1
|
Wagle SR, Kovacevic B, Sen LY, Diress M, Foster T, Ionescu CM, Lim P, Brunet A, James R, Carvalho L, Mooranian A, Al-Salami H. Revolutionizing drug delivery strategies with probucol to combat oxidative stress in retinal degeneration: A comprehensive review. Eur J Pharm Biopharm 2025; 210:114695. [PMID: 40089074 DOI: 10.1016/j.ejpb.2025.114695] [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: 12/03/2024] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
Localized oxidative stress plays a key role in the development of retinal degenerative diseases, with diabetic retinopathy (DR) being one of them, contributing significantly to this vision-threatening complication of diabetes. Increased oxidative burden leads to dysfunction across various retinal cell types, including vascular endothelial cells, neurons, glial cells and pericytes. Importantly, even after achieving normalized glycemia, the detrimental effects of oxidative stress persist. Nonetheless, growing data highlights the therapeutic potential of antioxidants in safeguarding vision. However, extensive clinical trials using traditional antioxidants have produced mixed results. Therefore, probucol, known for its ability to limit vascular oxidative stress, decrease superoxide generation, and improve endogenous antioxidant activity, is a promising candidate explored in this review. In addition to describing probucol, this review will explore novel therapeutic formulation strategies by incorporating bile acid into probucol-loaded nanoparticles to enhance drug delivery to the posterior segment of the eye for more effective management of DR. The integration of bio-nanotechnology with probucol and bile acids represents a promising avenue for developing effective therapies for DR, addressing the limitations of traditional antioxidant treatments.
Collapse
Affiliation(s)
- Susbin Raj Wagle
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
| | - Bozica Kovacevic
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
| | - Le Yang Sen
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
| | - Mengistie Diress
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia; Department of Human Physiology, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Thomas Foster
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
| | - Corina Mihaela Ionescu
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
| | - Patrick Lim
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
| | - Alicia Brunet
- Centre for Ophthalmology and Visual Science (incorporating the Lions Eye Institute), the University of Western Australia, Perth, Western Australia, Australia
| | - Rebekah James
- Centre for Ophthalmology and Visual Science (incorporating the Lions Eye Institute), the University of Western Australia, Perth, Western Australia, Australia
| | - Livia Carvalho
- Centre for Ophthalmology and Visual Science (incorporating the Lions Eye Institute), the University of Western Australia, Perth, Western Australia, Australia; Department of Optometry and Vision Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Armin Mooranian
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia; School of Pharmacy, University of Otago, Dunedin, Otago, New Zealand.
| | - Hani Al-Salami
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia; Medical School, University of Western Australia, Perth, Western Australia, Australia.
| |
Collapse
|
2
|
Jomova K, Alomar SY, Nepovimova E, Kuca K, Valko M. Heavy metals: toxicity and human health effects. Arch Toxicol 2025; 99:153-209. [PMID: 39567405 PMCID: PMC11742009 DOI: 10.1007/s00204-024-03903-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
Heavy metals are naturally occurring components of the Earth's crust and persistent environmental pollutants. Human exposure to heavy metals occurs via various pathways, including inhalation of air/dust particles, ingesting contaminated water or soil, or through the food chain. Their bioaccumulation may lead to diverse toxic effects affecting different body tissues and organ systems. The toxicity of heavy metals depends on the properties of the given metal, dose, route, duration of exposure (acute or chronic), and extent of bioaccumulation. The detrimental impacts of heavy metals on human health are largely linked to their capacity to interfere with antioxidant defense mechanisms, primarily through their interaction with intracellular glutathione (GSH) or sulfhydryl groups (R-SH) of antioxidant enzymes such as superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), glutathione reductase (GR), and other enzyme systems. Although arsenic (As) is believed to bind directly to critical thiols, alternative hydrogen peroxide production processes have also been postulated. Heavy metals are known to interfere with signaling pathways and affect a variety of cellular processes, including cell growth, proliferation, survival, metabolism, and apoptosis. For example, cadmium can affect the BLC-2 family of proteins involved in mitochondrial death via the overexpression of antiapoptotic Bcl-2 and the suppression of proapoptotic (BAX, BAK) mechanisms, thus increasing the resistance of various cells to undergo malignant transformation. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important regulator of antioxidant enzymes, the level of oxidative stress, and cellular resistance to oxidants and has been shown to act as a double-edged sword in response to arsenic-induced oxidative stress. Another mechanism of significant health threats and heavy metal (e.g., Pb) toxicity involves the substitution of essential metals (e.g., calcium (Ca), copper (Cu), and iron (Fe)) with structurally similar heavy metals (e.g., cadmium (Cd) and lead (Pb)) in the metal-binding sites of proteins. Displaced essential redox metals (copper, iron, manganese) from their natural metal-binding sites can catalyze the decomposition of hydrogen peroxide via the Fenton reaction and generate damaging ROS such as hydroxyl radicals, causing damage to lipids, proteins, and DNA. Conversely, some heavy metals, such as cadmium, can suppress the synthesis of nitric oxide radical (NO·), manifested by altered vasorelaxation and, consequently, blood pressure regulation. Pb-induced oxidative stress has been shown to be indirectly responsible for the depletion of nitric oxide due to its interaction with superoxide radical (O2·-), resulting in the formation of a potent biological oxidant, peroxynitrite (ONOO-). This review comprehensively discusses the mechanisms of heavy metal toxicity and their health effects. Aluminum (Al), cadmium (Cd), arsenic (As), mercury (Hg), lead (Pb), and chromium (Cr) and their roles in the development of gastrointestinal, pulmonary, kidney, reproductive, neurodegenerative (Alzheimer's and Parkinson's diseases), cardiovascular, and cancer (e.g. renal, lung, skin, stomach) diseases are discussed. A short account is devoted to the detoxification of heavy metals by chelation via the use of ethylenediaminetetraacetic acid (EDTA), dimercaprol (BAL), 2,3-dimercaptosuccinic acid (DMSA), 2,3-dimercapto-1-propane sulfonic acid (DMPS), and penicillamine chelators.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine The Philosopher University in Nitra, 949 74, Nitra, Slovakia
| | - Suliman Y Alomar
- Doping Research Chair, Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 708 00, Ostrava-Poruba, Czech Republic
| | - Kamil Kuca
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 708 00, Ostrava-Poruba, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia.
| |
Collapse
|
3
|
Soares MP, De Angelis CF, Taylor EW, Silva LM, Montanari BH, Azevedo VC, da Costa Souza I, Monferrán MV, Wunderlin DA, Fernandes MN, Leite CAC. Dynamics of metal/metalloid bioaccumulation and sensitivity in post-larvae shrimp (Macrobrachium rosenbergii) exposed to settleable atmospheric particulate matter from an industrial source. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177355. [PMID: 39489450 DOI: 10.1016/j.scitotenv.2024.177355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/27/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
The metallurgy industry is a potent global source of particulate matter (PM) atmospheric emissions. A portion of this PM may settle in aquatic (SePM) carrying metal/metalloid particles and metallic nanoparticles. Surprisingly, this form of contamination has not received due attention from most environmental monitoring agencies. We analyzed the effect of exposure to SePM on shrimp post-larvae, a critical stage for the viability of shrimp populations and for the trophic chain. After acclimation, shrimp were exposed to contaminants using a randomized experimental design-a 4 × 4 factorial arrangement with 2 factors: exposure time (24, 48, 72, and 96 h) and SePM concentration (0.00, 0.01, 0.10, and 1.00 g L-1). The bioaccumulation of metals, contamination rates, mortality, and ROS-related biomarkers (lipid peroxidation - LPO; DNA strand breakage DNA SB and metallothionein content - MTs;) were evaluated. After contamination, the water contained 27 different metals/metalloids. Post-larvae accumulated metals, such as Cd, Pb, Al, As, Se, Sr, Zr, Ba, La, Ce, W, and Hg. However, the rise in SePM did not result in a proportional bioaccumulation rise, indicating that effective biological barriers may work for some metals. Although the different levels of SePM changed mortality dynamics, they resulted in a similar final lethality (60-80 %). SePM caused significant damage to lipids (increased LPO), genetic material (DNA SB), and increased Mts. Such effects may reflect a particularly deleterious ecological problem as it is present at such an early stage of life. These results identified a clear environmental risk since the lower level of exposure used was 102 times lower than that measured in the habitats affected by local industry. Consequently, our results emphasize the need for clear protocols for monitoring the effects of SePM in aquatic environments.
Collapse
Affiliation(s)
- Michelly Pereira Soares
- Department of Physiological Sciences, Federal University of São Carlos, Rod Washington Luis km 235, 13565-905 São Carlos, SP, Brazil.
| | - Carolina Fernandes De Angelis
- Department of Physiological Sciences, Federal University of São Carlos, Rod Washington Luis km 235, 13565-905 São Carlos, SP, Brazil
| | - Edwin W Taylor
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Ludmila Mendes Silva
- Department of Physiological Sciences, Federal University of São Carlos, Rod Washington Luis km 235, 13565-905 São Carlos, SP, Brazil.
| | - Beatriz Helena Montanari
- Department of Physiological Sciences, Federal University of São Carlos, Rod Washington Luis km 235, 13565-905 São Carlos, SP, Brazil
| | | | - Iara da Costa Souza
- Department of Physiological Sciences, Federal University of São Carlos, Rod Washington Luis km 235, 13565-905 São Carlos, SP, Brazil.
| | - Magdalena V Monferrán
- ICYTAC, Instituto de Ciencia y Tecnología de Alimentos Córdoba, CONICET and Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cdad. Universitaria, 5000 Córdoba, Argentina; Departamento Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, CONICET, CIBICI, Ciudad Universitaria, Medina Allende Esq. Haya de La Torre S/n, 5000 Cordoba, Argentina.
| | - Daniel A Wunderlin
- ICYTAC, Instituto de Ciencia y Tecnología de Alimentos Córdoba, CONICET and Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cdad. Universitaria, 5000 Córdoba, Argentina
| | - Marisa Narciso Fernandes
- Department of Physiological Sciences, Federal University of São Carlos, Rod Washington Luis km 235, 13565-905 São Carlos, SP, Brazil.
| | - Cléo Alcantara Costa Leite
- Department of Physiological Sciences, Federal University of São Carlos, Rod Washington Luis km 235, 13565-905 São Carlos, SP, Brazil.
| |
Collapse
|
4
|
Kang B, Wang J, Guo S, Yang L. Mercury-induced toxicity: Mechanisms, molecular pathways, and gene regulation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 943:173577. [PMID: 38852866 DOI: 10.1016/j.scitotenv.2024.173577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/01/2024] [Accepted: 05/25/2024] [Indexed: 06/11/2024]
Abstract
Mercury is a well-known neurotoxicant for humans and wildlife. The epidemic of mercury poisoning in Japan has clearly demonstrated that chronic exposure to methylmercury (MeHg) results in serious neurological damage to the cerebral and cerebellar cortex, leading to the dysfunction of the central nervous system (CNS), especially in infants exposed to MeHg in utero. The occurrences of poisoning have caused a wide public concern regarding the health risk emanating from MeHg exposure; particularly those eating large amounts of fish may experience the low-level and long-term exposure. There is growing evidence that MeHg at environmentally relevant concentrations can affect the health of biota in the ecosystem. Although extensive in vivo and in vitro studies have demonstrated that the disruption of redox homeostasis and microtube assembly is mainly responsible for mercurial toxicity leading to adverse health outcomes, it is still unclear whether we could quantitively determine the occurrence of interaction between mercurial and thiols and/or selenols groups of proteins linked directly to outcomes, especially at very low levels of exposure. Furthermore, intracellular calcium homeostasis, cytoskeleton, mitochondrial function, oxidative stress, neurotransmitter release, and DNA methylation may be the targets of mercury compounds; however, the primary targets associated with the adverse outcomes remain to be elucidated. Considering these knowledge gaps, in this article, we conducted a comprehensive review of mercurial toxicity, focusing mainly on the mechanism, and genes/proteins expression. We speculated that comprehensive analyses of transcriptomics, proteomics, and metabolomics could enhance interpretation of "omics" profiles, which may reveal specific biomarkers obviously correlated with specific pathways that mediate selective neurotoxicity.
