcomes in childhood. Given the ubiquity of metals exposure, interventions aimed at reducing prenatal exposure may improve cognitive outcomes in children. This article is part of a Special Collection on Environmental Epidemiology.
Collapse
Affiliation(s)
- Maria José Rosa
- Correspondence to Dr. Maria José Rosa, Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1057, New York, NY 10029 (e-mail: )
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Chen Z, Ao C, Liu Y, Yang Y, Liu Y, Ming Q, Li C, Zhao H, Ban J, Li J. Manganese induces oxidative damage in the hippocampus by regulating the expression of oxidative stress-related genes via modulation of H3K18 acetylation. ENVIRONMENTAL TOXICOLOGY 2024; 39:2240-2253. [PMID: 38129942 DOI: 10.1002/tox.24102] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 10/25/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023]
Abstract
Prolonged exposure to manganese (Mn) contributes to hippocampal Mn accumulation, which leads to neurodegenerative diseases called manganese poisoning. However, the underlying molecular mechanisms remain unclear and there are no ideal biomarkers. Oxidative stress is the essential mechanisms of Mn-related neurotoxicity. Furthermore, histone acetylation has been identified as being engaged in the onset and development of neurodegenerative diseases. Therefore, the work aims to understand the molecular mechanisms of oxidative damage in the hippocampus due to Mn exposure from the aspect of histone acetylation modification and to assess whether H3K18 acetylation (H3K18ac) modification level in peripheral blood reflect Mn-induced oxidative damage in the hippocampus. Here, we randomly divided 60 male rats into four groups and injected them intraperitoneally with sterile pure water and MnCl2 ⋅4H2 O (5, 10, and 15 mg/kg) for 16 weeks, 5 days a week, once a day. The data confirmed that Mn exposure down-regulated superoxide dismutase activity and glutathione level as well as up-regulated malondialdehyde level in the hippocampus and plasma, and that there was a positive correlation between these indicators in the hippocampus and plasma. Besides, we noted that Mn treatment upregulated H3K18ac modification levels in the hippocampus and peripheral blood and that H3K18ac modification levels correlated with oxidative stress. Further studies demonstrated that Mn treatment decreased the amounts of H3K18ac enrichment in the manganese superoxide dismutase (SOD2) and glutathione transferase omega 1 (GSTO1) gene promoter regions, contributing to oxidative damage in the hippocampus. In short, our results demonstrate that Mn induces oxidative damage in the hippocampus by inhibiting the expression of SOD2 and GSTO1 genes via modulation of H3K18ac. In assessing Mn-induced hippocampal neurotoxicity, oxidative damage in plasma may reflect hippocampal oxidative damage in Mn-exposed groups.
Collapse
Affiliation(s)
- Zhi Chen
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China
| | - Chunyan Ao
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yan Liu
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yue Yang
- Guiyang Stomatological Hospital, Guiyang, Guizhou, China
| | - Ying Liu
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China
| | - Qian Ming
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China
| | - Changzhe Li
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China
| | - Hua Zhao
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jiaqi Ban
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jun Li
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
38
|
Zarandi AF, Shirkhanloo H, Rakhtshah J. An immobilization of 2-(Aminomethyl) thiazole on multi-walled carbon nanotubes used for rapid extraction of manganese ions in hepatic patients. J Pharm Biomed Anal 2024; 240:115941. [PMID: 38211517 DOI: 10.1016/j.jpba.2023.115941] [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: 10/16/2023] [Revised: 12/23/2023] [Accepted: 12/23/2023] [Indexed: 01/13/2024]
Abstract
A new method based on the immobilization of 2-(Aminomethyl) thiazole on the multi-walled carbon nanotubes (AMTZ@MWCNTs) was used to extract manganese (Mn) in the human blood, serum, and urine samples. First, 20 mg of AMTZ@MWCNTs, 0.2 mL of acetone, and 0.1 g of ionic liquid (IL) were completely mixed and injected into 2.0 mL human samples by a microscale syringe at pH 5.5. After shaking and centrifuging, the Mn ions were extracted and separated through the ultrasound-assisted- ionic liquid-dispersive micro solid-phase extraction (UAS-IL-D-μ-SPE) before being determined by the graphite furnace atomic absorption spectrometry (GF-AAS). According to the results, manganese in the blood of hepatic patients had higher concentrations than healthy people (Aged 25-60, 50 N). The Mn adsorption capacities for the AMTZ@MWCNTs and MWCNTs adsorbents were achieved at 192.5 mg/g and 26.3 mg/g, respectively. In the high enrichment factor (HEF), the limit of detection (LOD), linear range (LR), and mean relative standard division (RSD%) were calculated at 15 ng/L, 0.05-3.8 μg/L, and 2.34, respectively (n = 10). The methodology was validated using certified reference material (CRM) and spiking standard solutions to human samples.
