1
|
Tolstova T, Dotsenko E, Luzgina N, Rusanov A. Preconditioning of Mesenchymal Stem Cells Enhances the Neuroprotective Effects of Their Conditioned Medium in an Alzheimer's Disease In Vitro Model. Biomedicines 2024; 12:2243. [PMID: 39457556 PMCID: PMC11504366 DOI: 10.3390/biomedicines12102243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) develops as a result of oxidative damage to neurons and chronic inflammation of microglia. These processes can be influenced by the use of a conditioned medium (CM) derived from mesenchymal stem cells (MSCs). The CM contains a wide range of factors that have neurotrophic, antioxidant, and anti-inflammatory effects. In addition, the therapeutic potential of the CM can be further enhanced by pretreating the MSCs to increase their paracrine activity. The current study aimed to investigate the neuroprotective effects of CM derived from MSCs, which were either activated by a TLR3 ligand or exposed to CoCl2, a hypoxia mimetic (pCM or hCM, respectively), in an in vitro model of AD. METHODS We have developed a novel in vitro model of AD that allows us to investigate the neuroprotective and anti-inflammatory effects of MSCs on induced neurodegeneration in the PC12 cell line and the activation of microglia using THP-1 cells. RESULTS This study demonstrates for the first time that pCM and hCM exhibit more pronounced immunosuppressive effects on proinflammatory M1 macrophages compared to CM derived from untreated MSCs (cCM). This may help prevent the development of neuroinflammation by balancing the M1 and M2 microglial phenotypes via the decreased secretion of proinflammatory cytokines (IL-1β, IL-6, and TNF-α) and increased secretion of IL-4, as well as the expression of IL-10 and TGF-β by macrophages. Moreover, a previously unknown increase in the neurotrophic properties of hCM was discovered, which led to an increase in the viability of neuron-like PC12 cells under H2O2-induced oxidative-stress conditions. These results are likely associated with an increase in the production of growth factors, including vascular endothelial growth factor (VEGF). In addition, the neuroprotective effects of CM from preconditioned MSCs are also mediated by the activation of the Nrf2/ARE pathway in PC12 cells. CONCLUSIONS TLR3 activation in MSCs leads to more potent immunosuppressive effects of the CM against pro-inflammatory M1 macrophages, while the use of hCM led to increased neurotrophic effects after H2O2-induced damage to neuronal cells. These results are of interest for the potential treatment of AD with CM from preactivated MSCs.
Collapse
Affiliation(s)
- Tatiana Tolstova
- Institute of Biomedical Chemistry, Pogodinskaya 10, 119121 Moscow, Russia
| | | | | | - Alexander Rusanov
- Institute of Biomedical Chemistry, Pogodinskaya 10, 119121 Moscow, Russia
| |
Collapse
|
2
|
Wei R, Wei P, Yuan H, Yi X, Aschner M, Jiang YM, Li SJ. Inflammation in Metal-Induced Neurological Disorders and Neurodegenerative Diseases. Biol Trace Elem Res 2024; 202:4459-4481. [PMID: 38206494 DOI: 10.1007/s12011-023-04041-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024]
Abstract
Essential metals play critical roles in maintaining human health as they participate in various physiological activities. Nonetheless, both excessive accumulation and deficiency of these metals may result in neurotoxicity secondary to neuroinflammation and the activation of microglia and astrocytes. Activation of these cells can promote the release of pro-inflammatory cytokines. It is well known that neuroinflammation plays a critical role in metal-induced neurotoxicity as well as the development of neurological disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Initially seen as a defense mechanism, persistent inflammatory responses are now considered harmful. Astrocytes and microglia are key regulators of neuroinflammation in the central nervous system, and their excessive activation may induce sustained neuroinflammation. Therefore, in this review, we aim to emphasize the important role and molecular mechanisms underlying metal-induced neurotoxicity. Our objective is to raise the awareness on metal-induced neuroinflammation in neurological disorders. However, it is not only just neuroinflammation that different metals could induce; they can also cause harm to the nervous system through oxidative stress, apoptosis, and autophagy, to name a few. The primary pathophysiological mechanism by which these metals induce neurological disorders remains to be determined. In addition, given the various pathways through which individuals are exposed to metals, it is necessary to also consider the effects of co-exposure to multiple metals on neurological disorders.
Collapse
Affiliation(s)
- Ruokun Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Peiqi Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Haiyan Yuan
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Xiang Yi
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Michael Aschner
- The Department of Molecular Pharmacology at Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yue-Ming Jiang
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| | - Shao-Jun Li
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| |
Collapse
|
3
|
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
|
4
|
Sun Y, Wu D, Yang X, Tang B, Xia C, Luo C, Gong Q, Lui S, Hu N. The associations of peripheral interleukin alterations and hippocampal subfield volume deficits in schizophrenia. Cereb Cortex 2024; 34:bhae308. [PMID: 39077921 DOI: 10.1093/cercor/bhae308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/03/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024] Open
Abstract
The hippocampus is one of the brain regions most vulnerable to inflammatory insults, and the relationships between peripheral inflammation and hippocampal subfields in patients with schizophrenia remain unclear. In this study, forty-six stably medicated patients with schizophrenia and 48 demographically matched healthy controls (HCs) were recruited. The serum levels of IL - 1β, IL-6, IL-10, and IL-12p70 were measured, and 3D high-resolution T1-weighted magnetic resonance imaging was performed. The IL levels and hippocampal subfield volumes were both compared between patients and HCs. The associations of altered IL levels with hippocampal subfield volumes were assessed in patients. Patients with schizophrenia demonstrated higher serum levels of IL-6 and IL-10 but lower levels of IL-12p70 than HCs. In patients, the levels of IL-6 were positively correlated with the volumes of the left granule cell layer of the dentate gyrus (GCL) and cornu Ammonis (CA) 4, while the levels of IL-10 were negatively correlated with the volumes of those subfields. IL-6 and IL-10 might have antagonistic roles in atrophy of the left GCL and CA4. This suggests a complexity of peripheral cytokine dysregulation and the potential for its selective effects on hippocampal substructures, which might be related to the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Yuan Sun
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Dongsheng Wu
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 18, Section 3, South Renmin Road, Chengdu 610041, China
| | - Xiyue Yang
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Biqiu Tang
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Chao Xia
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Chunyan Luo
- Department of Radiology, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Su Lui
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Na Hu
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Department of Radiology, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| |
Collapse
|
5
|
Leal-Nazaré CG, Arrifano GP, Lopes-Araújo A, Santos-Sacramento L, Barthelemy JL, Soares-Silva I, Crespo-Lopez ME, Augusto-Oliveira M. Methylmercury neurotoxicity: Beyond the neurocentric view. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 920:170939. [PMID: 38365040 DOI: 10.1016/j.scitotenv.2024.170939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/09/2024] [Accepted: 02/10/2024] [Indexed: 02/18/2024]
Abstract
Mercury is a highly toxic metal widely used in human activities worldwide, therefore considered a global public health problem. Many cases of mercury intoxication have occurred in history and represent a huge challenge nowadays. Of particular importance is its methylated form, methylmercury (MeHg). This mercurial species induces damage to several organs in the human body, especially to the central nervous system. Neurological impairments such as executive, memory, motor and visual deficits are associated with MeHg neurotoxicity. Molecular mechanisms involved in MeHg-induced neurotoxicity include excitotoxicity due to glutamatergic imbalance, disturbance in calcium homeostasis and oxidative balance, failure in synaptic support, and inflammatory response. Although neurons are largely affected by MeHg intoxication, they only represent half of the brain cells. Glial cells represent roughly 50 % of the brain cells and are key elements in the functioning of the central nervous system. Particularly, astrocytes and microglia are deeply involved in MeHg-induced neurotoxicity, resulting in distinct neurological outcomes depending on the context. In this review, we discuss the main findings on astroglial and microglial involvement as mediators of neuroprotective and neurotoxic responses to MeHg intoxication. The literature shows that these responses depend on chemical and morphophysiological features, thus, we present some insights for future investigations, considering the particularities of the context, including time and dose of exposure, brain region, and species of study.
Collapse
Affiliation(s)
- Caio Gustavo Leal-Nazaré
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Gabriela P Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Amanda Lopes-Araújo
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Leticia Santos-Sacramento
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Jean Ludger Barthelemy
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Isabela Soares-Silva
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil.
| | - Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil.
| |
Collapse
|
6
|
Nunes C, Proença S, Ambrosini G, Pamies D, Thomas A, Kramer NI, Zurich MG. Integrating distribution kinetics and toxicodynamics to assess repeat dose neurotoxicity in vitro using human BrainSpheres: a case study on amiodarone. Front Pharmacol 2023; 14:1248882. [PMID: 37745076 PMCID: PMC10512064 DOI: 10.3389/fphar.2023.1248882] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 07/24/2023] [Indexed: 09/26/2023] Open
Abstract
For ethical, economical, and scientific reasons, animal experimentation, used to evaluate the potential neurotoxicity of chemicals before their release in the market, needs to be replaced by new approach methodologies. To illustrate the use of new approach methodologies, the human induced pluripotent stem cell-derived 3D model BrainSpheres was acutely (48 h) or repeatedly (7 days) exposed to amiodarone (0.625-15 µM), a lipophilic antiarrhythmic drug reported to have deleterious effects on the nervous system. Neurotoxicity was assessed using transcriptomics, the immunohistochemistry of cell type-specific markers, and real-time reverse transcription-polymerase chain reaction for various genes involved in the lipid metabolism. By integrating distribution kinetics modeling with neurotoxicity readouts, we show that the observed time- and concentration-dependent increase in the neurotoxic effects of amiodarone is driven by the cellular accumulation of amiodarone after repeated dosing. The development of a compartmental in vitro distribution kinetics model allowed us to predict the change in cell-associated concentrations in BrainSpheres with time and for different exposure scenarios. The results suggest that human cells are intrinsically more sensitive to amiodarone than rodent cells. Amiodarone-induced regulation of lipid metabolism genes was observed in brain cells for the first time. Astrocytes appeared to be the most sensitive human brain cell type in vitro. In conclusion, assessing readouts at different molecular levels after the repeat dosing of human induced pluripotent stem cell-derived BrainSpheres in combination with the compartmental modeling of in vitro kinetics provides a mechanistic means to assess neurotoxicity pathways and refine chemical safety assessment for humans.
Collapse
Affiliation(s)
- Carolina Nunes
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland
| | - Susana Proença
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
- Toxicology Division, Wageningen University, Wageningen, Netherlands
| | - Giovanna Ambrosini
- Bioinformatics Competence Center, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Bioinformatics Competence Center, University of Lausanne, Lausanne, Switzerland
| | - David Pamies
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland
| | - Aurélien Thomas
- Unit of Forensic Toxicology and Chemistry, CURML, Lausanne and Geneva University Hospitals, Geneva, Switzerland
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Nynke I. Kramer
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
- Toxicology Division, Wageningen University, Wageningen, Netherlands
| | - Marie-Gabrielle Zurich
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland
| |
Collapse
|
7
|
de Paula Arrifano G, Crespo-Lopez ME, Lopes-Araújo A, Santos-Sacramento L, Barthelemy JL, de Nazaré CGL, Freitas LGR, Augusto-Oliveira M. Neurotoxicity and the Global Worst Pollutants: Astroglial Involvement in Arsenic, Lead, and Mercury Intoxication. Neurochem Res 2023; 48:1047-1065. [PMID: 35997862 DOI: 10.1007/s11064-022-03725-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/01/2022] [Accepted: 08/09/2022] [Indexed: 10/15/2022]
Abstract
Environmental pollution is a global threat and represents a strong risk factor for human health. It is estimated that pollution causes about 9 million premature deaths every year. Pollutants that can cross the blood-brain barrier and reach the central nervous system are of special concern, because of their potential to cause neurological and development disorders. Arsenic, lead and mercury are usually ranked as the top three in priority lists of regulatory agencies. Against xenobiotics, astrocytes are recognised as the first line of defence in the CNS, being involved in virtually all brain functions, contributing to homeostasis maintenance. Here, we discuss the current knowledge on the astroglial involvement in the neurotoxicity induced by these pollutants. Beginning by the main toxicokinetic characteristics, this review also highlights the several astrocytic mechanisms affected by these pollutants, involving redox system, neurotransmitter and glucose metabolism, and cytokine production/release, among others. Understanding how these alterations lead to neurological disturbances (including impaired memory, deficits in executive functions, and motor and visual disfunctions), by revisiting the current knowledge is essential for future research and development of therapies and prevention strategies.
