1
|
Ulloa M, Macías F, Clapp C, Martínez de la Escalera G, Arnold E. Prolactin is an Endogenous Antioxidant Factor in Astrocytes That Limits Oxidative Stress-Induced Astrocytic Cell Death via the STAT3/NRF2 Signaling Pathway. Neurochem Res 2024; 49:1879-1901. [PMID: 38755517 PMCID: PMC11144156 DOI: 10.1007/s11064-024-04147-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/29/2024] [Accepted: 05/02/2024] [Indexed: 05/18/2024]
Abstract
Oxidative stress-induced death of neurons and astrocytes contributes to the pathogenesis of numerous neurodegenerative diseases. While significant progress has been made in identifying neuroprotective molecules against neuronal oxidative damage, little is known about their counterparts for astrocytes. Prolactin (PRL), a hormone known to stimulate astroglial proliferation, viability, and cytokine expression, exhibits antioxidant effects in neurons. However, its role in protecting astrocytes from oxidative stress remains unexplored. Here, we investigated the effect of PRL against hydrogen peroxide (H2O2)-induced oxidative insult in primary cortical astrocyte cultures. Incubation of astrocytes with PRL led to increased enzymatic activity of superoxide dismutase (SOD) and glutathione peroxidase (GPX), resulting in higher total antioxidant capacity. Concomitantly, PRL prevented H2O2-induced cell death, reactive oxygen species accumulation, and protein and lipid oxidation. The protective effect of PRL upon H2O2-induced cell death can be explained by the activation of both signal transducer and activator of transcription 3 (STAT3) and NFE2 like bZIP transcription factor 2 (NRF2) transduction cascades. We demonstrated that PRL induced nuclear translocation and transcriptional upregulation of Nrf2, concurrently with the transcriptional upregulation of the NRF2-dependent genes heme oxygenase 1, Sod1, Sod2, and Gpx1. Pharmacological blockade of STAT3 suppressed PRL-induced transcriptional upregulation of Nrf2, Sod1 and Gpx1 mRNA, and SOD and GPX activities. Furthermore, genetic ablation of the PRL receptor increased astroglial susceptibility to H2O2-induced cell death and superoxide accumulation, while diminishing their intrinsic antioxidant capacity. Overall, these findings unveil PRL as a potent antioxidant hormone that protects astrocytes from oxidative insult, which may contribute to brain neuroprotection.
Collapse
Affiliation(s)
- Miriam Ulloa
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, 76230, Querétaro, México
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Mexico City, México
| | - Fernando Macías
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, 76230, Querétaro, México
| | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, 76230, Querétaro, México
| | | | - Edith Arnold
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, 76230, Querétaro, México.
- CONAHCYT-Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, Querétaro, México.
| |
Collapse
|
2
|
Effects of Antioxidant Gene Overexpression on Stress Resistance and Malignization In Vitro and In Vivo: A Review. Antioxidants (Basel) 2022; 11:antiox11122316. [PMID: 36552527 PMCID: PMC9774954 DOI: 10.3390/antiox11122316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Reactive oxygen species (ROS) are normal products of a number of biochemical reactions and are important signaling molecules. However, at the same time, they are toxic to cells and have to be strictly regulated by their antioxidant systems. The etiology and pathogenesis of many diseases are associated with increased ROS levels, and many external stress factors directly or indirectly cause oxidative stress in cells. Within this context, the overexpression of genes encoding the proteins in antioxidant systems seems to have become a viable approach to decrease the oxidative stress caused by pathological conditions and to increase cellular stress resistance. However, such manipulations unavoidably lead to side effects, the most dangerous of which is an increased probability of healthy tissue malignization or increased tumor aggression. The aims of the present review were to collect and systematize the results of studies devoted to the effects resulting from the overexpression of antioxidant system genes on stress resistance and carcinogenesis in vitro and in vivo. In most cases, the overexpression of these genes was shown to increase cell and organism resistances to factors that induce oxidative and genotoxic stress but to also have different effects on cancer initiation and promotion. The last fact greatly limits perspectives of such manipulations in practice. The overexpression of GPX3 and SOD3 encoding secreted proteins seems to be the "safest" among the genes that can increase cell resistance to oxidative stress. High efficiency and safety potential can also be found for SOD2 overexpression in combinations with GPX1 or CAT and for similar combinations that lead to no significant changes in H2O2 levels. Accumulation, systematization, and the integral analysis of data on antioxidant gene overexpression effects can help to develop approaches for practical uses in biomedical and agricultural areas. Additionally, a number of factors such as genetic and functional context, cell and tissue type, differences in the function of transcripts of one and the same gene, regulatory interactions, and additional functions should be taken into account.
Collapse
|
3
|
Oyagbemi AA, Adebiyi OE, Adigun KO, Ogunpolu BS, Falayi OO, Hassan FO, Folarin OR, Adebayo AK, Adejumobi OA, Asenuga ER, Ola-Davies OE, Omobowale TO, Olopade JO, Saba AB, Adedapo AA, Nkadimeng SM, McGaw LJ, Oguntibeju OO, Yakubu MA. Clofibrate, a PPAR-α agonist, abrogates sodium fluoride-induced neuroinflammation, oxidative stress, and motor incoordination via modulation of GFAP/Iba-1/anti-calbindin signaling pathways. ENVIRONMENTAL TOXICOLOGY 2020; 35:242-253. [PMID: 31710167 DOI: 10.1002/tox.22861] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/20/2019] [Accepted: 10/10/2019] [Indexed: 06/10/2023]
Abstract
Fluoride is an environmental contaminant that is ubiquitously present in air, water, and soil. It is commonly added in minute quantity to drinking water, toothpaste, and mouth rinses to prevent tooth decay. Epidemiological findings have demonstrated that exposure to fluoride induced neurodevelopmental toxicity, developmental neurotoxicity, and motor disorders. The neuroprotective effect of clofibrate, a peroxisome proliferator-activated receptor alpha agonist, was investigated in the present study. Forty male Wistar rats were used for this study and randomly grouped into 10 rats per group as control, sodium fluoride (NaF) alone (300 ppm), NaF plus clofibrate (250 mg/kg), and NaF plus lisinopril (10 mg/kg), respectively, for 7 days. NaF was administered in drinking water while clofibrate and lisinopril were administered by oral gavage. Markers of neuronal inflammation and oxidative stress, acetylcholinesterase activity, and neurobehavioral (hanging wire and open field) tests were performed. Immunohistochemistry was performed on brain tissues, and they were probed with glial fibrillary acidic protein, ionized calcium-binding adaptor molecule 1, and cerebellar Ca2+ -binding protein calbindin-D28k. The results showed that NaF significantly increased of oxidative stress and neuroinflammation and inhibited AChE activity. Immunostaining showed reactive astrocytes, microgliosis, loss of dendritic spines, and arborization in Purkinje cells in rats administered only NaF. Neurobehavioral results showed that cotreatment of NaF with clofibrate improved muscular strength and locomotion, reduced anxiety, and significantly reduced astrocytic count. Overall, cotreatment of NaF with either clofibrate or lisinopril showed neuroprotective effects by mitigating neuronal inflammation and oxidative and motor incoordination. Hence, clofibrate could be seen as a novel drug candidate against neurodegeneration and motor disorders.
Collapse
Affiliation(s)
- Ademola A Oyagbemi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Olamide E Adebiyi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Kabirat O Adigun
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Blessing S Ogunpolu
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Olufunke O Falayi
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Fasilat O Hassan
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Oluwabusayo R Folarin
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Benin, Nigeria
- Department of Medical Laboratory Science, College of Health Sciences, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
| | - Adedeji K Adebayo
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Olumuyiwa A Adejumobi
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Ebunoluwa R Asenuga
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Benin, Nigeria
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Olufunke E Ola-Davies
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Temidayo O Omobowale
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - James O Olopade
- Department of Medical Laboratory Science, College of Health Sciences, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Adebowale B Saba
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Adeolu A Adedapo
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Sanah M Nkadimeng
- Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Faculty of Veterinary Science, Onderstepoort, South Africa
| | - Lyndy J McGaw
- Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Faculty of Veterinary Science, Onderstepoort, South Africa
| | - Oluwafemi O Oguntibeju
- Phytomedicine and Phytochemistry Group, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| | - Momoh A Yakubu
- Department of Environmental & Interdisciplinary Sciences, College of Science, Engineering & Technology, Texas Southern University, Houston, Texas
| |
Collapse
|
4
|
Carvalho-Silva M, Gomes LM, de Prá SDT, Wessler LB, Schuck PF, Scaini G, de Bem AF, Blum-Silva CH, Reginatto FH, de Oliveira J, Streck EL. Evidence of hippocampal astrogliosis and antioxidant imbalance after L-tyrosine chronic administration in rats. Metab Brain Dis 2020; 35:193-200. [PMID: 31705440 DOI: 10.1007/s11011-019-00511-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 10/25/2019] [Indexed: 12/13/2022]
Abstract
Tyrosinemia type II is a genetic disorder characterized by elevated blood levels of the amino acid tyrosine caused by the deficiency of tyrosine aminotransferase enzyme, resulting in neurologic and developmental difficulties in the patients. Although neurological sequelae are common in Tyrosinemia type II patients, the mechanisms involved are still poorly understood. The oxidative stress appears to be, at least in part, responsible for neurological complication in this inborn error metabolism. We observed that an acute injection of tyrosine in rats caused a massive oxidative stress in different brain structures. The glutathione system and superoxide dismutase enzyme are relevant antioxidant strategies of the cells and tissues, including in the brain. Other important point is the strong relation between oxidative damage and inflammatory events. Herein, we investigated the effects of chronic administration of tyrosine in the hippocampus of young rats, with emphasis in the activity of GSH related enzymes and superoxide dismutase enzyme, and the astrocytosis. We observed that rats exposed to high levels of tyrosine presented an increased content of tyrosine, which was associated with an increment in the activity of glutathione peroxidase and glutathione reductase as well as with a diminished activity of superoxide dismutase. This antioxidant imbalance was accompanied by enhanced glial fibrillary acidic protein immunoreactivity, a marker of astrocytes, in the brain area studied. In conclusion, hippocampus astrogliosis is also a characteristic of brain alteration in Tyrosinemia. In addition, the chronic exposition to high levels of tyrosine is associated with an alteration in the activity of fundamental antioxidant enzymes.
