1
|
Liao Y, Wen L, Zheng R, Shen Y, Ha TA, Lin M, Cheng R, Gao Y, Shang P. Novel Perspectives Focused on the Relationship Between Herpesvirus Encephalitis and Anti-GFAP-Antibody-Positive Astrocytopathy. Mol Neurobiol 2024:10.1007/s12035-024-04660-0. [PMID: 39731639 DOI: 10.1007/s12035-024-04660-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/03/2024] [Accepted: 12/02/2024] [Indexed: 12/30/2024]
Abstract
Virus encephalitis (VE), recognized as one of the common kinds of central nervous system (CNS) diseases after virus infection, has a surprising correlation with autoimmune encephalitis (AE) when autoimmune antibodies emerge in cerebrospinal fluid (CSF) or serum. Herpes simplex virus and Epstein-Barr virus are the most critical agents worldwide. By molecular mimicry, herpes viruses can invade the brain directly or indirectly. As a type-III intermediate filament, glial fibrillary acidic protein (GFAP) can be seen in both the central and peripheral nervous system and is regarded as a marker of astrocyte activation. Autoimmune glial fibrillary acidic protein astrocytopathy (GFAP-A), an autoimmune inflammatory CNS disorder with unearthed pathogenic mechanism yet, is correlated with CD8 + T cells and AQP4 astrocytopathy in TNF signaling. It brings a new concept of VE and GFAP coexisting, which has been documented in several case reports. Considering the infectious role of herpes viruses in CNS, EBV contributes to GFAP-IgG significantly and may result in GFAP-A. Coincidently, the existence of GFAP-IgG in patients with infection of herpes viruses has been documented as well. There exist multiple diagnoses of VE, ranging from traditional diagnostic criteria, such as CSF examination and electronic techniques, to a novel approach, according to case reports, the detection of GFAP-lgG. In terms of treatment, except for (IVIG), the explorations for new curative targets and optimal diagnostic time are of great necessity. In conclusion, emphasis given to the CNS autoimmune effect brought by the virus infection is highly worthy.
Collapse
Affiliation(s)
- Yuqiao Liao
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Linxin Wen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruoyi Zheng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yinan Shen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Teng-Ai Ha
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingkai Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Ruogu Cheng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Gao
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pei Shang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN, USA.
| |
Collapse
|
2
|
Costanza A, Amerio A, Aguglia A, Rossi M, Parise A, Magnani L, Serafini G, Amore M, Martins D, Nguyen KD. Reactive Astrocytosis-A Potential Contributor to Increased Suicide in Long COVID-19 Patients? Brain Sci 2024; 14:973. [PMID: 39451987 PMCID: PMC11505806 DOI: 10.3390/brainsci14100973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/19/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Long COVID-19 is an emerging chronic illness of significant public health concern due to a myriad of neuropsychiatric sequelae, including increased suicidal ideation (SI) and behavior (SB). METHODS This review provides a concise synthesis of clinical evidence that points toward the dysfunction of astrocytes, the most abundant glial cell type in the central nervous system, as a potential shared pathology between SI/SB and COVID-19. RESULTS Depression, a suicide risk factor, and SI/SB were both associated with reduced frequencies of various astrocyte subsets and complex proteomic/transcriptional changes of astrocyte-related markers in a brain-region-specific manner. Astrocyte-related circulating markers were increased in depressed subjects and, to a less consistent extent, in COVID-19 patients. Furthermore, reactive astrocytosis was observed in subjects with SI/SB and those with COVID-19. CONCLUSIONS Astrocyte dysfunctions occurred in depression, SI/SB, and COVID-19. Reactive-astrocyte-mediated loss of the blood-brain barrier (BBB) integrity and subsequent neuroinflammation-a factor previously linked to SI/SB development-might contribute to increased suicide in individuals with long COVID-19. As such, the formulation of new therapeutic strategies to restore astrocyte homeostasis, enhance BBB integrity, and mitigate neuroinflammation may reduce SI/SB-associated neuropsychiatric manifestations among long COVID-19 patients.
Collapse
Affiliation(s)
- Alessandra Costanza
- Department of Psychiatry, Faculty of Medicine, University of Geneva (UNIGE), 24 Rue du Général-Dufour, 1211 Geneva, Switzerland
- Department of Psychiatry, Faculty of Biomedical Sciences, University of Italian Switzerland (USI), Via Giuseppe Buffi 13, 6900 Lugano, Switzerland
- Department of Psychiatry, Adult Psychiatry Service, University Hospitals of Geneva (HUG), Rue Gabrielle-Perret-Gentil 4, 1205 Geneva, Switzerland
- “Nel Chiostro”, Medical and Study Center, Via Camillo Leone 29, 13100 Vercelli, Italy
| | - Andrea Amerio
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Via Balbi, 5, 16132 Genoa, Italy; (A.A.); (A.A.); (G.S.); (M.A.)
- IRCCS Polyclinic Hospital San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Andrea Aguglia
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Via Balbi, 5, 16132 Genoa, Italy; (A.A.); (A.A.); (G.S.); (M.A.)
- IRCCS Polyclinic Hospital San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Martina Rossi
- “Nel Chiostro”, Medical and Study Center, Via Camillo Leone 29, 13100 Vercelli, Italy
| | - Alberto Parise
- Geriatric-Rehabilitation Department, University Hospital of Parma, 43126 Parma, Italy;
| | - Luca Magnani
- Department of Psychiatry, San Maurizio Hospital of Bolzano, Via Lorenz Böhler, 5, 39100 Bolzano, Italy;
| | - Gianluca Serafini
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Via Balbi, 5, 16132 Genoa, Italy; (A.A.); (A.A.); (G.S.); (M.A.)
- IRCCS Polyclinic Hospital San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Mario Amore
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Via Balbi, 5, 16132 Genoa, Italy; (A.A.); (A.A.); (G.S.); (M.A.)
- IRCCS Polyclinic Hospital San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN)—King’s College London (KCL), Strand Campus, London WC2R 2LS, UK;
- NIHR Maudesley BRC, 16 De Crespigny Park, SE5 8AF South London and Maudesley NHS Trust, Denmark Hill, London SE5 8AZ, UK
| | - Khoa D. Nguyen
- Program in Immunology, Stanford University, 450 Jane Stanford Way, Stanford, CA 94305, USA;
- Department of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
3
|
Blaber AP, Sadeghian F, Naz Divsalar D, Scarisbrick IA. Elevated biomarkers of neural injury in older adults following head-down bed rest: links to cardio-postural deconditioning with spaceflight and aging. Front Hum Neurosci 2023; 17:1208273. [PMID: 37822710 PMCID: PMC10562592 DOI: 10.3389/fnhum.2023.1208273] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/21/2023] [Accepted: 08/29/2023] [Indexed: 10/13/2023] Open
Abstract
Introduction Prolonged physical inactivity with bed rest or spaceflight is associated with cardiovascular and neuromuscular deconditioning; however, its impact on neural integrity of cardio-postural reflexes and possible mitigation with exercise has not been examined. We assessed the association between the physiological deconditioning of bed rest immobilization with neural injury markers and the effects of 60-75 min of daily exercise. Methods Data were collected as part of a randomized clinical trial (clinicaltrials.gov identifier: NCT04964999) at the McGill University Medical Centre. Twenty-two 55- to 65-year-old healthy volunteers gave informed consent and took part. Within sex, participants were randomly assigned to exercise (60- to 75-min daily) or control (inactive) groups and spent 14 days in continuous 6° head-down tilt. Neural injury [neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), total tau (t-Tau), myelin basic protein (MBP), brain-derived neurotrophic factor (BDNF), ubiquitin carboxy-terminal hydrolase L1 (UCH-L1)], as well as interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), and insulin-like growth factor 1 (IGF-1) biomarkers were measured before, during, and after bed rest. The false discovery rate with Huber M-estimation was used to correlate changes in biomarkers with cardiovascular and muscular function changes over bed rest. Results Bed rest elevated NfL, GFAP, TNF-α, and IL-6 in all participants and reduced IGF-1 in females only. With standing, changes in heart rate, blood pressure, and lower limb muscle motoneuron activity correlated with changes in TNF-α and BDNF. Baroreflex control, leg muscle maximal voluntary contraction, and postural sway are correlated with GFAP and NfL. Exercise participants had fewer interactions than control participants, but significant correlations still existed, with both groups exhibiting similar reductions in orthostatic tolerance. Discussion An hour of daily exercise in older persons otherwise immobilized for 2 weeks did not abate bed rest-induced increases in serum signatures of neural injury or pro-inflammatory markers. Exercise reduced the number of physiological interactions of biomarkers, but significant cardio-postural correlations remained with no protection against post-bed rest orthostatic intolerance. The identification of associations of inflammatory and neural injury biomarkers with changes in cardio-postural physiology and exercise points to biotherapeutic opportunities and improved exercise interventions for astronauts and individuals in bed rest. Clinical trial registration https://www.clinicaltrials.gov/search?cond=NCT04964999, identifier: NCT04964999.
Collapse
Affiliation(s)
- Andrew P. Blaber
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Farshid Sadeghian
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Donya Naz Divsalar
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Isobel A. Scarisbrick
- Department of Physical Medicine and Rehabilitation, Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
4
|
Cellular Model of Malignant Transformation of Primary Human Astrocytes Induced by Deadhesion/Readhesion Cycles. Int J Mol Sci 2022; 23:ijms23094471. [PMID: 35562862 PMCID: PMC9103552 DOI: 10.3390/ijms23094471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/01/2021] [Revised: 04/03/2022] [Accepted: 04/12/2022] [Indexed: 01/07/2023] Open
Abstract
Astrocytoma is the most common and aggressive tumor of the central nervous system. Genetic and environmental factors, bacterial infection, and several other factors are known to be involved in gliomagenesis, although the complete underlying molecular mechanism is not fully understood. Tumorigenesis is a multistep process involving initiation, promotion, and progression. We present a human model of malignant astrocyte transformation established by subjecting primary astrocytes from healthy adults to four sequential cycles of forced anchorage impediment (deadhesion). After limiting dilution of the surviving cells obtained after the fourth deadhesion/readhesion cycle, three clones were randomly selected, and exhibited malignant characteristics, including increased proliferation rate and capacity for colony formation, migration, and anchorage-independent growth in soft agar. Functional assay results for these clonal cells, including response to temozolomide, were comparable to U87MG—a human glioblastoma-derived cell lineage—reinforcing malignant cell transformation. RNA-Seq analysis by next-generation sequencing of the transformed clones relative to the primary astrocytes revealed upregulation of genes involved in the PI3K/AKT and Wnt/β-catenin signaling pathways, in addition to upregulation of genes related to epithelial–mesenchymal transition, and downregulation of genes related to aerobic respiration. These findings, at a molecular level, corroborate the change in cell behavior towards mesenchymal-like cell dedifferentiation. This linear progressive model of malignant human astrocyte transformation is unique in that neither genetic manipulation nor treatment with carcinogens are used, representing a promising tool for testing combined therapeutic strategies for glioblastoma patients, and furthering knowledge of astrocytoma transformation and progression.