Collapse
Affiliation(s)
- Gabriela de Paula Arrifano
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Av. Augusto Corrêa, 01, Belém, PA, 66075-110, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Av. Augusto Corrêa, 01, Belém, PA, 66075-110, Brazil
| | - Amanda Lopes-Araújo
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Av. Augusto Corrêa, 01, Belém, PA, 66075-110, Brazil
| | - Letícia Santos-Sacramento
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Av. Augusto Corrêa, 01, Belém, PA, 66075-110, Brazil
| | - Jean L Barthelemy
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Av. Augusto Corrêa, 01, Belém, PA, 66075-110, Brazil
| | - Caio Gustavo Leal de Nazaré
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Av. Augusto Corrêa, 01, Belém, PA, 66075-110, Brazil
| | - Luiz Gustavo R Freitas
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Av. Augusto Corrêa, 01, Belém, PA, 66075-110, Brazil
| | - Marcus Augusto-Oliveira
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Av. Augusto Corrêa, 01, Belém, PA, 66075-110, Brazil.
| |
Collapse
|
8
|
Lenski M, Bruno C, Darrouzain F, Allorge D. Métabolomique : principes et applications en toxicologie biologique et médicolégale. TOXICOLOGIE ANALYTIQUE ET CLINIQUE 2023. [DOI: 10.1016/j.toxac.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
9
|
Shinoda Y, Akiyama M, Toyama T. Potential Association between Methylmercury Neurotoxicity and Inflammation. Biol Pharm Bull 2023; 46:1162-1168. [PMID: 37661394 DOI: 10.1248/bpb.b23-00075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Methylmercury (MeHg) is the causal substrate of Minamata disease and a major environmental toxicant. MeHg is widely distributed, mainly in the ocean, meaning its bioaccumulation in seafood is a considerable problem for human health. MeHg has been intensively investigated and is known to induce inflammatory responses and neurodegeneration. However, the relationship between MeHg-induced inflammatory responses and neurodegeneration is not understood. In the present review, we first describe recent findings showing an association between inflammatory responses and certain MeHg-unrelated neurological diseases caused by neurodegeneration. In addition, cell-specific MeHg-induced inflammatory responses are summarized for the central nervous system including those of microglia, astrocytes, and neurons. We also describe MeHg-induced inflammatory responses in peripheral cells and tissue, such as macrophages and blood. These findings provide a concept of the relationship between MeHg-induced inflammatory responses and neurodegeneration, as well as direction for future research of MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Yo Shinoda
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Masahiro Akiyama
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University
| | - Takashi Toyama
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University
| |
Collapse
|
10
|
Effect of Heat Stress on Hippocampal Neurogenesis: Insights into the Cellular and Molecular Basis of Neuroinflammation-Induced Deficits. Cell Mol Neurobiol 2023; 43:1-13. [PMID: 34767143 DOI: 10.1007/s10571-021-01165-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/01/2021] [Indexed: 01/07/2023]
Abstract
Heat stress is known to result in neuroinflammation, neuronal damage, and disabilities in learning and memory in animals and humans. It has previously been reported that cognitive impairment caused by neuroinflammation may at least in part be mediated by defective hippocampal neurogenesis, and defective neurogenesis has been linked to aberrantly activated microglial cells. Moreover, the release of cytokines within the brain has been shown to contribute to the disruption of cognitive functions in several conditions following neuroinflammation. In this review, we summarize evolving evidence for the current understanding of inflammation-induced deficits in hippocampal neurogenesis, and the resulting behavioral impairments after heat stress. Furthermore, we provide valuable insights into the molecular and cellular mechanisms underlying neuroinflammation-induced deficits in hippocampal neurogenesis, particularly relating to cognitive dysfunction following heat stress. Lastly, we aim to identify potential mechanisms through which neuroinflammation induces cognitive dysfunction, and elucidate how neuroinflammation contributes to defective hippocampal neurogenesis. This review may therefore help to better understand the relationship between hippocampal neurogenesis and heat stress.
Collapse
|
11
|
Martins B, Novo JP, Fonseca É, Raposo R, Sardão VA, Pereira F, Oriá RB, Fontes-Ribeiro C, Malva J. Necrotic-like BV-2 microglial cell death due to methylmercury exposure. Front Pharmacol 2022; 13:1003663. [PMID: 36408241 PMCID: PMC9667718 DOI: 10.3389/fphar.2022.1003663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/18/2022] [Indexed: 12/01/2022] Open
Abstract
Methylmercury (MeHg) is a dangerous environmental contaminant with strong bioaccumulation in the food chain and neurotoxic properties. In the nervous system, MeHg may cause neurodevelopment impairment and potentially interfere with immune response, compromising proper control of neuroinflammation and aggravating neurodegeneration. Human populations are exposed to environmental contamination with MeHg, especially in areas with strong mining or industrial activity, raising public health concerns. Taking this into consideration, this work aims to clarify pathways leading to acute toxic effects caused by MeHg exposure in microglial cells. BV-2 mouse microglial cells were incubated with MeHg at different concentrations (0.01, 0.1, 1 and 10 µM) for 1 h prior to continuous Lipopolysaccharide (LPS, 0.5 μg/ml) exposure for 6 or 24 h. After cell exposure, reactive oxygen species (ROS), IL-6 and TNF-α cytokines production, inducible nitric oxide synthase (iNOS) expression, nitric oxide (NO) release, metabolic activity, propidium iodide (PI) uptake, caspase-3 and -9 activities and phagocytic activity were assessed. MeHg 10 µM decreased ROS formation, the production and secretion of pro-inflammatory cytokines IL-6, TNF-α, iNOS immunoreactivity, the release of NO in BV-2 cells. Furthermore, MeHg 10 µM decreased the metabolic activity of BV-2 and increased the number of PI-positive cells (necrotic-like cell death) when compared to the respective control group. Besides, MeHg did not interfere with caspase activity or the phagocytic profile of cells. The short-term effects of a high concentration of MeHg on BV-2 microglial cells lead to impaired production of several pro-inflammatory mediators, as well as a higher microglial cell death via necrosis, compromising their neuroinflammatory response. Clarifying the mechanisms underlying MeHg-induced neurotoxicity and neurodegeneration in brain cells is relevant to better understand acute and long-term chronic neuroinflammatory responses following MeHg exposure.
Collapse
Affiliation(s)
- B. Martins
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - J. P. Novo
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - É. Fonseca
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - R. Raposo
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal,Experimental Biology Core, Health Sciences Center, University of Fortaleza, Fortaleza, Brazil
| | - V. A. Sardão
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal,Center for Neuroscience and Cell Biology (CNC), UC Biotech, University of Coimbra, Cantanhede, Portugal
| | - F. Pereira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - R. B. Oriá
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - C. Fontes-Ribeiro
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - J. Malva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal,*Correspondence: J. Malva,
| |
Collapse
|
12
|
Szpakowski P, Ksiazek-Winiarek D, Turniak-Kusy M, Pacan I, Glabinski A. Human Primary Astrocytes Differently Respond to Pro- and Anti-Inflammatory Stimuli. Biomedicines 2022; 10:biomedicines10081769. [PMID: 35892669 PMCID: PMC9331936 DOI: 10.3390/biomedicines10081769] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/03/2022] [Accepted: 07/18/2022] [Indexed: 12/15/2022] Open
Abstract
For a long time, astrocytes were considered a passive brain cell population. However, recently, many studies have shown that their role in the central nervous system (CNS) is more active. Previously, it was stated that there are two main functional phenotypes of astrocytes. However, nowadays, it is clear that there is rather a broad spectrum of these phenotypes. The major goal of this study was to evaluate the production of some inflammatory chemokines and neurotrophic factors by primary human astrocytes after pro- or anti-inflammatory stimulation. We observed that only astrocytes induced by inflammatory mediators TNFα/IL-1a/C1q produced CXCL10, CCL1, and CXCL13 chemokines. Unstimulated astrocytes and those cultured with anti-inflammatory cytokines (IL-4, IL-10, or TGF-β1) did not produce these chemokines. Interestingly, astrocytes cultured in proinflammatory conditions significantly decreased the release of neurotrophic factor PDGF-A, as compared to unstimulated astrocytes. However, in response to anti-inflammatory cytokine TGF-β1, astrocytes significantly increased PDGF-A production compared to the medium alone. The production of another studied neurotrophic factor BDNF was not influenced by pro- or anti-inflammatory stimulation. The secretory response was accompanied by changes in HLA-DR, CD83, and GFAP expression. Our study confirms that astrocytes differentially respond to pro- and anti-inflammatory stimuli, especially to inflammatory cytokines TNF-α, IL-1a, and C1q, suggesting their role in leukocyte recruitment.
Collapse
|
13
|
Olude MA, Mouihate A, Mustapha OA, Farina C, Quintana FJ, Olopade JO. Astrocytes and Microglia in Stress-Induced Neuroinflammation: The African Perspective. Front Immunol 2022; 13:795089. [PMID: 35707531 PMCID: PMC9190229 DOI: 10.3389/fimmu.2022.795089] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Africa is laden with a youthful population, vast mineral resources and rich fauna. However, decades of unfortunate historical, sociocultural and leadership challenges make the continent a hotspot for poverty, indoor and outdoor pollutants with attendant stress factors such as violence, malnutrition, infectious outbreaks and psychological perturbations. The burden of these stressors initiate neuroinflammatory responses but the pattern and mechanisms of glial activation in these scenarios are yet to be properly elucidated. Africa is therefore most vulnerable to neurological stressors when placed against a backdrop of demographics that favor explosive childbearing, a vast population of unemployed youths making up a projected 42% of global youth population by 2030, repressive sociocultural policies towards women, poor access to healthcare, malnutrition, rapid urbanization, climate change and pollution. Early life stress, whether physical or psychological, induces neuroinflammatory response in developing nervous system and consequently leads to the emergence of mental health problems during adulthood. Brain inflammatory response is driven largely by inflammatory mediators released by glial cells; namely astrocytes and microglia. These inflammatory mediators alter the developmental trajectory of fetal and neonatal brain and results in long-lasting maladaptive behaviors and cognitive deficits. This review seeks to highlight the patterns and mechanisms of stressors such as poverty, developmental stress, environmental pollutions as well as malnutrition stress on astrocytes and microglia in neuroinflammation within the African context.
