1
|
Cresto N, Forner-Piquer I, Baig A, Chatterjee M, Perroy J, Goracci J, Marchi N. Pesticides at brain borders: Impact on the blood-brain barrier, neuroinflammation, and neurological risk trajectories. CHEMOSPHERE 2023; 324:138251. [PMID: 36878369 DOI: 10.1016/j.chemosphere.2023.138251] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/11/2023] [Accepted: 02/24/2023] [Indexed: 06/18/2023]
Abstract
Pesticides are omnipresent, and they pose significant environmental and health risks. Translational studies indicate that acute exposure to high pesticide levels is detrimental, and prolonged contact with low concentrations of pesticides, as single and cocktail, could represent a risk factor for multi-organ pathophysiology, including the brain. Within this research template, we focus on pesticides' impact on the blood-brain barrier (BBB) and neuroinflammation, physical and immunological borders for the homeostatic control of the central nervous system (CNS) neuronal networks. We examine the evidence supporting a link between pre- and postnatal pesticide exposure, neuroinflammatory responses, and time-depend vulnerability footprints in the brain. Because of the pathological influence of BBB damage and inflammation on neuronal transmission from early development, varying exposures to pesticides could represent a danger, perhaps accelerating adverse neurological trajectories during aging. Refining our understanding of how pesticides influence brain barriers and borders could enable the implementation of pesticide-specific regulatory measures directly relevant to environmental neuroethics, the exposome, and one-health frameworks.
Collapse
Affiliation(s)
- Noemie Cresto
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Isabel Forner-Piquer
- Centre for Pollution Research and Policy, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, United Kingdom.
| | - Asma Baig
- Centre for Pollution Research and Policy, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, United Kingdom
| | - Mousumi Chatterjee
- Centre for Pollution Research and Policy, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, United Kingdom
| | - Julie Perroy
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Nicola Marchi
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
2
|
Miranda CA, Beretta EM, Ferreira LA, da Silva ES, Coimbra BZ, Pereira PT, Miranda RG, Dorta DJ, Rodrigues FTV, Mingatto FE. Role of biotransformation in the diazinon-induced toxicity in HepG2 cells and antioxidant protection by tetrahydrocurcumin. Toxicol Rep 2022; 10:32-39. [PMID: 36578673 PMCID: PMC9791245 DOI: 10.1016/j.toxrep.2022.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/27/2022] [Accepted: 12/11/2022] [Indexed: 12/14/2022] Open
Abstract
Diazinon (DZN) is an insecticide extensively used to control pests in crops and animals. However, its indicriminated use may lead to liver damage in animals and humans. This study aimed to evaluate the toxicity of DZN (25-150 µM) on human hepatoblastoma (HepG2) cells after 24 and 48 h of exposure and the role of its biotransformation on the toxicological potential. We also tested the protective effect of tetrahydrocurcumin (THC), an antioxidant agent, in the DZN-induced citotoxicity. DZN caused cytotoxicity in the HepG2 cells, inhibiting cell proliferation and reducing cell viability in a dose- and time-dependent manner. The pre-incubation of HepG2 cells with chemical inducers of cytochrome P450 monooxygenase 3-methylcholanthrene and phenobarbital resulted in a further decrease of cell viability associated with DZN exposure. In addition, the metabolite diazoxon was more toxic than DZN. Our results also revealed that THC alleviated DZN-induced cytotoxicity and reactive oxygen and nitrogen species (RONS) generation in HepG2 cells. In conclusion, our data provide novel insights into the involvement of biotransformation in the mechanisms of DZN-induced cytotoxicity and suggest that amelioration of RONS accumulation might be involved in the protective effect of THC on DZN-induced liver injury.
Collapse
Affiliation(s)
- Camila Araújo Miranda
- Department of Animal Science, College of Agricultural and Technological Sciences, São Paulo State University (Unesp), Dracena, SP, Brazil
| | - Eduardo Morais Beretta
- Department of Animal Science, College of Agricultural and Technological Sciences, São Paulo State University (Unesp), Dracena, SP, Brazil
- Medical School, Unifadra, Faculdades de Dracena, Dracena, SP, Brazil
| | - Layra Araújo Ferreira
- Department of Animal Science, College of Agricultural and Technological Sciences, São Paulo State University (Unesp), Dracena, SP, Brazil
| | | | | | | | - Raul Ghiraldelli Miranda
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Daniel Junqueira Dorta
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Flávia Thomaz Verechia Rodrigues
- Department of Animal Science, College of Agricultural and Technological Sciences, São Paulo State University (Unesp), Dracena, SP, Brazil
| | - Fábio Erminio Mingatto
- Department of Animal Science, College of Agricultural and Technological Sciences, São Paulo State University (Unesp), Dracena, SP, Brazil
- Correspondence to: Department of Animal Science, College of Agricultural and Technological Sciences, São Paulo State University (Unesp), Dracena, 17900–000, SP, Brazil.
| |
Collapse
|
3
|
Li J, Bi H. Integrated Strategy of Network Pharmacology and in vitro Screening to Identify Mechanism of Diazinon-induced Hippocampal Neurotoxicity. Neurotoxicology 2022; 92:122-130. [DOI: 10.1016/j.neuro.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 10/16/2022]
|
4
|
Polonikov A, Bocharova I, Azarova I, Klyosova E, Bykanova M, Bushueva O, Polonikova A, Churnosov M, Solodilova M. The Impact of Genetic Polymorphisms in Glutamate-Cysteine Ligase, a Key Enzyme of Glutathione Biosynthesis, on Ischemic Stroke Risk and Brain Infarct Size. Life (Basel) 2022; 12:life12040602. [PMID: 35455093 PMCID: PMC9032935 DOI: 10.3390/life12040602] [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: 04/07/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 11/16/2022] Open
Abstract
The purpose of this pilot study was to explore whether polymorphisms in genes encoding the catalytic (GCLC) and modifier (GCLM) subunits of glutamate-cysteine ligase, a rate-limiting enzyme in glutathione synthesis, play a role in the development of ischemic stroke (IS) and the extent of brain damage. A total of 1288 unrelated Russians, including 600 IS patients and 688 age- and sex-matched healthy subjects, were enrolled for the study. Nine common single nucleotide polymorphisms (SNPs) of the GCLC and GCLM genes were genotyped using the MassArray-4 system. SNP rs2301022 of GCLM was strongly associated with a decreased risk of ischemic stroke regardless of sex and age (OR = 0.39, 95%CI 0.24−0.62, p < 0.0001). Two common haplotypes of GCLM possessed protective effects against ischemic stroke risk (p < 0.01), but exclusively in nonsmoker patients. Infarct size was increased by polymorphisms rs636933 and rs761142 of GCLC. The mbmdr method enabled identifying epistatic interactions of GCLC and GCLM gene polymorphisms with known IS susceptibility genes that, along with environmental risk factors, jointly contribute to the disease risk and brain infarct size. Understanding the impact of genes and environmental factors on glutathione metabolism will allow the development of effective strategies for the treatment of ischemic stroke and disease prevention.
Collapse
Affiliation(s)
- Alexey Polonikov
- Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia; (E.K.); (M.B.); (O.B.); (A.P.); (M.S.)