Collapse
|
5
|
Schultz B, Taday J, Menezes L, Cigerce A, Leite MC, Gonçalves CA. Calpain-Mediated Alterations in Astrocytes Before and During Amyloid Chaos in Alzheimer's Disease. J Alzheimers Dis 2021; 84:1415-1430. [PMID: 34719501 DOI: 10.3233/jad-215182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022]
Abstract
One of the changes found in the brain in Alzheimer's disease (AD) is increased calpain, derived from calcium dysregulation, oxidative stress, and/or neuroinflammation, which are all assumed to be basic pillars in neurodegenerative diseases. The role of calpain in synaptic plasticity, neuronal death, and AD has been discussed in some reviews. However, astrocytic calpain changes sometimes appear to be secondary and consequent to neuronal damage in AD. Herein, we explore the possibility of calpain-mediated astroglial reactivity in AD, both preceding and during the amyloid phase. We discuss the types of brain calpains but focus the review on calpains 1 and 2 and some important targets in astrocytes. We address the signaling involved in controlling calpain expression, mainly involving p38/mitogen-activated protein kinase and calcineurin, as well as how calpain regulates the expression of proteins involved in astroglial reactivity through calcineurin and cyclin-dependent kinase 5. Throughout the text, we have tried to provide evidence of the connection between the alterations caused by calpain and the metabolic changes associated with AD. In addition, we discuss the possibility that calpain mediates amyloid-β clearance in astrocytes, as opposed to amyloid-β accumulation in neurons.
Collapse
Affiliation(s)
- Bruna Schultz
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jéssica Taday
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Leonardo Menezes
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Anderson Cigerce
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marina C Leite
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Carlos-Alberto Gonçalves
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
6
|
Ribeiro ACAF, Batista TH, Ferrari MS, Giusti-Paiva A, Vilela FC. The accentuation in post-traumatic stress disorder-like symptoms induced by diabetes in rats is not associated with a further increase in astrocyte activation in the hippocampus. Neurosci Lett 2021; 762:136174. [PMID: 34400287 DOI: 10.1016/j.neulet.2021.136174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/06/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 10/20/2022]
Abstract
Individuals with post-traumatic stress disorder (PTSD) show increased rates of several serious metabolic diseases. However, little is known about pre-existing metabolic diseases and the development of PTSD. Therefore, we aimed to evaluate the course of post-traumatic stress disorder (PTSD) development in rats with preexisting diabetes. In addition, we quantified glial fibrillary acidic protein (GFAP) in the hippocampus of the experimental animals. For this, we used male Wistar rats and divided them into two groups: saline and alloxan (150 mg/Kg, i.p.). The animals were weighed, and plasma glucose was measured after 48 h of diabetes induction by alloxan. The animals were either exposed to inescapable footshocks or not, followed by social isolation. After 14 days, the animals were re-exposed to the box, and the freezing time was evaluated for 10 min. Over the following days, the animals were tested on the open field, social interaction and forced swimming tests. In another group of animals, elevated plus maze and object recognition tests were performed. Our results demonstrated that animals with diabetes had more pronounced PTSD-like symptoms as a reduction in social interaction, an increase in immobility time in forced swimming, a reduction in permanence in the open arms of the elevated plus maze, and a deficit in the object recognition index more accentuated. However, this did not reflect astrocyte activation in the hippocampus. In conclusion, diabetes accentuates post-traumatic stress disorder-like symptoms but not astrocyte activation in the hippocampus.
Collapse
Affiliation(s)
- Ana Cláudia A F Ribeiro
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
| | - Tatiane H Batista
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
| | - Mariela S Ferrari
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
| | - Alexandre Giusti-Paiva
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
| | - Fabiana C Vilela
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil.
| |
Collapse
|
7
|
Borodinova AA, Balaban PM, Bezprozvanny IB, Salmina AB, Vlasova OL. Genetic Constructs for the Control of Astrocytes' Activity. Cells 2021; 10:cells10071600. [PMID: 34202359 PMCID: PMC8306323 DOI: 10.3390/cells10071600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/31/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022] Open
Abstract
In the current review, we aim to discuss the principles and the perspectives of using the genetic constructs based on AAV vectors to regulate astrocytes’ activity. Practical applications of optogenetic approaches utilizing different genetically encoded opsins to control astroglia activity were evaluated. The diversity of astrocytic cell-types complicates the rational design of an ideal viral vector for particular experimental goals. Therefore, efficient and sufficient targeting of astrocytes is a multiparametric process that requires a combination of specific AAV serotypes naturally predisposed to transduce astroglia with astrocyte-specific promoters in the AAV cassette. Inadequate combinations may result in off-target neuronal transduction to different degrees. Potentially, these constraints may be bypassed with the latest strategies of generating novel synthetic AAV serotypes with specified properties by rational engineering of AAV capsids or using directed evolution approach by searching within a more specific promoter or its replacement with the unique enhancer sequences characterized using modern molecular techniques (ChIP-seq, scATAC-seq, snATAC-seq) to drive the selective transgene expression in the target population of cells or desired brain regions. Realizing these strategies to restrict expression and to efficiently target astrocytic populations in specific brain regions or across the brain has great potential to enable future studies.
Collapse
Affiliation(s)
- Anastasia A. Borodinova
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia;
| | - Pavel M. Balaban
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia;
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (I.B.B.); (A.B.S.); (O.L.V.)
- Correspondence:
| | - Ilya B. Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (I.B.B.); (A.B.S.); (O.L.V.)
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Alla B. Salmina
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (I.B.B.); (A.B.S.); (O.L.V.)
- Research Institute of Molecular Medicine and Pathobiochemistry, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
- Research Center of Neurology, 125367 Moscow, Russia
| | - Olga L. Vlasova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (I.B.B.); (A.B.S.); (O.L.V.)
| |
Collapse
|
8
|
Hsu SS, Jan CR, Liang WZ. The investigation of the pyrethroid insecticide lambda-cyhalothrin (LCT)-affected Ca 2+ homeostasis and -activated Ca 2+-associated mitochondrial apoptotic pathway in normal human astrocytes: The evaluation of protective effects of BAPTA-AM (a selective Ca 2+ chelator). Neurotoxicology 2018; 69:97-107. [PMID: 30292652 DOI: 10.1016/j.neuro.2018.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/09/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 12/22/2022]
Abstract
Exposure to insecticides has been found to have deleterious effects on human health. Lambda-cyhalothrin (LCT), a mixture of isomers of cyhalothrin, is a pyrethroid insecticide routinely used in pest control programs. LCT was reported to cause neurotoxic effects in various models. However, the mechanism of underlying effect of LCT on cytotoxicity in normal human brain cells is still elusive. This study examined whether LCT affected Ca2+ homeostasis and Ca2+-related physiology in Gibco® Human Astrocytes (GHA cells), and explored whether BAPTA-AM (1,2-bis(2-aminophenoxy)ethane-N,N,N'N'-tetraacetic acid), a selective Ca2+ chelator, has protective effects on LCT-treated GHA cells. The data show that LCT (10-15 μM) concentration-dependently induced cytotoxicity in both GHA cells and DI TNC1 normal rat astrocytes but only induced intracellular Ca2+ concentration ([Ca2+]i) rises in GHA cells. In terms of Ca2+ signaling in GHA cells, LCT-induced [Ca2+]i rises were reduced by removing extracellular Ca2+ and were inhibited by store-operated Ca2+ channel modulators (2-APB, econazole or SKF96365). In Ca2+-free medium, pretreatment with the endoplasmic reticulum Ca2+ pump inhibitor thapsigargin abolished LCT-induced [Ca2+]i rises. Conversely, incubation with LCT abolished thapsigargin-induced [Ca2+]i rises. Regarding cytotoxicity, LCT evoked apoptosis by regulating apoptotic protein expressions (Bax, BCl-2, cleaved caspase-9/-3). This apoptotic response was significantly inhibited by prechelating cytosolic Ca2+ with BAPTA-AM. Together, in GHA cells, LCT induced [Ca2+]i rises by inducing Ca2+ entry via store-operated Ca2+ channels and Ca2+ release from the endoplasmic reticulum. Moreover, BAPTA-AM has a protective effect on inhibiting LCT-activated mitochondrial apoptotic pathway. This study provided new insights into the molecular protective mechanism of LCT-induced cytotoxicity in normal human astrocytes.
Collapse
Affiliation(s)
- Shu-Shong Hsu
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung, 81362, Taiwan; Department of Surgery, National Defense Medical Center, Taipei, 11490, Taiwan; Department of Nursing, Meiho University, Pingtung, 91202, Taiwan
| | - Chung-Ren Jan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 81362, Taiwan
| | - Wei-Zhe Liang
- Department of Pharmacy, Tajen University, Pingtung, 90741, Taiwan; Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 81362, Taiwan.
| |
Collapse
|
9
|
Characterization of 3D embryonic C57BL/6 and A/J mouse midbrain micromass in vitro culture systems for developmental neurotoxicity testing. Toxicol In Vitro 2018; 48:33-44. [DOI: 10.1016/j.tiv.2017.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/20/2017] [Accepted: 12/13/2017] [Indexed: 01/19/2023]
|
10
|
Chandrasekaran A, Avci HX, Leist M, Kobolák J, Dinnyés A. Astrocyte Differentiation of Human Pluripotent Stem Cells: New Tools for Neurological Disorder Research. Front Cell Neurosci 2016; 10:215. [PMID: 27725795 PMCID: PMC5035736 DOI: 10.3389/fncel.2016.00215] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/29/2016] [Accepted: 08/30/2016] [Indexed: 12/22/2022] Open
Abstract
Astrocytes have a central role in brain development and function, and so have gained increasing attention over the past two decades. Consequently, our knowledge about their origin, differentiation and function has increased significantly, with new research showing that astrocytes cultured alone or co-cultured with neurons have the potential to improve our understanding of various central nervous system diseases, such as amyotrophic lateral sclerosis, Alzheimer’s disease, or Alexander disease. The generation of astrocytes derived from pluripotent stem cells (PSCs) opens up a new area for studying neurologic diseases in vitro; these models could be exploited to identify and validate potential drugs by detecting adverse effects in the early stages of drug development. However, as it is now known that a range of astrocyte populations exist in the brain, it will be important in vitro to develop standardized protocols for the in vitro generation of astrocyte subsets with defined maturity status and phenotypic properties. This will then open new possibilities for co-cultures with neurons and the generation of neural organoids for research purposes. The aim of this review article is to compare and summarize the currently available protocols and their strategies to generate human astrocytes from PSCs. Furthermore, we discuss the potential role of human-induced PSCs derived astrocytes in disease modeling.
