1
|
Nunes LGA, Ma C, Hoffmann FW, Shay AE, Pitts MW, Hoffmann PR. Selenoprotein I is indispensable for ether lipid homeostasis and proper myelination. J Biol Chem 2024; 300:107259. [PMID: 38582453 PMCID: PMC11061234 DOI: 10.1016/j.jbc.2024.107259] [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: 01/19/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/08/2024] Open
Abstract
Selenoprotein I (SELENOI) catalyzes the final reaction of the CDP-ethanolamine branch of the Kennedy pathway, generating the phospholipids phosphatidylethanolamine (PE) and plasmenyl-PE. Plasmenyl-PE is a key component of myelin and is characterized by a vinyl ether bond that preferentially reacts with oxidants, thus serves as a sacrificial antioxidant. In humans, multiple loss-of-function mutations in genes affecting plasmenyl-PE metabolism have been implicated in hereditary spastic paraplegia, including SELENOI. Herein, we developed a mouse model of nervous system-restricted SELENOI deficiency that circumvents embryonic lethality caused by constitutive deletion and recapitulates phenotypic features of hereditary spastic paraplegia. Resulting mice exhibited pronounced alterations in brain lipid composition, which coincided with motor deficits and neuropathology including hypomyelination, elevated reactive gliosis, and microcephaly. Further studies revealed increased lipid peroxidation in oligodendrocyte lineage cells and disrupted oligodendrocyte maturation both in vivo and in vitro. Altogether, these findings detail a critical role for SELENOI-derived plasmenyl-PE in myelination that is of paramount importance for neurodevelopment.
Collapse
Affiliation(s)
- Lance G A Nunes
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Chi Ma
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - FuKun W Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Ashley E Shay
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA; Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Matthew W Pitts
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA.
| | - Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| |
Collapse
|
2
|
Li Q, Liu S, Zheng T, Li M, Qi B, Zhou L, Liu B, Ma D, Zhao C, Chen Z. Grafted human-induced pluripotent stem cells-derived oligodendrocyte progenitor cells combined with human umbilical vein endothelial cells contribute to functional recovery following spinal cord injury. Stem Cell Res Ther 2024; 15:35. [PMID: 38321505 PMCID: PMC10848469 DOI: 10.1186/s13287-024-03651-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 01/29/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is a devastating disease that causes extensive damage to oligodendrocytes and neurons leading to demyelination and axonal degeneration. In this study, we co-transplanted cell grafts containing oligodendrocyte progenitor cells (OPCs) derived from human-induced pluripotent stem cells (iPSCs) combined with human umbilical vein endothelial cells (HUVECs), which were reported to promote OPCs survival and migration, into rat contusion models to promote functional recovery after SCI. METHODS OPCs were derived from iPSCs and identified by immunofluorescence at different time points. Functional assays in vitro were performed to evaluate the effect of HUVECs on the proliferation, migration, and survival of OPCs by co-culture and migration assay, as well as on the neuronal axonal growth. A combination of OPCs and HUVECs was transplanted into the rat contusive model. Upon 8 weeks, immunofluorescence staining was performed to test the safety of transplanted cells and to observe the neuronal repairment, myelination, and neural circuit reconstruction at the injured area; also, the functional recovery was assessed by Basso, Beattie, and Bresnahan open-field scale, Ladder climb, SEP, and MEP. Furthermore, the effect of HUVECs on grafts was also determined in vivo. RESULTS Data showed that HUVECs promote the proliferation, migration, and survival of OPCs both in vitro and in vivo. Furthermore, 8 weeks upon engraftment, the rats with OPCs and HUVECs co-transplantation noticeably facilitated remyelination, enhanced functional connection between the grafts and the host and promoted functional recovery. In addition, compared with the OPCs-alone transplantation, the co-transplantation generated more sensory neurons at the lesion border and significantly improved the sensory functional recovery. CONCLUSIONS Our study demonstrates that transplantation of OPCs combined with HUVECs significantly enhances both motor and sensory functional recovery after SCI. No significance was observed between OPCs combined with HUVECs group and OPCs-alone group in motor function recovery, while the sensory function recovery was significantly promoted in OPCs combined with HUVECs groups compared with the other two groups. These findings provide novel insights into the field of SCI research.
Collapse
Affiliation(s)
- Qian Li
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Sumei Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Tianqi Zheng
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Mo Li
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Boling Qi
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Liping Zhou
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Bochao Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Dan Ma
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, B4 7ET, UK
| | - Chao Zhao
- Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China.
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China.
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China.
| |
Collapse
|
3
|
Pernin F, Kuhlmann T, Kennedy TE, Antel JP. Oligodendrocytes in multiple sclerosis. MECHANISMS OF DISEASE PATHOGENESIS IN MULTIPLE SCLEROSIS 2024:261-287. [DOI: 10.1016/b978-0-12-823848-6.00009-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
4
|
Baag S, Mahapatra S, Mandal S. An Integrated and Multibiomarker approach to delineate oxidative stress status of Bellamya bengalensis under the interactions of elevated temperature and chlorpyrifos contamination. CHEMOSPHERE 2021; 264:128512. [PMID: 33049511 DOI: 10.1016/j.chemosphere.2020.128512] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/15/2020] [Accepted: 09/29/2020] [Indexed: 05/12/2023]
Abstract
Synergistic effects of warming on bioconcentration and receptiveness of pollutants are still poorly unravelled in conjunction with cellular and molecular responses. The present study addressed the impact of an environmental relevant dose of chlorpyrifos (organophosphate pesticide), under control (25 °C) and elevated levels of temperature (30 °C, 35 °C) in Bellamya bengalensis, a freshwater gastropod for 60 days across various endpoints. Multiple levels of biomarkers were measured: growth conditions (organ to flesh weight ratio, condition index), oxidative stress status (SOD, CAT, GST, LPO) and DNA damage (Comet assay-3rd, 30th and 60th days only) after acute (24, 48 and 72 h) and long-term exposures (10th, 20th, 30th, 40th, 50th and 60th days). An integrated biomarker response (IBR) strategy was adapted to amalgamate results generated from various biomarkers to assess organism's vulnerability to pesticide pollution and how it may shift with warming climate. Significant changes were observed in growth conditions under longer exposure periods. Acute as well as long-term exposures enhanced the antioxidant and detoxification enzyme activity. DNA damage was extensive under longer exposure to stress howbeit was also significantly escalated under acute severe warming. Antioxidant and detoxification mechanisms fell short in counteracting cellular level damage. The IBR results indicated long-term acclimation of B. bengalensis to elevated temperatures and pesticide contamination lead to an improved tolerance level howbeit, acute stress was more detrimental. This study provided evidence for the efficiency of employing an integrated biomarker approach for B. bengalensis in future bio-monitoring studies.
Collapse
Affiliation(s)
- Sritama Baag
- Marine Ecology Laboratory, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata, 700073, India
| | - Sayantan Mahapatra
- Marine Ecology Laboratory, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata, 700073, India
| | - Sumit Mandal
- Marine Ecology Laboratory, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata, 700073, India.
| |
Collapse
|
5
|
Zheng W, Li Q, Zhao C, Da Y, Zhang HL, Chen Z. Differentiation of Glial Cells From hiPSCs: Potential Applications in Neurological Diseases and Cell Replacement Therapy. Front Cell Neurosci 2018; 12:239. [PMID: 30140204 PMCID: PMC6094089 DOI: 10.3389/fncel.2018.00239] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/17/2018] [Indexed: 12/20/2022] Open
Abstract
Glial cells are the most abundant cell type in the central nervous system (CNS) and play essential roles in maintaining brain homeostasis, forming myelin, and providing support and protection for neurons, etc. Over the past decade, significant progress has been made in the reprogramming field. Given the limited accessibility of human glial cells, in vitro differentiation of human induced pluripotent stem cells (hiPSCs) into glia may provide not only a valuable research tool for a better understanding of the functions of glia in the CNS but also a potential cellular source for clinical therapeutic purposes. In this review, we will summarize up-to-date novel strategies for the committed differentiation into the three major glial cell types, i.e., astrocyte, oligodendrocyte, and microglia, from hiPSCs, focusing on the non-neuronal cell effects on the pathology of some representative neurological diseases. Furthermore, the application of hiPSC-derived glial cells in neurological disease modeling will be discussed, so as to gain further insights into the development of new therapeutic targets for treatment of neurological disorders.
Collapse
Affiliation(s)
- Wei Zheng
- Cell Therapy Center, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, China
| | - Qian Li
- Cell Therapy Center, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, China
| | - Chao Zhao
- Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Yuwei Da
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hong-Liang Zhang
- Department of Life Sciences, National Natural Science Foundation of China, Beijing, China
| | - Zhiguo Chen
- Cell Therapy Center, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
6
|
Mazaheri N, Peymani M, Galehdari H, Ghaedi K, Ghoochani A, Kiani-Esfahani A, Nasr-Esfahani MH. Ameliorating Effect of Osteopontin on H 2O 2-Induced Apoptosis of Human Oligodendrocyte Progenitor Cells. Cell Mol Neurobiol 2018; 38:891-899. [PMID: 29110207 PMCID: PMC11481903 DOI: 10.1007/s10571-017-0563-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/30/2017] [Indexed: 12/22/2022]
Abstract
Recently our group used oligodendrocyte progenitor cells (OPCs) as appropriate model cells to pinpoint the mechanism of the progress of neurodegenerative disorders. In the present study, we focused on the therapeutic role of osteopontin (OPN), a secreted glycosylated phosphoprotein, involved in a number of physiological events including bone formation and remodeling, immune responses, and tumor progression. Protective role of OPN, as a negative regulator of tumorigenesis, has already been clarified. Human embryonic stem cell-derived OPCs were pretreated with OPN before induction of apoptosis by H2O2. Data indicated that OPN prohibited cell death and enhanced OPC viability. This effect is achieved through reduction of apoptosis and induction of anti-apoptosis markers. In addition OPN induces expression of several integrin subunits, responsible for OPN interaction. Notably, our findings showed that expression of αV β1/β3/β5 and β8 integrins increased in response to OPN, while treatment with H2O2 down-regulated αV β1/β5 and β8 integrins expression significantly. In conclusion, OPN may act via αV integrin signaling and trigger suppression of P53-dependent apoptotic cascades. Therefore OPN therapy may be considered as a feasible process to prevent progress of neurodegenerative diseases in human.
Collapse
Affiliation(s)
- Neda Mazaheri
- Department of Genetics, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, 816513-1378, Iran
| | - Hamid Galehdari
- Department of Genetics, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Kamran Ghaedi
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran.