- Correspondence:
| | - Iuliia Bocharova
- Department of Medical Biological Disciplines, Belgorod State University, 85 Pobedy Street, 308015 Belgorod, Russia; (I.B.); (M.C.)
- Division of Neurosurgery, Kursk Regional Clinical Hospital, 45a Sumskaya, 305027 Kursk, Russia
| | - Iuliia Azarova
- Department of Biological Chemistry, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia;
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| | - Elena Klyosova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia; (E.K.); (M.B.); (O.B.); (A.P.); (M.S.)
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| | - Marina Bykanova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia; (E.K.); (M.B.); (O.B.); (A.P.); (M.S.)
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| | - Olga Bushueva
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia; (E.K.); (M.B.); (O.B.); (A.P.); (M.S.)
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| | - Anna Polonikova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia; (E.K.); (M.B.); (O.B.); (A.P.); (M.S.)
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State University, 85 Pobedy Street, 308015 Belgorod, Russia; (I.B.); (M.C.)
| | - Maria Solodilova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia; (E.K.); (M.B.); (O.B.); (A.P.); (M.S.)
| |
Collapse
|
5
|
Sibomana I, Rohan JG, Mattie DR. 21-Day dermal exposure to aircraft engine oils: effects on esterase activities in brain and liver tissues, blood, plasma, and clinical chemistry parameters for Sprague Dawley rats. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2021; 84:357-388. [PMID: 33380269 DOI: 10.1080/15287394.2020.1867680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
This dermal study tested the potential toxicity of grade 3 (G3) and 4 (G4) organophosphate-containing aircraft engine oils in both new (G3-N, G4-N) and used states (G3-U, G4-U) to alter esterase activities in blood, brain and liver tissues, clinical chemistry parameters, and electrophysiology of hippocampal neurons. A 300 µl volume of undiluted oil was applied in Hill Top Chamber Systems®, then attached to fur-free test sites on backs of male and female Sprague Dawley rats for 6 hr/day, 5 days/week for 21 days. Recovery rats received similar treatments and kept for 14 days post-exposure to screen for reversibility, persistence, or delayed occurrence of toxicity. In brain, both versions of G3 and G4 significantly decreased (32-41%) female acetylcholinesterase (AChE) activity while in males only G3-N and G4-N reduced (33%) AChE activity. Oils did not markedly affect AChE in liver, regardless of gender. In whole blood, G3-U decreased female AChE (29%) which persisted during recovery (32%). G4-N significantly lowered (29%) butyrylcholinesterase (BChE) in male plasma, but this effect was resolved during recovery. For clinical chemistry indices, only globulin levels in female plasma significantly increased following G3-N or G4-N exposure. Preliminary electrophysiology data suggested that effects of both versions of G3 and G4 on hippocampal function may be gender dependent. Aircraft maintenance workers may be at risk if precautions are not taken to minimize long-term aircraft oil exposure.
Collapse
Affiliation(s)
- Isaie Sibomana
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Wright-Patterson Air Force Base, OH, USA
- Air Force Research Laboratory, 711 Human Performance Wing, Wright-Patterson Air Force Base, OH, USA
| | - Joyce G Rohan
- Environmental Health Effects Laboratory, Naval Medical Research Unit Dayton (NAMRU-D), Wright-Patterson Air Force Base, OH, USA
| | - David R Mattie
- Air Force Research Laboratory, 711 Human Performance Wing, Wright-Patterson Air Force Base, OH, USA
| |
Collapse
|
6
|
Gestational exposures to organophosphorus insecticides: From acute poisoning to developmental neurotoxicity. Neuropharmacology 2020; 180:108271. [PMID: 32814088 DOI: 10.1016/j.neuropharm.2020.108271] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/03/2020] [Accepted: 08/10/2020] [Indexed: 11/22/2022]
Abstract
For over three-quarters of a century, organophosphorus (OP) insecticides have been ubiquitously used in agricultural, residential, and commercial settings and in public health programs to mitigate insect-borne diseases. Their broad-spectrum insecticidal effectiveness is accounted for by the irreversible inhibition of acetylcholinesterase (AChE), the enzyme that catalyzes acetylcholine (ACh) hydrolysis, in the nervous system of insects. However, because AChE is evolutionarily conserved, OP insecticides are also toxic to mammals, including humans, and acute OP intoxication remains a major public health concern in countries where OP insecticide usage is poorly regulated. Environmental exposures to OP levels that are generally too low to cause marked inhibition of AChE and to trigger acute signs of intoxication, on the other hand, represent an insidious public health issue worldwide. Gestational exposures to OP insecticides are particularly concerning because of the exquisite sensitivity of the developing brain to these insecticides. The present article overviews and discusses: (i) the health effects and therapeutic management of acute OP poisoning during pregnancy, (ii) epidemiological studies examining associations between environmental OP exposures during gestation and health outcomes of offspring, (iii) preclinical evidence that OP insecticides are developmental neurotoxicants, and (iv) potential mechanisms underlying the developmental neurotoxicity of OP insecticides. Understanding how gestational exposures to different levels of OP insecticides affect pregnancy and childhood development is critical to guiding implementation of preventive measures and direct research aimed at identifying effective therapeutic interventions that can limit the negative impact of these exposures on public health.
Collapse
|
7
|
Farkhondeh T, Mehrpour O, Forouzanfar F, Roshanravan B, Samarghandian S. Oxidative stress and mitochondrial dysfunction in organophosphate pesticide-induced neurotoxicity and its amelioration: a review. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:24799-24814. [PMID: 32358751 DOI: 10.1007/s11356-020-09045-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 04/23/2020] [Indexed: 06/11/2023]
Abstract
Organophosphorus pesticides (OPs) are widely used for controlling pests worldwide. The inhibitory effects of these pesticides on acetylcholinesterase lead to neurotoxic damages. The oxidative stress is responsible for several neurological diseases, including Parkinson's disease, seizure, depression, and Alzheimer's disease. Strong evidence suggests that dysfunction of mitochondria and oxidative stress are involved in neurological diseases. OPs can disturb the function of mitochondria by inducing oxidative stress. In the present study, we tried to highlight the role of dysfunction of mitochondria and the induction of oxidative stress in the neurotoxicity induced by OPs. Additionally, the amelioration of OP-induced oxidative damage and mitochondrial dysfunctional through the chemical and natural antioxidants have been discussed.
