1
|
Yu KB, Chandra F, Coley-O'Rourke EJ, Paulson ET, Novoselov A, Zhang D, Finnigan D, Paramo J, Lopez-Romero A, Dong TS, Schartup AT, Hsiao EY. An engineered gut bacterium protects against dietary methylmercury exposure in pregnant mice. Cell Host Microbe 2025; 33:621-631.e7. [PMID: 40315838 DOI: 10.1016/j.chom.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/17/2025] [Accepted: 04/10/2025] [Indexed: 05/04/2025]
Abstract
Despite efforts to decrease mercury emissions, chronic exposure to the neurotoxicant methylmercury (MeHg) continues to be a global problem that contributes to disparities in risk for neurological and metabolic diseases. Herein we engineer a human commensal gut bacterium, Bacteroides thetaiotaomicron (Bt), to detoxify MeHg by heterologous expression of organomercury lyase (MerB) and mercuric reductase (MerA) genes derived from a resistant bacterium isolated from Hg-polluted mines. We demonstrate that BtmerA/B demethylates MeHg both in vitro and within the intestines of mice orally exposed to MeHg or diets containing MeHg-rich fish. In pregnant mice exposed to dietary MeHg, BtmerA/B decreases MeHg accumulation in the maternal liver, brain, placenta, and fetal brain, and attenuates the expression of cellular stress genes in the fetal brain. Overall, this work provides foundational proof-of-principle supporting the ability of an engineered gut bacterium to limit MeHg bioaccumulation and reduce adverse effects of chronic MeHg exposure.
Collapse
Affiliation(s)
- Kristie B Yu
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Franciscus Chandra
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Elena J Coley-O'Rourke
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Erik T Paulson
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA 92037, USA
| | - Anna Novoselov
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - David Zhang
- UCLA Goodman-Luskin Microbiome Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Delanie Finnigan
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jorge Paramo
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Arlene Lopez-Romero
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tien S Dong
- UCLA Goodman-Luskin Microbiome Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amina T Schartup
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA 92037, USA.
| | - Elaine Y Hsiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Goodman-Luskin Microbiome Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
2
|
Daguano Gastaldi V, Hindermann M, Wilke JBH, Ronnenberg A, Arinrad S, Kraus S, Wildenburg AF, Ntolkeras A, Provost MJ, Ye L, Curto Y, Cortés-Silva JA, Butt UJ, Nave KA, Miskowiak KW, Ehrenreich H. A comprehensive and standardized pipeline for automated profiling of higher cognition in mice. CELL REPORTS METHODS 2025; 5:101011. [PMID: 40101715 PMCID: PMC12049718 DOI: 10.1016/j.crmeth.2025.101011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 11/20/2024] [Accepted: 02/26/2025] [Indexed: 03/20/2025]
Abstract
In rodent behavior research, observer-independent methods, such as the IntelliCage, enhance data collection in a social, and thus stress-reduced, environment. The IntelliCage system allows experimenters to create cognitive challenges for mice motivated by rewards. Given the extensive and diverse data from IntelliCage, there is a high demand for automated analysis. Here, we introduce IntelliR, a free and standardized pipeline for analyzing IntelliCage data, including a cognition index for performance comparison across challenges. IntelliR supports the automatic analysis of three challenges that cover spatial, episodic-like, and working memory with their reversal tests and can also be adapted for other designs. Results from three cohorts of adult female C57B6 mice showed improved task proficiency over time. To validate cognitive impairment detection, we used adult female NexCreERT2xRosa26-eGFP-DTA mice after neuron ablation in cortex and hippocampus, in which we observed reduced learning capabilities. IntelliR integrates easily into research, improving time management and reproducibility.
Collapse
Affiliation(s)
- Vinicius Daguano Gastaldi
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany; Georg-August-University, Göttingen, Germany
| | - Martin Hindermann
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany; Georg-August-University, Göttingen, Germany
| | - Justus B H Wilke
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Anja Ronnenberg
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Sahab Arinrad
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Sabine Kraus
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Anne-Fleur Wildenburg
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany; Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Antonios Ntolkeras
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany; Georg-August-University, Göttingen, Germany
| | - Micah J Provost
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany; Georg-August-University, Göttingen, Germany
| | - Liu Ye
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany; Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning 530022, China
| | - Yasmina Curto
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Jonathan-Alexis Cortés-Silva
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Umer Javed Butt
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Kamilla Woznica Miskowiak
- Neurocognition and Emotion in Affective Disorders (NEAD) Centre, Copenhagen Affective Disorder Research Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital and Department of Psychology, University of Copenhagen, 2000 Frederiksberg, Denmark
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany; Georg-August-University, Göttingen, Germany; Experimental Medicine, Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J 5, 68159 Mannheim, Germany.
| |
Collapse
|
3
|
Yamagata R, Saito A, Fukushima R, Nakagawasai O, Yamashita N, Tan-No K, Hwang GW. Methylmercury exposure at dosage conditions that do not affect growth can impair memory in adolescent mice. Toxicol Res 2024; 40:441-448. [PMID: 38911546 PMCID: PMC11187021 DOI: 10.1007/s43188-024-00239-y] [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: 03/05/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 06/25/2024] Open
Abstract
Methylmercury is an environmental pollutant that can induce serious central nervous system damage. Its ubiquitous presence in the environment in trace amounts has raised concerns about potential adverse effects on human health. Although many studies have evaluated the effects of methylmercury on neural development in fetal and neonatal mice, there has been less focus on studies using adolescent mice. Therefore, in this study, the effects of methylmercury on brain neurodevelopment and maturation were evaluated by various neurobehavioral trials using adolescent mice exposed to 30 ppm methylmercuric chloride (approximately 24 ppm methylmercury) for up to 8 weeks. Under these administration conditions, weight gain in adolescent mice was unaffected by methylmercury exposure. Furthermore, methylmercury exposure in adolescent mice had no effect on sociability as assessed by the social interaction test, impulsivity as assessed by the cliff avoidance reaction test, depressive behavior as assessed by the tail-suspension test, or locomotor activity as assessed using the Supermex system. In contrast, short-term memory assessed by the Y-maze test, as well as long-term memory assessed by novel object recognition and passive avoidance tests, revealed impairments induced by methylmercury exposure in adolescent mice. These results suggest that long-term exposure to methylmercury during adolescence potentially impairs memory function, and the nervous pathway of brain areas involved in learning and memory are particularly vulnerable to the adverse effects of methylmercury. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-024-00239-y.
Collapse
Affiliation(s)
- Ryota Yamagata
- Division of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 981-8558 Japan
| | - Ai Saito
- Division of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 981-8558 Japan
| | - Ryoko Fukushima
- Division of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 981-8558 Japan
| | - Osamu Nakagawasai
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 981-8558 Japan
| | - Naoya Yamashita
- Division of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 981-8558 Japan
| | - Koichi Tan-No
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 981-8558 Japan
| | - Gi-Wook Hwang
- Division of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 981-8558 Japan
| |
Collapse
|
4
|
Lipp HP, Krackow S, Turkes E, Benner S, Endo T, Russig H. IntelliCage: the development and perspectives of a mouse- and user-friendly automated behavioral test system. Front Behav Neurosci 2024; 17:1270538. [PMID: 38235003 PMCID: PMC10793385 DOI: 10.3389/fnbeh.2023.1270538] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/18/2023] [Indexed: 01/19/2024] Open
Abstract
IntelliCage for mice is a rodent home-cage equipped with four corner structures harboring symmetrical double panels for operant conditioning at each of the two sides, either by reward (access to water) or by aversion (non-painful stimuli: air-puffs, LED lights). Corner visits, nose-pokes and actual licks at bottle-nipples are recorded individually using subcutaneously implanted transponders for RFID identification of up to 16 adult mice housed in the same home-cage. This allows for recording individual in-cage activity of mice and applying reward/punishment operant conditioning schemes in corners using workflows designed on a versatile graphic user interface. IntelliCage development had four roots: (i) dissatisfaction with standard approaches for analyzing mouse behavior, including standardization and reproducibility issues, (ii) response to handling and housing animal welfare issues, (iii) the increasing number of mouse models had produced a high work burden on classic manual behavioral phenotyping of single mice. and (iv), studies of transponder-chipped mice in outdoor settings revealed clear genetic behavioral differences in mouse models corresponding to those observed by classic testing in the laboratory. The latter observations were important for the development of home-cage testing in social groups, because they contradicted the traditional belief that animals must be tested under social isolation to prevent disturbance by other group members. The use of IntelliCages reduced indeed the amount of classic testing remarkably, while its flexibility was proved in a wide range of applications worldwide including transcontinental parallel testing. Essentially, two lines of testing emerged: sophisticated analysis of spontaneous behavior in the IntelliCage for screening of new genetic models, and hypothesis testing in many fields of behavioral neuroscience. Upcoming developments of the IntelliCage aim at improved stimulus presentation in the learning corners and videotracking of social interactions within the IntelliCage. Its main advantages are (i) that mice live in social context and are not stressfully handled for experiments, (ii) that studies are not restricted in time and can run in absence of humans, (iii) that it increases reproducibility of behavioral phenotyping worldwide, and (iv) that the industrial standardization of the cage permits retrospective data analysis with new statistical tools even after many years.
Collapse
Affiliation(s)
- Hans-Peter Lipp
- Faculty of Medicine, Institute of Evolutionary Medicine, University of Zürich, Zürich, Switzerland
| | - Sven Krackow
- Institute of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Emir Turkes
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Seico Benner
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Ibaraki, Japan
| | | | | |
Collapse
|
5
|
Oguro A, Fujiyama T, Ishihara Y, Kataoka C, Yamamoto M, Eto K, Komohara Y, Imaoka S, Sakuragi T, Tsuji M, Shibata E, Kotake Y, Yamazaki T. Maternal DHA intake in mice increased DHA metabolites in the pup brain and ameliorated MeHg-induced behavioral disorder. J Lipid Res 2023; 64:100458. [PMID: 37838304 PMCID: PMC10656226 DOI: 10.1016/j.jlr.2023.100458] [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: 02/17/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/16/2023] Open
Abstract
Although pregnant women's fish consumption is beneficial for the brain development of the fetus due to the DHA in fish, seafood also contains methylmercury (MeHg), which adversely affects fetal brain development. Epidemiological studies suggest that high DHA levels in pregnant women's sera may protect the fetal brain from MeHg-induced neurotoxicity, but the underlying mechanism is unknown. Our earlier study revealed that DHA and its metabolite 19,20-dihydroxydocosapentaenoic acid (19,20-DHDP) produced by cytochrome P450s (P450s) and soluble epoxide hydrolase (sEH) can suppress MeHg-induced cytotoxicity in mouse primary neuronal cells. In the present study, DHA supplementation to pregnant mice suppressed MeHg-induced impairments of pups' body weight, grip strength, motor function, and short-term memory. DHA supplementation also suppressed MeHg-induced oxidative stress and the decrease in the number of subplate neurons in the cerebral cortex of the pups. DHA supplementation to dams significantly increased the DHA metabolites 19,20-epoxydocosapentaenoic acid (19,20-EDP) and 19,20-DHDP as well as DHA itself in the fetal and infant brains, although the expression levels of P450s and sEH were low in the fetal brain and liver. DHA metabolites were detected in the mouse breast milk and in human umbilical cord blood, indicating the active transfer of DHA metabolites from dams to pups. These results demonstrate that DHA supplementation increased DHA and its metabolites in the mouse pup brain and alleviated the effects of MeHg on fetal brain development. Pregnant women's intake of fish containing high levels of DHA (or DHA supplementation) may help prevent MeHg-induced neurotoxicity in the fetus.