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, 816513-1378, Iran.
| | - Ali Ghoochani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, 816513-1378, Iran
| | - Abbas Kiani-Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, 816513-1378, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran.
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, 816513-1378, Iran.
| |
Collapse
|
7
|
Ibáñez-Contreras A, Hernández-Arciga U, Poblano A, Arteaga-Silva M, Hernández-Godínez B, Mendoza-Cuevas GI, Toledo-Pérez R, Alarcón-Aguilar A, González-Puertos VY, Konigsberg M. Electrical activity of sensory pathways in female and male geriatric Rhesus monkeys (Macaca mulatta), and its relation to oxidative stress. Exp Gerontol 2017; 101:80-94. [PMID: 29146475 DOI: 10.1016/j.exger.2017.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 10/21/2017] [Accepted: 11/07/2017] [Indexed: 10/18/2022]
Abstract
Synapses loss during aging has been related to decreased neuronal excitability and reduced electrophysiological activity in the nervous system, as well as to increased brain damage. Those physiological and biochemical alterations have been related to the oxidative stress increase associated with old age. The main substrate of lipid peroxidation (LPX) in the central and peripheral nervous systems are the myelin sheaths, and their damage generates a delayed nerve conduction velocity. However, studies in which the neural conduction velocity is related to changes in the redox state are still lacking. Therefore, our aim was to correlate the sensory neural pathways delay in healthy geriatric Rhesus monkeys (Macaca mulatta) with the oxidative stress associated with physiological aging. Twenty-four monkeys were divided into four groups according to age and gender. Auditory, visual, and somatosensory evoked potentials were obtained. Superoxide dismutase, catalase, and glutathione peroxidase enzymatic activity, as well as LPX, were determined from blood samples. Our results showed significant differences between the older and younger age groups in all neural generators of the different sensory pathways evaluated, along with an increase in LPX and the antioxidant enzymatic activities. It suggests that, even though the enzymatic activity was found to be higher in older monkeys, probably as a compensatory effect, it was not enough to avoid LPX damage and the declined electric activity associated with age.
Collapse
Affiliation(s)
- A Ibáñez-Contreras
- Posgrado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, México D.F., Mexico; APREXBIO S.A.S. de C.V., Laboratorio de Primatología, Ciudad de México, México D.F., Mexico; Biología Integral para Vertebrados (BIOINVERT®), Unidad de Experimentación Animal, Estado de México, Mexico; Centro de Investigación, Proyecto CAMINA A.C. Unidad de Primates No Humanos, Ciudad de México, México D.F., Mexico; Laboratorio de Bioenergética y envejecimiento celular, Depto. de Ciencias de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, México D.F., Mexico
| | - U Hernández-Arciga
- Laboratorio de Bioenergética y envejecimiento celular, Depto. de Ciencias de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, México D.F., Mexico
| | - A Poblano
- Laboratorio de Neurofisiología Cognoscitiva, Instituto Nacional de Rehabilitación, Ciudad de México, México D.F., Mexico
| | - M Arteaga-Silva
- Depto. Biología de la Reproducción, Universidad Autónoma Metropolitana, Unidad Iztapalapa, México D.F., Mexico
| | - B Hernández-Godínez
- Posgrado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, México D.F., Mexico; APREXBIO S.A.S. de C.V., Laboratorio de Primatología, Ciudad de México, México D.F., Mexico; Biología Integral para Vertebrados (BIOINVERT®), Unidad de Experimentación Animal, Estado de México, Mexico; Centro de Investigación, Proyecto CAMINA A.C. Unidad de Primates No Humanos, Ciudad de México, México D.F., Mexico; Centro Nacional de Investigación en Instrumentación e Imagenología Médica (CI3M), Universidad Autónoma Metropolitana-Unidad Iztapalapa (UAM-I), México D.F., Mexico
| | - G I Mendoza-Cuevas
- APREXBIO S.A.S. de C.V., Laboratorio de Primatología, Ciudad de México, México D.F., Mexico; Biología Integral para Vertebrados (BIOINVERT®), Unidad de Experimentación Animal, Estado de México, Mexico; Centro de Investigación, Proyecto CAMINA A.C. Unidad de Primates No Humanos, Ciudad de México, México D.F., Mexico
| | - R Toledo-Pérez
- Laboratorio de Bioenergética y envejecimiento celular, Depto. de Ciencias de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, México D.F., Mexico
| | - A Alarcón-Aguilar
- Laboratorio de Bioenergética y envejecimiento celular, Depto. de Ciencias de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, México D.F., Mexico
| | - V Y González-Puertos
- Laboratorio de Bioenergética y envejecimiento celular, Depto. de Ciencias de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, México D.F., Mexico
| | - M Konigsberg
- Laboratorio de Bioenergética y envejecimiento celular, Depto. de Ciencias de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, México D.F., Mexico.
| |
Collapse
|
8
|
Quraishe S, Wyttenbach A, Matinyarare N, Perry VH, Fern R, O'Connor V. Selective and compartmentalized myelin expression of HspB5. Neuroscience 2015; 316:130-42. [PMID: 26718604 DOI: 10.1016/j.neuroscience.2015.12.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/17/2015] [Accepted: 12/17/2015] [Indexed: 11/26/2022]
Abstract
In the present study, we reveal myelin-specific expression and targeting of mRNA and biochemical pools of HspB5 in the mouse CNS. Our observations are based on in situ hybridization, electron microscopy and co-localization with 2',3'-Cyclic-Nucleotide 3'-Phosphodiesterase (CNPase), reinforcing this myelin-selective expression. HspB5 mRNA might be targeted to these structures based on its presence in discrete clusters resembling RNA granules and the presence of a putative RNA transport signal. Further, sub-cellular fractionation of myelin membranes reveals a distinct sub-compartment-specific association and detergent solubility of HspB5. This is akin to other abundant myelin proteins and is consistent with HspB5's association with cytoskeletal/membrane assemblies. Oligodendrocytes have a pivotal role in supporting axonal function via generating and segregating the ensheathing myelin. This specialization places extreme structural and metabolic demands on this glial cell type. Our observations place HspB5 in oligodendrocytes which may require selective and specific chaperone capabilities to maintain normal function and neuronal support.
Collapse
Affiliation(s)
- S Quraishe
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, Building 85, University of Southampton, Southampton SO17 1BJ, UK.
| | - A Wyttenbach
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, Building 85, University of Southampton, Southampton SO17 1BJ, UK
| | - N Matinyarare
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, Building 85, University of Southampton, Southampton SO17 1BJ, UK
| | - V H Perry
- Centre for Biological Sciences, Mailpoint 840 (room LD80b), Level D Laboratories and Pathology Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - R Fern
- Peninsula School of Medicine and Dentistry, University of Plymouth, Devon, PL4 8AA, UK
| | - V O'Connor
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, Building 85, University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
9
|
Leomanni A, Schettino T, Calisi A, Gorbi S, Mezzelani M, Regoli F, Lionetto MG. Antioxidant and oxidative stress related responses in the Mediterranean land snail Cantareus apertus exposed to the carbamate pesticide Carbaryl. Comp Biochem Physiol C Toxicol Pharmacol 2015; 168:20-7. [PMID: 25451076 DOI: 10.1016/j.cbpc.2014.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/20/2014] [Accepted: 11/20/2014] [Indexed: 10/24/2022]
Abstract
The aim of the present work was to study the alterations of the antioxidant defenses and the overall susceptibility to oxidative stress of the terrestrial snail Cantareus apertus exposed to the carbamate pesticide Carbaryl at a low environmentally realistic concentration. The animals were exposed to Lactuca sativa soaked for 1h in 1μM Carbaryl. The temporal dynamics of the responses was assessed by measurements at 3, 7 and 14days of exposure. C. apertus exposed to Carbaryl activates a number of enzymatic antioxidant responses, represented by the early induction of catalase, glutathione peroxidase, glutathione reductase, followed by a delayed induction of superoxide dismutase. Concomitantly, a derangement of the total oxyradical scavenging of the tissues was observed, suggesting an overall impairment of the tissue capability to neutralize ROS probably resulting from the overall negative balance between enzymatic antioxidant defense capability and oxidative stress intensity. This negative balance exposed the animals to the risk of oxidative stress damages including genotoxic damage. Compared to acetylcholinesterase inhibition, the antioxidant responses developed to Carbaryl exposure at the low concentration utilized showed a greater percentage variation in exposed organisms. The results pointed out the high sensitivity of the antioxidant and oxidative stress related responses to Carbaryl exposure at an environmental realistic concentration, demonstrating their usefulness in environmental monitoring and risk assessment. The study highlights also the usefulness of the terrestrial snail C. apertus as potential bioindicator species for assessing the risk of pesticide environmental contamination.
Collapse
Affiliation(s)
- A Leomanni
- Dip.to di Scienze e Tecnologie Biologiche e Ambientali (DiSTeBA), Università del Salento, Via prov.le Lecce-Monteroni, 73100 Lecce, Italy
| | - T Schettino
- Dip.to di Scienze e Tecnologie Biologiche e Ambientali (DiSTeBA), Università del Salento, Via prov.le Lecce-Monteroni, 73100 Lecce, Italy
| | - A Calisi
- Dip.to di Scienze e Tecnologie Biologiche e Ambientali (DiSTeBA), Università del Salento, Via prov.le Lecce-Monteroni, 73100 Lecce, Italy
| | - S Gorbi
- Dip.to di Genetica Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Via Ranieri 65, Montedago, 60131 Ancona, Italy
| | - M Mezzelani
- Dip.to di Genetica Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Via Ranieri 65, Montedago, 60131 Ancona, Italy
| | - F Regoli
- Dip.to di Genetica Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Via Ranieri 65, Montedago, 60131 Ancona, Italy
| | - M G Lionetto
- Dip.to di Scienze e Tecnologie Biologiche e Ambientali (DiSTeBA), Università del Salento, Via prov.le Lecce-Monteroni, 73100 Lecce, Italy.
| |
Collapse
|
10
|
Kruska N, Schönfeld P, Pujol A, Reiser G. Astrocytes and mitochondria from adrenoleukodystrophy protein (ABCD1)-deficient mice reveal that the adrenoleukodystrophy-associated very long-chain fatty acids target several cellular energy-dependent functions. Biochim Biophys Acta Mol Basis Dis 2015; 1852:925-36. [PMID: 25583114 DOI: 10.1016/j.bbadis.2015.01.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 12/03/2014] [Accepted: 01/05/2015] [Indexed: 12/21/2022]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a severe neurodegenerative disorder resulting from defective ABCD1 transport protein. ABCD1 mediates peroxisomal uptake of free very-long-chain fatty acids (VLCFA) as well as their CoA-esters. Consequently, VLCFA accumulate in patients' plasma and tissues, which is considered as pathogenic X-ALD triggering factor. Clinical symptoms are mostly manifested in neural tissues and adrenal gland. Here, we investigate astrocytes from wild-type control and a genetic X-ALD mouse model (Abcd1-knockout), exposed to supraphysiological VLCFA (C22:0, C24:0 and C26:0) concentrations. They exhibit multiple impairments of energy metabolism. Furthermore, brain mitochondria from Abcd1(-/-) mice and wild-type control respond similarly to VLCFA with increased ROS generation, impaired oxidative ATP synthesis and diminished Ca(2+) uptake capacity, suggesting that a defective ABCD1 exerts no adaptive pressure on mitochondria. In contrast, astrocytes from Abcd1(-/-) mice respond more sensitively to VLCFA than wild-type control astrocytes. Moreover, long-term application of VLCFA induces high ROS generation, and strong in situ depolarization of mitochondria, and, in Abcd1(-/-) astrocytes, severely diminishes the capability to revert oxidized pyridine nucleotides to NAD(P)H. In addition, observed differences in responses of mitochondria and astrocytes to the hydrocarbon chain length of VLCFA suggest that detrimental VLCFA activities in astrocytes involve defective cellular functions other than mitochondria. In summary, we clearly demonstrate that VLCFA increase the vulnerability of Abcd1(-/-) astrocytes.
