51
|
Mills EA, Mao-Draayer Y. Understanding Progressive Multifocal Leukoencephalopathy Risk in Multiple Sclerosis Patients Treated with Immunomodulatory Therapies: A Bird's Eye View. Front Immunol 2018; 9:138. [PMID: 29456537 PMCID: PMC5801425 DOI: 10.3389/fimmu.2018.00138] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/16/2018] [Indexed: 12/14/2022] Open
Abstract
The increased use of newer potent immunomodulatory therapies for multiple sclerosis (MS), including natalizumab, fingolimod, and dimethyl fumarate, has expanded the patient population at risk for developing progressive multifocal leukoencephalopathy (PML). These MS therapies shift the profile of lymphocytes within the central nervous system (CNS) leading to increased anti-inflammatory subsets and decreased immunosurveillance. Similar to MS, PML is a demyelinating disease of the CNS, but it is caused by the JC virus. The manifestation of PML requires the presence of an active, genetically rearranged form of the JC virus within CNS glial cells, coupled with the loss of appropriate JC virus-specific immune responses. The reliability of metrics used to predict risk for PML could be improved if all three components, i.e., viral genetic strain, localization, and host immune function, were taken into account. Advances in our understanding of the critical lymphocyte subpopulation changes induced by these MS therapies and ability to detect viral mutation and reactivation will facilitate efforts to develop these metrics.
Collapse
Affiliation(s)
- Elizabeth A Mills
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States.,Graduate Program in Immunology, Program in Biomedical Sciences, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
52
|
THE ROLE OF ASTROCYTES IN THE HOMEOSTASIS MAINTAINING IN THE NERVE TISSUE. WORLD OF MEDICINE AND BIOLOGY 2018. [DOI: 10.26724/2079-8334-2018-2-64-220-225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
53
|
Chowdhury HH, Cerqueira SR, Sousa N, Oliveira JM, Reis RL, Zorec R. The uptake, retention and clearance of drug-loaded dendrimer nanoparticles in astrocytes – electrophysiological quantification. Biomater Sci 2018; 6:388-397. [DOI: 10.1039/c7bm00886d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Endocytosed dendrimer nanoparticles (NPs) are cleared from the astrocytes by an increased rate of transient exocytotic fusion events.
Collapse
Affiliation(s)
- Helena H. Chowdhury
- Laboratory of Neuroendocrinology – Molecular Cell Physiology
- Institute of Pathophysiology
- Faculty of Medicine
- 1000 Ljubljana
- Slovenia
| | - Susana R. Cerqueira
- 3B's Research Group – Biomaterials
- Biodegradables and Biomimetics
- University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- 4805-017 Barco GMR
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS)
- School of Health Sciences
- University of Minho
- 4710-057 Braga
- Portugal
| | - Joaquim M. Oliveira
- 3B's Research Group – Biomaterials
- Biodegradables and Biomimetics
- University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- 4805-017 Barco GMR
| | - Rui L. Reis
- 3B's Research Group – Biomaterials
- Biodegradables and Biomimetics
- University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- 4805-017 Barco GMR
| | - Robert Zorec
- Laboratory of Neuroendocrinology – Molecular Cell Physiology
- Institute of Pathophysiology
- Faculty of Medicine
- 1000 Ljubljana
- Slovenia
| |
Collapse
|
54
|
Martin-Jiménez CA, García-Vega Á, Cabezas R, Aliev G, Echeverria V, González J, Barreto GE. Astrocytes and endoplasmic reticulum stress: A bridge between obesity and neurodegenerative diseases. Prog Neurobiol 2017; 158:45-68. [DOI: 10.1016/j.pneurobio.2017.08.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/22/2017] [Accepted: 08/04/2017] [Indexed: 12/13/2022]
|
55
|
Salazar K, Martínez F, Pérez-Martín M, Cifuentes M, Trigueros L, Ferrada L, Espinoza F, Saldivia N, Bertinat R, Forman K, Oviedo MJ, López-Gambero AJ, Bonansco C, Bongarzone ER, Nualart F. SVCT2 Expression and Function in Reactive Astrocytes Is a Common Event in Different Brain Pathologies. Mol Neurobiol 2017; 55:5439-5452. [PMID: 28942474 DOI: 10.1007/s12035-017-0762-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/31/2017] [Indexed: 11/28/2022]
Abstract
Ascorbic acid (AA), the reduced form of vitamin C, acts as a neuroprotector by eliminating free radicals in the brain. Sodium/vitamin C co-transporter isoform 2 (SVCT2) mediates uptake of AA by neurons. It has been reported that SVCT2 mRNA is induced in astrocytes under ischemic damage, suggesting that its expression is enhanced in pathological conditions. However, it remains to be established if SVCT expression is altered in the presence of reactive astrogliosis generated by different brain pathologies. In the present work, we demonstrate that SVCT2 expression is increased in astrocytes present at sites of neuroinflammation induced by intracerebroventricular injection of a GFP-adenovirus or the microbial enzyme, neuraminidase. A similar result was observed at 5 and 10 days after damage in a model of traumatic injury and in the hippocampus and cerebral cortex in the in vivo kindling model of epilepsy. Furthermore, we defined that cortical astrocytes maintained in culture for long periods acquire markers of reactive gliosis and express SVCT2, in a similar way as previously observed in situ. Finally, by means of second harmonic generation and 2-photon fluorescence imaging, we analyzed brain necropsied material from patients with Alzheimer's disease (AD), which presented with an accumulation of amyloid plaques. Strikingly, although AD is characterized by focalized astrogliosis surrounding amyloid plaques, SVCT2 expression at the astroglial level was not detected. We conclude that SVCT2 is heterogeneously induced in reactive astrogliosis generated in different pathologies affecting the central nervous system (CNS).
