1
|
van Melis LVJ, Peerdeman AM, Huiberts EHW, van Kleef RGDM, de Groot A, Westerink RHS. Effects of acute insecticide exposure on neuronal activity in vitro in rat cortical cultures. Neurotoxicology 2024; 102:58-67. [PMID: 38599286 DOI: 10.1016/j.neuro.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/13/2024] [Accepted: 04/08/2024] [Indexed: 04/12/2024]
Abstract
Exposure to pesticides, such as carbamates, organophosphates, organochlorines and pyrethroids, has been linked to various health problems, including neurotoxicity. Although most in vivo studies use only male rodents, some studies have shown in vivo sex-specific effects after acute exposure. Since in vivo studies are costly and require a large number of animals, in vitro assays that take sex-specific effects into account are urgently needed. We therefore assessed the acute effects of exposure to different carbamates (methomyl, aldicarb and carbaryl), organophosphates (chlorpyrifos (CPF), chlorpyrifos-oxon (CPO) and 3,5,6-trichloropyridinol), organochlorines (endosulfan, dieldrin and lindane) and pyrethroids (permethrin, alpha-cypermethrin and 3-phenoxy-benzoic acid (3-PBA)) on neuronal network function in sex-separated rat primary cortical cultures using micro-electrode array (MEA) recordings. Our results indicate that exposure to the carbamate carbaryl and the organophosphates CPF and CPO decreased neuronal activity, with CPO being the most potent. Notably, (network) burst patterns differed between CPF and CPO, with CPO inducing fewer, but more intense (network) bursts. Exposure to low micromolar levels of endosulfan induced a hyperexcitation, most likely due to the antagonistic effects on GABA receptors. Interestingly, females were more sensitive to endosulfan than males. Exposure to dieldrin and lindane also increased neuronal activity, albeit less than endosulfan and without sex-specific effects. Exposure to type I pyrethroid permethrin increased neuronal activity, while exposure to type II pyrethroid alpha-cypermethrin strongly decreased neuronal activity. The increase seen after permethrin exposure was more pronounced in males than in females. Together, these results show that acute exposure to different classes of pesticides exerts differential effects on neuronal activity. Moreover, it shows that MEA recordings are suited to detect sex-specific neurotoxic effects in vitro.
Collapse
Affiliation(s)
- Lennart V J van Melis
- Neurotoxicology Research Group, Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, TD, Utrecht NL-3508, the Netherlands
| | - Anneloes M Peerdeman
- Neurotoxicology Research Group, Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, TD, Utrecht NL-3508, the Netherlands
| | - Eva H W Huiberts
- Neurotoxicology Research Group, Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, TD, Utrecht NL-3508, the Netherlands
| | - Regina G D M van Kleef
- Neurotoxicology Research Group, Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, TD, Utrecht NL-3508, the Netherlands
| | - Aart de Groot
- Neurotoxicology Research Group, Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, TD, Utrecht NL-3508, the Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, TD, Utrecht NL-3508, the Netherlands..
| |
Collapse
|
2
|
Sánchez-Yépez J, Acevedo-Huergo T, Mendoza-Trejo MS, Corona R, Hernández-Plata I, Viñuela-Berni V, Giordano M, Rodríguez VM. Early and transitory hypoactivity and olfactory alterations after chronic atrazine exposure in female Sprague-Dawley rats. Neurotoxicology 2024; 101:68-81. [PMID: 38340903 DOI: 10.1016/j.neuro.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/19/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
Several studies have shown that chronic exposure to the herbicide atrazine (ATR) causes alterations in locomotor activity and markers of the dopaminergic systems of male rats. However, few studies have evaluated the sex-dependent effects of atrazine exposure. The aim of the present study was to evaluate whether chronic ATR exposure causes alterations in behavioral performance and dopaminergic systems of female rats. At weaning, two groups of rats were exposed to 1 or 10 mg ATR/kg body weight daily thorough the food, while the control group received food without ATR for 14 months. Spontaneous locomotor activity was evaluated monthly for 12 months, while anxiety, egocentric and spatial memory, motor coordination, and olfactory function tasks were evaluated between 13 and 14 months of ATR exposure. Tyrosine hydroxylase (TH) and monoamine content in brain tissue were assessed at the end of ATR treatment. Female rats treated with 1 or 10 mg ATR showed vertical hypoactivity compared to the control group only in the first month of ATR exposure. Impairments in olfactory functions were found due to ATR exposure. Nevertheless, no alterations in anxiety, spatial and egocentric memory, or motor coordination tasks were observed, while the levels of TH and dopamine and its metabolites in brain tissue were similar among groups. These results suggest that female rats could present greater sensitivity to the neurotoxic effects of ATR on spontaneous locomotor activity in the early stages of development. However, they are unaffected by chronic ATR exposure later in life compared to male rats. More studies are necessary to unravel the sex-related differences observed after chronic ATR exposure.
Collapse
Affiliation(s)
- Jonathan Sánchez-Yépez
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Triana Acevedo-Huergo
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Maria Soledad Mendoza-Trejo
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Rebeca Corona
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Isela Hernández-Plata
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Verónica Viñuela-Berni
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Magda Giordano
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Verónica M Rodríguez
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico.
| |
Collapse
|
3
|
Rocha SM, Kirkley KS, Chatterjee D, Aboellail TA, Smeyne RJ, Tjalkens RB. Microglia-specific knock-out of NF-κB/IKK2 increases the accumulation of misfolded α-synuclein through the inhibition of p62/sequestosome-1-dependent autophagy in the rotenone model of Parkinson's disease. Glia 2023; 71:2154-2179. [PMID: 37199240 PMCID: PMC10330367 DOI: 10.1002/glia.24385] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/19/2023]
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder worldwide, with a greater prevalence in men than women. The etiology of PD is largely unknown, although environmental exposures and neuroinflammation are linked to protein misfolding and disease progression. Activated microglia are known to promote neuroinflammation in PD, but how environmental agents interact with specific innate immune signaling pathways in microglia to stimulate conversion to a neurotoxic phenotype is not well understood. To determine how nuclear factor kappa B (NF-κB) signaling dynamics in microglia modulate neuroinflammation and dopaminergic neurodegeneration, we generated mice deficient in NF-κB activation in microglia (CX3CR1-Cre::IKK2fl/fl ) and exposed them to 2.5 mg/kg/day of rotenone for 14 days, followed by a 14-day post-lesioning incubation period. We postulated that inhibition of NF-κB signaling in microglia would reduce overall inflammatory injury in lesioned mice. Subsequent analysis indicated decreased expression of the NF-κB-regulated autophagy gene, sequestosome 1 (p62), in microglia, which is required for targeting ubiquitinated α-synuclein (α-syn) for lysosomal degradation. Knock-out animals had increased accumulation of misfolded α-syn within microglia, despite an overall reduction in neurodegeneration. Interestingly, this occurred more prominently in males. These data suggest that microglia play key biological roles in the degradation and clearance of misfolded α-syn and this process works in concert with the innate immune response associated with neuroinflammation. Importantly, the accumulation of misfolded α-syn protein aggregates alone did not increase neurodegeneration following exposure to rotenone but required the NF-κB-dependent inflammatory response in microglia.