Collapse
Affiliation(s)
- Ali Faghihi Zarandi
- Environmental Health Engineering Research Center, Kerman university of Medical Science, Kerman, Iran; Department of Occupational Hygiene Engineering, Faculty of Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Shirkhanloo
- Research Institute of Petroleum Industry (RIPI), West Entrance Blvd., Olympic Village, Tehran, Iran.
| | - Jamshid Rakhtshah
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| |
Collapse
|
39
|
Guo X, Xu J, Tian Y, Ouyang F, Yu X, Liu J, Yan C, Zhang J. Interaction of prenatal maternal selenium and manganese levels on child neurodevelopmental trajectories-the Shanghai birth cohort study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 915:170095. [PMID: 38224892 DOI: 10.1016/j.scitotenv.2024.170095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/17/2024]
Abstract
OBJECTIVE The fetal brain is particularly plastic, and may be concurrently affected by chemical exposure and malnutritional factors. Selenium is essential for the developing brain, and excess manganese exposure may exert neurotoxic effects. However, few epidemiological studies have evaluated the interaction of manganese and selenium assessed in different prenatal stages on postnatal neurodevelopmental trajectories. METHODS This study contained 1024 mother-child pairs in the Shanghai-birth-cohort study from 2013 to 2016 recruited since early/before pregnancy with complete data on manganese and selenium levels in different prenatal stages and infant neurodevelopmental trajectories. Whole blood manganese and selenium in early pregnancy and around birth were measured by inductively-coupled-plasma-mass-spectrometry (ICP-MS), children's cognitive development was evaluated at 6, 12, and 24 months of age using Age & Stage-Questionnaire (ASQ)-3 and Bayley-III. Multiple linear regression was used to investigate the interaction of prenatal selenium and manganese on neurodevelopmental trajectories. RESULTS The prenatal manganese and selenium levels were 1.82 ± 0.98 μg/dL and 13.53 ± 2.70 μg/dL for maternal blood in early pregnancy, and 5.06 ± 1.67 μg/dL and 11.81 ± 3.35 μg/dL for umbilical cord blood, respectively. Higher prenatal Se levels were associated with better neurocognitive performances or the consistently-high-level trajectory (P < 0.05), with more significant associations observed in early pregnancy than around birth. However, such positive relationships became non-significant or even adverse in high (vs. low) manganese status, and the effect differences between low and high manganese were more significant in early pregnancy. CONCLUSIONS Prenatal Selenium was positively associated with child neurodevelopment, but prenatal high manganese may mitigate such favorable effects. The effects were mainly observed in earlier prenatal stage.
Collapse
Affiliation(s)
- Xiangrong Guo
- The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, China
| | - Jian Xu
- The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, China.
| | - Ying Tian
- MOE-Shanghai Key Lab of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fengxiu Ouyang
- MOE-Shanghai Key Lab of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiaodan Yu
- MOE-Shanghai Key Lab of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Junxia Liu
- MOE-Shanghai Key Lab of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Chonghuai Yan
- MOE-Shanghai Key Lab of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jun Zhang
- MOE-Shanghai Key Lab of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
40
|
Lu W, He J, Wei S, Tang C, Ma X, Li D, Chen H, Zou Y. Circular RNA circRest regulates manganese induced cell apoptosis by targeting the mmu-miR-6914-5p/Ephb3 axis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 344:123395. [PMID: 38266697 DOI: 10.1016/j.envpol.2024.123395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/23/2023] [Accepted: 01/17/2024] [Indexed: 01/26/2024]
Abstract
Overexposure to manganese (Mn) can lead to neurotoxicity, the underlying mechanisms remain incompletely understood. Circular RNAs (circRNAs) have emerged as important regulators in various biological processes. It is plausible that circRNAs may be involved in the biological mechanisms underlying Mn caused neurotoxicity. Here, circRest was downregulated in Mn-exposed mouse neuroblastoma cells (N2a cells) by RNA sequencing and quantitative real-time PCR. When circRest was overexpressed, it led to an increase in cell viability and a decrease in apoptosis following Mn exposure. Conversely, silencing circRest resulted in opposite effects in N2a cells. Further investigation revealed that circRest acts as a mmu-miR-6914-5p sponge, and mmu-miR-6914-5p could bind and inhibit Ephb3, thereby promoting apoptosis in N2a cells. This was confirmed through RNA antisense purification and dual luciferase reporter assays. Additionally, the circRest/mmu-miR-6914-5p/Ephb3 axis may influence memory and learning in mice following Mn exposure. In conclusion, our study uncovers a novel mechanism by which circRest may attenuate Mn caused neurotoxicity via the mmu-miR-6914-5p/Ephb3 axis.