Collapse
Affiliation(s)
- Katterine Salazar
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile.,Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepcion, Chile
| | - Fernando Martínez
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile.,Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepcion, Chile
| | - Margarita Pérez-Martín
- Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain
| | - Manuel Cifuentes
- Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain
| | - Laura Trigueros
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile.,Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain
| | - Luciano Ferrada
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Francisca Espinoza
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Natalia Saldivia
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Romina Bertinat
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Katherine Forman
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - María José Oviedo
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Antonio J López-Gambero
- Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain
| | - Christian Bonansco
- Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Gran Bretaña Avenida 1111, 2360102, Valparaíso, Chile
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois Chicago, Chicago, IL, USA.,Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Francisco Nualart
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile. .,Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepcion, Chile.
| |
Collapse
|
56
|
Lana D, Ugolini F, Nosi D, Wenk GL, Giovannini MG. Alterations in the Interplay between Neurons, Astrocytes and Microglia in the Rat Dentate Gyrus in Experimental Models of Neurodegeneration. Front Aging Neurosci 2017; 9:296. [PMID: 28955220 PMCID: PMC5601988 DOI: 10.3389/fnagi.2017.00296] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/29/2017] [Indexed: 11/13/2022] Open
Abstract
The hippocampus is negatively affected by aging and neurodegenerative diseases leading to impaired learning and memory abilities. A diverse series of progressive modifications in the intercellular communication among neurons, astrocytes and microglia occur in the hippocampus during aging or inflammation. A detailed understanding of the neurobiological modifications that contribute to hippocampal dysfunction may reveal new targets for therapeutic intervention. The current study focussed on the interplay between neurons and astroglia in the Granule Layer (GL) and the Polymorphic Layer (PL) of the Dentate Gyrus (DG) of adult, aged and LPS-treated rats. In GL and PL of aged and LPS-treated rats, astrocytes were less numerous than in adult rats. In GL of LPS-treated rats, astrocytes acquired morphological features of reactive astrocytes, such as longer branches than was observed in adult rats. Total and activated microglia increased in the aged and LPS-treated rats, as compared to adult rats. In the GL of aged and LPS-treated rats many neurons were apoptotic. Neurons decreased significantly in GL and PL of aged but not in rats treated with LPS. In PL of aged and LPS-treated rats many damaged neurons were embraced by microglia cells and were infiltrated by branches of astrocyte, which appeared to be bisecting the cell body, forming triads. Reactive microglia had a scavenging activity of dying neurons, as shown by the presence of neuronal debris within their cytoplasm. The levels of the chemokine fractalkine (CX3CL1) increased in hippocampal homogenates of aged rats and rats treated with LPS, and CX3CL1 immunoreactivity colocalized with activated microglia cells. Here we demonstrated that in the DG of aged and LPS-treated rats, astrocytes and microglia cooperate and participate in phagocytosis/phagoptosis of apoptotic granular neurons. The differential expression/activation of astroglia and the alteration of their intercommunication may be responsible for the different susceptibility of the DG in comparison to the CA1 and CA3 hippocampal areas to neurodegeneration during aging and inflammation.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of FlorenceFlorence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of FlorenceFlorence, Italy
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of FlorenceFlorence, Italy
| | - Gary L Wenk
- Department of Psychology, The Ohio State UniversityColumbus, OH, United States
| | - Maria G Giovannini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of FlorenceFlorence, Italy
| |
Collapse
|
57
|
Ereminas G, Majiene D, Sidlauskas K, Jakstas V, Ivanauskas L, Vaitiekaitis G, Liobikas J. Neuroprotective properties of anthocyanidin glycosides against H 2O 2-induced glial cell death are modulated by their different stability and antioxidant activity in vitro. Biomed Pharmacother 2017; 94:188-196. [PMID: 28759756 DOI: 10.1016/j.biopha.2017.07.077] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/31/2022] Open
Abstract
The neuroprotective effect of several anthocyanins in combination with their stability and antioxidant/pro-oxidant activity has been investigated against H2O2-induced oxidative stress in C6 glial cells. First it was found that delphinidin (Dp) 3-O-glucoside and 3-O-rutinoside were degraded within an hour, and at the same time stimulated the production of H2O2 in the micromolar concentration range. The stability of peonidin, pelargonidin (Pg), malvidin (Mv) and cyanidin (Cy) 3-O-glucosides and Cy 3-O-rutinoside was significantly higher than that of Dp 3-O-glycosides, with Pg3G showing the highest percent recovery over time. Based on these findings and chemical difference (according to the set of functional groups on the B-ring) of tested anthocyanins Cy3G, Mv3G and Pg3G were selected as candidates for the protection of glial cells against H2O2-induced oxidative stress. It was revealed that Cy3G (5-20μM) and Mv3G (10-20μM) but not Pg3G protected glial cells against H2O2-induced necrotic cell death. Moreover, these anthocyanins sustained the glutathione antioxidant defence system. Finally, to the extent of our knowledge we were the first to demonstrate the protective effect of Cy3G on the resting mitochondrial respiration rate in H2O2-affected glial cells. The results suggest that Cy3G, as the most prominent antioxidant among tested anthocyanins, could be a potential adjuvant for the prevention or reduction of necrotic glial cell death during the oxidative stress conditions met in neurodegenerative diseases. However, further elucidation of other possible mechanisms for anthocyanins to protect the nervous system is encouraged.