Collapse
Affiliation(s)
- Nicol Kruska
- Institut für Neurobiochemie, Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, D-39120 Magdeburg, Germany
| | - Peter Schönfeld
- Institut für Biochemie und Zellbiologie, Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, D-39120 Magdeburg, Germany
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, ICREA and Institut d'Investigació Biomedica de Bellvitge (IDIBELL), Hospitalet de Llobregat 08908 Barcelona, Spain; Center for Biomedical Research in Rare Diseases (CIBERER), ISCIII Madrid, Spain
| | - Georg Reiser
- Institut für Neurobiochemie, Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, D-39120 Magdeburg, Germany.
| |
Collapse
|
11
|
Glutathione-Dependent Detoxification Processes in Astrocytes. Neurochem Res 2014; 40:2570-82. [PMID: 25428182 DOI: 10.1007/s11064-014-1481-1] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 11/10/2014] [Accepted: 11/15/2014] [Indexed: 01/17/2023]
Abstract
Astrocytes have a pivotal role in brain as partners of neurons in homeostatic and metabolic processes. Astrocytes also protect other types of brain cells against the toxicity of reactive oxygen species and are considered as first line of defence against the toxic potential of xenobiotics. A key component in many of the astrocytic detoxification processes is the tripeptide glutathione (GSH) which serves as electron donor in the GSH peroxidase-catalyzed reduction of peroxides. In addition, GSH is substrate in the detoxification of xenobiotics and endogenous compounds by GSH-S-transferases which generate GSH conjugates that are efficiently exported from the cells by multidrug resistance proteins. Moreover, GSH reacts with the reactive endogenous carbonyls methylglyoxal and formaldehyde to intermediates which are substrates of detoxifying enzymes. In this article we will review the current knowledge on the GSH metabolism of astrocytes with a special emphasis on GSH-dependent detoxification processes.
Collapse
|
12
|
Hohnholt MC, Blumrich EM, Dringen R. Multiassay analysis of the toxic potential of hydrogen peroxide on cultured neurons. J Neurosci Res 2014; 93:1127-37. [PMID: 25354694 DOI: 10.1002/jnr.23502] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/19/2014] [Accepted: 09/25/2014] [Indexed: 11/10/2022]
Abstract
To clarify discrepancies in the literature on the adverse effects of hydrogen peroxide on neurons, this study investigated the application of this peroxide to cultured cerebellar granule neurons with six assays frequently used to test for viability. Cultured neurons efficiently cleared exogenous H2O2. Although viability was not affected by exposure to 10 µM hydrogen peroxide, an exposure to the peroxide in higher concentrations rapidly lowered, within 15 min, the cellular 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltertrazolium bromide (MTT) reduction capacity to 53% ± 1% (100 µM) and 31% ± 1% (1,000 µM) and the 3-amino-7-dimethylamino-2-methyl-phenazine hydrochloride (neutral red; NR) uptake to 84% ± 6% (100 µM) and 33% ± 1% (1,000 µM) of control cells. The release of glycolytically generated lactate was stopped within 30 min in neurons treated with 1,000 µM peroxide. In contrast, even hours after peroxide application, the cell morphology, the number of propidium iodide-positive cells, and the extracellular activity of the cytosolic enzyme lactate dehydrogenase (LDH) were not significantly altered. The rapid loss in MTT reduction and NR uptake after exposure of neurons to H2O2 for 5 or 15 min correlated well with a strongly compromised MTT reduction and a very high extracellular LDH activity observed after further incubation in peroxide-free medium for a total incubation period of 24 hr. These data demonstrate that cultured neurons do not recover from damage that is inflicted by a short exposure to H2O2 and that the rapid losses in the capacities of neurons for MTT reduction and NR uptake are good predictors of delayed cell damage.
Collapse
Affiliation(s)
- Michaela C Hohnholt
- Centre for Biomolecular Interactions Bremen and Centre for Environmental Research and Sustainable Technology, University of Bremen, Bremen, Germany
| | - Eva M Blumrich
- Centre for Biomolecular Interactions Bremen and Centre for Environmental Research and Sustainable Technology, University of Bremen, Bremen, Germany
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen and Centre for Environmental Research and Sustainable Technology, University of Bremen, Bremen, Germany
| |
Collapse
|
13
|
Konyalioglu S, Armagan G, Yalcin A, Atalayin C, Dagci T. Effects of resveratrol on hydrogen peroxide-induced oxidative stress in embryonic neural stem cells. Neural Regen Res 2014; 8:485-95. [PMID: 25206691 PMCID: PMC4146049 DOI: 10.3969/j.issn.1673-5374.2013.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 01/05/2013] [Indexed: 12/26/2022] Open
Abstract
Resveratrol, a natural phenolic compound, has been shown to prevent cardiovascular diseases and cancer and exhibit neuroprotective effects. In this study, we examined the neuroprotective and antioxidant effects of resveratrol against hydrogen peroxide in embryonic neural stem cells. Hydrogen peroxide treatment alone increased catalase and glutathione peroxidase activities but did not change superoxide dismutase levels compared with hydrogen peroxide + resveratrol treatment. Nitric oxide synthase activity and concomitant nitric oxide levels increased in response to hydrogen peroxide treatment. Conversely, resveratrol treatment decreased nitric oxide synthase activity and nitric oxide levels. Resveratrol also attenuated hydrogen peroxide-induced nuclear or mitochondrial DNA damage. We propose that resveratrol may be a promising agent for protecting embryonic neural stem cells because of its potential to decrease oxidative stress by inducing higher activity of antioxidant enzymes, decreasing nitric oxide production and nitric oxide synthase activity, and alleviating both nuclear and mitochondrial DNA damage.
Collapse
Affiliation(s)
- Sibel Konyalioglu
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Bornova-Izmir 35100, Turkey
| | - Guliz Armagan
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Bornova-Izmir 35100, Turkey
| | - Ayfer Yalcin
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Bornova-Izmir 35100, Turkey
| | - Cigdem Atalayin
- Department of Restorative Dentistry and Endodontics, Faculty of Dentistry, Ege University, Bornova-Izmir 35100, Turkey
| | - Taner Dagci
- Department of Physiology, School of Medicine, Ege University, Bornova-Izmir 35100, Turkey ; Center for Brain Research, Ege University, Bornova-Izmir 35100, Turkey
| |
Collapse
|
14
|
Hirrlinger J, Nave KA. Adapting brain metabolism to myelination and long-range signal transduction. Glia 2014; 62:1749-61. [DOI: 10.1002/glia.22737] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 07/23/2014] [Accepted: 07/23/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Johannes Hirrlinger
- Department of Neurogenetics; Max-Planck-Institute for Experimental Medicine; Göttingen Germany
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig; Leipzig Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics; Max-Planck-Institute for Experimental Medicine; Göttingen Germany
| |
Collapse
|
15
|
Jenkins SI, Yiu HHP, Rosseinsky MJ, Chari DM. Magnetic nanoparticles for oligodendrocyte precursor cell transplantation therapies: progress and challenges. MOLECULAR AND CELLULAR THERAPIES 2014; 2:23. [PMID: 26056590 PMCID: PMC4452053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/20/2014] [Indexed: 11/21/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) have shown high promise as a transplant population to promote regeneration in the central nervous system, specifically, for the production of myelin - the protective sheath around nerve fibers. While clinical trials for these cells have commenced in some areas, there are currently key barriers to the translation of neural cell therapies. These include the ability to (a) image transplant populations in vivo; (b) genetically engineer transplant cells to augment their repair potential; and (c) safely target cells to sites of pathology. Here, we review the evidence that magnetic nanoparticles (MNPs) are a 'multifunctional nanoplatform' that can aid in safely addressing these translational challenges in neural cell/OPC therapy: by facilitating real-time and post-mortem assessment of transplant cell biodistribution, and biomolecule delivery to transplant cells, as well as non-invasive 'magnetic cell targeting' to injury sites by application of high gradient fields. We identify key issues relating to the standardization and reporting of physicochemical and biological data in the field; we consider that it will be essential to systematically address these issues in order to fully evaluate the utility of the MNP platform for neural cell transplantation, and to develop efficacious neurocompatible particles for translational applications.
Collapse
Affiliation(s)
- Stuart I Jenkins
- />Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine Keele University, Stoke-on-Trent, Staffordshire ST5 5BG UK
| | - Humphrey H P Yiu
- />School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS UK
| | | | - Divya M Chari
- />Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine Keele University, Stoke-on-Trent, Staffordshire ST5 5BG UK
| |
Collapse
|
16
|
Jenkins SI, Yiu HHP, Rosseinsky MJ, Chari DM. Magnetic nanoparticles for oligodendrocyte precursor cell transplantation therapies: progress and challenges. MOLECULAR AND CELLULAR THERAPIES 2014; 2:23. [PMID: 26056590 PMCID: PMC4452053 DOI: 10.1186/2052-8426-2-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/20/2014] [Indexed: 01/12/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) have shown high promise as a transplant population to promote regeneration in the central nervous system, specifically, for the production of myelin – the protective sheath around nerve fibers. While clinical trials for these cells have commenced in some areas, there are currently key barriers to the translation of neural cell therapies. These include the ability to (a) image transplant populations in vivo; (b) genetically engineer transplant cells to augment their repair potential; and (c) safely target cells to sites of pathology. Here, we review the evidence that magnetic nanoparticles (MNPs) are a ‘multifunctional nanoplatform’ that can aid in safely addressing these translational challenges in neural cell/OPC therapy: by facilitating real-time and post-mortem assessment of transplant cell biodistribution, and biomolecule delivery to transplant cells, as well as non-invasive ‘magnetic cell targeting’ to injury sites by application of high gradient fields. We identify key issues relating to the standardization and reporting of physicochemical and biological data in the field; we consider that it will be essential to systematically address these issues in order to fully evaluate the utility of the MNP platform for neural cell transplantation, and to develop efficacious neurocompatible particles for translational applications.