Collapse
Affiliation(s)
- Ami Oguro
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Taichi Fujiyama
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yasuhiro Ishihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | | | | | - Komyo Eto
- National Institute for Minamata Disease, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Susumu Imaoka
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo, Japan
| | - Toshihide Sakuragi
- Department of Environmental Health, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan; Department of Obstetrics and Gynecology, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Mayumi Tsuji
- Department of Environmental Health, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Eiji Shibata
- Department of Obstetrics and Gynecology, Dokkyo Medical University, Tochigi, Japan
| | - Yaichiro Kotake
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takeshi Yamazaki
- Program of Life and Environmental Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
6
|
Bramati G, Stauffer P, Nigri M, Wolfer DP, Amrein I. Environmental enrichment improves hippocampus-dependent spatial learning in female C57BL/6 mice in novel IntelliCage sweet reward-based behavioral tests. Front Behav Neurosci 2023; 17:1256744. [PMID: 37791111 PMCID: PMC10543696 DOI: 10.3389/fnbeh.2023.1256744] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/11/2023] [Indexed: 10/05/2023] Open
Abstract
The IntelliCage is an automated home-cage system that allows researchers to investigate the spontaneous behavior and learning abilities of group-housed mice. The IntelliCage enables us to increase the standardization and reproducibility of behavioral outcomes by the omission of experimenter-mouse interactions. Although the IntelliCage provides a less stressful environment for animals, standard IntelliCage protocols use controlled water access as the motivational driver for learning. To overcome possible water restrictions in slow learners, we developed a series of novel protocols based on appetitive learning, in which mice had permanent access to plain water but were additionally rewarded with sweetened water upon solving the task. C57BL/6NCrl female mice were used to assess the efficacy of these sweet reward-based protocols in a series of learning tasks. Compared to control mice tested with standard protocols, mice motivated with a sweet reward did equal to or better in operant performance and place learning tasks. Learning of temporal rules was slower than that in controls. When faced with a combined temporal x spatial working memory task, sweet-rewarded mice learned little and chose plain water. In a second set of experiments, the impact of environmental enrichment on appetitive learning was tested. Mice kept under enriched environment (EE) or standard housing (SH) conditions prior to the IntelliCage experiments performed similarly in the sweet-rewarded place learning task. EE mice performed better in the hippocampus-dependent spatial working memory task. The improved performance of EE mice in the hippocampus-dependent spatial working memory task might be explained by the observed larger volume of their mossy fibers. Our results confirm that environmental enrichment increases complex spatial learning abilities and leads to long-lasting morphological changes in the hippocampus. Furthermore, simple standard IntelliCage protocols could easily be adapted to sweet rewards, which improve animal welfare by removing the possibility of water restriction. However, complex behavioral tasks motivated by sweet reward-based learning need further adjustments to reach the same efficacy as standard protocols.
Collapse
Affiliation(s)
- Giulia Bramati
- Division Functional Neuroanatomy, Institute of Anatomy, University Zurich, Zürich, Switzerland
| | - Pia Stauffer
- Division Functional Neuroanatomy, Institute of Anatomy, University Zurich, Zürich, Switzerland
| | - Martina Nigri
- Division Functional Neuroanatomy, Institute of Anatomy, University Zurich, Zürich, Switzerland
- Department of Health Sciences and Technology, ETH, Zürich, Switzerland
| | - David P. Wolfer
- Division Functional Neuroanatomy, Institute of Anatomy, University Zurich, Zürich, Switzerland
- Department of Health Sciences and Technology, ETH, Zürich, Switzerland
| | - Irmgard Amrein
- Division Functional Neuroanatomy, Institute of Anatomy, University Zurich, Zürich, Switzerland
- Department of Health Sciences and Technology, ETH, Zürich, Switzerland
| |
Collapse
|
7
|
Bose R, Spulber S, Ceccatelli S. The Threat Posed by Environmental Contaminants on Neurodevelopment: What Can We Learn from Neural Stem Cells? Int J Mol Sci 2023; 24:4338. [PMID: 36901772 PMCID: PMC10002364 DOI: 10.3390/ijms24054338] [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: 01/08/2023] [Revised: 02/03/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Exposure to chemicals may pose a greater risk to vulnerable groups, including pregnant women, fetuses, and children, that may lead to diseases linked to the toxicants' target organs. Among chemical contaminants, methylmercury (MeHg), present in aquatic food, is one of the most harmful to the developing nervous system depending on time and level of exposure. Moreover, certain man-made PFAS, such as PFOS and PFOA, used in commercial and industrial products including liquid repellants for paper, packaging, textile, leather, and carpets, are developmental neurotoxicants. There is vast knowledge about the detrimental neurotoxic effects induced by high levels of exposure to these chemicals. Less is known about the consequences that low-level exposures may have on neurodevelopment, although an increasing number of studies link neurotoxic chemical exposures to neurodevelopmental disorders. Still, the mechanisms of toxicity are not identified. Here we review in vitro mechanistic studies using neural stem cells (NSCs) from rodents and humans to dissect the cellular and molecular processes changed by exposure to environmentally relevant levels of MeHg or PFOS/PFOA. All studies show that even low concentrations dysregulate critical neurodevelopmental steps supporting the idea that neurotoxic chemicals may play a role in the onset of neurodevelopmental disorders.
Collapse
Affiliation(s)
| | | | - Sandra Ceccatelli
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
8
|
Kendricks DR, Bhattacharya S, Reed MN, Newland MC. Impacts of Neonatal Methylmercury on Behavioral Flexibility and Learning in Spatial Discrimination Reversal and Visual Signal Detection Tasks. Neurotoxicology 2022; 93:9-21. [PMID: 36055519 DOI: 10.1016/j.neuro.2022.08.013] [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: 06/14/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/19/2022]
Abstract
Early postnatal development in rodents is sensitive to neurotoxic effects of the environmental contaminant, methylmercury. While juvenile and adolescent exposure also produce long-term impairments in behavior, the outcome of neonatal exposure is less understood. Neural development during the neonatal period in rodents is akin to that seen in humans during the third trimester of pregnancy but methylmercury exposure occurring during the neonatal period has not been modeled, partly because breast milk is a poor source of bioavailable methylmercury. To examine this developmental period, male Long-Evans rats were exposed to 0, 80, or 350µg/kg/day methylmercuric chloride from postnatal days 1 to 10, the rodent neonatal period. As adults, behavioral flexibility, attention, memory, and expression of the dopamine transporter in these rats was assessed. Rats exhibited changes in behavioral flexibility assessed in a spatial discrimination reversal procedure. Those rats exposed to the highest dose of methylmercury displayed subtly altered patterns of perseveration compared to control animals. During acquisition of the attention/memory procedure, rats exposed to this dose also had slower acquisition, and achieved lower overall accuracy during training, compared to controls despite neither attention nor memory being affected once the task was acquired. Finally, dopamine transporter expression in the striatum, prefrontal cortex, and hippocampus was unchanged in these adult rats. The results of this study replicate the trend of findings seen with exposure during gestation or during adolescence.
Collapse
Affiliation(s)
- Dalisa R Kendricks
- Department of Psychological Sciences, Auburn University, Auburn, AL, USA
| | - Subhrajit Bhattacharya
- Keck Graduate Institute, School of Pharmacy and Health Sciences, Claremont Colleges, Claremont, CA, USA
| | - Miranda N Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA; Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA
| | | |
Collapse
|
9
|
Akiyama M, Shinkai Y, Yamakawa H, Kim YG, Kumagai Y. Potentiation of methylmercury toxicity by combined metal exposure: In vitro and in vivo models of a restricted metal exposome. CHEMOSPHERE 2022; 299:134374. [PMID: 35318019 DOI: 10.1016/j.chemosphere.2022.134374] [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: 12/22/2021] [Revised: 03/05/2022] [Accepted: 03/17/2022] [Indexed: 06/14/2023]
Abstract
Methylmercury (MeHg) is a prevalent toxic metal that readily modifies protein thiols. Reactive persulfides that play a role in redox homeostasis are able to inactivate this metal through sulfur adduct formation. Although humans are exposed to other metals that could consume reactive persulfides on a daily basis, the health effects of combined exposure to MeHg and other metals remain unexplored. This study aimed to examine potential MeHg toxicity during exposure to MeHg with other metals capable of consuming reactive persulfides. We designed a simple system to assess the risk of combined exposure to metals based on reactivity to reactive persulfides and mercury accumulation. Among the metals examined in a cell-free system, copper, cadmium, nickel, and MeHg consumed Na2S2, used as a model of reactive persulfides, whereas zinc, iron, lithium, strontium, tin, and aluminum did not. In HepG2 cells, binary exposure to MeHg and copper, but not aluminum, increased the consumption of extracellular reactive persulfides. Binary exposure exacerbated MeHg-induced cytotoxicity by promoting the modification of intracellular proteins by MeHg. In a mouse model, binary exposure to MeHg and copper resulted in elevated mercury accumulation in the fetuses and placenta of pregnant mice, as well as the brain and liver of non-pregnant mice. Our study suggests that MeHg sensitivity can be increased by combined exposure with other electrophilic metals. In particular, binary exposure to MeHg and copper during pregnancy exacerbated mercury accumulation in offspring.
Collapse
Affiliation(s)
- Masahiro Akiyama
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan; Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, 105-8512, Japan
| | - Yasuhiro Shinkai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hiroto Yamakawa
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yun-Gi Kim
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, 105-8512, Japan
| | - Yoshito Kumagai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
10
|
Kendricks DR, Boomhower SR, Newland MC. Adolescence as a sensitive period for neurotoxicity: Lifespan developmental effects of methylmercury. Pharmacol Biochem Behav 2022; 217:173389. [PMID: 35452710 DOI: 10.1016/j.pbb.2022.173389] [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: 10/19/2021] [Revised: 04/04/2022] [Accepted: 04/11/2022] [Indexed: 10/18/2022]
Abstract
Neurotoxicity resulting from the environmental contaminant, methylmercury (MeHg), is a source of concern for many human populations that rely heavily on the consumption of fish and rice as stable ingredients in the diet. The developmental period of exposure is important both to the qualitative effects of MeHg and to the dose required to produce those effects. MeHg exposure during the sensitive prenatal period causes deleterious and long-lasting changes in neurodevelopment at particularly low doses. The effects include a wide host of cognitive and behavioral outcomes expressed in adulthood and sometimes not until aging. However, neurotoxic outcomes of methylmercury when exposure occurs during adolescence are only recently revealing impacts on human populations and animal models. This review examines the current body of work and showcases the sensitivity of adolescence, a period that straddles early development and adulthood, to methylmercury neurotoxicity and the implications such toxicity has in our understanding of methylmercury's effects in human populations and animal models.
Collapse
Affiliation(s)
- Dalisa R Kendricks
- Department of Psychology, Auburn University, Auburn, AL, United States of America.
| | - Steven R Boomhower
- Gradient, Boston, MA, United States of America; Harvard Division of Continuing Education, Harvard University, Cambridge, MA, United States of America
| | | |
Collapse
|
11
|
Abu-Taweel GM, Rudayni HA. Curcumin ameliorated the mercuric chloride induced depression and anxiety in female mice offspring. ENVIRONMENTAL RESEARCH 2022; 204:112031. [PMID: 34534522 DOI: 10.1016/j.envres.2021.112031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/25/2021] [Accepted: 08/29/2021] [Indexed: 06/13/2023]
Abstract
In the present investigation, the effect of mercuric chloride on gestation and lactation periods in mice was studied. The animals were treated with 10 ppm of HgCl2 and its complications were evaluated by supplementing 150 and 300 ppm of curcumin, respectively. Results indicated that HgCl2 increased depression-like behavior in treated animals compared to control and effects of depression in offspring significantly (p˂0.001) enhanced. Interestingly, the Tail suspension test clearly confirmed that the administration of curcumin enhanced the immobility (p˂0.001). The results confirmed that the curcumin administered mice spent less time in the closed arm (P < 0.001), whereas spent a very long time (P < 0.001) in the open arm. Related to the locomotor behaviors, number of squares crossed, wall rear, rear, and locomotion duration were decreased significantly (P < 0.001) while immobility duration was increased (P < 0.001) significantly compared to control. The anxiety and depression behaviors disorder due to mercuric chloride exposure indicated its availability via placenta or/and milk during lactation. The treatment with curcumin improved anxiety and depression behaviors compared to Hg experimental group.
Collapse
Affiliation(s)
- Gasem Mohammad Abu-Taweel
- Department of Biology, College of Sciences, Jazan University, P.O. Box 2079, Jazan, 45142, Saudi Arabia.
| | - Hassan Ahmed Rudayni
- Biology Department, College of Science, Imam Muhammad bin Saud Islamic University, B.o.box 90950, Riyadh, 11623, Saudi Arabia
| |
Collapse
|
12
|
Methylmercury chloride exposure exacerbates existing neurobehavioral and immune dysfunctions in the BTBR T+ Itpr3tf/J mouse model of autism. Immunol Lett 2022; 244:19-27. [DOI: 10.1016/j.imlet.2022.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/03/2022] [Accepted: 03/04/2022] [Indexed: 12/15/2022]
|
13
|
Methylmercury exposure during prenatal and postnatal neurodevelopment promotes oxidative stress associated with motor and cognitive damages in rats: an environmental-experimental toxicology study. Toxicol Rep 2022; 9:563-574. [PMID: 35392159 PMCID: PMC8980556 DOI: 10.1016/j.toxrep.2022.02.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/29/2022] [Accepted: 02/25/2022] [Indexed: 12/11/2022] Open
Abstract
The environmental contamination by methylmercury (MeHg) is a major concern for public health. The effects of MeHg in the central nervous system (CNS) of adult animals have been extensively investigated; however, little is known about the effects of MeHg exposure during intrauterine and lactation periods on motor and cognitive functions of adolescent rats. Therefore, this study aimed to investigate the effect of MeHg exposure during intrauterine life and lactation on both motor and cognitive functions of offspring rats. Ten female Wistar rats were exposed to 40 μg/kg/day of MeHg through cookie treats from the first day of pregnancy until the last day of breastfeeding. Both motor and cognitive functions of offspring male rats were assessed by open field, rotarod, and step-down inhibitory avoidance tests. Forty-one days after birth, the hippocampus and cerebellum were collected to determine total Hg content, antioxidant capacity against peroxyl radicals (ACAP), reduced glutathione (GSH) levels, lipid peroxidation (LPO), and nitrite levels. MeHg exposure during CNS development increased Hg levels in both hippocampal and cerebellar parenchymas, triggered oxidative stress throughout ACAP and GSH decrease, increased LPO and nitrite levels. These alterations resulted in reduced spontaneous and stimulated locomotion and short- and long-term memory deficits. Therefore, damages triggered by MeHg exposure during intrauterine life and lactation had detrimental effects on oxidative biochemistry and motor and cognitive functions of offspring rats. The MeHg exposure during CNS development increased mercury levels in hippocampal and cerebellar parenchyma. The MeHg intoxication during pregnancy and lactation impairs the redox status of hippocampus and cerebellum of the offspring. MeHg exposure causes behavioral effects in motor ability and cognition of offspring rats.