Collapse
Affiliation(s)
- Bolun Kang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012 Beijing, China
| | - Jinghan Wang
- 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
| | - Lixin Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012 Beijing, China.
| |
Collapse
|
5
|
Morán-Serradilla C, Plano D, Sanmartín C, Sharma AK. Selenization of Small Molecule Drugs: A New Player on the Board. J Med Chem 2024; 67:7759-7787. [PMID: 38716896 DOI: 10.1021/acs.jmedchem.3c02426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
There is an urgent need to develop safer and more effective modalities for the treatment of a wide range of pathologies due to the increasing rates of drug resistance, undesired side effects, poor clinical outcomes, etc. Throughout the years, selenium (Se) has attracted a great deal of attention due to its important role in human health. Besides, a growing body of work has unveiled that the inclusion of Se motifs into a great number of molecules is a promising strategy for obtaining novel therapeutic agents. In the current Perspective, we have gathered the most recent literature related to the incorporation of different Se moieties into the scaffolds of a wide range of known drugs and their feasible pharmaceutical applications. In addition, we highlight different representative examples as well as provide our perspective on Se drugs and the possible future directions, promises, opportunities, and challenges of this ground-breaking area of research.
Collapse
Affiliation(s)
| | - Daniel Plano
- Department of Pharmaceutical Sciences, University of Navarra, Irunlarrea 1, Pamplona E-31008, Spain
| | - Carmen Sanmartín
- Department of Pharmaceutical Sciences, University of Navarra, Irunlarrea 1, Pamplona E-31008, Spain
| | - Arun K Sharma
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, United States
- Penn State Cancer Institute, 400 University Drive,Hershey, Pennsylvania 17033, United States
| |
Collapse
|
6
|
Del Rio Naiz SC, Varela KG, de Carvalho D, Remor AP. Probucol neuroprotection against manganese-induced damage in adult Wistar rat brain slices. Toxicol Ind Health 2023; 39:638-650. [PMID: 37705340 DOI: 10.1177/07482337231201565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Manganese (Mn) is an abundant element used for commercial purposes and is essential for the proper function of biological systems. Chronic exposure to high Mn concentrations causes Manganism, a Parkinson's-like neurological disorder. The pathophysiological mechanism of Manganism remains unknown; however, it involves mitochondrial dysfunction and oxidative stress. This study assessed the neuroprotective effect of probucol, a hypolipidemic agent with anti-inflammatory and antioxidant properties, on cell viability and oxidative stress in slices of the cerebral cortex and striatum from adult male Wistar rats. Brain structure slices were kept separately and incubated with manganese chloride (MnCl2) and probucol to evaluate the cell viability and oxidative parameters. Probucol prevented Mn toxicity in the cerebral cortex and striatum, as evidenced by the preservation of cell viability observed with probucol (10 and 30 μM) pre-treatment, as well as the prevention of mitochondrial complex I inhibition in the striatum (30 μM). These findings support the protective antioxidant action of probucol, attributed to its ability to prevent cell death and mitigate Mn-induced mitochondrial dysfunction.
Collapse
Affiliation(s)
| | - Karina Giacomini Varela
- Programa de Pós-Graduação em Biociências e Saúde (PPGBS), Área de Ciências da Vida e Saúde, Universidade do Oeste de Santa Catarina (UNOESC), Joaçaba, Brazil
| | - Diego de Carvalho
- Programa de Pós-Graduação em Biociências e Saúde (PPGBS), Área de Ciências da Vida e Saúde, Universidade do Oeste de Santa Catarina (UNOESC), Joaçaba, Brazil
| | - Aline Pertile Remor
- Programa de Pós-Graduação em Biociências e Saúde (PPGBS), Área de Ciências da Vida e Saúde, Universidade do Oeste de Santa Catarina (UNOESC), Joaçaba, Brazil
| |
Collapse
|
7
|
Gamit N, Patil M, B Sundrappa S, Sundaram SM, Sethi G, Dharmarajan A, Warrier S. Mesenchymal stem cell-derived rapid drug screening system for Alzheimer's disease for the identification of novel drugs. Drug Dev Res 2023; 84:1496-1512. [PMID: 37571798 DOI: 10.1002/ddr.22102] [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: 02/14/2023] [Revised: 06/11/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023]
Abstract
A reliable and efficient in vitro model is needed to screen drugs for Alzheimer's disease (AD), as many drugs are currently in the developmental stage. To address this, we developed an in vitro model using amniotic membrane-derived mesenchymal stem cells (AM-MSCs) to screen novel drugs for AD. We differentiated AM-MSCs into neurons and degenerated them using beta amyloid1-42 (Aß). We then tested AD drugs, which are commercially available such as donepezil, rivastigmine, memantine, citicoline, and two novel drugs, that is, probucol, an anti-hyperlipidaemic drug, and NMJ-2, a cinnamic acid analogue for their potential to protect the cells against neurodegeneration. We used gene expression and immunofluorescence staining to assess the neuroprotective ability of these drugs. We also measured the ability of these drugs to reduce lactate dehydrogenase, reactive oxygen species, and nitric oxide levels, as well as their ability to stabilize the mitochondrial membrane potential and increase acetylcholine (ACh) levels. The AD drugs and novel drugs reduced cytotoxicity and oxidative stress, stabilized mitochondrial membrane potential, and restored ACh levels. Furthermore, they reduced BACE1 expression, with a concomitant increase in the expression of cholinergic markers. This AM-MSCs-based AD-like model has immense potential to be an accurate human model and an alternative to animal models for testing a large number of lead compounds in a short time. Our results also suggest that the novel drugs probucol and NMJ-2 may protect against Aß-induced neurodegeneration.
Collapse
Affiliation(s)
- Naisarg Gamit
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, India
| | - Manasi Patil
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, India
| | - Soumya B Sundrappa
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, India
| | - S Mohana Sundaram
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, India
- Cuor Stem Cellutions Pvt Ltd, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, India
- Department of Biotechnology, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| |
Collapse
|
8
|
Ye Y, Chen Y, Wu H, Fu Y, Sun Y, Wang X, Li P, Wu Z, Wang J, Yang Z, Zhou E. Investigations into ferroptosis in methylmercury-induced acute kidney injury in mice. ENVIRONMENTAL TOXICOLOGY 2023; 38:1372-1383. [PMID: 36880449 DOI: 10.1002/tox.23770] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/10/2023] [Accepted: 02/20/2023] [Indexed: 05/18/2023]
Abstract
Methylmercury (MeHg) is a highly poisonous form of mercury and a risk factor for kidney impairment in humans that currently has no effective means of therapy. Ferroptosis is a non-apoptotic metabolic cell death linked to numerous diseases. It is currently unknown whether ferroptosis takes part in MeHg-induced kidney damage. Here, we established a model of acute kidney injury (AKI) in mice by gavage with different doses of MeHg (0, 40, 80, 160 μmol/kg). Serological analysis revealed elevated levels of UA, UREA, and CREA; H&E staining showed variable degrees of renal tubule injury; qRT-PCR detection displayed increased expression of KIM-1 and NGAL in the groups with MeHg treatment, indicated that MeHg successfully induced AKI. Furthermore, MDA levels enhanced in renal tissues of mice with MeHg exposure whereas GSH levels decreased; ACSL4 and PTGS2 nucleic acid levels elevated while SLC7A11 levels reduced; transmission electron microscopy illustrated that the density of the mitochondrial membrane thickened and the ridge reduced considerably; protein levels for 4HNE and TfR1 improved since GPX4 levels declined, all these results implying the involvement of ferroptosis as a result of MeHg exposure. Additionally, the observed elevation in the protein levels of NLRP3, p-p65, p-p38, p-ERK1/2, and KEAP1 in tandem with downregulated Nrf2 expression levels indicate the involvement of the NF-κB/NLRP3/MAPK/Nrf2 pathways. All the above findings suggested that ferroptosis and the NF-κB/NLRP3/MAPK/Nrf2 pathways are implicated in MeHg-induced AKI, thereby providing a theoretical foundation and reference for future investigations into the prevention and treatment of MeHg-induced kidney injury.
Collapse
Affiliation(s)
- Yingrong Ye
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Yichun Chen
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Hanpeng Wu
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Yiwu Fu
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Youpeng Sun
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Xia Wang
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Peixuan Li
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Zhikai Wu
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Jingjing Wang
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Zhengtao Yang
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Ershun Zhou
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| |
Collapse
|
9
|
Ishida K, Takeda K, Takehara Y, Takabayashi T, Miyara M, Sanoh S, Kawai H, Ohta S, Kotake Y. Methylmercury Decreases AMPA Receptor Subunit GluA2 Levels in Cultured Rat Cortical Neurons. Biol Pharm Bull 2023; 46:292-300. [PMID: 36724957 DOI: 10.1248/bpb.b22-00744] [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: 02/03/2023]
Abstract
Methylmercury (MeHg) is a well-known environmental pollutant that has harmful effects on the central nervous systems of humans and animals. The molecular mechanisms of MeHg-induced neurotoxicity at low concentrations are not fully understood. Here, we investigated the effects of low-concentration MeHg on the cell viability, Ca2+ homeostasis, and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA2 levels, which determine Ca2+ permeability of AMPA receptors, in rat primary cortical neurons. Exposure of cortical neurons to 100 and 300 nM MeHg for 7 d resulted in a decrease in GluA2 levels, an increase in basal intracellular Ca2+ concentration, increased phosphorylation levels of extracellular signal-regulated kinase (ERK)1/2 and p38, and decreased cell viability. Moreover, glutamate stimulation exacerbated the decrease in cell viability and increased intracellular Ca2+ levels in MeHg-treated neurons compared to control neurons. MeHg-induced neuronal cell death was ameliorated by 1-naphthyl acetyl spermine, a specific antagonist of Ca2+-permeable, GluA2-lacking AMPA receptors. Our findings raise the possibility that decreased neuronal GluA2 levels and the subsequent increase in intracellular Ca2+ concentration may contribute to MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Keishi Ishida
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Kazuki Takeda
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Yuki Takehara
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | | | - Masatsugu Miyara
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Seigo Sanoh
- Graduate School of Biomedical and Health Sciences, Hiroshima University.,Wakayama Medical University
| | - Hidehiko Kawai
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Shigeru Ohta
- Graduate School of Biomedical and Health Sciences, Hiroshima University.,Wakayama Medical University
| | - Yaichiro Kotake
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| |
Collapse
|
10
|
Bjørklund G, Antonyak H, Polishchuk A, Semenova Y, Lesiv M, Lysiuk R, Peana M. Effect of methylmercury on fetal neurobehavioral development: an overview of the possible mechanisms of toxicity and the neuroprotective effect of phytochemicals. Arch Toxicol 2022; 96:3175-3199. [PMID: 36063174 DOI: 10.1007/s00204-022-03366-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 08/17/2022] [Indexed: 11/25/2022]
Abstract
Methylmercury (MeHg) is a global environmental pollutant with neurotoxic effects. Exposure to MeHg via consumption of seafood and fish can severely impact fetal neurobehavioral development even when MeHg levels in maternal blood are as low as about 5 μg/L, which the mother tolerates well. Persistent motor dysfunctions and cognitive deficits may result from trans-placental exposure. The present review summarizes current knowledge on the mechanisms of MeHg toxicity during the period of nervous system development. Although cerebellar Purkinje cells are MeHg targets, the actions of MeHg on thiol components in the neuronal cytoskeleton as well as on mitochondrial enzymes and induction of disturbances of glutamate signaling can impair extra-cerebellar functions, also at levels well tolerated by adult individuals. Numerous herbal substances possess neuroprotective effects, predominantly represented by natural polyphenolic molecules that might be utilized to develop natural drugs to alleviate neurotoxicity symptoms caused by MeHg or other Hg compounds.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Toften 24, 8610, Mo i Rana, Norway.
| | | | | | | | - Marta Lesiv
- Ivan Franko National University of Lviv, Lviv, Ukraine
| | - Roman Lysiuk
- Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Massimiliano Peana
- Department of Chemical, Physics, Mathematics and Natural Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
11
|
Kumar Rai R, Shankar Pati R, Islam A, Roy G. Detoxification of organomercurials by thiones and selones: A short review. Inorganica Chim Acta 2022. [DOI: 10.1016/j.ica.2022.120980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
12
|
Ferroptosis as a mechanism of non-ferrous metal toxicity. Arch Toxicol 2022; 96:2391-2417. [PMID: 35727353 DOI: 10.1007/s00204-022-03317-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/11/2022] [Indexed: 11/02/2022]
Abstract
Ferroptosis is a recently discovered form of regulated cell death, implicated in multiple pathologies. Given that the toxicity elicited by some metals is linked to alterations in iron metabolism and induction of oxidative stress and lipid peroxidation, ferroptosis might be involved in such toxicity. Although direct evidence is insufficient, certain pioneering studies have demonstrated a crosstalk between metal toxicity and ferroptosis. Specifically, the mechanisms underlying metal-induced ferroptosis include induction of ferritinophagy, increased DMT-1 and TfR cellular iron uptake, mitochondrial dysfunction and mitochondrial reactive oxygen species (mitoROS) generation, inhibition of Xc-system and glutathione peroxidase 4 (GPX4) activity, altogether resulting in oxidative stress and lipid peroxidation. In addition, there is direct evidence of the role of ferroptosis in the toxicity of arsenic, cadmium, zinc, manganese, copper, and aluminum exposure. In contrast, findings on the impact of cobalt and nickel on ferroptosis are scant and nearly lacking altogether for mercury and especially lead. Other gaps in the field include limited studies on the role of metal speciation in ferroptosis and the critical cellular targets. Although further detailed studies are required, it seems reasonable to propose even at this early stage that ferroptosis may play a significant role in metal toxicity, and its modulation may be considered as a potential therapeutic tool for the amelioration of metal toxicity.