Collapse
Affiliation(s)
- Matthew Ayokunle Olude
- Vertebrate Morphology, Environmental Toxicology and Neuroscience Unit, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
- *Correspondence: Matthew Ayokunle Olude,
| | - Abdeslam Mouihate
- Department of Physiology, Faculty of Medicine, Health Sciences Centre, Kuwait University, Kuwait City, Kuwait
| | - Oluwaseun Ahmed Mustapha
- Vertebrate Morphology, Environmental Toxicology and Neuroscience Unit, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
| | - Cinthia Farina
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) San Raffaele Scientific Institute, Institute of Experimental Neurology (INSPE) and Division of Neuroscience, Milan, Italy
| | - Francisco Javier Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - James Olukayode Olopade
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
14
|
Salkov VN, Voronkov DN, Khudoerkov RM. [The role of mercury and arsenic in the etiology and pathogenesis of Parkinson's and Alzheimer's diseases]. Arkh Patol 2022; 84:59-64. [PMID: 36178224 DOI: 10.17116/patol20228405159] [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: 06/16/2023]
Abstract
A critical review of literature data on the toxic effects of mercury and arsenic on the human brain and their relationship with the etiology and pathogenesis of neurodegenerative diseases such as Parkinson's and Alzheimer's diseases is presented. In the first case, the toxic effect of mercury and arsenic on the brain stimulates oxidative stress, which leads to the formation of free oxygen species and a decrease in the antioxidant defense of neurons. In the second case, the harmful effect of mercury changes the structure and properties of β-amyloid, and the toxic effect of arsenic contributes to its accumulation. In the pathogenesis of the diseases under consideration, particular importance is attached to the reaction of astrocytes that initiate neuroinflammation, which is also characteristic of mercury and arsenic intoxication. Considering that the symptoms recorded during intoxication with mercury and arsenic are in many respects similar to those of Parkinson's and Alzheimer's diseases, and their pathogenetic mechanisms (oxidative stress and neuroinflammation) coincide, then the toxic effects of mercury and arsenic in neurodegenerative diseases analyzed in this review can be characterized as the influence of the most significant risk factors.
Collapse
Affiliation(s)
- V N Salkov
- Research Center of Neurology, Brain Institute, Moscow, Russia
| | - D N Voronkov
- Research Center of Neurology, Brain Institute, Moscow, Russia
| | - R M Khudoerkov
- Research Center of Neurology, Brain Institute, Moscow, Russia
| |
Collapse
|
15
|
Revisiting Astrocytic Roles in Methylmercury Intoxication. Mol Neurobiol 2021; 58:4293-4308. [PMID: 33990914 DOI: 10.1007/s12035-021-02420-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
Intoxication by heavy metals such as methylmercury (MeHg) is recognized as a global health problem, with strong implications in central nervous system pathologies. Most of these neuropathological conditions involve vascular, neurotransmitter recycling, and oxidative balance disruption leading to accelerated decline in fine balance, and learning, memory, and visual processes as main outcomes. Besides neurons, astrocytes are involved in virtually all the brain processes and perform important roles in neurological response following injuries. Due to astrocytes' strategic functions in brain homeostasis, these cells became the subject of several studies on MeHg intoxication. The most heterogenous glial cells, astrocytes, are composed of plenty of receptors and transporters to dialogue with neurons and other cells and to monitor extracellular environment responding tightly through fluctuation of cytosolic ions. The overall toxicity of MeHg might be determined on the basis of the balance between MeHg-mediated injury to neurons and protective responses from astrocytes. Although the role of neurons in MeHg intoxication is relatively well-established, the role of the astrocytes is only beginning to be understood. In this review, we update the information on astroglial modulation of the MeHg-induced neurotoxicity, providing remarks on their protective and deleterious roles and insights for future studies.
Collapse
|
16
|
Novo JP, Martins B, Raposo RS, Pereira FC, Oriá RB, Malva JO, Fontes-Ribeiro C. Cellular and Molecular Mechanisms Mediating Methylmercury Neurotoxicity and Neuroinflammation. Int J Mol Sci 2021; 22:ijms22063101. [PMID: 33803585 PMCID: PMC8003103 DOI: 10.3390/ijms22063101] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/06/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Methylmercury (MeHg) toxicity is a major environmental concern. In the aquatic reservoir, MeHg bioaccumulates along the food chain until it is consumed by riverine populations. There has been much interest in the neurotoxicity of MeHg due to recent environmental disasters. Studies have also addressed the implications of long-term MeHg exposure for humans. The central nervous system is particularly susceptible to the deleterious effects of MeHg, as evidenced by clinical symptoms and histopathological changes in poisoned humans. In vitro and in vivo studies have been crucial in deciphering the molecular mechanisms underlying MeHg-induced neurotoxicity. A collection of cellular and molecular alterations including cytokine release, oxidative stress, mitochondrial dysfunction, Ca2+ and glutamate dyshomeostasis, and cell death mechanisms are important consequences of brain cells exposure to MeHg. The purpose of this review is to organize an overview of the mercury cycle and MeHg poisoning events and to summarize data from cellular, animal, and human studies focusing on MeHg effects in neurons and glial cells. This review proposes an up-to-date compendium that will serve as a starting point for further studies and a consultation reference of published studies.
Collapse
Affiliation(s)
- João P. Novo
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
| | - Beatriz Martins
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
| | - Ramon S. Raposo
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
- Experimental Biology Core, University of Fortaleza, Health Sciences, Fortaleza 60110-001, Brazil
| | - Frederico C. Pereira
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
| | - Reinaldo B. Oriá
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil;
| | - João O. Malva
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
- Correspondence: (J.O.M.); (C.F.-R.)
| | - Carlos Fontes-Ribeiro
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
- Correspondence: (J.O.M.); (C.F.-R.)
| |
Collapse
|
17
|
Branco V, Aschner M, Carvalho C. Neurotoxicity of mercury: an old issue with contemporary significance. ADVANCES IN NEUROTOXICOLOGY 2021; 5:239-262. [PMID: 34263092 PMCID: PMC8276940 DOI: 10.1016/bs.ant.2021.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Mercury exerts a variety of toxic effects, depending on the specific compound and route of exposure. However, neurotoxicity in virtue of its consequence to health causes the greatest concern for toxicologists. This is particularly true regarding fetal development, where neurotoxic effects are much more severe than in adults, and the toxicity threshold is lower. Here, we review the major concepts regarding the neurotoxicity of mercury compounds (mercury vapor; methylmercury and ethylmercury), from exposure routes to toxicokinetic particularities leading to brain deposition and the development of neurotoxic effects. Albeit research on the neurotoxicity of mercury compounds has significantly advanced from the second half of the twentieth century onwards, several grey areas regarding the mechanism of toxicity still exist. Thus, we emphasize research advances during the last two decades concerning the molecular interactions of mercury which cause neurotoxic effects. Highlights include the disruption of glutamate signaling and excitotoxicity resulting from exposure to mercury and the interaction with redox active residues such as cysteines and selenocysteines which are the premise accounting for the disruption of redox homeostasis caused by mercurials. We also address how immunotoxic effects at the CNS, namely microglia and astrocyte activation modulate developmental neurotoxicity, a major topic in contemporary research.
Collapse
Affiliation(s)
- Vasco Branco
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, USA
| | - Cristina Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
18
|
Kabir MT, Uddin MS, Zaman S, Begum Y, Ashraf GM, Bin-Jumah MN, Bungau SG, Mousa SA, Abdel-Daim MM. Molecular Mechanisms of Metal Toxicity in the Pathogenesis of Alzheimer’s Disease. Mol Neurobiol 2020; 58:1-20. [DOI: 10.1007/s12035-020-02096-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/25/2020] [Indexed: 12/24/2022]
|
19
|
The role of neuroglia in autism spectrum disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 173:301-330. [PMID: 32711814 DOI: 10.1016/bs.pmbts.2020.04.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neuroglia are a large class of neural cells of ectodermal (astroglia, oligodendroglia, and peripheral glial cells) and mesodermal (microglia) origin. Neuroglial cells provide homeostatic support, protection, and defense to the nervous tissue. Pathological potential of neuroglia has been acknowledged since their discovery. Research of the recent decade has shown the key role of all classes of glial cells in autism spectrum disorders (ASD), although molecular mechanisms defining glial contribution to ASD are yet to be fully characterized. This narrative conceptualizes recent findings of the broader roles of glial cells, including their active participation in the control of cerebral environment and regulation of synaptic development and scaling, highlighting their putative involvement in the etiopathogenesis of ASD.
Collapse
|
20
|
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: 179] [Impact Index Per Article: 44.8] [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
|
21
|
Muniroh M, Gumay AR, Indraswari DA, Bahtiar Y, Hardian H, Bakri S, Maharani N, Karlowee V, Koriyama C, Yamamoto M. Activation of MIP-2 and MCP-5 Expression in Methylmercury-Exposed Mice and Their Suppression by N-Acetyl-L-Cysteine. Neurotox Res 2020; 37:827-834. [PMID: 32040762 DOI: 10.1007/s12640-020-00174-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 01/15/2020] [Accepted: 02/02/2020] [Indexed: 01/11/2023]
Abstract
Methylmercury (MeHg) is a well-known neurotoxin of the central nervous system (CNS). Neuroinflammation is one of the main pathways of MeHg-induced CNS impairment. This study aims to investigate the expressions of IL-6, MIP-2, and MCP-5, as biomarkers in relation with MeHg-induced CNS impairment and N-acetyl-L-cysteine (NAC) treatment in mice, as well as histopathological changes of brain tissue and clinical symptom such as ataxia. Twenty male Balb/c mice, aged 8-9 weeks, were divided into 4 groups and treated with saline (control), NAC [150 mg/kg body weight (BW) day], MeHg (4 mg Hg/kg BW), or a combination of MeHg and NAC for 17 days. MeHg induced the expression of IL-6, MIP-2, and MCP-5 in the serum, with median values (those in controls) of 55.06 (9.44), 15.94 (9.30), and 458.91 (239.91) mg/dl, respectively, and a statistical significance was observed only in IL-6 expression (p < 0.05). MIP-2 and MCP-5 expressions tended to increase in the cerebrum of MeHg-treated group compared with controls; however, the difference was not statistically significant. MeHg treatment also increased IL-6 expression in the cerebellum (7.73 and 4.81 mg/dl in MeHg-treated group and controls, respectively), with a marginal significance. NAC significantly suppressed MeHg-induced IL-6 and MIP-2 expressions in the serum (p < 0.05 for both), and slightly reduced MCP-5 expression in the cerebrum. Ataxia was observed in all MeHg-treated mice after 9-day exposure as well as the decrease of intact Purkinje cells in brain tissue (p < 0.05). These findings suggest that MeHg induced neurotoxicity by elevating the expression of IL-6, MIP-2, and MCP-5 and causing ataxia symptoms, and NAC reduced MeHg-mediated effects on the CNS.