Collapse
Affiliation(s)
- Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Omid Mehrpour
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences(BUMS), Birjand, Iran
- Rocky Mountain Poison and Drug Safety, Denver Health, Denver, CO, USA
| | - Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Babak Roshanravan
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
8
|
Liu L, Koo Y, Russell T, Gay E, Li Y, Yun Y. Three-dimensional brain-on-chip model using human iPSC-derived GABAergic neurons and astrocytes: Butyrylcholinesterase post-treatment for acute malathion exposure. PLoS One 2020; 15:e0230335. [PMID: 32163499 PMCID: PMC7067464 DOI: 10.1371/journal.pone.0230335] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/26/2020] [Indexed: 12/24/2022] Open
Abstract
Organophosphates (OPs) induce acute and chronic neurotoxicity, primarily by inhibiting acetylcholinesterase (AChE) activity as well as by necrosis, and apoptosis. Butyrylcholinesterase (BuChE), an exogenous bioscavenger of OPs, can be used as a treatment for OP exposure. It is prerequisite to develop in vitro brain models that can study BuChE post-treatment for acute OP exposure. In this study, we developed a three-dimensional (3D) brain-on-chip platform with human induced pluripotent stem cell (iPSC)-derived neurons and astrocytes to simulate human brain behavior. The platform consists of two compartments: 1) a hydrogel embedded with human iPSC-derived GABAergic neurons and astrocytes and 2) a perfusion channel with dynamic medium flow. The brain tissue constructs were exposed to Malathion (MT) at various concentrations and then treated with BuChE after 20 minutes of MT exposure. Results show that the iPSC-derived neurons and astrocytes directly interacted and formed synapses in the 3D matrix, and that treatment with BuChE improved viability after MT exposure up to a concentration of 10−3 M. We conclude that the 3D brain-on-chip platform with human iPSC-derived brain cells is a suitable model to study the neurotoxicity of OP exposure and evaluate therapeutic compounds for treatment.
Collapse
Affiliation(s)
- Lumei Liu
- FIT BEST Laboratory, Department of Chemical, Biological, and Bio Engineering, North Carolina Agricultural and Technical State University, Greensboro, North Carolina, United States of America
| | - Youngmi Koo
- FIT BEST Laboratory, Department of Chemical, Biological, and Bio Engineering, North Carolina Agricultural and Technical State University, Greensboro, North Carolina, United States of America
| | - Teal Russell
- FIT BEST Laboratory, Department of Chemical, Biological, and Bio Engineering, North Carolina Agricultural and Technical State University, Greensboro, North Carolina, United States of America
| | - Elaine Gay
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, North Carolina, United States of America
| | - Yan Li
- Chemical Engineering, Florida A&M University-Florida State University, Tallahassee, Florida, United States of America
| | - Yeoheung Yun
- FIT BEST Laboratory, Department of Chemical, Biological, and Bio Engineering, North Carolina Agricultural and Technical State University, Greensboro, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
9
|
Gonzalez A. Antioxidants and Neuron-Astrocyte Interplay in Brain Physiology: Melatonin, a Neighbor to Rely on. Neurochem Res 2020; 46:34-50. [PMID: 31989469 DOI: 10.1007/s11064-020-02972-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 12/19/2022]
Abstract
This manuscript is a review focused onto the role of astrocytes in the protection of neurons against oxidative stress and how melatonin can contribute to the maintenance of brain homeostasis. The first part of the review is dedicated to the dependence of neurons on astrocytes by terms of survival under oxidative stress conditions. Additionally, the effects of melatonin against oxidative stress in the brain and its putative role in the protection against diseases affecting the brain are highlighted. The effects of melatonin on the physiology of neurons and astrocytes also are reviewed.
Collapse
Affiliation(s)
- Antonio Gonzalez
- Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, Avenida de las Ciencias s/n, 10003, Cáceres, Spain.
| |
Collapse
|
10
|
Braidy N, Zarka M, Jugder BE, Welch J, Jayasena T, Chan DKY, Sachdev P, Bridge W. The Precursor to Glutathione (GSH), γ-Glutamylcysteine (GGC), Can Ameliorate Oxidative Damage and Neuroinflammation Induced by Aβ 40 Oligomers in Human Astrocytes. Front Aging Neurosci 2019; 11:177. [PMID: 31440155 PMCID: PMC6694290 DOI: 10.3389/fnagi.2019.00177] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
Glutathione (GSH) is one of the most abundant thiol antioxidants in cells. Many chronic and age-related diseases are associated with a decline in cellular GSH levels or impairment in the catalytic activity of the GSH biosynthetic enzyme glutamate cysteine ligase (GCL). γ-glutamylcysteine (GGC), a precursor to glutathione (GSH), can replenish depleted GSH levels under oxidative stress conditions, by circumventing the regulation of GSH biosynthesis and providing the limiting substrate. Soluble amyloid-β (Aβ) oligomers have been shown to induce oxidative stress, synaptic dysfunction and memory deficits which have been reported in Alzheimer’s disease (AD). Calcium ions, which are increased with age and in AD, have been previously reported to enhance the formation of Aβ40 oligomers, which have been casually associated with the pathogenesis of the underlying neurodegenerative condition. In this study, we examined the potential beneficial effects of GGC against exogenous Aβ40 oligomers on biomarkers of apoptosis and cell death, oxidative stress, and neuroinflammation, in human astrocytes. Treatment with Aβ40 oligomers significantly reduced the cell viability and apoptosis of astrocyte brain cultures and increased oxidative modifications of DNA, lipids, and protein, enhanced pro-inflammatory cytokine release and increased the activity of the proteolytic matrix metalloproteinase enzyme, matric metalloproteinase (MMP)-2 and reduced the activity of MMP-9 after 24 h. Co-treatment of Aβ40 oligomers with GGC at 200 μM increased the activity of the antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GPx) and led to significant increases in the levels of the total antioxidant capacity (TAC) and GSH and reduced the GSSG/GSH ratio. GGC also upregulated the level of the anti-inflammatory cytokine IL-10 and reduced the levels of the pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) and attenuated the changes in metalloproteinase activity in oligomeric Aβ40-treated astrocytes. Our data provides renewed insight on the beneficial effects of increased GSH levels by GGC in human astrocytes, and identifies yet another potential therapeutic strategy to attenuate the cytotoxic effects of Aβ oligomers in AD.
Collapse
Affiliation(s)
- Nady Braidy
- Centre for Healthy Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Martin Zarka
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, NSW, Australia
| | - Bat-Erdene Jugder
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, NSW, Australia
| | - Jeffrey Welch
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, NSW, Australia
| | - Tharusha Jayasena
- Centre for Healthy Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Daniel K Y Chan
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,Department of Aged Care and Rehabilitation, Bankstown Hospital, Bankstown, NSW, Australia
| | - Perminder Sachdev
- Neuropsychiatric Institute, Euroa Centre, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Wallace Bridge
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
11
|
Effects of astrocyte conditioned medium on neuronal AChE expression upon 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure. Chem Biol Interact 2019; 309:108686. [DOI: 10.1016/j.cbi.2019.05.052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/24/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022]
|
12
|
Abdel-Daim MM, Abushouk AI, Alkhalf MI, Toraih EA, Fawzy MS, Ijaz H, Aleya L, Bungau SG. Antagonistic effects of Spirulina platensis on diazinon-induced hemato-biochemical alterations and oxidative stress in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:27463-27470. [PMID: 30043347 DOI: 10.1007/s11356-018-2761-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 07/13/2018] [Indexed: 06/08/2023]
Abstract
Spirulina platensis (SP) is a traditionally used microalga for a wide range of pharmacological activities, including amelioration of heavy metals and pesticides toxicity. This study evaluated the antioxidant and organoprotective effects of SP against diazinon (DZN)-induced subacute toxicity on the blood, heart, liver, and kidneys of male Wistar albino rats. Diazinon (20 mg/kg, subcutaneous) was administered to animals either alone or along with an oral pure SP powder at doses of 500 and 1000 mg/kg. Alterations in hematological and serum biochemical parameters, as well as oxidative stress markers in the hepatic, renal, and cardiac tissues were evaluated, using colorimetric spectrophotometric techniques. The obtained results revealed that in comparison to the control group, DZN-treated rats exhibited significantly lower (p < 0.05) red blood cells and platelets counts, hemoglobin and hematocrit values, and activities of serum acetylcholinesterase and tissue antioxidant enzymes (glutathione peroxidase, superoxide dismutase, and catalase). Meanwhile, biochemical analysis showed significantly higher (p < 0.05) white blood cells count, serum concentrations of tumor necrosis factor-α and cardiac [creatine kinase (CK) and CK-muscle/brain fraction], hepatic [transaminases and alkaline phosphatase], and renal [uric acid, urea and creatinine] injury markers, and tissue levels of malondialdehyde (a marker of lipid peroxidation) in the DZN-intoxicated group, compared to normal controls. Interestingly, the administration of SP significantly ameliorated the previous hemato-biochemical alterations and mitigated DZN-induced organ injuries and oxidative stress. In conclusion, the natural antioxidant microalga (SP) effectively alleviated the DZN-induced hematologic alterations and organ injuries, probably through its antioxidant and anti-inflammatory activities.