Collapse
|
13
|
Dong L, Yang B, Zhang Y, Wang S, Li F, Xing G, Farina M, Zhang Y, Appiah-Kubi K, Tinkov AA, Aschner M, Shi H, Liu T, Lu R. Ferroptosis contributes to methylmercury-induced cytotoxicity in rat primary astrocytes and Buffalo rat liver cells. Neurotoxicology 2022; 90:228-236. [DOI: 10.1016/j.neuro.2022.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/27/2022] [Accepted: 04/14/2022] [Indexed: 01/18/2023]
|
14
|
A Novel Diselenide-Probucol-Analogue Protects Against Methylmercury-Induced Toxicity in HT22 Cells by Upregulating Peroxide Detoxification Systems: a Comparison with Diphenyl Diselenide. Neurotox Res 2022; 40:127-139. [PMID: 35043379 DOI: 10.1007/s12640-021-00466-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/24/2021] [Accepted: 12/20/2021] [Indexed: 10/19/2022]
Abstract
Methylmercury (MeHg) is a ubiquitous environmental neurotoxicant whose mechanisms of action involve oxidation of endogenous nucleophilic groups (mainly thiols and selenols), depletion of antioxidant defenses, and disruption of neurotransmitter homeostasis. Diphenyl diselenide-(PhSe)2-a model diaryl diselenide, has been reported to display significant protective effects against MeHg-induced neurotoxicity under both in vitro and in vivo experimental conditions. In this study, we compared the protective effects of (PhSe)2 with those of RC513 (4,4'-diselanediylbis(2,6-di-tert-butylphenol), a novel diselenide-probucol-analog) against MeHg-induced toxicity in the neuronal (hippocampal) cell line HT22. Although both (PhSe)2 and RC513 significantly mitigated MeHg- and tert-butylhydroperoxide (t-BuOOH)-cytotoxicity, the probucol analog exhibited superior protective effects, which were observed earlier and at lower concentrations compared to (PhSe)2. RC513 treatment (at either 0.5 µM or 2 µM) significantly increased glutathione peroxidase (GPx) activity, which has been reported to counteract MeHg-toxicity. (PhSe)2 was also able to increase GPx activity, but only at 2 µM. Although both compounds increased the Gpx1 transcripts at 6 h after treatments, only RC513 was able to increase mRNA levels of Prx2, Prx3, Prx5, and Txn2, which are also involved in peroxide detoxification. RC513 (at 2 µM) significantly increased GPx-1 protein expression in HT22 cells, although (PhSe)2 displayed a minor (nonsignificant) effect in this parameter. In agreement, RC513 induced a faster and superior capability to cope with exogenously-added peroxide (t-BuOOH). In summary, when compared to the prototypical organic diaryl diselenide [(PhSe)2], RC513 displayed superior protective properties against MeHg-toxicity in vitro; this was paralleled by a more pronounced upregulation of defenses related to detoxification of peroxides, which are well-known MeHg-derived intermediate oxidant species.
Collapse
|
15
|
da Silva EB, Eichwald T, Glaser V, Varela KG, Baptistella AR, de Carvalho D, Remor AP. Protective Effects of Probucol on Different Brain Cells Exposed to Manganese. Neurotox Res 2022; 40:276-285. [PMID: 35043377 DOI: 10.1007/s12640-021-00458-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/11/2021] [Accepted: 12/03/2021] [Indexed: 10/19/2022]
Abstract
Manganese (Mn) is an essential metal for many functions in the body. However, in excess, it can be neurotoxic and cause a Parkinson-like syndrome, known as manganism. Here, we aimed to identify a protective effect of probucol, a lipid-lowering agent with anti-inflammatory and antioxidant properties, against Mn-induced toxicity in human neuroblastoma (SH-SY5Y) and glioblastoma (C6) cell lines. The cells were incubated with increasing concentrations of Mn followed by probucol addition 1, 3, 6, and/or 24 h to assess the metal toxic doses and measure the protective effect of probucol against Mn-induced oxidative damage. Longer exposition to Mn showed decreased SH-SY5Y cellular viability in concentrations higher than 100 µM, and probucol was able to prevent this effect. The C6 cells were more sensitive to the Mn deleterious actions, decreasing the cell viability after 6 h of 500 µM Mn exposure. In addition, probucol prevents the complex I and II of the mitochondrial respiratory chain (MRC) inhibition caused by Mn and decreased the intracellular ROS production. Taken together, our results showed that Mn toxicity affects differently both cell lines and probucol has a protective effect against the oxidative imbalance in the central nervous system.
Collapse
Affiliation(s)
- Erica Blenda da Silva
- Graduada em Medicina, Área de Ciências da Vida e Saúde, Universidade do Oeste de Santa Catarina (UNOESC), Campus de Joaçaba, SC, 89600-000, Joaçaba, Brazil
| | - Tuany Eichwald
- Programa de Pós-Graduação em Biociências e Saúde (PPGBS), Área de Ciências da Vida e Saúde, Universidade do Oeste de Santa Catarina (UNOESC), Campus de Joaçaba, SC, 89600-000, Joaçaba, Brazil
| | - Viviane Glaser
- Centro de Ciências Rurais, Coordenadoria Especial de Ciências Biológicas e Agronômicas, Universidade Federal de Santa Catarina (UFSC), Campus de Curitibanos, SC, Curitibanos, Brazil
| | - Karina Giacomini Varela
- Graduada em Ciências Biológicas, Universidade do Oeste de Santa Catarina (UNOESC), Campus de Joaçaba, Biotério, 89600-000, Joaçaba, Brazil
| | - Antuani Rafael Baptistella
- Programa de Pós-Graduação em Biociências e Saúde (PPGBS), Área de Ciências da Vida e Saúde, Universidade do Oeste de Santa Catarina (UNOESC), Campus de Joaçaba, SC, 89600-000, Joaçaba, Brazil
| | - Diego de Carvalho
- Programa de Pós-Graduação em Biociências e Saúde (PPGBS), Área de Ciências da Vida e Saúde, Universidade do Oeste de Santa Catarina (UNOESC), Campus de Joaçaba, SC, 89600-000, Joaçaba, Brazil
| | - Aline Pertile Remor
- Programa de Pós-Graduação em Biociências e Saúde (PPGBS), Área de Ciências da Vida e Saúde, Universidade do Oeste de Santa Catarina (UNOESC), Campus de Joaçaba, SC, 89600-000, Joaçaba, Brazil.
| |
Collapse
|
16
|
Naime AA, Barbosa FVAR, Bueno DC, Curi Pedrosa R, Canto RFS, Colle D, Braga AL, Farina M. Prevention of ferroptosis in acute scenarios: an in vitro study with classic and novel anti-ferroptotic compounds. Free Radic Res 2021; 55:1062-1079. [PMID: 34895012 DOI: 10.1080/10715762.2021.2017912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Ferroptosis, an iron-dependent form of cell death, has critical roles in diverse pathologies. Data on the temporal events mediating the prevention of ferroptosis are lacking. Focused on temporal aspects of cytotoxicity/protection, we investigated the effects of classic (Fer-1) and novel [2,6-di-tert-butyl-4-(2-thienylthio)phenol (C1) and 2,6-di-tert-butyl-4-(2-thienylselano)phenol (C2)] anti-ferroptotic agents against RSL3-, BSO- or glutamate-induced ferroptosis in cultured HT22 neuronal cell line, comparing their effects with those of the antioxidants trolox, ebselen and probucol. Glutamate (5 mM), BSO (25 μM) and RSL3 (50 nM) decreased approximately 40% of cell viability at 24 h. At these concentrations, none of these agents changed cell viability at 6 h after treatments; RSL3 increased lipoperoxidation from 6 h, although BSO and glutamate only did so at 12 h after treatments. At similar conditions, BSO and glutamate (but not RSL3) decreased GSH levels at 6 h after treatments. Fer-1, C1 and C2 exhibited similar protective effects against glutamate-, BSO- and RSL3-cytotoxicity, but this protection was limited when the protective agents were delivered to cells at time-points characterized by increased lipoperoxidation (but not glutathione depletion). Compared to Fer-1, C1 and C2, the anti-ferroptotic effects of trolox, ebselen and probucol were minor. Cytoprotective effects were not associated with direct antioxidant efficacies. These results indicate that the temporal window is central in affecting the efficacies of anti-ferroptotic drugs in acute scenarios; ferroptosis prevention is improbable when significant rates of lipoperoxidation were already achieved. C1 and C2 displayed remarkable cytoprotective effects, representing a promising new class of compounds to treat ferroptosis-related pathologies.
Collapse
Affiliation(s)
- Aline Aita Naime
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | | | - Diones Caeran Bueno
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Rozangela Curi Pedrosa
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Rômulo Faria Santos Canto
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
| | - Dirleise Colle
- Department of Clinical Analyses, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Antônio Luiz Braga
- Department of Chemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| |
Collapse
|
17
|
Xie Y, Song A, Zhu Y, Jiang A, Peng W, Zhang C, Meng X. Effects and mechanisms of probucol on aging-related hippocampus-dependent cognitive impairment. Biomed Pharmacother 2021; 144:112266. [PMID: 34634555 DOI: 10.1016/j.biopha.2021.112266] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND In the present study, we aimed to investigate the effects of probucol on aging-related hippocampus-dependent cognitive impairment and explore the potential mechanisms. METHODS D-galactose (100 mg/kg, once daily for 6 weeks) was subcutaneously injected to induce aging in mice. Then the mice were administered with probucol or vehicle once a day for 2 weeks. The hippocampus-related cognition was evaluated with Morris water maze test, novel object recognition test, and contextual fear conditioning test. Moreover, synaptic plasticity was assessed, and RNA-sequencing was applied to further explore the molecular mechanisms. RESULTS Aging mice induced by D-galactose showed conspicuous learning and memory impairment, which was significantly ameliorated by probucol. Meanwhile, probucol enhanced the spine density and dendritic branches, improved long-term potentiation, and increased the expression of PSD95 of aging mice. Probucol regulated 70 differentially expressed genes compared to D-galactose group, of which 38 genes were upregulated and 32 genes were downregulated. At last, RNA-sequencing results were verified by quantitative reverse transcription-polymerase chain reaction. CONCLUSIONS Probucol improved learning and memory in aging mice through enhancing synaptic plasticity and regulating gene expression, indicating the potential application of probucol to prevent and treat aging-related disorders.
Collapse
Affiliation(s)
- Yaru Xie
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Anni Song
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuting Zhu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Anni Jiang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenpeng Peng
- Department of cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xianfang Meng
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
18
|
Xie J, Tao L, Wu Q, Li T, Yang C, Lin T, Liu B, Li G, Chen D. Mercury and selenium in squids from the Pacific Ocean and Indian Ocean: The distribution and human health implications. MARINE POLLUTION BULLETIN 2021; 173:112926. [PMID: 34536705 DOI: 10.1016/j.marpolbul.2021.112926] [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: 08/01/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 06/13/2023]
Abstract
Squids are globally distributed. Hg-contaminated squids may have high risks on humans. With abundant Se (antagonistic effect on Hg), the risks can be reduced. We collected squids around the world (Northwest Pacific Ocean, Southeast Pacific Ocean and Indian Ocean). Concentrations of Hg and Se were region-based and tissue-based. The higher content of Se were, the lower relative Hg levels were. The correlation between Se:Hg and Se was the strongest in the digestive gland. The values of Se:Hg and THQ all confirm that the health risk was lower in samples with higher concentrations of Se. Despite the risk assessment by Se:Hg, BRV and THQ analysis showed no risk when consumed in moderation, the maximum daily intake is provided based on Monte Carlo simulation. In future, when evaluating the risks cause by Hg exposure and providing the recommended daily amount, it may need to concurrent consideration of Se levels.