Collapse
Affiliation(s)
- Savannah M. Rocha
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523
| | - Kelly S. Kirkley
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523
| | - Debotri Chatterjee
- Jefferson Comprehensive Parkinson’s Center, Vickie & Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Tawfik A. Aboellail
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523
| | - Richard J. Smeyne
- Jefferson Comprehensive Parkinson’s Center, Vickie & Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Ronald B. Tjalkens
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523
| |
Collapse
|
4
|
Neuroprotective Effect of Vitamin D on Behavioral and Oxidative Parameters of Male and Female Adult Wistar Rats Exposed to Mancozeb (manganese/zinc ethylene bis-dithiocarbamate). Mol Neurobiol 2023; 60:3724-3740. [PMID: 36940076 DOI: 10.1007/s12035-023-03298-8] [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: 09/14/2022] [Accepted: 03/02/2023] [Indexed: 03/21/2023]
Abstract
The constant exposure of rural workers to pesticides is a serious public health problem. Mancozeb (MZ) is a pesticide linked to hormonal, behavioral, genetic, and neurodegenerative effects, mainly related to oxidative stress. Vitamin D is a promising molecule that acts as a protector against brain aging. This study aimed to evaluate the neuroprotective role of vitamin D in adult male and female Wistar rats exposed to MZ. Animals received 40 mg/kg of MZ i.p. and 12.5 μg/kg or 25 μg/kg vitamin D by gavage, twice a week, for 6 weeks. The concentration of manganese had a significant increase in the hippocampus of both sexes and in the striatum of females, unlike zinc, which did not show a significant increase. MZ poisoning led to mitochondrial changes in brain tissues and promoted anxiogenic effects, especially in females. Alterations in antioxidant enzymes, mainly in the catalase activity were observed in intoxicated rats. Taken together, our results showed that exposure to MZ leads to the accumulation of manganese in brain tissues, and the behavior and metabolic/oxidative impairment were different between the sexes. Furthermore, the administration of Vitamin D was effective in preventing the damage caused by the pesticide.
Collapse
|
5
|
Human-Induced Pluripotent Stem Cell-Based Models for Studying Sex-Specific Differences in Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1387:57-88. [PMID: 34921676 DOI: 10.1007/5584_2021_683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The prevalence of neurodegenerative diseases is steadily increasing worldwide, and epidemiological studies strongly suggest that many of the diseases are sex-biased. It has long been suggested that biological sex differences are crucial for neurodegenerative diseases; however, how biological sex affects disease initiation, progression, and severity is not well-understood. Sex is a critical biological variable that should be taken into account in basic research, and this review aims to highlight the utility of human-induced pluripotent stem cells (iPSC)-derived models for studying sex-specific differences in neurodegenerative diseases, with advantages and limitations. In vitro systems utilizing species-specific, renewable, and physiologically relevant cell sources can provide powerful platforms for mechanistic studies, toxicity testings, and drug discovery. Matched healthy, patient-derived, and gene-corrected human iPSCs, from both sexes, can be utilized to generate neuronal and glial cell types affected by specific neurodegenerative diseases to study sex-specific differences in two-dimensional (2D) and three-dimensional (3D) human culture systems. Such relatively simple and well-controlled systems can significantly contribute to the elucidation of molecular mechanisms underlying sex-specific differences, which can yield effective, and potentially sex-based strategies, against neurodegenerative diseases.
Collapse
|
6
|
Posillico CK. Three's Company: Neuroimmune activation, sex, and memory at the tripartite synapse. Brain Behav Immun Health 2021; 16:100326. [PMID: 34589812 PMCID: PMC8474433 DOI: 10.1016/j.bbih.2021.100326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 12/30/2022] Open
Abstract
The neuroimmune system is required for normal cognitive functions such as learning and memory in addition to its critical role in detecting and responding to invading pathogens and injury. Understanding the functional convergence of neurons, astrocytes, and microglia at the synapse, particularly in the hippocampus, is key to understanding the nuances of such diverse roles. In the healthy brain, communication between all three cells is important for regulating neuronal activation and synaptic plasticity mechanisms, and during neuroinflammation, the activity and functions of all three cells can produce and be modulated by inflammatory cytokines. An important remaining component to this system is the conclusive evidence of sex differences in hippocampal plasticity mechanisms, hormone modulation of synaptic plasticity, functional properties of hippocampal neurons, and in neuroimmune activation. Sex as a biological variable here is necessary to consider given sex differences in the prevalence of memory-related disorders such as Alzheimer's disease and Post-Traumatic Stress disorder, both of which present with neuroimmune dysregulation. To make meaningful progress towards a deeper understanding of sex biases in memory-related disease prevalence, I propose that the next chapter of psychoneuroimmune research must focus on the signal integration and transduction at the synapse between experience-dependent plasticity mechanisms, neuroimmune activation, and the influence of biological sex.
Collapse
|
7
|
Posillico CK, Garcia-Hernandez RE, Tronson NC. Sex differences and similarities in the neuroimmune response to central administration of poly I:C. J Neuroinflammation 2021; 18:193. [PMID: 34488804 PMCID: PMC8418962 DOI: 10.1186/s12974-021-02235-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/04/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The neuroimmune system is required for normal neural processes, including modulation of cognition, emotion, and adaptive behaviors. Aberrant neuroimmune activation is associated with dysregulation of memory and emotion, though the precise mechanisms at play are complex and highly context dependent. Sex differences in neuroimmune activation and function further complicate our understanding of its roles in cognitive and affective regulation. METHODS Here, we characterized the physiological sickness and inflammatory response of the hippocampus following intracerebroventricular (ICV) administration of a synthetic viral mimic, polyinosinic:polycytidylic acid (poly I:C), in both male and female C57Bl/6N mice. RESULTS We observed that poly I:C induced weight loss, fever, and elevations of cytokine and chemokines in the hippocampus of both sexes. Specifically, we found transient increases in gene expression and protein levels of IL-1α, IL-1β, IL-4, IL-6, TNFα, CCL2, and CXCL10, where males showed a greater magnitude of response compared with females. Only males showed increased IFNα and IFNγ in response to poly I:C, whereas both males and females exhibited elevations of IFNβ, demonstrating a specific sex difference in the anti-viral response in the hippocampus. CONCLUSION Our data suggest that type I interferons are one potential node mediating sex-specific cytokine responses and neuroimmune effects on cognition. Together, these findings highlight the importance of using both males and females and analyzing a broad set of inflammatory markers in order to identify the precise, sex-specific roles for neuroimmune dysregulation in neurological diseases and disorders.
Collapse
Affiliation(s)
- Caitlin K Posillico
- Psychology Department, University of Michigan, 530 Church St., Ann Arbor, MI 48109, USA
| | | | - Natalie C Tronson
- Psychology Department, University of Michigan, 530 Church St., Ann Arbor, MI 48109, USA.
| |
Collapse
|
8
|
Lombardi C, Thompson S, Ritz B, Cockburn M, Heck JE. Residential proximity to pesticide application as a risk factor for childhood central nervous system tumors. ENVIRONMENTAL RESEARCH 2021; 197:111078. [PMID: 33798513 PMCID: PMC8212567 DOI: 10.1016/j.envres.2021.111078] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/18/2021] [Accepted: 03/21/2021] [Indexed: 05/16/2023]
Abstract
BACKGROUND Pesticide exposures have been examined previously as risk factors for childhood brain cancers, but few studies were able to assess risk from specific agents. OBJECTIVE To evaluate risks for childhood central nervous system tumors associated with residential proximity to agricultural pesticide applications. METHODS Using the California Cancer Registry, we identified cancer cases less than 6 years of age and frequency matched them by year of birth to 20 cancer-free controls identified from birth certificates. We restricted analyses to mothers living in rural areas and births occurring between 1998 and 2011, resulting in 667 cases of childhood central nervous system tumors and 123,158 controls. Possible carcinogens were selected per the Environmental Protection Agency's (US. EPA) classifications, and prenatal exposure was assessed according to pesticides reported by the California Department of Pesticide Regulation's (CDPR) Pesticide Use Reporting (PUR) system as being applied within 4000m of the maternal residence at birth. We computed odds ratios for individual pesticide associations using unconditional logistic and hierarchical regression models. RESULTS We observed elevated risks in the hierarchical models for diffuse astrocytoma with exposure to bromacil (OR: 2.12, 95% CI: 1.13-3.97), thiophanate-methyl (OR: 1.64, 95% CI: 1.02-2.66), triforine (OR: 2.38, 95% CI: 1.44-3.92), and kresoxim methyl (OR: 2.09, 95% CI: 1.03-4.21); elevated risks for medulloblastoma with exposure to chlorothalonil (OR: 1.78, 95% CI: 1.15-2.76), propiconazole (OR: 1.60, 95% CI: 1.02, 2.53), dimethoate (OR: 1.60, 95% CI: 1.06, 2.43), and linuron (OR: 2.52, 95% CI: 1.25, 5.11); and elevated risk for ependymoma with exposure to thiophanate-methyl (OR: 1.72, 95% CI: 1.10-2.68). CONCLUSION Our study suggests that exposure to certain pesticides through residential proximity to agricultural applications during pregnancy may increase the risk of childhood central nervous system tumors.