Collapse
Affiliation(s)
- Mohamed M Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| | | | - Maha I Alkhalf
- Biochemistry Department, Faculty of Science, Al Faisaliah King Abdulaziz University, Jeddah, Saudi Arabia
| | - Eman A Toraih
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
- Center of Excellence of Molecular and Cellular Medicine, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Manal S Fawzy
- Department of Medical Biochemistry, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Hira Ijaz
- College of Pharmacy, Government College University, Faisalabad, Pakistan
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, 25030 11, Besançon Cedex, France
| | - Simona G Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
13
|
Chen H, Streifel KM, Singh V, Yang D, Mangini L, Wulff H, Lein PJ. From the Cover: BDE-47 and BDE-49 Inhibit Axonal Growth in Primary Rat Hippocampal Neuron-Glia Co-Cultures via Ryanodine Receptor-Dependent Mechanisms. Toxicol Sci 2018; 156:375-386. [PMID: 28003438 DOI: 10.1093/toxsci/kfw259] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are widespread environmental contaminants associated with adverse neurodevelopmental outcomes in children and preclinical models; however, the mechanisms by which PBDEs cause developmental neurotoxicity remain speculative. The structural similarity between PBDEs and nondioxin-like (NDL) polychlorinated biphenyls (PCBs) suggests shared toxicological properties. Consistent with this, both NDL PCBs and PBDEs have been shown to stabilize ryanodine receptors (RyRs) in the open configuration. NDL PCB effects on RyR activity are causally linked to increased dendritic arborization, but whether PBDEs similarly enhance dendritic growth is not known. In this study, we quantified the effects of individual PBDE congeners on not only dendritic but also axonal growth since both are regulated by RyR-dependent mechanisms, and both are critical determinants of neuronal connectivity. Neuronal-glial co-cultures dissociated from the neonatal rat hippocampus were exposed to BDE-47 or BDE-49 in the culture medium. At concentrations ranging from 20 pM to 2 µM, neither PBDE congener altered dendritic arborization. In contrast, at concentrations ≥ 200 pM, both congeners delayed neuronal polarization resulting in significant inhibition of axonal outgrowth during the first few days in vitro. The axon inhibitory effects of these PBDE congeners occurred independent of cytotoxicity, and were blocked by pharmacological antagonism of RyR or siRNA knockdown of RyR2. These results demonstrate that the molecular and cellular mechanisms by which PBDEs interfere with neurodevelopment overlap with but are distinct from those of NDL PCBs, and suggest that altered patterns of neuronal connectivity may contribute to the developmental neurotoxicity of PBDEs.
Collapse
Affiliation(s)
- Hao Chen
- Department of Molecular Biosciences, School of Veterinary Medicine
| | - Karin M Streifel
- Department of Molecular Biosciences, School of Veterinary Medicine
| | - Vikrant Singh
- Department of Pharmacology, School of Medicine, University of California-Davis, Davis, California 95616
| | - Dongren Yang
- Department of Molecular Biosciences, School of Veterinary Medicine
| | - Linley Mangini
- Department of Molecular Biosciences, School of Veterinary Medicine
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California-Davis, Davis, California 95616
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine
| |
Collapse
|
14
|
Saraji M, Jafari MT, Amooshahi MM. Sol-gel/nanoclay composite as a sorbent for microextraction in packed syringe combined with corona discharge ionization ion mobility spectrometry for the determination of diazinon in water samples. J Sep Sci 2017; 41:493-500. [DOI: 10.1002/jssc.201700967] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/16/2017] [Accepted: 10/16/2017] [Indexed: 02/04/2023]
Affiliation(s)
- Mohammad Saraji
- Department of Chemistry; Isfahan University of Technology; Isfahan Iran
| | | | | |
Collapse
|
15
|
Vahidirad M, Arab-Nozari M, Mohammadi H, Zamani E, Shaki F. Protective effect of captopril against diazinon induced nephrotoxicity and neurotoxicity via inhibition of ROS-NO pathway. Drug Chem Toxicol 2017; 41:287-293. [PMID: 29115169 DOI: 10.1080/01480545.2017.1391830] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Diazinon (Dz) is a widely used insecticide. It can induce nephrotoxicity and neurotoxicity via oxidative stress. Captopril, an angiotensin-converting enzyme inhibitor, is known for its antioxidant properties. In this study, we used captopril for ameliorating of Dz-induced kidney and brain toxicity in rats. Animals were divided into five groups as follows: negative control (olive oil), Dz (150 mg kg-1), captopril (60 and 100 mg kg-1) and positive control (N-acetylcysteine 200 mg kg-1) were injected intraperitoneally 30 min before Dz. After 24 h, animals were anesthetized and the brain and kidney tissues were separated. Then oxidative stress factors were evaluated. Also, blood was collected for assessment of blood urea nitrogen (BUN), creatinine (Cr) and nitric oxide (NO) levels. Dz significantly increased oxidative stress markers such as reactive oxygen species (ROS), lipid peroxidation, and protein carbonyl as well as glutathione (GSH) oxidation in both tissues. Increased levels of the BUN, Cr and NO were observed after Dz injection. Interestingly, captopril administration significantly decreased ROS production in both tissues. Captopril significantly protected kidney and brain against lipid peroxidation and GSH oxidation. Administration of captopril could markedly inhibit protein carbonyl production in kidney and brain after Dz injection. Furthermore, captopril ameliorated the increased level of BUN, Cr and NO. These results suggested that captopril can prevent Dz-induced oxidative stress, nephrotoxicity and neurotoxicity because of its antioxidant activity.