Collapse
Affiliation(s)
- Jingqian Xie
- College of Marine Ecology and Environment, Shanghai Ocean University, Shanghai 201306, China; Skate Key Laboratory of Organic Geochemistry, Guangdong Key Laboratory of Environment and Resources, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China
| | - Ling Tao
- College of Marine Ecology and Environment, Shanghai Ocean University, Shanghai 201306, China
| | - Qiang Wu
- College of Marine Ecology and Environment, Shanghai Ocean University, Shanghai 201306, China
| | - Tiejun Li
- Zhejiang Marine Fisheries Research Institute, Key Laboratory of Sustainable Utilization of Technology Research for Fishery Resource of Zhejiang Province, Zhoushan, 316021, China
| | - Chenghu Yang
- Zhejiang Marine Fisheries Research Institute, Key Laboratory of Sustainable Utilization of Technology Research for Fishery Resource of Zhejiang Province, Zhoushan, 316021, China
| | - Tian Lin
- College of Marine Ecology and Environment, Shanghai Ocean University, Shanghai 201306, China
| | - Bilin Liu
- College of Marine Science, Shanghai Ocean University, Shanghai 201306, China; The Key Laboratory of Sustainable Exploitation of Oceanic Fisheries Resources, Ministry of Education, Shanghai 201306, China
| | - Gang Li
- College of Marine Science, Shanghai Ocean University, Shanghai 201306, China; The Key Laboratory of Sustainable Exploitation of Oceanic Fisheries Resources, Ministry of Education, Shanghai 201306, China.
| | - Duofu Chen
- College of Marine Science, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
19
|
Wei Y, Ni L, Pan J, Li X, Xu B, Deng Y, Yang T, Liu W. The Roles of Oxidative Stress in Regulating Autophagy in Methylmercury-induced Neurotoxicity. Neuroscience 2021; 469:175-190. [PMID: 34174372 DOI: 10.1016/j.neuroscience.2021.06.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 12/24/2022]
Abstract
Methylmercury (MeHg) is a potential neurotoxin that is highly toxic to the human central nervous system. Although MeHg neurotoxicity has been widely studied, the mechanism of MeHg neurotoxicity has not yet been fully elucidated. Some research evidence suggests that oxidative stress and autophagy are important molecular mechanisms of MeHg-induced neurotoxicity. Researchers have widely accepted that oxidative stress regulates the autophagy pathway. The current study reviews the activation of Nuclear factor-erythroid-2-related factor (Nrf2)-related oxidative stress pathways and autophagy signaling pathways in the case of MeHg neurotoxicity. In addition, autophagy mainly plays a role in the neurotoxicity of MeHg through mTOR-dependent and mTOR-independent autophagy signaling pathways. Finally, the regulation of autophagy by reactive oxygen species (ROS) and Nrf2 in MeHg neurotoxicity was explored in this review, providing a new concept for the study of the neurotoxicity mechanism of MeHg.
Collapse
Affiliation(s)
- Yanfeng Wei
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Linlin Ni
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Jingjing Pan
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Xiaoyang Li
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Tianyao Yang
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China.
| |
Collapse
|
20
|
Environmentally relevant developmental methylmercury exposures alter neuronal differentiation in a human-induced pluripotent stem cell model. Food Chem Toxicol 2021; 152:112178. [PMID: 33831500 DOI: 10.1016/j.fct.2021.112178] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/15/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022]
Abstract
Developmental methylmercury (MeHg) exposure selectively targets the cerebral and cerebellar cortices, as seen by disruption of cytoarchitecture and glutamatergic (GLUergic) neuron hypoplasia. To begin to understand the mechanisms of this loss of GLUergic neurons, we aimed to develop a model of developmental MeHg neurotoxicity in human-induced pluripotent stem cells differentiating into cortical GLUergic neurons. Three dosing paradigms at 0.1 μM and 1.0 μM MeHg, which span different stages of neurodevelopment and reflect toxicologically relevant accumulation levels seen in human studies and mammalian models, were established. With these exposure paradigms, no changes were seen in commonly studied endpoints of MeHg toxicity, including viability, proliferation, and glutathione levels. However, MeHg exposure induced changes in mitochondrial respiration and glycolysis and in markers of neuronal differentiation. Our novel data suggests that GLUergic neuron hypoplasia seen with MeHg toxicity may be due to the partial inhibition of neuronal differentiation, given the increased expression of the early dorsal forebrain marker FOXG1 and corresponding decrease in expression on neuronal markers MAP2 and DCX and the deep layer cortical neuronal marker TBR1. Future studies should examine the persistent and latent functional effects of this MeHg-induced disruption of neuronal differentiation as well as transcriptomic and metabolomic alterations that may mediate MeHg toxicity.
Collapse
|
21
|
Methylmercury, oxidative stress, and neurodegeneration. Toxicology 2021. [DOI: 10.1016/b978-0-12-819092-0.00015-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
22
|
Zieniewska I, Maciejczyk M, Zalewska A. The Effect of Selected Dental Materials Used in Conservative Dentistry, Endodontics, Surgery, and Orthodontics as Well as during the Periodontal Treatment on the Redox Balance in the Oral Cavity. Int J Mol Sci 2020; 21:ijms21249684. [PMID: 33353105 PMCID: PMC7767252 DOI: 10.3390/ijms21249684] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/31/2022] Open
Abstract
Oxidative stress (OS) is a redox homeostasis disorder that results in oxidation of cell components and thus disturbs cell metabolism. OS is induced by numerous internal as well as external factors. According to recent studies, dental treatment may also be one of them. The aim of our work was to assess the effect of dental treatment on the redox balance of the oral cavity. We reviewed literature available in PubMed, Medline, and Scopus databases, including the results from 2010 to 2020. Publications were searched according to the keywords: oxidative stress and dental monomers; oxidative stress and amalgam; oxidative stress and periodontitis, oxidative stress and braces, oxidative stress and titanium; oxidative stress and dental implants, oxidative stress and endodontics treatment, oxidative stress and dental treatment; and oxidative stress and dental composite. It was found that dental treatment with the use of composites, amalgams, glass-ionomers, materials for root canal filling/rinsing, orthodontic braces (made of various metal alloys), titanium implants, or whitening agents can disturb oral redox homeostasis by affecting the antioxidant barrier and increasing oxidative damage to salivary proteins, lipids, and DNA. Abnormal saliva secretion/composition was also observed in dental patients in the course of OS. It is suggested that the addition of antioxidants to dental materials or antioxidant therapy applied during dental treatment could protect the patient against harmful effects of OS in the oral cavity.
Collapse
Affiliation(s)
- Izabela Zieniewska
- Doctoral Studies, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-274 Bialystok, Poland
- Correspondence: (I.Z.); (A.Z.)
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, 15-022 Bialystok, Poland;
| | - Anna Zalewska
- Experimental Dentistry Laboratory, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-274 Bialystok, Poland
- Correspondence: (I.Z.); (A.Z.)
| |
Collapse
|
23
|
Fujimura M, Usuki F. Methylmercury-Mediated Oxidative Stress and Activation of the Cellular Protective System. Antioxidants (Basel) 2020; 9:antiox9101004. [PMID: 33081221 PMCID: PMC7602710 DOI: 10.3390/antiox9101004] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 12/17/2022] Open
Abstract
Methylmercury (MeHg) is a well-known neurotoxicant that causes severe intoxication in humans. In Japan, it is referred to as Minamata disease, which involves two characteristic clinical forms: fetal type and adult type depending on the exposed age. In addition to MeHg burden level, individual susceptibility to MeHg plays a role in the manifestation of MeHg toxicity. Research progress has pointed out the importance of oxidative stress in the pathogenesis of MeHg toxicity. MeHg has a high affinity for selenohydryl groups, sulfhydryl groups, and selenides. It has been clarified that such affinity characteristics cause the impairment of antioxidant enzymes and proteins, resulting in the disruption of antioxidant systems. Furthermore, MeHg-induced intracellular selenium deficiency due to the greater affinity of MeHg for selenohydryl groups and selenides leads to failure in the recoding of a UGA codon for selenocysteine and results in the degradation of antioxidant selenoenzyme mRNA by nonsense-mediated mRNA decay. The defect of antioxidant selenoenzyme replenishment exacerbates MeHg-mediated oxidative stress. On the other hand, it has also been revealed that MeHg can directly activate the antioxidant Keap1/Nrf2 signaling pathway. This review summarizes the incidence of MeHg-mediated oxidative stress from the viewpoint of the individual intracellular redox system interactions and the MeHg-mediated aforementioned intracellular events. In addition, the mechanisms of cellular stress pathways and neuronal cell death triggered by MeHg-mediated oxidative stress and direct interactions of MeHg with reactive residues of proteins are mentioned.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease, Kumamoto 867-0008, Japan;
| | - Fusako Usuki
- Division of Neuroimmunology, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima 890-8544, Japan
- Correspondence: ; Tel.: +81-99-275-6246; Fax: +81-99-275-5942
| |
Collapse
|
24
|
Fujimura M, Usuki F, Unoki T. Decreased plasma thiol antioxidant capacity precedes neurological signs in a rat methylmercury intoxication model. Food Chem Toxicol 2020; 146:111810. [PMID: 33058990 DOI: 10.1016/j.fct.2020.111810] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/13/2022]
Abstract
The main target organ for MeHg is the nervous system, and its neurological dysfunction remains irreversible. Therefore, predictive biomarkers associated with individual susceptibility to MeHg and future clinical severity are needed to protect against the progression of MeHg toxicity. In this study, we demonstrated that plasma thiol antioxidant capacity (-SHp) is a useful predictive biomarker associated with future clinical severity using MeHg-intoxicated rats administered 1 mg/kg/day for 4 weeks. Blood samples were collected from the subclavian vein of each rat once a week to examine total blood mercury concentrations and the levels of plasma oxidative stress markers. Time course analyses of the correlation between these weekly blood examination values and hind limb crossing signs score after 4 weeks of MeHg exposure were performed, and plasma -SHp levels after 2 weeks of MeHg exposure showed strong correlations with future hind limb crossing sign scores. Neuropathological changes also developed in parallel with hind limb crossing sign scores. Quantitative analysis of vacuolar areas in the spinal cord showed a strong correlation with hind limb crossing sign scores. In conclusion, evaluation of plasma -SHp levels allowed us to detect individuals at risk for health damage and could protect the sensitive population against MeHg toxicity.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Science, National Institute for Minamata Disease, Kumamoto, Japan.
| | - Fusako Usuki
- Division of Neuroimmunology, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan
| | - Takamitsu Unoki
- Department of Basic Medical Science, National Institute for Minamata Disease, Kumamoto, Japan
| |
Collapse
|
25
|
Ajsuvakova OP, Tinkov AA, Aschner M, Rocha JB, Michalke B, Skalnaya MG, Skalny AV, Butnariu M, Dadar M, Sarac I, Aaseth J, Bjørklund G. Sulfhydryl groups as targets of mercury toxicity. Coord Chem Rev 2020; 417:213343. [PMID: 32905350 PMCID: PMC7470069 DOI: 10.1016/j.ccr.2020.213343] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The present study addresses existing data on the affinity and conjugation of sulfhydryl (thiol; -SH) groups of low- and high-molecular-weight biological ligands with mercury (Hg). The consequences of these interactions with special emphasis on pathways of Hg toxicity are highlighted. Cysteine (Cys) is considered the primary target of Hg, and link its sensitivity with thiol groups and cellular damage. In vivo, Hg complexes play a key role in Hg metabolism. Due to the increased affinity of Hg to SH groups in Cys residues, glutathione (GSH) is reactive. The geometry of Hg(II) glutathionates is less understood than that with Cys. Both Cys and GSH Hg-conjugates are important in Hg transport. The binding of Hg to Cys mediates multiple toxic effects of Hg, especially inhibitory effects on enzymes and other proteins that contain free Cys residues. In blood plasma, albumin is the main Hg-binding (Hg2+, CH3Hg+, C2H5Hg+, C6H5Hg+) protein. At the Cys34 residue, Hg2+ binds to albumin, whereas other metals likely are bound at the N-terminal site and multi-metal binding sites. In addition to albumin, Hg binds to multiple Cys-containing enzymes (including manganese-superoxide dismutase (Mn-SOD), arginase I, sorbitol dehydrogenase, and δ-aminolevulinate dehydratase, etc.) involved in multiple processes. The affinity of Hg for thiol groups may also underlie the pathways of Hg toxicity. In particular, Hg-SH may contribute to apoptosis modulation by interfering with Akt/CREB, Keap1/Nrf2, NF-κB, and mitochondrial pathways. Mercury-induced oxidative stress may ensue from Cys-Hg binding and inhibition of Mn-SOD (Cys196), thioredoxin reductase (TrxR) (Cys497) activity, as well as limiting GSH (GS-HgCH3) and Trx (Cys32, 35, 62, 65, 73) availability. Moreover, Hg-thiol interaction also is crucial in the neurotoxicity of Hg by modulating the cytoskeleton and neuronal receptors, to name a few. However, existing data on the role of Hg-SH binding in the Hg toxicity remains poorly defined. Therefore, more research is needed to understand better the role of Hg-thiol binding in the molecular pathways of Hg toxicology and the critical role of thiols to counteract negative effects of Hg overload.