Collapse
Affiliation(s)
| | - Hasan X Avci
- BioTalentum LtdGödöllő, Hungary; Department of Medical Chemistry, University of SzegedSzeged, Hungary
| | - Marcel Leist
- Dorenkamp-Zbinden Chair, Faculty of Mathematics and Sciences, University of Konstanz Konstanz, Germany
| | | | - Andras Dinnyés
- BioTalentum LtdGödöllő, Hungary; Molecular Animal Biotechnology Laboratory, Szent Istvan UniversityGödöllő, Hungary
| |
Collapse
|
11
|
|
12
|
Yang Z, Wang KKW. Glial fibrillary acidic protein: from intermediate filament assembly and gliosis to neurobiomarker. Trends Neurosci 2015; 38:364-74. [PMID: 25975510 PMCID: PMC4559283 DOI: 10.1016/j.tins.2015.04.003] [Citation(s) in RCA: 600] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/15/2015] [Revised: 04/03/2015] [Accepted: 04/07/2015] [Indexed: 12/20/2022]
Abstract
Glial fibrillary acidic protein (GFAP) is an intermediate filament (IF) III protein uniquely found in astrocytes in the central nervous system (CNS), non-myelinating Schwann cells in the peripheral nervous system (PNS), and enteric glial cells. GFAP mRNA expression is regulated by several nuclear-receptor hormones, growth factors, and lipopolysaccharides (LPSs). GFAP is also subject to numerous post-translational modifications (PTMs), while GFAP mutations result in protein deposits known as Rosenthal fibers in Alexander disease. GFAP gene activation and protein induction appear to play a critical role in astroglial cell activation (astrogliosis) following CNS injuries and neurodegeneration. Emerging evidence also suggests that, following traumatic brain and spinal cord injuries and stroke, GFAP and its breakdown products are rapidly released into biofluids, making them strong candidate biomarkers for such neurological disorders.
Collapse
Affiliation(s)
- Zhihui Yang
- Program for Neurotrauma, Neuroproteomics, and Biomarkers Research, Departments of Psychiatry and Neuroscience, McKnight Brain Institute, L4-100, University of Florida, 1149 South Newell Drive, Gainesville, FL 32611, USA
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics, and Biomarkers Research, Departments of Psychiatry and Neuroscience, McKnight Brain Institute, L4-100, University of Florida, 1149 South Newell Drive, Gainesville, FL 32611, USA.
| |
Collapse
|
13
|
Yang X, Zhu F, Zhang X, Gao Z, Cao Y. Ipsilateral versus bilateral limb-training in promoting the proliferation and differentiation of endogenous neural stem cells following cerebral infarction in rats. Neural Regen Res 2014; 7:2698-704. [PMID: 25337116 PMCID: PMC4200738 DOI: 10.3969/j.issn.1673-5374.2012.34.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/20/2012] [Accepted: 11/02/2012] [Indexed: 12/11/2022] Open
Abstract
We investigated the effects of ipsilateral versus bilateral limb-training on promotion of endogenous neural stem cells in the peripheral infarct zone and the corresponding cerebral region in the unaffected hemisphere of rats with cerebral infarction. Middle cerebral artery occlusion was induced in Wistar rats. The rat forelimb on the unaffected side was either wrapped up with tape to force the use of the paretic forelimb in rats or not braked to allow bilateral forelimbs to participate in training. Daily training consisted of mesh drum training, balance beam training, and stick rolling training for a total of 40 minutes, once per day. Control rats received no training. At 14 days after functional training, rats receiving bilateral limb-training exhibited milder neurological impairment than that in the ipsilateral limb-training group or the control group. The number of nestin/glial fibrillary acidic protein-positive and nestin/microtubule-associated protein 2-positive cells in the peripheral infarct zone and in the corresponding cerebral region in the unaffected hemisphere was significantly higher in rats receiving bilateral limb-training than in rats receiving ipsilateral limb-training. These data suggest that bilateral limb-training can promote the proliferation and differentiation of endogenous neural stem cells in the bilateral hemispheres after cerebral infarction and accelerate the recovery of neurologic function. In addition, bilateral limb-training produces better therapeutic effects than ipsilateral limb-training.
Collapse
Affiliation(s)
- Xiyao Yang
- Department of Neurology, the First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Feng Zhu
- Department of Neurology, Shenyang Brain Hospital, Shenyang 110001, Liaoning Province, China
| | - Xiaomei Zhang
- The First People's Hospital of Dandong, Dandong 118000, Liaoning Province, China
| | - Zhuo Gao
- Department of Neurology, Shenyang Brain Hospital, Shenyang 110001, Liaoning Province, China
| | - Yunpeng Cao
- Department of Neurology, the First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
14
|
Schreiner AE, Durry S, Aida T, Stock MC, Rüther U, Tanaka K, Rose CR, Kafitz KW. Laminar and subcellular heterogeneity of GLAST and GLT-1 immunoreactivity in the developing postnatal mouse hippocampus. J Comp Neurol 2014; 522:204-24. [PMID: 23939750 DOI: 10.1002/cne.23450] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/11/2012] [Revised: 05/23/2013] [Accepted: 08/02/2013] [Indexed: 11/06/2022]
Abstract
Astrocytes express two sodium-coupled transporters, glutamate-aspartate transporter (GLAST) and glutamate transporter-1 (GLT-1), which are essential for the maintenance of low extracellular glutamate levels. We performed a comparative analysis of the laminar and subcellular expression profile of GLAST and GLT-1 in the developing postnatal mouse hippocampus by using immunohistochemistry and western blotting and employing high-resolution fluorescence microscopy. Astrocytes were identified by costaining with glial fibrillary acidic protein (GFAP) or S100β. In CA1, the density of GFAP-positive cells and GFAP expression rose during the first 2 weeks after birth, paralleled by a steady increase in GLAST immunoreactivity and protein content. Upregulation of GLT-1 was completed only at postnatal days (P) P20-25 and was thus delayed by about 10 days. GLAST staining was highest along the stratum pyramidale and was especially prominent in astrocytes at P3-5. GLAST immunoreactivity indicated no preferential localization to a specific cellular compartment. GLT-1 exhibited a laminar expression pattern from P10-15 on, with the highest immunoreactivity in the stratum lacunosum-moleculare. At the cellular level, GLT-1 immunoreactivity did not entirely cover astrocyte somata and exhibited clusters at processes. In neonatal and juvenile animals, discrete clusters of GLT-1 were also detected at perivascular endfeet. From these results, we conclude there is a remarkable subcellular heterogeneity of GLAST and GLT-1 expression in the developing hippocampus. The clustering of GLT-1 at astrocyte endfeet indicates that it might serve a specialized functional role at the blood-brain barrier during formation of the hippocampal network.
Collapse
Affiliation(s)
- Alexandra E Schreiner
- Institute of Neurobiology, Heinrich Heine University Duesseldorf, 40225, Duesseldorf, Germany
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Desmet AS, Cirillo C, Vanden Berghe P. Distinct subcellular localization of the neuronal marker HuC/D reveals hypoxia-induced damage in enteric neurons. Neurogastroenterol Motil 2014; 26:1131-43. [PMID: 24861242 DOI: 10.1111/nmo.12371] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/31/2014] [Accepted: 04/29/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Correct neuronal identification is essential to study neurons in health and disease. Although commonly used as pan-neuronal marker, HuC/D's expression pattern varies substantially between healthy and (patho)physiological conditions. This heterogenic labeling has received very little attention. We sought to investigate the subcellular HuC/D localization in enteric neurons in different conditions. METHODS The localization of neuronal RNA-binding proteins HuC/D was investigated by immunohistochemistry in the mouse myenteric plexus using different toxins and caustic agents. Preparations were also stained with Sox10 and glial fibrillary acidic protein (GFAP) antibodies to assess enteric glial cell appearance. KEY RESULTS Mechanically induced tissue damage, interference with the respiratory chain and oxygen (O2 ) deprivation increased nuclear HuC/D immunoreactivity. This effect was paralleled by a distortion of the GFAP-labeled glial network along with a loss of Sox10 expression and coincided with the activation of a non-apoptotic genetic program. Chemically induced damage and specific noxious stimuli did not induce a change in HuC/D immunoreactivity, supporting the specific nature of the nuclear HuC/D localization. CONCLUSIONS & INFERENCES HuC/D is not merely a pan-neuronal marker but its subcellular localization also reflects the condition of a neuron at the time of fixation. The functional meaning of this change in HuC/D localization is not entirely clear, but disturbance in O2 supply in combination with the support of enteric glial cells seems to play a crucial role. The molecular consequence of changes in HuC/D expression needs to be further investigated.
Collapse
Affiliation(s)
- A-S Desmet
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for GastroIntestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | | | | |
Collapse
|
16
|
Phosphoproteomics and bioinformatics analyses of spinal cord proteins in rats with morphine tolerance. PLoS One 2014; 9:e83817. [PMID: 24392096 PMCID: PMC3879267 DOI: 10.1371/journal.pone.0083817] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/24/2013] [Accepted: 11/08/2013] [Indexed: 12/19/2022] Open
Abstract
Introduction Morphine is the most effective pain-relieving drug, but it can cause unwanted side effects. Direct neuraxial administration of morphine to spinal cord not only can provide effective, reliable pain relief but also can prevent the development of supraspinal side effects. However, repeated neuraxial administration of morphine may still lead to morphine tolerance. Methods To better understand the mechanism that causes morphine tolerance, we induced tolerance in rats at the spinal cord level by giving them twice-daily injections of morphine (20 µg/10 µL) for 4 days. We confirmed tolerance by measuring paw withdrawal latencies and maximal possible analgesic effect of morphine on day 5. We then carried out phosphoproteomic analysis to investigate the global phosphorylation of spinal proteins associated with morphine tolerance. Finally, pull-down assays were used to identify phosphorylated types and sites of 14-3-3 proteins, and bioinformatics was applied to predict biological networks impacted by the morphine-regulated proteins. Results Our proteomics data showed that repeated morphine treatment altered phosphorylation of 10 proteins in the spinal cord. Pull-down assays identified 2 serine/threonine phosphorylated sites in 14-3-3 proteins. Bioinformatics further revealed that morphine impacted on cytoskeletal reorganization, neuroplasticity, protein folding and modulation, signal transduction and biomolecular metabolism. Conclusions Repeated morphine administration may affect multiple biological networks by altering protein phosphorylation. These data may provide insight into the mechanism that underlies the development of morphine tolerance.