Collapse
Affiliation(s)
- Stuart I Jenkins
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine Keele University, Stoke-on-Trent, Staffordshire ST5 5BG UK
| | - Humphrey H P Yiu
- School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS UK
| | | | - Divya M Chari
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine Keele University, Stoke-on-Trent, Staffordshire ST5 5BG UK
| |
Collapse
|
17
|
Hohnholt MC, Dringen R. Short time exposure to hydrogen peroxide induces sustained glutathione export from cultured neurons. Free Radic Biol Med 2014; 70:33-44. [PMID: 24524999 DOI: 10.1016/j.freeradbiomed.2014.02.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 01/31/2014] [Accepted: 02/03/2014] [Indexed: 12/18/2022]
Abstract
Hydrogen peroxide is a normal by-product of cellular metabolism that in higher concentrations can cause oxidative stress. Cultured cerebellar granule neurons efficiently disposed of micromolar concentrations of hydrogen peroxide with half-times in the minute range in a process that predominately involved catalase. Application of up to 100 µM hydrogen peroxide did not affect the cell viability for up to 4h, but caused a time- and concentration-dependent increase in the extracellular glutathione (GSH) content that was accompanied by a matching decrease in the cellular GSH content. Hydrogen peroxide at 100 µM stimulated maximally the GSH export from viable neurons, but did not affect GSH export from cultured astrocytes. The peroxide-induced extracellular GSH accumulation from neurons was lowered by 70% in the presence of MK571, an inhibitor of multidrug resistance protein (Mrp) 1. The extracellular GSH content determined after 4h of incubation was already significantly increased after a 5-min exposure of neurons to hydrogen peroxide and became maximal after 15 min of peroxide application. These data demonstrate that just a short exposure of viable cerebellar granule neurons to micromolar concentrations of hydrogen peroxide stimulates a prolonged Mrp1-mediated export of cellular GSH. This process may compromise the antioxidative potential of neurons and increase their sensitivity toward drugs and toxins.
Collapse
Affiliation(s)
- Michaela C Hohnholt
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, 28334 Bremen, Germany; Centre for Environmental Research, and Sustainable Technology, University of Bremen, 28334 Bremen, Germany.
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, 28334 Bremen, Germany; Centre for Environmental Research, and Sustainable Technology, University of Bremen, 28334 Bremen, Germany
| |
Collapse
|
18
|
Revisiting the neuropathogenesis of Zellweger syndrome. Neurochem Int 2014; 69:1-8. [DOI: 10.1016/j.neuint.2014.02.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/11/2014] [Accepted: 02/24/2014] [Indexed: 01/27/2023]
|
19
|
Primary Cultures of Astrocytes and Neurons as Model Systems to Study the Metabolism and Metabolite Export from Brain Cells. BRAIN ENERGY METABOLISM 2014. [DOI: 10.1007/978-1-4939-1059-5_3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
20
|
Miljković D, Spasojević I. Multiple sclerosis: molecular mechanisms and therapeutic opportunities. Antioxid Redox Signal 2013; 19:2286-334. [PMID: 23473637 PMCID: PMC3869544 DOI: 10.1089/ars.2012.5068] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 02/09/2012] [Accepted: 03/09/2013] [Indexed: 12/15/2022]
Abstract
The pathophysiology of multiple sclerosis (MS) involves several components: redox, inflammatory/autoimmune, vascular, and neurodegenerative. All of them are supported by the intertwined lines of evidence, and none of them should be written off. However, the exact mechanisms of MS initiation, its development, and progression are still elusive, despite the impressive pace by which the data on MS are accumulating. In this review, we will try to integrate the current facts and concepts, focusing on the role of redox changes and various reactive species in MS. Knowing the schedule of initial changes in pathogenic factors and the key turning points, as well as understanding the redox processes involved in MS pathogenesis is the way to enable MS prevention, early treatment, and the development of therapies that target specific pathophysiological components of the heterogeneous mechanisms of MS, which could alleviate the symptoms and hopefully stop MS. Pertinent to this, we will outline (i) redox processes involved in MS initiation; (ii) the role of reactive species in inflammation; (iii) prooxidative changes responsible for neurodegeneration; and (iv) the potential of antioxidative therapy.
Collapse
Affiliation(s)
- Djordje Miljković
- Department of Immunology, Institute for Biological Research “Siniša Stanković,” University of Belgrade, Belgrade, Serbia
| | - Ivan Spasojević
- Life Sciences Department, Institute for Multidisciplinary Research, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
21
|
Abstract
Genetic deficits and loss of function for the triggering receptor expressed in myeloid cells 2 (TREM2; encoded at chr6p21.1), a transmembrane spanning stimulatory receptor of the immunoglobulin/lectin-like gene superfamily, have been associated with deficiencies in phagocytosis and the innate immune system in Alzheimer's disease. In this study, we provide evidence that TREM2 is downregulated in samples of sporadic Alzheimer hippocampal CA1 compared with age-matched controls. A nuclear factor-кB (NF-кB)-sensitive miRNA-34a (encoded at chr1p36.22), upregulated in Alzheimer's disease, was found to target the 299 nucleotide human TREM2 mRNA 3'-untranslated region (3'-UTR) and downregulate the expression of a TREM2-3'-UTR reporter vector. A stabilized anti-miRNA-34a (AM-34a) quenched this pathogenic response. The results suggest that an epigenetic mechanism involving an NF-кB-mediated, miRNA-34a-regulated downregulation of TREM2 expression may shape innate immune and phagocytic responses that contribute to inflammatory neurodegeneration.
Collapse
|
22
|
Dowell JA, Johnson JA. Mechanisms of Nrf2 protection in astrocytes as identified by quantitative proteomics and siRNA screening. PLoS One 2013; 8:e70163. [PMID: 23922950 PMCID: PMC3726381 DOI: 10.1371/journal.pone.0070163] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 06/16/2013] [Indexed: 12/30/2022] Open
Abstract
The Nrf2 (NF-E2 related factor 2)-ARE (antioxidant response element) pathway controls a powerful array of endogenous cellular antioxidant systems and is an important pathway in the detoxification of reactive oxygen species (ROS) in the brain. Using a combination of quantitative proteomics and siRNA screening, we have identified novel protective mechanisms of the Nrf2-ARE pathway against oxidative stress in astrocytes. Studies from our lab and others have shown Nrf2 overexpression protects astrocytes from oxidative stress. However, the exact mechanisms by which Nrf2 elicits these effects are unknown. In this study, we show that induction of Nrf2 reduces levels of reactive oxygen species (ROS) produced by various oxidative stressors and results in robust cytoprotection. To identify the enzymes responsible for these effects, we used stable isotope labeling by amino acids in cell culture (SILAC) and quantitative shotgun proteomics to identify 72 Nrf2-regulated proteins in astrocytes. We hypothesized a subset of these proteins might play a critical role in Nrf2 protection. In order to identify these critical proteins, we used bioinformatics to narrow our target list of proteins and then systematically screened each candidate with siRNA to assess the role of each in Nrf2 protection. We screened each target against H2O2, tert-butyl hydroperoxide, and 4-hydroxynonenal and subsequently identified three enzymes-catalase, prostaglandin reductase-1, and peroxiredoxin-6-that are critical for Nrf2-mediated protection in astrocytes.
Collapse
Affiliation(s)
- James A. Dowell
- Division of Pharmaceutical Science, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jeffrey A. Johnson
- Division of Pharmaceutical Science, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Center of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
23
|
Amaral AI, Meisingset TW, Kotter MR, Sonnewald U. Metabolic aspects of neuron-oligodendrocyte-astrocyte interactions. Front Endocrinol (Lausanne) 2013; 4:54. [PMID: 23717302 PMCID: PMC3651962 DOI: 10.3389/fendo.2013.00054] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 04/24/2013] [Indexed: 11/26/2022] Open
Abstract
Whereas astrocytes have been in the limelight of scientific interest in brain energy metabolism for a while, oligodendrocytes are still waiting for a place on the metabolic stage. We propose to term the interaction of oligodendrocytes with astrocytes and neurons: NOA (neuron-oligodendrocyte-astrocyte) interactions. One of the reasons to find out more about metabolic interactions between oligodendrocytes, neurons, and astrocytes is to establish markers of healthy oligodendrocyte metabolism that could be used for the diagnosis and assessment of white matter disease. The vesicular release of glutamate in the white matter has received considerable attention in the past. Oligodendrocyte lineage cells express glutamate receptors and glutamate toxicity has been implicated in diseases affecting oligodendrocytes such as hypoxic-ischaemic encephalopathy, inflammatory diseases and trauma. As oligodendrocyte precursor cells vividly react to injury it is also important to establish whether cells recruited into damaged areas are able to regenerate lost myelin sheaths or whether astrocytic scarring occurs. It is therefore important to consider metabolic aspects of astrocytes and oligodendrocytes separately. The present review summarizes the limited evidence available on metabolic cycles in oligodendrocytes and so hopes to stimulate further research interests in this important field.