Collapse
Affiliation(s)
- Wenmin Lu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jiacheng He
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Shengtao Wei
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chuanqiao Tang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiaoli Ma
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Danni Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Hao Chen
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Yunfeng Zou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China; Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Nanning, 530021, Guangxi, China.
| |
Collapse
|
41
|
Takemura Mariano MV, Paganotto Leandro L, Gomes KK, Dos Santos AB, de Rosso VO, Dafre AL, Farina M, Posser T, Franco JL. Assessing the disparity: comparative toxicity of Copper in zebrafish larvae exposes alarming consequences of permissible concentrations in Brazil. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:166-184. [PMID: 38073470 DOI: 10.1080/15287394.2023.2290630] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Copper (Cu) is a naturally occurring metal with essential micronutrient properties. However, this metal might also pose increased adverse environmental and health risks due to industrial and agricultural activities. In Brazil, the maximum allowable concentration of Cu in drinking water is 2 mg/L. Despite this standard, the impact of such concentrations on aquatic organisms remains unexplored. This study aimed to evaluate the toxicity of CuSO4 using larval zebrafish at environmentally relevant concentrations. Zebrafish (Danio rerio) larvae at 72 hr post-fertilization (hpf) were exposed to nominal CuSO4 concentrations ranging from 0.16 to 48 mg/L to determine the median lethal concentration (LC50), established at 8.4 mg/L. Subsequently, non-lethal concentrations of 0.16, 0.32, or 1.6 mg/L were selected for assessing CuSO4 -induced toxicity. Morphological parameters, including body length, yolk sac area, and swim bladder area, were adversely affected by CuSO4 exposure, particularly at 1.6 mg/L (3.31 mm ±0.1, 0.192 mm2 ±0.01, and 0.01 mm2 ±0.05, respectively). In contrast, the control group exhibited values of 3.62 mm ±0.09, 0.136 mm2 ±0.013, and 0.3 mm2 ±0.06, respectively. Behavioral assays demonstrated impairments in escape response and swimming capacity, accompanied by increased levels of reactive oxygen species (ROS) and lipid peroxidation. In addition, decreased levels of non-protein thiols and reduced cellular viability were noted. Data demonstrated that exposure to CuSO4 at similar concentrations as those permitted in Brazil for Cu adversely altered morphological, biochemical, and behavioral endpoints in zebrafish larvae. This study suggests that the permissible Cu concentrations in Brazil need to be reevaluated, given the potential enhanced adverse health risks of exposure to environmental metal contamination.
Collapse
Affiliation(s)
- Maria Vitória Takemura Mariano
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
| | - Luana Paganotto Leandro
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
- Department of Molecular Biology and Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Karen Kich Gomes
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
| | - Ana Beatriz Dos Santos
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
| | - Vitor Oliveira de Rosso
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
| | - Alcir Luiz Dafre
- Department of Biochemistry, Center for Biological Sciences, Federal University of Santa Catarina, Santa Catarina, Brazil
| | - Marcelo Farina
- Department of Biochemistry, Center for Biological Sciences, Federal University of Santa Catarina, Santa Catarina, Brazil
| | - Thaís Posser
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
| | - Jeferson Luis Franco
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
| |
Collapse
|
42
|
Latronico T, Rossano R, Miniero DV, Casalino E, Liuzzi GM. Neuroprotective Effect of Resveratrol against Manganese-Induced Oxidative Stress and Matrix Metalloproteinase-9 in an "In Vivo" Model of Neurotoxicity. Int J Mol Sci 2024; 25:2142. [PMID: 38396818 PMCID: PMC10888573 DOI: 10.3390/ijms25042142] [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: 12/24/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Chronic exposure to manganese (Mn) leads to its accumulation in the central nervous system (CNS) and neurotoxicity with not well-known mechanisms. We investigated the involvement of matrix metalloproteinase (MMP)-2 and -9 in Mn neurotoxicity in an in vivo model of rats treated through an intraperitoneal injection, for 4 weeks, with 50 mg/kg of MnCl2 in the presence or in the absence of 30 mg/kg of resveratrol (RSV). A loss of weight was observed in Mn-treated rats compared with untreated and RSV-treated rats. A progressive recovery of body weight was detected in rats co-treated with Mn and RSV. The analysis of brain homogenates indicated that RSV counteracted the Mn-induced increase in MMP-9 levels and reactive oxygen species production as well as the Mn-induced decrease in superoxide dismutase activity and glutathione content. In conclusion, Mn exposure, resulting in MMP-9 induction with mechanisms related to oxidative stress, represents a risk factor for the development of CNS diseases.