Collapse
Affiliation(s)
- Milad Vahidirad
- a Department of Toxicology and Pharmacology, Faculty of Pharmacy , Pharmaceutical Sciences Branch, Islamic Azad University , Tehran , Iran
| | - Milad Arab-Nozari
- b Department of Toxicology and Pharmacology, Faculty of Pharmacy , Mazandaran University of Medical Sciences , Sari , Iran
| | - Hamidreza Mohammadi
- b Department of Toxicology and Pharmacology, Faculty of Pharmacy , Mazandaran University of Medical Sciences , Sari , Iran
| | - Ehsan Zamani
- b Department of Toxicology and Pharmacology, Faculty of Pharmacy , Mazandaran University of Medical Sciences , Sari , Iran
| | - Fatemeh Shaki
- b Department of Toxicology and Pharmacology, Faculty of Pharmacy , Mazandaran University of Medical Sciences , Sari , Iran
| |
Collapse
|
16
|
Gleixner AM, Hutchison DF, Sannino S, Bhatia TN, Leak LC, Flaherty PT, Wipf P, Brodsky JL, Leak RK. N-Acetyl-l-Cysteine Protects Astrocytes against Proteotoxicity without Recourse to Glutathione. Mol Pharmacol 2017; 92:564-575. [PMID: 28830914 DOI: 10.1124/mol.117.109926] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023] Open
Abstract
N-acetyl-l-cysteine (NAC) exhibits protective properties in brain injury models and has undergone a number of clinical trials. Most studies of NAC have focused on neurons. However, neuroprotection may be complemented by the protection of astrocytes because healthier astrocytes can better support the viability of neurons. Here, we show that NAC can protect astrocytes against protein misfolding stress (proteotoxicity), the hallmark of neurodegenerative disorders. Although NAC is thought to be a glutathione precursor, NAC protected primary astrocytes from the toxicity of the proteasome inhibitor MG132 without eliciting any increase in glutathione. Furthermore, glutathione depletion failed to attenuate the protective effects of NAC. MG132 elicited a robust increase in the folding chaperone heat shock protein 70 (Hsp70), and NAC mitigated this effect. Nevertheless, three independent inhibitors of Hsp70 function ablated the protective effects of NAC, suggesting that NAC may help preserve Hsp70 chaperone activity and improve protein quality control without need for Hsp70 induction. Consistent with this view, NAC abolished an increase in ubiquitinated proteins in MG132-treated astrocytes. However, NAC did not affect the loss of proteasome activity in response to MG132, demonstrating that it boosted protein homeostasis and cell viability without directly interfering with the efficacy of this proteasome inhibitor. The thiol-containing molecules l-cysteine and d-cysteine both mimicked the protective effects of NAC, whereas the thiol-lacking molecule N-acetyl-S-methyl-l-cysteine failed to exert protection or blunt the rise in ubiquitinated proteins. Collectively, these findings suggest that the thiol group in NAC is required for its effects on glial viability and protein quality control.
Collapse
Affiliation(s)
- Amanda M Gleixner
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Daniel F Hutchison
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Sara Sannino
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Tarun N Bhatia
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Lillian C Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Patrick T Flaherty
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Peter Wipf
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Jeffrey L Brodsky
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| |
Collapse
|
17
|
Wang YH, Chang SH, Mau-Hsu D, Wang JH, Young TH. Poly(N-(4-aminobutyl)-acrylamide) as mimetic polylysine for improving survival and differentiation of cerebellar granule neurons. J Biomed Mater Res B Appl Biomater 2017; 106:1194-1201. [PMID: 28561950 DOI: 10.1002/jbm.b.33932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 04/11/2017] [Accepted: 05/12/2017] [Indexed: 11/06/2022]
Abstract
Poly(N-(4-aminobutyl)-acrylamide) (P4Am) and poly-d-lysine (PDL) possess the same butylamine side chain. The main difference is the peptide structure, which is in the main chain of PDL but in the side chain of P4Am. PDL has been extensively used in the preparation of neuronal cultures. We assumed neurons are sensitive enough to distinguish such structure difference so these two cationic polymers were compared at serial coating concentrations for culturing cerebellar granule neurons from 7-day-old Wistar rats in this study. Cellular viability and morphology assay showed no obvious difference for neurons cultured at high coating concentrations (>0.31 µg/mL) of these two polymers. In contrast, the difference in the peptide structure between P4Am and PDL could be distinguished by neurons at low coating concentrations (< 0.16 µg/mL). P4Am at low coating concentration could keep aggregates with three or four thick processes to support a more complete neural network with higher cellular viability than PDL. This suggests the interaction between neurons and the specific peptide structure of P4Am at low coating concentration was able to improve survival and differentiation of cultured cerebellar granule neurons. Although neural cells exhibited different morphologies and activities on high and low P4Am coating concentrations, immunofluorescence imaging confirmed most of cultured cells were functionally mature neurons, stained by neurofilament, synapsin I, and GAP43. The information should be useful for designing new biomaterials for regeneration of damaged circuits following disease or trauma. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1194-1201, 2018.
Collapse
Affiliation(s)
- Yu-Hsin Wang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Shao-Hsuan Chang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Daxen Mau-Hsu
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Jyh-Horng Wang
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Tai-Horng Young
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
18
|
Wu X, Yang X, Majumder A, Swetenburg R, Goodfellow FT, Bartlett MG, Stice SL. From the Cover: AstrocytesAre Protective Against Chlorpyrifos Developmental Neurotoxicity in Human Pluripotent Stem Cell-Derived Astrocyte-Neuron Cocultures. Toxicol Sci 2017; 157:410-420. [DOI: 10.1093/toxsci/kfx056] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Xian Wu
- Department of Animal and Dairy Science, Interdisciplinary Toxicology Program
- Department of Animal and Dairy Science, Regenerative Bioscience Center
| | - Xiangkun Yang
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia 30602
| | | | | | - Forrest T. Goodfellow
- Department of Animal and Dairy Science, Interdisciplinary Toxicology Program
- Department of Animal and Dairy Science, Regenerative Bioscience Center
| | - Michael G. Bartlett
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia 30602
| | - Steven L. Stice
- Department of Animal and Dairy Science, Interdisciplinary Toxicology Program
- Department of Animal and Dairy Science, Regenerative Bioscience Center
- ArunA Biomedical, Athens, Georgia 30602
| |
Collapse
|
19
|
Voorhees JR, Rohlman DS, Lein PJ, Pieper AA. Neurotoxicity in Preclinical Models of Occupational Exposure to Organophosphorus Compounds. Front Neurosci 2017; 10:590. [PMID: 28149268 PMCID: PMC5241311 DOI: 10.3389/fnins.2016.00590] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 12/08/2016] [Indexed: 01/06/2023] Open
Abstract
Organophosphorus (OPs) compounds are widely used as insecticides, plasticizers, and fuel additives. These compounds potently inhibit acetylcholinesterase (AChE), the enzyme that inactivates acetylcholine at neuronal synapses, and acute exposure to high OP levels can cause cholinergic crisis in humans and animals. Evidence further suggests that repeated exposure to lower OP levels insufficient to cause cholinergic crisis, frequently encountered in the occupational setting, also pose serious risks to people. For example, multiple epidemiological studies have identified associations between occupational OP exposure and neurodegenerative disease, psychiatric illness, and sensorimotor deficits. Rigorous scientific investigation of the basic science mechanisms underlying these epidemiological findings requires valid preclinical models in which tightly-regulated exposure paradigms can be correlated with neurotoxicity. Here, we review the experimental models of occupational OP exposure currently used in the field. We found that animal studies simulating occupational OP exposures do indeed show evidence of neurotoxicity, and that utilization of these models is helping illuminate the mechanisms underlying OP-induced neurological sequelae. Still, further work is necessary to evaluate exposure levels, protection methods, and treatment strategies, which taken together could serve to modify guidelines for improving workplace conditions globally.