Collapse
Affiliation(s)
- Milena Carvalho-Silva
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
| | - Lara M Gomes
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
| | - Samira Dal-Toé de Prá
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
| | - Leticia B Wessler
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
| | - Patricia F Schuck
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
| | - Giselli Scaini
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Andreza Fabro de Bem
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Carlos H Blum-Silva
- Programa de Pós-graduação em Farmácia, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, Brazil
| | - Flávio H Reginatto
- Programa de Pós-graduação em Farmácia, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, Brazil
| | - Jade de Oliveira
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90035-000, Brazil
| | - Emilio L Streck
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil.
| |
Collapse
|
5
|
Yang Q, Zhao Z, Bai Z, Hou H, Yuan Y, Guo A, Li Y. Effects of mycorrhizae and water conditions on perennial ryegrass growth in rare earth tailings. RSC Adv 2019; 9:10881-10888. [PMID: 35515284 PMCID: PMC9062534 DOI: 10.1039/c8ra10442e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/27/2019] [Indexed: 11/21/2022] Open
Abstract
Mycorrhizal symbioses, which include plant roots and arbuscular mycorrhizal fungi (AMF), can significantly enhance plant resistance and promote the absorption of soil nutrients by plants. A greenhouse experiment was conducted to investigate the effects of three AMF species (Glomus mosses, Glomus etunicatum and Glomus versiforme) on the height, biomass, malondialdehyde (MDA) and proline contents and antioxidant enzyme activities of perennial ryegrass (Lolium perenne) under different water supply treatments. Potted experimental soil samples were collected from the abandoned rare earth tailings in Ganzhou, Jiangxi. The results showed that all three AMF species infected ryegrass under the different treatments. Under severe drought stress, G. mosses had the most significant effects on the promotion of ryegrass performance. After inoculation, the height and whole-plant biomass of ryegrass increased by 60.44% and 150%, respectively. In addition, inoculation with AMF significantly reduced the content of MDA and proline in the ryegrass leaves in all water supply treatments except the moderate drought stress treatment, in which there was no effect. The leaf antioxidant enzyme activity was also measured. The results showed that under severe drought stress, inoculation with Glomus mosses significantly increased the activities of CAT and SOD in ryegrass and enhanced the resistance of plants. A possible reason that AMF promotes host plant growth and enhances drought resistance is that AMF directly increases the absorption of soil water and minerals by host plant roots and indirectly improves the physiological metabolism of plants.
Collapse
Affiliation(s)
- Qiao Yang
- School of Land Science and Technology, China University of Geosciences Beijing 100083 P. R. China
| | - Zhongqiu Zhao
- School of Land Science and Technology, China University of Geosciences Beijing 100083 P. R. China
- Key Laboratory of Land Consolidation and Rehabilitation Ministry of Land and Resources Beijing 100035 China
| | - Zhongke Bai
- School of Land Science and Technology, China University of Geosciences Beijing 100083 P. R. China
- Key Laboratory of Land Consolidation and Rehabilitation Ministry of Land and Resources Beijing 100035 China
| | - Hong Hou
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences Beijing 100012 China
| | - Ye Yuan
- School of Land Science and Technology, China University of Geosciences Beijing 100083 P. R. China
| | - Anning Guo
- School of Land Science and Technology, China University of Geosciences Beijing 100083 P. R. China
| | - Yufeng Li
- School of Land Science and Technology, China University of Geosciences Beijing 100083 P. R. China
| |
Collapse
|
6
|
Zehlila A, Schaumann A, Mlouka AB, Bourguiba I, Hardouin J, Masmoudi O, Cosette P, Amri M, Jouenne T. Glioprotective effect of Ulva rigida extract against UVB cellular damages. ALGAL RES 2017. [DOI: 10.1016/j.algal.2017.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
7
|
Douiri S, Bahdoudi S, Hamdi Y, Cubì R, Basille M, Fournier A, Vaudry H, Tonon MC, Amri M, Vaudry D, Masmoudi-Kouki O. Involvement of endogenous antioxidant systems in the protective activity of pituitary adenylate cyclase-activating polypeptide against hydrogen peroxide-induced oxidative damages in cultured rat astrocytes. J Neurochem 2016; 137:913-30. [DOI: 10.1111/jnc.13614] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 02/09/2016] [Accepted: 02/24/2016] [Indexed: 02/04/2023]
Affiliation(s)
- Salma Douiri
- Laboratory of Functional Neurophysiology and Pathology; Research Unit UR/11ES09; Department of Biological Sciences; Faculty of Science of Tunis; University Tunis El Manar; Tunis Tunisia
| | - Seyma Bahdoudi
- Laboratory of Functional Neurophysiology and Pathology; Research Unit UR/11ES09; Department of Biological Sciences; Faculty of Science of Tunis; University Tunis El Manar; Tunis Tunisia
- Inserm U982; Laboratory of Neuronal and Neuroendocrine Communication and Differentiation; University of Rouen; Mont-Saint-Aignan France
| | - Yosra Hamdi
- Laboratory of Functional Neurophysiology and Pathology; Research Unit UR/11ES09; Department of Biological Sciences; Faculty of Science of Tunis; University Tunis El Manar; Tunis Tunisia
| | - Roger Cubì
- Inserm U982; Laboratory of Neuronal and Neuroendocrine Communication and Differentiation; University of Rouen; Mont-Saint-Aignan France
| | - Magali Basille
- Inserm U982; Laboratory of Neuronal and Neuroendocrine Communication and Differentiation; University of Rouen; Mont-Saint-Aignan France
- Regional Platform for Cell Imaging of Normandie (PRIMACEN); Institute for Biomedical Research and Innovation; University of Rouen; Mont-Saint-Aignan France
| | - Alain Fournier
- INRS - Institut Armand-Frappier; Laval Quebec Canada
- Laboratoire International Associé Samuel de Champlain; Institut Armand-Frappier; Laval Quebec Canada
- International Associated Laboratory Samuel de Champlain; University of Rouen; Mont-Saint-Aignan France
| | - Hubert Vaudry
- Inserm U982; Laboratory of Neuronal and Neuroendocrine Communication and Differentiation; University of Rouen; Mont-Saint-Aignan France
- Regional Platform for Cell Imaging of Normandie (PRIMACEN); Institute for Biomedical Research and Innovation; University of Rouen; Mont-Saint-Aignan France
- International Associated Laboratory Samuel de Champlain; University of Rouen; Mont-Saint-Aignan France
| | - Marie-Christine Tonon
- Inserm U982; Laboratory of Neuronal and Neuroendocrine Communication and Differentiation; University of Rouen; Mont-Saint-Aignan France
- Regional Platform for Cell Imaging of Normandie (PRIMACEN); Institute for Biomedical Research and Innovation; University of Rouen; Mont-Saint-Aignan France
| | - Mohamed Amri
- Laboratory of Functional Neurophysiology and Pathology; Research Unit UR/11ES09; Department of Biological Sciences; Faculty of Science of Tunis; University Tunis El Manar; Tunis Tunisia
| | - David Vaudry
- Inserm U982; Laboratory of Neuronal and Neuroendocrine Communication and Differentiation; University of Rouen; Mont-Saint-Aignan France
- Regional Platform for Cell Imaging of Normandie (PRIMACEN); Institute for Biomedical Research and Innovation; University of Rouen; Mont-Saint-Aignan France
- International Associated Laboratory Samuel de Champlain; University of Rouen; Mont-Saint-Aignan France
| | - Olfa Masmoudi-Kouki
- Laboratory of Functional Neurophysiology and Pathology; Research Unit UR/11ES09; Department of Biological Sciences; Faculty of Science of Tunis; University Tunis El Manar; Tunis Tunisia
| |
Collapse
|
8
|
Elmann A, Telerman A, Erlank H, Ofir R, Kashman Y, Beit-Yannai E. Achillolide A Protects Astrocytes against Oxidative Stress by Reducing Intracellular Reactive Oxygen Species and Interfering with Cell Signaling. Molecules 2016; 21:301. [PMID: 26950103 PMCID: PMC6274406 DOI: 10.3390/molecules21030301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 02/23/2016] [Accepted: 02/25/2016] [Indexed: 11/17/2022] Open
Abstract
Achillolide A is a natural sesquiterpene lactone that we have previously shown can inhibit microglial activation. In this study we present evidence for its beneficial effects on astrocytes under oxidative stress, a situation relevant to neurodegenerative diseases and brain injuries. Viability of brain astrocytes (primary cultures) was determined by lactate dehydrogenase (LDH) activity, intracellular ROS levels were detected using 2',7'-dichlorofluorescein diacetate, in vitro antioxidant activity was measured by differential pulse voltammetry, and protein phosphorylation was determined using specific ELISA kits. We have found that achillolide A prevented the H₂O₂-induced death of astrocytes, and attenuated the induced intracellular accumulation of reactive oxygen species (ROS). These activities could be attributed to the inhibition of the H₂O₂-induced phosphorylation of MAP/ERK kinase 1 (MEK1) and p44/42 mitogen-activated protein kinases (MAPK), and to the antioxidant activity of achillolide A, but not to H₂O₂ scavenging. This is the first study that demonstrates its protective effects on brain astrocytes, and its ability to interfere with MAPK activation. We propose that achillolide A deserves further evaluation for its potential to be developed as a drug for the prevention/treatment of neurodegenerative diseases and brain injuries where oxidative stress is part of the pathophysiology.