Collapse
|
17
|
Abstract
Motor unit (MU) enlargement by sprouting is an important compensatory mechanism for loss of functional MUs during normal aging and neuromuscular disease. Perisynaptic Schwann cells at neuromuscular junctions extend processes that bridge between denervated and reinnervated endplates, and guide axonal sprouts to reinnervate the denervated endplates. In a rat model of partial denervation, high levels of daily neuromuscular activity have been shown to inhibit the outgrowth of sprouts by preventing Schwann cell bridging. In this review, we consider (1) the relative roles of increasing levels of oxidative stress and neuromuscular activity to the destabilization of neuromuscular junctions with age and disease, and (2) how a progressive increase in the neuromuscular activity of declining numbers of functional MUs contributes to the progressive failure of adaptive sprouting and, in turn, to the progressive muscle weakness in the motoneuron diseases of post-polio syndrome and amyotrophic lateral sclerosis. We conclude that there is a time-related progression of MU loss, adaptive sprouting followed by maladaptive sprouting, and continuing recession of terminals during normal aging. The progression is accelerated in motoneuron disease, progressing more rapidly in the post-polio syndrome after prolonged denervation and extremely rapidly in ALS.
Collapse
Affiliation(s)
- Tessa Gordon
- Centre of Neuroscience, Division of Physical Medicine and Rehabilitation, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
| | | | | |
Collapse
|
18
|
Physical exercise increases GFAP expression and induces morphological changes in hippocampal astrocytes. Brain Struct Funct 2013; 219:293-302. [PMID: 23288255 DOI: 10.1007/s00429-012-0500-8] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/06/2012] [Accepted: 12/20/2012] [Indexed: 12/12/2022]
Abstract
Physical exercise has an important influence on brain plasticity, which affects the neuron-glia interaction. Astrocytes are susceptible to plasticity, and induce and stabilize synapses, regulate the concentration of various molecules, and support neuronal energy metabolism. The aim of our study was to investigate whether physical exercise is capable of altering the morphology, density and expression of glial fibrillary acidic protein (GFAP) in astrocytes from the CA1 region of rat hippocampus. Thirteen male rats were divided in two groups: sedentary (n = 6) and exercise (n = 7). The animals in the exercise group were submitted to a protocol of daily physical exercise on a treadmill for four consecutive weeks. GFAP immunoreactivity was evaluated using optical densitometry and the morphological analyses were an adaptation of Sholl's concentric circles method. Our results show that physical exercise is capable of increasing the density of GFAP-positive astrocytes as well as the regional and cellular GFAP expression. In addition, physical exercise altered astrocytic morphology as shown by the increase observed in the degree of ramification in the lateral quadrants and in the length of the longest astrocytic processes in the central quadrants. Our data demonstrate important changes in astrocytes promoted by physical exercise, supporting the idea that these cells are involved in regulating neural activity and plasticity.
Collapse
|
19
|
Rodnight RB, Gottfried C. Morphological plasticity of rodent astroglia. J Neurochem 2012; 124:263-75. [PMID: 23278277 DOI: 10.1111/jnc.12087] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/14/2012] [Revised: 11/04/2012] [Accepted: 11/04/2012] [Indexed: 11/29/2022]
Abstract
In the past two decades, there has been an explosion of research on the role of neuroglial interactions in the control of brain homeostasis in both physiological and pathological conditions. Astrocytes, a subtype of glia in the central nervous system, are dynamic signaling elements that regulate neurogenesis and development of brain circuits, displaying intimate dynamic relationships with neurons, especially at synaptic sites where they functionally integrate the tripartite synapse. When astrocytes are isolated from the brain and maintained in culture, they exhibit a polygonal shape unlike their precursors in vivo. However, cultured astrocytes can be induced to undergo morphological plasticity leading to process formation, either by interaction with neurons or by the influence of pharmacological agents. This review highlights studies on the molecular mechanisms underlying morphological plasticity in astrocyte cultures and intact brain tissue, both in situ and in vivo.
Collapse
Affiliation(s)
- Richard Burnard Rodnight
- School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, The University of Newcastle, Callaghan, Australia
| | | |
Collapse
|
20
|
Rosenkranz K, May C, Meier C, Marcus K. Proteomic analysis of alterations induced by perinatal hypoxic-ischemic brain injury. J Proteome Res 2012; 11:5794-803. [PMID: 23153068 DOI: 10.1021/pr3005869] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/28/2023]
Abstract
Perinatal hypoxic-ischemic brain injury is an important cause of neurological deficits still causing mortality and morbidity in the early period of life. As efficient clinical or pharmaceutical strategies to prevent or reduce the outcome of perinatal hypoxic-ischemic brain damage are limited, the development of new therapies is of utmost importance. To evolve innovative therapeutic concepts, elucidation of the mechanisms contributing to the neurological impairments upon hypoxic-ischemic brain injury is necessary. Therefore, we aimed for the identification of proteins that are affected by hypoxic-ischemic brain injury in neonatal rats. To assess changes in protein expression two days after induction of brain damage, a 2D-DIGE based proteome analysis was performed. Among the proteins altered after hypoxic-ischemic brain injury, Calcineurin A, Coronin-1A, as well as GFAP were identified, showing higher expression in lesioned hemispheres. Validation of the changes in Calcineurin A expression by Western Blot analysis demonstrated several truncated forms of this protein generated by limited proteolysis after hypoxia-ischemia. Further analysis revealed activation of calpain, which is involved in the limited proteolysis of Calcineurin. Active forms of Calcineurin are associated with the dephosphorylation of Darpp-32, an effect that was also demonstrated in lesioned hemispheres after perinatal brain injury.
Collapse
Affiliation(s)
- Katja Rosenkranz
- Department of Functional Proteomics, Ruhr-University Bochum, Germany.
| | | | | | | |
Collapse
|
21
|
Affiliation(s)
- Peter Dunkley
- School of Biomedical Sciences and Pharmacy; Faculty of Health, The University of Newcastle; University Drive; Callaghan; Australia
| |
Collapse
|
22
|
Ricoult SG, Goldman JS, Stellwagen D, Juncker D, Kennedy TE. Generation of microisland cultures using microcontact printing to pattern protein substrates. J Neurosci Methods 2012; 208:10-7. [PMID: 22561087 DOI: 10.1016/j.jneumeth.2012.04.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/18/2011] [Revised: 04/18/2012] [Accepted: 04/19/2012] [Indexed: 11/25/2022]
Abstract
The capacity to isolate small numbers of neurons in vitro is an essential tool to study the cell biology of synapses and the development of neuronal networks by specific cell types. Microisland culture assays allow for single neurons, or simple neural networks, to be isolated on islands of glial cells; however, the techniques commonly used to produce microisland substrates are expensive, challenging to control, and typically result in many discarded substrates. Here, we used microcontact printing to pattern a glass surface with islands of extracellular matrix proteins known to support neural cell growth and differentiation. To promote segregation of the cells to the islands, the substrate surrounding the islands was backfilled with polyethylene glycol (PEG), forming a relatively non-permissive surface on which cell attachment is limited. Astrocytes, and subsequently hippocampal neurons, were then seeded onto the islands of patterned protein. Using this method, readily reproducible patterns of protein islands were produced that permit cell attachment, differentiation, and growth. The technique is a rapid, inexpensive, and reliable means to generate patterned substrates appropriate for microisland cultures.
Collapse
Affiliation(s)
- Sébastien G Ricoult
- McGill Program in NeuroEngineering, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Ave., Montreal, Quebec H3A 2B4, Canada
| | | | | | | | | |
Collapse
|
23
|
Phosphorylation of GFAP is Associated with Injury in the Neonatal Pig Hypoxic-Ischemic Brain. Neurochem Res 2012; 37:2364-78. [DOI: 10.1007/s11064-012-0774-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/09/2012] [Revised: 03/23/2012] [Accepted: 03/29/2012] [Indexed: 12/24/2022]
|
24
|
Ye W, Huang X, Sun Y, Liu H, Jiang J, Cao Y. Ultrasound microbubbles combined with liposome-mediated pNogo-R shRNA delivery into neural stem cells. Neural Regen Res 2012; 7:54-9. [PMID: 25806059 PMCID: PMC4354117 DOI: 10.3969/j.issn.1673-5374.2012.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/25/2011] [Accepted: 12/23/2011] [Indexed: 11/18/2022] Open
Abstract
In the present study, ultrasound-mediated microbubble destruction (UMMD) alone and combined with liposome technology was used as a novel nonviral technique to transfect a Nogo receptor (Nogo-R) shRNA plasmid (pNogo-R shRNA) into neural stem cells (NSCs). Using green fluorescent protein as a reporter gene, transfection efficiency of NSCs was significantly higher in the group transfected with UMMD combined with liposomes compared with that of the group transfected with UMMD or liposomes alone, and did not affect cell vitality. In addition, Nogo-R mRNA and protein expression was dramatically decreased in the UMMD combined with liposome-mediated group compared with that of other groups after 24 hours of transfection. The UMMD technique combined with liposomes is a noninvasive gene transfer method, which showed minimal effects on cell viability and effectively increased transfer of Nogo-R shRNA into NSCs.
Collapse
Affiliation(s)
- Weixia Ye
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Xueping Huang
- Department of Orthopaedics, Luzhou People's Hospital, Luzhou 646000, Sichuan Province, China
| | - Yangyang Sun
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Hao Liu
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Jin Jiang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Youde Cao
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
25
|
|
26
|
Thacker M, Rivera LR, Cho HJ, Furness JB. The relationship between glial distortion and neuronal changes following intestinal ischemia and reperfusion. Neurogastroenterol Motil 2011; 23:e500-9. [PMID: 21410600 DOI: 10.1111/j.1365-2982.2011.01696.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Damage to mucosal epithelial cells, muscle cells and enteric neurons has been extensively studied following intestinal ischemia and reperfusion (I/R). Interestingly, the effects of intestinal I/R on enteric glia remains unexplored, despite knowledge that glia contribute to neuronal maintenance. Here, we describe structural damage to enteric glia and associated changes in distribution and immunoreactivity of the neuronal protein Hu. METHODS The mouse small intestine was made ischemic for 3 h and reperfused from 1 to 12 h. Immunohistochemical localisation of glial fibrillary acidic protein (GFAP), Hu and TUNEL were used to evaluate changes. KEY RESULTS At all time points glial cells became distorted, which was evident by their altered GFAP immunoreactivity, including an unusual appearance of bright perinuclear GFAP staining and the presence of GFAP globules. The numbers of neurons per ganglion area were significantly fewer in ganglia that contained distorted glia when compared with ganglia that contained glia of normal appearance. The distribution of Hu immunoreactivity was altered at all reperfusion time points. The presence of vacuoles and Hu granules in neurons was evident and an increase in nuclear Hu, relative to cytoplasmic Hu, was observed in ganglia that contained both normal and distorted glial cells. A number of neurons appeared to lose their Hu immunoreactivity, most noticeably in ganglia that contained distorted glial cells. TUNEL reaction occurred in a minority of glial cells and neurons. CONCLUSIONS & INFERENCES Structural damage to gliofilaments occurs following I/R and may be associated with damage to neighboring neurons.