Collapse
Affiliation(s)
- Muflihatul Muniroh
- Department of Physiology, Faculty of Medicine Diponegoro University, Tembalang Semarang, 50275, Indonesia.
| | - Ainun Rahmasari Gumay
- Department of Physiology, Faculty of Medicine Diponegoro University, Tembalang Semarang, 50275, Indonesia
| | - Darmawati Ayu Indraswari
- Department of Physiology, Faculty of Medicine Diponegoro University, Tembalang Semarang, 50275, Indonesia
| | - Yuriz Bahtiar
- Department of Physiology, Faculty of Medicine Diponegoro University, Tembalang Semarang, 50275, Indonesia
| | - Hardian Hardian
- Department of Physiology, Faculty of Medicine Diponegoro University, Tembalang Semarang, 50275, Indonesia
| | - Saekhol Bakri
- Department of Public Health, Faculty of Medicine Diponegoro University, Semarang, 50275, Indonesia
| | - Nani Maharani
- Department of Pharmacology and Therapy, Faculty of Medicine Diponegoro University, Semarang, 50275, Indonesia
| | - Vega Karlowee
- Department of Anatomical Pathology, Faculty of Medicine Diponegoro University, Semarang, 50275, Indonesia
| | - Chihaya Koriyama
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Health Sciences, Kagoshima, 890-8520, Japan
| | - Megumi Yamamoto
- Department of Environment and Public Health, National Institute for Minamata Disease, Kumamoto, 867-0008, Japan
| |
Collapse
|
22
|
The Nuclear Protein HOXB13 Enhances Methylmercury Toxicity by Inducing Oncostatin M and Promoting Its Binding to TNFR3 in Cultured Cells. Cells 2019; 9:cells9010045. [PMID: 31878059 PMCID: PMC7017003 DOI: 10.3390/cells9010045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 02/06/2023] Open
Abstract
Homeobox protein B13 (HOXB13), a transcription factor, is related to methylmercury toxicity; however, the downstream factors involved in enhancing methylmercury toxicity remain unknown. We performed microarray analysis to search for downstream factors whose expression is induced by methylmercury via HOXB13 in human embryonic kidney cells (HEK293), which are useful model cells for analyzing molecular mechanisms. Methylmercury induced the expression of oncostatin M (OSM), a cytokine of the interleukin-6 family, and this was markedly suppressed by HOXB13 knockdown. OSM knockdown also conferred resistance to methylmercury in HEK293 cells, and no added methylmercury resistance was observed when both HOXB13 and OSM were knocked down. Binding of HOXB13 to the OSM gene promoter was increased by methylmercury, indicating the involvement of HOXB13 in the enhancement of its toxicity. Because addition of recombinant OSM to the medium enhanced methylmercury toxicity in OSM-knockdown cells, extracellularly released OSM was believed to enhance methylmercury toxicity via membrane receptors. We discovered tumor necrosis factor receptor (TNF) receptor 3 (TNFR3) to be a potential candidate involved in the enhancement of methylmercury toxicity by OSM. This toxicity mechanism was also confirmed in mouse neuronal stem cells. We report, for the first time, that HOXB13 is involved in enhancement of methylmercury toxicity via OSM-expression induction and that the synthesized OSM causes cell death by binding to TNFR3 extracellularly.
Collapse
|
23
|
Ishihara Y, Itoh K, Oguro A, Chiba Y, Ueno M, Tsuji M, Vogel CFA, Yamazaki T. Neuroprotective activation of astrocytes by methylmercury exposure in the inferior colliculus. Sci Rep 2019; 9:13899. [PMID: 31554907 PMCID: PMC6761145 DOI: 10.1038/s41598-019-50377-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/11/2019] [Indexed: 11/17/2022] Open
Abstract
Methylmercury (MeHg) is well known to induce auditory disorders such as dysarthria. When we performed a global analysis on the brains of mice exposed to MeHg by magnetic resonance imaging, an increase in the T1 signal in the inferior colliculus (IC), which is localized in the auditory pathway, was observed. Therefore, the purpose of this study is to examine the pathophysiology and auditory dysfunction induced by MeHg, focusing on the IC. Measurement of the auditory brainstem response revealed increases in latency and decreases in threshold in the IC of mice exposed to MeHg for 4 weeks compared with vehicle mice. Incoordination in MeHg-exposed mice was noted after 6 weeks of exposure, indicating that IC dysfunction occurs earlier than incoordination. There was no change in the number of neurons or microglial activity, while the expression of glial fibrillary acidic protein, a marker for astrocytic activity, was elevated in the IC of MeHg-exposed mice after 4 weeks of exposure, indicating that astrogliosis occurs in the IC. Suppression of astrogliosis by treatment with fluorocitrate exacerbated the latency and threshold in the IC evaluated by the auditory brainstem response. Therefore, astrocytes in the IC are considered to play a protective role in the auditory pathway. Astrocytes exposed to MeHg increased the expression of brain-derived neurotrophic factor in the IC, suggesting that astrocytic brain-derived neurotrophic factor is a potent protectant in the IC. This study showed that astrogliosis in the IC could be an adaptive response to MeHg toxicity. The overall toxicity of MeHg might be determined on the basis of the balance between MeHg-mediated injury to neurons and protective responses from astrocytes.
Collapse
Affiliation(s)
- Yasuhiro Ishihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8521, Japan. .,Center for Health and the Environment, University of California, Davis, CA, 95616, USA.
| | - Kouichi Itoh
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa, 769-2193, Japan
| | - Ami Oguro
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8521, Japan
| | - Yoichi Chiba
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa, 761-0793, Japan
| | - Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa, 761-0793, Japan
| | - Mayumi Tsuji
- Department of Environmental Health, University of Occupational and Environmental Health, Fukuoka, 807-8555, Japan
| | - Christoph F A Vogel
- Center for Health and the Environment, University of California, Davis, CA, 95616, USA.,Department of Environmental Toxicology, University of California, Davis, CA, 95616, USA
| | - Takeshi Yamazaki
- Program of Life and Environmental Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8521, Japan
| |
Collapse
|
24
|
Oriolo G, Blanco-Hinojo L, Navines R, Mariño Z, Martín-Hernández D, Cavero M, Gimenez D, Caso J, Capuron L, Forns X, Pujol J, Sola R, Martin-Santos R. Association of chronic inflammation and perceived stress with abnormal functional connectivity in brain areas involved with interoception in hepatitis C patients. Brain Behav Immun 2019; 80:204-218. [PMID: 30872094 DOI: 10.1016/j.bbi.2019.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/06/2019] [Accepted: 03/09/2019] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Sickness behavioral changes elicited by inflammation may become prolonged and dysfunctional in patients with chronic disease, such as chronic hepatitis C (CHC). Neuroimaging studies show that the basal ganglia and insula are sensitive to systemic inflammation. AIM To elucidate the clinical and neurobiological aspects of prolonged illnesses in patients with CHC. METHODS Thirty-five CHC patients not treated with interferon-α or other antiviral therapy, and 30 control subjects matched for age and sex, were evaluated for perceived stress (perceived stress scale; PSS), depression (PHQ-9), fatigue and irritability through a visual analog scale (VAS), as well as serum levels of interleukin-6 (IL-6), prostaglandin E2 (PGE2) and oxidative stress markers. Functional MRI was performed, measuring resting-state functional connectivity using a region-of-interest (seed)-based approach focusing on the bilateral insula, subgenual anterior cingulate cortex and bilateral putamen. Between-group differences in functional connectivity patterns were assessed with two-sample t-tests, while the associations between symptoms, inflammatory markers and functional connectivity patterns were analyzed with multiple regression analyses. RESULTS CHC patients had higher PSS, PHQ-9 and VAS scores for fatigue and irritability, as well as increased IL-6 levels, PGE2 concentrations and antioxidant system activation compared to controls. PSS scores positively correlated with functional connectivity between the right anterior insula and right putamen, whereas PHQ-9 scores correlated with functional connectivity between most of the seeds and the right anterior insula. PGE2 (positively) and IL-6 (negatively) correlated with functional connectivity between the right anterior insula and right caudate nucleus and between the right ventral putamen and right putamen/globus pallidus. PGE2 and PSS scores accounted for 46% of the variance in functional connectivity between the anterior insula and putamen. CONCLUSIONS CHC patients exhibited increased perceived stress and depressive symptoms, which were associated with changes in inflammatory marker levels and in functional connectivity between the insula and putamen, areas involved in interoceptive integration, emotional awareness, and orientation of motivational state.
Collapse
Affiliation(s)
- Giovanni Oriolo
- Department of Psychiatry and Psychology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigacion Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain; Department of Medicine, Faculty of Medicine, and Institute of Neuroscience, University of Barcelona (UB), Barcelona, Spain
| | - Laura Blanco-Hinojo
- MRI Research Unit, Department of Radiology, Hospital del Mar, CIBERSAM, G21, Barcelona, Spain
| | - Ricard Navines
- Department of Psychiatry and Psychology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigacion Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain; Department of Medicine, Faculty of Medicine, and Institute of Neuroscience, University of Barcelona (UB), Barcelona, Spain
| | - Zoe Mariño
- Liver Unit, Hospital Clinic, IDIBAPS, Centro Investigacion Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| | - David Martín-Hernández
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, CIBERSAM, Imas12, IUINQ, Madrid, Spain
| | - Myriam Cavero
- Department of Psychiatry and Psychology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigacion Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain; Department of Medicine, Faculty of Medicine, and Institute of Neuroscience, University of Barcelona (UB), Barcelona, Spain
| | - Dolors Gimenez
- Liver Section, Hospital del Mar, Parc de Salut Mar, Grup de Recerca Hepatológica, FIMIM, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Javier Caso
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, CIBERSAM, Imas12, IUINQ, Madrid, Spain
| | - Lucile Capuron
- INRA, Laboratory of Nutrition and Integrative Neurobiology (NutriNeuro), UMR 1286, F-33076 Bordeaux, France; University of Bordeaux, Laboratory of Nutrition and Integrative Neurobiology (NutriNeuro), UMR 1286, F-33076 Bordeaux, France
| | - Xavier Forns
- Liver Unit, Hospital Clinic, IDIBAPS, Centro Investigacion Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| | - Jesus Pujol
- MRI Research Unit, Department of Radiology, Hospital del Mar, CIBERSAM, G21, Barcelona, Spain
| | - Ricard Sola
- Liver Section, Hospital del Mar, Parc de Salut Mar, Grup de Recerca Hepatológica, FIMIM, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Rocio Martin-Santos
- Department of Psychiatry and Psychology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigacion Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain; Department of Medicine, Faculty of Medicine, and Institute of Neuroscience, University of Barcelona (UB), Barcelona, Spain.
| |
Collapse
|
25
|
Freire MAM, Santana LNS, Bittencourt LO, Nascimento PC, Fernandes RM, Leão LKR, Fernandes LMP, Silva MCF, Amado LL, Gomes-Leal W, Crespo-Lopez ME, Maia CDSF, Lima RR. Methylmercury intoxication and cortical ischemia: Pre-clinical study of their comorbidity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 174:557-565. [PMID: 30865911 DOI: 10.1016/j.ecoenv.2019.03.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/01/2019] [Accepted: 03/03/2019] [Indexed: 06/09/2023]
Abstract
Stroke is one of the main causes of human disability worldwide. Ischemic stroke is mostly characterized by metabolic collapse and fast tissue damage, followed by secondary damage in adjacent regions not previously affected. Heavy metals intoxication can be associated with stroke incidence, because of their damaging action in the vascular system. Mercury, in particular, possesses a high tropism by metabolically active regions, such as the brain. In the present study we sought to evaluate whether methylmercury (MeHg) intoxication can aggravate the tissue damage caused by an ischemic stroke induced by microinjections of endothelin-1 (ET-1) into the motor cortex of adult rats. Following MeHg intoxication by gavage (0.04 mg/kg/day) during 60 days, the animals were injected with ET-1 (1 μl, 40 pmol/μl) or vehicle (1 μl). After 7 days, all animals were submitted to behavioral tests and then their brains were processed to biochemical and immunohistochemical analyses. We observed that long-term MeHg intoxication promoted a significant Hg deposits in the motor cortex, with concomitant increase of microglial response, followed by reduction of the neuronal population following ischemia and MeHg intoxication, as well as disturbance in the antioxidant defense mechanisms by misbalance of oxidative biochemistry with increase of both lipid peroxidation and nitrite levels, associated to behavioral deficits. MeHg exposure and cortical ischemia demonstrated that both injuries are able of causing significant neurobehavioural impairments in motor coordination and learning accompanied of an exacerbated microglial activation, oxidative stress and neuronal loss in the motor cortex, indicating that MeHg as a source of metabolic disturbance can act as an important increasing factor of ischemic events in the brain.