Collapse
Affiliation(s)
- Ana I. Amaral
- Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust and Medical Research Council Cambridge Stem Cell Institute, Department of Clinical Neurosciences, University of CambridgeCambridge, UK
| | - Tore W. Meisingset
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and TechnologyTrondheim, Norway
| | - Mark R. Kotter
- Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust and Medical Research Council Cambridge Stem Cell Institute, Department of Clinical Neurosciences, University of CambridgeCambridge, UK
| | - Ursula Sonnewald
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and TechnologyTrondheim, Norway
- *Correspondence: Ursula Sonnewald, Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and Technology, PO Box 8905, MTFS, 7491 Trondheim, Norway. e-mail:
| |
Collapse
|
24
|
Role of peroxisomes in ROS/RNS-metabolism: Implications for human disease. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1363-73. [DOI: 10.1016/j.bbadis.2011.12.001] [Citation(s) in RCA: 412] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 11/25/2011] [Accepted: 12/02/2011] [Indexed: 12/27/2022]
|
25
|
The role of amino acid transporters in GSH synthesis in the blood-brain barrier and central nervous system. Neurochem Int 2012; 61:405-14. [PMID: 22634224 DOI: 10.1016/j.neuint.2012.05.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 04/17/2012] [Accepted: 05/15/2012] [Indexed: 12/22/2022]
Abstract
Glutathione (GSH) plays a critical role in protecting cells from oxidative stress and xenobiotics, as well as maintaining the thiol redox state, most notably in the central nervous system (CNS). GSH concentration and synthesis are highly regulated within the CNS and are limited by availability of the sulfhydryl amino acid (AA) l-cys, which is mainly transported from the blood, through the blood-brain barrier (BBB), and into neurons. Several antiporter transport systems (e.g., x(c)(-), x(-)(AG), and L) with clearly different luminal and abluminal distribution, Na(+), and pH dependency have been described in brain endothelial cells (BEC) of the BBB, as well as in neurons, astrocytes, microglia and oligodendrocytes from different brain structures. The purpose of this review is to summarize information regarding the different AA transport systems for l-cys and its oxidized form l-cys(2) in the CNS, such as expression and activity in blood-brain barrier endothelial cells, astrocytes and neurons and environmental factors that modulate transport kinetics.
Collapse
|
26
|
Park SJ, Lee JH, Kim HY, Choi YH, Park JS, Suh YH, Park SM, Joe EH, Jou I. Astrocytes, but not microglia, rapidly sense H₂O₂via STAT6 phosphorylation, resulting in cyclooxygenase-2 expression and prostaglandin release. THE JOURNAL OF IMMUNOLOGY 2012; 188:5132-41. [PMID: 22504638 DOI: 10.4049/jimmunol.1101600] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Emerging evidence has established that astrocytes, once considered passive supporting cells that maintained extracellular ion levels and served as a component of the blood-brain barrier, play active regulatory roles during neurogenesis and in brain pathology. In the current study, we demonstrated that astrocytes sense H(2)O(2) by rapidly phosphorylating the transcription factor STAT6, a response not observed in microglia. STAT6 phosphorylation was induced by generators of other reactive oxygen species (ROS) and reactive nitrogen species, as well as in the reoxygenation phase of hypoxia/reoxygenation, during which ROS are generated. Src-JAK pathways mediated STAT6 phosphorylation upstream. Experiments using lipid raft disruptors and analyses of detergent-fractionated cells demonstrated that H(2)O(2)-induced STAT6 phosphorylation occurred in lipid rafts. Under experimental conditions in which H(2)O(2) did not affect astrocyte viability, H(2)O(2)-induced STAT6 phosphorylation resulted in STAT6-dependent cyclooxygenase-2 expression and subsequent release of PGE(2) and prostacyclin, an effect also observed in hypoxia/reoxygenation. Finally, PGs released from H(2)O(2)-stimulated astrocytes inhibited microglial TNF-α expression. Accordingly, our results indicate that ROS-induced STAT6 phosphorylation in astrocytes can modulate the functions of neighboring cells, including microglia, through cyclooxygenase-2 induction and subsequent release of PGs. Differences in the sensitivity of STAT6 in astrocytes (highly sensitive) and microglia (insensitive) to phosphorylation following brief exposure to H(2)O(2) suggest that astrocytes can act as sentinels for certain stimuli, including H(2)O(2) and ROS, refining the canonical notion that microglia are the first line of defense against external stimuli.
Collapse
Affiliation(s)
- Soo Jung Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon 442-721, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
|
28
|
Increased neuronal glutathione and neuroprotection in GTRAP3-18-deficient mice. Neurobiol Dis 2011; 45:973-82. [PMID: 22210510 DOI: 10.1016/j.nbd.2011.12.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 10/12/2011] [Accepted: 12/04/2011] [Indexed: 11/22/2022] Open
Abstract
Glutathione (GSH) is an important neuroprotective molecule in the brain. The strategy to increase neuronal GSH level is a promising approach to the treatment of neurodegenerative diseases. However, the regulatory mechanism by which neuron-specific GSH synthesis is facilitated remains elusive. Glutamate transporter-associated protein 3-18 (GTRAP3-18) is an endoplasmic reticulum protein interacting with excitatory amino acid carrier 1 (EAAC1), which is a neuronal glutamate/cysteine transporter. To investigate the potential regulatory mechanism to increase neuronal GSH level in vivo, we generated GTRAP3-18-deficient (GTRAP3-18(-/-)) mice using a gene-targeting approach. Disruption of the GTRAP3-18 gene resulted in increased EAAC1 expression in the plasma membrane, increased neuronal GSH content and neuroprotection against oxidative stress. In addition, GTRAP3-18(-/-) mice performed better in motor/spatial learning and memory tests than wild-type mice. Therefore, the suppression of GTRAP3-18 increases neuronal resistance to oxidative stress by increasing GSH content and also facilitates cognitive function. The present results may provide a molecular basis for the development of treatments for neurodegenerative diseases.
Collapse
|
29
|
Backos DS, Franklin CC, Reigan P. The role of glutathione in brain tumor drug resistance. Biochem Pharmacol 2011; 83:1005-12. [PMID: 22138445 DOI: 10.1016/j.bcp.2011.11.016] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 11/18/2011] [Accepted: 11/18/2011] [Indexed: 12/24/2022]
Abstract
Chemotherapy is central to the current treatment modality for primary human brain tumors, but despite high-dose and intensive treatment regimens there has been little improvement in patient outcome. The development of tumor chemoresistance has been proposed as a major contributor to this lack of response. While there have been some improvements in our understanding of the molecular mechanisms underlying brain tumor drug resistance over the past decade, the contribution of glutathione (GSH) and the GSH-related enzymes to drug resistance in brain tumors have been largely overlooked. GSH constitutes a major antioxidant defense system in the brain and together with the GSH-related enzymes plays an important role in protecting cells against free radical damage and dictating tumor cell response to adjuvant cancer therapies, including irradiation and chemotherapy. Glutamate cysteine ligase (GCL), glutathione synthetase (GS), glutathione peroxidase (GPx), glutathione reductase (GR), glutathione-S-transferases (GST), and GSH complex export transporters (GS-X pumps) are major components of the GSH-dependent enzyme system that function in a dynamic cascade to maintain redox homeostasis. In many tumors, the GSH system is often dysregulated, resulting in a more drug resistant phenotype. This is commonly associated with GST-mediated GSH conjugation of various anticancer agents leading to the formation of less toxic GSH-drug complexes, which can be readily exported from the cell. Advances in our understanding of the mechanisms of drug resistance and patient selection based on biomarker profiles will be crucial to adapt therapeutic strategies and improve outcomes for patients with primary malignant brain tumors.
Collapse
Affiliation(s)
- Donald S Backos
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, 80045, United States
| | | | | |
Collapse
|
30
|
Hohnholt MC, Geppert M, Dringen R. Treatment with iron oxide nanoparticles induces ferritin synthesis but not oxidative stress in oligodendroglial cells. Acta Biomater 2011; 7:3946-54. [PMID: 21763792 DOI: 10.1016/j.actbio.2011.06.052] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 06/23/2011] [Accepted: 06/29/2011] [Indexed: 11/18/2022]
Abstract
Magnetic iron oxide nanoparticles (IONPs) have been used for a variety of neurobiological applications, although little is yet known as to the fate of such particles in brain cells. To address these questions, we have exposed oligodendroglial OLN-93 cells to dimercaptosuccinate-coated IONPs. Treatment of the cells strongly increased the specific cellular iron content proportional to the IONP concentrations applied (0-1000 μM total iron as IONPs) up to 300-fold, but did not cause any acute cytotoxicity or induce oxidative stress. To investigate the potential of OLN-93 cells to liberate iron from the accumulated IONPs, we have studied the upregulation of the iron storage protein ferritin and the cell proliferation as cellular processes that depend on the availability of low-molecular-weight iron. The presence of IONPs caused a concentration-dependent increase in the amount of cellular ferritin and partially bypassed the inhibition of cell proliferation by the iron chelator deferoxamine. These data demonstrate that viable OLN-93 cells efficiently take up IONPs and suggest that these cells are able to use iron liberated from accumulated IONPs for their metabolism.
Collapse
Affiliation(s)
- Michaela C Hohnholt
- Centre for Biomolecular Interactions Bremen, University of Bremen, D-28334 Bremen, Germany
| | | | | |
Collapse
|
31
|
Mitozo PA, de Souza LF, Loch-Neckel G, Flesch S, Maris AF, Figueiredo CP, Dos Santos ARS, Farina M, Dafre AL. A study of the relative importance of the peroxiredoxin-, catalase-, and glutathione-dependent systems in neural peroxide metabolism. Free Radic Biol Med 2011; 51:69-77. [PMID: 21440059 DOI: 10.1016/j.freeradbiomed.2011.03.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 03/09/2011] [Accepted: 03/11/2011] [Indexed: 01/01/2023]
Abstract
Cells are endowed with several overlapping peroxide-degrading systems whose relative importance is a matter of debate. In this study, three different sources of neural cells (rat hippocampal slices, rat C6 glioma cells, and mouse N2a neuroblastoma cells) were used as models to understand the relative contributions of individual peroxide-degrading systems. After a pretreatment (30 min) with specific inhibitors, each system was challenged with either H₂O₂ or cumene hydroperoxide (CuOOH), both at 100 μM. Hippocampal slices, C6 cells, and N2a cells showed a decrease in the H₂O₂ decomposition rate (23-28%) by a pretreatment with the catalase inhibitor aminotriazole. The inhibition of glutathione reductase (GR) by BCNU (1,3-bis(2-chloroethyl)-1-nitrosourea) significantly decreased H₂O₂ and CuOOH decomposition rates (31-77%). Inhibition of catalase was not as effective as BCNU at decreasing cell viability (MTT assay) and cell permeability or at increasing DNA damage (comet test). Impairing the thioredoxin (Trx)-dependent peroxiredoxin (Prx) recycling by thioredoxin reductase (TrxR) inhibition with auranofin neither potentiated peroxide toxicity nor decreased the peroxide-decomposition rate. The results indicate that neural peroxidatic systems depending on Trx/TrxR for recycling are not as important as those depending on GSH/GR. Dimer formation, which leads to Prx2 inactivation, was observed in hippocampal slices and N2a cells treated with H₂O₂, but not in C6 cells. However, Prx-SO₃ formation, another form of Prx inactivation, was observed in all neural cell types tested, indicating that redox-mediated signaling pathways can be modulated in neural cells. These differences in Prx2 dimerization suggest specific redox regulation mechanisms in glia-derived (C6) compared to neuron-derived (N2a) cells and hippocampal slices.