Collapse
Affiliation(s)
- Christina Lombardi
- Department of Cancer Prevention and Control, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 700 N. San Vicente Blvd., Pacific Design Center, G599, West Hollywood, CA, 90069, USA
| | - Shiraya Thompson
- Department of Epidemiology, Fielding School of Public Health, University of California, 650 Charles E. Young Dr. S, Box 951772, Los Angeles, CA, 90095-1772, USA
| | - Beate Ritz
- Department of Epidemiology, Fielding School of Public Health, University of California, 650 Charles E. Young Dr. S, Box 951772, Los Angeles, CA, 90095-1772, USA
| | - Myles Cockburn
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, 2001, N. Soto Street, Suite 318-A, Los Angeles, CA, USA
| | - Julia E Heck
- Department of Epidemiology, Fielding School of Public Health, University of California, 650 Charles E. Young Dr. S, Box 951772, Los Angeles, CA, 90095-1772, USA; Jonsson Comprehensive Cancer Center, University of California, Box 951781, Los Angeles, CA, 90095-1781, USA; College of Health and Public Service, University of North Texas, 1155 Union Circle #311340, Denton, TX, 76203-5017, USA.
| |
Collapse
|
9
|
Kianpour F, Mohseni M, Beigmohamadi M, Yazdinezhad A, Ramazani A, Hosseini MJ, Sharafi A. The protective effects of Ziziphora tenuior L. against chlorpyrifos induced toxicity: Involvement of inflammatory and cell death signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 272:113959. [PMID: 33636318 DOI: 10.1016/j.jep.2021.113959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ziziphora tenuior L. is used as a medicinal plant in treatment of various diseases such as gastric disorders, stomach ache, dysentery, uterus infection, gut inflammation and menstruation. AIM OF THE STUDY In the present study, the protective effects of Ziziphora tenuior extract against chlorpyrifos (CPF), the most commonly or popularly used insecticide in Asia and Africa were investigated in liver and lung tissues with emphasis in apoptotic and inflammatory pathways in rat model. MATERIALS AND METHODS The experiments were performed by gavage of male rats for 8 weeks. The extract of Z. tenuior was administrated at three different doses (40, 80, 160 mg/kg). 6.75 mg/kg CPF was administrated as the maximum tolerable dose based on our previous study. RESULTS Our data indicated that CPF can increase the expression of some inflammatory genes (IL-6, TLR-2, IL-1β, TNF-α, and NLPR3) and apoptosis genes (Caspase 3, Caspase 9, Caspase 8 and Bax). On the other hand, it can down regulate Bcl-2 gene expression. Post-treatment of Z. tenuior extract in CPF- treated rats showed significant decrease in apoptotic and inflammatory gene expression in the liver and lung due to its anti-apoptotic effects which confirmed by Bcl-2 gene overexpression. CONCLUSION The present study suggested that Z. tenuior extract, as a traditional treatment can be able to moderate CPF toxicity via significant effect on inflammatory and apoptotic cell death signaling pathway. Also, based on our preliminary data, it is suggested that Z. tenuior extract can prevent the adverse effects of CPF in liver and lung tissues.
Collapse
Affiliation(s)
- Fatemeh Kianpour
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahtab Mohseni
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mina Beigmohamadi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Alireza Yazdinezhad
- Department of Pharmacognosy, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Ramazani
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mir-Jamal Hosseini
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Ali Sharafi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
10
|
Chowen JA, Garcia-Segura LM. Role of glial cells in the generation of sex differences in neurodegenerative diseases and brain aging. Mech Ageing Dev 2021; 196:111473. [PMID: 33766745 DOI: 10.1016/j.mad.2021.111473] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/11/2022]
Abstract
Diseases and aging-associated alterations of the nervous system often show sex-specific characteristics. Glial cells play a major role in the endogenous homeostatic response of neural tissue, and sex differences in the glial transcriptome and function have been described. Therefore, the possible role of these cells in the generation of sex differences in pathological alterations of the nervous system is reviewed here. Studies have shown that glia react to pathological insults with sex-specific neuroprotective and regenerative effects. At least three factors determine this sex-specific response of glia: sex chromosome genes, gonadal hormones and neuroactive steroid hormone metabolites. The sex chromosome complement determines differences in the transcriptional responses in glia after brain injury, while gonadal hormones and their metabolites activate sex-specific neuroprotective mechanisms in these cells. Since the sex-specific neuroprotective and regenerative activity of glial cells causes sex differences in the pathological alterations of the nervous system, glia may represent a relevant target for sex-specific therapeutic interventions.
Collapse
Affiliation(s)
- Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, and IMDEA Food Institute, CEIUAM+CSIC, Madrid, Spain.
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC) and Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
11
|
Lennol MP, Canelles S, Guerra-Cantera S, Argente J, García-Segura LM, de Ceballos ML, Chowen JA, Frago LM. Amyloid-β 1-40 differentially stimulates proliferation, activation of oxidative stress and inflammatory responses in male and female hippocampal astrocyte cultures. Mech Ageing Dev 2021; 195:111462. [PMID: 33609535 DOI: 10.1016/j.mad.2021.111462] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and has a higher incidence in women. The main component of the senile plaques characteristic of AD is amyloid-beta (Aβ), with surrounding astrocytes contributing to the degenerative process. We hypothesized that the sex difference in the incidence of AD could be partially due to differential astrocytic responses to Aβ. Thus, the effect of Aβ1-40 on cell viability, the inflammatory response, and oxidative status was studied in cultures of hippocampal astrocytes from male and female rats. Aβ1-40 increased astrocyte viability in both female and male cultures by activating proliferation and survival pathways. Pro-inflammatory and anti-inflammatory responses were induced in astrocytes from both sexes. Aβ1-40 did not affect endoplasmic reticulum stress although it induced oxidative stress in male and female astrocytes. Interestingly, male astrocytes had an increase in cell number and significantly lower cell death in response to Aβ1-40. Conversely, astrocytes from females displayed a greater inflammatory response after the Aβ1-40 challenge. These results suggest that the inflammatory and oxidative environment induced by Aβ1-40 in female astrocytes may contribute to enhance the vulnerability to AD and warrants further studies to unveil the mechanisms underlying sex differences in astrocytic responses.
Collapse
Affiliation(s)
- Matthew P Lennol
- Department of Paediatrics, Facultad de Medicina, Universidad Autónoma de Madrid, Arzobispo Morcillo, 4, Madrid, 28029, Spain; Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Av. Menéndez Pelayo, 65, Madrid, 28009, Spain
| | - Sandra Canelles
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Av. Menéndez Pelayo, 65, Madrid, 28009, Spain; Instituto de Investigación Sanitaria Princesa, IIS-IP, Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5 Pabellón 11, Planta 0, Madrid, 28029, Spain
| | - Santiago Guerra-Cantera
- Department of Paediatrics, Facultad de Medicina, Universidad Autónoma de Madrid, Arzobispo Morcillo, 4, Madrid, 28029, Spain; Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Av. Menéndez Pelayo, 65, Madrid, 28009, Spain; Instituto de Investigación Sanitaria Princesa, IIS-IP, Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5 Pabellón 11, Planta 0, Madrid, 28029, Spain
| | - Jesús Argente
- Department of Paediatrics, Facultad de Medicina, Universidad Autónoma de Madrid, Arzobispo Morcillo, 4, Madrid, 28029, Spain; Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Av. Menéndez Pelayo, 65, Madrid, 28009, Spain; Instituto de Investigación Sanitaria Princesa, IIS-IP, Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5 Pabellón 11, Planta 0, Madrid, 28029, Spain; IMDEA Food Institute, CEI UAM + CSIC, Carretera de Cantoblanco 8, Madrid, 28049, Spain
| | - Luis Miguel García-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Av. Doctor Arce, 37, Madrid, 28002, Spain; CIBER de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5 Pabellón 11, Planta 0, Madrid, 28029, Spain
| | - María L de Ceballos
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Av. Doctor Arce, 37, Madrid, 28002, Spain; CIBER de Investigación Biomédica en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5 Pabellón 11, Planta 0, Madrid, 28029, Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Av. Menéndez Pelayo, 65, Madrid, 28009, Spain; Instituto de Investigación Sanitaria Princesa, IIS-IP, Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5 Pabellón 11, Planta 0, Madrid, 28029, Spain; IMDEA Food Institute, CEI UAM + CSIC, Carretera de Cantoblanco 8, Madrid, 28049, Spain
| | - Laura M Frago
- Department of Paediatrics, Facultad de Medicina, Universidad Autónoma de Madrid, Arzobispo Morcillo, 4, Madrid, 28029, Spain; Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Av. Menéndez Pelayo, 65, Madrid, 28009, Spain; Instituto de Investigación Sanitaria Princesa, IIS-IP, Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5 Pabellón 11, Planta 0, Madrid, 28029, Spain.