Collapse
Affiliation(s)
- Olga P. Ajsuvakova
- Yaroslavl State University, Yaroslavl, Russia
- Federal Scientific Center of Biological Systems and Agrotechnologies of the Russian Academy of Sciences, Orenburg, Russia
- IM Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexey A. Tinkov
- Yaroslavl State University, Yaroslavl, Russia
- Federal Scientific Center of Biological Systems and Agrotechnologies of the Russian Academy of Sciences, Orenburg, Russia
- IM Sechenov First Moscow State Medical University, Moscow, Russia
| | - Michael Aschner
- IM Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - João B.T. Rocha
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | | | | | - Anatoly V. Skalny
- Yaroslavl State University, Yaroslavl, Russia
- Federal Scientific Center of Biological Systems and Agrotechnologies of the Russian Academy of Sciences, Orenburg, Russia
- IM Sechenov First Moscow State Medical University, Moscow, Russia
| | - Monica Butnariu
- Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timișoara, Timişoara, Romania
- CONEM Romania Biotechnology and Environmental Sciences Group, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timișoara, Timişoara, Romania
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Ioan Sarac
- Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timișoara, Timişoara, Romania
- CONEM Romania Biotechnology and Environmental Sciences Group, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timișoara, Timişoara, Romania
| | - Jan Aaseth
- IM Sechenov First Moscow State Medical University, Moscow, Russia
- Research Department, Innlandet Hospital Trust, Brumunddal, Norway
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| |
Collapse
|
26
|
Santos DB, Colle D, Moreira ELG, Santos AA, Hort MA, Santos K, Oses JP, Razzera G, Farina M. Probucol Protects Neuronal Cells Against Peroxide-Induced Damage and Directly Activates Glutathione Peroxidase-1. Mol Neurobiol 2020; 57:3245-3257. [PMID: 32506382 DOI: 10.1007/s12035-020-01963-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/28/2020] [Indexed: 10/24/2022]
Abstract
Experimental evidence has shown that probucol, a hypocholesterolemic agent, is also able to increase glutathione peroxidase (GPx) activity. However, there is a lack of knowledge about the mechanism(s) involved in this event. In this study, in vitro experiments with purified GPx1 from bovine erythrocytes and cultured SH-SY5Y neuroblastoma cells, as well as in silico studies with GPx1, were performed in order to elucidate mechanisms mediating the stimulatory effect of probucol on GPx activity and to investigate the relevance of this event in terms of susceptibility against peroxide-induced cytotoxicity. In vitro experiments with purified GPx1 showed a direct stimulatory effect of probucol on the activity of GPx1, which was related to an increase in Vmax with no changes in KM. Probucol also increased GPx activity in cultured SH-SY5Y neuroblastoma cells, while the levels of GPx1 expression were not changed, corroborating the results found with the purified enzyme. In addition, probucol rendered SH-SY5Y cells more resistant to hydroperoxide-induced cytotoxicity, and this event was abolished in GPx1 knocked-down cells. In silico studies with GPx1 pointed to a potential binding site for probucol at the close vicinity of the GSH pocket. Collectively, the results presented herein indicate that GPx1 plays a central role in the probucol-induced protective effects against peroxide toxicity. This highlights a novel target (GPx1) and a new mechanism of action (direct activation) for an "old drug." The relevance of such results for in vivo conditions deserves further investigation.
Collapse
Affiliation(s)
- Danúbia B Santos
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, 88040-900, Brazil.
| | - Dirleise Colle
- Department of Clinical Analyses, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Eduardo L G Moreira
- Department of Physiological Sciences, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Alessandra A Santos
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mariana A Hort
- Institute of Biological Sciences, Federal University of Rio Grande, Rio Grande, Rio Grande do Sul, Brazil
| | - Karin Santos
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, 88040-900, Brazil
| | - Jean P Oses
- Institute of Bioscience, Sector of Biochemistry, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Guilherme Razzera
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, 88040-900, Brazil
| | - Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, 88040-900, Brazil.
| |
Collapse
|
27
|
Bueno DC, Canto RFS, de Souza V, Andreguetti RR, Barbosa FAR, Naime AA, Dey PN, Wüllner V, Lopes MW, Braga AL, Methner A, Farina M. New Probucol Analogues Inhibit Ferroptosis, Improve Mitochondrial Parameters, and Induce Glutathione Peroxidase in HT22 Cells. Mol Neurobiol 2020; 57:3273-3290. [DOI: 10.1007/s12035-020-01956-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023]
|
28
|
Oxidative Stress Biomarkers in Erythrocytes of Captive Pre-Juvenile Loggerhead Turtles Following Acute Exposure to Methylmercury. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10103602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This study describes the use of erythrocytes (RBCs) of loggerhead turtles as in vitro models for evaluating their toxicity to methylmercury. Blood samples of loggerhead turtles that were born in the Colombian Caribbean were used. The LC50 of RBCs to methylmercury was determined at 96 h using methylmercury concentrations of 0.5–100 mg L−1. Next, the viability of the RBCs and the activity of the enzymes superoxide dismutase (SOD), glutathione S-transferase (GST), and lipid peroxidation by malondialdehyde (MDA) at 6 and 12 h of exposure to acute concentrations of 0, 1, and 5 mg L−1 were evaluated. The LC50 for loggerhead turtle RBCs was 8.32 mg L−1. The cell viability bioassay of RBCs exposed for 12 h only showed 100% cell viability. Increasing in vitro MeHg concentrations caused a corresponding increase in MDA concentration as well as decreases in the activities of SOD and GST. The RBCs represent an excellent model for ecotoxicological studies and SOD, GST, and MDA are biomarkers of environmental pollution and oxidative stress in loggerhead turtles. This was the first study conducted on loggerhead turtle where the response of RBCs to MeHg-induced oxidative stress is evaluated.
Collapse
|
29
|
Bittarello AC, Vieira JCS, Braga CP, da Cunha Bataglioli I, de Oliveira G, Rocha LC, Zara LF, Buzalaf MAR, de Oliveira LCS, Adamec J, de Magalhães Padilha P. Metalloproteomic approach of mercury-binding proteins in liver and kidney tissues of Plagioscion squamosissimus (corvina) and Colossoma macropomum (tambaqui) from Amazon region: Possible identification of mercury contamination biomarkers. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 711:134547. [PMID: 31812405 DOI: 10.1016/j.scitotenv.2019.134547] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 06/10/2023]
Abstract
Fish is an important source of protein, vitamins, and minerals. However, this food is also a major source of human exposure to toxic contaminants such as mercury. Thus, this paper aimed to evaluate mercury-binding proteins for possible application as biomarkers of mercury contamination in hepatic and renal tissues of Plagioscion squamosissimus (carnivorous fish) and Colossoma macropomum (omnivorous fish) from the Amazon region using metalloproteomic approach. The proteome of hepatic and renal tissues of fish species was separated by two-dimensional polyacrylamide gel electrophoresis (2D-PAGE), and the mercury concentrations in protein spots were determined by graphite furnace atomic absorption spectrometry (GFAAS). Finally, the protein spots associated to mercury were characterized by electrospray ionization mass spectrometry (ESI-MS/MS). The activity of antioxidant enzymes (SOD, CAT, GPx, and GST) and lipid peroxidation (LPO) were also determined. The results showed that the highest concentrations of mercury were found in the carnivorous species (P. squamosissimus) and that the accumulation pattern of this metal was higher in hepatic tissues than in renal tissues for both species. A tendency was observed for greater enzymatic activity in the hepatic and renal tissues of P. squamosissimus, the species with the highest concentration of mercury. Only GPx activity in the kidney and GST in the liver were lower for the P. squamosissimus species, and this finding can be explained by the interaction of mercury with these enzymes. The data obtained by ESI-MS/MS allowed for the characterization of the protein spots associated to mercury, revealing proteins involved in energy metabolism, biomolecules transport, protein synthesis and degradation, cell differentiation, gene regulation, and the antioxidant system. The results obtained in the present study can contribute to understanding the physiological processes underlying mercury toxicity and have provided new perspectives on possible candidates for mercury contamination biomarkers in fish.
Collapse
Affiliation(s)
- Alis Correia Bittarello
- São Paulo State University (UNESP), School of Veterinary Medicine and Animal Science, Botucatu, Brazil
| | - José Cavalcante Souza Vieira
- São Paulo State University (UNESP), Institute of Biosciences, Botucatu, Brazil; Institute of Chemistry (INQUI), Federal University of Mato Grosso do Sul, Campo Grande (UFMS), Brazil.
| | | | | | | | - Leone Campos Rocha
- São Paulo State University (UNESP), School of Veterinary Medicine and Animal Science, Botucatu, Brazil
| | - Luiz Fabrício Zara
- University of Brasília (UNB), College of Planaltina, Distrito Federal, Brazil
| | | | | | - Jiri Adamec
- University of Nebraska (UNL), Lincoln, United States
| | - Pedro de Magalhães Padilha
- São Paulo State University (UNESP), School of Veterinary Medicine and Animal Science, Botucatu, Brazil; São Paulo State University (UNESP), Institute of Biosciences, Botucatu, Brazil.
| |
Collapse
|
30
|
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
|
31
|
Oliveira LF, Rodrigues LD, Cardillo GM, Nejm MB, Guimarães-Marques M, Reyes-Garcia SZ, Zuqui K, Vassallo DV, Fiorini AC, Scorza CA, Scorza FA. Deleterious effects of chronic mercury exposure on in vitro LTP, memory process, and oxidative stress. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:7559-7569. [PMID: 31885058 DOI: 10.1007/s11356-019-06625-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 09/25/2019] [Indexed: 05/21/2023]
Abstract
Heavy metal contamination in aquatic environments plays an important role in the exposure of humans to these toxicants. Among these pollutants, mercury (Hg) is one main concern due to its high neurotoxicity and environmental persistence. Even in low concentrations, Hg bioaccumulation is a major threat to human health, with higher impact on populations whose diet has fish as chief consumption. Mercury compounds have high affinity for neuronal receptors and proteins, which gives Hg its cumulative feature and have the ability to cross cell membranes and blood-brain barrier to show their neurotoxicity. Intoxication with Hg increases levels of reactive oxygen species (ROS), thus depleting faster the resource of antioxidant proteins. To evaluate Hg-induced hippocampal ROS production, synaptic plasticity, anxiety, and memory, a total of 11 male Wistar rats were exposed to HgCl2 (Hg30 group) to produce a residual concentration of 8 ng/mL at the end of 30 days. Behavioral tests (plus-maze discriminative avoidance task), in vitro electrophysiology, and ROS assays were performed. Western blot assay showed decreased levels of antioxidant proteins GPx and SOD in Hg30 group. Increased ROS production was observed in the CA1 and CA3 regions in the Hg-exposed group. Plus-maze task detected long-term memory impairment in Hg30 group, linked to poorer in vitro long-term potentiation as compared to control group. Hg intoxication also promoted higher anxiety-like behavior in the exposed animals. In conclusion, our data suggests that low doses of HgCl2 resulted in impaired long-term memory and unbalance between decreased antioxidant protein expression and increased ROS production in the hippocampus.