Collapse
Affiliation(s)
| | - Luana Nazaré S Santana
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Priscila Cunha Nascimento
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Rafael Monteiro Fernandes
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Luana Ketlen R Leão
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Luanna Melo P Fernandes
- Laboratory of Pharmacology of Inflammation and Behavior, Institute of Health Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Marcia Cristina F Silva
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Lílian Lund Amado
- Laboratory of Ecotoxicology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Walace Gomes-Leal
- Laboratory of Experimental Neuroprotection and Neuroregeneration, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Cristiane do Socorro F Maia
- Laboratory of Pharmacology of Inflammation and Behavior, Institute of Health Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil.
| |
Collapse
|
26
|
Leardini-Tristao M, Charles AL, Lejay A, Pizzimenti M, Meyer A, Estato V, Tibiriçá E, Andres E, Geny B. Beneficial Effect of Exercise on Cognitive Function during Peripheral Arterial Disease: Potential Involvement of Myokines and Microglial Anti-Inflammatory Phenotype Enhancement. J Clin Med 2019; 8:jcm8050653. [PMID: 31083472 PMCID: PMC6571759 DOI: 10.3390/jcm8050653] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/05/2019] [Accepted: 05/07/2019] [Indexed: 12/25/2022] Open
Abstract
Peripheral arterial disease (PAD), leading to intermittent claudication, critical ischemia with rest pain, and/or tissue damage, is a public health issue associated with significant morbidity and mortality. Little is known about the link between PAD, cognitive function, and whether exercise might reduce cognitive dysfunction in PAD patients, as previously observed concerning both quality of life and prognosis. This review highlights the fact that patients suffering from PAD often demonstrate cognitive dysfunction characterized by reduced performance in nonverbal reasoning, reduced verbal fluency, and decreased information processing speed and a greater risk for progression toward dementia. Further, the data presented support that physical exercise, likely through myokine secretion and microglial anti-inflammatory phenotype enhancement, might participate in the cognition protection in common clinical settings.
Collapse
Affiliation(s)
- Marina Leardini-Tristao
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Avenida Brasil 4365, 21040-360, Rio de Janeiro, Brazil.
| | - Anne-Laure Charles
- Université de Strasbourg, Fédération de médecine translationnelle de Strasbourg (FMTS), Faculté de Médecine, Equipe d'accueil 3072, 11 Rue Humann, 67000 Strasbourg, France.
| | - Anne Lejay
- Université de Strasbourg, Fédération de médecine translationnelle de Strasbourg (FMTS), Faculté de Médecine, Equipe d'accueil 3072, 11 Rue Humann, 67000 Strasbourg, France.
- Service de Chirurgie Cardiovasculaire, Pôle de Pathologie Cardiaque, Hôpitaux Universitaires, CHRU Strasbourg, 67000 CEDEX, France.
| | - Mégane Pizzimenti
- Université de Strasbourg, Fédération de médecine translationnelle de Strasbourg (FMTS), Faculté de Médecine, Equipe d'accueil 3072, 11 Rue Humann, 67000 Strasbourg, France.
| | - Alain Meyer
- Université de Strasbourg, Fédération de médecine translationnelle de Strasbourg (FMTS), Faculté de Médecine, Equipe d'accueil 3072, 11 Rue Humann, 67000 Strasbourg, France.
- Service de Physiologie et d'explorations Fonctionnelles, Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, 1 place de l'Hôpital, 67091 Strasbourg CEDEX, France.
| | - Vanessa Estato
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Avenida Brasil 4365, 21040-360, Rio de Janeiro, Brazil.
| | - Eduardo Tibiriçá
- Instituto Nacional de Cardiologia, Ministério da Saúde, Rua das lanjeiras 374, 22240-006, Rio de Janeiro 22240-006, Brazil.
| | - Emmanuel Andres
- Service de Médecine Interne, Diabète et Maladies Métaboliques, Pôle M.I.R.N.E.D., Hôpitaux Universitaires, CHRU Strasbourg, 67000 CEDEX, France.
| | - Bernard Geny
- Université de Strasbourg, Fédération de médecine translationnelle de Strasbourg (FMTS), Faculté de Médecine, Equipe d'accueil 3072, 11 Rue Humann, 67000 Strasbourg, France.
- Service de Physiologie et d'explorations Fonctionnelles, Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, 1 place de l'Hôpital, 67091 Strasbourg CEDEX, France.
| |
Collapse
|
27
|
Shinoda Y, Ehara S, Tatsumi S, Yoshida E, Takahashi T, Eto K, Kaji T, Fujiwara Y. Methylmercury-induced neural degeneration in rat dorsal root ganglion is associated with the accumulation of microglia/macrophages and the proliferation of Schwann cells. J Toxicol Sci 2019; 44:191-199. [PMID: 30842371 DOI: 10.2131/jts.44.191] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Exposure to organic mercury, especially methylmercury (MeHg), causes Minamata disease, a severe chronic neurological disorder. Minamata disease predominantly affects the central nervous system, and therefore, studies on the mechanisms of MeHg neurotoxicity have focused primarily on the brain. Although the peripheral nervous system is also affected by the organometallic compound and shows signs of neural degeneration, the mechanisms of peripheral MeHg neurotoxicity remain unclear. In the present study, we performed quantitative immunohistochemical analyses of the dorsal root ganglion (DRG) and associated sensory and motor fibers to clarify the mechanisms of MeHg-induced peripheral neurotoxicity in Wistar rats. Methylmercury chloride (6.7 mg/kg/day) was orally administrated for 5 days, followed by 2 days without administration, and this cycle was repeated once again. Seven and 14 days after the beginning of MeHg exposure, rats were anesthetized, and their DRGs and sensory and motor nerve fibers were removed and processed for immunohistochemical analyses. The frozen sections were immunostained for neuronal, Schwann cell, microglial and macrophage markers. DRG sensory neuron somata and axons showed significant degeneration on day 14. At the same time, an accumulation of microglia and the infiltration of macrophages were observed in the DRGs and sensory nerve fibers. In addition, MeHg caused significant Schwann cell proliferation in the sensory nerve fibers. In comparison, there was no noticeable change in the motor fibers. Our findings suggest that in the peripheral nervous system, MeHg toxicity is associated with neurodegenerative changes to DRG sensory neurons and the induction of a neuroprotective and/or enhancement of neurodegenerative host response.
Collapse
Affiliation(s)
- Yo Shinoda
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Shunsuke Ehara
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Satoshi Tatsumi
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Eiko Yoshida
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Tsutomu Takahashi
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Komyo Eto
- Health and Nursing Facilities for the Aged, Jushindai, Shinwakai
| | - Toshiyuki Kaji
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Yasuyuki Fujiwara
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|
28
|
Santana LNDS, Bittencourt LO, Nascimento PC, Fernandes RM, Teixeira FB, Fernandes LMP, Freitas Silva MC, Nogueira LS, Amado LL, Crespo-Lopez ME, Maia CDSF, Lima RR. Low doses of methylmercury exposure during adulthood in rats display oxidative stress, neurodegeneration in the motor cortex and lead to impairment of motor skills. J Trace Elem Med Biol 2019; 51:19-27. [PMID: 30466930 DOI: 10.1016/j.jtemb.2018.09.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/21/2018] [Accepted: 09/10/2018] [Indexed: 12/20/2022]
Abstract
Despite the vast distribution among tissues, the central nervous system (CNS) represents the main target of methylmercury (MeHg) toxicity. However, few studies have evaluated the effects of MeHg exposure on the CNS at equivalent doses to human environmental exposure. In our study, we evaluated the motor cortex, an important area of motor control, in adult rats chronically exposed to MeHg in a concentration equivalent to those found in fish-eating populations exposed to mercury (Hg). The parameters evaluated were total Hg accumulation, oxidative stress, tissue damage, and behavioral assessment in functional actions that involved this cortical region. Our results show in exposed animals a significantly greater level of Hg in the motor cortex; increase of nitrite levels and lipid peroxidation, associated with decreased antioxidant capacity against peroxyl radicals; reduction of neuronal and astrocyte density; and poor coordination and motor learning impairment. Our data showed that chronic exposure at low doses to MeHg is capable of promoting damages to the motor cortex of adult animals, with changes in oxidative biochemistry misbalance, neurodegeneration, and motor function impairment.
Collapse
Affiliation(s)
- Luana Nazaré da Silva Santana
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Priscila Cunha Nascimento
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Rafael Monteiro Fernandes
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Francisco Bruno Teixeira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Luanna Melo Pereira Fernandes
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Marcia Cristina Freitas Silva
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Lygia Sega Nogueira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Lílian Lund Amado
- Laboratory of Ecotoxicology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Cristiane do Socorro Ferraz Maia
- Laboratory of Pharmacology of Inflammation and Behavior, Institute of Health Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil.
| |
Collapse
|
29
|
Puga S, Cardoso V, Pinto-Ribeiro F, Pacheco M, Almeida A, Pereira P. Brain morphometric profiles and their seasonal modulation in fish (Liza aurata) inhabiting a mercury contaminated estuary. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 237:318-328. [PMID: 29499575 DOI: 10.1016/j.envpol.2018.02.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/09/2018] [Accepted: 02/16/2018] [Indexed: 06/08/2023]
Abstract
Mercury (Hg) is a potent neurotoxicant known to induce important adverse effects on fish, but a deeper understanding is lacking regarding how environmental exposure affects the brain morphology and neural plasticity of specific brain regions in wild specimens. In this work, it was evaluated the relative volume and cell density of the lateral pallium, hypothalamus, optic tectum and molecular layer of the cerebellum on wild Liza aurata captured in Hg-contaminated (LAR) and non-contaminated (SJ) sites of a coastal system (Ria de Aveiro, Portugal). Given the season-related variations in the environment that fish are naturally exposed, this assessment was performed in the winter and summer. Hg triggered a deficit in cell density of hypothalamus during the winter that could lead to hormonal dysfunctions, while in the summer Hg promoted larger volumes of the optic tectum and cerebellum, indicating the warm period as the most critical for the manifestation of putative changes in visual acuity and motor-dependent tasks. Moreover, in fish from the SJ site, the lateral pallium relative volume and the cell density of the hypothalamus and optic tectum were higher in the winter than in summer. Thus, season-related stimuli strongly influence the size and/or cell density of specific brain regions in the non-contaminated area, pointing out the ability of fish to adapt to environmental and physiological demands. Conversely, fish from the Hg-contaminated site showed a distinct seasonal profile of brain morphology, presenting a larger optic tectum in the summer, as well as a larger molecular layer of the cerebellum with higher cell density. Moreover, Hg exposure impaired the winter-summer variation of the lateral pallium relative size (as observed at SJ). Altogether, seasonal variations in fish neural morphology and physiology should be considered when performing ecotoxicological studies in order to better discriminate the Hg neurotoxicity.