Collapse
Affiliation(s)
- Anat Elmann
- Department of Food Quality and Safety, The Volcani Center, Agricultural Research Organization, Bet Dagan 50250, Israel.
| | - Alona Telerman
- Department of Food Quality and Safety, The Volcani Center, Agricultural Research Organization, Bet Dagan 50250, Israel.
| | - Hilla Erlank
- Department of Food Quality and Safety, The Volcani Center, Agricultural Research Organization, Bet Dagan 50250, Israel.
| | - Rivka Ofir
- Dead Sea & Arava Science Center and Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheba 84105, Israel.
| | - Yoel Kashman
- Raymond and Beverly Sackler Faculty of Exact Sciences, School of chemistry, Tel Aviv University, Ramat Aviv 69978, Israel.
| | - Elie Beit-Yannai
- Clinical Biochemistry and Pharmacology Department, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheba 84105, Israel.
| |
Collapse
|
9
|
Lei XG, Zhu JH, Cheng WH, Bao Y, Ho YS, Reddi AR, Holmgren A, Arnér ESJ. Paradoxical Roles of Antioxidant Enzymes: Basic Mechanisms and Health Implications. Physiol Rev 2016; 96:307-64. [PMID: 26681794 DOI: 10.1152/physrev.00010.2014] [Citation(s) in RCA: 262] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are generated from aerobic metabolism, as a result of accidental electron leakage as well as regulated enzymatic processes. Because ROS/RNS can induce oxidative injury and act in redox signaling, enzymes metabolizing them will inherently promote either health or disease, depending on the physiological context. It is thus misleading to consider conventionally called antioxidant enzymes to be largely, if not exclusively, health protective. Because such a notion is nonetheless common, we herein attempt to rationalize why this simplistic view should be avoided. First we give an updated summary of physiological phenotypes triggered in mouse models of overexpression or knockout of major antioxidant enzymes. Subsequently, we focus on a series of striking cases that demonstrate "paradoxical" outcomes, i.e., increased fitness upon deletion of antioxidant enzymes or disease triggered by their overexpression. We elaborate mechanisms by which these phenotypes are mediated via chemical, biological, and metabolic interactions of the antioxidant enzymes with their substrates, downstream events, and cellular context. Furthermore, we propose that novel treatments of antioxidant enzyme-related human diseases may be enabled by deliberate targeting of dual roles of the pertaining enzymes. We also discuss the potential of "antioxidant" nutrients and phytochemicals, via regulating the expression or function of antioxidant enzymes, in preventing, treating, or aggravating chronic diseases. We conclude that "paradoxical" roles of antioxidant enzymes in physiology, health, and disease derive from sophisticated molecular mechanisms of redox biology and metabolic homeostasis. Simply viewing antioxidant enzymes as always being beneficial is not only conceptually misleading but also clinically hazardous if such notions underpin medical treatment protocols based on modulation of redox pathways.
Collapse
Affiliation(s)
- Xin Gen Lei
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jian-Hong Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Wen-Hsing Cheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Yongping Bao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ye-Shih Ho
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Amit R Reddi
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Arne Holmgren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Elias S J Arnér
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Ouyang YB, Stary CM, White RE, Giffard RG. The use of microRNAs to modulate redox and immune response to stroke. Antioxid Redox Signal 2015; 22:187-202. [PMID: 24359188 PMCID: PMC4281877 DOI: 10.1089/ars.2013.5757] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SIGNIFICANCE Cerebral ischemia is a major cause of death and disability throughout the world, yet therapeutic options remain limited. The interplay between the cellular redox state and the immune response plays a critical role in determining the extent of neural cell injury after ischemia and reperfusion. Excessive amounts of reactive oxygen species (ROS) generated by mitochondria and other sources act both as triggers and effectors of inflammation. This review will focus on the interplay between these two mechanisms. RECENT ADVANCES MicroRNAs (miRNAs) are important post-transcriptional regulators that interact with multiple target messenger RNAs coordinately regulating target genes, including those involved in controlling mitochondrial function, redox state, and inflammatory pathways. This review will focus on the regulation of mitochondria, ROS, and inflammation by miRNAs in the chain of deleterious intra- and intercellular events that lead to brain cell death after cerebral ischemia. CRITICAL ISSUES Although pretreatment using miRNAs was effective in cerebral ischemia in rodents, testing treatment after the onset of ischemia is an essential next step in the development of acute stroke treatment. In addition, miRNA formulation and delivery into the CNS remain a challenge in the clinical translation of miRNA therapy. FUTURE DIRECTIONS Future research should focus on post-treatment and potential clinical use of miRNAs.
Collapse
Affiliation(s)
- Yi-Bing Ouyang
- Department of Anesthesia, Stanford University School of Medicine , Stanford, California
| | | | | | | |
Collapse
|
11
|
Oxaliplatin neurotoxicity involves peroxisome alterations. PPARγ agonism as preventive pharmacological approach. PLoS One 2014; 9:e102758. [PMID: 25036594 PMCID: PMC4103888 DOI: 10.1371/journal.pone.0102758] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 06/23/2014] [Indexed: 02/07/2023] Open
Abstract
The development of neuropathic syndromes is an important, dose limiting side effect of anticancer agents like platinum derivates, taxanes and vinca alkaloids. The causes of neurotoxicity are still unclear but the impairment of the oxidative equilibrium is strictly related to pain. Two intracellular organelles, mitochondria and peroxisomes cooperate to the maintaining of the redox cellular state. Whereas a relationship between chemotherapy-dependent mitochondrial alteration and neuropathy has been established, the role of peroxisome is poor explored. In order to study the mechanisms of oxaliplatin-induced neurotoxicity, peroxisomal involvement was evaluated in vitro and in vivo. In primary rat astrocyte cell culture, oxaliplatin (10 µM for 48 h or 1 µM for 5 days) increased the number of peroxisomes, nevertheless expression and functionality of catalase, the most important antioxidant defense enzyme in mammalian peroxisomes, were significantly reduced. Five day incubation with the selective Peroxisome Proliferator Activated Receptor-γ (PPAR-γ) antagonist G3335 (30 µM) induced a similar peroxisomal impairment suggesting a relationship between PPARγ signaling and oxaliplatin neurotoxicity. The PPARγ agonist rosiglitazone (10 µM) reduced the harmful effects induced both by G3335 and oxaliplatin. In vivo, in a rat model of oxaliplatin induced neuropathy, a repeated treatment with rosiglitazone (3 and 10 mg kg−1 per os) significantly reduced neuropathic pain evoked by noxious (Paw pressure test) and non-noxious (Cold plate test) stimuli. The behavioral effect paralleled with the prevention of catalase impairment induced by oxaliplatin in dorsal root ganglia. In the spinal cord, catalase protection was showed by the lower rosiglitazone dosage without effect on the astrocyte density increase induced by oxaliplatin. Rosiglitazone did not alter the oxaliplatin-induced mortality of the human colon cancer cell line HT-29. These results highlight the role of peroxisomes in oxaliplatin-dependent nervous damage and suggest PPARγ stimulation as a candidate to counteract oxaliplatin neurotoxicity.
Collapse
|
12
|
Le HT, Sin WC, Lozinsky S, Bechberger J, Vega JL, Guo XQ, Sáez JC, Naus CC. Gap junction intercellular communication mediated by connexin43 in astrocytes is essential for their resistance to oxidative stress. J Biol Chem 2013; 289:1345-54. [PMID: 24302722 DOI: 10.1074/jbc.m113.508390] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Oxidative stress induced by reactive oxygen species (ROS) is associated with various neurological disorders including aging, neurodegenerative diseases, as well as traumatic and ischemic insults. Astrocytes have an important role in the anti-oxidative defense in the brain. The gap junction protein connexin43 (Cx43) forms intercellular channels as well as hemichannels in astrocytes. In the present study, we investigated the contribution of Cx43 to astrocytic death induced by the ROS hydrogen peroxide (H2O2) and the mechanism by which Cx43 exerts its effects. Lack of Cx43 expression or blockage of Cx43 channels resulted in increased ROS-induced astrocytic death, supporting a cell protective effect of functional Cx43 channels. H2O2 transiently increased hemichannel activity, but reduced gap junction intercellular communication (GJIC). GJIC in wild-type astrocytes recovered after 7 h, but was absent in Cx43 knock-out astrocytes. Blockage of Cx43 hemichannels incompletely inhibited H2O2-induced hemichannel activity, indicating the presence of other hemichannel proteins. Panx1, which is predicted to be a major hemichannel contributor in astrocytes, did not appear to have any cell protective effect from H2O2 insults. Our data suggest that GJIC is important for Cx43-mediated ROS resistance. In contrast to hypoxia/reoxygenation, H2O2 treatment decreased the ratio of the hypophosphorylated isoform to total Cx43 level. Cx43 has been reported to promote astrocytic death induced by hypoxia/reoxygenation. We therefore speculate the increase in Cx43 dephosphorylation may account for the facilitation of astrocytic death. Our findings suggest that the role of Cx43 in response to cellular stress is dependent on the activation of signaling pathways leading to alteration of Cx43 phosphorylation states.
Collapse
Affiliation(s)
- Hoa T Le
- From the Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3 Canada
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Di Cesare Mannelli L, Zanardelli M, Failli P, Ghelardini C. Oxaliplatin-induced oxidative stress in nervous system-derived cellular models: could it correlate with in vivo neuropathy? Free Radic Biol Med 2013; 61:143-50. [PMID: 23548635 DOI: 10.1016/j.freeradbiomed.2013.03.019] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 02/11/2013] [Accepted: 03/25/2013] [Indexed: 01/17/2023]
Abstract
Oxaliplatin is a platinum-organic drug with antineoplastic properties used for colorectal cancer. With respect to the other platinum derivates oxaliplatin induces only a mild hematological and gastrointestinal toxicity. Its limiting side effect is its neurotoxicity, which results in a sensory neuropathy. Repeated oxaliplatin treatment in the rat led to a neuropathic pain characterized by a significant oxidative damage throughout the nervous system. The natural antioxidants silibinin and α-tocopherol reduce redox alteration and prevent pain. Starting from the "oxidative hypothesis" as a molecular basis of chemotherapy-induced neurotoxicity, we decided to explore deep inside the mechanisms of oxaliplatin neurotoxicity and search for a cellular system useful for screening antioxidant compounds that can reduce oxaliplatin neurotoxicity. Focusing on various constituents of the central nervous system, we used the neuronal-derived cell line SH-SY5Y and primary cultures of rat cortical astrocytes. Oxaliplatin significantly increased superoxide anion production and induced lipid peroxidation (malonyldialdehyde levels) and protein (carbonylated proteins) and DNA oxidation (8-OH-dG levels). Silibinin and α-tocopherol (10µM) were able to reduce the oxidative damage in both cell types. These antioxidants fully protected astrocytes from the caspase 3 apoptotic signaling activation induced by oxaliplatin. The damage prevention effects of silibinin and α-tocopherol on nervous system-derived cells did not interfere with the oxaliplatin antineoplastic in vitro mechanism as evaluated on a human colon adenocarcinoma cell line (HT29). Moreover, neither silibinin nor α-tocopherol modified the oxaliplatin-induced apoptosis in HT29 cells, suggesting a different antiapoptotic profile in normal vs tumoral cells for these antioxidant compounds. In conclusion, because data obtained in in vitro cellular models parallel the in vivo study we propose cell models to investigate oxaliplatin neurotoxicity and to screen possible therapeutic adjuvant agents.