Collapse
|
14
|
Qiu Q, Huang Y, Zhang B, Huang D, Chen X, Fan Z, Lin J, Yang W, Wang K, Qu N, Li J, Li Z, Huang J, Li S, Zhang J, Liu G, Rui G, Chen X, Zhao Q. Noninvasive Dual-Modality Photoacoustic-Ultrasonic Imaging to Detect Mammalian Embryo Abnormalities after Prenatal Exposure to Methylmercury Chloride (MMC): A Mouse Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:27002. [PMID: 35108087 PMCID: PMC8809665 DOI: 10.1289/ehp8907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Severe environmental pollution and contaminants left in the environment due to the abuse of chemicals, such as methylmercury, are associated with an increasing number of embryonic disorders. Ultrasound imaging has been widely used to investigate embryonic development malformation and dysorganoplasia in both research and clinics. However, this technique is limited by its low contrast and lacking functional parameters such as the ability to measure blood oxygen saturation (SaO 2 ) and hemoglobin content (HbT) in tissues, measures that could be early vital indicators for embryonic development abnormality. Herein, we proposed combining two highly complementary techniques into a photoacoustic-ultrasound (PA-US) dual-modality imaging approach to noninvasively detect early mouse embryo abnormalities caused by methylmercury chloride (MMC) in real time. OBJECTIVES This study aimed to assess the use of PA-US dual-modality imaging for noninvasive detection of embryonic toxicity at different stages of growth following prenatal MMC exposure. Additionally, we compared the PA-US imagining results to traditional histological methods to determine whether this noninvasive method could detect early developmental defects in utero. METHODS Different dosages of MMC were administrated to pregnant mice by gavage to establish models of different levels of embryonic malformation. Ultrasound, photoacoustic signal intensity (PSI), blood oxygen saturation (SaO 2 ), and hemoglobin content (HbT) were quantified in all experimental groups. Furthermore, the embryos were sectioned and examined for pathological changes. RESULTS Using PA-US imaging, we detected differences in PSI, SaO 2 , HbT, and heart volume at embryonic day (E)14.5 and E11.5 for low and high dosages of MMC, respectively. More important, our results showed that differences between control and treated embryos identified by in utero PA-US imaging were consistent with those identified in ex vivo embryos using histological methods. CONCLUSION Our results suggest that noninvasive dual-modality PA-US is a promising strategy for detecting developmental toxicology in the uterus. Overall, this study presents a new approach for detecting embryonic toxicities, which could be crucial in clinics when diagnosing aberrant embryonic development. https://doi.org/10.1289/EHP8907.
Collapse
Affiliation(s)
- Qi Qiu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Yali Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Bei Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Doudou Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Xin Chen
- Department of Orthopedics, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Zhongxiong Fan
- Department of Biomaterials, College of Materials, Research Center of Biomedical Engineering of Xiamen & Key Laboratory of Biomedical Engineering of Fujian Province & Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Xiamen, China
| | - Jinpei Lin
- Department of Integrated TCM & Western Medicine Department, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Wensheng Yang
- Department of Pathology Affiliated Chenggong Hospital, Xiamen University, Xiamen, China
| | - Kai Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Ning Qu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Juan Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Zhihong Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Jingyu Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Shenrui Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Jiaxing Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Gang Rui
- Department of Orthopedics, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Qingliang Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
15
|
Pan J, Li X, Wei Y, Ni L, Xu B, Deng Y, Yang T, Liu W. Advances on the Influence of Methylmercury Exposure during Neurodevelopment. Chem Res Toxicol 2022; 35:43-58. [PMID: 34989572 DOI: 10.1021/acs.chemrestox.1c00255] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Mercury (Hg) is a toxic heavy-metal element, which can be enriched in fauna and flora and transformed into methylmercury (MeHg). MeHg is a widely distributed environmental pollutant that may be harmful to fish-eating populations through enrichment of aquatic food chains. The central nervous system is a primary target of MeHg. Embryos and infants are more sensitive to MeHg, and exposure to MeHg during gestational feeding can significantly impair the homeostasis of offspring, leading to long-term neurodevelopmental defects. At present, MeHg-induced neurodevelopmental toxicity has become a hotspot in the field of neurotoxicology, but its mechanisms are not fully understood. Some evidence point to oxidative damage, excitotoxicity, calcium ion imbalance, mitochondrial dysfunction, epigenetic changes, and other molecular mechanisms that play important roles in MeHg-induced neurodevelopmental toxicity. In this review, advances in the study of neurodevelopmental toxicity of MeHg exposure during pregnancy and the molecular mechanisms of related pathways are summarized, in order to provide more scientific basis for the study of neurodevelopmental toxicity of MeHg.
Collapse
Affiliation(s)
- Jingjing Pan
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Xiaoyang Li
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Yanfeng Wei
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Linlin Ni
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Tianyao Yang
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| |
Collapse
|
16
|
Unoki T, Akiyama M, Shinkai Y, Kumagai Y, Fujimura M. Spatio-temporal distribution of reactive sulfur species during methylmercury exposure in the rat brain. J Toxicol Sci 2022; 47:31-37. [PMID: 34987139 DOI: 10.2131/jts.47.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Brain susceptibility to methylmercury (MeHg) is developmentally and regionally specific in both humans and rodents, but the mechanism is not well clarified. Reactive sulfur species (RSS) with high nucleophilicity can react with MeHg, leading to the formation of a less toxic metabolite bismethylmercury sulfide, thus exerting cytoprotection. In this study, we assessed the variation of RSS content in the rat brain and evaluated its relevance in sensitivity to MeHg. Analyses of fetal/juvenile rat brains showed low RSS levels in early developmental stages. Site-specific analysis of adult rat brains revealed that cerebellar RSS levels were lower than those of the hippocampus. Microscopically, RSS levels of the granular cell layer were lower than those of the molecular layer in the cerebellum. Thus, low RSS levels corresponded with age and site of the brain that is vulnerable to MeHg. Taken together with the finding that brain RSS were consumed during MeHg exposure, these results indicate that RSS is a factor that defines the specificity of MeHg vulnerability in the brain.
Collapse
Affiliation(s)
- Takamitsu Unoki
- Department of Basic Medical Sciences, National Institute for Minamata Disease
| | - Masahiro Akiyama
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University
| | - Yasuhiro Shinkai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba
| | - Yoshito Kumagai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba
| | - Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease
| |
Collapse
|
17
|
Luz DA, Cartágenes SDC, da Silveira CCSDM, Pinheiro BG, Ferraro KMMM, Fernandes LDMP, Fontes-Júnior EA, Maia CDSF. Methylmercury plus Ethanol Exposure: How Much Does This Combination Affect Emotionality? Int J Mol Sci 2021; 22:ijms222313131. [PMID: 34884935 PMCID: PMC8658096 DOI: 10.3390/ijms222313131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022] Open
Abstract
Mercury is a heavy metal found in organic and inorganic forms that represents an important toxicant with impact on human health. Mercury can be released in the environment by natural phenoms (i.e., volcanic eruptions), industrial products, waste, or anthropogenic actions (i.e., mining activity). Evidence has pointed to mercury exposure inducing neurological damages related to emotional disturbance, such as anxiety, depression, and insomnia. The mechanisms that underlie these emotional disorders remain poorly understood, although an important role of glutamatergic pathways, alterations in HPA axis, and disturbance in activity of monoamines have been suggested. Ethanol (EtOH) is a psychoactive substance consumed worldwide that induces emotional alterations that have been strongly investigated, and shares common pathophysiological mechanisms with mercury. Concomitant mercury and EtOH intoxication occur in several regions of the world, specially by communities that consume seafood and fish as the principal product of nutrition (i.e., Amazon region). Such affront appears to be more deleterious in critical periods of life, such as the prenatal and adolescence period. Thus, this review aimed to discuss the cellular and behavioral changes displayed by the mercury plus EtOH exposure during adolescence, focused on emotional disorders, to answer the question of whether mercury plus EtOH exposure intensifies depression, anxiety, and insomnia observed by the toxicants in isolation.
Collapse
Affiliation(s)
- Diandra Araújo Luz
- Laboratório de Farmacologia da Inflamação e do Comportamento, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66075-110, PA, Brazil; (D.A.L.); (S.d.C.C.); (C.C.S.d.M.d.S.); (B.G.P.); (K.M.M.M.F.); (E.A.F.-J.)
| | - Sabrina de Carvalho Cartágenes
- Laboratório de Farmacologia da Inflamação e do Comportamento, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66075-110, PA, Brazil; (D.A.L.); (S.d.C.C.); (C.C.S.d.M.d.S.); (B.G.P.); (K.M.M.M.F.); (E.A.F.-J.)
| | - Cinthia Cristina Sousa de Menezes da Silveira
- Laboratório de Farmacologia da Inflamação e do Comportamento, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66075-110, PA, Brazil; (D.A.L.); (S.d.C.C.); (C.C.S.d.M.d.S.); (B.G.P.); (K.M.M.M.F.); (E.A.F.-J.)
| | - Bruno Gonçalves Pinheiro
- Laboratório de Farmacologia da Inflamação e do Comportamento, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66075-110, PA, Brazil; (D.A.L.); (S.d.C.C.); (C.C.S.d.M.d.S.); (B.G.P.); (K.M.M.M.F.); (E.A.F.-J.)
| | - Kissila Márvia Matias Machado Ferraro
- Laboratório de Farmacologia da Inflamação e do Comportamento, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66075-110, PA, Brazil; (D.A.L.); (S.d.C.C.); (C.C.S.d.M.d.S.); (B.G.P.); (K.M.M.M.F.); (E.A.F.-J.)
| | - Luanna de Melo Pereira Fernandes
- Departamento de Ciências Morfológicas e Fisiológicas, Centro das Ciências Biológicas e da Saúde (CCBS), Universidade Estadual do Pará, Belém 66095-100, PA, Brazil;
| | - Enéas Andrade Fontes-Júnior
- Laboratório de Farmacologia da Inflamação e do Comportamento, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66075-110, PA, Brazil; (D.A.L.); (S.d.C.C.); (C.C.S.d.M.d.S.); (B.G.P.); (K.M.M.M.F.); (E.A.F.-J.)
| | - Cristiane do Socorro Ferraz Maia
- Laboratório de Farmacologia da Inflamação e do Comportamento, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66075-110, PA, Brazil; (D.A.L.); (S.d.C.C.); (C.C.S.d.M.d.S.); (B.G.P.); (K.M.M.M.F.); (E.A.F.-J.)
- Correspondence:
| |
Collapse
|
18
|
Glazer L, Brennan CH. Developmental Exposure to Low Concentrations of Methylmercury Causes Increase in Anxiety-Related Behaviour and Locomotor Impairments in Zebrafish. Int J Mol Sci 2021; 22:10961. [PMID: 34681620 PMCID: PMC8535691 DOI: 10.3390/ijms222010961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/22/2021] [Accepted: 09/29/2021] [Indexed: 11/17/2022] Open
Abstract
Methylmercury (MeHg) is a ubiquitous pollutant shown to cause developmental neurotoxicity, even at low levels. However, there is still a large gap in our understanding of the mechanisms linking early-life exposure to life-long behavioural impairments. Our aim was to characterise the short- and long-term effects of developmental exposure to low doses of MeHg on anxiety-related behaviours in zebrafish, and to test the involvement of neurological pathways related to stress-response. Zebrafish embryos were exposed to sub-acute doses of MeHg (0, 5, 10, 15, 30 nM) throughout embryo-development, and tested for anxiety-related behaviours and locomotor activity at larval (light/dark locomotor activity) and adult (novel tank and tap assays) life-stages. Exposure to all doses of MeHg caused increased anxiety-related responses; heightened response to the transition from light to dark in larvae, and a stronger dive response in adults. In addition, impairment in locomotor activity was observed in the higher doses in both larvae and adults. Finally, the expressions of several neural stress-response genes from the HPI-axis and dopaminergic system were found to be disrupted in both life-stages. Our results provide important insights into dose-dependent differences in exposure outcomes, the development of delayed effects over the life-time of exposed individuals and the potential mechanisms underlying these effects.