Collapse
Affiliation(s)
- Jaymie R. Voorhees
- Department of Psychiatry, University of Iowa Carver College of MedicineIowa City, IA, USA
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa Carver College of MedicineIowa City, IA, USA
| | - Diane S. Rohlman
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa Carver College of MedicineIowa City, IA, USA
- Department of Occupational and Environmental Health, University of Iowa College of Public HealthIowa City, IA, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, DavisDavis, CA, USA
| | - Andrew A. Pieper
- Department of Psychiatry, University of Iowa Carver College of MedicineIowa City, IA, USA
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa Carver College of MedicineIowa City, IA, USA
- Department of Neurology, University of Iowa Carver College of MedicineIowa City, IA, USA
- Department of Free Radical and Radiation Biology Program, University of Iowa Carver College of MedicineIowa City, IA, USA
- Department of Radiation Oncology Holden Comprehensive Cancer Center, University of Iowa Carver College of MedicineIowa City, IA, USA
- Department of Veteran Affairs, University of Iowa Carver College of MedicineIowa City, IA, USA
- Weill Cornell Autism Research Program, Weill Cornell Medical CollegeNew York, NY, USA
| |
Collapse
|
20
|
Merwin SJ, Obis T, Nunez Y, Re DB. Organophosphate neurotoxicity to the voluntary motor system on the trail of environment-caused amyotrophic lateral sclerosis: the known, the misknown, and the unknown. Arch Toxicol 2017; 91:2939-2952. [PMID: 28070599 DOI: 10.1007/s00204-016-1926-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 12/20/2016] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common adult-onset paralytic disorder. It is characterized by progressive degeneration of the motor neurons controlling voluntary movement. The underlying mechanisms remain elusive, a fact that has precluded development of effective treatments. ALS presents as a sporadic condition 90-95% of the time, i.e., without familial history or obvious genetic mutation. This suggests that ALS has a strong environmental component. Organophosphates (OPs) are prime candidate neurotoxicants in the etiology of ALS, as exposure to OPs was linked to higher ALS incidence among farmers, soccer players, and Gulf War veterans. In addition, polymorphisms in paraoxonase 1, an enzyme that detoxifies OPs, may increase individual vulnerability both to OP poisoning and to the risk of developing ALS. Furthermore, exposure to high doses of OPs can give rise to OP-induced delayed neuropathy (OPIDN), a debilitating condition akin to ALS characterized by similar motor impairment and paralysis. The question we pose in this review is: "what can we learn from acute exposure to high doses of neurotoxicants (OPIDN) that could help our understanding of chronic diseases resulting from potentially decades of silent exposure (ALS)?" The resemblances between OPIDN and ALS are striking at the clinical, etiological, neuropathological, cellular, and potentially molecular levels. Here, we critically present available evidence, discuss current limitations, and posit future research. In the search for the environmental origin of ALS, OPIDN offers an exciting trail to follow, which can hopefully lead to the development of novel strategies to prevent and cure these dreadful disorders.
Collapse
Affiliation(s)
- Samantha J Merwin
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA.,NIEHS Center for Environmental Health Sciences in Northern Manhattan, Columbia University, New York, NY, 10032, USA.,Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
| | - Teresa Obis
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA.,NIEHS Center for Environmental Health Sciences in Northern Manhattan, Columbia University, New York, NY, 10032, USA.,Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
| | - Yanelli Nunez
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA.,NIEHS Center for Environmental Health Sciences in Northern Manhattan, Columbia University, New York, NY, 10032, USA.,Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA.,Toxicology Ph.D. Program, Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Diane B Re
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA. .,NIEHS Center for Environmental Health Sciences in Northern Manhattan, Columbia University, New York, NY, 10032, USA. .,Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA. .,, 722 W 168th street Suite 1107B, New York, NY, 10032, USA.
| |
Collapse
|
21
|
Shiri M, Navaei-Nigjeh M, Baeeri M, Rahimifard M, Mahboudi H, Shahverdi AR, Kebriaeezadeh A, Abdollahi M. Blockage of both the extrinsic and intrinsic pathways of diazinon-induced apoptosis in PaTu cells by magnesium oxide and selenium nanoparticles. Int J Nanomedicine 2016; 11:6239-6250. [PMID: 27920530 PMCID: PMC5125760 DOI: 10.2147/ijn.s119680] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Diazinon (DZ) is an organophosphorus insecticide that acts as an acetylcholinesterase inhibitor. It is important to note that it can induce oxidative stress, lipid peroxidation, diabetic disorders, and cytotoxicity. Magnesium oxide (MgO) and selenium nanoparticles (Se NPs) showed promising protection against oxidative stress, lipid peroxidation, cytotoxicity, and diabetic disorders. Therefore, this study was conducted to explore the possible protective mechanisms of MgO and Se NPs against DZ-induced cytotoxicity in PaTu cell line. Cytotoxicity of DZ, in the presence or absence of effective doses of MgO and Se NPs, was determined in human pancreatic cancer cell line (PaTu cells) after 24 hours of exposure by using mitochondrial activity and mitochondrial membrane potential assays. Then, the insulin, proinsulin, and C-peptide release; caspase-3 and -9 activities; and total thiol molecule levels were assessed. Determination of cell viability, including apoptotic and necrotic cells, was assessed via acridine orange/ethidium bromide double staining. Furthermore, expression of 15 genes associated with cell death/apoptosis in various phenomena was examined after 24 hours of contact with DZ and NPs by using real-time polymerase chain reaction. Compared to the individual cases, the group receiving the combination of MgO and Se NPs showed more beneficial effects in reducing the toxicity of DZ. Cotreatment of PaTu cell lines with MgO and Se NPs counteracts the toxicity of DZ on insulin-producing cells.
Collapse
Affiliation(s)
- Mahdi Shiri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences; School of Medicine, Artesh University of Medical Sciences
| | - Mona Navaei-Nigjeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Baeeri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences
| | - Mahban Rahimifard
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences
| | - Hossein Mahboudi
- Department of Biotechnology, Faculty of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Shahverdi
- Department of Biotechnology, Faculty of Pharmacy and Biotechnology Research Center
| | - Abbas Kebriaeezadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences; Toxicology Interest Group, USERN; Endocrinology & Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Li Z, Liu Q, Liu C, Li C, Li Y, Li S, Liu X, Shao J. Evaluation of PFOS-mediated neurotoxicity in rat primary neurons and astrocytes cultured separately or in co-culture. Toxicol In Vitro 2016; 38:77-90. [PMID: 27825932 DOI: 10.1016/j.tiv.2016.11.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 01/28/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a potential neurotoxicant reported by epidemiological investigations and experimental studies, while the underlying mechanisms are still unclear. Astrocytes not only support for the construction of neurons, but also conduct neuronal functions through glutamate-glutamine cycle in astrocyte-neuron crosstalk. In the present study, the effect of PFOS exposure on rat primary hippocampal neurons or cortex astrocytes was evaluated. Then the role of the astrocytes in PFOS-induced toxic effect on neurons was explored with astrocyte-neuron co-culture system. Exposure of rat primary hippocampal neurons to PFOS has led to oxidation-antioxidation imbalance, increased apoptosis and abnormal autophagy. The adverse effect of PFOS on rat primary cortex astrocytes manifested in the form of altered extracellular glutamate and glutamine concentrations, decreased glutamine synthase activity, as well as decreased gene expression of glutamine synthase, glutamate transporters and glutamine transporters in the glutamate-glutamine cycle. Especially, the alleviation of PFOS-inhibited neurite outgrowth in neurons could be observed in astrocyte-neuron co-culture system, though the ability of astrocytes in fostering neurite outgrowth was affected by PFOS. These results indicated that both astrocytes and neurons might be the targets of PFOS-induced neurotoxicity, and astrocytes could protect against PFOS-inhibited neurite outgrowth in primary cultured neurons. Our research might render some information in explaining the mechanisms of PFOS-induced neurotoxicity.