Collapse
Affiliation(s)
- Sónia Puga
- Life and Health Sciences Research Institute (ICVS), School of Medicine (EM), Campus of Gualtar, University of Minho, 4750-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Vera Cardoso
- Life and Health Sciences Research Institute (ICVS), School of Medicine (EM), Campus of Gualtar, University of Minho, 4750-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Filipa Pinto-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine (EM), Campus of Gualtar, University of Minho, 4750-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Mário Pacheco
- Department of Biology and CESAM, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Armando Almeida
- Life and Health Sciences Research Institute (ICVS), School of Medicine (EM), Campus of Gualtar, University of Minho, 4750-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Patrícia Pereira
- Department of Biology and CESAM, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
30
|
The Putative Role of Environmental Mercury in the Pathogenesis and Pathophysiology of Autism Spectrum Disorders and Subtypes. Mol Neurobiol 2017; 55:4834-4856. [DOI: 10.1007/s12035-017-0692-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 07/13/2017] [Indexed: 01/28/2023]
|
31
|
Risher JF, Tucker P. Alkyl Mercury-Induced Toxicity: Multiple Mechanisms of Action. REVIEWS OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2017; 240:105-149. [PMID: 27161558 PMCID: PMC10508330 DOI: 10.1007/398_2016_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
There are a number of mechanisms by which alkylmercury compounds cause toxic action in the body. Collectively, published studies reveal that there are some similarities between the mechanisms of the toxic action of the mono-alkyl mercury compounds methylmercury (MeHg) and ethylmercury (EtHg). This paper represents a summary of some of the studies regarding these mechanisms of action in order to facilitate the understanding of the many varied effects of alkylmercurials in the human body. The similarities in mechanisms of toxicity for MeHg and EtHg are presented and compared. The difference in manifested toxicity of MeHg and EtHg are likely the result of the differences in exposure, metabolism, and elimination from the body, rather than differences in mechanisms of action between the two.
Collapse
Affiliation(s)
- John F Risher
- Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry, 1600 Clifton Road (MS F-58), Atlanta, GA, 30333, USA.
| | - Pamela Tucker
- Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry, 1600 Clifton Road (MS F-58), Atlanta, GA, 30333, USA
| |
Collapse
|
32
|
Sawyer TW, Lee JJ, Villanueva M, Wang Y, Nelson P, Song Y, Fan C, Barnes J, McLaws L. The Effect of Underwater Blast on Aggregating Brain Cell Cultures. J Neurotrauma 2017; 34:517-528. [PMID: 27163293 DOI: 10.1089/neu.2016.4430] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Although the deleterious effects of primary blast on gas-filled organs are well accepted, the effect of blast-induced shock waves on the brain is less clear because of factors that complicate the interpretation of clinical and experimental data. Brain cell aggregate cultures are comprised of multiple differentiated brain cell types and were used to examine the effects of underwater blast. Suspensions of these cultures encased in dialysis tubing were exposed to explosive-generated underwater blasts of low (∼300 kPa), medium (∼2,700 kPa), or high (∼14,000 kPa) intensities and harvested at 1-28 days post-exposure. No changes in gross morphology were noted immediately or weeks after blast wave exposure, and no increases in either apoptotic (caspase-3) or necrotic (lactate dehydrogenase) cell death were observed. Changes in neuronal (neurofilament H, acetylcholinesterase, and choline acetyltransferase) and glial (glial fibrillary acidic protein, glutamine synthetase) endpoints did not occur. However, significant time- and pressure-related increases in Akt (protein kinase B) phosphorylation were noted, as well as declines in vascular endothelial growth factor levels, implicating pathways involved in cellular survival mechanisms. The free-floating nature of the aggregates during blast wave exposure, coupled with their highly hydrolyzed dialysis tubing containment, results in minimized boundary effects, thus enabling accurate assessment of brain cell response to a simplified shock-induced stress wave. This work shows that, at its simplest, blast-induced shock waves produce subtle changes in brain tissue. This study has mechanistic implications for the study of primary blast-induced traumatic brain injury and supports the thesis that underwater blast may cause subtle changes in the brains of submerged individuals.
Collapse
Affiliation(s)
- Thomas W Sawyer
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Julian J Lee
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Mercy Villanueva
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Yushan Wang
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Peggy Nelson
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Yanfeng Song
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Chengyang Fan
- 2 Canada West Biosciences , Calgary, Alberta, Canada
| | - Julia Barnes
- 3 Hyland Quality Systems , Medicine Hat, Alberta, Canada
| | - Lori McLaws
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| |
Collapse
|
33
|
Ringer C, Tune S, Bertoune MA, Schwarzbach H, Tsujikawa K, Weihe E, Schütz B. Disruption of calcitonin gene-related peptide signaling accelerates muscle denervation and dampens cytotoxic neuroinflammation in SOD1 mutant mice. Cell Mol Life Sci 2017; 74:339-358. [PMID: 27554772 PMCID: PMC11107523 DOI: 10.1007/s00018-016-2337-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 08/06/2016] [Accepted: 08/08/2016] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease. Neuronal vacuolization and glial activation are pathologic hallmarks in the superoxide dismutase 1 (SOD1) mouse model of ALS. Previously, we found the neuropeptide calcitonin gene-related peptide (CGRP) associated with vacuolization and astrogliosis in the spinal cord of these mice. We now show that CGRP abundance positively correlated with the severity of astrogliosis, but not vacuolization, in several motor and non-motor areas throughout the brain. SOD1 mice harboring a genetic depletion of the βCGRP isoform showed reduced CGRP immunoreactivity associated with vacuolization, while motor functions, body weight, survival, and astrogliosis were not altered. When CGRP signaling was completely disrupted through genetic depletion of the CGRP receptor component, receptor activity-modifying protein 1 (RAMP1), hind limb muscle denervation, and loss of muscle performance were accelerated, while body weight and survival were not affected. Dampened neuroinflammation, i.e., reduced levels of astrogliosis in the brain stem already in the pre-symptomatic disease stage, and reduced microgliosis and lymphocyte infiltrations during the late disease phase were additional neuropathology features in these mice. On the molecular level, mRNA expression levels of brain-derived neurotrophic factor (BDNF) and those of the anti-inflammatory cytokine interleukin 6 (IL-6) were elevated, while those of several pro-inflammatory cytokines found reduced in the brain stem of RAMP1-deficient SOD1 mice at disease end stage. Our results thus identify an important, possibly dual role of CGRP in ALS pathogenesis.
Collapse
Affiliation(s)
- Cornelia Ringer
- Department of Molecular Neurosciences, Institute of Anatomy and Cell Biology, Philipps-University, Robert-Koch-Strasse 8, 35037, Marburg, Germany
- Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | - Sarah Tune
- Department of Physiology, University of Lübeck, Lübeck, Germany
| | - Mirjam A Bertoune
- Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, Philipps-University, Marburg, Germany
| | - Hans Schwarzbach
- Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, Philipps-University, Marburg, Germany
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Yamadaoka, Suita, Osaka, Japan
| | - Eberhard Weihe
- Department of Molecular Neurosciences, Institute of Anatomy and Cell Biology, Philipps-University, Robert-Koch-Strasse 8, 35037, Marburg, Germany.
| | - Burkhard Schütz
- Department of Molecular Neurosciences, Institute of Anatomy and Cell Biology, Philipps-University, Robert-Koch-Strasse 8, 35037, Marburg, Germany.
| |
Collapse
|
34
|
Yamamoto M, Khan N, Muniroh M, Motomura E, Yanagisawa R, Matsuyama T, Vogel CFA. Activation of interleukin-6 and -8 expressions by methylmercury in human U937 macrophages involves RelA and p50. J Appl Toxicol 2016; 37:611-620. [DOI: 10.1002/jat.3411] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 09/22/2016] [Accepted: 10/03/2016] [Indexed: 01/30/2023]
Affiliation(s)
- Megumi Yamamoto
- Integrated Physiology Section, Department of Basic Medical Science; National Institute for Minamata Disease; 4058-18 Hama Minamata, Kumamoto 867-0008 Japan
| | - Noureen Khan
- Department of Epidemiology and Preventive Medicine; Kagoshima University Graduate School of Medical and Dental Sciences; 8-35-1 Sakuragaoka Kagoshima 890-8544 Japan
| | - Muflihatul Muniroh
- Department of Epidemiology and Preventive Medicine; Kagoshima University Graduate School of Medical and Dental Sciences; 8-35-1 Sakuragaoka Kagoshima 890-8544 Japan
| | - Eriko Motomura
- Integrated Physiology Section, Department of Basic Medical Science; National Institute for Minamata Disease; 4058-18 Hama Minamata, Kumamoto 867-0008 Japan
| | - Rie Yanagisawa
- Integrated Physiology Section, Department of Basic Medical Science; National Institute for Minamata Disease; 4058-18 Hama Minamata, Kumamoto 867-0008 Japan
| | - Takami Matsuyama
- Department of Immunology; Kagoshima University Graduate School of Medical and Dental Sciences; 8-35-1 Sakuragaoka Kagoshima 890-8544 Japan
| | - Christoph F. A. Vogel
- Department of Environmental Toxicology and Center for Health and the Environment; University of California; Davis CA 95616 USA
| |
Collapse
|
35
|
Kapelski P, Skibinska M, Maciukiewicz M, Pawlak J, Permoda-Osip A, Twarowska-Hauser J. Family-based association study of interleukin 6 (IL6) and its receptor (IL6R) functional polymorphisms in schizophrenia in the Polish population. J Neuroimmunol 2015. [DOI: 10.1016/j.jneuroim.2014.09.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Muniroh M, Khan N, Koriyama C, Akiba S, Vogel CFA, Yamamoto M. Suppression of methylmercury-induced IL-6 and MCP-1 expressions by N-acetylcysteine in U-87MG human astrocytoma cells. Life Sci 2015; 134:16-21. [PMID: 26006043 DOI: 10.1016/j.lfs.2015.04.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 04/09/2015] [Accepted: 04/23/2015] [Indexed: 10/23/2022]
Abstract
AIMS The aim of this study was to clarify the involvement of oxidative stress in methylmercury (MeHg)-induced pro-inflammatory cytokine expressions and the suppressive effects of N-acetylcysteine (NAC) in MeHg-induced cytokine expression. MATERIALS AND METHODS Using U-87-MG human astrocytoma cell line, interleukin (IL)-6 and monocyte chemoattractant protein (MCP)-1 expressions induced by 4 μM MeHg were measured at mRNA and protein levels. Hydrogen peroxide (H2O2) and superoxide anion (O2(-)) were quantified by flow-cytometry analysis. To examine the suppressive effects of NAC on the cytokine expressions among different timing of NAC treatment, cells were treated with 0.5 or 5mM NAC before, simultaneously, or after MeHg administration. KEY FINDINGS MeHg exposure at 4 μM, a non-cytotoxic concentration, significantly induced MCP-1 and IL-6 expressions at both mRNA and protein levels. A significant increase of H2O2 production but not O2(-) was observed. MeHg-induced expression of MCP-1 and IL-6 mRNA was reduced by 10-20% in the presence of 5mM NAC (co-treatment experiment) compared to cells treated with MeHg only. Pre-treatment of cells with 0.5 or 5mM NAC at 0.5 or 1h and its subsequent washout before MeHg addition suppressed MCP-1 and IL-6 cytokine expressions. Post-treatment of cells with NAC after MeHg addition also suppressed the cytokine induction, but the magnitude of suppression was evidently lower than in co-treated cells even though the H2O2 generation was almost completely suppressed by NAC. SIGNIFICANCE NAC may effectively suppress the MeHg-induced cytokine production through both, inhibition of reactive oxygen species as well as extracellular chelation of MeHg in astrocytes.