Collapse
Affiliation(s)
- Tiziana Latronico
- Department of Biosciences, Biotechnologies and Environment, University of Bari “A. Moro”, 70126 Bari, Italy; (D.V.M.); (G.M.L.)
| | - Rocco Rossano
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy;
| | - Daniela Valeria Miniero
- Department of Biosciences, Biotechnologies and Environment, University of Bari “A. Moro”, 70126 Bari, Italy; (D.V.M.); (G.M.L.)
| | - Elisabetta Casalino
- Department of Veterinary Medicine, University of Bari “A. Moro”, 70010 Bari, Italy;
| | - Grazia Maria Liuzzi
- Department of Biosciences, Biotechnologies and Environment, University of Bari “A. Moro”, 70126 Bari, Italy; (D.V.M.); (G.M.L.)
| |
Collapse
|
43
|
Zeylan M, Senyuz S, Picón-Pagès P, García-Elías A, Tajes M, Muñoz FJ, Oliva B, Garcia-Ojalvo J, Barbu E, Vicente R, Nattel S, Ois A, Puig-Pijoan A, Keskin O, Gursoy A. Shared Proteins and Pathways of Cardiovascular and Cognitive Diseases: Relation to Vascular Cognitive Impairment. J Proteome Res 2024; 23:560-573. [PMID: 38252700 PMCID: PMC10846560 DOI: 10.1021/acs.jproteome.3c00289] [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: 05/12/2023] [Revised: 09/29/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024]
Abstract
One of the primary goals of systems medicine is the detection of putative proteins and pathways involved in disease progression and pathological phenotypes. Vascular cognitive impairment (VCI) is a heterogeneous condition manifesting as cognitive impairment resulting from vascular factors. The precise mechanisms underlying this relationship remain unclear, which poses challenges for experimental research. Here, we applied computational approaches like systems biology to unveil and select relevant proteins and pathways related to VCI by studying the crosstalk between cardiovascular and cognitive diseases. In addition, we specifically included signals related to oxidative stress, a common etiologic factor tightly linked to aging, a major determinant of VCI. Our results show that pathways associated with oxidative stress are quite relevant, as most of the prioritized vascular cognitive genes and proteins were enriched in these pathways. Our analysis provided a short list of proteins that could be contributing to VCI: DOLK, TSC1, ATP1A1, MAPK14, YWHAZ, CREB3, HSPB1, PRDX6, and LMNA. Moreover, our experimental results suggest a high implication of glycative stress, generating oxidative processes and post-translational protein modifications through advanced glycation end-products (AGEs). We propose that these products interact with their specific receptors (RAGE) and Notch signaling to contribute to the etiology of VCI.