Collapse
Affiliation(s)
- L Di Cesare Mannelli
- Department of Preclinical and Clinical Pharmacology, University of Florence, 50139 Florence, Italy.
| | - M Zanardelli
- Department of Preclinical and Clinical Pharmacology, University of Florence, 50139 Florence, Italy
| | - P Failli
- Department of Preclinical and Clinical Pharmacology, University of Florence, 50139 Florence, Italy
| | - C Ghelardini
- Department of Preclinical and Clinical Pharmacology, University of Florence, 50139 Florence, Italy
| |
Collapse
|
14
|
Parabucki AB, Bozić ID, Bjelobaba IM, Lavrnja IC, Brkić PD, Jovanović TS, Savić DZ, Stojiljković MB, Peković SM. Hyperbaric oxygenation alters temporal expression pattern of superoxide dismutase 2 after cortical stab injury in rats. Croat Med J 2013; 53:586-97. [PMID: 23275324 PMCID: PMC3547292 DOI: 10.3325/cmj.2012.53.586] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Aim To evaluate the effect of hyperbaric oxygen therapy (HBOT) on superoxide dismutase 2 (SOD2) expression pattern after the cortical stab injury (CSI). Methods CSI was performed on 88 male Wistar rats, divided into control, sham, lesioned, and HBO groups. HBOT protocol was the following: pressure applied was 2.5 absolute atmospheres, for 60 minutes, once a day for consecutive 3 or 10 days. The pattern of SOD2 expression and cellular localization was analyzed using real-time polymerase chain reaction, Western blot, and double-label fluorescence immunohistochemistry. Neurons undergoing degeneration were visualized with Fluoro-Jade®B. Results CSI induced significant transient increase in SOD2 protein levels at day 3 post injury, which was followed by a reduction toward control levels at post-injury day 10. At the same time points, mRNA levels for SOD2 in the injured cortex were down-regulated. Exposure to HBO for 3 days considerably down-regulated SOD2 protein levels in the injured cortex, while after 10 days of HBOT an up-regulation of SOD2 was observed. HBOT significantly increased mRNA levels for SOD2 at both time points compared to the corresponding L group, but they were still lower than in controls. Double immunofluorescence staining revealed that 3 days after CSI, up-regulation of SOD2 was mostly due to an increased expression in reactive astrocytes surrounding the lesion site. HBOT attenuated SOD2 expression both in neuronal and astroglial cells. Fluoro-Jade®B labeling showed that HBOT significantly decreased the number of degenerating neurons in the injured cortex. Conclusion HBOT alters SOD2 protein and mRNA levels after brain injury in a time-dependent manner.
Collapse
Affiliation(s)
- Ana B Parabucki
- Department of Neurobiology, Institute for Biological Research Sinisa Stankovic, University of Belgrade, Blvd Despota Stefana 142, 11060 Belgrade, Serbia.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Genetic evidence of an evolutionarily conserved role for Nrf2 in the protection against oxidative stress. Mol Cell Biol 2012; 32:4455-61. [PMID: 22949501 DOI: 10.1128/mcb.00481-12] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Transcription factor Nrf2 is considered a master regulator of antioxidant defense in mammals. However, it is unclear whether this concept is applicable to nonmammalian vertebrates, because no animal model other than Nrf2 knockout mice has been generated to examine the effects of Nrf2 deficiency. Here, we characterized a recessive loss-of-function mutant of Nrf2 (nrf2(fh318)) in a lower vertebrate, the zebrafish (Danio rerio). In keeping with the findings in the mouse model, nrf2(fh318) mutants exhibited reduced induction of the Nrf2 target genes in response to oxidative stress and electrophiles but were viable and fertile, and their embryos developed normally. The nrf2(fh318) larvae displayed enhanced sensitivity to oxidative stress and electrophiles, especially peroxides, and pretreatment with an Nrf2-activating compound, sulforaphane, decreased peroxide-induced lethality in the wild type but not nrf2(fh318) mutants, indicating that resistance to oxidative stress is highly dependent on Nrf2 functions. These results reveal an evolutionarily conserved role of vertebrate Nrf2 in protection against oxidative stress. Interestingly, there were no significant differences between wild-type and nrf2(fh318) larvae with regard to their sensitivity to superoxide and singlet oxygen generators, suggesting that the importance of Nrf2 in oxidative stress protection varies based on the type of reactive oxygen species (ROS).
Collapse
|
16
|
Hamdi Y, Kaddour H, Vaudry D, Bahdoudi S, Douiri S, Leprince J, Castel H, Vaudry H, Tonon MC, Amri M, Masmoudi-Kouki O. The octadecaneuropeptide ODN protects astrocytes against hydrogen peroxide-induced apoptosis via a PKA/MAPK-dependent mechanism. PLoS One 2012; 7:e42498. [PMID: 22927932 PMCID: PMC3424241 DOI: 10.1371/journal.pone.0042498] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 07/06/2012] [Indexed: 12/18/2022] Open
Abstract
Astrocytes synthesize and release endozepines, a family of regulatory peptides, including the octadecaneuropeptide (ODN) an endogenous ligand of both central-type benzodiazepine (CBR) and metabotropic receptors. We have recently shown that ODN exerts a protective effect against hydrogen peroxide (H2O2)-induced oxidative stress in astrocytes. The purpose of the present study was to determine the type of receptor and the transduction pathways involved in the protective effect of ODN in cultured rat astrocytes. We have first observed a protective activity of ODN at very low concentrations that was abrogated by the metabotropic ODN receptor antagonist cyclo1–8[DLeu5]OP, but not by the CBR antagonist flumazenil. We have also found that the metabotropic ODN receptor is positively coupled to adenylyl cyclase in astrocytes and that the glioprotective action of ODN upon H2O2-induced astrocyte death is PKA- and MEK-dependent, but PLC/PKC-independent. Downstream of PKA, ODN induced ERK phosphorylation, which in turn activated the expression of the anti-apoptotic gene Bcl-2 and blocked the stimulation by H2O2 of the pro-apoptotic gene Bax. The effect of ODN on the Bax/Bcl-2 balance contributed to abolish the deleterious action of H2O2 on mitochondrial membrane integrity and caspase-3 activation. Finally, the inhibitory effect of ODN on caspase-3 activity was shown to be PKA and MEK-dependent. In conclusion, the present results demonstrate that the potent glioprotective action of ODN against oxidative stress involves the metabotropic ODN receptor coupled to the PKA/ERK-kinase pathway to inhibit caspase-3 activation.
Collapse
Affiliation(s)
- Yosra Hamdi
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Hadhemi Kaddour
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El Manar, Tunis, Tunisia
| | - David Vaudry
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of Rouen, Mont-Saint-Aignan, France
- International Associated Laboratory Samuel de Champlain, Mont-Saint-Aignan, France
- Regional Platform for Cell Imaging of Haute-Normandie (PRIMACEN), Institute for Medical Research and Innovation (IRIB), University of Rouen, Mont-Saint-Aignan, France
| | - Seyma Bahdoudi
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Salma Douiri
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Jérôme Leprince
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of Rouen, Mont-Saint-Aignan, France
- International Associated Laboratory Samuel de Champlain, Mont-Saint-Aignan, France
- Regional Platform for Cell Imaging of Haute-Normandie (PRIMACEN), Institute for Medical Research and Innovation (IRIB), University of Rouen, Mont-Saint-Aignan, France
| | - Helene Castel
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of Rouen, Mont-Saint-Aignan, France
| | - Hubert Vaudry
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of Rouen, Mont-Saint-Aignan, France
- International Associated Laboratory Samuel de Champlain, Mont-Saint-Aignan, France
- Regional Platform for Cell Imaging of Haute-Normandie (PRIMACEN), Institute for Medical Research and Innovation (IRIB), University of Rouen, Mont-Saint-Aignan, France
- * E-mail: (MA), (HV)
| | - Marie-Christine Tonon
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of Rouen, Mont-Saint-Aignan, France
| | - Mohamed Amri
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El Manar, Tunis, Tunisia
- * E-mail: (MA), (HV)
| | - Olfa Masmoudi-Kouki
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
17
|
Fisahn A, Lavebratt C, Canlon B. Acoustic startle hypersensitivity in Mceph mice and its effect on hippocampal excitability. Eur J Neurosci 2012; 34:1121-30. [PMID: 21966978 DOI: 10.1111/j.1460-9568.2011.07834.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Current therapies and research for epilepsy concentrate mainly on controlling the disease, but not on prevention of its development and progression. This is partly due to the under-appreciated heterogeneity of the different epileptic syndromes, and a lack of knowledge about the underlying mechanisms of hypersensitivity and hypersynchrony in epilepsy development and spread. In this study we investigate mechanisms underlying the increased susceptibility to acoustic startle in a mouse model homozygous for the spontaneous megencephaly (mceph) mutation, which results in a lack of the functional potassium channel Kv1.1. Mceph mice are hypersensitive to acoustic startle, a response that is not seen in the wild-type (WT) littermates. After acoustic startle, a strong activation of astrocytes, as indicated by glial fibrillary acidic protein, occurred in the inferior colliculus and hippocampus. Both the hypersensitivity of acoustic startle as well as activation of astrocytes could be maintained at WT levels by pre-treating the Mceph mice with the anti-epileptic drug valproate. Furthermore, we utilized the Mceph mouse model to investigate whether acoustic startle-induced hypersensitivity has negative consequences for synchronous neuronal activity in other, non-auditory, systems and networks in the brain, such as the hippocampus. Our findings show that acoustic startle-induced hypersensitivity primes hippocampal networks by increasing their excitability, which results in increased strength of rhythmic network activity. Our results provide novel insights into the mechanisms that underlie the spread of hypersensitivity and hypersynchrony across functionally different parts of the brain.