| |
Collapse
|
12
|
Garcia JM, Meek EC, Chambers JE. Novel pyridinium oximes enhance 24-h survivability against a lethal dose of nerve agent surrogate in adult female rats. Toxicology 2020; 446:152626. [PMID: 33159982 DOI: 10.1016/j.tox.2020.152626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/08/2020] [Accepted: 11/01/2020] [Indexed: 02/06/2023]
Abstract
Our laboratory has developed novel substituted phenoxyalkyl pyridinium oximes (US Patent 9,227,937) designed to more efficiently penetrate the central nervous system to enhance survivability and attenuate seizure-like signs and neuropathology. Previous studies with male Sprague-Dawley rats indicated that survivability was enhanced against the nerve agent (sarin) surrogate, 4-nitrophenyl isopropyl methylphosphonate (NIMP). In this study, female adult Sprague-Dawley rats, tested specifically in diestrus, were challenged subcutaneously with lethal concentrations of NIMP (0.6 mg/kg). After development of seizure-like behavior and other signs of cholinergic toxicity, human equivalent dosages of atropine (0.65 mg/kg) and one of four oximes (2-PAM, or novel oxime 15, 20, or 55; 0.146 mmol/kg) or Multisol vehicle was administered alone or in binary oxime combinations intramuscularly. Animals were closely monitored for signs of cholinergic toxicity and 24 h survivability. Percentages of animals surviving the 24 h NIMP challenge dose were 35 % for 2-PAM and 55 %, 70 %, and 25 % for novel oximes 15, 20, and 55, respectively. Improvements in survival were also observed over 2-PAM alone with binary combinations of 2-PAM and either oxime 15 or oxime 20. Additionally, administration of novel oximes decreased the duration of seizure-like behavior as compared to 2-PAM suggesting that these oximes better penetrate the blood-brain barrier to mitigate central nervous system hypercholinergic activity. Efficacies were similar between females and previously reported males. These data indicate that the novel pyridinium oximes enhance survivability against lethal OP toxicity as compared to 2-PAM in adult female rats.
Collapse
Affiliation(s)
- Jason M Garcia
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, P.O. Box 6100, Mississippi State, MS, 39762, USA.
| | - Edward C Meek
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, P.O. Box 6100, Mississippi State, MS, 39762, USA.
| | - Janice E Chambers
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, P.O. Box 6100, Mississippi State, MS, 39762, USA.
| |
Collapse
|
13
|
Saint-Amour D, Muckle G, Gagnon-Chauvin A, Rouget F, Monfort C, Michineau L, Thomé JP, Kadhel P, Multigner L, Cordier S. Visual contrast sensitivity in school-age Guadeloupean children exposed to chlordecone. Neurotoxicology 2020; 78:195-201. [DOI: 10.1016/j.neuro.2020.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 02/25/2020] [Indexed: 10/24/2022]
|
14
|
Chang GQ, Collier AD, Karatayev O, Gulati G, Boorgu DSSK, Leibowitz SF. Moderate Prenatal Ethanol Exposure Stimulates CXCL12/CXCR4 Chemokine System in Radial Glia Progenitor Cells in Hypothalamic Neuroepithelium and Peptide Neurons in Lateral Hypothalamus of the Embryo and Postnatal Offspring. Alcohol Clin Exp Res 2020; 44:866-879. [PMID: 32020622 DOI: 10.1111/acer.14296] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/21/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Prenatal exposure to ethanol (EtOH) has lasting effects on neuropeptide and neuroimmune systems in the brain alongside detrimental alcohol-related behaviors. At low-to-moderate doses, prenatal EtOH stimulates neurogenesis in lateral hypothalamus (LH) and increases neurons that express the orexigenic peptides hypocretin/orexin (Hcrt/OX) and melanin-concentrating hormone (MCH), and the proinflammatory chemokine CCL2, which through its receptor CCR2 stimulates cell differentiation and movement. Our recent studies demonstrated that CCL2 and CCR2 colocalize with MCH neurons and are involved in EtOH's stimulatory effect on their development but show no relation to Hcrt/OX. Here, we investigated another chemokine, CXCL12, and its receptor, CXCR4, which promote neurogenesis and neuroprogenitor cell proliferation, to determine if they also exhibit peptide specificity in their response to EtOH exposure. METHODS Pregnant rats were intraorally administered a moderate dose of EtOH (2 g/kg/d) from embryonic day 10 (E10) to E15. Their embryos and postnatal offspring were examined using real-time quantitative PCR and immunofluorescence histochemistry, to determine if EtOH affects CXCL12 and CXCR4 and the colocalization of CXCR4 with Hcrt/OX and MCH neurons in the LH and with radial glia neuroprogenitor cells in the hypothalamic neuroepithelium (NEP). RESULTS Prenatal EtOH strongly stimulated CXCL12 and CXCR4 in LH neurons of embryos and postnatal offspring. This stimulation was significantly stronger in Hcrt/OX than MCH neurons in LH and also occurred in radial glia neuroprogenitor cells dense in the NEP. These effects were sexually dimorphic, consistently stronger in females than males. CONCLUSIONS While showing prenatal EtOH exposure to have a sexually dimorphic, stimulatory effect on CXCL12 and CXCR4 in LH similar to CCL2 and its receptor, these results reveal their distinct relationship to the peptide neurons, with the former closely related to Hcrt/OX and the latter to MCH, and they link EtOH's actions in LH to a stimulatory effect on neuroprogenitor cells in the NEP.
Collapse
Affiliation(s)
- Guo-Qing Chang
- From the, Laboratory of Behavioral Neurobiology, (GQC, ADC, OK, GG, SFL), The Rockefeller University, New York, New York
| | - Adam D Collier
- From the, Laboratory of Behavioral Neurobiology, (GQC, ADC, OK, GG, SFL), The Rockefeller University, New York, New York
| | - Olga Karatayev
- From the, Laboratory of Behavioral Neurobiology, (GQC, ADC, OK, GG, SFL), The Rockefeller University, New York, New York
| | - Gazal Gulati
- From the, Laboratory of Behavioral Neurobiology, (GQC, ADC, OK, GG, SFL), The Rockefeller University, New York, New York
| | | | - Sarah F Leibowitz
- From the, Laboratory of Behavioral Neurobiology, (GQC, ADC, OK, GG, SFL), The Rockefeller University, New York, New York
| |
Collapse
|
15
|
Bollinger JL, Salinas I, Fender E, Sengelaub DR, Wellman CL. Gonadal hormones differentially regulate sex-specific stress effects on glia in the medial prefrontal cortex. J Neuroendocrinol 2019; 31:e12762. [PMID: 31228875 PMCID: PMC6715499 DOI: 10.1111/jne.12762] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/20/2019] [Accepted: 06/18/2019] [Indexed: 12/21/2022]
Abstract
Women are more susceptible to various stress-linked psychopathologies, including depression. Dysfunction of the medial prefrontal cortex (mPFC) has been implicated in depression, and studies indicate sex differences in stress effects on mPFC structure and function. For example, chronic stress induces dendritic atrophy in the mPFC in male rats, yet dendritic growth in females. Recent findings suggest glial pathways toward depression. Glia are highly responsive to neuronal activity and function as critical regulators of synaptic plasticity. Preclinical models demonstrate stress-induced microglial activation in mPFC in males, yet deactivation in females. By contrast, stress reduces astrocyte complexity in mPFC in male rats, whereas the effects in females are unknown. Glia possess receptors for most gonadal hormones and gonadal hormones are known to modulate neuronal activity. Thus, gonadal hormones represent a potential mechanism underlying sex differences in glia, as well as divergent stress effects. Therefore, we examined the role of gonadal hormones in sex-specific stress effects on neuronal activity (ie FosB/ ΔFosB induction) and glia in the mPFC. The findings obtained indicate greater microglial activation in mPFC in females and a greater astrocyte area in males. Basal astrocyte morphology is modulated by androgens, whereas androgens or oestrogens dampen the microglial state in males. Astrocyte morphology is associated with neuronal activity in both sexes, regardless of hormonal condition. Chronic stress induced astrocytic atrophy in males, yet hypertrophy in females, with gonadal hormones partly regulating this difference. Stress effects on microglia are oestradiol-dependent in females. Taken together, these data suggest sex-specific, gonadal hormone-dependent stress effects on astrocytes and microglia in the mPFC.