Collapse
Affiliation(s)
- Melisa
E. Zeylan
- Computational
Sciences and Engineering, Graduate School of Science and Engineering, Koç University, Istanbul 34450, Türkiye
| | - Simge Senyuz
- Computational
Sciences and Engineering, Graduate School of Science and Engineering, Koç University, Istanbul 34450, Türkiye
| | - Pol Picón-Pagès
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Anna García-Elías
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Marta Tajes
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Francisco J. Muñoz
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Baldomero Oliva
- Laboratory
of Structural Bioinformatics (GRIB), Department of Medicine and Life
Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Jordi Garcia-Ojalvo
- Laboratory
of Dynamical Systems Biology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Eduard Barbu
- Institute
of Computer Science, University of Tartu, Tartu, 50090, Estonia
| | - Raul Vicente
- Institute
of Computer Science, University of Tartu, Tartu, 50090, Estonia
| | - Stanley Nattel
- Department
of Medicine and Research Center, Montreal Heart Institute and Université
de Montréal; Institute of Pharmacology, West German Heart and
Vascular Center, University Duisburg-Essen,
Germany; IHU LIRYC and Fondation Bordeaux Université, Bordeaux 33000, France
| | - Angel Ois
- Department
of Neurology, Hospital Del Mar. Hospital
Del Mar - Medical Research Institute and Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Albert Puig-Pijoan
- Department
of Neurology, Hospital Del Mar. Hospital
Del Mar - Medical Research Institute and Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Ozlem Keskin
- Department
of Chemical and Biological Engineering, Koç University, Istanbul 34450, Türkiye
| | - Attila Gursoy
- Department
of Computer Engineering, Koç University, Istanbul 34450, Türkiye
| |
Collapse
|
44
|
Liu Y, Zhao H, Yang Y, Liu Y, Ao CY, Zeng JM, Ban JQ, Li J. Mechanism by which HDAC3 regulates manganese induced H3K27ac in SH-SY5Y cells and intervention by curcumin. Arch Biochem Biophys 2024; 752:109878. [PMID: 38151197 DOI: 10.1016/j.abb.2023.109878] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/29/2023]
Abstract
Long-term excessive exposure to manganese can impair neuronal function in the brain, but the underlying pathological mechanism remains unclear. Oxidative stress plays a central role in manganese-induced neurotoxicity. Numerous studies have established a strong link between abnormal histone acetylation levels and the onset of various diseases. Histone deacetylase inhibitors and activators, such as TSA and ITSA-1, are often used to investigate the intricate mechanisms of histone acetylation in disease. In addition, recent experiments have provided substantial evidence demonstrating that curcumin (Cur) can act as an epigenetic regulator. Given these findings, this study aims to investigate the mechanisms underlying oxidative damage in SH-SY5Y cells exposed to MnCl2·4H2O, with a particular focus on histone acetylation, and to assess the potential therapeutic efficacy of Cur. In this study, SH-SY5Y cells were exposed to manganese for 24 h, were treated with TSA or ITSA-1, and were treated with or without Cur. The results suggested that manganese exposure, which leads to increased expression of HDAC3, induced H3K27 hypoacetylation, inhibited the transcription of antioxidant genes, decreased antioxidant enzyme activities, and induced oxidative damage in cells. Pretreatment with an HDAC3 inhibitor (TSA) increased the acetylation of H3K27 and the transcription of antioxidant genes and thus slowed manganese exposure-induced cellular oxidative damage. In contrast, an HDAC3 activator (ITSA-1) partially increased manganese-induced cellular oxidative damage, while Cur prevented manganese-induced oxidative damage. In summary, these findings suggest that inhibiting H3K27ac is a possible mechanism for ameliorating manganese-induced damage to dopaminergic neurons and that Cur exerts a certain protective effect against manganese-induced damage to dopaminergic neurons.
Collapse
Affiliation(s)
- Ying Liu
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Hua Zhao
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Yue Yang
- Guiyang Stomatological Hospital, Guiyang, Guizhou, 550002, China
| | - Yan Liu
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Chun-Yan Ao
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Jia-Min Zeng
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Jia-Qi Ban
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China.
| | - Jun Li
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China.
| |
Collapse
|
45
|
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
|
46
|
Fang YY, Teng MJ, Peng JC, Zheng XW, Mo YQ, Ho TT, Lin JJ, Luo JJ, Aschner M, Jiang YM. Combined exposure to manganese and iron decreases oxidative stress-induced nerve damage by increasing Nrf2/HO-1/NQO1 expression. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115853. [PMID: 38128313 DOI: 10.1016/j.ecoenv.2023.115853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/27/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Manganese (Mn) and iron (Fe) are essential trace elements for humans, yet excessive exposure to Mn or Fe can accumulate in the central nervous system (CNS) and cause neurotoxicity. The purpose of this study was to investigate the effects of Mn and Fe exposure, alone or in combination, on inducing oxidative stress-induced neurological damage in rat cortical and SH-SY5Y cells, and to determine whether combined exposure to these metals increases their individual toxicity. METHODS SH-SY5Y cells and male Sprague-Dawley rats were used to observe the effects of oxidative stress-induced neurological damage induced by exposure to manganese and iron alone or in combination. To detect the expression of anti-oxidative stress-related proteins, Nrf2, HO-1, and NQO1, and the apoptosis-related proteins, Bcl2 and Bax, and the neurological damage-related protein, α-syn. To detect reactive oxygen species generation and apoptosis. To detect the expression of the rat cortical protein Nrf2. To detect the production of proinflammatory cytokines. RESULTS We demonstrate that juvenile developmental exposure to Mn and Fe and their combination impairs cognitive performance in rats by inducing oxidative stress causing neurodegeneration in the cortex. Mn, Fe, and their combined exposure increased the expression of ROS, Bcl2, Bax, and α-syn, activated the inflammatory factors IL-6 and IL-12, inhibited the activities of SOD and GSH, and induced oxidative stress-induced neurodegeneration both in rats and SH-SY5Y cells. Combined Mn-Fe exposure attenuated the oxidative stress induced by Mn and Fe exposure alone by increasing the expression of antioxidant factors Nrf2, HO-1, and NQO1. CONCLUSION In both in vivo and in vitro studies, manganese and iron alone or in combination induced oxidative stress, leading to neuronal damage. In contrast, combined exposure to manganese and iron mitigated the oxidative stress induced by exposure to manganese and iron alone by increasing the expression of antioxidant factors. Therefore, studies to elucidate the main causes of toxicity and establish the molecular mechanisms of toxicity should help to develop more effective therapeutic modalities in the future.