Collapse
Affiliation(s)
- André Fisahn
- Neuronal Oscillations Laboratory, KI-Alzheimer's Disease Research Center, NVS, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
18
|
Hamdi Y, Kaddour H, Vaudry D, Douiri S, Bahdoudi S, Leprince J, Castel H, Vaudry H, Amri M, Tonon MC, Masmoudi-Kouki O. The stimulatory effect of the octadecaneuropeptide ODN on astroglial antioxidant enzyme systems is mediated through a GPCR. Front Endocrinol (Lausanne) 2012; 3:138. [PMID: 23181054 PMCID: PMC3502939 DOI: 10.3389/fendo.2012.00138] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Astroglial cells possess an array of cellular defense systems, including superoxide dismutase (SOD) and catalase antioxidant enzymes, to prevent damage caused by oxidative stress on the central nervous system. Astrocytes specifically synthesize and release endozepines, a family of regulatory peptides including the octadecaneuropeptide (ODN). ODN is the ligand of both central-type benzodiazepine receptors (CBR), and an adenylyl cyclase- and phospholipase C-coupled receptor. We have recently shown that ODN is a potent protective agent that prevents hydrogen peroxide (H(2)O(2))-induced inhibition of SOD and catalase activities and stimulation of cell apoptosis in astrocytes. The purpose of the present study was to investigate the type of receptor involved in ODN-induced inhibition of SOD and catalase in cultured rat astrocytes. We found that ODN induced a rapid stimulation of SOD and catalase gene transcription in a concentration-dependent manner. In addition, 0.1 nM ODN blocked H(2)O(2)-evoked reduction of both mRNA levels and activities of SOD and catalase. Furthermore, the inhibitory actions of ODN on the deleterious effects of H(2)O(2) on SOD and catalase were abrogated by the metabotropic ODN receptor antagonist cyclo(1-8)[Dleu(5)]OP, but not by the CBR antagonist flumazenil. Finally, the protective action of ODN against H(2)O(2)-evoked inhibition of endogenous antioxidant systems in astrocytes was protein kinase A (PKA)-dependent, but protein kinase C-independent. Taken together, these data demonstrate for the first time that ODN, acting through its metabotropic receptor coupled to the PKA pathway, prevents oxidative stress-induced alteration of antioxidant enzyme expression and activities. The peptide ODN is thus a potential candidate for the development of specific agonists that would selectively mimic its protective activity.
Collapse
Affiliation(s)
- Yosra Hamdi
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
| | - Hadhemi Kaddour
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
| | - David Vaudry
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of RouenMont-Saint-Aignan, France
- International Associated Laboratory Samuel de ChamplainMont-Saint-Aignan, France
- Regional Platform for Cell Imaging of Haute-Normandie, Institute for Medical Research and Innovation, University of RouenMont-Saint-Aignan, France
| | - Salma Douiri
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
| | - Seyma Bahdoudi
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
| | - Jérôme Leprince
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of RouenMont-Saint-Aignan, France
- International Associated Laboratory Samuel de ChamplainMont-Saint-Aignan, France
| | - Hélène Castel
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of RouenMont-Saint-Aignan, France
| | - Hubert Vaudry
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of RouenMont-Saint-Aignan, France
- International Associated Laboratory Samuel de ChamplainMont-Saint-Aignan, France
- *Correspondence: Mohamed Amri, Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El Manar, 2092 Tunis, Tunisia. e-mail: ; Hubert Vaudry, Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, International Associated Laboratory Samuel de Champlain, Regional Platform for Cell Imaging of Haute-Normandie, Institute for Medical Research and Innovation, University of Rouen, 76821 Mont-Saint-Aignan, France. e-mail:
| | - Mohamed Amri
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
- *Correspondence: Mohamed Amri, Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El Manar, 2092 Tunis, Tunisia. e-mail: ; Hubert Vaudry, Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, International Associated Laboratory Samuel de Champlain, Regional Platform for Cell Imaging of Haute-Normandie, Institute for Medical Research and Innovation, University of Rouen, 76821 Mont-Saint-Aignan, France. e-mail:
| | - Marie-Christine Tonon
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of RouenMont-Saint-Aignan, France
| | - Olfa Masmoudi-Kouki
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
| |
Collapse
|
19
|
Hamdi Y, Masmoudi-Kouki O, Kaddour H, Belhadj F, Gandolfo P, Vaudry D, Mokni M, Leprince J, Hachem R, Vaudry H, Tonon MC, Amri M. Protective effect of the octadecaneuropeptide on hydrogen peroxide-induced oxidative stress and cell death in cultured rat astrocytes. J Neurochem 2011; 118:416-28. [DOI: 10.1111/j.1471-4159.2011.07315.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
20
|
Ourednik V, Ourednik J, Xu Y, Zhang Y, Lynch WP, Snyder EY, Schachner M. Cross-Talk Between Stem Cells and the Dysfunctional Brain is Facilitated by Manipulating the Niche: Evidence from an Adhesion Molecule. Stem Cells 2009; 27:2846-56. [DOI: 10.1002/stem.227] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
21
|
Targeting superoxide dismutase 1 to overcome cisplatin resistance in human ovarian cancer. Cancer Chemother Pharmacol 2008; 63:723-30. [DOI: 10.1007/s00280-008-0791-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2008] [Accepted: 06/17/2008] [Indexed: 10/21/2022]
|
22
|
Li Y, Bao Y, Jiang B, Wang Z, Liu Y, Zhang C, An L. Catalpol protects primary cultured astrocytes from in vitro ischemia-induced damage. Int J Dev Neurosci 2008; 26:309-17. [PMID: 18337048 DOI: 10.1016/j.ijdevneu.2008.01.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Revised: 01/12/2008] [Accepted: 01/14/2008] [Indexed: 12/22/2022] Open
Abstract
Catalpol, an iridoid glycoside abundant in the roots of Rehmannia glutinosa, has been previously found to prevent the loss of CA1 hippocampal neurons and to reduce working errors in gerbils after ischemia-reperfusion injury. In the present study, we investigated the effects of catalpol on astrocytes in an ischemic model to further characterize its neuroprotective mechanisms. Primary cultured astrocytes exposed to oxygen-glucose deprivation (OGD) followed by reperfusion (adding back oxygen and glucose, OGD-R), were used as an in vitro ischemic model. Treatment of the astrocytes with catalpol during ischemia-reperfusion increased astrocyte survival significantly in a concentration-dependent manner, as demonstrated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, lactate dehydrogenase (LDH) release and morphological observation. In addition, catalpol prevented the decrease in mitochondrial membrane potential, inhibited the formation of reactive oxygen species (ROS) and the production of nitric oxide (NO), decreased the level of lipid peroxide and the activity of inducible nitric oxide synthase (iNOS), and elevated the activities of superoxide dismutase (SOD), glutathione peroxidase (GPx) and the content of glutathione (GSH). Our results suggest that catalpol exerts the most significant cytoprotective effect on astrocytes by suppressing the production of free radicals and elevating antioxidant capacity.
Collapse
Affiliation(s)
- Yachen Li
- School of Environmental and Biological Science & Technology, Dalian University of Technology, Dalian, Liaoning, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Tripanichkul W, Sripanichkulchai K, Duce JA, Finkelstein DI. 17β-Estradiol reduces nitrotyrosine immunoreactivity and increases SOD1 and SOD2 immunoreactivity in nigral neurons in male mice following MPTP insult. Brain Res 2007; 1164:24-31. [PMID: 17640623 DOI: 10.1016/j.brainres.2007.05.076] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 05/15/2007] [Accepted: 05/21/2007] [Indexed: 10/23/2022]
Abstract
Emerging evidence suggests the beneficial effects of estrogen on Parkinson's disease (PD), yet the mechanisms of action implicated remain elusive. While experimental evidence suggests that estrogen possesses potent antioxidative properties, it is still unknown whether the hormone exhibits a neuroprotection in a PD animal model through its antioxidant activities. This study therefore investigated the effects of 17beta-estradiol (E2) on the immunoreactivity of nigral neurons and glia for nitrotyrosine (NT, a stable marker for oxidative stress), Cu/Zn superoxide dismutase (SOD1) and Mn superoxide dismutase (SOD2) in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model. Adult male mice were treated with E2 or vehicle for 11 days during which they were injected with MPTP or saline on the sixth day. The brains were collected on day 11 and quantitative immunohistochemistry was used to assess the number of NT-, SOD1- and SOD2-immunoreactive (IR) cells in the substantia nigra pars compacta (SNpc). In saline-treated group, E2 decreased NT-IR neuronal number and raised SOD1 and SOD2 expression in neurons and glia in the SNpc. MPTP induced a significant increase in the number of NT- and SOD2-IR neurons, but decreased the number of SOD1-IR neurons. MPTP also triggered a significant increase of SOD2- and SOD1-IR glial number. E2 pretreatment in MPTP mice reduced the number of NT-IR neurons, increased the number of SOD1- and SOD2-IR neurons, but did not alter the MPTP effect on glia immunoreactive to either SOD. Stimulation of SOD1 and SOD2 expression in nigral neurons suggests that E2 provides neuroprotection against MPTP-induced oxidative stress, partly through its ability to act as an antioxidant.