Collapse
Affiliation(s)
- Leandro F Oliveira
- Department of Neurology and Neurosurgery, UNIFESP/EPM, São Paulo, Brazil
| | - Laís D Rodrigues
- Department of Neurology and Neurosurgery, UNIFESP/EPM, São Paulo, Brazil
| | | | - Mariana B Nejm
- Department of Neurology and Neurosurgery, UNIFESP/EPM, São Paulo, Brazil
| | | | - Selvin Z Reyes-Garcia
- Department of Neurology and Neurosurgery, UNIFESP/EPM, São Paulo, Brazil
- Department of Morphological Science, Faculty of Medical Sciences, National Autonomous University of Honduras, San Pedro Sula, Honduras
| | - Karolini Zuqui
- Department of Physiological Sciences, Federal University of Espírito Santo, Espírito Santo, Brazil
| | - Dalton V Vassallo
- Department of Physiological Sciences, Federal University of Espírito Santo, Espírito Santo, Brazil
| | - Ana C Fiorini
- Department of Department of Speech-Language Pathology, Audiology, UNIFESP/EPM, Brazil and Pontifical Catholic University, São Paulo, Brazil
| | - Carla A Scorza
- Department of Neurology and Neurosurgery, UNIFESP/EPM, São Paulo, Brazil
| | - Fulvio A Scorza
- Department of Neurology and Neurosurgery, UNIFESP/EPM, São Paulo, Brazil.
| |
Collapse
|
32
|
Aaseth J, Wallace DR, Vejrup K, Alexander J. Methylmercury and developmental neurotoxicity: A global concern. CURRENT OPINION IN TOXICOLOGY 2020. [DOI: 10.1016/j.cotox.2020.01.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
33
|
Stability and biological testing of taurine-conjugated bile acid antioxidant microcapsules for diabetes treatment. Ther Deliv 2020; 10:99-106. [PMID: 30729887 DOI: 10.4155/tde-2018-0034] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM Taurine-conjugated bile acids possess positive formulation-stabilization effects, which are desirable in diabetes treatments. The taurine-conjugated bile acid, taurocholic acid (TCA), has shown promising formulation-stabilizing effects on the delivery of the antioxidant drug, probucol (PB), but success is limited due to its poor release profile. This study aimed to design new PB-TCA formulations using new polymers, and examine antioxidant and antidiabetic effects using β-cells for PB with or without TCA. MATERIALS AND METHODS Different formulations using alginate-insoluble esters of polymethylacrylate polymers encapsulating PB and TCA were developed, microencapsulated and examined for stability and biological activity. RESULTS TCA addition to new PB matrices improved osmotic and mechanical properties, and this effect was dependent on polymethylacrylate composition and concentration. CONCLUSION TCA can optimize the oral delivery of anti-diabetic compounds.
Collapse
|
34
|
Huang JL, Yu C, Su M, Yang SM, Zhang F, Chen YY, Liu JY, Jiang YF, Zhong ZG, Wu DP. Probucol, a "non-statin" cholesterol-lowering drug, ameliorates D-galactose induced cognitive deficits by alleviating oxidative stress via Keap1/Nrf2 signaling pathway in mice. Aging (Albany NY) 2019; 11:8542-8555. [PMID: 31590160 PMCID: PMC6814622 DOI: 10.18632/aging.102337] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022]
Abstract
Oxidative stress plays a vital role in the initiation and progression of age-related neurodegenerative diseases. Ameliorating oxidative damage is therefore considered as a beneficial strategy for the treatment of age-related neurodegenerative disorders. Probucol (Prob), a lipid-lowering prototype agent, was reported to treat cardiovascular diseases, chronic kidney disease and diabetes mellitus. However, whether Prob has an effect on age-related neurodegenerative diseases remains unknown. In the study, it was found that Prob ameliorated D-galactose (D-gal) induced cognitive deficits and neuronal loss in the hippocampal CA1 region. Moreover, Prob alleviated ROS and MDA levels by elevating SOD, GSH-PX and HO-1 mRNA and protein expressions, and improving plasmic and cerebral SOD and GSH-PX activities in D-gal treated mice. Furthermore, Prob promoted the dissociation of Keap1/Nrf2 complex leading to the accumulation of Nrf2 in nucleus, implying that the improved anti-oxidant property of Prob is mediated by Keap1/Nrf2 pathway. The study firstly demonstrates the favorable effects of Prob against cognitive impairments in a senescent mouse model, rendering this compound a promising agent for the treatment or prevention of age-related neurodegenerative disease.
Collapse
Affiliation(s)
- Jin-Lan Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Chao Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Min Su
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Si-Man Yang
- Scientific research center of traditional Chinese medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Fan Zhang
- Scientific research center of traditional Chinese medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Yuan-Yuan Chen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jin-Yuan Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yi-Fan Jiang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Zhen-Guo Zhong
- Scientific research center of traditional Chinese medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Deng-Pan Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| |
Collapse
|
35
|
Takahashi T, Shimohata T. Vascular Dysfunction Induced by Mercury Exposure. Int J Mol Sci 2019; 20:E2435. [PMID: 31100949 PMCID: PMC6566353 DOI: 10.3390/ijms20102435] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/10/2019] [Accepted: 05/16/2019] [Indexed: 12/15/2022] Open
Abstract
Methylmercury (MeHg) causes severe damage to the central nervous system, and there is increasing evidence of the association between MeHg exposure and vascular dysfunction, hemorrhage, and edema in the brain, but not in other organs of patients with acute MeHg intoxication. These observations suggest that MeHg possibly causes blood-brain barrier (BBB) damage. MeHg penetrates the BBB into the brain parenchyma via active transport systems, mainly the l-type amino acid transporter 1, on endothelial cell membranes. Recently, exposure to mercury has significantly increased. Numerous reports suggest that long-term low-level MeHg exposure can impair endothelial function and increase the risks of cardiovascular disease. The most widely reported mechanism of MeHg toxicity is oxidative stress and related pathways, such as neuroinflammation. BBB dysfunction has been suggested by both in vitro and in vivo models of MeHg intoxication. Therapy targeted at both maintaining the BBB and suppressing oxidative stress may represent a promising therapeutic strategy for MeHg intoxication. This paper reviews studies on the relationship between MeHg exposure and vascular dysfunction, with a special emphasis on the BBB.
Collapse
Affiliation(s)
- Tetsuya Takahashi
- Department of Neurology, National Hospital Organization Nishiniigata Chuo Hospital, Niigata 950-2085, Japan.
| | - Takayoshi Shimohata
- Department of Neurology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan.
| |
Collapse
|
36
|
Kuras R, Kozlowska L, Reszka E, Wieczorek E, Jablonska E, Gromadzinska J, Stanislawska M, Janasik B, Wasowicz W. Environmental mercury exposure and selenium-associated biomarkers of antioxidant status at molecular and biochemical level. A short-term intervention study. Food Chem Toxicol 2019; 130:187-198. [PMID: 31078725 DOI: 10.1016/j.fct.2019.04.056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/16/2019] [Accepted: 04/29/2019] [Indexed: 11/30/2022]
Abstract
Mercury (Hg) is a potent toxicant. In the field of public health a chronic-low-level environmental Hg exposure resulting from fish consumption in general population is still being discussed. The objective of the study was to assess the influence of real Hg exposure on biomarkers of selenium (Se) status and selected biomarkers of pro-oxidant/anti-oxidant effects in healthy men (n = 67) who participated in the short-term intervention study consisting in daily fish consumption for two weeks. The analysis included Se level, Se-associated antioxidants at molecular (profile of 7 genes encoding selected proteins related to antioxidant defense) and biochemical levels (Se-dependent glutathione peroxidases activities and plasma selenoprotein P concentration). A pro-oxidant/anti-oxidant balance was explored using a biomarker of plasma lipid peroxidation and total antioxidant activity. The study revealed significant correlations (p < 0.05) between the biomarkers of exposure to Hg, Se level and Se-dependent antioxidants. Even though the risk of adverse effects of Hg for volunteers was substantially low, biomarkers of Hg altered levels of circulation selenoproteins and their genes expression. Changes in genes expression during study differed between the main enzymes involved in two systems: downregulation of thioredoxin reductase1 and upregulation of glutathione peroxidases. Hg exposure caused imbalance between the biomarkers of pro-oxidant/anti-oxidant effects.
Collapse
Affiliation(s)
- Renata Kuras
- Nofer Institute of Occupational Medicine, Department of Biological and Environmental Monitoring, 8 Teresy St, 91-348, Lodz, Poland.
| | - Lucyna Kozlowska
- Department of Dietetics, Faculty of Human Nutrition and Consumer Sciences, University of Life Sciences, Nowoursynowska 159c St., Building 32, 02-776, Warsaw, Poland
| | - Edyta Reszka
- Nofer Institute of Occupational Medicine, Department of Molecular Genetics and Epigenetics, 8 Teresy St, 91-348, Lodz, Poland
| | - Edyta Wieczorek
- Nofer Institute of Occupational Medicine, Department of Molecular Genetics and Epigenetics, 8 Teresy St, 91-348, Lodz, Poland
| | - Ewa Jablonska
- Nofer Institute of Occupational Medicine, Department of Molecular Genetics and Epigenetics, 8 Teresy St, 91-348, Lodz, Poland
| | - Jolanta Gromadzinska
- Nofer Institute of Occupational Medicine, Department of Biological and Environmental Monitoring, 8 Teresy St, 91-348, Lodz, Poland
| | - Magdalena Stanislawska
- Nofer Institute of Occupational Medicine, Department of Biological and Environmental Monitoring, 8 Teresy St, 91-348, Lodz, Poland
| | - Beata Janasik
- Nofer Institute of Occupational Medicine, Department of Biological and Environmental Monitoring, 8 Teresy St, 91-348, Lodz, Poland
| | - Wojciech Wasowicz
- Nofer Institute of Occupational Medicine, Department of Biological and Environmental Monitoring, 8 Teresy St, 91-348, Lodz, Poland
| |
Collapse
|
37
|
Prince LM, Aschner M, Bowman AB. Human-induced pluripotent stems cells as a model to dissect the selective neurotoxicity of methylmercury. Biochim Biophys Acta Gen Subj 2019; 1863:129300. [PMID: 30742955 DOI: 10.1016/j.bbagen.2019.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/09/2019] [Accepted: 02/01/2019] [Indexed: 01/07/2023]
Abstract
Methylmercury (MeHg) is a potent neurotoxicant affecting both the developing and mature central nervous system (CNS) with apparent indiscriminate disruption of multiple homeostatic pathways. However, genetic and environmental modifiers contribute significant variability to neurotoxicity associated with human exposures. MeHg displays developmental stage and neural lineage selective neurotoxicity. To identify mechanistic-based neuroprotective strategies to mitigate human MeHg exposure risk, it will be critical to improve our understanding of the basis of MeHg neurotoxicity and of this selective neurotoxicity. Here, we propose that human-based pluripotent stem cell cellular approaches may enable mechanistic insight into genetic pathways that modify sensitivity of specific neural lineages to MeHg-induced neurotoxicity. Such studies are crucial for the development of novel disease modifying strategies impinging on MeHg exposure vulnerability.
Collapse
Affiliation(s)
- Lisa M Prince
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, United States.
| |
Collapse
|
38
|
Nogara PA, Oliveira CS, Schmitz GL, Piquini PC, Farina M, Aschner M, Rocha JBT. Methylmercury's chemistry: From the environment to the mammalian brain. Biochim Biophys Acta Gen Subj 2019; 1863:129284. [PMID: 30659885 DOI: 10.1016/j.bbagen.2019.01.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 12/14/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023]
Abstract
Methylmercury is a neurotoxicant that is found in fish and rice. MeHg's toxicity is mediated by blockage of -SH and -SeH groups of proteins. However, the identification of MeHg's targets is elusive. Here we focus on the chemistry of MeHg in the abiotic and biotic environment. The toxicological chemistry of MeHg is complex in metazoans, but at the atomic level it can be explained by exchange reactions of MeHg bound to -S(e)H with another free -S(e)H group (R1S(e)-HgMe + R2-S(e)H ↔ R1S(e)H + R2-S(e)-HgMe). This reaction was first studied by professor Rabenstein and here it is referred as the "Rabenstein's Reaction". The absorption, distribution, and excretion of MeHg in the environment and in the body of animals will be dictated by Rabenstein's reactions. The affinity of MeHg by thiol and selenol groups and the exchange of MeHg by Rabenstein's Reaction (which is a diffusion controlled reaction) dictates MeHg's neurotoxicity. However, it is important to emphasize that the MeHg exchange reaction velocity with different types of thiol- and selenol-containing proteins will also depend on protein-specific structural and thermodynamical factors. New experimental approaches and detailed studies about the Rabenstein's reaction between MeHg with low molecular mass thiol (LMM-SH) molecules (cysteine, GSH, acetyl-CoA, lipoate, homocysteine) with abundant high molecular mass thiol (HMM-SH) molecules (albumin, hemoglobin) and HMM-SeH (GPxs, Selenoprotein P, TrxR1-3) are needed. The study of MeHg migration from -S(e)-Hg- bonds to free -S(e)H groups (Rabenstein's Reaction) in pure chemical systems and neural cells (with special emphasis to the LMM-SH and HMM-S(e)H molecules cited above) will be critical to developing realistic constants to be used in silico models that will predict the distribution of MeHg in humans.