Collapse
Affiliation(s)
- Zhenwei Li
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Qi Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Chang Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Chunna Li
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Yachen Li
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Shuangyue Li
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Xiaohui Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China.
| | - Jing Shao
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
23
|
Pearson JN, Patel M. The role of oxidative stress in organophosphate and nerve agent toxicity. Ann N Y Acad Sci 2016; 1378:17-24. [PMID: 27371936 DOI: 10.1111/nyas.13115] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 05/03/2016] [Accepted: 05/05/2016] [Indexed: 01/06/2023]
Abstract
Organophosphate (OP) nerve agents exert their toxicity through inhibition of acetylcholinesterase. The excessive stimulation of cholinergic receptors rapidly causes neuronal damage, seizures, death, and long-term neurological impairment in those that survive. Owing to the lethality of organophosphorus agents and the growing risk they pose, medical interventions that prevent OP toxicity and the delayed injury response are much needed. Studies have shown that oxidative stress occurs in models of subacute, acute, and chronic exposure to OP agents. Key findings of these studies include alterations in mitochondrial function and increased free radical-mediated injury, such as lipid peroxidation. This review focuses on the role of reactive oxygen species in OP neurotoxicity and its dependence on seizure activity. Understanding the sources, mechanisms, and pathological consequences of OP-induced oxidative stress can lead to the development of rational therapies for treating toxic exposures.
Collapse
Affiliation(s)
| | - Manisha Patel
- Neuroscience Program. .,Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
24
|
Ajibade TO, Oyagbemi AA, Omobowale TO, Asenuga ER, Afolabi JM, Adedapo AA. Mitigation of diazinon-induced cardiovascular and renal dysfunction by gallic acid. Interdiscip Toxicol 2016; 9:66-77. [PMID: 28652848 PMCID: PMC5458106 DOI: 10.1515/intox-2016-0008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 03/31/2016] [Accepted: 04/04/2016] [Indexed: 01/20/2023] Open
Abstract
Studies of the link between environmental pollutants and cardiovascular dysfunction, neglected for decades, have recently provided new insights into the pathology and consequences of these killers. In this study, rats were divided into four groups, each containing 10 rats. The rats in group one served as controls and were administered normal saline, whereas the rats in group two were orally gavaged with 3 mg/kg of diazinon (DZN) alone for twenty one consecutive days. The rats in groups 3 and 4 were administered respective 60 mg/kg and 120 mg/kg gallic acid (GA) in addition to DZN for twenty one consecutive days. Exposure of rats to diazinon significantly (p<0.05) reduced the activities of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione-S-transferase (GST) and reduced glutathione (GSH) content. Malondialdehyde, hydrogen peroxide (H2O2) and nitric oxide (NO) contents were also significantly (p<0.05) elevated following DZN exposure. DZN further caused a significant (p<0.05) decrease of heart rate and QT interval prolongation. Hematologic analysis revealed significant reduction (p<0.05) in packed cell volume (PCV), hemoglobin concentration (Hb), red blood cell (RBC) count, and total white blood cell count of rats administered only DZN. Observations in this study suggest a modulatory role of gallic acid in diazinon-induced anemia and associated cardiovascular dysfunction in rats. Treatment with gallic acid reversed the oxidative stress markers studied, increased the antioxidant defence system and reduced deleterious effects on hematological parameters in rats. Pathologic findings of the heart and kidney were also found to be lessened.
Collapse
Affiliation(s)
- Temitayo Olabisi Ajibade
- Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Ademola Adetokunbo Oyagbemi
- Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | | | | | | | - Adeolu Alex Adedapo
- Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| |
Collapse
|
25
|
Switching from astrocytic neuroprotection to neurodegeneration by cytokine stimulation. Arch Toxicol 2016; 91:231-246. [PMID: 27052459 DOI: 10.1007/s00204-016-1702-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/21/2016] [Indexed: 12/12/2022]
Abstract
Astrocytes, the largest cell population in the human brain, are powerful inflammatory effectors. Several studies have examined the interaction of activated astrocytes with neurons, but little is known yet about human neurotoxicity under such situations and about strategies of neuronal rescue. To address this question, immortalized murine astrocytes (IMA) were combined with human LUHMES neurons and stimulated with an inflammatory (TNF, IL-1) cytokine mix (CM). Neurotoxicity was studied both in co-cultures and in monocultures after transfer of conditioned medium from activated IMA. Interventions with >20 drugs were used to profile the model system. Control IMA supported neurons and protected them from neurotoxicants. Inflammatory activation reduced this protection, and prolonged exposure of co-cultures to CM triggered neurotoxicity. Neither the added cytokines nor the release of NO from astrocytes were involved in this neurodegeneration. The neurotoxicity-mediating effect of IMA was faithfully reproduced by human astrocytes. Moreover, glia-dependent toxicity was also observed, when IMA cultures were stimulated with CM, and the culture medium was transferred to neurons. Such neurotoxicity was prevented when astrocytes were treated by p38 kinase inhibitors or dexamethasone, whereas such compounds had no effect when added to neurons. Conversely, treatment of neurons with five different drugs, including resveratrol and CEP1347, prevented toxicity of astrocyte supernatants. Thus, the sequential IMA-LUHMES neuroinflammation model is suitable for separate profiling of both glial-directed and directly neuroprotective strategies. Moreover, direct evaluation in co-cultures of the same cells allows for testing of therapeutic effectiveness in more complex settings, in which astrocytes affect pharmacological properties of neurons.
Collapse
|
26
|
Degradation of organophosphorus pesticide diazinon using activated persulfate: Optimization of operational parameters and comparative study by Taguchi's method. J Taiwan Inst Chem Eng 2015. [DOI: 10.1016/j.jtice.2015.05.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
27
|
Kubik LL, Philbert MA. The role of astrocyte mitochondria in differential regional susceptibility to environmental neurotoxicants: tools for understanding neurodegeneration. Toxicol Sci 2015; 144:7-16. [PMID: 25740792 DOI: 10.1093/toxsci/kfu254] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In recent decades, there has been a significant expansion in our understanding of the role of astrocytes in neuroprotection, including spatial buffering of extracellular ions, secretion of metabolic coenzymes, and synaptic regulation. Astrocytic neuroprotective functions require energy, and therefore require a network of functional mitochondria. Disturbances to astrocytic mitochondrial homeostasis and their ability to produce ATP can negatively impact neural function. Perturbations in astrocyte mitochondrial function may accrue as the result of physiological aging processes or as a consequence of neurotoxicant exposure. Hydrophobic environmental neurotoxicants, such as 1,3-dinitrobenzene and α-chlorohydrin, cause regionally specific spongiform lesions mimicking energy deprivation syndromes. Astrocyte involvement includes mitochondrial damage that either precedes or is accompanied by neuronal damage. Similarly, environmental neurotoxicants that are implicated in the etiology of age-related neurodegenerative conditions cause regionally specific damage in the brain. Based on the regioselective nature of age-related neurodegenerative lesions, chemically induced models of regioselective lesions targeting astrocyte mitochondria can provide insight into age-related susceptibilities in astrocyte mitochondria. Most of the available research to date focuses on neuronal damage in cases of age-related neurodegeneration; however, there is a body of evidence that supports a central mechanistic role for astrocyte mitochondria in the expression of neural injury. Regional susceptibility to neuronal damage induced by aging by exposure to neurotoxicants may be a reflection of highly variable regional energy requirements. This review identifies region-specific vulnerabilities in astrocyte mitochondria in examples of exposure to neurotoxicants and in age-related neurodegeneration.