Collapse
Affiliation(s)
- Yuan-Yuan Fang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China
| | - Meng-Jun Teng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China
| | - Jian-Chao Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China
| | - Xiao-Wei Zheng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China
| | - Ya-Qi Mo
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China
| | - Thanh-Tung Ho
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, 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
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, 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
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, 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
- Department of Molecular Pharmacology at Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China.
| |
Collapse
|
47
|
Song R, Chen H, Zhan R, Han M, Zhao L, Shen X. Vitamin E protects dopaminergic neurons against manganese-induced neurotoxicity through stimulation of CHRM1 and KCNJ4. J Trace Elem Med Biol 2024; 81:127326. [PMID: 37939525 DOI: 10.1016/j.jtemb.2023.127326] [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: 05/14/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Manganese (Mn) overexposure can induce neurotoxicity and lead to manganism. Vitamin E (Vit E) has neuroprotective effects by acting as an ROS scavenger, preventing mitochondrial dysfunction and neuronal apoptosis. However, the effects of Vit E on Mn-induced nigrostriatal system lesions remains unknown. OBJECTIVES We aim to investigate whether Vit E has protective effects on Mn-induced nigrostriatal system lesions and mRNA expression profiles in the SN of mice. METHODS Sixty 8-week-old C57BL/6 male mice were randomly divided into the Control, MnCl2, MnCl2 +Vit E, and Vit E group. Twenty-four hours after the last injection, the behaviour test was performed. The numbers of dopaminergic neurons in Substantia nigra (SN), the contents of dopamine and its metabolite levels in striatium, and the morphology of mitochondria and nuclei in the dopaminergic neurons in SN were detected by immunofluorescence staining, high-performance liquid chromatography, and transmission electron microscopy. Transcriptome analysis was used to analyze the signaling pathways and RT-PCR was used to verify the mRNA levels. RESULTS Vit E ameliorates behavioral disorders and attenuates the loss of nigral dopaminergic neurons in the Mn-induced mouse model. In addition, Vit E antagonized Mn-induced toxicity by restoring mitochondrial function. The results of transcriptome sequencing and RTPCR show that the protective effect of Vit E was related to the upregulation of CHRM1 and KCNJ4 mRNA in the SN. CONCLUSIONS Vit E has neuroprotective effects on Mn-induced neurodegeneration in the nigrostriatal system. This effect may be related to the upregulation of CHRM1 and KCNJ4 mRNA stimulated by Vit E in the SN.