Collapse
Affiliation(s)
- Lilah Glazer
- Nanchang Joint Programme, School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Caroline H. Brennan
- Department of Biological and Experimental Psychology, School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK;
| |
Collapse
|
19
|
Iman IN, Yusof NAM, Talib UN, Ahmad NAZ, Norazit A, Kumar J, Mehat MZ, Jayabalan N, Muthuraju S, Stefaniuk M, Kaczmarek L, Muzaimi M. The IntelliCage System: A Review of Its Utility as a Novel Behavioral Platform for a Rodent Model of Substance Use Disorder. Front Behav Neurosci 2021; 15:683780. [PMID: 34149373 PMCID: PMC8211779 DOI: 10.3389/fnbeh.2021.683780] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
The use of animal models for substance use disorder (SUD) has made an important contribution in the investigation of the behavioral and molecular mechanisms underlying substance abuse and addiction. Here, we review a novel and comprehensive behavioral platform to characterize addiction-like traits in rodents using a fully automated learning system, the IntelliCage. This system simultaneously captures the basic behavioral navigation, reward preference, and aversion, as well as the multi-dimensional complex behaviors and cognitive functions of group-housed rodents. It can reliably capture and track locomotor and cognitive pattern alterations associated with the development of substance addiction. Thus, the IntelliCage learning system offers a potentially efficient, flexible, and sensitive tool for the high-throughput screening of the rodent SUD model.
Collapse
Affiliation(s)
- Ismail Nurul Iman
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Nurul Aiman Mohd Yusof
- Department of Anatomy, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Ummi Nasrah Talib
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Nur Aimi Zawami Ahmad
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Anwar Norazit
- Department of Biomedical Sciences, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Muhammad Zulfadli Mehat
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Nanthini Jayabalan
- Translational Neuroscience Lab, UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Sangu Muthuraju
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Marzena Stefaniuk
- BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Leszek Kaczmarek
- BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Mustapha Muzaimi
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
20
|
Lian M, Field CL, van Wijngaarden E, Rios C, Castellini JM, Greig DJ, Rea LD, Coleman DJ, Thomson CE, Gulland FMD, O'Hara TM. Assessment of clinical outcomes associated with mercury concentrations in harbor seal pups (Phoca vitulina richardii) in central California. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 758:143686. [PMID: 33279198 DOI: 10.1016/j.scitotenv.2020.143686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 06/12/2023]
Abstract
Monomethyl mercury (MeHg+) from the diet can cause mild to severe neurotoxicosis in fish-eating mammals. Chronic and low-level in utero exposure also can be neurotoxic, as documented in laboratory animal studies and epidemiologic investigations. In free-ranging animals, it is challenging to study low-level exposure related neurotoxicosis, and few studies have investigated the relationship between mercury (Hg) and adverse outcomes in wild populations. Relative to Hg concentrations on admission we evaluated different types of behaviors for 267 Pacific harbor seal (HS; Phoca vitulina richardii) pups at The Marine Mammal Center from 2015 to 2019 during rehabilitation after stranding and maternal separation. Admitted HS pups underwent a clinical exam; including sex and weight determination, and hair (partly lanugo grown in utero) and blood samples were collected for total Hg concentration ([THg]) determination. All pups were monitored weekly (behavior assessments included response to tactile stimulation, movement, swimming, interactions with other seals, hand feeding, and feeding independently), and days in rehabilitation and survival were recorded. There was a significant negative correlation between [THg] and responses to tactile stimulation and movements, measured in both hair and whole blood (p < 0.05). This relationship was found both during the intensive care unit (ICU) stage, and during the pool stage of rehabilitation. Additionally, there was a significant association between greater [THg] and number of days spent in rehabilitation, although there was no relationship between [THg] and survival. There was a significant sex difference, with greater [THg] in female pups, which contrasts with previously published findings in juvenile and adult harbor seals. Our findings support small, but significant associations between gestational THg exposure and clinical effects for tactile sensory response and movement, and longer rehabilitation durations for HS pups, although there was considerable variability among animals.
Collapse
Affiliation(s)
- Marianne Lian
- Department of Veterinary Medicine, University of Alaska Fairbanks, 2141 Koyokuk Dr, Fairbanks, AK 99775-7750, USA; Department of Chemistry and Biochemistry, University of Alaska Fairbanks, 900 Yukon Dr Rm. 194, Fairbanks, AK 99775-6160, USA.
| | - Cara L Field
- The Marine Mammal Center, 2000 Bunker Road, Fort Cronkhite, Sausalito, CA 94965, USA
| | - Edwin van Wijngaarden
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Carlos Rios
- The Marine Mammal Center, 2000 Bunker Road, Fort Cronkhite, Sausalito, CA 94965, USA
| | - J Margaret Castellini
- Department of Veterinary Medicine, University of Alaska Fairbanks, 2141 Koyokuk Dr, Fairbanks, AK 99775-7750, USA
| | - Denise J Greig
- The Marine Mammal Center, 2000 Bunker Road, Fort Cronkhite, Sausalito, CA 94965, USA
| | - Lorrie D Rea
- Water and Environmental Research Center, Institute of Northern Engineering, University of Alaska Fairbanks, 1764 Tanana Loop, Fairbanks, AK 99775, USA
| | - Denver J Coleman
- The Marine Mammal Center, 2000 Bunker Road, Fort Cronkhite, Sausalito, CA 94965, USA
| | - Christine E Thomson
- Department of Veterinary Medicine, University of Alaska Fairbanks, 2141 Koyokuk Dr, Fairbanks, AK 99775-7750, USA; Animal Referral Hospital Brisbane, Sinnamon Park, Brisbane, Queensland, Australia
| | - Frances M D Gulland
- The Marine Mammal Center, 2000 Bunker Road, Fort Cronkhite, Sausalito, CA 94965, USA
| | - Todd M O'Hara
- Department of Veterinary Medicine, University of Alaska Fairbanks, 2141 Koyokuk Dr, Fairbanks, AK 99775-7750, USA; Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
21
|
DNA methyltransferase- and histone deacetylase-mediated epigenetic alterations induced by low-level methylmercury exposure disrupt neuronal development. Arch Toxicol 2021; 95:1227-1239. [PMID: 33454822 DOI: 10.1007/s00204-021-02984-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 01/06/2021] [Indexed: 10/22/2022]
Abstract
Methylmercury (MeHg) is a chemical substance that causes adverse effects on fetal development. However, the molecular mechanisms by which environmental MeHg affects fetal development have not been clarified. Recently, it has been suggested that the toxic effects of chemicals on fetal development are related alterations in epigenetics, such as DNA methylation and histone modification. In order to analyze the epigenetic effects of low-level MeHg exposure on neuronal development, we evaluated neuronal development both in vivo and in vitro. Pregnant mice (C57BL/6J) were orally administrated 3 mg/kg of MeHg once daily from embryonic day 12-14. Fetuses were removed on embryonic day 19 and brain tissues were collected. LUHMES cells were treated with 1 nM of MeHg for 6 days and collected on the last day of treatment. In both in vivo and in vitro samples, MeHg significantly suppressed neurite outgrowth. Decreased acetylated histone H3 (AcH3) levels and increased histone deacetylase (HDAC) 3 and HDAC6 levels were observed in response to MeHg treatment in both in vivo and in vitro experiments. In addition, increased DNA methylation and DNA methyltransferase 1 (DNMT1) levels were observed in both in vivo and in vitro experiments. The inhibition of neurite outgrowth resulting from MeHg exposure was restored by co-treatment with DNMT inhibitor or HDAC inhibitors. Our results suggest that neurological effects such as reduced neurite outgrowth due to low-level MeHg exposure result from epigenetic changes, including a decrease in AcH3 via increased HDAC levels and an increase in DNA methylation via increased DNMT1 levels.
Collapse
|
22
|
Dridi I, Soulimani R, Bouayed J. Chronic depression-like phenotype in male offspring mice following perinatal exposure to naturally contaminated eels with a mixture of organic and inorganic pollutants. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:156-165. [PMID: 32297116 DOI: 10.1007/s11356-020-08799-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/06/2020] [Indexed: 06/11/2023]
Abstract
Previously, we demonstrated that maternal exposure to high, intermediate, or lowly contaminated European eels with a mixture of chemicals, during pregnancy and lactation, resulted in adult despair-like behavior, selectively in male offspring mice. Here, we investigate if depression-like behavior in offspring males was transient or permanent by monitoring immobility behavior, a measure of behavioral despair, at three distinct stages of life, including young adult (post-natal day (PND) 55), mature adult (PND 200) and middle (PNDs 335-336) age, in the forced swimming (FST) and the tail suspension (TST) tests. Oxidative stress markers including malondialdehyde (MDA) levels and superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) activities were evaluated in the hippocampus, prefrontal cortex, and cerebellum of middle-aged animals. Findings showed a significant enhancement of immobility behavior in the TST performed at young adult age (all p < 0.05) in the FST carried out at mature adult age (all p < 0.001) and in both behavioral tests realized at middle age (all p < 0.05, except one p = 0.06) in mice perinatally exposed to eels compared with non-exposed controls. Antioxidant-related enzyme activities, including SOD and CAT, were only elevated in the hippocampus of middle-aged males perinatally exposed to the two more polluted eels (all p < 0.05). Further, lipid peroxidation, assessed by MDA levels, was not found to be differentially regulated in the selected areas of middle-aged brains of exposed mice (all p > 0.05). Collectively, this suggested limited oxidative metabolism disturbances in middle-aged brains exposed to eels. In summary, our results highlighted that offspring males perinatally exposed to naturally contaminated reared and river eels with persistent organic pollutants (POPs) and heavy metals displayed chronic depression-like phenotype. As extrapolation of data to humans should be done with precaution, retrospective and prospective epidemiological studies are needed to clarify this potential relationship, stressed in our animal model, between maternal polluted fish consumption and chronically low mood in offspring.
Collapse
Affiliation(s)
- Imen Dridi
- LCOMS/Neurotoxicologie Alimentaire et Bioactivité, Université de Lorraine, 57000, Metz, France
| | - Rachid Soulimani
- LCOMS/Neurotoxicologie Alimentaire et Bioactivité, Université de Lorraine, 57000, Metz, France
| | - Jaouad Bouayed
- LCOMS/Neurotoxicologie Alimentaire et Bioactivité, Université de Lorraine, 57000, Metz, France.
| |
Collapse
|
23
|
Ijomone OM, Ijomone OK, Iroegbu JD, Ifenatuoha CW, Olung NF, Aschner M. Epigenetic influence of environmentally neurotoxic metals. Neurotoxicology 2020; 81:51-65. [PMID: 32882300 PMCID: PMC7708394 DOI: 10.1016/j.neuro.2020.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
Continuous globalization and industrialization have ensured metals are an increasing aspect of daily life. Their usefulness in manufacturing has made them vital to national commerce, security and global economy. However, excess exposure to metals, particularly as a result of environmental contamination or occupational exposures, has been detrimental to overall health. Excess exposure to several metals is considered environmental risk in the aetiology of several neurological and neurodegenerative diseases. Metal-induced neurotoxicity has been a major health concern globally with intensive research to unravel the mechanisms associated with it. Recently, greater focus has been directed at epigenetics to better characterize the underlying mechanisms of metal-induced neurotoxicity. Epigenetic changes are those modifications on the DNA that can turn genes on or off without altering the DNA sequence. This review discusses how epigenetic changes such as DNA methylation, post translational histone modification and noncoding RNA-mediated gene silencing mediate the neurotoxic effects of several metals, focusing on manganese, arsenic, nickel, cadmium, lead, and mercury.