Collapse
Affiliation(s)
- M Thacker
- Department of Anatomy & Cell Biology, University of Melbourne, Parkville, Vic., Australia.
| | | | | | | |
Collapse
|
27
|
Santin K, da Rocha RF, Cechetti F, Quincozes-Santos A, de Souza DF, Nardin P, Rodrigues L, Leite MC, Moreira JCF, Salbego CG, Gonçalves CA. Moderate exercise training and chronic caloric restriction modulate redox status in rat hippocampus. Brain Res 2011; 1421:1-10. [PMID: 21974860 DOI: 10.1016/j.brainres.2011.08.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/17/2011] [Revised: 07/27/2011] [Accepted: 08/02/2011] [Indexed: 01/29/2023]
Abstract
Physical activity has been related to antioxidant adaptations, which is associated with health benefits, including those to the nervous system. Additionally, available data suggest exercise and a caloric restriction regimen may reduce both the incidence and severity of neurological disorders. Therefore, our aim was to compare hippocampal redox status and glial parameters among sedentary, trained, caloric-restricted sedentary and caloric-restricted trained rats. Forty male adult rats were divided into 4 groups: ad libitum-fed sedentary (AS), ad libitum-fed exercise training (AE), calorie-restricted sedentary (RS) and calorie-restricted exercise training (RE). The caloric restriction (decrease of 30% in food intake) and exercise training (moderate in a treadmill) were carried out for 3 months. Thereafter hippocampus was surgically removed, and then redox and glial parameters were assessed. Increases in reduced glutathione (GSH) levels and total antioxidant reactivity (TAR) were observed in AE, RS and RE. The nitrite/nitrate levels decreased only in RE. We found a decrease in carbonyl content in AE, RS and RE, while no modifications were detected in thiobarbituric acid reactive substances (TBARS). Total reactive antioxidant potential (TRAP), superoxide dismutase (SOD) activity, S100B and glial fibrilary acid protein (GFAP) content did not change, but caloric restriction was able to increase glutamine synthetase (GS) activity in RS and glutamate uptake in RS and RE. Exercise training, caloric restriction and both combined can decrease oxidative damage in the hippocampus, possibly involving modulation of astroglial function, and could be used as a strategy for the prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
- Katiane Santin
- Department of Biochemistry, Institute of Health Basic Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Wang C, Kane MA, Napoli JL. Multiple retinol and retinal dehydrogenases catalyze all-trans-retinoic acid biosynthesis in astrocytes. J Biol Chem 2010; 286:6542-53. [PMID: 21138835 DOI: 10.1074/jbc.m110.198382] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/05/2023] Open
Abstract
All-trans-retinoic acid (atRA) stimulates neurogenesis, dendritic growth of hippocampal neurons, and higher cognitive functions, such as spatial learning and memory formation. Although astrocyte-derived atRA has been considered a key factor in neurogenesis, little direct evidence identifies hippocampus cell types and the enzymes that biosynthesize atRA. Here we show that primary rat astrocytes, but not neurons, biosynthesize atRA using multiple retinol dehydrogenases (Rdh) of the short chain dehydrogenase/reductase gene family and retinaldehyde dehydrogenases (Raldh). Astrocytes secrete atRA into their medium; neurons sequester atRA. The first step, conversion of retinol into retinal, is rate-limiting. Neurons and astrocytes both synthesize retinyl esters and reduce retinal into retinol. siRNA knockdown indicates that Rdh10, Rdh2 (mRdh1), and Raldh1, -2, and -3 contribute to atRA production. Knockdown of the Rdh Dhrs9 increased atRA synthesis ∼40% by increasing Raldh1 expression. Immunocytochemistry revealed cytosolic and nuclear expression of Raldh1 and cytosol and perinuclear expression of Raldh2. atRA autoregulated its concentrations by inducing retinyl ester synthesis via lecithin:retinol acyltransferase and stimulating its catabolism via inducing Cyp26B1. These data show that adult hippocampus astrocytes rely on multiple Rdh and Raldh to provide a paracrine source of atRA to neurons, and atRA regulates its own biosynthesis in astrocytes by directing flux of retinol. Observation of cross-talk between Dhrs9 and Raldh1 provides a novel mechanism of regulating atRA biosynthesis.
Collapse
Affiliation(s)
- Chao Wang
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California 94720, USA
| | | | | |
Collapse
|
29
|
Morphological changes in white matter astrocytes in response to hypoxia/ischemia in the neonatal pig. Brain Res 2010; 1319:164-74. [DOI: 10.1016/j.brainres.2010.01.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/22/2009] [Revised: 01/05/2010] [Accepted: 01/06/2010] [Indexed: 11/18/2022]
|
30
|
Steffek AE, McCullumsmith RE, Haroutunian V, Meador-Woodruff JH. Cortical expression of glial fibrillary acidic protein and glutamine synthetase is decreased in schizophrenia. Schizophr Res 2008; 103:71-82. [PMID: 18562176 PMCID: PMC3774017 DOI: 10.1016/j.schres.2008.04.032] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 07/19/2007] [Revised: 04/24/2008] [Accepted: 04/29/2008] [Indexed: 11/30/2022]
Abstract
Altered expression of structural and functional molecules expressed by astrocytes may play a role in the pathophysiology of schizophrenia. We investigated the hypothesis that the astrocytic enzyme glutamine synthetase, involved in maintaining the glutamate-glutamine cycle, and the cytoskeletal molecule glial fibrillary acidic protein (GFAP) are abnormally expressed in schizophrenia. We used Western blot analysis to measure levels of glutamine synthetase and GFAP in several brain regions of subjects with schizophrenia and a comparison group. We found that glutamine synthetase protein expression was significantly decreased in the superior temporal gyrus, and both glutamine synthetase and GFAP were significantly reduced in the anterior cingulate cortex in schizophrenia. Neither molecule demonstrated altered expression in the dorsolateral prefrontal cortex, primary visual cortex, or hippocampus. Chronic treatment with haloperidol did not alter the expression of these molecules in the rat brain, suggesting that our findings are not due to a medication effect. These data support an astrocytic component to the pathophysiology of schizophrenia and suggest that astrocytic molecules involved in enzymatic activity and cytoskeletal integrity may have a role in disease-related abnormalities in this illness.
Collapse
Affiliation(s)
- Amy E. Steffek
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI
| | - Robert E. McCullumsmith
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL
| | | | - James H. Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
31
|
Loureiro SO, Heimfarth L, Pelaez PDL, Vanzin CS, Viana L, Wyse ATS, Pessoa-Pureur R. Homocysteine activates calcium-mediated cell signaling mechanisms targeting the cytoskeleton in rat hippocampus. Int J Dev Neurosci 2008; 26:447-55. [PMID: 18406095 DOI: 10.1016/j.ijdevneu.2008.03.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/29/2008] [Revised: 03/03/2008] [Accepted: 03/03/2008] [Indexed: 01/13/2023] Open
Abstract
Homocysteine is considered to be neurotoxic and a risk factor for neurodegenerative diseases. Despite the increasing evidences of excitotoxic mechanisms of homocysteine (Hcy), little is known about the action of Hcy on the cytoskeleton. In this context, the aim of the present work was to investigate the signaling pathways involved in the mechanism of action of Hcy on cytoskeletal phosphorylation in cerebral cortex and hippocampus of rats during development. Results showed that 100 microM Hcy increased the intermediate filament (IF) phosphorylation only in 17-day-old rat hippocampal slices without affecting the cerebral cortex from 9- to 29-day-old animals. Stimulation of (45)Ca(2+) uptake supported the involvement of NMDA receptors and voltage-dependent channels in extracellular Ca(2+) flux, as well as Ca(2+) release from intracellular stores through inositol-3-phosphate and ryanodine receptors. Moreover, the mechanisms underlying the Hcy effect on hippocampus cytoskeleton involved the participation of phospholipase C, protein kinase C, mitogen-activated protein kinase, phosphoinositol-3 kinase and calcium/calmodulin-dependent protein kinase II. The Hcy-induced IF hyperphosphorylation was also related to G(i) protein and inhibition of cAMP levels. These findings demonstrate that Hcy at a concentration described to induce neurotoxicity activates the IF-associated phosphorylating system during development in hippocampal slices of rats through different cell signaling mechanisms. These results probably suggest that hippocampal rather than cortical cytoskeleton is susceptible to neurotoxical concentrations of Hcy during development and this could be involved in the neural damage characteristic of mild homocystinuric patients.
Collapse
Affiliation(s)
- Samanta Oliveira Loureiro
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande so Sul, Rua Ramiro Barcelos 2600, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
32
|
Tramontina F, Leite MC, Cereser K, de Souza DF, Tramontina AC, Nardin P, Andreazza AC, Gottfried C, Kapczinski F, Gonçalves CA. Immunoassay for glial fibrillary acidic protein: antigen recognition is affected by its phosphorylation state. J Neurosci Methods 2007; 162:282-6. [PMID: 17303248 DOI: 10.1016/j.jneumeth.2007.01.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2006] [Revised: 12/20/2006] [Accepted: 01/04/2007] [Indexed: 11/29/2022]
Abstract
Glial fibrillary acid protein (GFAP) is used commonly as a marker of astrogliosis and astrocyte activation in several situations involving brain injury. Its content may be measured by immunocytochemistry, immunoblotting or enzyme-linked immunosorbent assay (ELISA), usually employing commercial antibodies. Two major post-translational modifications in GFAP (phosphorylation and proteolysis) may alter the interpretation of results or for immunoassay standardization. This study using a non-sandwich ELISA aimed to investigate the putative changes in the immunorecognition due to the phosphorylated state of the antigen by a routinely used polyclonal anti-GFAP antibody from DAKO. Results involving in vitro phosphorylation of purified GFAP or biological samples (brain tissue, cell culture and cerebrospinal fluid) mediated by protein kinase dependent on cAMP indicate that GFAP phosphorylation improves the recognition by the used antibody. These results provide support to the understanding of fast changes in the GFAP-immunoreactivity and suggest that caution is necessary in the interpretation of results using this antibody, as well as indicate that the effect of post-translational modifications must be considered during the standardization of immunoassays with other antibodies.