Collapse
Affiliation(s)
- Ruihan Song
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, Shandong, China
| | - Huanhuan Chen
- Qingdao Municipal Center for Disease Control and Prevention/Qingdao Institute of Preventive Medicine, Qingdao, Shandong, China
| | - Runqing Zhan
- Qingdao University Affiliated Hiser Hospital, Qingdao, Shandong, China
| | - Miaomiao Han
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, Shandong, China
| | - Longzhu Zhao
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, Shandong, China
| | - Xiaoli Shen
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
48
|
Ogunro OB, Olasehinde OR. Neuroinflammatory Response and Redox-regulation Activity of Hyperoside in Manganese-induced Neurotoxicity Model of Wistar Rats. Curr Aging Sci 2024; 17:220-236. [PMID: 38500281 DOI: 10.2174/0118746098277166231204103616] [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: 09/04/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 03/20/2024]
Abstract
BACKGROUND Excessive manganese exposure can lead to neurotoxicity with detrimental effects on the brain. Neuroinflammatory responses and redox regulation play pivotal roles in this process. Exploring the impact of hyperoside in a Wistar rat model offers insights into potential neuroprotective strategies against manganese-induced neurotoxicity. OBJECTIVE The study investigated the neuroprotective efficacy of hyperoside isolated from the ethanol leaf extract of Gongronema latifolium (HELEGL), in the brain tissue of Wistar rats following 15 consecutive days of exposure to 30 mg/L of MnCl2. METHODS Control animals in Group 1 had access to regular drinking water, while animals in groups 2-4 were exposed to MnCl2 in their drinking water. Groups 3 and 4 also received additional HELEGL at doses of 100 mg/kg and 200 mg/kg of body weight, respectively. In Group 5, HELEGL at a dose of 100 mg/kg of body weight was administered alone. Treatment with HELEGL commenced on day 8 via oral administration. RESULTS HELEGL effectively mitigated MnCl2-induced memory impairment, organ-body weight discrepancies, and fluid intake deficits. Exposure to MnCl2 increased the activities or levels of various markers such as acyl peptide hydrolase, tumour necrosis factor-α, dipeptidyl peptidase IV, nitric oxide, IL-1β, prolyl oligopeptidase, caspase-3, myeloperoxidase, H2O2, and malondialdehyde, while it decreased the activities or levels of others, including AChE, BChE, DOPA, serotonin, epinephrine, norepinephrine, GST, GPx, CAT, SOD, GSH, and T-SH (p < 0.05). In contrast, HELEGL effectively counteracted the adverse effects of MnCl2 by alleviating oxidative stress, inflammation, apoptosis, mitochondrial dysfunction, cognitive deficits, and bolstering the antioxidant status. Moreover, HELEGL restored the normal histoarchitecture of the brain, which had been distorted by MnCl2. CONCLUSION In summary, HELEGL reversed the causative factors of neurodegenerative diseases induced by MnCl2 exposure, suggesting its potential for further exploration as a prospective therapeutic agent in the management of Alzheimer's disease and related forms of dementia.
Collapse
Affiliation(s)
- Olalekan Bukunmi Ogunro
- Department of Biological Sciences, Reproductive & Endocrinology, Toxicology, and Bioinformatics Research Laboratory, KolaDaisi University, Ibadan, Nigeria
- Department of Biochemistry, University of Ilorin, Ilorin, Nigeria
| | - Oluwaseun Ruth Olasehinde
- Department of Biochemistry, University of Ilorin, Ilorin, Nigeria
- Department of Medical Biochemistry, College of Medicine and Health Science, Afe Babalola University, Ado-Ekiti, Nigeria
| |
Collapse
|
49
|
Friedman A, Boselli E, Ogneva-Himmelberger Y, Heiger-Bernays W, Brochu P, Burgess M, Schildroth S, Denehy A, Downs T, Papautsky I, Clauss Henn B. Manganese in residential drinking water from a community-initiated case study in Massachusetts. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2024; 34:58-67. [PMID: 37301899 PMCID: PMC10727146 DOI: 10.1038/s41370-023-00563-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Manganese (Mn) is a metal commonly found in drinking water, but the level that is safe for consumption is unknown. In the United States (U.S.), Mn is not regulated in drinking water and data on water Mn concentrations are temporally and spatially sparse. OBJECTIVE Examine temporal and spatial variability of Mn concentrations in repeated tap water samples in a case study of Holliston, Massachusetts (MA), U.S., where drinking water is pumped from shallow aquifers that are vulnerable to Mn contamination. METHODS We collected 79 residential tap water samples from 21 households between September 2018 and December 2019. Mn concentrations were measured using inductively coupled plasma mass spectrometry. We calculated descriptive statistics and percent of samples exceeding aesthetic (secondary maximum containment level; SMCL) and lifetime health advisory (LHA) guidelines of 50 µg/L and 300 µg/L, respectively. We compared these concentrations to concurrent and historic water Mn concentrations from publicly available data across MA. RESULTS The median Mn concentration in Holliston residential tap water was 2.3 µg/L and levels were highly variable (range: 0.03-5,301.8 µg/L). Mn concentrations exceeded the SMCL and LHA in 14% and 12% of samples, respectively. Based on publicly available data across MA from 1994-2022, median Mn concentration was 17.0 µg/L (N = 37,210; range: 1-159,000 µg/L). On average 40% of samples each year exceeded the SMCL and 9% exceeded the LHA. Samples from publicly available data were not evenly distributed between MA towns or across sampling years. IMPACT STATEMENT This study is one of the first to examine Mn concentrations in drinking water both spatially and temporally in the U.S. Findings suggest that concentrations of Mn in drinking water frequently exceed current guidelines and occur at concentrations shown to be associated with adverse health outcomes, especially for vulnerable and susceptible subpopulations like children. Future studies that comprehensively examine exposure to Mn in drinking water and its associations with children's health are needed to protect public health.