Collapse
Affiliation(s)
- Omamuyovwi M Ijomone
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria.
| | - Olayemi K Ijomone
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria; Department of Anatomy, University of Medical Sciences, Ondo, Nigeria
| | - Joy D Iroegbu
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Chibuzor W Ifenatuoha
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Nzube F Olung
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Michael Aschner
- Departments of Molecular Pharmacology and Neurosciences, Albert Einstein College of Medicine, NY, USA.
| |
Collapse
|
24
|
Rand MD, Conrad K, Marvin E, Harvey K, Henderson D, Tawil R, Sobolewski M, Cory-Slechta DA. Developmental exposure to methylmercury and resultant muscle mercury accumulation and adult motor deficits in mice. Neurotoxicology 2020; 81:1-10. [PMID: 32735808 PMCID: PMC7708410 DOI: 10.1016/j.neuro.2020.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/06/2020] [Accepted: 07/17/2020] [Indexed: 11/20/2022]
Abstract
Developmental methylmercury (MeHg) exposure can have lasting consequences on neural development and motor function across the lifespan. Recent evidence for MeHg targeting of myogenic pathways has drawn attention to the possibility that developing skeletal muscle plays a role in the motor deficits stemming from early life MeHg exposure. In this study we examined a potential role for muscle in influencing MeHg developmental toxicity in offspring of female mice exposed to MeHg via drinking water. Dams had access to 0, 0.5 or 5.0 ppm MeHg chloride in drinking water from two weeks prior to mating through weaning. Blood, brain and muscle tissue was harvested from dams at weaning and pups at postnatal days (PND) 6, 21 and 60 for analysis of total Hg. Muscle tissue sections were examined with histological stains. Behavioral testing of offspring was conducted at PND 60 and included locomotor activity, inverted screen, grip strength and rotarod tests to assess motor function. Total Hg (tHg) levels in dam muscles at weaning were 1.7-3-fold higher than Hg levels in blood or brain. In PND6 male and female pups, muscle and brain tHg levels were 2 to 4-fold higher than blood tHg. Brain tHg levels decreased more rapidly than muscle tHg levels between PND 6 and 21. Premised on modeling of growth dilution, brain tissue demonstrated an elimination of tHg while muscle tissue exhibited a net uptake of tHg between PND 6 and 21. Despite overall elevated Hg levels in developing muscle, no gross morphological or cytological phenotypes were observed in muscle at PND 60. At the higher MeHg dose, grip strength was reduced in both females and males at PND 60, whereas only male specific deficits were observed in locomotor activity and inverted screen tests with marginally significant deficits on rotarod. These findings highlight a potential role for developing skeletal muscle in mediating the neuromuscular insult of early life MeHg exposure.
Collapse
Affiliation(s)
| | | | - Elena Marvin
- Department of Environmental Medicine, United States
| | | | - Don Henderson
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Rabi Tawil
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | | | | |
Collapse
|
25
|
Rossa-Roccor V, Karim ME. Are US adults with low-exposure to methylmercury at increased risk for depression? A study based on 2011-2016 National Health and Nutrition Examination Surveys (NHANES). Int Arch Occup Environ Health 2020; 94:419-431. [PMID: 33104857 DOI: 10.1007/s00420-020-01592-9] [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: 02/04/2020] [Accepted: 10/09/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Depression is a highly-prevalent disorder among US adults and despite advancements in treatment options, prevalence rates are increasing. With the emerging recommendations of dietary interventions such as high fish intake come potential risks, for example, exposure to methylmercury (MeHg). Case reports and animal models have suggested a possible association of high doses of MeHg with psychiatric symptoms; the impact of low-dose exposure on depression remains unknown. METHODS In this cross-sectional study, survey-weighted logistic regression models were built to assess the relationship between low-dose MeHg blood levels and depression in a sample of n = 3930 adults from the National Health and Nutrition Examination Survey (NHANES) from 2011 to 2016. RESULTS 9.1% (n = 1335) of the respondents screened positive for depression; all participants had MeHg blood levels below the US Environmental Protection Agency's reference dose. The adjusted multivariate logistic regression model showed no statistically significant association between MeHg blood levels and depression. CONCLUSION Low-dose MeHg does not seem to be associated with depression in this study. However, dietary recommendations with regards to fish intake should be made cautiously. Further studies are needed, especially considering predicted increasing environmental pollution of our food webs and the potentially higher vulnerability of subpopulations such as pregnant women.
Collapse
Affiliation(s)
- Verena Rossa-Roccor
- School of Population and Public Health, University of British Columbia, 2206 East Mall, Vancouver, BC, V6T1Z3, Canada.
| | - M Ehsan Karim
- School of Population and Public Health, University of British Columbia, 2206 East Mall, Vancouver, BC, V6T1Z3, Canada.,Centre for Health Evaluation and Outcome Sciences, University of British Columbia, 1081 Burrard Street, Vancouver, BC, V6Z1Y6, Canada
| |
Collapse
|
26
|
Kimura E, Suzuki G, Uramaru N, Endo T, Maekawa F. Behavioral impairments in infant and adult mouse offspring exposed to 2,3,7,8-tetrabromodibenzofuran in utero and via lactation. ENVIRONMENT INTERNATIONAL 2020; 142:105833. [PMID: 32559560 DOI: 10.1016/j.envint.2020.105833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/19/2020] [Accepted: 05/22/2020] [Indexed: 06/11/2023]
Abstract
Polybrominated dibenzo-p-dioxins and dibenzofurans (PBDD/DFs) have been unintentionally produced and emitted from the lifecycle of products containing brominated flame retardants, such as polybrominated diphenyl ether, which is suspected to cause developmental neurotoxicity (DNT). Although it is plausible that PBDD/DFs can also induce DNT, information regarding their neurotoxic potential is currently limited. Hence, in the present study, we examined the effects of in utero and lactational exposure to brominated dibenzofurans on infant and adult offspring behavior to understand the mechanism of PBDD/DFs toxicity and detect effective behavioral endpoints in DNT assessment. We analyzed the behavior of mouse offspring born to dams administered 2,3,7,8-tetrabromodibenzofuran (2,3,7,8-TeBDF; dose of 0, 9, or 45 μg/kg) or 2,3,8-tribromodibenzofuran (2,3,8-TrBDF; dose of 0, 75.6, or 378 μg/kg) on gestational day 12.5. In mouse offspring born to dams exposed to 2,3,7,8-TeBDF, the exploratory behavior in a novel environment in adulthood and ultrasonic vocalization (USV) during infancy were significantly reduced. Additionally, AhR-target genes, such as Cyp1a1, were induced in the liver of 2,3,7,8-TeBDF-exposed offspring in a dose-dependent manner. Conversely, no significant changes in the infant and adult behaviors and expression level of AhR-target genes were observed in the 2,3,8-TrBDF-exposed offspring. These results suggest that 2,3,7,8-TeBDF can induce DNT and that the analysis of exploratory behavior in a novel environment and USV may be useful endpoints to assess DNT of dioxin-related substances.
Collapse
Affiliation(s)
- Eiki Kimura
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba 305-8506, Japan; Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo 102-0083, Japan
| | - Go Suzuki
- Center for Material Cycles and Waste Management Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba 305-8506, Japan
| | - Naoto Uramaru
- Nihon Pharmaceutical University, 10281 Komuro Ina-machi, Kitaadachi-gun, Saitama 362-0806, Japan
| | - Toshihiro Endo
- Phenovance Research & Technology, 5-4-19-302A, Kashiwanoha, Kashiwa, Chiba 277-0882, Japan
| | - Fumihiko Maekawa
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba 305-8506, Japan.
| |
Collapse
|
27
|
Hagiwara T, Hagino H, Ueda K, Nakama M, Minami T. Maternal exposure to methylmercury causes an impairment in ependymal cilia motility in the third ventricle and dilation of lateral ventricles in mice offspring. Birth Defects Res 2020; 112:1253-1259. [PMID: 32648687 DOI: 10.1002/bdr2.1750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 05/25/2020] [Accepted: 06/09/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND Although maternal MeHg-exposure causes hydrocephalus in the offspring of mice, its pathogenesis has not been fully explained. In the present study, we examined the issue of how maternal MeHg-exposure in mice affects ependymal ciliary movement in the offspring and whether the lateral ventricles in offspring show dilation. METHODS Pregnant mice were given drinking water containing 0, 10, or 20 mg/L MeHg, or a single dose of 2 mg/kg MeHg. Brain slices were prepared from the offspring and the ependymal ciliary movement of ependymal cells in the third ventricle were observed by a high-speed digital camera. The dilation of the lateral ventricles in the offspring was assessed by histological examination. RESULTS The administration of MeHg in the drinking water of pregnant mice at levels of 10 mg/L and 20 mg/L MeHg from GD10 to birth caused a significant decrease of ciliary beating frequency (CBF) in ependymal cells of the third ventricle in the weaned offspring. The ependymal ciliary movement of the weaned offspring was particularly sensitive in the case of the administration of MeHg at GD10. Moreover, there was a significant dilation of cross-sectional areas of lateral ventricles in weaned offspring from the pregnant mice that had been administered MeHg. The CBF and the cross-sectional areas of the lateral ventricles improved with time. CONCLUSIONS These results suggest that the impairment of ependymal ciliary movement by maternal MeHg-exposure contributes to the development of hydrocephalus in the offspring.
Collapse
Affiliation(s)
- Teruki Hagiwara
- Department of Life Science, School of Science & Engineering, Kindai University, Higashiosaka, Japan
| | - Hajime Hagino
- Department of Science, Interdisciplinary Graduate School of Science and Engineering, Kindai University, Higashiosaka, Japan
| | - Kaho Ueda
- Department of Science, Interdisciplinary Graduate School of Science and Engineering, Kindai University, Higashiosaka, Japan
| | - Mina Nakama
- Department of Science, Interdisciplinary Graduate School of Science and Engineering, Kindai University, Higashiosaka, Japan.,Division of Clinical Genetics, Gifu University Hospital, Gifu, Japan
| | - Takeshi Minami
- Department of Life Science, School of Science & Engineering, Kindai University, Higashiosaka, Japan.,Department of Science, Interdisciplinary Graduate School of Science and Engineering, Kindai University, Higashiosaka, Japan
| |
Collapse
|
28
|
Kiryk A, Janusz A, Zglinicki B, Turkes E, Knapska E, Konopka W, Lipp HP, Kaczmarek L. IntelliCage as a tool for measuring mouse behavior - 20 years perspective. Behav Brain Res 2020; 388:112620. [PMID: 32302617 DOI: 10.1016/j.bbr.2020.112620] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 03/23/2020] [Indexed: 12/21/2022]
Abstract
Since the 1980s, we have witnessed the rapid development of genetically modified mouse models of human diseases. A large number of transgenic and knockout mice have been utilized in basic and applied research, including models of neurodegenerative and neuropsychiatric disorders. To assess the biological function of mutated genes, modern techniques are critical to detect changes in behavioral phenotypes. We review the IntelliCage, a high-throughput system that is used for behavioral screening and detailed analyses of complex behaviors in mice. The IntelliCage was introduced almost two decades ago and has been used in over 150 studies to assess both spontaneous and cognitive behaviors. We present a critical analysis of experimental data that have been generated using this device.
Collapse
Affiliation(s)
- Anna Kiryk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Artur Janusz
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Bartosz Zglinicki
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Emir Turkes
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, Irving Medical Center, New York, NY, USA
| | - Ewelina Knapska
- BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Witold Konopka
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Hans-Peter Lipp
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland
| | - Leszek Kaczmarek
- BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
29
|
Yang L, Zhang Y, Wang F, Luo Z, Guo S, Strähle U. Toxicity of mercury: Molecular evidence. CHEMOSPHERE 2020; 245:125586. [PMID: 31881386 DOI: 10.1016/j.chemosphere.2019.125586] [Citation(s) in RCA: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/28/2019] [Accepted: 12/08/2019] [Indexed: 05/25/2023]
Abstract
Minamata disease in Japan and the large-scale poisoning by methylmercury (MeHg) in Iraq caused wide public concerns about the risk emanating from mercury for human health. Nowadays, it is widely known that all forms of mercury induce toxic effects in mammals, and increasing evidence supports the concern that environmentally relevant levels of MeHg could impact normal biological functions in wildlife. The information of mechanism involved in mercurial toxicity is growing but knowledge gaps still exist between the adverse effects and mechanisms of action, especially at the molecular level. A body of data obtained from experimental studies on mechanisms of mercurial toxicity in vivo and in vitro points to that disruption of the antioxidant system may play an important role in the mercurial toxic effects. Moreover, the accumulating evidence indicates that signaling transduction, protein or/and enzyme activity, and gene regulation are involving in mediating toxic and adaptive response to mercury exposure. We conducted here a comprehensive review of mercurial toxic effects on wildlife and human, in particular synthesized key findings of molecular pathways involved in mercurial toxicity from the cells to human. We discuss the molecular evidence related mercurial toxicity to the adverse effects, with particular emphasis on the gene regulation. The further studies relying on Omic analysis connected to adverse effects and modes of action of mercury will aid in the evaluation and validation of causative relationship between health outcomes and gene expression.