Collapse
Affiliation(s)
- Muflihatul Muniroh
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan; Department of Physiology, Faculty of Medicine, Diponegoro University, Tembalang, Semarang 50725, Indonesia
| | - Noureen Khan
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - Chihaya Koriyama
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan.
| | - Suminori Akiba
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - Christoph F A Vogel
- Department of Environmental Toxicology and Center for Health and the Environment, University of California, Davis, CA 95616, USA
| | - Megumi Yamamoto
- Integrated Physiology Section, Department of Basic Medical Science, National Institute for Minamata Disease, 4058-18 Hama, Minamata, Kumamoto 867-0008, Japan
| |
Collapse
|
37
|
Yang XT, Huang GH, Feng DF, Chen K. Insight into astrocyte activation after optic nerve injury. J Neurosci Res 2014; 93:539-48. [PMID: 25257183 DOI: 10.1002/jnr.23487] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 08/25/2014] [Accepted: 08/29/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Xi-Tao Yang
- Department of Neurosurgery, No. 3 People's Hospital; Shanghai Jiaotong University School of Medicine; Shanghai China
| | - Guo-Hui Huang
- Department of Neurosurgery, No. 3 People's Hospital; Shanghai Jiaotong University School of Medicine; Shanghai China
| | - Dong-Fu Feng
- Department of Neurosurgery, No. 3 People's Hospital; Shanghai Jiaotong University School of Medicine; Shanghai China
- Institute of Traumatic Medicine; Shanghai Jiaotong University School of Medicine; Shanghai China
| | - Kui Chen
- Department of Neurosurgery, No. 3 People's Hospital; Shanghai Jiaotong University School of Medicine; Shanghai China
| |
Collapse
|
38
|
Alépée N, Bahinski A, Daneshian M, De Wever B, Fritsche E, Goldberg A, Hansmann J, Hartung T, Haycock J, Hogberg H, Hoelting L, Kelm JM, Kadereit S, McVey E, Landsiedel R, Leist M, Lübberstedt M, Noor F, Pellevoisin C, Petersohn D, Pfannenbecker U, Reisinger K, Ramirez T, Rothen-Rutishauser B, Schäfer-Korting M, Zeilinger K, Zurich MG. State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2014. [PMID: 25027500 DOI: 10.14573/altex1406111] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Integrated approaches using different in vitro methods in combination with bioinformatics can (i) increase the success rate and speed of drug development; (ii) improve the accuracy of toxicological risk assessment; and (iii) increase our understanding of disease. Three-dimensional (3D) cell culture models are important building blocks of this strategy which has emerged during the last years. The majority of these models are organotypic, i.e., they aim to reproduce major functions of an organ or organ system. This implies in many cases that more than one cell type forms the 3D structure, and often matrix elements play an important role. This review summarizes the state of the art concerning commonalities of the different models. For instance, the theory of mass transport/metabolite exchange in 3D systems and the special analytical requirements for test endpoints in organotypic cultures are discussed in detail. In the next part, 3D model systems for selected organs--liver, lung, skin, brain--are presented and characterized in dedicated chapters. Also, 3D approaches to the modeling of tumors are presented and discussed. All chapters give a historical background, illustrate the large variety of approaches, and highlight up- and downsides as well as specific requirements. Moreover, they refer to the application in disease modeling, drug discovery and safety assessment. Finally, consensus recommendations indicate a roadmap for the successful implementation of 3D models in routine screening. It is expected that the use of such models will accelerate progress by reducing error rates and wrong predictions from compound testing.
Collapse
|
39
|
Shinozaki Y, Nomura M, Iwatsuki K, Moriyama Y, Gachet C, Koizumi S. Microglia trigger astrocyte-mediated neuroprotection via purinergic gliotransmission. Sci Rep 2014; 4:4329. [PMID: 24710318 PMCID: PMC3948352 DOI: 10.1038/srep04329] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/21/2014] [Indexed: 12/21/2022] Open
Abstract
Microglia are highly sensitive to even small changes in the brain environment, such as invasion of non-hazardous toxicants or the presymptomatic state of diseases. However, the physiological or pathophysiological consequences of their responses remain unknown. Here, we report that cultured microglia sense low concentrations of the neurotoxicant methylmercury (MeHglow) and provide neuroprotection against MeHg, for which astrocytes are also required. When exposed to MeHglow, microglia exocytosed ATP via p38 MAPK- and vesicular nucleotide transporter (VNUT)-dependent mechanisms. Astrocytes responded to the microglia-derived ATP via P2Y1 receptors and released interleukin-6 (IL-6), thereby protecting neurons against MeHglow. These neuroprotective actions were also observed in organotypic hippocampal slices from wild-type mice, but not in slices prepared from VNUT knockout or P2Y1 receptor knockout mice. These findings suggest that microglia sense and respond to even non-hazardous toxicants such as MeHglow and change their phenotype into a neuroprotective one, for which astrocytic support is required.
Collapse
Affiliation(s)
- Youichi Shinozaki
- 1] Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan [2] Japan Science and Technology Agency, CREST, Tokyo 102-0076, Japan
| | - Masatoshi Nomura
- Department of Endocrine and Metabolic Diseases/Diabetes Mellitus Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Ken Iwatsuki
- Institute for Innovation, Ajinomoto Co. Inc., Kawasaki 210-8681, Japan
| | - Yoshinori Moriyama
- Advanced Science Research Center, Okayama University, Okayama 700-8530, Japan
| | - Christian Gachet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U.311, Etablissement de Transfusion Sanguine, 10, rue Spielmann, B.P. 36, 67065 Strasbourg, France
| | - Schuichi Koizumi
- 1] Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan [2] Japan Science and Technology Agency, CREST, Tokyo 102-0076, Japan
| |
Collapse
|
40
|
Na KS, Jung HY, Kim YK. The role of pro-inflammatory cytokines in the neuroinflammation and neurogenesis of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2014; 48:277-86. [PMID: 23123365 DOI: 10.1016/j.pnpbp.2012.10.022] [Citation(s) in RCA: 294] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/11/2012] [Accepted: 10/26/2012] [Indexed: 12/22/2022]
Abstract
Schizophrenia is a serious mental illness with chronic symptoms and significant impairment in psychosocial functioning. Although novel antipsychotics have been developed, the negative and cognitive symptoms of schizophrenia are still unresponsive to pharmacotherapy. The high level of social impairment and a chronic deteriorating course suggest that schizophrenia likely has neurodegenerative characteristics. Inflammatory markers such as pro-inflammatory cytokines are well-known etiological factors for psychiatric disorders, including schizophrenia. Inflammation in the central nervous system is closely related to neurodegeneration. In addition to pro-inflammatory cytokines, microglia also play an important role in the inflammatory process in the CNS. Uncontrolled activity of pro-inflammatory cytokines and microglia can induce schizophrenia in tandem with genetic vulnerability and glutamatergic neurotransmitters. Several studies have investigated the possible effects of antipsychotics on inflammation and neurogenesis. Additionally, anti-inflammatory adjuvant therapy has been under investigation as a treatment option for schizophrenia. Further studies should consider the confounding effects of systemic factors such as metabolic syndrome and smoking. In addition, the unique mechanisms by which pro-inflammatory cytokines are involved in the etiopathology of schizophrenia should be investigated. In this article, we aimed to review (1) major findings regarding neuroinflammation and pro-inflammatory cytokine alterations in schizophrenia, (2) interactions between neuroinflammation and neurogenesis as possible neural substrates for schizophrenia, and (3) novel pharmacological approaches.
Collapse
Affiliation(s)
- Kyoung-Sae Na
- Department of Psychiatry, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | | | | |
Collapse
|
41
|
Alépée N, Bahinski A, Daneshian M, De Wever B, Fritsche E, Goldberg A, Hansmann J, Hartung T, Haycock J, Hogberg HT, Hoelting L, Kelm JM, Kadereit S, McVey E, Landsiedel R, Leist M, Lübberstedt M, Noor F, Pellevoisin C, Petersohn D, Pfannenbecker U, Reisinger K, Ramirez T, Rothen-Rutishauser B, Schäfer-Korting M, Zeilinger K, Zurich MG. State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology. ALTEX 2014; 31:441-77. [PMID: 25027500 PMCID: PMC4783151 DOI: 10.14573/altex.1406111] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 06/30/2014] [Indexed: 02/02/2023]
Abstract
Integrated approaches using different in vitro methods in combination with bioinformatics can (i) increase the success rate and speed of drug development; (ii) improve the accuracy of toxicological risk assessment; and (iii) increase our understanding of disease. Three-dimensional (3D) cell culture models are important building blocks of this strategy which has emerged during the last years. The majority of these models are organotypic, i.e., they aim to reproduce major functions of an organ or organ system. This implies in many cases that more than one cell type forms the 3D structure, and often matrix elements play an important role. This review summarizes the state of the art concerning commonalities of the different models. For instance, the theory of mass transport/metabolite exchange in 3D systems and the special analytical requirements for test endpoints in organotypic cultures are discussed in detail. In the next part, 3D model systems for selected organs--liver, lung, skin, brain--are presented and characterized in dedicated chapters. Also, 3D approaches to the modeling of tumors are presented and discussed. All chapters give a historical background, illustrate the large variety of approaches, and highlight up- and downsides as well as specific requirements. Moreover, they refer to the application in disease modeling, drug discovery and safety assessment. Finally, consensus recommendations indicate a roadmap for the successful implementation of 3D models in routine screening. It is expected that the use of such models will accelerate progress by reducing error rates and wrong predictions from compound testing.
Collapse
Affiliation(s)
| | - Anthony Bahinski
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, USA
| | - Mardas Daneshian
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany
| | | | - Ellen Fritsche
- Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Alan Goldberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - Jan Hansmann
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Thomas Hartung
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany,Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - John Haycock
- Department of Materials Science of Engineering, University of Sheffield, Sheffield, UK
| | - Helena T. Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - Lisa Hoelting
- Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | | | - Suzanne Kadereit
- Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Emily McVey
- Board for the Authorization of Plant Protection Products and Biocides, Wageningen, The Netherlands
| | | | - Marcel Leist
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany,Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Marc Lübberstedt
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Fozia Noor
- Biochemical Engineering, Saarland University, Saarbruecken, Germany
| | | | | | | | | | - Tzutzuy Ramirez
- BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
| | | | - Monika Schäfer-Korting
- Institute for Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Berlin, Germany
| | - Katrin Zeilinger
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Marie-Gabriele Zurich
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland,Swiss Center for Applied Human Toxicology (SCAHT), University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
42
|
Zeidán-Chuliá F, Salmina AB, Malinovskaya NA, Noda M, Verkhratsky A, Moreira JCF. The glial perspective of autism spectrum disorders. Neurosci Biobehav Rev 2014; 38:160-72. [DOI: 10.1016/j.neubiorev.2013.11.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 11/03/2013] [Accepted: 11/21/2013] [Indexed: 01/01/2023]
|
43
|
Maternal immune activation during gestation interacts with Disc1 point mutation to exacerbate schizophrenia-related behaviors in mice. J Neurosci 2013; 33:7654-66. [PMID: 23637159 DOI: 10.1523/jneurosci.0091-13.2013] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Schizophrenia is thought to result from interactions between susceptible genotypes and environmental risk factors. DISC1 is an important gene for schizophrenia and mood disorders based on both human and animal studies. In the present study we sought to investigate interactions between two distinct point mutations in the mouse Disc1 gene (L100P and Q31L) and maternal immune activation (MIA) during pregnancy with polyinosinic:polycytidylic acid (polyI:C). PolyI:C given at 5 mg/kg impaired cognitive and social behavior in both wild-type (WT) and Disc1-Q31L(+/-) offspring, and reduced prepulse inhibition at 16 but not 8 weeks of age. Disc1-L100P(+/-) mutants were more sensitive to MIA than WT or Disc1-Q31L(+/-) mice. Interleukin-6 (IL-6) is a critical cytokine for mediating the behavioral and transcriptional effects of polyI:C. We found a more pronounced increase of IL-6 in response to polyI:C in fetal brain in Disc1-L100P(+/-) mice compared with WT or Disc1-Q31L(+/-) mice. Coadministration of an anti-IL-6 antibody with polyI:C reversed schizophrenia-related behavioral phenotypes in Disc1-L100P(+/-) mice. In summary, we found specific interactions between discrete genetic (Disc1-L100P(+/-)) and environmental factors (MIA) that exacerbate schizophrenia-related phenotypes. IL-6 may be important in the pathophysiology of this interaction.