Collapse
Affiliation(s)
- Alexa Friedman
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA.
| | - Elena Boselli
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA
| | - Yelena Ogneva-Himmelberger
- Department of International Development, Community, and Environment, Clark University, Worcester, MA, USA
| | - Wendy Heiger-Bernays
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Paige Brochu
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Mayah Burgess
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Samantha Schildroth
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | | | - Timothy Downs
- Department of International Development, Community, and Environment, Clark University, Worcester, MA, USA
| | - Ian Papautsky
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA
| | - Birgit Clauss Henn
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| |
Collapse
|
50
|
Akbar WA, Rahim HU, Irfan M, Sehrish AK, Mudassir M. Assessment of heavy metal distribution and bioaccumulation in soil and plants near coal mining areas: implications for environmental pollution and health risks. ENVIRONMENTAL MONITORING AND ASSESSMENT 2023; 196:97. [PMID: 38153601 DOI: 10.1007/s10661-023-12258-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 12/14/2023] [Indexed: 12/29/2023]
Abstract
Monitoring heavy metals (HMs) across source distance and depth distribution near coal mining sites is essential for preventing environmental pollution and health risks. This study investigated the distribution of selected HMs, cadmium (Cd2+), chromium (Cr2+), copper (Cu2+), manganese (Mn2+), nickel (Ni2+), lead (Pb2+), and zinc (Zn2+), in soil samples collected from ten sites (S-1-S-10) at two different depths (0-15 and 15-30 cm) and distances of 50, 100, and 200 m from a mining source. Additionally, three plant species, Prosopis spp., Justicia spp., and wheat, were collected to assess HM bioavailability and leaf accumulation. Coal mine activities' impact on soil properties and their HM associations were also explored. Results reveal HM concentrations except for Cr2+ exceeding World Health Organization (WHO) limits. In surface soil, Cd2+ (58%), Cu2+ (93%), Mn2+ (68%), Ni2+ (80%), Pb2+ (35%), and Zn2+ (88%) surpassed permissible limits. Subsurface soil also exhibited elevated Cd2+ (53%), Cu2+ (83%), Mn2+ (60%), Ni2+ (80%), Pb2+ (35%), and Zn2+ (77%). Plant species displayed varying HM levels, exceeding permissible limits, with average concentrations of 1.4, 1.34, 1.42, 4.1, 2.74, 2.0, and 1.98 mg kg-1 for Cd2+, Pb2+, Cr2+, Cu2+, Mn2+, Ni2+, and Zn2+, respectively. Bioaccumulation factors were highest in wheat, Prosopis spp., and Justicia spp. Source distance and depth distribution significantly influenced soil pH, electrical conductivity (EC), and soil organic carbon (SOC). Soil pH and EC increased with an increase in soil depth, while SOC decreased. Pearson correlation analysis revealed varying relationships between soil properties and HMs, showing a considerably negative correlation. Concentrations of HMs decreased with increasing depth and distance from mining activities, validated by regression analysis. Findings suggest crops from these soils may pose health risks for consumption.
Collapse
Affiliation(s)
- Waqas Ali Akbar
- Department of Soil and Environmental Sciences, The University of Agriculture, Peshawar, 25130, Pakistan
| | - Hafeez Ur Rahim
- Department of Chemical, Pharmaceutical and Agricultural Sciences (DOCPAS), University of Ferrara, 44121, Ferrara, Italy.
| | - Muhammad Irfan
- Department of Soil and Environmental Sciences, The University of Agriculture, Peshawar, 25130, Pakistan
| | - Adiba Khan Sehrish
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University (Xianlin Campus), 163 Xianlin Road, Jiangsu Province, Qixia District, Nanjing, 210023, People's Republic of China
| | - Muhammad Mudassir
- Department of Soil and Environmental Sciences, The University of Agriculture, Peshawar, 25130, Pakistan
| |
Collapse
|