Collapse
Affiliation(s)
- Justin L Bollinger
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, USA
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, USA
| | - Isabella Salinas
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, USA
| | - Emily Fender
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Dale R Sengelaub
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, USA
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, USA
| | - Cara L Wellman
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, USA
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, USA
| |
Collapse
|
16
|
Arbo MD, Altknecht LF, Cattani S, Braga WV, Peruzzi CP, Cestonaro LV, Göethel G, Durán N, Garcia SC. In vitro cardiotoxicity evaluation of graphene oxide. Mutat Res 2019; 841:8-13. [PMID: 31138412 DOI: 10.1016/j.mrgentox.2019.03.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 02/28/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023]
Abstract
Graphene is a two-dimensional (2D) monolayer of carbon atoms, tightly packed, forming a honey comb crystal lattice, with physical, chemical, and mechanical properties greatly used for energy storage, electrochemical devices, and in nanomedicine. Many studies showed that nanomaterials have side-effects on health. At present, there is a lack of information regarding graphene and its derivatives including their cardiotoxic properties. The aim of the present study was to evaluate the toxicity of nano-graphene oxide (nano-GO) in the rat cardiomyoblast cell line H9c2 and the involvement of oxidative processes. The cell viability was evaluated with the fluorescein diacetate (FDA)/propidium iodide (PI) and in the trypan blue exclusion assay, furthermore mitochondrial membrane potential and production of free radicals were measured. Genotoxicity was evaluated in comet assay and low molecular weight DNA experiment. Reduction of cell viability with 20, 40, 60, 80, and 100 μg/mL nano-GO was observed after 24 h incubation. Besides, nano-GO induced a mitochondrial hyperpolarization and a significant increase of free radicals production in the same concentrations. DNA breaks were observed at 40, 60, 80, and 100 μg/mL. This DNA damage was accompanied by a significant increase in LMW DNA only at 40 μg/mL. In conclusion, the nano-GO caused cardiotoxicity in our in vitro model, with mitochondrial disturbances, generation of reactive species and interactions with DNA, indicating the importance of the further evaluation of the safety of nanomaterials.
Collapse
Affiliation(s)
- Marcelo Dutra Arbo
- Laboratório de Toxicologia, Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
| | - Louise F Altknecht
- Laboratório de Toxicologia, Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
| | - Shanda Cattani
- Laboratório de Toxicologia, Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
| | - Wesley V Braga
- Laboratório de Toxicologia, Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
| | - Caroline P Peruzzi
- Laboratório de Toxicologia, Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
| | - Larissa V Cestonaro
- Laboratório de Toxicologia, Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
| | - Gabriela Göethel
- Laboratório de Toxicologia, Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
| | - Nelson Durán
- Laboratório Nacional de Nanotecnologia - LNNano, Instituto de Quimica-UNICAMP, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Solange Cristina Garcia
- Laboratório de Toxicologia, Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil.
| |
Collapse
|
17
|
Alarcón-Aguilar A, Luna-López A, López-Diazguerrero NE, Königsberg M. The Effect of MPP+ on the Viability of Primary Cortical Astrocytes Isolated from Female and Male Wistar Rats of Different Ages. Cell Mol Neurobiol 2019; 39:321-328. [PMID: 30539419 PMCID: PMC11469902 DOI: 10.1007/s10571-018-0643-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023]
Abstract
Although age is known to be the main risk for developing chronic and neurodegenerative diseases, those illnesses have a different prevalence depending on the sex. It has been questioned whether genetic and hormonal differences are preserved in primary cultures from individuals of different genders. Therefore, here we studied the susceptibility of astrocytes, obtained from female and male Wistar rats of different ages (newborn, 9 and 24 months-old), to the well-known toxin MPP+ after 2 weeks in vitro, at different concentrations and exposure times. Our results showed that there are no variances due to gender, but that there are important differences associated to age in terms of the viability against this toxin.
Collapse
Affiliation(s)
- Adriana Alarcón-Aguilar
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, Ciudad de México, Mexico
| | | | | | - Mina Königsberg
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, Ciudad de México, Mexico.
- División de Ciencias Biológicas y de la Salud, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, A.P. 55-535, C.P 09340, Ciudad de México, Mexico.
| |
Collapse
|
18
|
Martínez-Morcillo S, Pérez-López M, Soler-Rodríguez F, González A. The organophosphorus pesticide dimethoate decreases cell viability and induces changes in different biochemical parameters of rat pancreatic stellate cells. Toxicol In Vitro 2019; 54:89-97. [PMID: 30243730 DOI: 10.1016/j.tiv.2018.09.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 09/15/2018] [Accepted: 09/18/2018] [Indexed: 12/14/2022]
Abstract
In the present study we employed cultured pancreatic stellate cells to study the effect of the organophosphorus insecticide dimethoate on pancreatic cell physiology. Esterase activity, cell viability, reactive oxygen species generation and Ca2+ mobilization were examined. Our results show that dimethoate (0.1, 1 and 10 μM) induced a concentration-dependent inhibition of cholinesterase enzymatic activity at all concentrations tested. A drop in carboxylesterase activity was noted in the presence of 10 μM dimethoate. In the presence of the pesticide a decrease in cell viability was detected. The clearer effect could be observed when the cells had been incubated during 96 h in the presence of dimethoate. The pesticide induced a slight but statistically significant increase in the production of reactive oxygen species in the mitochondria. Incubation of cells with dimethoate, in the presence of Ca2+ in the extracellular medium, led to a slow and progressive increase in [Ca2+]c towards an elevated value over the prestimulation level. A similar behavior was observed in the absence of extracellular Ca2+, indicating that dimethoate releases Ca2+ from the intracellular stores. Our results suggest that dimethoate might alter intracellular pathways that are critical for pancreatic physiology, creating a situation potentially leading to dysfunction in the exocrine pancreas.