Collapse
Affiliation(s)
- Lixin Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China.
| | - Yuanyuan Zhang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Feifei Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Zidie Luo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Shaojuan Guo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Uwe Strähle
- Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
30
|
Pregnant rats exposed to low-level methylmercury exhibit cerebellar synaptic and neuritic remodeling during the perinatal period. Arch Toxicol 2020; 94:1335-1347. [DOI: 10.1007/s00204-020-02696-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 03/02/2020] [Indexed: 12/14/2022]
|
31
|
Bacteria affect Caenorhabditis elegans responses to MeHg toxicity. Neurotoxicology 2019; 75:129-135. [PMID: 31542475 DOI: 10.1016/j.neuro.2019.09.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/02/2019] [Indexed: 01/06/2023]
Abstract
The organic form of mercurial complex, methylmercury (MeHg), is a neurotoxin that bioaccumulates in the food web. Studies in model organisms, such as Caenorhabditis elegans (C. elegans), provide powerful insights on the role of genetic factors in MeHg-induced toxicity. C. elegans is a free living worm that is commonly cultured in nematode growth medium (NGM) agar plates seeded with bacteria. The bacteria have broad impact on C. elegans biology, including development, reproduction and lifespan, as well as metabolism of experimental chemicals. We hypothesized that MeHg toxicity in C. elegans could be modified by the bacterial food. Using a C. elegans wild-type (WT) strain and transgenic reporter strains, we found that bacterial food reduced the chronic toxicity of MeHg in C. elegans in a dose- and live-status-dependent manner. The MeHg-induced death rate in C. elegans was highest in fasted worms, followed by dehydrated dead bacteria, dead bacteria and live bacteria fed worms. Among the different bacterial foods, dehydrated dead bacteria fed worms were most sensitive to the toxicity of MeHg. The distinct bacteria food (dehydrated dead bacteria food) attenuated oxidative stress and development delay in C. elegans exposed to MeHg. The FOXO transcriptional factor DAF16 was not changed by MeHg but modified by the distinct bacteria food. Independent of MeHg treatment, daf-16 expression in fed worms migrated from the intestine to muscle. We conclude that, in chronic exposure studies in C. elegans, the effects of bacteria on toxicological outcomes should be considered.
Collapse
|
32
|
Reardon AJF, Karathra J, Ribbenstedt A, Benskin JP, MacDonald AM, Kinniburgh DW, Hamilton TJ, Fouad K, Martin JW. Neurodevelopmental and Metabolomic Responses from Prenatal Coexposure to Perfluorooctanesulfonate (PFOS) and Methylmercury (MeHg) in Sprague-Dawley Rats. Chem Res Toxicol 2019; 32:1656-1669. [PMID: 31340646 DOI: 10.1021/acs.chemrestox.9b00192] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Methylmercury (MeHg) and perfluorooctanesulfonate (PFOS) are major contaminants of human blood that are both common in dietary fish, thereby raising questions about their combined impact on human development. Here, pregnant Sprague-Dawley rats ingested a daily dose, from gestational day 1 through to weaning, of either 1 mg/kg bw PFOS (PFOS-only), 1 mg/kg MeHg (MeHg-only), a mixture of 0.1 mg/kg PFOS and 1 mg/kg MeHg (Low-Mix), or of 1 mg/kg of PFOS and 1 mg/kg MeHg (High-Mix). Newborns were monitored for physical milestones and reflexive developmental responses, and in juveniles the spontaneous activity, anxiety, memory, and cognition were assessed. Targeted metabolomics of 199 analytes was applied to sectioned brain regions of juvenile offspring. Newborns in the High-Mix group had decreased weight gain as well as delayed reflexes and innate behavioral responses compared to controls and individual chemical groups indicating a toxicological interaction on early development. In juveniles, cumulative mixture effects increased in a dose-dependent manner in tests of anxiety-like behavior. However, other developmental test results suggested antagonism, as PFOS-only and MeHg-only juveniles had increased hyperactivity and thigmotaxic behavior, respectively, but fewer effects in Low-Mix and High-Mix groups. Consistent with these behavioral observations, a pattern of antagonism was also observed in neurochemicals measured in rat cortex, as PFOS-only and MeHg-only juveniles had altered concentrations of metabolites (e.g., lipids, amino acids, and biogenic amines), while no changes were evident in the combined exposures. The cortical metabolites altered in PFOS-only and MeHg-only exposed groups are involved in inhibitory and excitatory neurotransmission. These proof-of-principle findings at relatively high doses indicate the potential for toxicological interaction between PFOS and MeHg, with developmental-stage specific effects. Future mixture studies at lower doses are warranted, and prospective human birth cohorts should consider possible confounding effects from PFOS and mercury exposure on neurodevelopment.
Collapse
Affiliation(s)
- Anthony J F Reardon
- Department of Laboratory Medicine and Pathology , University of Alberta , Edmonton , Alberta T6G 2G3 , Canada
| | - Jacqueline Karathra
- Department of Laboratory Medicine and Pathology , University of Alberta , Edmonton , Alberta T6G 2G3 , Canada
| | - Anton Ribbenstedt
- Department of Environmental Science and Analytical Chemistry (ACES) , Stockholm University , Stockholm SE-11418 , Sweden
| | - Jonathan P Benskin
- Department of Environmental Science and Analytical Chemistry (ACES) , Stockholm University , Stockholm SE-11418 , Sweden
| | - Amy M MacDonald
- Alberta Centre for Toxicology , University of Calgary , Calgary , Alberta T2N 1N4 , Canada
| | - David W Kinniburgh
- Alberta Centre for Toxicology , University of Calgary , Calgary , Alberta T2N 1N4 , Canada
| | - Trevor J Hamilton
- Department of Psychology , MacEwan University , Edmonton , Alberta T5J 4S2 , Canada
| | - Karim Fouad
- Department of Physical Therapy , University of Alberta , Edmonton , Alberta T6G 2G4 , Canada
| | - Jonathan W Martin
- Department of Laboratory Medicine and Pathology , University of Alberta , Edmonton , Alberta T6G 2G3 , Canada.,Department of Environmental Science and Analytical Chemistry (ACES) , Stockholm University , Stockholm SE-11418 , Sweden
| |
Collapse
|
33
|
Goodrich JM, Ingle ME, Domino SE, Treadwell MC, Dolinoy DC, Burant C, Meeker JD, Padmanabhan V. First trimester maternal exposures to endocrine disrupting chemicals and metals and fetal size in the Michigan Mother-Infant Pairs study. J Dev Orig Health Dis 2019; 10:447-458. [PMID: 30696509 PMCID: PMC6660406 DOI: 10.1017/s204017441800106x] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Exposures to endocrine disrupting chemicals and metals are near ubiquitous worldwide, and their potential impact on children is a major public health concern. This pilot study was designed to characterize exposures to phthalates, phenols and metals among pregnant women in the first trimester, and to examine associations with fetal biometrics and birth weight. A total of 41 chemicals and elements were analyzed in urine from 56 mothers with full-term newborns from the Michigan Mother-Infant Pairs study. Bivariate analyses identified predictors of exposure biomarkers. Associations between birth weight, Fenton z-scores and second trimester fetal biometrics with toxicants were examined via multivariable linear regression. An average of 30 toxicants were detected in maternal urine. Fast food consumption was associated with several phthalate metabolites, phenols and metals, and canned food consumption with bisphenol F (P <0.05). Mono (3-carboxypropyl) phthalate was significantly associated with higher birth weight and Fenton z-score while the opposite was observed for bisphenol S. Estimated femur length from ultrasonography was significantly inversely associated with arsenic, barium and lead. While limited by sample size, this study is one of the first to evaluate birth outcomes with respect to emerging endocrine disrupting chemicals and to examine associations between toxicants and fetal biometrics. Exposure assessment was provided by the National Institute of Environmental Health Sciences' Children's Health Exposure Analysis Resource (NIEHS CHEAR), a resource available to children's studies with the goal of combining data across cohorts in an effort to characterize the impact of toxicants on child health from birth and beyond.
Collapse
Affiliation(s)
- Jaclyn M. Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
| | - Mary E. Ingle
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
| | - Steven E. Domino
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Marjorie C. Treadwell
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Dana C. Dolinoy
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
| | - Charles Burant
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - John D. Meeker
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
| | - Vasantha Padmanabhan
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
34
|
Perez-Fernandez C, Flores P, Sánchez-Santed F. A Systematic Review on the Influences of Neurotoxicological Xenobiotic Compounds on Inhibitory Control. Front Behav Neurosci 2019; 13:139. [PMID: 31333425 PMCID: PMC6620897 DOI: 10.3389/fnbeh.2019.00139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/06/2019] [Indexed: 01/24/2023] Open
Abstract
Background: Impulsive and compulsive traits represent a variety of maladaptive behaviors defined by the difficulties to stop an improper response and the control of a repeated behavioral pattern without sensitivity to changing contingencies, respectively. Otherwise, human beings are continuously exposed to plenty neurotoxicological agents which have been systematically linked to attentional, learning, and memory dysfunctions, both preclinical and clinical studies. Interestingly, the link between both impulsive and compulsive behaviors and the exposure to the most important xenobiotic compounds have been extensively developed; although the information has been rarely summarized. For this, the present systematic review schedule and analyze in depth the most important works relating different subtypes of the above-mentioned behaviors with 4 of the most important xenobiotic compounds: Lead (Pb), Methylmercury (MeHg), Polychlorinated biphenyls (PCB), and Organophosphates (OP) in both preclinical and clinical models. Methods: Systematic search strategy on PubMed databases was developed, and the most important information was structured both in text and in separate tables based on rigorous methodological quality assessment. Results: For Lead, Methylmercury, Polychlorinated biphenyls and organophosphates, a total of 44 (31 preclinical), 34 (21), 38 (23), and 30 (17) studies were accepted for systematic synthesis, respectively. All the compounds showed an important empirical support on their role in the modulation of impulsive and, in lesser degree, compulsive traits, stronger and more solid in animal models with inconclusive results in humans in some cases (i.e., MeHg). However, preclinical and clinical studies have systematically focused on different subtypes of the above-mentioned behaviors, as well as impulsive choice or habit conformations have been rarely studied. Discussion: The strong empirical support in preclinical studies contrasts with the lack of connection between preclinical and clinical models, as well as the different methodologies used. Further research should be focused on dissipate these differences as well as deeply study impulsive choice, decision making, risk taking, and cognitive flexibility, both in experimental animals and humans.
Collapse
Affiliation(s)
| | - Pilar Flores
- Department of Psychology and Health Research Center, University of Almería, Almería, Spain
| | | |
Collapse
|
35
|
Raciti M, Salma J, Spulber S, Gaudenzi G, Khalajzeyqami Z, Conti M, Anderlid BM, Falk A, Hermanson O, Ceccatelli S. NRXN1 Deletion and Exposure to Methylmercury Increase Astrocyte Differentiation by Different Notch-Dependent Transcriptional Mechanisms. Front Genet 2019; 10:593. [PMID: 31316548 PMCID: PMC6610538 DOI: 10.3389/fgene.2019.00593] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 06/05/2019] [Indexed: 01/11/2023] Open
Abstract
Controversial evidence points to a possible involvement of methylmercury (MeHg) in the etiopathogenesis of autism spectrum disorders (ASD). In the present study, we used human neuroepithelial stem cells from healthy donors and from an autistic patient bearing a bi-allelic deletion in the gene encoding for NRXN1 to evaluate whether MeHg would induce cellular changes comparable to those seen in cells derived from the ASD patient. In healthy cells, a subcytotoxic concentration of MeHg enhanced astroglial differentiation similarly to what observed in the diseased cells (N1), as shown by the number of GFAP positive cells and immunofluorescence signal intensity. In both healthy MeHg-treated and N1 untreated cells, aberrations in Notch pathway activity seemed to play a critical role in promoting the differentiation toward glia. Accordingly, treatment with the established Notch inhibitor DAPT reversed the altered differentiation. Although our data are not conclusive since only one of the genes involved in ASD is considered, the results provide novel evidence suggesting that developmental exposure to MeHg, even at subcytotoxic concentrations, induces alterations in astroglial differentiation similar to those observed in ASD.
Collapse
Affiliation(s)
- Marilena Raciti
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jahan Salma
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Stefan Spulber
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Giulia Gaudenzi
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Mirko Conti
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Britt-Marie Anderlid
- Centre for Molecular Medicine, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Ceccatelli
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
36
|
Akiyama M, Unoki T, Shinkai Y, Ishii I, Ida T, Akaike T, Yamamoto M, Kumagai Y. Environmental Electrophile-Mediated Toxicity in Mice Lacking Nrf2, CSE, or Both. ENVIRONMENTAL HEALTH PERSPECTIVES 2019; 127:67002. [PMID: 31166132 PMCID: PMC6794492 DOI: 10.1289/ehp4949] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 06/01/2023]
Abstract
BACKGROUND Transcription factor Nrf2 (nuclear factor-erythroid 2-related factor 2) plays a key role in detoxification of electrophiles via formation of glutathione (GSH) adducts and subsequent excretion into extracellular spaces. We found that reactive sulfur species (RSS), such as cysteine persulfides produced by cystathionine [Formula: see text] (CSE), capture environmental electrophiles through formation of sulfur adducts. However, contributions of Nrf2 and CSE to the blockage of environmental electrophile-mediated toxicity remain to be evaluated. OBJECTIVES The aim of this study was to clarify roles that CSE and Nrf2 play in the protection against various environmental electrophiles. We also wished to clarify the molecular basis of the developmental window of toxicity through investigating expression levels of Nrf2, RSS-producing enzymes, and sulfur nucleophiles during developmental stages of mice. METHODS Wild-type (WT), CSE knockout (KO), Nrf2 KO, Nrf2/CSE double KO (DKO) mice, and their primary hepatocytes were analyzed in this study. Cadmium (Cd), methylmercury (MeHg), 1,4-naphthoquinone, crotonaldehyde, and acrylamide were used. We conducted Western blotting, real-time polymerase chain reaction (PCR), 3-(4,5-dimethylthiazol-2-yl)-2,5-triphenyl tetrazolium bromide (MTT) assays, liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) analysis, alanine transaminase (ALT) activity, histopathological analysis, and rotarod test. RESULTS Primary hepatocytes from DKO mice were significantly more sensitive to the environmental electrophiles than each single KO counterpart. Both Nrf2 and CSE single KO mice were highly susceptible to Cd and MeHg, and such sensitivity was further exacerbated in the DKO mice. Lower-level expressions of CSE and sulfur nucleophiles than those in adult mice were observed in a window of developmental stage. CONCLUSIONS Our mouse model provided new insights into the response to environmental electrophiles; while Nrf2 is recognized as a key transcription factor for detoxification of environmental electrophiles, CSE is crucial factor to repress their toxicity in a parallel mode. In addition, the sensitivity of fetuses to MeHg appears to be, at least in part, associated with the restricted production of RSS due to low-level expression of CSE. https://doi.org/10.1289/EHP4949.