Collapse
Affiliation(s)
- Francine Tramontina
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos, 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Williams SM, Sullivan RKP, Scott HL, Finkelstein DI, Colditz PB, Lingwood BE, Dodd PR, Pow DV. Glial glutamate transporter expression patterns in brains from multiple mammalian species. Glia 2005; 49:520-41. [PMID: 15578656 DOI: 10.1002/glia.20139] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/06/2022]
Abstract
It is generally assumed that rodent brains can be used as representative models of neurochemical function in other species, such as humans. We have compared the distributions of the predominant glial glutamate transporters in rodents, rabbits, cats, pigs, monkeys, and humans. We identify similarities but also significant differences between species. GLT-1v, which is abundantly expressed by rodent astrocytes, is expressed only in a rare subset of astrocytes of cats and humans, and appears to be absent from brains of rabbits and monkeys. Conversely, in the pig brain GLT-1v is expressed only by oligodendrocytes. GLAST and GLT-1alpha expression differed significantly between species; while rodents and rabbits exhibited uniform expression patterns in cortex, higher species, including cats, pigs, monkeys, and humans, exhibited heterogeneities in cortical and hippocampal expression. Patches devoid of labeling intermingling with patches of strong labeling were evident in areas such as temporal cortex and frontal cortex. In addition, we noted that in human motor cortex, there were inconsistencies in labeling for the C-terminal of GLT-1alpha and common domains of GLT-1, suggesting that the C-terminal region may be missing or that an unidentified splicing is present in many human astrocytes. Collectively our data suggest that assumptions as to the roles of glutamate transporters in any species may need to be tested empirically.
Collapse
Affiliation(s)
- Susan M Williams
- Department of Physiology and Pharmacology, School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Oppelt D, Rodnight R, Horn J, Fitarelli D, Kommers T, Oliveira D, Wofchuk S. Role of intracellular calcium stores on the effect of metabotropic glutamate receptors on phosphorylation of glial fibrillary acidic protein in hippocampal slices from immature rats. Neurochem Res 2004; 29:1541-5. [PMID: 15260132 DOI: 10.1023/b:nere.0000029567.68068.ab] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/12/2022]
Abstract
Phosphorylation of glial fibrillary acidic protein (GFAP) in slices from immature rats is stimulated by glutamate via a group II metabotropic glutamate receptor (mGluR II) and by absence of external Ca2+ in reactions that are not additive (Wofchuk and Rodnight, Neurochem. Int. 24:517-523, 1994). These observations suggested that glutamate, via an mGluR, inhibits Ca(2+)-entry through L-type Ca2+ channels and down-regulates a Ca(2+)-dependent dephosphorylation event coupled to GFAP. Because ryanodine receptors are present on internal Ca2+ stores and are associated with L-type Ca(2+)-channels, we investigated the possibility that the glutamatergic modulation of GFAP phosphorylation involves internal Ca2+ stores regulated by ryanodine receptors and whether the Ca2+ originating from these stores acts in a similar manner to external Ca2+. The results showed that the ryanodine receptor-agonists, caffeine and ryanodine and thapsigargin, all of which in appropriate doses increase cytoplasmic Ca2+, reversed the stimulation of GFAP phosphorylation given by 1S,3R-ACPD, an mGluR II agonist.
Collapse
Affiliation(s)
- D Oppelt
- Departamento de Bioquimica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
35
|
Frizzo JK, Tramontina F, Bortoli E, Gottfried C, Leal RB, Lengyel I, Donato R, Dunkley PR, Gonçalves CA. S100B-mediated inhibition of the phosphorylation of GFAP is prevented by TRTK-12. Neurochem Res 2004; 29:735-40. [PMID: 15098935 DOI: 10.1023/b:nere.0000018844.51009.40] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/12/2022]
Abstract
S100B belongs to a family of calcium-binding proteins involved in cell cycle and cytoskeleton regulation. We observed an inhibitory effect of S100B on glial fibrillary acidic protein (GFAP) phosphorylation, when stimulated by cAMP or Ca2+/calmodulin, in a cytoskeletal fraction from primary astrocyte cultures. We found that S100B has no direct effect on CaM KII activity, the major kinase in this cytoskeletal fraction able to phosphorylate GFAP. The inhibition of GFAP phosphorylation is most likely due to the binding of S100B to the phosphorylation sites on this protein and blocking the access of these sites to the protein kinases. This inhibition was dependent on Ca2+. However, Zn2+ could substitute for Ca2+. The inhibitory effect of S100B was prevented by TRTK-12, a peptide that blocks S100B interaction with several target proteins including glial fibrillary acidic protein. These data suggest a role for S100B in the assembly of intermediate filaments in astrocytes.
Collapse
Affiliation(s)
- Juliana Karl Frizzo
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Asklund T, Appelskog IB, Ammerpohl O, Ekström TJ, Almqvist PM. Histone deacetylase inhibitor 4-phenylbutyrate modulates glial fibrillary acidic protein and connexin 43 expression, and enhances gap-junction communication, in human glioblastoma cells. Eur J Cancer 2004; 40:1073-81. [PMID: 15093585 DOI: 10.1016/j.ejca.2003.11.034] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/13/2003] [Accepted: 11/17/2003] [Indexed: 11/21/2022]
Abstract
Human glioblastoma cell cultures were established and the expression of glial fibrillary acidic protein (GFAP) and the gap-junction protein connexin 43 (Cx43) was confirmed by Western blot. Following treatment with 4-phenylbutyrate (4-PB), increased concentrations of non-phosphorylated GFAP were seen, while phosphorylated isoforms remained intact. Immunocytochemical staining of glioblastoma cells revealed an intracellular redistribution of GFAP. In addition to cytoplasmic immunostaining, GFAP immunoreactivity was also associated with the nucleus and/or the nuclear membrane. Phosphorylated and non-phosphorylated Cx43 proteins were increased 2- to 5-fold following 4-PB treatment, and were redistributed to areas of the cell surface, participating in cell-to-cell contacts. In addition, functional gap-junction coupling was amplified, as indicated by increased fluorescent dye transfer, and elevated levels of Cx43 protein were detected in parallel with enhanced gap-junction communication. Induced cell differentiation, with improved functional coupling of tumour cells, may be of importance for therapeutic strategies involving intercellular transport of low molecular-weight compounds.
Collapse
Affiliation(s)
- T Asklund
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
37
|
Tam SL, Gordon T. Neuromuscular activity impairs axonal sprouting in partially denervated muscles by inhibiting bridge formation of perisynaptic Schwann cells. ACTA ACUST UNITED AC 2003; 57:221-34. [PMID: 14556287 DOI: 10.1002/neu.10276] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/10/2022]
Abstract
Following partial denervation of rat hindlimb muscle, terminal Schwann cells extend processes from denervated endplates to induce and guide sprouting from the remaining intact axons. Increased neuromuscular activity significantly reduces motor unit enlargement and sprouting during the acute phase of sprouting. These findings led to the hypothesis that increased neuromuscular activity perturbs formation of Schwann cell bridges and thereby reduces sprouting. Adult rat tibialis anterior (TA) muscles were extensively denervated by avulsion of L4 spinal root and were immediately subjected to normal caged activity or running exercise (8 h daily) for 3, 7, 14, 21, and 28 days. Combined silver/cholinesterase histochemical staining revealed that the progressive reinnervation of denervated endplates by sprouts over a 1 month period in the extensively partially denervated TA muscles was completely abolished by increased neuromuscular activity. Immunohistochemical staining and triple immunofluorescence revealed that the increased neuromuscular activity did not perturb the production of Schwann cell processes, but prevented bridging between Schwann cell processes at innervated and denervated endplates. Our findings suggest that failure of Schwann cell processes to bridge between endplates accounts, at least in part, for the inhibitory effect of increased neuromuscular activity on sprouting.
Collapse
Affiliation(s)
- S L Tam
- Centre of Neuroscience, Division of Physical Medicine and Rehabilitation, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada T6G 2S2.
| | | |
Collapse
|
38
|
Simonová Z, Sterbová K, Brozek G, Komárek V, Syková E. Postnatal hypobaric hypoxia in rats impairs water maze learning and the morphology of neurones and macroglia in cortex and hippocampus. Behav Brain Res 2003; 141:195-205. [PMID: 12742256 DOI: 10.1016/s0166-4328(02)00366-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/21/2022]
Abstract
Newborn rats were exposed to intermittent hypobaric hypoxia from birth until the age of 19 days. Spatial memory was tested in a Morris water maze from postnatal day (P) 23 to P32 and from P100 to P109. From P24 to P27 and on days P100 and P101, the escape latencies of hypoxic animals were longer than those of controls. At P24, the number of neuronal bodies increased in cortical layer II of the somatosensory, motor, and auditory areas, and in layer V of the motor area, but the number of neuronal bodies throughout the whole cortical thickness was unchanged. Decreases in the immunostaining density for neurofilaments (anti-NF 160), astrocytes (anti-GFAP), and oligodendrocytes (RIP) were found in the hippocampus, and the typical parallel organisation of neuronal and macroglial processes was lost. Decreases in immunostaining for neurofilaments and oligodendrocytes were also found in the somatosensory cortex and motor cortex. In adult hypoxic rats, at P114-P240, the number of neuronal bodies and the immunostaining density for neurofilaments, astrocytes, and oligodendrocytes in the examined areas were similar to adult controls; however, in the hippocampus we found hypertrophy of fine astrocytic processes and a decreased number of oligodendrocytic processes. We conclude that the neonatal brain damage induced by hypobaric hypoxia impairs spatial memory in infant as well as adult rats. Hypobaric hypoxia delays the maturation of neurones and substantially affects macroglia in the cortex and hippocampus.