Collapse
Affiliation(s)
| | - Marcos Pérez-López
- Unit of Toxicology, Veterinary Faculty, University of Extremadura, Caceres, Spain.
| | | | - Antonio González
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain.
| |
Collapse
|
19
|
Chistyakov DV, Azbukina NV, Astakhova AA, Goriainov SV, Chistyakov VV, Sergeeva MG. Sex-Mediated Differences in LPS Induced Alterations of TNFα, IL-10 Expression, and Prostaglandin Synthesis in Primary Astrocytes. Int J Mol Sci 2018; 19:ijms19092793. [PMID: 30227622 PMCID: PMC6164227 DOI: 10.3390/ijms19092793] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 09/12/2018] [Accepted: 09/14/2018] [Indexed: 01/14/2023] Open
Abstract
Although many neurological and psychiatric disorders reveal clear sex-dependent variations, the molecular mechanism of this process is not clear enough. Astrocytes are involved in the response of neural tissue to injury and inflammation, produce steroid hormones, and sense steroid presence. To explore the hypothesis that astrocytes may participate in sex-mediated differences of inflammatory responses, we have examined whether male and female primary rat astrocytes show different responses to lipopolysaccharide (LPS) as a toll-like receptor 4 (TLR4) agonist. Levels of mRNA and proteins of tumor necrosis factor alpha (TNFα), interleukin-10 (IL-10), and cyclooxygenase (COX)-2 were assessed using qPCR, immunoblotting, and ELISA. UPLC-MS/MS was used to detect prostaglandins (PGs). LPS stimulation resulted in different levels of cytokine production; more TNFα and less IL-10 were produced in female cells compared with male astrocytes. Although the levels of the COX-2 expression were not altered, LPS significantly induced the synthesis of PGs with notable sex-related differences. PGE2 and PGD2 were less and 6-keto-PGF1α was more upregulated in female astrocytes, and TXB2 had similar levels in cells obtained from males and females. Trilostane, an inhibitor of 3β-Hydroxysteroid dehydrogenase (3β-HSD), inhibited the LPS-induced TNFα production and the release of PGE2, PGD2, and 6-keto-PGF1α in female astrocytes. Thus, male and female astrocytes differentially respond to inflammatory challenges on the level of production of cytokines and steroid hormones. Sex-mediated differences in pro- and anti-inflammatory responses should be taken into consideration for the effective treatment of disorders with neuroinflammation.
Collapse
Affiliation(s)
- Dmitry V Chistyakov
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119992, Russia.
- Laboratory of electrophysiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia.
| | - Nadezda V Azbukina
- Faculty of Bioengineering and Bioinformatics, Moscow Lomonosov State University, Moscow 119234, Russia.
| | - Alina A Astakhova
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119992, Russia.
| | - Sergei V Goriainov
- SREC PFUR, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia.
| | - Viktor V Chistyakov
- SREC PFUR, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia.
| | - Marina G Sergeeva
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119992, Russia.
| |
Collapse
|
20
|
Lipotoxic Effects of Palmitic Acid on Astrocytes Are Associated with Autophagy Impairment. Mol Neurobiol 2018; 56:1665-1680. [PMID: 29916142 DOI: 10.1007/s12035-018-1183-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/07/2018] [Indexed: 12/23/2022]
|
21
|
Torres-Rojas C, Jones BC. Sex Differences in Neurotoxicogenetics. Front Genet 2018; 9:196. [PMID: 29922331 PMCID: PMC5996082 DOI: 10.3389/fgene.2018.00196] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/15/2018] [Indexed: 12/02/2022] Open
Abstract
A major development in biomedical research is the recognition that the sex of an individual plays a key role in susceptibility, treatment, and outcomes of most diseases. In this contribution, we present evidence that sex is also important in the toxicity of many environmental toxicants and contributes to the effect of genetics. Thus, individual differences in response to toxicants includes genetic makeup, the environment and sex; in fact, sex differences may be considered a part of genetic constitution. In this review, we present evidence for sex contribution to susceptibility for a number of toxicants.
Collapse
Affiliation(s)
- Carolina Torres-Rojas
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Byron C Jones
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
22
|
Sex modulated effects of sarin exposure in rats: Toxicity, hypothermia and inflammatory markers. Neurotoxicology 2018; 66:121-127. [DOI: 10.1016/j.neuro.2018.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/28/2018] [Accepted: 04/02/2018] [Indexed: 12/31/2022]
|
23
|
Comfort N, Re DB. Sex-Specific Neurotoxic Effects of Organophosphate Pesticides Across the Life Course. Curr Environ Health Rep 2018; 4:392-404. [PMID: 29063415 DOI: 10.1007/s40572-017-0171-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW This review discusses the sex-specific effects of exposure to various organophosphate (OP) pesticides throughout the life course and potential reasons for the differential vulnerabilities observed across sexes. RECENT FINDINGS Sex is a crucial factor in the response to toxicants, yet the sex-specific effects of OP exposure, particularly in juveniles and adults, remain unresolved. This is largely due to study design and inconsistencies in exposure and outcome assessments. Exposure to OPs results in multiple adverse outcomes influenced by many factors including sex. Reported sex-specific effects suggest that males are more susceptible to OPs, which reflects the sex-dependent prevalence of various neurodevelopmental and neurodegenerative disorders such as autism and amyotrophic lateral sclerosis (ALS), in which males are at greater risk. Thus, this review proposes that the biological sex-specific effects elicited by OP exposure may in part underlie the dimorphic susceptibilities observed in neurological disorders. Understanding the immediate and long-term effects of OP exposure across sexes will be critical in advancing our understanding of OP-induced neurotoxicity and disease.
Collapse
Affiliation(s)
- Nicole Comfort
- 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. .,, 722 W 168th Street, 11th floor, 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
|
24
|
Heitzer M, Kaiser S, Kanagaratnam M, Zendedel A, Hartmann P, Beyer C, Johann S. Administration of 17β-Estradiol Improves Motoneuron Survival and Down-regulates Inflammasome Activation in Male SOD1(G93A) ALS Mice. Mol Neurobiol 2016; 54:8429-8443. [PMID: 27957680 DOI: 10.1007/s12035-016-0322-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease manifested by the progressive loss of upper and lower motoneurons. The pathomechanism of ALS is complex and not yet fully understood. Neuroinflammation is believed to significantly contribute to disease progression. Inflammasome activation was recently shown in the spinal cord of human sporadic ALS patients and in the SOD1(G93A) mouse model for ALS. In the present study, we investigated the neuroprotective and anti-inflammatory effects of 17β-estradiol (E2) treatment in pre-symptomatic and symptomatic male SOD1(G93A) mice. Symptomatic mice with E2 substitution exhibited improved motor performance correlating with an increased survival of motoneurons in the lumbar spinal cord. Expression of NLRP3 inflammasome proteins and levels of activated caspase 1 and mature interleukin 1 beta were significantly reduced in SOD1(G93A) mice supplemented with E2.
Collapse
Affiliation(s)
- Marius Heitzer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Sarah Kaiser
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Mithila Kanagaratnam
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Adib Zendedel
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.,Department of Anatomical Sciences, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Philipp Hartmann
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.,JARA-BRAIN, 52074, Aachen, Germany
| | - Sonja Johann
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
25
|
Acaz-Fonseca E, Avila-Rodriguez M, Garcia-Segura LM, Barreto GE. Regulation of astroglia by gonadal steroid hormones under physiological and pathological conditions. Prog Neurobiol 2016; 144:5-26. [DOI: 10.1016/j.pneurobio.2016.06.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 06/05/2016] [Indexed: 01/07/2023]
|
26
|
Arbo BD, Marques CV, Ruiz-Palmero I, Ortiz-Rodriguez A, Ghorbanpoor S, Arevalo MA, Garcia-Segura LM, Ribeiro MF. 4'-Chlorodiazepam is neuroprotective against amyloid-beta through the modulation of survivin and bax protein expression in vitro. Brain Res 2015; 1632:91-7. [PMID: 26707976 DOI: 10.1016/j.brainres.2015.12.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 12/06/2015] [Accepted: 12/11/2015] [Indexed: 12/12/2022]
Abstract
The translocator protein of 18kDa (TSPO) is located in the outer mitochondrial membrane and is involved in the cholesterol transport into the mitochondria and in the regulation of steroidogenesis, mitochondrial permeability transition pore opening and apoptosis. TSPO ligands have been investigated as therapeutic agents that promote neuroprotective effects in experimental models of brain injury and neurodegenerative diseases. The aim of this study was to identify the neuroprotective effects of 4'-chlorodiazepam (4'-CD), a ligand of TSPO, against amyloid-beta (Aβ) in SHSY-5Y neuroblastoma cells and its mechanisms of action. Aβ decreased the viability of SHSY-5Y neuroblastoma cells, while 4'-CD had a neuroprotective effect at the doses of 1nM and 10nM. The neuroprotective effects of 4'-CD against Aβ were associated with the inhibition of Aβ-induced upregulation of Bax and downregulation of survivin. In summary, our findings indicate that 4'-CD is neuroprotective against Aβ-induced neurotoxicity by a mechanism that may involve the regulation of Bax and survivin expression.