Collapse
Affiliation(s)
- Wanida Tripanichkul
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10110, Thailand
| | | | | | | |
Collapse
|
24
|
Lee YS, Song YS, Giffard RG, Chan PH. Biphasic role of nuclear factor-kappa B on cell survival and COX-2 expression in SOD1 Tg astrocytes after oxygen glucose deprivation. J Cereb Blood Flow Metab 2006; 26:1076-88. [PMID: 16395278 DOI: 10.1038/sj.jcbfm.9600261] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In cytoplasm, nuclear factor-kappaB (NF-kappaB) is associated with the inhibitory protein, IkappaBalpha. On activation by H2O2, IkappaBalpha is phosphorylated and degraded, exposing the nuclear localization signals on the NF-kappaB heterodimer. Cyclooxygenase-2 (COX-2), which mediates prostaglandin synthesis during inflammation, is induced by oxidative stress mediated by NF-kappaB. We investigated whether the NF-kappaB signaling pathway affected cell death and COX-2 expression after hypoxia-induced oxidative stress in wild-type (WT) and copper/zinc-superoxide dismutase transgenic (SOD1 Tg) astrocytes. In WT astrocytes, phospho-IkappaBalpha was highly expressed after oxygen-glucose deprivation (OGD) and 2 h of reperfusion, concomitant with the decrease in IkappaBalpha. The NF-kappaB p50 level increased similarly in WT and SOD1 Tg astrocytes (1.2-/1.4-fold) after OGD. Electrophoretic mobility shift assay showed higher DNA-binding activity of NF-kappaB p50 in WT than in SOD1 Tg astrocytes 6 h after 4 h of OGD. The COX-2 level was induced by 2.7- and 1.3-fold after OGD in WT and SOD1 Tg astrocytes, and an antioxidant protected both groups against OGD injury. Superoxide dismutase transgenic cells were 23% more protective against OGD injury than WTs when assessed by lactate dehydrogenase release. However, transfection of NF-kappaB small interfering RNAs in SOD1 Tg astrocytes aggravated cell death and increased COX-2 expression. These results suggest that the NF-kappaB signaling pathway induced COX-2 expression and promoted cell death in WTs after OGD injury; however, NF-kappaB activation protected cells and decreased COX-2 expression in SOD1 Tg astrocytes. This biphasic role of NF-kappaB might be coordinately regulated by reactive oxygen species levels in astrocytes, thereby functioning as a regulator of cell death/survival.
Collapse
Affiliation(s)
- Yong-Sun Lee
- Department of Neurosurgery, Department of Neurology and Neurological Sciences, and Program in Neurosciences, Stanford University School of Medicine, Stanford, California 94305-5487, USA
| | | | | | | |
Collapse
|
25
|
Laurent J, Paly E, Marche PN, London J. Early thymic T cell development in young transgenic mice overexpressing human Cu/Zn superoxide dismutase, a model of Down syndrome. Free Radic Biol Med 2006; 40:1971-80. [PMID: 16716898 DOI: 10.1016/j.freeradbiomed.2006.01.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Revised: 12/05/2005] [Accepted: 01/24/2006] [Indexed: 12/20/2022]
Abstract
Previous studies have shown that transgenic mice overexpressing Cu/Zn superoxide dismutase, a model of Down syndrome, exhibit premature thymic involution. We have performed a flow cytometry analysis of the developing thymus in these homozygous transgenic mice (hSOD1/hSOD1: Tg-SOD). Longitudinal follow-up analysis from day 3 to day 280 showed an early thymic development in Tg-SOD mice compared with controls. This early thymic development was associated with an increased migration of mature T cells to peripheral lymphoid organs. BrdU labeling showed no difference between Tg-SOD and control mice, confirming that the greater number of peripheral T cells in Tg-SOD mice was not due to extensive proliferation of these cells but rather to a greater pool of emigrant T cells in Tg-SOD.
Collapse
Affiliation(s)
- Julien Laurent
- CePo-Centre Pluridisciplinaire d'Oncology, Avenue PierreDecker, 4CH Lausanne, Switzerland
| | | | | | | |
Collapse
|
26
|
Peluffo H, Acarin L, Arís A, González P, Villaverde A, Castellano B, González B. Neuroprotection from NMDA excitotoxic lesion by Cu/Zn superoxide dismutase gene delivery to the postnatal rat brain by a modular protein vector. BMC Neurosci 2006; 7:35. [PMID: 16638118 PMCID: PMC1462999 DOI: 10.1186/1471-2202-7-35] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Accepted: 04/25/2006] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Superoxide mediated oxidative stress is a key neuropathologic mechanism in acute central nervous system injuries. We have analyzed the neuroprotective efficacy of the transient overexpression of antioxidant enzyme Cu/Zn Superoxide dismutase (SOD) after excitotoxic injury to the immature rat brain by using a recently constructed modular protein vector for non-viral gene delivery termed NLSCt. For this purpose, animals were injected with the NLSCt vector carrying the Cu/Zn SOD or the control GFP transgenes 2 hours after intracortical N-methyl-D-aspartate (NMDA) administration, and daily functional evaluation was performed. Moreover, 3 days after, lesion volume, neuronal degeneration and nitrotyrosine immunoreactivity were evaluated. RESULTS Overexpression of Cu/Zn SOD transgene after NMDA administration showed improved functional outcome and a reduced lesion volume at 3 days post lesion. In secondary degenerative areas, increased neuronal survival as well as decreased numbers of degenerating neurons and nitrotyrosine immunoreactivity was seen. Interestingly, injection of the NLSCt vector carrying the control GFP transgene also displayed a significant neuroprotective effect but less pronounced. CONCLUSION When the appropriate levels of Cu/Zn SOD are expressed transiently after injury using the non-viral modular protein vector NLSCt a neuroprotective effect is seen. Thus recombinant modular protein vectors may be suitable for in vivo gene therapy, and Cu/Zn SOD should be considered as an interesting therapeutic transgene.
Collapse
Affiliation(s)
- Hugo Peluffo
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| | - Laia Acarin
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| | - Anna Arís
- Institut de Biotecnologia i de Biomedicina and Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193, Spain
| | - Pau González
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| | - Antoni Villaverde
- Institut de Biotecnologia i de Biomedicina and Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193, Spain
| | - Bernardo Castellano
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| | - Berta González
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| |
Collapse
|
27
|
Makarov P, Kropf S, Wiswedel I, Augustin W, Schild L. Consumption of redox energy by glutathione metabolism contributes to hypoxia/ reoxygenation-induced injury in astrocytes. Mol Cell Biochem 2006; 286:95-101. [PMID: 16583144 DOI: 10.1007/s11010-005-9098-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2005] [Accepted: 11/30/2005] [Indexed: 11/30/2022]
Abstract
The role of glutathione during ischemia/reperfusion is still a controversial issue. Glutathione should exert beneficial effects in the situation of ischemia/reperfusion due to its antioxidative potency. However, increasing survival time after transient ischemia and hypoxia has been reported for glutathione depleted cells. This work was aimed to analyse whether glutathione metabolism essentially contributes to redox energy failure and subsequent cell damage during ischemia/reperfusion. For this purpose, primary astrocyte rich cell cultures were subjected to 1 h hypoxia followed by up to 4 h reoxygenation in combination with substrate deprivation and glutathione depletion. The ability of the cells to reduce MTT was used to quantify the redox power of the cells. Inhibition of glutathione synthesis by L-buthionine-(S,R)-sulfoximine (BSO) caused depletion of cellular glutathione within 24 h and increase in MTT reduction by about 10% under normoxic conditions. Reoxygenation following 1 h of hypoxia was associated with decrease in MTT reduction which was enhanced by substrate deprivation. Glutathione depletion reduced hypoxia-induced decrease in MTT reduction. Three hours of substrate deprivation prior hypoxia resulted in lower levels of MTT reduction during reoxygenaton. Our data suggest that in situations of oxidative stress such as ischemia/reperfusion, glutathione metabolism may causes decrease of the cellular redox energy below a threshold level required for basic cellular functions finally resulting in cell injury.
Collapse
Affiliation(s)
- Petr Makarov
- Institute of Clinical Chemistry and Pathological Biochemistry, Department of Pathological Biochemistry, Medical Faculty, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | | | | | | | | |
Collapse
|
28
|
Takemura S, Kayama T, Kuge A, Ali H, Kokubo Y, Sato S, Kamii H, Goto K, Yoshimoto T. Correlation between copper/zinc superoxide dismutase and the proliferation of neural stem cells in aging and following focal cerebral ischemia. J Neurosurg 2006; 104:129-36. [PMID: 16509156 DOI: 10.3171/jns.2006.104.1.129] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Object
Neural stem cells (NSCs) have been demonstrated in the subventricular zone (SVZ) of the lateral ventricle and the subgranular zone of the hippocampal dentate gyrus (DG). Although aging rats manifest a decrease in NSCs, rats exposed to stress (for example, ischemia, epilepsy, radiation, and trauma) show an increase in these cells. In transgenic mice, the overexpression of human copper/zinc superoxide dismutase (SOD1), an endogenous antioxidant, has been reported to be a protective enzyme against transient focal cerebral ischemia. The authors investigated the correlation between SOD1 and the proliferation of NSCs in aging as chronic oxidative stress (Experiment 1) and acute oxidative stress induced by transient focal cerebral ischemia (Experiment 2) in mice.
Methods
Bromodeoxyuridine (BrdU) was used in the evaluation of NSCs. In Experiment 1, NSCs in the SVZ significantly increased in 16-month-old transgenic mice compared with wild-type mice (p = 0.0001). In Experiment 2, mice were subjected to 30-minute occlusions of the middle cerebral artery. The increase in NSCs in the DG in transgenic mice was significantly greater than that in wild-type mice (p < 0.05).
Conclusions
Results in this study suggest that chronic and acute oxidative stress may inhibit the proliferation of NSCs and that SOD1 may play a key role in NSC proliferation.