Collapse
Affiliation(s)
- Zuzana Simonová
- Department of Neuroscience, Center for Cell Therapy and Tissue Repair, Charles University, Second Medical Faculty, 150 18 Prague 5, Czech Republic
| | | | | | | | | |
Collapse
|
39
|
Funchal C, de Lima Pelaez P, Loureiro SO, Vivian L, Dall Bello Pessutto F, de Almeida LMV, Tchernin Wofchuk S, Wajner M, Pessoa Pureur R. alpha-Ketoisocaproic acid regulates phosphorylation of intermediate filaments in postnatal rat cortical slices through ionotropic glutamatergic receptors. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2002; 139:267-76. [PMID: 12480141 DOI: 10.1016/s0165-3806(02)00578-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 10/27/2022]
Abstract
In this study we investigated the effects of alpha-ketoisocaproic acid (KIC), the main keto acid accumulating in the inherited neurometabolic disorder maple syrup urine disease (MSUD), on the in vitro incorporation of 32P into intermediate filament (IF) proteins from cerebral cortex of rats during development. KIC decreased the in vitro incorporation of 32P into the IF proteins studied up to day 12, had no effect on day 15, and increased this phosphorylation in cortical slices of 17- and 21-day-old rats. A similar effect on IF phosphorylation was achieved along development by incubating cortical slices with glutamate. Furthermore, the altered phosphorylation caused by the presence of KIC in the incubation medium was mediated by the ionotropic receptors NMDA, AMPA and kainate up to day 12 and by NMDA and AMPA in tissue slices from 17- and 21-day-old rats. The results suggest that alterations of IF phosphorylation may be associated with the neuropathology of MSUD.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Cerebral Cortex/drug effects
- Cerebral Cortex/growth & development
- Cerebral Cortex/metabolism
- Excitatory Amino Acid Agonists/pharmacology
- Excitatory Amino Acid Antagonists/pharmacology
- Glial Fibrillary Acidic Protein/drug effects
- Glial Fibrillary Acidic Protein/metabolism
- Glutamic Acid/metabolism
- Glutamic Acid/pharmacology
- Intermediate Filament Proteins/drug effects
- Intermediate Filament Proteins/metabolism
- Intermediate Filaments/drug effects
- Intermediate Filaments/metabolism
- Keto Acids/metabolism
- Keto Acids/pharmacology
- Maple Syrup Urine Disease/metabolism
- Maple Syrup Urine Disease/physiopathology
- Neurons/drug effects
- Neurons/metabolism
- Organ Culture Techniques
- Phosphorylation/drug effects
- Rats
- Rats, Wistar
- Receptors, AMPA/drug effects
- Receptors, AMPA/metabolism
- Receptors, Glutamate/drug effects
- Receptors, Glutamate/metabolism
- Receptors, Kainic Acid/drug effects
- Receptors, Kainic Acid/metabolism
- Receptors, Metabotropic Glutamate/drug effects
- Receptors, Metabotropic Glutamate/metabolism
- Receptors, N-Methyl-D-Aspartate/drug effects
- Receptors, N-Methyl-D-Aspartate/metabolism
- Synaptic Transmission/drug effects
- Synaptic Transmission/physiology
- Vimentin/drug effects
- Vimentin/metabolism
Collapse
Affiliation(s)
- Cláudia Funchal
- Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Departamento de Bioquímica, Rua Ramiro Barcelos 2600 anexo, 90035-003, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Vivian L, Pessutto FDB, de Almeida LMV, Loureiro SDO, Pelaez PDL, Funchal C, Wajner M, Pessoa-Pureur R. Effect of propionic and methylmalonic acids on the high molecular weight neurofilament subunit (NF-H) in rat cerebral cortex. Neurochem Res 2002; 27:1691-7. [PMID: 12515324 DOI: 10.1023/a:1021699314007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/12/2022]
Abstract
Propionic and methylmalonic acidemias are inherited neurometabolic disorders biochemically characterized by tissue accumulation of propionic (PA) and methylmalonic (MMA) acids, respectively. Neurofilaments (NF) are important cytoskeletal proteins and phosphorylation/dephosphorylation of NF is important to stabilize the cytoskeleton. We investigated the effects of PA and MMA on the high molecular weight neurofilament subunit associated with the cytoskeletal fraction of rat cerebral cortex along development. Cortical slices from 9- to 60-day-old rats were incubated with 2.5 mM PA or MMA. The cytoskeletal fraction was extracted and the immunoreactivity for phosphorylated or total NF-H was analyzed by immunoblotting using specific antibodies. Results showed that treatment of tissue slices with the acids induced an increased Triton-insoluble phosphorylated NF-H immunoreactivity in up to 17-day-old rats. Furthermore, treatments significantly increased the total amount of NF-H in 12-day-old rats. These findings indicate that PA and MMA alter the dynamic regulation of NF-H assembly in the cytoskeletal fraction.
Collapse
Affiliation(s)
- L Vivian
- Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Departamento de Bioquímica, 90035-003 Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Kommers T, Rodnight R, Boeck C, Vendite D, Oliveira D, Horn J, Oppelt D, Wofchuk S. Phosphorylation of glial fibrillary acidic protein is stimulated by glutamate via NMDA receptors in cortical microslices and in mixed neuronal/glial cell cultures prepared from the cerebellum. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2002; 137:139-48. [PMID: 12220706 DOI: 10.1016/s0165-3806(02)00434-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/19/2022]
Abstract
In previous work we showed that phosphorylation of glial fibrillary acidic protein (GFAP), an astrocyte marker, is increased by glutamate in hippocampal slices from immature rats via a type II metabotropic receptor. In the present work we show that glutamate also stimulates GFAP phosphorylation in microslices prepared from immature cerebellar cortex, but by a different receptor mechanism from that observed in the hippocampus. Thus, in cerebellar microslices, NMDA consistently stimulated GFAP phosphorylation, whereas no effect of metabotropic or non-NMDA ionotropic agonists was observed. Glutamate and NMDA also stimulated GFAP phosphorylation in mixed neuronal/glial cell cultures from the cerebellum, although no effect of these agonists was observed in primary cultures of cerebellar astrocytes. In both models, the effects of glutamate and NMDA were dependent on external Ca(2+), were reversed by the NMDA receptor antagonist AP5 and were not blocked by tetrodotoxin. In the slice study the effect of NMDA was confined to a period starting with the first detectable expression of GFAP at 10 days and finishing at 16 days postnatal, as previously observed with metabotropic agonists in hippocampal slices. This period in the rat corresponds to the start of synaptogenesis when astrocyte hypertrophy is occurring. The results are discussed in the light of information in the literature on the occurrence of functional NMDA receptor subunits in glia.
Collapse
Affiliation(s)
- Trícia Kommers
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600, Anexo 90035003, RS, Porto Alegre, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Glutamine synthetase (GS) in brain is located mainly in astrocytes. One of the primary roles of astrocytes is to protect neurons against excitotoxicity by taking up excess ammonia and glutamate and converting it into glutamine via the enzyme GS. Changes in GS expression may reflect changes in astroglial function, which can affect neuronal functions. Hyperammonemia is an important factor responsible of hepatic encephalopathy (HE) and causes astroglial swelling. Hyperammonemia can be experimentally induced and an adaptive astroglial response to high levels of ammonia and glutamate seems to occur in long-term studies. In hyperammonemic states, astroglial cells can experience morphological changes that may alter different astrocyte functions, such as protein synthesis or neurotransmitters uptake. One of the observed changes is the increase in the GS expression in astrocytes located in glutamatergic areas. The induction of GS expression in these specific areas would balance the increased ammonia and glutamate uptake and protect against neuronal degeneration, whereas, decrease of GS expression in non-glutamatergic areas could disrupt the neuron-glial metabolic interactions as a consequence of hyperammonemia. Induction of GS has been described in astrocytes in response to the action of glutamate on active glutamate receptors. The over-stimulation of glutamate receptors may also favour nitric oxide (NO) formation by activation of NO synthase (NOS), and NO has been implicated in the pathogenesis of several CNS diseases. Hyperammonemia could induce the formation of inducible NOS in astroglial cells, with the consequent NO formation, deactivation of GS and dawn-regulation of glutamate uptake. However, in glutamatergic areas, the distribution of both glial glutamate receptors and glial glutamate transporters parallels the GS location, suggesting a functional coupling between glutamate uptake and degradation by glutamate transporters and GS to attenuate brain injury in these areas. In hyperammonemia, the astroglial cells located in proximity to blood-vessels in glutamatergic areas show increased GS protein content in their perivascular processes. Since ammonia freely crosses the blood-brain barrier (BBB) and astrocytes are responsible for maintaining the BBB, the presence of GS in the perivascular processes could produce a rapid glutamine synthesis to be released into blood. It could, therefore, prevent the entry of high amounts of ammonia from circulation to attenuate neurotoxicity. The changes in the distribution of this critical enzyme suggests that the glutamate-glutamine cycle may be differentially impaired in hyperammonemic states.
Collapse
Affiliation(s)
- I Suárez
- Departamento de Biología Celular y Genética, Facultad de Biología, Universidad de Alcalá, 28871, Madrid, Spain.
| | | | | |
Collapse
|
43
|
Ziegler DR, Araújo E, Rotta LN, Perry ML, Gonçalves CA. A ketogenic diet increases protein phosphorylation in brain slices of rats. J Nutr 2002; 132:483-7. [PMID: 11880575 DOI: 10.1093/jn/132.3.483] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/14/2022] Open
Abstract
Ketogenic diets have been used to treat seizure disorders of children and recently it was shown to increase the drug-induced seizure threshold in rats. Protein phosphorylation is a major regulatory mechanism of signal transduction that has been implicated in modulating neuronal excitability. We investigated the basal protein phosphorylation in microslices from different brain regions (hippocampus, cerebral cortex and cerebellum) of young rats fed a ketogenic diet, and we evaluated the effect of this diet on weight development and health of these rats based on serum biochemistry. Thirty-day-old rats consumed ad libitum ketogenic (high fat) or control diets for 8 wk. Rats consuming the high fat diet had ketonemia without signs of undernutrition or illness. Microslices were incubated in media containing (32)P-phosphate, and (32)P-phosphoprotein content was analyzed by one- or two-dimensional electrophoresis followed by autoradiography. Basal protein phosphorylation was greater in brain slices from ketogenic rats. Different increments of synapsin I, GAP-43 and GFAP phosphorylation were observed in two-dimensional autoradiography. A ketogenic diet induced metabolic changes affecting the basal status of protein phosphorylation. This change could affect the mechanisms of signal transduction in neural cells involved in the increase in the seizure threshold.