Collapse
Affiliation(s)
- Péricles Arruda Mitozo
- Departamento de Ciências Fisiológicas, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, SC, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Aoyama K, Watabe M, Nakaki T. Modulation of neuronal glutathione synthesis by EAAC1 and its interacting protein GTRAP3-18. Amino Acids 2011; 42:163-9. [PMID: 21373771 DOI: 10.1007/s00726-011-0861-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 02/17/2011] [Indexed: 01/17/2023]
Abstract
Glutathione (GSH) plays essential roles in different processes such as antioxidant defenses, cell signaling, cell proliferation, and apoptosis in the central nervous system. GSH is a tripeptide composed of glutamate, cysteine, and glycine. The concentration of cysteine in neurons is much lower than that of glutamate or glycine, so that cysteine is the rate-limiting substrate for neuronal GSH synthesis. Most neuronal cysteine uptake is mediated through the neuronal sodium-dependent glutamate transporter, known as excitatory amino acid carrier 1 (EAAC1). Glutamate transporters are vulnerable to oxidative stress and EAAC1 dysfunction impairs neuronal GSH synthesis by reducing cysteine uptake. This may start a vicious circle leading to neurodegeneration. Intracellular signaling molecules functionally regulate EAAC1. Glutamate transporter-associated protein 3-18 (GTRAP3-18) activation down-regulates EAAC1 function. Here, we focused on the interaction between EAAC1 and GTRAP3-18 at the plasma membrane to investigate their effects on neuronal GSH synthesis. Increased level of GTRAP3-18 protein induced a decrease in GSH level and, thereby, increased the vulnerability to oxidative stress, while decreased level of GTRAP3-18 protein induced an increase in GSH level in vitro. We also confirmed these results in vivo. Our studies demonstrate that GTRAP3-18 regulates neuronal GSH level by controlling the EAAC1-mediated uptake of cysteine.
Collapse
Affiliation(s)
- Koji Aoyama
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8605, Japan
| | | | | |
Collapse
|
33
|
Fumaric acid dialkyl esters deprive cultured rat oligodendroglial cells of glutathione and upregulate the expression of heme oxygenase 1. Neurosci Lett 2010; 475:56-60. [DOI: 10.1016/j.neulet.2010.03.048] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2010] [Revised: 03/10/2010] [Accepted: 03/18/2010] [Indexed: 11/17/2022]
|
34
|
Chu PWY, Beart PM, Jones NM. Preconditioning protects against oxidative injury involving hypoxia-inducible factor-1 and vascular endothelial growth factor in cultured astrocytes. Eur J Pharmacol 2010; 633:24-32. [PMID: 20153315 DOI: 10.1016/j.ejphar.2010.02.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 01/18/2010] [Accepted: 02/02/2010] [Indexed: 10/19/2022]
Abstract
Tolerance to brain injury involves hypoxia-inducible factor-1 (HIF-1) and its target genes as the key pathway mediating a cascade of events including cell survival, energetics, and angiogenesis. In this study, we established the treatment paradigms for an in vitro model of tolerance to oxidative injury in primary astrocytic cultures and further examined the roles for the HIF-1 signalling cascade. Isolated murine astrocytes were preconditioned with sub-toxic concentrations of HIF-1 inducers and subsequently exposed to a H(2)O(2) insult, where changes in cell viability and protein expression were determined. Preconditioning with non-damaging concentrations of desferrioxamine (DFO) and ethyl-3,4-dihydroxybenzoate (EDHB) significantly improved cellular viability after H(2)O(2) injury treatment. Time course studies revealed that DFO and EDHB treatments alone induced sequential activation of HIF-1 signal transduction where nuclear HIF-1alpha protein accumulation was detected as early as 2h, followed by downstream upregulation of intracellular and released VEGF from 4h and 8h onwards, respectively. The protective effects of DFO and EDHB preconditioning against H(2)O(2) injury were abolished by co-treatment with cycloheximide, an inhibitor of protein synthesis. Importantly, when the anti-HIF-1 compound, 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole (YC-1) was used, the cytoprotection and VEGF accumulation produced by DFO and EDHB preconditioning were diminished. These results indicate the essential role of the HIF-1 pathway in our model of tolerance against oxidative injury in cultured astrocytes, and suggest roles for astrocytic HIF-1 expression and VEGF release which may influence the function of surrounding cells and vasculature during oxidative stress-related brain diseases.
Collapse
Affiliation(s)
- Percy W Y Chu
- Molecular Neuropharmacology, Florey Neuroscience Institutes, University of Melbourne, VIC 3010, Australia
| | | | | |
Collapse
|
35
|
Bartzokis G. Alzheimer's disease as homeostatic responses to age-related myelin breakdown. Neurobiol Aging 2009; 32:1341-71. [PMID: 19775776 DOI: 10.1016/j.neurobiolaging.2009.08.007] [Citation(s) in RCA: 405] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2008] [Revised: 08/13/2009] [Accepted: 08/17/2009] [Indexed: 12/11/2022]
Abstract
The amyloid hypothesis (AH) of Alzheimer's disease (AD) posits that the fundamental cause of AD is the accumulation of the peptide amyloid beta (Aβ) in the brain. This hypothesis has been supported by observations that genetic defects in amyloid precursor protein (APP) and presenilin increase Aβ production and cause familial AD (FAD). The AH is widely accepted but does not account for important phenomena including recent failures of clinical trials to impact dementia in humans even after successfully reducing Aβ deposits. Herein, the AH is viewed from the broader overarching perspective of the myelin model of the human brain that focuses on functioning brain circuits and encompasses white matter and myelin in addition to neurons and synapses. The model proposes that the recently evolved and extensive myelination of the human brain underlies both our unique abilities and susceptibility to highly prevalent age-related neuropsychiatric disorders such as late onset AD (LOAD). It regards oligodendrocytes and the myelin they produce as being both critical for circuit function and uniquely vulnerable to damage. This perspective reframes key observations such as axonal transport disruptions, formation of axonal swellings/sphenoids and neuritic plaques, and proteinaceous deposits such as Aβ and tau as by-products of homeostatic myelin repair processes. It delineates empirically testable mechanisms of action for genes underlying FAD and LOAD and provides "upstream" treatment targets. Such interventions could potentially treat multiple degenerative brain disorders by mitigating the effects of aging and associated changes in iron, cholesterol, and free radicals on oligodendrocytes and their myelin.
Collapse
Affiliation(s)
- George Bartzokis
- Department of Psychiatry and Biobehavioral Sciences, The David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
36
|
Miller VM, Lawrence DA, Mondal TK, Seegal RF. Reduced glutathione is highly expressed in white matter and neurons in the unperturbed mouse brain--implications for oxidative stress associated with neurodegeneration. Brain Res 2009; 1276:22-30. [PMID: 19393633 PMCID: PMC2744579 DOI: 10.1016/j.brainres.2009.04.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 03/13/2009] [Accepted: 04/12/2009] [Indexed: 10/20/2022]
Abstract
Oxidative stress is implicated in the pathogenesis of many neurodegenerative diseases, including Parkinson's disease and Alzheimer's disease. The depletion of glutathione (GSH) a powerful antioxidant renders cells particularly vulnerable to oxidative stress. Isolated neuronal and glial cell culture studies suggest that glia rather than neurons have greatest reserves of GSH, implying that neurons are most sensitive to oxidative stress. However, pathological in vivo studies suggest that GSH associated enzymes are elevated in neurons rather than astrocytes. The active, reduced form of GSH is rapidly degraded thus making it difficult to identify the location of GSH in post-mortem tissue. Therefore, to determine whether GSH is more highly expressed in neurons or astrocytes we perfused mouse brains with a solution containing NEM which reacts with the sulfhydryl group of GSH, thus locking the active form in situ, prior to immunostaining with an anti-GS-NEM antibody. We obtained brightfield and fluorescent digital images of sections stained with DAPI and antibodies directed against GS-NEM, glial fibrillary acidic protein (GFAP) in regions containing the hippocampus, striatum, frontal cortex, midbrain nuclei, cerebellum and reticular formation neurons. GSH was most abundant in neurons and white matter in all brain regions, and only in occasional astrocytes lining the third and fourth ventricles. High levels of GSH in neurons and white matter, suggests astrocytes rather than neurons may be particularly vulnerable to oxidative stress.
Collapse
Affiliation(s)
- V M Miller
- Biggs Laboratory, Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY 12201, USA.
| | | | | | | |
Collapse
|
37
|
Murin R, Cesar M, Kowtharapu BS, Verleysdonk S, Hamprecht B. Expression of Pyruvate Carboxylase in Cultured Oligodendroglial, Microglial and Ependymal Cells. Neurochem Res 2008; 34:480-9. [DOI: 10.1007/s11064-008-9806-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2008] [Accepted: 07/07/2008] [Indexed: 11/28/2022]
|
38
|
Ferrero-Gutiérrez A, Pérez-Gómez A, Novelli A, Fernández-Sánchez MT. Inhibition of protein phosphatases impairs the ability of astrocytes to detoxify hydrogen peroxide. Free Radic Biol Med 2008; 44:1806-16. [PMID: 18313406 DOI: 10.1016/j.freeradbiomed.2008.01.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Revised: 01/11/2008] [Accepted: 01/17/2008] [Indexed: 10/22/2022]
Abstract
We have used protein phosphatase (PP) inhibitors and rat cerebellar glial cells in primary culture to investigate the role of PP activity in the ability of glial cells to detoxify exogenously applied hydrogen peroxide (H2O2). The marine toxin okadaic acid (OKA), a potent PP1 and PP2A inhibitor, caused a concentration-dependent degeneration of astrocytes and increased the formation of hydroperoxide radicals significantly. Subtoxic exposures to OKA significantly potentiated toxicity by exogenous H2O2. The concentration of H2O2 that reduced by 50% the survival of astrocytes after 3 h was estimated at 720+/-40 microM in the absence and 85+/-30 microM in the presence of the toxin. The PP inhibitors calyculin A and endothall also potentiated H2O2 toxicity in cerebellar astrocytes. OKA caused a time-dependent inhibition of both glial catalase and glutathione peroxidase, reducing by approximately 50% the activity of these enzymes after 3 h, whereas other enzymatic activities remained unaffected. Also, OKA reduced the cellular content of total glutathione and elevated oxidized glutathione to about 25% of total glutathione. OKA-treated astrocytes cleared H2O2 from the incubation medium approximately two times more slowly than control cultures. Our results suggest a prominent role for PP activity in the antioxidant mechanisms protecting astrocytes against damage by H2O2.