Collapse
|
44
|
Bozkurt A, Yardan T, Ciftcioglu E, Baydin A, Hakligor A, Bitigic M, Bilge S. Time course of serum S100B protein and neuron-specific enolase levels of a single dose of chlorpyrifos in rats. Basic Clin Pharmacol Toxicol 2012; 107:893-8. [PMID: 20456333 DOI: 10.1111/j.1742-7843.2010.00593.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Organophosphate (OP) compounds are a large class of chemicals, many of which are used as pesticides. It is suggested that OPs specifically affect glia and neurons. Effects of acute exposure to chlorpyrifos (CPF), which is a common organophosphorus pesticide used worldwide, on neuron-specific enolase (NSE) and S100B levels in rat blood during 7 days were assessed. Rats were evaluated either before (0 hr) or 2, 12, 24, 48 and 168 hr (7 days) after injection of CPF (279 mg/kg, s.c.) or vehicle (peanut oil, 2 ml/kg, s.c.) for clinical signs of toxicity. Immediately after the evaluation of toxicity, blood samples were taken for biochemical assays. CPF administration produced decreases in body-weight and temperature, which were observed for first time at 12 hr after CPF administration and continued for 168 hr (p < 0.05-0.001). Serum S100B and NSE levels were acutely increased 2 hr after CPF administration and remained high at 12 hr (p < 0.01-0.001). NSE and S100B levels were not different in either CPF or vehicle groups at following time points. Serum butyrylcholinesterase (EC 3.1.1.8; BuChE) activity was dramatically reduced at 2 hr after CPF and remained low at each time points during 7 days (p < 0.01-0.001). Our results suggest that the usefulness of serum levels of these glia- and neuron-specific marker proteins in assessing OP toxicity, specifically CPF-induced toxicity.
Collapse
Affiliation(s)
- Ayhan Bozkurt
- Department of Physiology, Ondokuz Mayıs University, School of Medicine, Samsun, Turkey.
| | | | | | | | | | | | | |
Collapse
|
45
|
Ni M, Li X, Rocha JBT, Farina M, Aschner M. Glia and methylmercury neurotoxicity. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2012; 75:1091-1101. [PMID: 22852858 PMCID: PMC4059390 DOI: 10.1080/15287394.2012.697840] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Methylmercury (MeHg) is a global environmental pollutant with significant adverse effects on human health. As the major target of MeHg, the central nervous system (CNS) exhibits the most recognizable poisoning symptoms. The role of the two major nonneuronal cell types, astrocytes and microglia, in response to MeHg exposure was recently compared. These two cell types share several common features in MeHg toxicity, but interestingly, these cells types also exhibit distinct response kinetics, indicating a cell-specific role in mediating MeHg-induced neurotoxicity. The aim of this study was to review the most recent literature and summarize key features of glial responses to this organometal.
Collapse
Affiliation(s)
- Mingwei Ni
- Department of Surgery, New York Hospital Medical Center Queens, New York City, New York, USA
| | - Xin Li
- Neuroscience Graduate Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - João B. T. Rocha
- Departamento de Química, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, 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 Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
46
|
Monnet‐Tschudi F, Defaux A, Braissant O, Cagnon L, Zurich M. Methods to Assess Neuroinflammation. ACTA ACUST UNITED AC 2011; Chapter 12:Unit12.19. [DOI: 10.1002/0471140856.tx1219s50] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Florianne Monnet‐Tschudi
- Department of Physiology, University of Lausanne Lausanne Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT) Lausanne Switzerland
| | - Antoinette Defaux
- Department of Physiology, University of Lausanne Lausanne Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT) Lausanne Switzerland
| | - Olivier Braissant
- Clinical Chemistry Laboratory, Centre Hospitalier Universitaire Vaudois Lausanne Switzerland
| | - Laurène Cagnon
- Clinical Chemistry Laboratory, Centre Hospitalier Universitaire Vaudois Lausanne Switzerland
| | - Marie‐Gabrielle Zurich
- Department of Physiology, University of Lausanne Lausanne Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT) Lausanne Switzerland
| |
Collapse
|
47
|
Farina M, Aschner M, Rocha JBT. Oxidative stress in MeHg-induced neurotoxicity. Toxicol Appl Pharmacol 2011; 256:405-17. [PMID: 21601588 PMCID: PMC3166649 DOI: 10.1016/j.taap.2011.05.001] [Citation(s) in RCA: 241] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 05/01/2011] [Accepted: 05/02/2011] [Indexed: 12/20/2022]
Abstract
Methylmercury (MeHg) is an environmental toxicant that leads to long-lasting neurological and developmental deficits in animals and humans. Although the molecular mechanisms mediating MeHg-induced neurotoxicity are not completely understood, several lines of evidence indicate that oxidative stress represents a critical event related to the neurotoxic effects elicited by this toxicant. The objective of this review is to summarize and discuss data from experimental and epidemiological studies that have been important in clarifying the molecular events which mediate MeHg-induced oxidative damage and, consequently, toxicity. Although unanswered questions remain, the electrophilic properties of MeHg and its ability to oxidize thiols have been reported to play decisive roles to the oxidative consequences observed after MeHg exposure. However, a close examination of the relationship between low levels of MeHg necessary to induce oxidative stress and the high amounts of sulfhydryl-containing antioxidants in mammalian cells (e.g., glutathione) have led to the hypothesis that nucleophilic groups with extremely high affinities for MeHg (e.g., selenols) might represent primary targets in MeHg-induced oxidative stress. Indeed, the inhibition of antioxidant selenoproteins during MeHg poisoning in experimental animals has corroborated this hypothesis. The levels of different reactive species (superoxide anion, hydrogen peroxide and nitric oxide) have been reported to be increased in MeHg-exposed systems, and the mechanisms concerning these increments seem to involve a complex sequence of cascading molecular events, such as mitochondrial dysfunction, excitotoxicity, intracellular calcium dyshomeostasis and decreased antioxidant capacity. This review also discusses potential therapeutic strategies to counteract MeHg-induced toxicity and oxidative stress, emphasizing the use of organic selenocompounds, which generally present higher affinity for MeHg when compared to the classically studied agents.
Collapse
Affiliation(s)
- Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Michael Aschner
- Department of Pediatrics and Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - João B. T. Rocha
- Departamento de Química, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| |
Collapse
|
48
|
Bassett T, Bach P, Chan HM. Effects of methylmercury on the secretion of pro-inflammatory cytokines from primary microglial cells and astrocytes. Neurotoxicology 2011; 33:229-34. [PMID: 22037494 DOI: 10.1016/j.neuro.2011.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 10/10/2011] [Accepted: 10/13/2011] [Indexed: 10/16/2022]
Abstract
Glial cells, including oligodendrocytes, astrocytes and microglia are important to proper central nervous system (CNS) function. Deregulation or changes to CNS populations of astrocytes and microglia in particular are expected to play a role in many neurodegenerative diseases, including Parkinson's disease, amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD). Previous studies have reported methylmercury (MeHg) induced changes in glial cell function; however, the effects of MeHg on these cells remains poorly understood. This study aims to examine the effect of MeHg on the secretion of pro-inflammatory cytokines from microglia and astrocytes. The impact of the microglia/astrocyte ratio on cytokine secretion was also examined. Microglia and astrocytes were cultured from the brains of neo-natal BALB/C mice and dosed with MeHg (0-1 μM) and stimulated with PAM(3)CSK(4) (PAM(3)), a toll-like receptor (TLR) ligand. After this, the secretion of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α) and interleukin-1-beta (IL-1β) was measured by ELISA. MeHg reduced the secretion of IL-6 in a dose dependant manner but did not effect the secretion of TNF-α. No change in IL-1β was observed in any treatments, indicating that PAM(3) cannot induce the secretion of this cytokine from glial cells. Additionally, the ratio of microglia/astrocyte had an effect on the secretion of IL-6 but not TNF-α. These results indicate that MeHg can modify the response of glial cells and the interactions with astrocytes can affect the response of the microglia cells in culture. These results are significant in understanding the potential relationship with MeHg and neurodegenerative diseases and for the interpretation of results of future in vitro studies using monoculture.
Collapse
Affiliation(s)
- Tyler Bassett
- Community Health Sciences Program, University of Northern British Columbia, Prince George, BC, Canada
| | | | | |
Collapse
|
49
|
Silva SL, Osório C, Vaz AR, Barateiro A, Falcão AS, Silva RFM, Brites D. Dynamics of neuron-glia interplay upon exposure to unconjugated bilirubin. J Neurochem 2011; 117:412-24. [PMID: 21275990 DOI: 10.1111/j.1471-4159.2011.07200.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Microglia are the main players of the brain immune response. They act as active sensors that rapidly respond to injurious insults by shifting into different activated states. Elevated levels of unconjugated bilirubin (UCB) induce cell death, immunostimulation and oxidative stress in both neurons and astrocytes. We recently reported that microglial phagocytic phenotype precedes the release of pro-inflammatory cytokines upon UCB exposure. We investigated whether and how microglia microenvironment influences the response to UCB. Our findings revealed that conditioned media derived from UCB-treated astrocytes reduce microglial inflammatory reaction and cell death, suggesting an attempt to curtail microglial over activation. Conditioned medium from UCB-challenged neurons, although down-regulating tumor necrosis factor-α and interleukin-1β promoted the release of interleukin-6 and nitric oxide, the activation of matrix metalloproteinase-9, and cell death, as compared with UCB-direct effects on microglia. Moreover, soluble factors released by UCB-treated neurons intensified the phagocytic properties manifested by microglia under direct exposure to UCB. Results from neuron-microglia mixed cultures incubated with UCB evidenced that sensitized microglia were able to prevent neurite outgrowth impairment and cell death. In conclusion, our data indicate that stressed neurons signal microglial clearance functions, but also overstimulate its inflammatory potential ultimately leading to microglia demise.
Collapse
Affiliation(s)
- Sandra L Silva
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | | | | | | | | | | | | |
Collapse
|
50
|
Ni M, Li X, Yin Z, Sidoryk-Węgrzynowicz M, Jiang H, Farina M, Rocha JBT, Syversen T, Aschner M. Comparative study on the response of rat primary astrocytes and microglia to methylmercury toxicity. Glia 2011; 59:810-20. [PMID: 21351162 DOI: 10.1002/glia.21153] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 01/11/2011] [Indexed: 12/16/2022]
Abstract
As the two major glial cell types in the brain, astrocytes and microglia play pivotal but different roles in maintaining optimal brain function. Although both cell types have been implicated as major targets of methylmercury (MeHg), their sensitivities and adaptive responses to this metal can vary given their distinctive properties and physiological functions. This study was carried out to compare the responses of astrocytes and microglia following MeHg treatment, specifically addressing the effects of MeHg on cell viability, reactive oxygen species (ROS) generation and glutathione (GSH) levels, as well as mercury (Hg) uptake and the expression of NF-E2-related factor 2 (Nrf2). Results showed that microglia are more sensitive to MeHg than astrocytes, a finding that is consistent with their higher Hg uptake and lower basal GSH levels. Microglia also demonstrated higher ROS generation compared with astrocytes. Nrf2 and its downstream genes were upregulated in both cell types, but with different kinetics (much faster in microglia). In summary, microglia and astrocytes each exhibit a distinct sensitivity to MeHg, resulting in their differential temporal adaptive responses. These unique sensitivities appear to be dependent on the cellular thiol status of the particular cell type.
Collapse
Affiliation(s)
- Mingwei Ni
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | | | |
Collapse
|