Collapse
Affiliation(s)
- Denize R Ziegler
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | |
Collapse
|
44
|
Hassouna IA, Rahmy TR. REACTIVE ASTROCYTIC RESPONSE AND INCREASED PROLIFERATING CELL NUCLEAR ANTIGEN EXPRESSION IN CEREBRAL CORTEX OF ENVENOMED RATS. ACTA ACUST UNITED AC 2001. [DOI: 10.1081/txr-100108559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/03/2022]
|
45
|
Elezgarai I, Bilbao A, Mateos JM, Azkue JJ, Benítez R, Osorio A, Díez J, Puente N, Doñate-Oliver F, Grandes P. Group II metabotropic glutamate receptors are differentially expressed in the medial nucleus of the trapezoid body in the developing and adult rat. Neuroscience 2001; 104:487-98. [PMID: 11377849 DOI: 10.1016/s0306-4522(01)00080-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/18/2022]
Abstract
The existence of a neuronal-glial signalling through the activation of neurotransmitter receptors expressed in glia is well-documented. In excitatory synapses, glutamate released from presynaptic terminals activates not only postsynaptic receptors, but also ionotropic and metabotropic glutamate receptors localized in the glia ensheathing the synapses. The medial nucleus of the trapezoid body of the auditory system is involved in the localization of sounds in the space. In this nucleus, the large excitatory synaptic terminals formed by the calyces of Held on the principal globular cell bodies are wrapped by astrocytic processes. Since these synapses are functional from early postnatal days, glia receiving excitatory synaptic signals from the calyces may participate in modulating the maturation and development of the system. Groups I and II of metabotropic glutamate receptors (mGluRs) have been localized in glial cells in different brain regions. To investigate whether group II mGluRs are present in the medial nucleus of the trapezoid body, we have studied the pattern of expression of mGluR2/3 in the developing and mature nucleus by means of immunocytochemichal methods. The most remarkable finding was the switch in the occurrence of mGluR2/3 from glial to neuronal compartments. Thus, a preferential localization of mGluR2/3 immunoreactivity was observed in astrocytic processes surrounding the calyces of Held during the early postnatal development. In contrast, the main feature in adult rats was the presence of the group II mGluRs in presynaptic calyces of Held and postsynaptic principal globular cells.From these observations we suggest a role for group II mGluRs in neuronal-glial signalling in the calyx of Held-principal globular neuron synapses. Activation of these receptors might be relevant to the maturation and modulation of synaptic transmission in the medial nucleus of the trapezoid body.
Collapse
Affiliation(s)
- I Elezgarai
- Department of Neurosciences, Faculty of Medicine and Dentistry, Basque Country University, 699-48080, Bilbao, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
de Mattos-Dutra A, Meirelles R, Bevilaqua da Rocha B, Kommers T, Wofchuk ST, Wajner M, Pessoa-Pureur R. Methylmalonic and propionic acids increase the in vitro incorporation of 32P into cytoskeletal proteins from cerebral cortex of young rats through NMDA glutamate receptors. Brain Res 2000; 856:111-8. [PMID: 10677617 DOI: 10.1016/s0006-8993(99)02380-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/25/2022]
Abstract
In this study we investigated the effects of methylmalonic acid (MMA) and propionic acid (PA) on the phosphorylation of cytoskeletal proteins of cerebral cortex of rats. Slices of tissue were incubated with 32P-orthophosphate in the presence or absence of glutamate, MMA, PA and ionotropic or metabotropic glutamate receptor agonists. The cytoskeletal fraction was isolated and the radioactivity incorporated into the cytoskeletal proteins was measured. Results demonstrated that the acids, glutamate and NMDA increased the phosphorylation of the proteins studied. However, this effect was not observed for non-NMDA ionotropic agonists or metabotropic agonists. Experiments using glutamate receptor antagonists confirmed that MMA and PA at the same concentrations as found in tissues from propionic or methylmalonic acidemic children increase the phosphorylation of cytoskeletal proteins, possibly via NMDA glutamate receptors. Therefore, it is feasible that these findings may be related to the neurological dysfunction characteristic of these disorders.
Collapse
Affiliation(s)
- A de Mattos-Dutra
- Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Departamento de Bioquímica, Rua Ramiro Barcelos 2600 anexo, 90035-003, Porto Alegre, Brazil
| | | | | | | | | | | | | |
Collapse
|
47
|
Karl J, Gottfried C, Tramontina F, Dunkley P, Rodnight R, Gonçalves CA. GFAP phosphorylation studied in digitonin-permeabilized astrocytes: standardization of conditions. Brain Res 2000; 853:32-40. [PMID: 10627305 DOI: 10.1016/s0006-8993(99)02224-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/17/2022]
Abstract
Cycles of assembly/disassembly of the intermediate filaments of astrocytes are modulated by the phosphorylation of glial fibrillary acidic protein (GFAP). The sites on GFAP are localized at the N-terminal where they are phosphorylated by cAMP-dependent and Ca(2+)-dependent protein kinases. Phosphorylation of GFAP has been investigated in brain slices, astrocyte cultures, cytoskeletal fractions and purified systems. Here we describe a different approach to study GFAP phosphorylation. We show that permeabilization of astrocytes in culture with digitonin allows direct access to the systems phosphorylating GFAP. Conditions for the permeabilization were established with an assay based on the exclusion of Trypan blue. Incubation of permeabilized cells with cAMP and Ca(2+) increased the phosphorylation state of GFAP. Immunocytochemistry with anti-GFAP showed that permeabilized astrocytes retained their typical flat, fibroblast morphology and exhibited well preserved glial filaments. On incubation with cAMP the filaments apparently condensed to form long processes. The results suggest the approach of studying structural changes in glial filaments in parallel to protein phosphorylation, in the presence of specific modulators of protein kinases and phosphatases has considerable potential.
Collapse
Affiliation(s)
- J Karl
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos, 2600-Anexo, 90035-003, Porto Alegre, Brazil
| | | | | | | | | | | |
Collapse
|
48
|
Gottfried C, Valentim L, Salbego C, Karl J, Wofchuk ST, Rodnight R. Regulation of protein phosphorylation in astrocyte cultures by external calcium ions: specific effects on the phosphorylation of glial fibrillary acidic protein (GFAP), vimentin and heat shock protein 27 (HSP27). Brain Res 1999; 833:142-9. [PMID: 10375689 DOI: 10.1016/s0006-8993(99)01503-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/23/2022]
Abstract
The effect of external Ca2+ ([Ca2+]e) on the incorporation of [32P] into total protein, cytoskeletal proteins and the heat shock protein HSP27, was studied in primary cultures of astrocytes from the rat hippocampus. Zero [Ca2+]e increased total 32P-incorporation into astrocyte protein and when this was normalized to 100%, incorporation was significantly increased into glial fibrillary acidic protein (GFAP), vimentin (VIM) and HSP27. The difference in total 32P-incorporation between zero [Ca2+]e and 1 mM [Ca2+]e was reversed by incubation of the cells with the protein phosphatase inhibitor okadaic acid in the range 1-10 nM; higher concentrations of okadaic acid (50-100 nM) further increased total 32P-incorporation. In zero [Ca2+]e the non-specific channel blocker Co2+ (1 mM) decreased total 32P-incorporation by approximately 30%. The results were compared with a previous study [S.T. Wofchuk, R. Rodnight, Age-dependent changes in the regulation by external calcium ions of the phosphorylation of glial fibrillary acidic protein in slices of rat hippocampus, Dev. Brain Res. 85 (1995) 181-186] in which it was shown that in immature hippocampal slices zero [Ca2+]e compared with 1 mM [Ca2+]e increased 32P-incorporation into GFAP without changing total incorporation. The difference between the results for total 32P-incorporation obtained in cultured astrocytes and immature brain tissue was found to be related to the concentration of [Ca2+]e in the medium since in slices concentrations of [Ca2+]e higher than 1 mM progressively decreased total incorporation. The difference may reflect a higher Ca2+-permeability of the plasma membrane in cultured astrocytes and/or to the complex structure of the slice tissue. In two-dimensional electrophoresis HSP27, in contrast to GFAP and VIM, was separated into 3 immunodetectable isoforms only two of which were normally phosphorylated. After labelling in the presence of okadaic acid both immunodetectable and phosphorylated HSP27 focussed as a single polypeptide. Phorbol dibutyrate (1 microM) and zero [Ca2+]e stimulated the phosphorylation of both isoforms, but in the case of zero [Ca2+]e the effect on the more acidic isoform was proportionally greater.
Collapse
Affiliation(s)
- C Gottfried
- Departamento de Bioquímica, UFRGS, Instituto de Ciências Básicas da Saúde, Rua Ramiro Barcelos 2600-Anexo, 90.035.003, Porto Alegre, RS, Brazil
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Similar to astrocytes at CNS synapses, perisynaptic Schwann cells (PSCs) surround nerve terminals at the neuromuscular junction (NMJ). These special teloglial cells are sensitive to neurotransmitters and upregulate glial fibrillary acidic protein (GFAP) when deprived of synaptic activity. We found that activation of muscarinic acetylcholine receptors (mAChRs) at PSCs, but not purinergic (ATP and adenosine) or peptidergic [substance P (SP) and calcitonin gene-related peptide (CGRP)] receptors, prevented this upregulation. When applied onto single PSCs, muscarine evoked Ca2+ responses that fatigued but prevented upregulation of this glial cytoskeletal protein. Application of ATP onto single PSCs evoked Ca2+ signals that showed little fatigue, and GFAP upregulation occurred. Thus, Ca2+ signals alone cannot prevent GFAP upregulation in the PSCs. After blockade of cholinergic receptors by gallamine, neuronal activity was not effective in maintaining low GFAP levels in the perisynaptic glia. Last, immunohistochemistry disclosed mAChRs on PSCs and nearby fibroblasts. Thus, acetylcholine secreted by the nerve terminal acts on the PSCs via mAChRs to regulate GFAP. Cytoskeletal changes may influence perisynaptic glial functions, including growth, remodeling, and modulation of the synapse.
Collapse
|
50
|
Ziegler DR, Innocente CE, Leal RB, Rodnight R, Gonçalves CA. The S100B protein inhibits phosphorylation of GFAP and vimentin in a cytoskeletal fraction from immature rat hippocampus. Neurochem Res 1998; 23:1259-63. [PMID: 9804281 DOI: 10.1023/a:1020740115790] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/12/2022]
Abstract
The S100B protein belongs to a family of small Ca2+-binding proteins involved in several functions including cytoskeletal reorganization. The effect of S 100B on protein phosphorylation was investigated in a cytoskeletal fraction prepared from immature rat hippocampus. An inhibitory effect of 5 microM S100B on total protein phosphorylation, ranging from 25% to 40%, was observed in the presence of Ca2+ alone, Ca2+ plus calmodulin or Ca2+ plus cAMP. Analysis by two dimensional electrophoresis revealed a Ca2+/calmodulin-dependent and a Ca2+/cAMP-dependent inhibitory effect of S100B, ranging from 62% to 67% of control, on the phosphorylation of the intermediate filament proteins glial fibrillary acidic protein (GFAP) and vimentin. The fact that S100B binds to the N-terminal domain of GFAP and that the two proteins are co-localized in astrocytes suggests a potential in vivo role for S100B in modulating the phosphorylation of intermediate filament proteins in glia.
Collapse
Affiliation(s)
- D R Ziegler
- Departamento de Bioquímica, ICBS, UFRGS, Porto Alegre, Brazil
| | | | | | | | | |
Collapse
|