Collapse
Affiliation(s)
- B D Arbo
- Laboratório de Interação Neuro-Humoral, Department of Physiology, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil; Instituto Cajal - CSIC, Avenida Doctor Arce, 37, 28002 Madrid, Spain.
| | - C V Marques
- Laboratório de Interação Neuro-Humoral, Department of Physiology, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil; Instituto Cajal - CSIC, Avenida Doctor Arce, 37, 28002 Madrid, Spain
| | - I Ruiz-Palmero
- Instituto Cajal - CSIC, Avenida Doctor Arce, 37, 28002 Madrid, Spain
| | - A Ortiz-Rodriguez
- Instituto Cajal - CSIC, Avenida Doctor Arce, 37, 28002 Madrid, Spain
| | - S Ghorbanpoor
- Instituto Cajal - CSIC, Avenida Doctor Arce, 37, 28002 Madrid, Spain
| | - M A Arevalo
- Instituto Cajal - CSIC, Avenida Doctor Arce, 37, 28002 Madrid, Spain
| | - L M Garcia-Segura
- Instituto Cajal - CSIC, Avenida Doctor Arce, 37, 28002 Madrid, Spain
| | - M F Ribeiro
- Laboratório de Interação Neuro-Humoral, Department of Physiology, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| |
Collapse
|
27
|
Grassi D, Ghorbanpoor S, Acaz-Fonseca E, Ruiz-Palmero I, Garcia-Segura LM. The Selective Estrogen Receptor Modulator Raloxifene Regulates Arginine-Vasopressin Gene Expression in Human Female Neuroblastoma Cells Through G Protein-Coupled Estrogen Receptor and ERK Signaling. Endocrinology 2015. [PMID: 26200092 DOI: 10.1210/en.2014-2010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The selective estrogen receptor modulator raloxifene reduces blood pressure in hypertensive postmenopausal women. In the present study we have explored whether raloxifene regulates gene expression of arginine vasopressin (AVP), which is involved in the pathogenesis of hypertension. The effect of raloxifene was assessed in human female SH-SY5Y neuroblastoma cells, which have been recently identified as a suitable cellular model to study the estrogenic regulation of AVP. Raloxifene, within a concentration ranging from 10(-10) M to 10(-6) M, decreased the mRNA levels of AVP in SH-SY5Y cells with maximal effect at 10(-7) M. This effect of raloxifene was imitated by an agonist (±)-1-[(3aR*,4S*,9bS*)-4-(6-bromo-1,3-benzodioxol-5-yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinolin-8-yl]-ethanone of G protein-coupled estrogen receptor-1 (GPER) and blocked by an antagonist (3aS*,4R*,9bR*)-4-(6-bromo-1,3-benzodioxol-5-yl)-3a,4,5,9b-3H-cyclopenta[c]quinoline of GPER and by GPER silencing. Raloxifene induced a time-dependent increase in the level of phosphorylated ERK1 and ERK2, by a mechanism blocked by the GPER antagonist. The treatment of SH-SY5Y cells with either a MAPK/ERK kinase 1/2-specific inhibitor (1,4-diamino-2, 3-dicyano-1,4-bis(2-aminophenylthio)butadine) or a protein kinase C inhibitor (sotrastaurin) blocked the effects of raloxifene on the phosphorylation of ERK1/2 and the regulation of AVP mRNA levels. These results reveal a mechanism mediating the regulation of AVP expression by raloxifene, involving the activation of GPER, which in turn activates protein kinase C, MAPK/ERK kinase, and ERK. The regulation of AVP by raloxifene and GPER may have implications for the treatment of blood hypertension(.).
Collapse
Affiliation(s)
- Daniela Grassi
- Instituto Cajal (D.G., S.G., E.A.-F., I.R.P., L.M.G.-S.), Consejo Superior de Investigaciones Científicas, E-28002 Madrid, Spain; Institute of Anatomy and Cell Biology (D.G.), Department of Molecular Embryology, Albert-Ludwigs-Universität Freiburg, D-79104 Freiburg, Germany; and Department of Cell and Molecular Biology (S.G.), School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Samar Ghorbanpoor
- Instituto Cajal (D.G., S.G., E.A.-F., I.R.P., L.M.G.-S.), Consejo Superior de Investigaciones Científicas, E-28002 Madrid, Spain; Institute of Anatomy and Cell Biology (D.G.), Department of Molecular Embryology, Albert-Ludwigs-Universität Freiburg, D-79104 Freiburg, Germany; and Department of Cell and Molecular Biology (S.G.), School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Estefania Acaz-Fonseca
- Instituto Cajal (D.G., S.G., E.A.-F., I.R.P., L.M.G.-S.), Consejo Superior de Investigaciones Científicas, E-28002 Madrid, Spain; Institute of Anatomy and Cell Biology (D.G.), Department of Molecular Embryology, Albert-Ludwigs-Universität Freiburg, D-79104 Freiburg, Germany; and Department of Cell and Molecular Biology (S.G.), School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Isabel Ruiz-Palmero
- Instituto Cajal (D.G., S.G., E.A.-F., I.R.P., L.M.G.-S.), Consejo Superior de Investigaciones Científicas, E-28002 Madrid, Spain; Institute of Anatomy and Cell Biology (D.G.), Department of Molecular Embryology, Albert-Ludwigs-Universität Freiburg, D-79104 Freiburg, Germany; and Department of Cell and Molecular Biology (S.G.), School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Luis M Garcia-Segura
- Instituto Cajal (D.G., S.G., E.A.-F., I.R.P., L.M.G.-S.), Consejo Superior de Investigaciones Científicas, E-28002 Madrid, Spain; Institute of Anatomy and Cell Biology (D.G.), Department of Molecular Embryology, Albert-Ludwigs-Universität Freiburg, D-79104 Freiburg, Germany; and Department of Cell and Molecular Biology (S.G.), School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| |
Collapse
|
28
|
Wilhelm CJ, Hashimoto JG, Roberts ML, Bloom SH, Andrew MR, Wiren KM. Astrocyte Dysfunction Induced by Alcohol in Females but Not Males. Brain Pathol 2015; 26:433-51. [PMID: 26088166 DOI: 10.1111/bpa.12276] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 06/10/2015] [Indexed: 01/08/2023] Open
Abstract
Chronic alcohol abuse is associated with brain damage in a sex-specific fashion, but the mechanisms involved are poorly described and remain controversial. Previous results have suggested that astrocyte gene expression is influenced by ethanol intoxication and during abstinence in vivo. Here, bioinformatic analysis of astrocyte-enriched ethanol-regulated genes in vivo revealed ubiquitin pathways as an ethanol target, but with sexually dimorphic cytokine signaling and changes associated with brain aging in females and not males. Consistent with this result, astrocyte activation was observed after exposure in female but not male animals, with reduced S100β levels in the anterior cingulate cortex and increased GFAP(+) cells in the hippocampus. In primary culture, the direct effects of chronic ethanol exposure followed by recovery on sex-specific astrocyte function were examined. Male astrocyte responses were consistent with astrocyte deactivation with reduced GFAP expression during ethanol exposure. In contrast, female astrocytes exhibited increased expression of Tnf, reduced expression of the neuroprotective cytokine Tgfb1, disrupted bioenergetics and reduced excitatory amino acid uptake following exposure or recovery. These results indicate widespread astrocyte dysfunction in ethanol-exposed females and suggest a mechanism that may underlie increased vulnerability to ethanol-induced neurotoxicity in females.