Collapse
Affiliation(s)
- Amaia Ferrero-Gutiérrez
- Department of Biochemistry and Molecular Biology, Institute of Biotechnology of Asturias, University of Oviedo, 33006 Oviedo, Spain
| | | | | | | |
Collapse
|
39
|
Kippert A, Trajkovic K, Fitzner D, Opitz L, Simons M. Identification of Tmem10/Opalin as a novel marker for oligodendrocytes using gene expression profiling. BMC Neurosci 2008; 9:40. [PMID: 18439243 PMCID: PMC2396630 DOI: 10.1186/1471-2202-9-40] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Accepted: 04/25/2008] [Indexed: 01/06/2023] Open
Abstract
Background During the development of the central nervous system, oligodendrocytes generate large amounts of myelin, a multilayered insulating membrane that ensheathes axons, thereby allowing the fast conduction of the action potential and maintaining axonal integrity. Differentiation of oligodendrocytes to myelin-forming cells requires the downregulation of RhoA GTPase activity. Results To gain insights into the molecular mechanisms of oligodendrocyte differentiation, we performed microarray expression profiling of the oligodendroglial cell line, Oli-neu, treated with the Rho kinase (ROCK) inhibitor, Y-27632 or with conditioned neuronal medium. This resulted in the identification of the transmembrane protein 10 (Tmem10/Opalin), a novel type I transmembrane protein enriched in differentiating oligodendrocytes. In primary cultures, Tmem10 was abundantly expressed in O4-positive oligodendrocytes, but not in oligodendroglial precursor cells, astrocytes, microglia or neurons. In mature oligodendrocytes Tmem10 was enriched in the rims and processes of the cells and was only found to a lesser extent in the membrane sheets. Conclusion Together, our results demonstrate that Tmem10 is a novel marker for in vitro generated oligodendrocytes.
Collapse
Affiliation(s)
- Angelika Kippert
- Centre for Biochemistry and Molecular Cell Biology, University of Göttingen Humboldtallee 23, Göttingen, Germany.
| | | | | | | | | |
Collapse
|
40
|
Murín R, Schaer A, Kowtharapu BS, Verleysdonk S, Hamprecht B. Expression of 3-hydroxyisobutyrate dehydrogenase in cultured neural cells. J Neurochem 2008; 105:1176-86. [PMID: 18284611 DOI: 10.1111/j.1471-4159.2008.05298.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The branched-chain amino acids (BCAAs)--isoleucine, leucine, and valine--belong to the limited group of substances transported through the blood-brain barrier. One of the functions they are thought to have in brain is to serve as substrates for meeting parenchymal energy demands. Previous studies have shown the ubiquitous expression of a branched-chain alpha-keto acid dehydrogenase among neural cells. This enzyme catalyzes the initial and rate-limiting step in the irreversible degradative pathway for the carbon skeleton of valine and the other two branched-chain amino acids. Unlike the acyl-CoA derivates in the irreversible part of valine catabolism, 3-hydroxyisobutyrate could be expected to be released from cells by transport across the mitochondrial and plasma membranes. This could indeed be demonstrated for cultured astroglial cells. Therefore, to assess the ability of neural cells to make use of this valine-derived carbon skeleton as a metabolic substrate for the generation of energy, we investigated the expression in cultured neural cells of the enzyme processing this hydroxy acid, 3-hydroxyisobutyrate dehydrogenase (HIBDH). To achieve this, HIBDH was purified from bovine liver to serve as antigen for the production of an antiserum. Affinity-purified antibodies against HIBDH specifically recognized the enzyme in liver and brain homogenates. Immunocytochemistry demonstrated the ubiquitous expression of HIBDH among cultured glial (astroglial, oligodendroglial, microglial, and ependymal cells) and neuronal cells. Using an RT-PCR technique, these findings were corroborated by the detection of HIBDH mRNA in these cells. Furthermore, immunofluorescence double-labeling of astroglial cells with antisera against HIBDH and the mitochondrial marker pyruvate dehydrogenase localized HIBDH to mitochondria. The expression of HIBDH in neural cells demonstrates their potential to utilize valine imported into the brain for the generation of energy.
Collapse
Affiliation(s)
- Radovan Murín
- Interfaculty Institute for Biochemistry, University of Tuebingen, Tuebingen, Germany.
| | | | | | | | | |
Collapse
|
41
|
Furian AF, Oliveira MS, Royes LFF, Fiorenza NG, Fighera MR, Myskiw JC, Weiblen R, Rubin MA, Frussa-Filho R, Mello CF. GM1 ganglioside induces vasodilation and increases catalase content in the brain. Free Radic Biol Med 2007; 43:924-32. [PMID: 17697937 DOI: 10.1016/j.freeradbiomed.2007.05.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2006] [Revised: 05/27/2007] [Accepted: 05/31/2007] [Indexed: 10/23/2022]
Abstract
Monosialoganglioside (GM1) is a glycosphingolipid present in most cell membranes that displays antioxidant and neuroprotective properties. GM1 increases catalase activity in cerebral cortices in vivo, but the mechanisms underlying this effect of GM1 are not known. In the current study we investigated the effect of GM1 (50 mg/kg, ip) on the content of hemoglobin and catalase activity of hippocampus, cortex, and striatum of rats. GM1 administration increased catalase activity and hemoglobin content in brain samples after 30 min, but had no effect on blood catalase activity. GM1-induced increase in catalase activity was abolished by brain perfusion with heparinized saline. Brain catalase activity in the absence of blood, estimated by regression analysis of data from perfused and nonperfused animals, was not altered by the systemic injection of GM1. Moreover, the addition of GM1 (30 or 100 microM) did not increase catalase activity in slices of cerebral cortex in situ, further suggesting that blood circulation is required for this effect. The GM1-induced vasodilation was confirmed in vivo, because the systemic injection of GM1 (50 mg/kg, ip) increased (1.2-1.6 times) the width of pial vessels.
Collapse
Affiliation(s)
- Ana Flávia Furian
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kassmann CM, Lappe-Siefke C, Baes M, Brügger B, Mildner A, Werner HB, Natt O, Michaelis T, Prinz M, Frahm J, Nave KA. Axonal loss and neuroinflammation caused by peroxisome-deficient oligodendrocytes. Nat Genet 2007; 39:969-76. [PMID: 17643102 DOI: 10.1038/ng2070] [Citation(s) in RCA: 263] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Accepted: 05/14/2007] [Indexed: 01/02/2023]
Abstract
Oligodendrocytes myelinate axons for rapid impulse conduction and contribute to normal axonal functions in the central nervous system. In multiple sclerosis, demyelination is caused by autoimmune attacks, but the role of oligodendroglial cells in disease progression and axon degeneration is unclear. Here we show that oligodendrocytes harbor peroxisomes whose function is essential for maintaining white matter tracts throughout adult life. By selectively inactivating the import factor PEX5 in myelinating glia, we generated mutant mice that developed normally, but within several months showed ataxia, tremor and premature death. Absence of functional peroxisomes from oligodendrocytes caused widespread axonal degeneration and progressive subcortical demyelination, but did not interfere with glial survival. Moreover, it caused a strong proinflammatory milieu and, unexpectedly, the infiltration of B and activated CD8+ T cells into brain lesions. We conclude that peroxisomes provide oligodendrocytes with an essential neuroprotective function against axon degeneration and neuroinflammation, which is relevant for human demyelinating diseases.
Collapse
Affiliation(s)
- Celia M Kassmann
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, D-37075 Göttingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Muyderman H, Wadey AL, Nilsson M, Sims NR. Mitochondrial glutathione protects against cell death induced by oxidative and nitrative stress in astrocytes. J Neurochem 2007; 102:1369-82. [PMID: 17484727 DOI: 10.1111/j.1471-4159.2007.04641.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The major cellular antioxidant, glutathione, is mostly localized in the cytosol but a small portion is found in mitochondria. We have recently shown that highly selective depletion of mitochondrial glutathione in astrocytes in culture markedly increased cell death induced by the peroxynitrite donor, 3-morpholino-syndnonimine. The present study was aimed at characterizing the increase in susceptibility arising from mitochondrial glutathione loss and testing the possibility that elevating this metabolite pool above normal values could be protective. The increased vulnerability of astrocytes with depleted mitochondrial glutathione to Sin-1 was confirmed. Furthermore, these cells showed marked increases in sensitivity to hydrogen peroxide and also to high concentrations of the nitric oxide donor, S-nitroso-N-acetyl-penicillamine. The increase in cell death was mostly due to necrosis as indicated by substantially increased release of lactate dehydrogenase and staining of nuclei with propidium iodide but little change in annexin V staining and caspase 3 activation. The enhanced cell loss was blocked by prior restoration of the mitochondrial glutathione content. It was also essentially fully inhibited by treatment with cyclosporin A, consistent with a role for the mitochondrial permeability transition in the development of cell death. Susceptibility to the classical apoptosis inducer, staurosporine, was only affected to a small extent in contrast to the response to the other substances tested. Incubation of normal astrocytes with glutathione monoethylester produced large and long-lasting increases in mitochondrial glutathione content with much smaller effects on the cytosolic glutathione pool. This treatment reduced cell death on exposure to 3-morpholino-syndnonimine or hydrogen peroxide but not S-nitroso-N-acetyl-pencillamine or staurosporine. These findings provide evidence for an important role for mitochondrial glutathione in preserving cell viability during periods of oxidative or nitrative stress and indicate that increases in this glutathione pool can confer protection against some of these stressors.
Collapse
Affiliation(s)
- Håkan Muyderman
- Centre for Neuroscience and Department of Medical Biochemistry, School of Medicine, Flinders University, Adelaide, Australia.
| | | | | | | |
Collapse
|
44
|
Masuda T, Hida H, Kanda Y, Aihara N, Ohta K, Yamada K, Nishino H. Oral administration of metal chelator ameliorates motor dysfunction after a small hemorrhage near the internal capsule in rat. J Neurosci Res 2007; 85:213-22. [PMID: 17061255 DOI: 10.1002/jnr.21089] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Cerebral hemorrhage leads to local production of free iron, radicals, cytokines, etc. To investigate whether a decrease of iron-mediated radical production influences functional recovery after intracerebral hemorrhage (ICH), a modified ICH rat model with a small hemorrhage near the internal capsule (IC) accompanied with relatively severe motor dysfunction was first developed. Then clioquinol (CQ), an iron chelator that reduces hydroxyl radical production, was orally administrated. Injection of different doses of Type IV collagenase (1.4 mul 1-200 U/ml) into the left striatum near the IC in Wistar rats showed that injection of 7.5 U/ml collagenase resulted in a small hemorrhoidal lesion near the IC with relatively severe motor dysfunction (IC model). Retrograde labeling of neurons in the sensory-motor cortex and axons in the corticospinal tract using Fluoro-gold (FG) injection into the spinal cord (C3-C4) showed that few labeled neurons in the sensory-motor cortex were detected in the IC model, FG-labeled axons disappeared, and FG-including ED-1-positive cells appeared within 24 hr in the IC. Assessments of behavior and histologic analysis after oral administration of CQ in the IC model indicated that oral administration of CQ prevented a decrease of FG-labeled neurons, and resulted in better motor-function recovery. CQ inhibited hydrogen peroxide-induced cell toxicity in oligodendrocytes in vitro, but not in neurons. Our data suggests that CQ ameliorated motor dysfunction after a small hemorrhage near the IC by a mechanism that is related to reduction of chain-reactive hydroxyl radical production in oligodendrocytes.