Collapse
Affiliation(s)
- Sunao Takemura
- Department of Neurosurgery, Yamagata University School of Medicine, Yamagata, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Lu KT, Wang YW, Yang JT, Yang YL, Chen HI. Effect of Interleukin-1 on Traumatic Brain Injury–Induced Damage to Hippocampal Neurons. J Neurotrauma 2005; 22:885-95. [PMID: 16083355 DOI: 10.1089/neu.2005.22.885] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Interleukin-1 (IL-1) has many roles in the brain in addition to mediating inflammatory processes in the glia, and has also been implicated in neurodegenerative disease. Traumatic brain injury (TBI) is one of the most prevalent causes of morbidity and mortality in young persons. We conducted a study to assess the effect of IL-1 on the TBI-induced death of hippocampal neurons. After TBI was induced in adult male Sprague-Dawley rats under anesthesia, we evaluated neuronal damage score through microscopic examination and Pulsinelli's grading system. Reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting were used to measure the levels of IL-1alpha and IL- 1beta in brain tissue at different points after the induction of TBI. Over a 4-day period, the specific sites of release of IL-1alpha and IL-1beta in the brain were elucidated by immunocytochemistry with double- labeling. TBI to the hippocampus was followed by disruption of the blood-brain barrier and severe neuronal loss. Levels of IL-1alpha RNA and protein were significantly elevated at 3 h after TBI, peaked at 12 h, and remained elevated for 168 h. IL-1beta RNA and protein expression were also elevated at 3 h after TBI, but remained so only for 48 h. Our findings indicate that the observed TBI-induced increases in IL-1alpha and IL-1beta occur largely through release of these cytokines from neurons and astrocytes, respectively. Intraventricular administration of antibodies to IL-1alpha and IL-1beta before TBI significantly attenuated the TBI-induced loss of hippocampal neurons. These results show that IL-1alpha and IL-1beta play important roles in the TBI-induced loss of hippocampal neurons.
Collapse
Affiliation(s)
- Kwok-Tung Lu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
30
|
Peluffo H, Acarin L, Faiz M, Castellano B, Gonzalez B. Cu/Zn superoxide dismutase expression in the postnatal rat brain following an excitotoxic injury. J Neuroinflammation 2005; 2:12. [PMID: 15929797 PMCID: PMC1164430 DOI: 10.1186/1742-2094-2-12] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2005] [Accepted: 06/01/2005] [Indexed: 01/11/2023] Open
Abstract
Background In the nervous system, as in other organs, Cu/Zn superoxide dismutase (Cu/Zn SOD) is a key antioxidant enzyme involved in superoxide detoxification in normal cellular metabolism and after cell injury. Although it has been suggested that immature brain has a different susceptibility to oxidative damage than adult brain, the distribution and cell-specific expression of this enzyme in immature brain and after postnatal brain damage has not been documented. Methods In this study, we used immunohistochemistry and western blot to analyze the expression of Cu/Zn SOD in intact immature rat brain and in immature rat brain after an NMDA-induced excitotoxic cortical injury performed at postnatal day 9. Double immunofluorescence labelling was used to identify Cu/Zn SOD-expressing cell populations. Results In intact immature brain, Cu/Zn SOD enzyme was widely expressed at high levels in neurons mainly located in cortical layers II, III and V, in the sub-plate, in the pyriform cortex, in the hippocampus, and in the hypothalamus. Glial fibrillary acidic protein-positive cells only showed Cu/Zn SOD expression in the glia limitans and in scattered cells of the ventricle walls. No expression was detected in interfascicular oligodendroglia, microglia or endothelial cells. Following excitotoxic damage, neuronal Cu/Zn SOD was rapidly downregulated (over 2–4 hours) at the injection site before neurodegeneration signals and TUNEL staining were observed. Later, from 1 day post-lesion onward, an upregulation of Cu/Zn SOD was found due to increased expression in astroglia. A further increase was observed at 3, 5 and 7 days that corresponded to extensive induction of Cu/Zn SOD in highly reactive astrocytes and in the astroglial scar. Conclusion We show here that, in the intact immature brain, the expression of Cu/Zn SOD was mainly found in neurons. When damage occurs, a strong and very rapid downregulation of this enzyme precedes neuronal degeneration, and is followed by an upregulation of Cu/Zn SOD in astroglial cells.
Collapse
Affiliation(s)
- Hugo Peluffo
- Unit of Histology, Department Of Cell Biology, Physiology, and Immunology; Autonomous University of Barcelona, 08193, Spain
- Institute of Neuroscience, Autonomous University of Barcelona, 08193, Spain
| | - Laia Acarin
- Unit of Histology, Department Of Cell Biology, Physiology, and Immunology; Autonomous University of Barcelona, 08193, Spain
- Institute of Neuroscience, Autonomous University of Barcelona, 08193, Spain
| | - Maryam Faiz
- Unit of Histology, Department Of Cell Biology, Physiology, and Immunology; Autonomous University of Barcelona, 08193, Spain
- Institute of Neuroscience, Autonomous University of Barcelona, 08193, Spain
| | - Bernardo Castellano
- Unit of Histology, Department Of Cell Biology, Physiology, and Immunology; Autonomous University of Barcelona, 08193, Spain
- Institute of Neuroscience, Autonomous University of Barcelona, 08193, Spain
| | - Berta Gonzalez
- Unit of Histology, Department Of Cell Biology, Physiology, and Immunology; Autonomous University of Barcelona, 08193, Spain
- Institute of Neuroscience, Autonomous University of Barcelona, 08193, Spain
| |
Collapse
|
31
|
Acarin L, Peluffo H, Barbeito L, Castellano B, González B. Astroglial nitration after postnatal excitotoxic damage: correlation with nitric oxide sources, cytoskeletal, apoptotic and antioxidant proteins. J Neurotrauma 2005; 22:189-200. [PMID: 15665612 DOI: 10.1089/neu.2005.22.189] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Oxygen free radicals and nitric oxide (NO) participate in the pathogenesis of acute central nervous system (CNS) injury by forming peroxynitrite, which promotes oxidative damage and tyrosine nitration. Neuronal nitration is associated with cell death, but little is known of the characteristics and cell fate of nitrated astrocytes. In this study, we have used a postnatal excitotoxic lesion model (intracortical NMDA injection) and our aims were (i) to evaluate the temporal and spatial pattern of astroglial nitration in correlation with the neuropathological process and the sources of NO; and (ii) to establish, if any, the correlation among astrocyte nitration and other events such as expression of cytoskeletal proteins, antioxidant enzymes, and cell death markers to cope with nitration and/or undergo cell death. Our results show that after postnatal excitotoxic damage two distinct waves of nitration were observed in relation to astrocytes. At 24 h post-lesion, early-nitrated astrocytes were found within the neurodegenerating area, coinciding with the time of maximal cell death. These early-nitrated astrocytes are highly ramified protoplasmic cells, showing diffuse glial fibrillary acidic protein (GFAP) content and expressing inducible NOS. At later time-points, when astrogliosis is morphologically evident, nitrated hypertrophied reactive astrocytes are observed in the penumbra and the neurodegenerated area, displaying increased expression of GFAP and vimentin cytoskeletal proteins and of metallothionein I-II and Cu/Zn superoxide dismutase antioxidant proteins. Moreover, despite revealing activated caspase-3, they do not show TUNEL labeling. In summary, we show that nitrated astrocytes in vivo constitute a subpopulation of highly reactive astrocytes which display high resistance towards oxidative stress induced cell death.
Collapse
Affiliation(s)
- Laia Acarin
- Unit of Histology, School of Medicine, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autonoma Barcelona, Bellaterra 08193, Spain.
| | | | | | | | | |
Collapse
|
32
|
Chen Z, Duan RS, Lepécheur M, Paly E, London J, Zhu J. SOD-1 inhibits FAS expression in cortex of APP transgenic mice. Apoptosis 2005; 10:499-502. [PMID: 15909112 DOI: 10.1007/s10495-005-1879-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Peptides derived from proteolytic processing of the amyloid precursor protein (APP) are important for the pathogenesis of Alzheimer's disease (AD). In the present study, we found that transgenic mice overexpressing wild-type human APP gene (hAPP/+) displayed a much higher expression of FAS, one of the death receptor subfamily. This FAS overexpression was significantly reduced in the cortex of mice overexpressing both wild-type hAPP gene and wild-type human superoxide dismutase-1 gene (hSOD-1). Moreover hSOD-1 transgenic expression was associated with an increase of Glial fibrillary acidic protein (GFAP) production. This study indicates that SOD-1 overexpression can inhibit FAS expression, which may be beneficial in AD.
Collapse
Affiliation(s)
- Z Chen
- Division of Experimental Geriatrics, Department of Neurotec, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
33
|
Wang J, Ma JH, Giffard RG. Overexpression of copper/zinc superoxide dismutase decreases ischemia-like astrocyte injury. Free Radic Biol Med 2005; 38:1112-8. [PMID: 15780769 DOI: 10.1016/j.freeradbiomed.2005.01.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2004] [Revised: 01/06/2005] [Accepted: 01/11/2005] [Indexed: 11/23/2022]
Abstract
Overexpression of copper/zinc superoxide dismutase (SOD1) in transgenic mice protects from transient focal cerebral ischemia in adult animals, but increases oxidative injury in perinatal mice. The effect of SOD1 overexpression on astrocytes subjected to ischemia-like insults has not yet been determined. Overexpression of human SOD1 in astrocytes resulted in a 3-fold increase in SOD1 activity without coupled up-regulation of catalase or glutathione peroxidase activities. Cells subjected to oxygen-glucose deprivation (OGD) or glucose deprivation to mimic ischemic injury were protected by SOD1 overexpression. OGD injury was reduced 47.6+/-9.3%, assessed by release of lactate dehydrogenase. OGD also caused a significant increase in catalase activity which was moderated by SOD1 overexpression. The level of glutathione in astrocytes overexpressing SOD1 was maintained at higher levels following 5 h OGD compared to control cultures under the same conditions. Reduction of glutathione prior to OGD significantly increased cell death of SOD1-overexpressing astrocytes as well as controls, but SOD1 still provided significant protection, suggesting that both GSH-dependent scavenging and GSH-independent scavenging are relevant to SOD1 protection in astrocytes.