Collapse
Affiliation(s)
- Masahiro Akiyama
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takamitsu Unoki
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Basic Medical Sciences, National Institute for Minamata Disease, Minamata, Japan
| | - Yasuhiro Shinkai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Isao Ishii
- Laboratory of Health Chemistry, Showa Pharmaceutical University, Tokyo, Japan
| | - Tomoaki Ida
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takaaki Akaike
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshito Kumagai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
37
|
Maqbool F, Bahadar H, Hassani S, Niaz K, Baeeri M, Rahimifard M, Ghasemi-Niri SF, Abdollahi M. Biochemical evidence on the potential role of methyl mercury in hepatic glucose metabolism through inflammatory signaling and free radical pathways. J Cell Biochem 2019; 120:16195-16205. [PMID: 31081130 DOI: 10.1002/jcb.28899] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 03/12/2019] [Accepted: 03/22/2019] [Indexed: 12/18/2022]
Abstract
Methylmercury (MeHg) is an extremely important environmental toxicant posing serious health risks to human health and a big source of environmental pollutant. Numerous evidence available showing a link between nervous system toxicity and MeHg exposure. Other forms of mercury are reason of metabolic toxic effects and alteration of DNA in the human body. The sources of exposure could be occupational or other environmental settings. In the present study MeHg was orally gavaged to mice, at doses of 2.5, 5, and 10 mg/kg for 4 weeks. Fasting hyperglycemia, activity of hepatic phoshphoenolpyruvate carboxykinase and glucose 6-phoshphate were reported high as compared to control group. Inflammatory markers like, tumor necrosis factor α, the actual end product of inflammatory mediators' cascade pathway was also raised in comparison to control group. Hyperinsulinemia observed in serum showed clear understanding of mercury induced insulin resistance. Moreover, tissue damage due to increased oxidative stress markers like, hepatic lipid peroxidation, 8-deoxygunosine, reactive oxygen species, and carbonyl groups was significantly higher as compared to control group. MeHg caused a significant reduction in antioxidant markers like ferric reducing antioxidant power and total thiol molecules. The present study highlighted that activity of key enzymes involved in glucose metabolism is changed, owing to MeHg induced toxicity in the liver. Induction of similar toxic effects assumed to be stimulated by the production of high quantity free radicals.
Collapse
Affiliation(s)
- Faheem Maqbool
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Haji Bahadar
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Shokoufeh Hassani
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Kamal Niaz
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Baeeri
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahban Rahimifard
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyedeh Farnaz Ghasemi-Niri
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Kim W, Yoo DY, Jung HY, Kim JW, Hahn KR, Kwon HJ, Yoo M, Lee S, Nam SM, Yoon YS, Kim DW, Hwang IK. Leaf extracts from Dendropanax morbifera Léveille mitigate mercury-induced reduction of spatial memory, as well as cell proliferation, and neuroblast differentiation in rat dentate gyrus. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:94. [PMID: 31046739 PMCID: PMC6498467 DOI: 10.1186/s12906-019-2508-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 04/17/2019] [Indexed: 01/09/2023]
Abstract
Background The brain is susceptible to methylmercury toxicity, which causes irreversible damage to neurons and glia and the leaf extract Dendropanax morbifera Léveille (DML) has various biological functions in the nervous system. In this study, we examined the effects of DML on mercury-induced proliferating cells and differentiated neuroblasts. Methods Dimethylmercury (5 μg/kg) and galantamine (5 mg/kg) was administered intraperitoneally and/or DML (100 mg/kg) was orally to 7-week-old rats every day for 36 days. One hour after the treatment, novel object recognition test was examined. In addition, spatial probe tests were conducted on the 6th day after 5 days of continuous training in the Morris swim maze. Thereafter, the rats were euthanized for immunohistochemical staining analysis with Ki67 and doublecortin and measurement for acetylcholinesterase (AChE) activity. Results Dimethylmercury-treated rats showed reduced discrimination index in novel object recognition test and took longer to find the platform than did control group. Compared with dimethylmercury treatment alone, supplementation with DML or galatamine significantly ameliorated the reduction of discrimination index and reduced the time spent to find the platform. In addition, the number of platform crossings was lower in the dimethylmercury-treated group than in controls, while the administration of DML or galantamine significantly increased the number of crossings than did dimethylmercury treatment alone. Proliferating cells and differentiated neuroblasts, assessed by Ki67 and doublecortin immunohistochemical staining was significantly decreased in the dimethylmercury treated group versus controls. Supplementation with DML or galantamine significantly increased the number of proliferating cells and differentiated neuroblasts in the dentate gyrus. In addition, treatment with dimethylmercury significantly increased AChE activity in hippocampal homogenates, while treatment with dimethylmercury+DML or dimethylmercury+galantamine significantly ameliorated this increase. Conclusions These results suggest that DML may be a functional food that improves dimethylmercury-induced memory impairment and ameliorates dimethylmercury-induced reduction in proliferating cells and differentiated neuroblasts, and demonstrates corresponding activation of AChE activity in the dentate gyrus.
Collapse
|
39
|
The epigenetic dimension of psychological trauma — Prevention and psychotherapy. EUROPEAN JOURNAL OF TRAUMA & DISSOCIATION 2019. [DOI: 10.1016/j.ejtd.2019.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
40
|
Basnet RM, Zizioli D, Taweedet S, Finazzi D, Memo M. Zebrafish Larvae as a Behavioral Model in Neuropharmacology. Biomedicines 2019; 7:biomedicines7010023. [PMID: 30917585 PMCID: PMC6465999 DOI: 10.3390/biomedicines7010023] [Citation(s) in RCA: 215] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/22/2019] [Accepted: 03/22/2019] [Indexed: 12/21/2022] Open
Abstract
Zebrafish larvae show a clear and distinct pattern of swimming in response to light and dark conditions, following the development of a swim bladder at 4 days post fertilization. This swimming behavior is increasingly employed in the screening of neuroactive drugs. The recent emergence of high-throughput techniques for the automatic tracking of zebrafish larvae has further allowed an objective and efficient way of finding subtle behavioral changes that could go unnoticed during manual observations. This review highlights the use of zebrafish larvae as a high-throughput behavioral model for the screening of neuroactive compounds. We describe, in brief, the behavior repertoire of zebrafish larvae. Then, we focus on the utilization of light-dark locomotion test in identifying and screening of neuroactive compounds.
Collapse
Affiliation(s)
- Ram Manohar Basnet
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Daniela Zizioli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Somrat Taweedet
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Dario Finazzi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
- Clinical Chemistry Laboratory, ASST-Spedali Civili di Brescia, 25123 Brescia, Italy.
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| |
Collapse
|
41
|
Zhou CC, Gao ZY, He YQ, Wu MQ, Chen F, Wang J, Liu JX, Yan CH. Effects of lead, mercury, aluminium and manganese co-exposure on the serum BDNF concentration of pre-school children in Taizhou, China. CHEMOSPHERE 2019; 217:158-165. [PMID: 30415114 DOI: 10.1016/j.chemosphere.2018.11.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/18/2018] [Accepted: 11/02/2018] [Indexed: 06/09/2023]
Abstract
Previous studies have shown that toxic metal exposure can have adverse effects on the nervous system of children, but the toxicology of metal co-exposure on neurodevelopment remains to be clarified. Brain derived neurotrophic factor (BDNF) plays an important role in nervous system development, but the possible effects of metal co-exposure on the serum BDNF concentrations of children remain unknown. A total of 561 children living in Taizhou City, China were recruited to participate in our cross-sectional multicenter survey. We measured their blood Pb, Hg, Al and Mn levels and serum BDNF concentrations as well as determined their associations in the total and within sex subgroups. The geometric means of the blood Pb, Hg, Al and Mn levels in all the participants were 67.18 μg/L, 1.01 μg/L, 52.03 μg/L and 18.26 μg/L, respectively. The serum BDNF concentration in children was 19.45 ng/mL. After adjusting for confounders, the blood Pb levels were significantly negatively associated with the serum BDNF concentrations in all the subjects and boys but not in girls. In addition, a significantly negative interaction between blood Pb and blood Hg and a positive interaction between blood Pb and blood Al on serum BDNF concentrations were also observed in boys but not in girls. Our findings highlight the toxic effects of metal co-exposure on serum BDNF levels in pre-school children and indicate that these effects might differ by gender, which suggest that special attention should be paid to the sex-specific effects of metal exposure.
Collapse
Affiliation(s)
- Can-Can Zhou
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Zhen-Yan Gao
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China; Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, 200040, PR China
| | - Yu-Qiong He
- Department of Pharmacognosy, Second Military Medical University School of Pharmacy, Shanghai, 200433, PR China
| | - Mei-Qin Wu
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Fei Chen
- East China University of Science and Technology, Shanghai, 200237, PR China
| | - Ju Wang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Jun-Xia Liu
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Chong-Huai Yan
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China.
| |
Collapse
|
42
|
Ke T, Gonçalves FM, Gonçalves CL, Dos Santos AA, Rocha JBT, Farina M, Skalny A, Tsatsakis A, Bowman AB, Aschner M. Post-translational modifications in MeHg-induced neurotoxicity. Biochim Biophys Acta Mol Basis Dis 2018; 1865:2068-2081. [PMID: 30385410 DOI: 10.1016/j.bbadis.2018.10.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 12/29/2022]
Abstract
Mercury (Hg) exposure remains a major public health concern due to its widespread distribution in the environment. Organic mercurials, such as MeHg, have been extensively investigated especially because of their congenital effects. In this context, studies on the molecular mechanism of MeHg-induced neurotoxicity are pivotal to the understanding of its toxic effects and the development of preventive measures. Post-translational modifications (PTMs) of proteins, such as phosphorylation, ubiquitination, and acetylation are essential for the proper function of proteins and play important roles in the regulation of cellular homeostasis. The rapid and transient nature of many PTMs allows efficient signal transduction in response to stress. This review summarizes the current knowledge of PTMs in MeHg-induced neurotoxicity, including the most commonly PTMs, as well as PTMs induced by oxidative stress and PTMs of antioxidant proteins. Though PTMs represent an important molecular mechanism for maintaining cellular homeostasis and are involved in the neurotoxic effects of MeHg, we are far from understanding the complete picture on their role, and further research is warranted to increase our knowledge of PTMs in MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| | - Filipe Marques Gonçalves
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Cinara Ludvig Gonçalves
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | | | - João B T Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, 97105900 Santa Maria, RS, Brazil
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88040900 Florianópolis, SC, Brazil
| | - Anatoly Skalny
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl 150000, Russia; Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya St., 6, Moscow 105064, Russia; Orenburg State University, Pobedy Ave., 13, Orenburg 460352, Russia
| | - Aristidis Tsatsakis
- Center of Toxicology Science & Research, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, United States.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| |
Collapse
|
43
|
Sub-Nanomolar Methylmercury Exposure Promotes Premature Differentiation of Murine Embryonic Neural Precursor at the Expense of Their Proliferation. TOXICS 2018; 6:toxics6040061. [PMID: 30308979 PMCID: PMC6315723 DOI: 10.3390/toxics6040061] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/28/2018] [Accepted: 10/03/2018] [Indexed: 11/17/2022]
Abstract
Methylmercury (MeHg) is a ubiquitous environmental pollutant that is known to be neurotoxic, particularly during fetal development. However, the mechanisms responsible for MeHg-induced changes in adult neuronal function, when their exposure occurred primarily during fetal development, are not yet understood. We hypothesized that fetal MeHg exposure could affect neural precursor development leading to long-term neurotoxic effects. Primary cortical precursor cultures obtained from embryonic day 12 were exposed to 0 µM, 0.25 µM, 0.5 µM, 2.5 µM, and 5 µM MeHg for 48 or 72 h. All of the concentrations tested in the study did not affect cell viability. Intriguingly, we observed that cortical precursor exposed to 0.25 µM MeHg showed increased neuronal differentiation, while its proliferation was inhibited. Reduced neuronal differentiation, however, was observed in the higher dose groups. Our results suggest that micromolar MeHg exposure may deplete the pool of neural precursors by increasing premature neuronal differentiation, which can lead to long-term neurological effects in adulthood as opposed to the higher MeHg doses that cause more immediate toxicity during infant development.