Collapse
Affiliation(s)
- Clare J Wilhelm
- VA Portland Health Care System, Portland, OR.,Department of Psychiatry, Oregon Health & Science University, Portland, OR
| | - Joel G Hashimoto
- VA Portland Health Care System, Portland, OR.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
| | | | | | - Melissa R Andrew
- Cincinnati Center for Growth Disorders, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Kristine M Wiren
- VA Portland Health Care System, Portland, OR.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
| |
Collapse
|
29
|
Acaz-Fonseca E, Duran JC, Carrero P, Garcia-Segura LM, Arevalo MA. Sex differences in glia reactivity after cortical brain injury. Glia 2015; 63:1966-1981. [DOI: 10.1002/glia.22867] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/12/2015] [Accepted: 05/13/2015] [Indexed: 12/11/2022]
Affiliation(s)
| | - Juan C. Duran
- Consejo Superior De Investigaciones Cientificas (CSIC); Instituto Cajal; Madrid Spain
| | - Paloma Carrero
- Consejo Superior De Investigaciones Cientificas (CSIC); Instituto Cajal; Madrid Spain
| | - Luis M. Garcia-Segura
- Consejo Superior De Investigaciones Cientificas (CSIC); Instituto Cajal; Madrid Spain
| | - M. Angeles Arevalo
- Consejo Superior De Investigaciones Cientificas (CSIC); Instituto Cajal; Madrid Spain
| |
Collapse
|
30
|
Chen D, Zhang Z, Yao H, Cao Y, Xing H, Xu S. Pro- and anti-inflammatory cytokine expression in immune organs of the common carp exposed to atrazine and chlorpyrifos. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2014; 114:8-15. [PMID: 25175644 DOI: 10.1016/j.pestbp.2014.07.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 07/26/2014] [Accepted: 07/27/2014] [Indexed: 06/03/2023]
Abstract
Atrazine (ATR) and chlorpyrifos (CPF) are toxic and subject to long-term in vivo accumulation in different aquatic species throughout the world. The purpose of the present study was to examine the effect of ATR, CPF and combined ATR/CPF exposure on cytokines in the head kidney and spleen of common carp (Cyprinus carpio L.). The carp were sampled after a 40-d exposure to CPF and ATR, individually or in combination, followed by a 40-d recovery to measure the mRNA expression of IL-6fam (IL-6), IL-8, TNF-α, IL-10 and TGF-β1 (TGF-β) in the head kidney and spleen tissues. These results showed that the expression of cytokines IL-6, IL-8 and TNF-α in the head kidney and spleen was upregulated following ATR, CPF and mixed ATR/CPF exposure compared with the control group. The expression of IL-10 and TGF-β mRNA was significantly inhibited in both head kidney and spleen of carp exposed to ATR, CPF and the ATR/CPF mixture. The results suggested that long-term exposure of ATR, CPF and the ATR/CPF mixture in aquatic environments can induce the dysregulation of pro-/anti-inflammatory cytokine expression. The information regarding the effects of ATR and CPF on cytokine mRNA expression generated in this study will be important information for pesticides toxicology evaluation.
Collapse
Affiliation(s)
- Dechun Chen
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China; Department of Biological Engineering, Jilin Engineering Vocational College, 1299 Changfa Road, Siping 136001, China
| | - Ziwei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Haidong Yao
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Ye Cao
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Houjuan Xing
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China; Animal Health Supervision Institute of Heilongjiang Province, 243 Haping Road, Xiangfang District, Harbin 150069, China.
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China.
| |
Collapse
|
31
|
Acaz-Fonseca E, Sanchez-Gonzalez R, Azcoitia I, Arevalo MA, Garcia-Segura LM. Role of astrocytes in the neuroprotective actions of 17β-estradiol and selective estrogen receptor modulators. Mol Cell Endocrinol 2014; 389:48-57. [PMID: 24444786 DOI: 10.1016/j.mce.2014.01.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/08/2014] [Accepted: 01/08/2014] [Indexed: 01/04/2023]
Abstract
Neuroprotective actions of 17β-estradiol (estradiol) are in part mediated by direct actions on neurons. Astrocytes, which play an essential role in the maintenance of the homeostasis of neural tissue, express estrogen receptors and are also involved in the neuroprotective actions of estradiol in the brain. Estradiol controls gliosis and regulates neuroinflammation, edema and glutamate transport acting on astrocytes. In addition, the hormone regulates the release of neurotrophic factors and other neuroprotective molecules by astrocytes. In addition, reactive astrocytes are a local source of neuroprotective estradiol for the injured brain. Since estradiol therapy is not free from peripheral risks, alternatives for the hormone have been explored. Some selective estrogen receptor modulators (SERMs), which are already in use in clinical practice for the treatment of breast cancer, osteoporosis or menopausal symptoms, exert similar actions to estradiol on astrocytes. Therefore, SERMs represent therapeutic alternatives to estradiol for the activation of astroglia-mediated neuroprotective mechanisms.
Collapse
Affiliation(s)
| | | | - Iñigo Azcoitia
- Departamento de Biología Celular, Facultad de Biología, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | | | | |
Collapse
|
32
|
Ge Y, Wu F, Sun X, Xiang Z, Yang L, Huang S, Lu Z, Sun Y, Yu WF. Intrathecal infusion of hydrogen-rich normal saline attenuates neuropathic pain via inhibition of activation of spinal astrocytes and microglia in rats. PLoS One 2014; 9:e97436. [PMID: 24857932 PMCID: PMC4032255 DOI: 10.1371/journal.pone.0097436] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 04/17/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Reactive oxygen and nitrogen species are key molecules that mediate neuropathic pain. Although hydrogen is an established antioxidant, its effect on chronic pain has not been characterized. This study was to investigate the efficacy and mechanisms of hydrogen-rich normal saline induced analgesia. METHODOLOGY/PRINCIPAL FINDINGS In a rat model of neuropathic pain induced by L5 spinal nerve ligation (L5 SNL), intrathecal injection of hydrogen-rich normal saline relieved L5 SNL-induced mechanical allodynia and thermal hyperalgesia. Importantly, repeated administration of hydrogen-rich normal saline did not lead to tolerance. Preemptive treatment with hydrogen-rich normal saline prevented development of neuropathic pain behavior. Immunofluorochrome analysis revealed that hydrogen-rich normal saline treatment significantly attenuated L5 SNL-induced increase of 8-hydroxyguanosine immunoreactive cells in the ipsilateral spinal dorsal horn. Western blot analysis of SDS/PAGE-fractionated tyrosine-nitrated proteins showed that L5 SNL led to increased expression of tyrosine-nitrated Mn-containing superoxide dismutase (MnSOD) in the spinal cord, and hydrogen-rich normal saline administration reversed the tyrosine-nitrated MnSOD overexpression. We also showed that the analgesic effect of hydrogen-rich normal saline was associated with decreased activation of astrocytes and microglia, attenuated expression of interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) in the spinal cord. CONCLUSION/SIGNIFICANCE Intrathecal injection of hydrogen-rich normal saline produced analgesic effect in neuropathic rat. Hydrogen-rich normal saline-induced analgesia in neuropathic rats is mediated by reducing the activation of spinal astrocytes and microglia, which is induced by overproduction of hydroxyl and peroxynitrite.
Collapse
Affiliation(s)
- Yanhu Ge
- Department of Anesthesiology, Eastern Hepatobiliary Surgery Hospital, Changhai Hospital, Second Military Medical University, Shanghai, China
- Department of Anesthesiology, 309th Hospital of CPLA, Beijing, China
| | - Feixiang Wu
- Department of Anesthesiology, Eastern Hepatobiliary Surgery Hospital, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xuejun Sun
- Department of Diving Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Zhenghua Xiang
- Department of Neurobiology, Second Military Medical University, Shanghai, China
| | - Liqun Yang
- Department of Anesthesiology, Eastern Hepatobiliary Surgery Hospital, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Shengdong Huang
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhijie Lu
- Department of Anesthesiology, Eastern Hepatobiliary Surgery Hospital, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuming Sun
- Department of Anesthesiology, Eastern Hepatobiliary Surgery Hospital, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Wei-Feng Yu
- Department of Anesthesiology, Eastern Hepatobiliary Surgery Hospital, Changhai Hospital, Second Military Medical University, Shanghai, China
- * E-mail:
| |
Collapse
|