Collapse
Affiliation(s)
- Jian Wang
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | |
Collapse
|
34
|
Sebastià J, Cristòfol R, Pertusa M, Vílchez D, Torán N, Barambio S, Rodríguez-Farré E, Sanfeliu C. Down's syndrome astrocytes have greater antioxidant capacity than euploid astrocytes. Eur J Neurosci 2004; 20:2355-66. [PMID: 15525277 DOI: 10.1111/j.1460-9568.2004.03686.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Down's syndrome (trisomy 21) brain tissue is considered to be susceptible to oxidative injury, mainly because its increased Cu/Zn-superoxide dismutase (SOD1) activity is not followed by an adaptive rise in hydrogen peroxide metabolizing enzymes. In vitro, trisomic neurons suffer oxidative stress and degenerate. We studied the response of trisomy 21 neuron and astrocyte cultures to hydrogen peroxide injury and found that they were, respectively, more and less vulnerable than their euploid counterparts. Differences were detected 24 h after exposures in the region of 50 microm and 500 microm hydrogen peroxide for neuron and astrocyte cultures, respectively. Cytotoxicity results were paralleled by a decrease in cellular glutathione. In addition, trisomic astrocytes showed a lower basal content of superoxide ion and a higher clearance of hydrogen peroxide from the culture medium. In the presence of hydrogen peroxide, trisomic astrocytes maintained their concentration of intracellular superoxide and hydroperoxides at a lower level than euploid astrocytes. Consistent with these results, trisomic astrocytes in neuron coculture were more neuroprotective than euploid astrocytes against hydrogen peroxide injury. We suggest that SOD1 overexpression has beneficial effects on astrocytes, as it does in other systems with similarly high disposal of hydroperoxides. In addition to a higher enzymatic activity of SOD1, cultures of trisomic astrocytes showed slightly higher glutathione reductase activity than euploid cultures. Thus, trisomy 21 astrocytes showed a greater antioxidant capacity against hydrogen peroxide than euploid astrocytes, and they partially counteracted the oxidative vulnerability of trisomic neurons in culture.
Collapse
Affiliation(s)
- Jordi Sebastià
- Departament de Farmacologia i Toxicologia. Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC-IDIBAPS, Rosselló 161, E-08036 Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Tortarolo M, Crossthwaite AJ, Conforti L, Spencer JP, Williams RJ, Bendotti C, Rattray M. Expression of SOD1 G93A or wild-type SOD1 in primary cultures of astrocytes down-regulates the glutamate transporter GLT-1: lack of involvement of oxidative stress. J Neurochem 2004; 88:481-93. [PMID: 14690536 DOI: 10.1046/j.1471-4159.2003.02208.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Glutamate excitotoxicity is implicated in the aetiology of amyotrophic lateral sclerosis (ALS) with impairment of glutamate transport into astrocytes a possible cause of glutamate-induced injury to motor neurons. It is possible that mutations of Cu/Zn superoxide dismutase (SOD1), responsible for about 20% of familial ALS, down-regulates glutamate transporters via oxidative stress. We transfected primary mouse astrocytes to investigate the effect of the FALS-linked mutant hSOD1(G93A) and wild-type SOD1 (hSOD1wt) on the glutamate uptake system. Using western blotting, immunocytochemistry and RT-PCR it was shown that expression of either hSOD1(G93A) or hSOD1wt in astrocytes produced down-regulation of the levels of a glutamate transporter GLT-1, without alterations in its mRNA level. hSOD1(G93A) or hSOD1wt expression caused a decrease of the monomeric form of GLT-1 without increasing oxidative multimers of GLT-1. The effects were selective to GLT-1, since another glutamate transporter GLAST protein and mRNA levels were not altered. Reflecting the decrease in GLT-1 protein, [3H]d-aspartate uptake was reduced in cultures expressing hSOD1(G93A) or hSOD1wt. The hSOD1-induced decline in GLT-1 protein and [3H]d-aspartate uptake was not blocked by the antioxidant Trolox nor potentiated by antioxidant depletion using catalase and glutathione peroxidase inhibitors. Measurement of 2',7'-dichlorofluorescein (DCF)-induced fluorescence revealed that expression of hSOD1(G93A) or hSOD1wt in astrocytes does not lead to detectable increase of intracellular reactive oxygen species. This study suggests that levels of GLT-1 protein in astrocytes are reduced rapidly by overexpression of hSOD1, and is due to a property shared between the wild-type and G93A mutant form, but does not involve the production of intracellular oxidative stress.
Collapse
Affiliation(s)
- Massimo Tortarolo
- Biochemical Neuropharmacology Group, GKT Centre for Neuroscience Research, King's College London, Guy's Hospital Campus, London, UK
| | | | | | | | | | | | | |
Collapse
|
36
|
Haque ME, Asanuma M, Higashi Y, Miyazaki I, Tanaka KI, Ogawa N. Overexpression of Cu-Zn superoxide dismutase protects neuroblastoma cells against dopamine cytotoxicity accompanied by increase in their glutathione level. Neurosci Res 2003; 47:31-7. [PMID: 12941444 DOI: 10.1016/s0168-0102(03)00166-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dopamine (DA) was shown to exert toxic effects on cultured neurons through autoxidation or oxidative deamination, followed by formation of highly reactive quinone compounds and superoxide radicals. In the present study, therefore, any involvement of Cu-Zn superoxide dismutase (SOD) in DA toxicity was evaluated by transfection of Cu-Zn SOD cDNA. The transient transfection of Cu-Zn SOD cDNA inhibited the DA-induced decrease of dopaminergic neuroblastoma cells. Moreover, Cu-Zn SOD cDNA-transfection significantly increased the glutathione (GSH) level when the cells were exposed to DA. However, such Cu-Zn SOD-overexpression failed to show any protective effects against hydrogen peroxide. The Cu-Zn SOD-overexpressing cells also showed significantly higher levels of GSH upon DA exposure than did the empty vector-transfected cells. The increase in the GSH level in response to hydrogen peroxide remained almost identical in empty vector-transfected or Cu-Zn SOD-overexpressed cells. The level of GSH in DA-treated Cu-Zn SOD-overexpressing cells was 2.5-fold higher than that increased by hydrogen peroxide exposure. The catechol structure of DA molecule is probably involved in the mechanism of increasing GSH level. Furthermore, the Cu-Zn SOD-overexpressing cells inhibited the activation of caspase-3 upon DA exposure. Therefore, Cu-Zn SOD overexpression may temporarily inhibit or delay DA autoxidation and consequently increase the GSH level, which then prevents the activation of apoptotic pathway and subsequent cell death.
Collapse
Affiliation(s)
- M Emdadul Haque
- Department of Brain Science, Okayama University Graduate School of Medicine and Dentistry, 2-5-1 Shikatacho, Okayama 700-8558, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
Astrocytes are the most numerous cell type in the central nervous system. They provide structural, trophic, and metabolic support to neurons and modulate synaptic activity. Accordingly, impairment in these astrocyte functions during brain ischemia and other insults can critically influence neuron survival. Astrocyte functions that are known to influence neuronal survival include glutamate uptake, glutamate release, free radical scavenging, water transport, and the production of cytokines and nitric oxide. Long-term recovery after brain injury, through neurite outgrowth, synaptic plasticity, or neuron regeneration, is influenced by astrocyte surface molecule expression and trophic factor release. In addition, the death or survival of astrocytes themselves may affect the ultimate clinical outcome and rehabilitation through effects on neurogenesis and synaptic reorganization.
Collapse
Affiliation(s)
- Yongmei Chen
- Department of Neurology, University of California at San Francisco, California 94121, USA
| | | |
Collapse
|
38
|
Leker RR, Shohami E. Cerebral ischemia and trauma-different etiologies yet similar mechanisms: neuroprotective opportunities. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2002; 39:55-73. [PMID: 12086708 DOI: 10.1016/s0165-0173(02)00157-1] [Citation(s) in RCA: 285] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Cerebral ischemia leads to brain damage caused by pathogenetic mechanisms that are also activated by neurotrauma. These mechanisms include among others excitotoxicity, over production of free radicals, inflammation and apoptosis. Furthermore, cerebral ischemia and trauma both trigger similar auto-protective mechanisms including the production of heat shock proteins, anti-inflammatory cytokines and endogenous antioxidants. Neuroprotective therapy aims at minimizing the activation of toxic pathways and at enhancing the activity of endogenous neuroprotective mechanisms. The similarities in the damage-producing and endogenous auto-protective mechanisms may imply that neuroprotective compounds found to be active against one of these conditions may indeed be also protective in the other. This review summarizes the pathogenetic events of ischemic and traumatic brain injury and reviews the neuroprotective strategies employed thus far in each of these conditions with a special emphasize on their clinical relevance and on future directions in the field of neuronal protection.
Collapse
Affiliation(s)
- Ronen R Leker
- Department of Neurology and the Agnes Ginges Center for Human Neurogenetics, Hebrew University-Hadassah Medical School and Hadassah University Hospital, Jerusalem, Israel.
| | | |
Collapse
|
39
|
Matés JM, Pérez-Gómez C, Núñez de Castro I, Asenjo M, Márquez J. Glutamine and its relationship with intracellular redox status, oxidative stress and cell proliferation/death. Int J Biochem Cell Biol 2002; 34:439-58. [PMID: 11906817 DOI: 10.1016/s1357-2725(01)00143-1] [Citation(s) in RCA: 238] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Glutamine is a multifaceted amino acid used for hepatic urea synthesis, renal ammoniagenesis, gluconeogenesis in both liver and kidney, and as a major respiratory fuel for many cells. Decreased glutamine concentrations are found during catabolic stress and are related to susceptibility to infections. Besides, glutamine is not only an important energy source in mitochondria, but is also a precursor of the brain neurotransmitter glutamate, which is likewise used for biosynthesis of the cellular antioxidant glutathione. Reactive oxygen species, such as superoxide anions and hydrogen peroxide, function as intracellular second messengers activating, among others, apoptosis, whereas glutamine is an apoptosis suppressor. In fact, it could contribute to block apoptosis induced by exogenous agents or by intracellular stimuli. In conclusion, this article shows evidences for the important role of glutamine in the regulation of the cellular redox balance, including brain oxidative metabolism, apoptosis and tumour cell proliferation.
Collapse
Affiliation(s)
- José M Matés
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Campus de Teatinos, s/n 29071 Málaga, Spain.
| | | | | | | | | |
Collapse
|