Collapse
Affiliation(s)
- Pablo A Nogara
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Cláudia S Oliveira
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Gabriela L Schmitz
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Paulo C Piquini
- Departamento de Física, CCNE, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - João B T Rocha
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
39
|
Farina M, Aschner M. Glutathione antioxidant system and methylmercury-induced neurotoxicity: An intriguing interplay. Biochim Biophys Acta Gen Subj 2019; 1863:129285. [PMID: 30659883 DOI: 10.1016/j.bbagen.2019.01.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/09/2018] [Accepted: 01/09/2019] [Indexed: 01/20/2023]
Abstract
Methylmercury (MeHg) is a toxic chemical compound naturally produced mainly in the aquatic environment through the methylation of inorganic mercury catalyzed by aquatic microorganisms. MeHg is biomagnified in the aquatic food chain and, consequently, piscivorous fish at the top of the food chain possess huge amounts of MeHg (at the ppm level). Some populations that have fish as main protein's source can be exposed to exceedingly high levels of MeHg and develop signs of toxicity. MeHg is toxic to several organs, but the central nervous system (CNS) represents a preferential target, especially during development (prenatal and early postnatal periods). Though the biochemical events involved in MeHg-(neuro)toxicity are not yet entirely comprehended, a vast literature indicates that its pro-oxidative properties explain, at least partially, several of its neurotoxic effects. As result of its electrophilicity, MeHg interacts with (and oxidize) nucleophilic groups, such as thiols and selenols, present in proteins or low-molecular weight molecules. It is noteworthy that such interactions modify the redox state of these groups and, therefore, lead to oxidative stress and impaired function of several molecules, culminating in neurotoxicity. Among these molecules, glutathione (GSH; a major thiol antioxidant) and thiol- or selenol-containing enzymes belonging to the GSH antioxidant system represent key molecular targets involved in MeHg-neurotoxicity. In this review, we firstly present a general overview concerning the neurotoxicity of MeHg. Then, we present fundamental aspects of the GSH-antioxidant system, as well as the effects of MeHg on this system.
Collapse
Affiliation(s)
- Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, USA
| |
Collapse
|
40
|
Colle D, Santos DB, de Souza V, Lopes MW, Leal RB, de Souza Brocardo P, Farina M. Sodium selenite protects from 3-nitropropionic acid-induced oxidative stress in cultured primary cortical neurons. Mol Biol Rep 2018; 46:751-762. [PMID: 30511305 DOI: 10.1007/s11033-018-4531-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/28/2018] [Indexed: 12/21/2022]
Abstract
Selenium (Se) is an essential trace element for humans; its intake is needed to allow the proper synthesis of 25 different selenoproteins that are necessary to the normal functioning of several organs, including the brain. Accordingly, decreased Se levels have been associated with neurological disorders. In the present study, we investigated the potential beneficial effects of Se, as sodium selenite, against 3-nitropropionic acid (3-NP)-induced oxidative stress in primary cultures of mouse cortical neurons. 3-NP treatment caused a significant decrease in cellular viability, which was accompanied by decreases in mitochondrial complex II activity and reduced glutathione (GSH) content, as well as increases in reactive oxygen species (ROS) generation and oxidized glutathione (GSSG) levels. Sodium selenite pretreatment (6 days) attenuated 3-NP-induced decrease in cell viability. In addition, sodium selenite pretreatment significantly protected against 3-NP-induced increase in ROS generation and decrease in GSH/GSSG ratio. Of note, sodium selenite pretreatment did not change 3-NP-induced decrease of mitochondrial complex II activity, suggesting that Se modulates secondary events resultant from 3-NP-induced mitochondrial dyshomeostasis. In addition, sodium selenite pretreatment significantly increased glutathione peroxidase (GPx) activity. Our data provide insights into the mechanism of protection by sodium selenite, which is related, at least in part, to GPx induction.
Collapse
Affiliation(s)
- Dirleise Colle
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil. .,Departamento de Análises Clínicas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, CEP 88040-900, Brazil.
| | - Danúbia Bonfanti Santos
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil
| | - Viviane de Souza
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil
| | - Mark William Lopes
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil
| | - Rodrigo Bainy Leal
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil
| | - Patricia de Souza Brocardo
- Departamento de Ciências Morfológicas, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil.
| |
Collapse
|
41
|
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
|
42
|
Ghizoni H, Ventura M, Colle D, Gonçalves CL, de Souza V, Hartwig JM, Santos DB, Naime AA, Cristina de Oliveira Souza V, Lopes MW, Barbosa F, Brocardo PS, Farina M. Effects of perinatal exposure to n-3 polyunsaturated fatty acids and methylmercury on cerebellar and behavioral parameters in mice. Food Chem Toxicol 2018; 120:603-615. [DOI: 10.1016/j.fct.2018.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022]
|
43
|
Antidepressant Effects of Probucol on Early-Symptomatic YAC128 Transgenic Mice for Huntington's Disease. Neural Plast 2018; 2018:4056383. [PMID: 30186318 PMCID: PMC6112232 DOI: 10.1155/2018/4056383] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/28/2018] [Accepted: 07/26/2018] [Indexed: 11/17/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a trinucleotide expansion in the HD gene, resulting in an extended polyglutamine tract in the protein huntingtin. HD is traditionally viewed as a movement disorder, but cognitive and neuropsychiatric symptoms also contribute to the clinical presentation. Depression is one of the most common psychiatric disturbances in HD, present even before manifestation of motor symptoms. Diagnosis and treatment of depression in HD-affected individuals are essential aspects of clinical management in this population, especially owing to the high risk of suicide. This study investigated whether chronic administration of the antioxidant probucol improved motor and affective symptoms as well as hippocampal neurogenic function in the YAC128 transgenic mouse model of HD during the early- to mild-symptomatic stages of disease progression. The motor performance and affective symptoms were monitored using well-validated behavioral tests in YAC128 mice and age-matched wild-type littermates at 2, 4, and 6 months of age, after 1, 3, or 5 months of treatment with probucol (30 mg/kg/day via water supplementation, starting on postnatal day 30). Endogenous markers were used to assess the effect of probucol on cell proliferation (Ki-67 and proliferation cell nuclear antigen (PCNA)) and neuronal differentiation (doublecortin (DCX)) in the hippocampal dentate gyrus (DG). Chronic treatment with probucol reduced the occurrence of depressive-like behaviors in early- and mild-symptomatic YAC128 mice. Functional improvements were not accompanied by increased progenitor cell proliferation and neuronal differentiation. Our findings provide evidence that administration of probucol may be of clinical benefit in the management of early- to mild-symptomatic HD.
Collapse
|
44
|
Antunes Dos Santos A, Ferrer B, Marques Gonçalves F, Tsatsakis AM, Renieri EA, Skalny AV, Farina M, Rocha JBT, Aschner M. Oxidative Stress in Methylmercury-Induced Cell Toxicity. TOXICS 2018; 6:toxics6030047. [PMID: 30096882 PMCID: PMC6161175 DOI: 10.3390/toxics6030047] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/03/2018] [Accepted: 08/07/2018] [Indexed: 02/06/2023]
Abstract
Methylmercury (MeHg) is a hazardous environmental pollutant, which elicits significant toxicity in humans. The accumulation of MeHg through the daily consumption of large predatory fish poses potential health risks, and the central nervous system (CNS) is the primary target of toxicity. Despite well-described neurobehavioral effects (i.e., motor impairment), the mechanisms of MeHg-induced toxicity are not completely understood. However, several lines of evidence point out the oxidative stress as an important molecular mechanism in MeHg-induced intoxication. Indeed, MeHg is a soft electrophile that preferentially interacts with nucleophilic groups (mainly thiols and selenols) from proteins and low-molecular-weight molecules. Such interaction contributes to the occurrence of oxidative stress, which can produce damage by several interacting mechanisms, impairing the function of various molecules (i.e., proteins, lipids, and nucleic acids), potentially resulting in modulation of different cellular signal transduction pathways. This review summarizes the general aspects regarding the interaction between MeHg with regulators of the antioxidant response system that are rich in thiol and selenol groups such as glutathione (GSH), and the selenoenzymes thioredoxin reductase (TrxR) and glutathione peroxidase (Gpx). A particular attention is directed towards the role of the PI3K/Akt signaling pathway and the nuclear transcription factor NF-E2-related factor 2 (Nrf2) in MeHg-induced redox imbalance.
Collapse
Affiliation(s)
| | - Beatriz Ferrer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Filipe Marques Gonçalves
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Aristides M Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece.
| | - Elisavet A Renieri
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece.
| | - Anatoly V Skalny
- Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), Moscow 150000, Russia.
- Laboratory of Biotechnology and Applied Bioelementology, Yaroslavl State University, Yaroslavl 150014, Russia.
- All-Russian Research Institute of Medicinal and Aromatic Plants (VILAR), Moscow 150000, Russia.
| | - Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis 88040-900, Santa Catarina, Brazil.
| | - João B T Rocha
- Department of Biochemistry, Federal University of Santa Maria, Santa Maria 97105-900, Rio Grande do Sul, Brazil.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
45
|
Olguín N, Müller ML, Rodríguez-Farré E, Suñol C. Neurotransmitter amines and antioxidant agents in neuronal protection against methylmercury-induced cytotoxicity in primary cultures of mice cortical neurons. Neurotoxicology 2018; 69:278-287. [PMID: 30075218 DOI: 10.1016/j.neuro.2018.07.020] [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: 11/28/2017] [Revised: 07/26/2018] [Accepted: 07/30/2018] [Indexed: 12/23/2022]
Abstract
Methylmercury (MeHg) is an environmental toxicant with detrimental effects on the developing brain and adult nervous system. The main mechanisms identified include oxidative stress, changes in intracellular calcium, mitochondrial changes, inhibition of glutamate uptake, of protein synthesis and disruption of microtubules. However, little is known about mechanisms of protection against MeHg neurotoxicity. We found that resveratrol (10 μM) and ascorbic acid (200 μM) protected MeHg-induced cell death in primary cultures of cortical neurons. In this work, we aimed at finding additional targets that may be related to MeHg mode of action in cell toxicity with special emphasis in cell protection. We wonder whether neurotransmitters may affect the MeHg effects on neuronal death. Our findings show that neurons exposed to low MeHg concentrations exhibit less mortality if co-exposed to 10 μM dopamine (DA). However, DA metabolites, HVA (homovanillic acid) and DOPAC (3,4-dihydroxyphenylacetic acid) are not responsible for such protection. Furthermore, both DA D1 and D2 receptors agonists showed a protective effect against MeHg toxicity. It is striking though that DA receptor antagonists SKF83566 (10 μM) and haloperidol (10 μM) did not inhibit DA protection against MeHg. In addition, the protective effect of 10 μM DA against MeHg-induced toxicity was not affected by additional organochlorine pollutants exposure. Our results also demonstrate that cells exposed to MeHg in presence of 100 μM acetylcholine (ACh), show an increase in cell mortality at the "threshold value" of 100 nM MeHg. Finally, norepinephrine (10 μM) and serotonin (20 μM) also had an effect on cell protection. Altogether, we propose to further investigate the additional mechanisms that may be playing an important role in MeHg-induced cytotoxicity.
Collapse
Affiliation(s)
- Nair Olguín
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC - IDIBAPS, CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Marie-Lena Müller
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC - IDIBAPS, CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Eduard Rodríguez-Farré
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC - IDIBAPS, CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Cristina Suñol
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC - IDIBAPS, CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain.
| |
Collapse
|
46
|
Martinez CS, Peçanha FM, Brum DS, Santos FW, Franco JL, Zemolin APP, Anselmo-Franci JA, Junior FB, Alonso MJ, Salaices M, Vassallo DV, Leivas FG, Wiggers GA. Reproductive dysfunction after mercury exposure at low levels: evidence for a role of glutathione peroxidase (GPx) 1 and GPx4 in male rats. Reprod Fertil Dev 2018; 29:1803-1812. [PMID: 27755963 DOI: 10.1071/rd16310] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 09/22/2016] [Indexed: 01/26/2023] Open
Abstract
Mercury is a ubiquitous environmental pollutant and mercury contamination and toxicity are serious hazards to human health. Some studies have shown that mercury impairs male reproductive function, but less is known about its effects following exposure at low doses and the possible mechanisms underlying its toxicity. Herein we show that exposure of rats to mercury chloride for 30 days (first dose 4.6µgkg-1, subsequent doses 0.07µgkg-1day-1) resulted in mean (±s.e.m.) blood mercury concentrations of 6.8±0.3ngmL-1, similar to that found in human blood after occupational exposure or released from removal of amalgam fillings. Even at these low concentrations, mercury was deposited in reproductive organs (testis, epididymis and prostate), impaired sperm membrane integrity, reduced the number of mature spermatozoa and, in the testes, promoted disorganisation, empty spaces and loss of germinal epithelium. Mercury increased levels of reactive oxygen species and the expression of glutathione peroxidase (GPx) 1 and GPx4. These results suggest that the toxic effects of mercury on the male reproductive system are due to its accumulation in reproductive organs and that the glutathione system is its potential target. The data also suggest, for the first time, a possible role of the selenoproteins GPx1 and GPx4 in the reproductive toxicity of mercury chloride.