Collapse
Affiliation(s)
- Laura L Kubik
- Toxicology Program, Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109
| | - Martin A Philbert
- Toxicology Program, Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
28
|
Ferulic Acid Protects Against Lead Acetate-Induced Inhibition of Neurite Outgrowth by Upregulating HO-1 in PC12 Cells: Involvement of ERK1/2-Nrf2 Pathway. Mol Neurobiol 2015; 53:6489-6500. [DOI: 10.1007/s12035-015-9555-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/19/2015] [Indexed: 12/28/2022]
|
29
|
Efremova L, Schildknecht S, Adam M, Pape R, Gutbier S, Hanf B, Bürkle A, Leist M. Prevention of the degeneration of human dopaminergic neurons in an astrocyte co-culture system allowing endogenous drug metabolism. Br J Pharmacol 2015; 172:4119-32. [PMID: 25989025 DOI: 10.1111/bph.13193] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 05/04/2015] [Accepted: 05/12/2015] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND AND PURPOSE Few neuropharmacological model systems use human neurons. Moreover, available test systems rarely reflect functional roles of co-cultured glial cells. There is no human in vitro counterpart of the widely used 1-methyl-4-phenyl-tetrahydropyridine (MPTP) mouse model of Parkinson's disease EXPERIMENTAL APPROACH We generated such a model by growing an intricate network of human dopaminergic neurons on a dense layer of astrocytes. In these co-cultures, MPTP was metabolized to 1-methyl-4-phenyl-pyridinium (MPP(+) ) by the glial cells, and the toxic metabolite was taken up through the dopamine transporter into neurons. Cell viability was measured biochemically and by quantitative neurite imaging, siRNA techniques were also used. KEY RESULTS We initially characterized the activation of PARP. As in mouse models, MPTP exposure induced (poly-ADP-ribose) synthesis and neurodegeneration was blocked by PARP inhibitors. Several different putative neuroprotectants were then compared in mono-cultures and co-cultures. Rho kinase inhibitors worked in both models; CEP1347, ascorbic acid or a caspase inhibitor protected mono-cultures from MPP(+) toxicity, but did not protect co-cultures, when used alone or in combination. Application of GSSG prevented degeneration in co-cultures, but not in mono-cultures. The surprisingly different pharmacological profiles of the models suggest that the presence of glial cells, and the in situ generation of the toxic metabolite MPP(+) within the layered cultures played an important role in neuroprotection. CONCLUSIONS AND IMPLICATIONS Our new model system is a closer model of human brain tissue than conventional cultures. Its use for screening of candidate neuroprotectants may increase the predictiveness of a test battery.
Collapse
Affiliation(s)
- Liudmila Efremova
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany.,Research Training Group 1331 (RTG1331), University of Konstanz, Konstanz, Germany
| | - Stefan Schildknecht
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Martina Adam
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Regina Pape
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Simon Gutbier
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany.,Research Training Group 1331 (RTG1331), University of Konstanz, Konstanz, Germany
| | - Benjamin Hanf
- Research Training Group 1331 (RTG1331), University of Konstanz, Konstanz, Germany.,Molecular Toxicology Group, University of Konstanz, Konstanz, Germany
| | - Alexander Bürkle
- Molecular Toxicology Group, University of Konstanz, Konstanz, Germany
| | - Marcel Leist
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| |
Collapse
|
30
|
Glutathione-Dependent Detoxification Processes in Astrocytes. Neurochem Res 2014; 40:2570-82. [PMID: 25428182 DOI: 10.1007/s11064-014-1481-1] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 11/10/2014] [Accepted: 11/15/2014] [Indexed: 01/17/2023]
Abstract
Astrocytes have a pivotal role in brain as partners of neurons in homeostatic and metabolic processes. Astrocytes also protect other types of brain cells against the toxicity of reactive oxygen species and are considered as first line of defence against the toxic potential of xenobiotics. A key component in many of the astrocytic detoxification processes is the tripeptide glutathione (GSH) which serves as electron donor in the GSH peroxidase-catalyzed reduction of peroxides. In addition, GSH is substrate in the detoxification of xenobiotics and endogenous compounds by GSH-S-transferases which generate GSH conjugates that are efficiently exported from the cells by multidrug resistance proteins. Moreover, GSH reacts with the reactive endogenous carbonyls methylglyoxal and formaldehyde to intermediates which are substrates of detoxifying enzymes. In this article we will review the current knowledge on the GSH metabolism of astrocytes with a special emphasis on GSH-dependent detoxification processes.
Collapse
|
31
|
Ultrafine carbon black induces glutamate and ATP release by activating connexin and pannexin hemichannels in cultured astrocytes. Toxicology 2014; 323:32-41. [PMID: 24932759 DOI: 10.1016/j.tox.2014.06.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 05/21/2014] [Accepted: 06/11/2014] [Indexed: 12/22/2022]
Abstract
Ultrafine particles could enter central nervous system and were associated with brain damage. The underlying mechanisms have not been fully elucidated. Glutamate and ATP are important signaling molecules in brain physiology and pathology. We investigated whether ultrafine carbon black (ufCB) could regulate the release of glutamate and ATP from cultured cortical astrocytes and the involvement of hemichannels in the release mechanism. Our results showed that ufCB dose-dependently increased glutamate and ATP release and activated hemichannels in astrocytes. ufCB-activated hemichannels were attributed to the activation of both connexin 43 (Cx43) and pannexin1 (Panx1) hemichannels, which was based on the finding of increased protein expression and distribution on cell surface of Cx43 and Panx1, and the inhibiting effects of hemichannel inhibitor carbenoxolone, Cx43 hemichannel inhibitor (43)Gap27 and Panx1 hemichannel inhibitor (10)Panx1 on hemichannel activation. Furthermore, ufCB-induced glutamate and ATP release were dependent on Cx43 and Panx1 hemichannels, because carbenoxolone and (43)Gap27 inhibited ufCB-induced glutamate and ATP release, and (10)Panx1 inhibited ufCB-induced ATP release. Taken together, we demonstrated, for the first time, that ufCB could induce glutamate and ATP release by activating Cx43 and Panx1 hemchannels in astrocytes. Our findings suggest a novel mechanism for neurotoxicity caused by ultrafine particles.
Collapse
|