Collapse
|
44
|
Go S, Kurita H, Matsumoto K, Hatano M, Inden M, Hozumi I. Methylmercury causes epigenetic suppression of the tyrosine hydroxylase gene in an in vitro neuronal differentiation model. Biochem Biophys Res Commun 2018; 502:435-441. [PMID: 29856999 DOI: 10.1016/j.bbrc.2018.05.162] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 05/24/2018] [Indexed: 10/14/2022]
Abstract
Methylmercury (MeHg) is the causative substance of Minamata disease, which is associated with various neurological disorders such as sensory disturbance and ataxia. It has been suggested low-level dietary intake of MeHg from MeHg-containing fish during gestation adversely affects the fetus. In our study, we investigated the toxicological effects of MeHg exposure on neuronal differentiation focusing on epigenetics. We used human fetal brain-derived immortalized cells (LUHMES cells) as a human neuronal differentiation model. Cell viability, neuronal, and catecholamine markers in LUHMES cells were assessed after exposure to MeHg (0-1000 nM) for 6 days (from day 2 to day 8 of neuronal differentiation). Cell viability on day 8 was not affected by exposure to 1 nM MeHg for 6 days. mRNA levels of AADC, DBH, TUJ1, and SYN1 also were unaffected by MeHg exposure. In contrast, levels of TH, the rate-limiting enzyme for dopamine synthesis, were significantly decreased after MeHg exposure. Acetylated histone H3, acetylated histone H3 lysine 9, and tri-methyl histone H3 lysine 9 levels at the TH gene promoter were not altered by MeHg exposure. However, tri-methylation of histone H3 lysine 27 levels, related to transcriptional repression, were significantly increased at the TH gene promotor after MeHg exposure. In summary, MeHg exposure during neuronal differentiation led to epigenetic changes that repressed TH gene expression. This study provides useful insights into the toxicological mechanisms underlying the effects of developmental MeHg exposure during neuronal differentiation.
Collapse
Affiliation(s)
- Suzuna Go
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Department Biomedical Pharmaceutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi Gifu City, Gifu, 501-1196, Japan
| | - Hisaka Kurita
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Department Biomedical Pharmaceutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi Gifu City, Gifu, 501-1196, Japan
| | - Kana Matsumoto
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Department Biomedical Pharmaceutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi Gifu City, Gifu, 501-1196, Japan
| | - Manami Hatano
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Department Biomedical Pharmaceutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi Gifu City, Gifu, 501-1196, Japan
| | - Masatoshi Inden
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Department Biomedical Pharmaceutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi Gifu City, Gifu, 501-1196, Japan
| | - Isao Hozumi
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Department Biomedical Pharmaceutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi Gifu City, Gifu, 501-1196, Japan.
| |
Collapse
|
45
|
Belém-Filho IJA, Ribera PC, Nascimento AL, Gomes ARQ, Lima RR, Crespo-Lopez ME, Monteiro MC, Fontes-Júnior EA, Lima MO, Maia CSF. Low doses of methylmercury intoxication solely or associated to ethanol binge drinking induce psychiatric-like disorders in adolescent female rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 60:184-194. [PMID: 29734102 DOI: 10.1016/j.etap.2018.04.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/24/2018] [Accepted: 04/27/2018] [Indexed: 06/08/2023]
Abstract
Methylmercury (MeHg) is an environmental contaminant that provokes damage to developing brain. Simultaneously, the consumption of ethanol among adolescents has increased. Evidence concerning the effects of MeHg low doses per se or associated with ethanol during adolescence are scarce. Thus, we investigate behavioral disorders resulted from exposure to MeHg low doses and co-intoxicated with ethanol in adolescent rats. Wistar rats received chronic exposure to low doses of MeHg (40 μg/kg/day for 5 weeks) and/or ethanol binge drinking (3 g/kg/day at 3 days per week for 5 weeks). Animals were submitted to behavioral assays to assess emotionality and cognitive function. Total mercury content was evaluated in the brain and hair. Oxidative parameters were analyzed in blood samples. MeHg at low doses or associated to ethanol binge drinking produced psychiatric-like disorders and cognitive impairment. Peripherally, MeHg altered oxidative parameters when associated to ethanol. Ethanol administration reduced brain mercury deposit. We proposed that ethanol reduces the necessity of mercury tissue levels to display psychiatric-like disorders/cognitive impairment.
Collapse
Affiliation(s)
| | - Paula Cardoso Ribera
- Laboratório de Farmacologia da Inflamação e Comportamento, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Aline Lima Nascimento
- Laboratório de Farmacologia da Inflamação e Comportamento, Universidade Federal do Pará, Belém, Pará, Brazil
| | | | - Rafael Rodrigues Lima
- Laboratório de Biologia Funcional e Estrutural, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratório de Ensaios In Vitro, Imunologia e Microbiologia, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Marta Chagas Monteiro
- Laboratório de Farmacologia Molecular, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Enéas Andrade Fontes-Júnior
- Laboratório de Farmacologia da Inflamação e Comportamento, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Marcelo Oliveira Lima
- Laboratório de Toxicologia, Seção de Meio Ambiente, Instituto Evandro Chagas, Belém, Pará, Brazil
| | | |
Collapse
|
46
|
Marwari S, Dawe GS. (R)-fluoxetine enhances cognitive flexibility and hippocampal cell proliferation in mice. J Psychopharmacol 2018; 32:441-457. [PMID: 29458297 DOI: 10.1177/0269881118754733] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fluoxetine is a clinically successful antidepressant. It is a racemic mixture of (R) and (S) stereoisomers. In preclinical studies, chronic treatment with fluoxetine (10 mg/kg) had antidepressant effects correlated with increased hippocampal cell proliferation in adult rodents. However, the contribution of the enantiomers of fluoxetine is largely unknown. We investigated the effects of treatment with (R)- and (S)-fluoxetine on cognitive behavioral paradigms and examined cell proliferation in the hippocampus of C57BL/6J female mice. In a behavioral sequencing task using the IntelliCage system in which discriminated spatial patterns of rewarded and never-rewarded corners were reversed serially, (R)-fluoxetine-treated mice showed rapid acquisition of behavioral sequencing (compared with S-fluoxetine) and cognitive flexibility in subsequent reversal stages in intra- and inter-session analysis. (R)-fluoxetine also increased cell proliferation in the hippocampus, in particular in the suprapyramidal blade of the dentate gyrus. (R)-fluoxetine had superior effects to (S)-fluoxetine in elevated plus maze, forced-swim and tail-suspension tests. These results suggest that (R)-fluoxetine, which has been reported to have a shorter half-life than (S)-fluoxetine, has superior antidepressant effects and more consistently improves spatial learning and memory. This profile offers advantages in depression treatment and may also aid management of the neurocognitive impairments associated with depression.
Collapse
Affiliation(s)
- Subhi Marwari
- 1 Department of Pharmacology, National University of Singapore, Singapore
| | - Gavin S Dawe
- 1 Department of Pharmacology, National University of Singapore, Singapore.,2 Neurobiology and Ageing Programme, Life Sciences Institute, University of Singapore, Singapore
| |
Collapse
|
47
|
Raciti M, Ceccatelli S. Epigenetic mechanisms in developmental neurotoxicity. Neurotoxicol Teratol 2018; 66:94-101. [DOI: 10.1016/j.ntt.2017.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/28/2017] [Accepted: 12/04/2017] [Indexed: 12/28/2022]
|
48
|
Spulber S, Raciti M, Dulko-Smith B, Lupu D, Rüegg J, Nam K, Ceccatelli S. Methylmercury interferes with glucocorticoid receptor: Potential role in the mediation of developmental neurotoxicity. Toxicol Appl Pharmacol 2018; 354:94-100. [PMID: 29499248 DOI: 10.1016/j.taap.2018.02.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 02/25/2018] [Accepted: 02/26/2018] [Indexed: 12/14/2022]
Abstract
Methylmercury (MeHg) is a widespread environmental contaminant with established developmental neurotoxic effects. Computational models have identified glucocorticoid receptor (GR) signaling to be a key mediator behind the birth defects induced by Hg, but the mechanisms were not elucidated. Using molecular dynamics simulations, we found that MeHg can bind to the GR protein at Cys736 (located close to the ligand binding site) and distort the conformation of the ligand binging site. To assess the functional consequences of MeHg interaction with GR, we used a human cell line expressing a luciferase reporter system (HeLa AZ-GR). We found that 100 nM MeHg does not have any significant effect on GR activity alone, but the transactivation of gene expression by GR upon Dex (a synthetic GR agonist) administration was reduced in cells pre-treated with MeHg. Similar effects were found in transgenic zebrafish larvae expressing a GR reporter system (SR4G). Next we asked whether the effects of developmental exposure to MeHg are mediated by the effects on GR. Using a mutant zebrafish line carrying a loss-of-function mutation in the GR (grS357) we could show that the effects of developmental exposure to 2.5 nM MeHg are mitigated in absence of functional GR signaling. Taken together, our data indicate that inhibition of GR signaling may have a role in the developmental neurotoxic effects of MeHg.
Collapse
Affiliation(s)
- S Spulber
- Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden.
| | - M Raciti
- Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden
| | - B Dulko-Smith
- Umeå University, Faculty of Science and Technology, Department of Chemistry, Umeå, Sweden; University of Texas at Arlington, Department of Chemistry and Biochemistry, Arlington, TX, USA
| | - D Lupu
- Swetox, Karolinska Institutet, Unit of Toxicology Science, Södertälje, Sweden; "Iuliu Hatieganu" University of Medicine and Pharmacy, Department of Toxicology, Cluj-Napoca, Romania
| | - J Rüegg
- Swetox, Karolinska Institutet, Unit of Toxicology Science, Södertälje, Sweden; Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden
| | - K Nam
- Umeå University, Faculty of Science and Technology, Department of Chemistry, Umeå, Sweden; University of Texas at Arlington, Department of Chemistry and Biochemistry, Arlington, TX, USA
| | - S Ceccatelli
- Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden
| |
Collapse
|
49
|
Kalm M, Andreasson U, Björk-Eriksson T, Zetterberg H, Pekny M, Blennow K, Pekna M, Blomgren K. C3 deficiency ameliorates the negative effects of irradiation of the young brain on hippocampal development and learning. Oncotarget 2017; 7:19382-94. [PMID: 27029069 PMCID: PMC4991390 DOI: 10.18632/oncotarget.8400] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 03/09/2016] [Indexed: 02/06/2023] Open
Abstract
Radiotherapy in the treatment of pediatric brain tumors is often associated with debilitating late-appearing adverse effects, such as intellectual impairment. Areas in the brain harboring stem cells are particularly sensitive to irradiation (IR) and loss of these cells may contribute to cognitive deficits. It has been demonstrated that IR-induced inflammation negatively affects neural progenitor differentiation. In this study, we used mice lacking the third complement component (C3−/−) to investigate the role of complement in a mouse model of IR-induced injury to the granule cell layer (GCL) of the hippocampus. C3−/− and wild type (WT) mice received a single, moderate dose of 8 Gy to the brain on postnatal day 10. The C3−/− mice displayed 55 % more microglia (Iba-1+) and a trend towards increase in proliferating cells in the GCL compared to WT mice 7 days after IR. Importantly, months after IR C3−/− mice made fewer errors than WT mice in a reversal learning test indicating better learning capacity in C3−/− mice after IR. Notably, months after IR C3−/− and WT mice had similar GCL volumes, survival of newborn cells (BrdU), microglia (Iba-1) and astrocyte (S100β) numbers in the GCL. In summary, our data show that the complement system contributes to IR-induced loss of proliferating cells and maladaptive inflammatory responses in the acute phase after IR, leading to impaired learning capacity in adulthood. Targeting the complement system is hence promising for future strategies to reduce the long-term adverse consequences of IR in the young brain.
Collapse
Affiliation(s)
- Marie Kalm
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Ulf Andreasson
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Milos Pekny
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.,Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Hunter Medical Research Institute, University of Newcastle, New South Wales, Australia
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Marcela Pekna
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.,Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Hunter Medical Research Institute, University of Newcastle, New South Wales, Australia
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
50
|
Depressive-like phenotype induced by prenatal dexamethasone in mice is reversed by desipramine. Neuropharmacology 2017; 126:242-249. [DOI: 10.1016/j.neuropharm.2017.09.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 09/08/2017] [Accepted: 09/09/2017] [Indexed: 12/16/2022]
|