Collapse
Affiliation(s)
- Caroline S Martinez
- Postgraduate Program in Biochemistry, Postgraduate Program in Animal Science and Postgraduate Program in Biological Science, Universidade Federal do Pampa, BR 472 - Km 592 -118, 97500-970 Uruguaiana, Rio Grande do Sul, Brazil
| | - Franck M Peçanha
- Postgraduate Program in Biochemistry, Postgraduate Program in Animal Science and Postgraduate Program in Biological Science, Universidade Federal do Pampa, BR 472 - Km 592 -118, 97500-970 Uruguaiana, Rio Grande do Sul, Brazil
| | - Daniela S Brum
- Postgraduate Program in Biochemistry, Postgraduate Program in Animal Science and Postgraduate Program in Biological Science, Universidade Federal do Pampa, BR 472 - Km 592 -118, 97500-970 Uruguaiana, Rio Grande do Sul, Brazil
| | - Francielli W Santos
- Postgraduate Program in Biochemistry, Postgraduate Program in Animal Science and Postgraduate Program in Biological Science, Universidade Federal do Pampa, BR 472 - Km 592 -118, 97500-970 Uruguaiana, Rio Grande do Sul, Brazil
| | - Jeferson L Franco
- Postgraduate Program in Biochemistry, Postgraduate Program in Animal Science and Postgraduate Program in Biological Science, Universidade Federal do Pampa, BR 472 - Km 592 -118, 97500-970 Uruguaiana, Rio Grande do Sul, Brazil
| | - Ana Paula P Zemolin
- Postgraduate Program in Biochemistry, Postgraduate Program in Animal Science and Postgraduate Program in Biological Science, Universidade Federal do Pampa, BR 472 - Km 592 -118, 97500-970 Uruguaiana, Rio Grande do Sul, Brazil
| | - Janete A Anselmo-Franci
- Department of Physiology, School of Medicine, Universidade de São Paulo, Av. do Café s/n, 14040904, Ribeirão Preto, São Paulo, Brazil
| | - Fernando B Junior
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, Universidade de São Paulo, Av. do Café s/n, 14049-903, Ribeirão Preto, São Paulo, Brazil
| | - María J Alonso
- Department of Biochemistry, Physiology and Molecular Genetics, Universidad Rey Juan Carlos, Avda. Atenas s/n, 28922, Alcorcón, Spain
| | - Mercedes Salaices
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Arzobispo Morcillo 4, 28029, Madrid, Spain
| | - Dalton V Vassallo
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Av. Marechal Campos 1468, 29040-090, Vitória, Espírito Santo, Brazil
| | - Fábio G Leivas
- Postgraduate Program in Biochemistry, Postgraduate Program in Animal Science and Postgraduate Program in Biological Science, Universidade Federal do Pampa, BR 472 - Km 592 -118, 97500-970 Uruguaiana, Rio Grande do Sul, Brazil
| | - Giulia A Wiggers
- Postgraduate Program in Biochemistry, Postgraduate Program in Animal Science and Postgraduate Program in Biological Science, Universidade Federal do Pampa, BR 472 - Km 592 -118, 97500-970 Uruguaiana, Rio Grande do Sul, Brazil
| |
Collapse
|
47
|
Li Y, He B, Gao J, Liu QS, Liu R, Qu G, Shi J, Hu L, Jiang G. Methylmercury exposure alters RNA splicing in human neuroblastoma SK-N-SH cells: Implications from proteomic and post-transcriptional responses. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 238:213-221. [PMID: 29554569 DOI: 10.1016/j.envpol.2018.03.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/03/2018] [Accepted: 03/08/2018] [Indexed: 06/08/2023]
Abstract
The neurotoxic effects of methylmercury (MeHg) have been intensively studied. However, the molecular mechanisms responsible for the neurotoxicity of MeHg are not fully understood. To decipher these mechanisms, proteomic and high-throughput mRNA sequencing (RNA-seq) technique were utilized, comprehensively evaluating the cellular responses of human neuroblastoma SK-N-SH cells to MeHg exposure. Proteomic results revealed that MeHg exposure interfered with RNA splicing via splicesome, along with the known molecular mechanisms of mercury-related neurotoxicity (e.g. oxidative stress, protein folding, immune system processes, and cytoskeletal organization). The effects of MeHg on RNA splicing were further verified using RNA-seq. Compared to control, a total of 658 aberrant RNA alternative splicing (AS) events were observed after MeHg exposure. Proteomics and RNA-seq results also demonstrated that mercury chloride (HgCl2) influenced the expression levels of several RNA splicing related proteins and 676 AS events compared to control. These results suggested that RNA splicing could be a new molecular mechanism involved in MeHg and HgCl2 neurotoxicity.
Collapse
Affiliation(s)
- Yiling Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Bin He
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Jiejun Gao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Qian S Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Runzeng Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Guangbo Qu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Jianbo Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Ligang Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; Institute of Environment and Health, Jianghan University, Wuhan, 430056, China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
48
|
Fujimura M, Usuki F. Methylmercury induces oxidative stress and subsequent neural hyperactivity leading to cell death through the p38 MAPK-CREB pathway in differentiated SH-SY5Y cells. Neurotoxicology 2018; 67:226-233. [PMID: 29913201 DOI: 10.1016/j.neuro.2018.06.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/13/2018] [Accepted: 06/13/2018] [Indexed: 12/20/2022]
Abstract
Methylmercury (MeHg) induces site-specific cerebrocortical neuronal cell death. In our previous study using an in vivo mouse model, we reported that MeHg-induced cerebrocortical neuronal cell death may be due to neural hyperactivity triggered by activation of kinase pathways. However, the detailed molecular mechanism remained to be completely understood. In this study, we analyzed detailed signaling pathways for MeHg-induced neuronal cell death using all-trans-retinoic acid (RA) differentiated SH-SY5Y cells, which show neuron-like morphological changes and express neuron/synapse markers for cerebrocortical neurons. Time course studies revealed that MeHg-induced upregulation of c-fos, a marker of neural activation, preceded neuronal cell death. These results were similar to those observed in a MeHg-intoxicated mouse model. We observed early expression of the oxidative stress marker thymidine glycol followed by activation of p44/42 mitogen-activated protein kinase (MAPK) and p38 MAPK, and an increase in cAMP response element binding protein (CREB). Investigation of the effects of specific kinase inhibitors revealed that SB203580, a specific inhibitor for p38 MAPK, significantly blocked the upregulation of c-fos and the subsequent neuronal cell death. In contrast, PD98059 and U0126, specific inhibitors for p44/p42 MAPK, showed no effects on MeHg-induced neurotoxicity. Furthermore, the antioxidants Trolox and edaravone significantly suppressed MeHg-induced thymidine glycol expression, p38 MAPK-CREB pathway activation, and neurotoxicity. Altogether, these results suggest that MeHg-induced oxidative stress and subsequent activation of the p38 MAPK-CREB pathway contribute to cerebrocortical neuronal hyperactivity and subsequent neuronal cell death.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease, Kumamoto, Japan.
| | - Fusako Usuki
- Department of Clinical Medicine, National Institute for Minamata Disease, Kumamoto, Japan
| |
Collapse
|
49
|
Colle D, Farina M, Ceccatelli S, Raciti M. Paraquat and Maneb Exposure Alters Rat Neural Stem Cell Proliferation by Inducing Oxidative Stress: New Insights on Pesticide-Induced Neurodevelopmental Toxicity. Neurotox Res 2018; 34:820-833. [PMID: 29859004 DOI: 10.1007/s12640-018-9916-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/25/2018] [Accepted: 05/21/2018] [Indexed: 01/01/2023]
Abstract
Pesticide exposure has been linked to the pathogenesis of neurodevelopmental and neurodegenerative disorders including autism spectrum disorders, attention deficit/hyperactivity, and Parkinson's disease (PD). Developmental exposure to pesticides, even at low concentrations not harmful for the adult brain, can lead to neuronal loss and functional deficits. It has been shown that prenatal or early postnatal exposure to the herbicide paraquat (PQ) and the fungicide maneb (MB), alone or in combination, causes permanent toxicity in the nigrostriatal dopamine system, supporting the idea that early exposure to these pesticides may contribute to the pathophysiology of PD. However, the mechanisms mediating PQ and MB developmental neurotoxicity are not yet understood. Therefore, we investigated the neurotoxic effect of low concentrations of PQ and MB in primary cultures of rat embryonic neural stem cells (NSCs), with particular focus on cell proliferation and oxidative stress. Exposure to PQ alone or in combination with MB (PQ + MB) led to a significant decrease in cell proliferation, while the cell death rate was not affected. Consistently, PQ + MB exposure altered the expression of major genes regulating the cell cycle, namely cyclin D1, cyclin D2, Rb1, and p19. Moreover, PQ and PQ + MB exposures increased the reactive oxygen species (ROS) production that could be neutralized upon N-acetylcysteine (NAC) treatment. Notably, in the presence of NAC, Rb1 expression was normalized and a normal cell proliferation pattern could be restored. These findings suggest that exposure to PQ + MB impairs NSCs proliferation by mechanisms involving alterations in the redox state.
Collapse
Affiliation(s)
- Dirleise Colle
- Department of Clinical Analysis, Federal University of Santa Catarina, Florianópolis, Brazil. .,Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Brazil. .,Departamento de Análises Clínicas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil.
| | - Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Sandra Ceccatelli
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Marilena Raciti
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
50
|
Quispe RL, Canto RFS, Jaramillo ML, Barbosa FAR, Braga AL, de Bem AF, Farina M. Design, Synthesis, and In Vitro Evaluation of a Novel Probucol Derivative: Protective Activity in Neuronal Cells Through GPx Upregulation. Mol Neurobiol 2018; 55:7619-7634. [PMID: 29430618 DOI: 10.1007/s12035-018-0939-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/28/2018] [Indexed: 12/23/2022]
Abstract
Recent studies have shown that probucol (PB), a hipocholesterolemic agent with antioxidant and anti-inflammatory properties, presents neuroprotective properties. On the other hand, adverse effects have limited PB's clinical application. Thus, the search for PB derivatives with no or less adverse effects has been a topic of research. In this study, we present a novel organoselenium PB derivative (RC513) and investigate its potential protective activity in an in vitro experimental model of oxidative toxicity induced by tert-butyl hydroperoxide (tBuOOH) in HT22 neuronal cells, as well as exploit potential protective mechanisms. tBuOOH exposure caused a significant decrease in the cell viability, which was preceded by (i) increased reactive species generation and (ii) decreased mitochondrial maximum oxygen consumption rate. RC513 pretreatment (48 h) significantly prevented the tBuOOH-induced decrease of cell viability, RS generation, and mitochondrial dysfunction. Of note, RC513 significantly increased glutathione peroxidase (GPx) activity and mRNA expression of GPx1, a key enzyme involved in peroxide detoxification. The use of mercaptosuccinic acid, an inhibitor of GPx, significantly decreased the protective activity of RC513 against tBuOOH-induced cytotoxicity in HT22 cells, highlighting the importance of GPx upregulation in the observed protection. In summary, the results showed a significant protective activity of a novel PB derivative against tBuOOH-induced oxidative stress and mitochondrial dysfunction, which was related to the upregulation of GPx. Our results point to RC513 as a promising neuroprotective molecule, even though studies concerning potential beneficial effects and safety aspects of RC513 under in vivo conditions are well warranted.
Collapse
Affiliation(s)
- Ruth Liliám Quispe
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina, Campus Universitário, Florianópolis, SC, Brazil.
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Bloco C, CEP, Florianópolis, Santa Catarina, Brazil.
| | - Rômulo Faria Santos Canto
- Departamento de Farmacociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Michael Lorenz Jaramillo
- Departamento de Biologia Celular, Embriologia e Genética, Universidade Federal de Santa Catarina, Campus Universitário, Florianópolis, SC, Brazil
| | - Flavio Augusto Rocha Barbosa
- Departamento de Química, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Antônio Luiz Braga
- Departamento de Química, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Andreza Fabro de Bem
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Bloco C, CEP, Florianópolis, Santa Catarina, Brazil
| | - Marcelo Farina
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina, Campus Universitário, Florianópolis, SC, Brazil.
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Bloco C, CEP, Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|