Collapse
Affiliation(s)
- Tadashi Masuda
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Diaz-Hernandez JI, Moncada S, Bolaños JP, Almeida A. Poly(ADP-ribose) polymerase-1 protects neurons against apoptosis induced by oxidative stress. Cell Death Differ 2007; 14:1211-21. [PMID: 17347665 DOI: 10.1038/sj.cdd.4402117] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In neurons, DNA is prone to free radical damage, although repair mechanisms preserve the genomic integrity. However, activation of the DNA repair system, poly(ADP-ribose) polymerase (PARP-1), is thought to cause neuronal death through NAD+ depletion and mitochondrial membrane potential (delta psi(m)) depolarization. Here, we show that abolishing PARP-1 activity in primary cortical neurons can either enhance or prevent apoptotic death, depending on the intensity of an oxidative stress. Only in severe oxidative stress does PARP-1 activation result in NAD+ and ATP depletion and neuronal death. To investigate the role of PARP-1 in an endogenous model of oxidative stress, we used an RNA interference (RNAi) strategy to specifically knock down glutamate-cysteine ligase (GCL), the rate-limiting enzyme of glutathione biosynthesis. GCL RNAi spontaneously elicited a mild type of oxidative stress that was enough to stimulate PARP-1 in a Ca2+-calmodulin kinase II-dependent manner. GCL RNAi-mediated PARP-1 activation facilitated DNA repair, although neurons underwent delta psi(m) loss followed by some apoptotic death. PARP-1 inhibition did not prevent delta psi(m) loss, but enhanced the vulnerability of neurons to apoptosis upon GCL silencing. Conversely, mild expression of PARP-1 partially prevented to GCL RNAi-dependent apoptosis. Thus, in the mild progressive damage likely occur in neurodegenerative diseases, PARP-1 activation plays a neuroprotective role that should be taken into account when considering therapeutic strategies.
Collapse
Affiliation(s)
- J I Diaz-Hernandez
- Unidad de Investigación, Hospital Universitario de Salamanca-Instituto de Estudios Ciencias de la Salud de Castilla y León, Salamanca, Spain
| | | | | | | |
Collapse
|
46
|
Abstract
The redox environment within neural cells is dependent on a series of redox couples. The glutathione disulfide/ glutathione (GSSG/GSH) redox pair forms the major redox couple in cells and as such plays a critical role in regulating redox-dependent cellular functions. Not only does GSH act as an antioxidant but it also can modulate the activity of a variety of different proteins via S-glutathionylation of cysteine sulfhydryl groups. The thioredoxin system also makes a significant contribution to the redox environment by reducing inter- and intrachain protein disulfide bonds as well as maintaining the activity of important antioxidant enzymes such as peroxiredoxins and methionine sulfoxide reductases. The redox environment affects the activity and function of a number of different protein phosphatases, protein kinases, and transcription factors. The sum of these effects will determine how changes in the redox environment alter overall cellular function, thereby playing a fundamental role in regulating neural cell fate and physiology.
Collapse
Affiliation(s)
- Pamela Maher
- The Salk Institute for Biological Studies, La Jolla, California 92037, USA.
| |
Collapse
|
47
|
Murín R, Verleysdonk S, Raeymaekers L, Kaplán P, Lehotský J. Distribution of secretory pathway Ca2+ ATPase (SPCA1) in neuronal and glial cell cultures. Cell Mol Neurobiol 2006; 26:1355-65. [PMID: 16758324 PMCID: PMC11520639 DOI: 10.1007/s10571-006-9042-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2005] [Accepted: 02/27/2006] [Indexed: 10/24/2022]
Abstract
1. Secretory pathway Ca(2+) ATPase type 1 (SPCA1) is a newly recognized Ca(2+)/Mn(2+)-transporting pump localized in membranes of the Golgi apparatus. 2. The expression level of SPCA1 in brain tissue is relatively high in comparison with other tissues. 3. With the aim to determine the expression of SPCA1 within the different types of neural cells, we investigated the distribution of SPCA1 in neuronal, astroglial, oligodendroglial, ependymal, and microglial cell cultures derived from rat brains. 4. Western Blot analysis with rabbit anti-SPCA1 antibodies revealed the presence of SPCA1 in homogenates derived from neuronal, astroglial, ependymal, and oligodendroglial, but not from microglial cells. 5. Cell cultures that gave rise to positive signal in the immunoblot analysis were also examined immunocytochemically. 6. Immunocytochemical double-labeling experiments with anti-SPCA1 serum in combination with antibodies against cell-type specific proteins showed a localization of the SPCA1signal within cells stained positively also for GFAP, alpha-tubulin or MBP. 7. These results definitely established the expression of SPCA1 in astroglial, ependymal, and oligodendroglial cells. 8. In addition, the evaluation of neuronal cultures for the presence of SPCA1 revealed an SPCA1-specific immunofluorescence signal in cells identified as neurons.
Collapse
Affiliation(s)
- Radovan Murín
- Interfaculty Institute of Biochemistry, University of Tuebingen, Hoppe-Seyler-Str. 4, D-72076 Tuebingen, Germany
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University, Mala Hora 4, SK-03601 Martin, Slovakia
| | - Stephan Verleysdonk
- Interfaculty Institute of Biochemistry, University of Tuebingen, Hoppe-Seyler-Str. 4, D-72076 Tuebingen, Germany
| | - Luc Raeymaekers
- Department of Physiology, Catholic University Leuven, Gasthuisberg, B-3000 Leuven, Belgium
| | - Peter Kaplán
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University, Mala Hora 4, SK-03601 Martin, Slovakia
| | - Ján Lehotský
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University, Mala Hora 4, SK-03601 Martin, Slovakia
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University, Mala Hora 4, SK-03601 Martin, Slovakia
| |
Collapse
|
48
|
Abstract
Multiple sclerosis (MS) is an immune-mediated disease, with inflammation and neurodegeneration contributing to neuronal demyelination and axonal injury. Current therapies for MS are directed toward modulation of the immune response; however, there is increasing evidence that oxidative stress is an important component in the pathogenesis of MS. The inflammatory environment in demyelinating lesions is conducive to the generation of reactive oxygen species. When these species are generated in MS and animal models of MS, products such as peroxynitrite and superoxide are formed that are highly toxic to cells. There are several examples of potential beneficial effects from various antioxidants in animal models of MS, but the efficacy may vary between different agents and, in some instances, may yield deleterious effects. Despite these promising results in animal models, there is limited and conflicting evidence of potential therapeutic effects of antioxidants such as vitamins C and E in treating MS. However, clinical trials in MS patients with more potent antioxidants, identified in animal studies, have been initiated.
Collapse
Affiliation(s)
- Noel G Carlson
- GRECC, VASLCHCS, Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, USA
| | | |
Collapse
|
49
|
Murín R, Verleysdonk S, Rapp M, Hamprecht B. Immunocytochemical localization of 3-methylcrotonyl-CoA carboxylase in cultured ependymal, microglial and oligodendroglial cells. J Neurochem 2006; 97:1393-402. [PMID: 16696850 DOI: 10.1111/j.1471-4159.2006.03819.x] [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: 11/28/2022]
Abstract
To evaluate the ability of ependymal, microglial and oligodendroglial cells to degrade leucine, the presence of 3-methylcrotonyl-CoA carboxylase (MCC) was investigated in cultures of these cells. MCC is a biotin-containing heterodimeric enzyme that is specific for the irreversible part of the leucine catabolic pathway. It has been reported previously that in cell culture MCC is expressed in astrocytes and a subpopulation of neurones. In the present study ependymal, microglial and oligodendroglial cell cultures, derived from the brains of newborn rats, were examined for the expression of MCC by RT-PCR, western blotting and immunocytochemistry. The results of RT-PCR and western blotting showed the presence of mRNA as well as protein of both subunits of MCC in ependymal, microglial and oligodendroglial cell cultures. Immunocytochemical investigation of the cellular and subcellular distribution of MCC demonstrated a mitochondrial location of MCC in all neuroglial cell types investigated. The ubiquitous expression of MCC in glial cells demonstrates the ability of the cells to engage in the catabolism of leucine transported into the brain, mainly for the generation of energy.
Collapse
Affiliation(s)
- Radovan Murín
- Interfaculty Institute for Biochemistry, University of Tuebingen, Tuebingen, Germany
| | | | | | | |
Collapse
|
50
|
Dallas S, Miller DS, Bendayan R. Multidrug resistance-associated proteins: expression and function in the central nervous system. Pharmacol Rev 2006; 58:140-61. [PMID: 16714484 DOI: 10.1124/pr.58.2.3] [Citation(s) in RCA: 214] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Drug delivery to the brain is highly restricted, since compounds must cross a series of structural and metabolic barriers to reach their final destination, often a cellular compartment such as neurons, microglia, or astrocytes. The primary barriers to the central nervous system are the blood-brain and blood-cerebrospinal fluid barriers. Through structural modifications, including the presence of tight junctions that greatly limit paracellular transport, the cells that make up these barriers restrict diffusion of many pharmaceutically active compounds. In addition, the cells that comprise the blood-brain and blood-cerebrospinal fluid barriers express multiple ATP-dependent, membrane-bound, efflux transporters, such as members of the multidrug resistance-associated protein (MRP) family, which contribute to lowered drug accumulation. A relatively new concept in brain drug distribution just beginning to be explored is the possibility that cellular components of the brain parenchyma could act as a "second" barrier to brain permeation of pharmacological agents via expression of many of the same transporters. Indeed, efflux transporters expressed in brain parenchyma may facilitate the overall export of xenobiotics from the central nervous system, essentially handing them off to the barrier tissues. We propose that these primary and secondary barriers work in tandem to limit overall accumulation and distribution of xenobiotics in the central nervous system. The present review summarizes recent knowledge in this area and emphasizes the clinical significance of MRP transporter expression in a variety of neurological disorders.
Collapse
Affiliation(s)
- Shannon Dallas
- Laboratory of Pharmacology and Chemistry, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | | | | |
Collapse
|