Collapse
Affiliation(s)
- Gedas Ereminas
- Laboratory of Biochemistry, Neuroscience Institute, Lithuanian University of Health Sciences, Eiveniu str. 4 LT, 50161, Kaunas, Lithuania
| | - Daiva Majiene
- Laboratory of Biochemistry, Neuroscience Institute, Lithuanian University of Health Sciences, Eiveniu str. 4 LT, 50161, Kaunas, Lithuania
| | - Kastytis Sidlauskas
- Laboratory of Biochemistry, Neuroscience Institute, Lithuanian University of Health Sciences, Eiveniu str. 4 LT, 50161, Kaunas, Lithuania
| | - Valdas Jakstas
- Department of Pharmacognosy, Faculty of Pharmacy, Lithuanian University of Health Sciences, Eiveniu str. 4, LT, 50161 Kaunas, Lithuania
| | - Liudas Ivanauskas
- Department of Analytical and Toxicological Chemistry, Faculty of Pharmacy, Lithuanian University of Health Sciences, Eiveniu str. 4, LT, 50161, Kaunas, Lithuania
| | - Gintautas Vaitiekaitis
- Department of Physics, Mathematics and Biophysics, Faculty of Medicine, Lithuanian University of Health Sciences, Eiveniu str. 4, LT, 50161, Kaunas, Lithuania
| | - Julius Liobikas
- Laboratory of Biochemistry, Neuroscience Institute, Lithuanian University of Health Sciences, Eiveniu str. 4 LT, 50161, Kaunas, Lithuania.
| |
Collapse
|
58
|
French JA, Koepp M, Naegelin Y, Vigevano F, Auvin S, Rho JM, Rosenberg E, Devinsky O, Olofsson PS, Dichter MA. Clinical studies and anti-inflammatory mechanisms of treatments. Epilepsia 2017; 58 Suppl 3:69-82. [PMID: 28675558 PMCID: PMC5679081 DOI: 10.1111/epi.13779] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2017] [Indexed: 02/06/2023]
Abstract
In this exciting era, we are coming closer and closer to bringing an anti-inflammatory therapy to the clinic for the purpose of seizure prevention, modification, and/or suppression. At present, it is unclear what this approach might entail, and what form it will take. Irrespective of the therapy that ultimately reaches the clinic, there will be some commonalities with regard to clinical trials. A number of animal models have now been used to identify inflammation as a major underlying mechanism of both chronic seizures and the epileptogenic process. These models have demonstrated that specific anti-inflammatory treatments can be effective at both suppressing chronic seizures and interfering with the process of epileptogenesis. Some of these have already been evaluated in early phase clinical trials. It can be expected that there will soon be more clinical trials of both "conventional, broad spectrum" anti-inflammatory agents and novel new approaches to utilizing specific anti-inflammatory therapies with drugs or other therapeutic interventions. A summary of some of those approaches appears below, as well as a discussion of the issues facing clinical trials in this new domain.
Collapse
Affiliation(s)
- Jacqueline A. French
- Comprehensive Epilepsy Center, NYU Langone School of Medicine, New York City, New York, U.S.A
| | - Matthias Koepp
- Institute of Neurology, University College London, London, United Kingdom
| | - Yvonne Naegelin
- Department of Neurology, University Hospital Basel, Basel, Switzerland
| | - Federico Vigevano
- Neurology Unit, Department of Neuroscience, Bambino Gesù Children Hospital, Rome, Italy
| | - Stéphane Auvin
- Pediatric Neurology, Robert Debré University Hospital, Paris, France
| | - Jong M. Rho
- Alberta Children’s Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Evan Rosenberg
- Comprehensive Epilepsy Center, NYU Langone School of Medicine, New York City, New York, U.S.A
| | - Orrin Devinsky
- Comprehensive Epilepsy Center, NYU Langone School of Medicine, New York City, New York, U.S.A
| | - Peder S. Olofsson
- Center for Bioelectronic Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marc A. Dichter
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| |
Collapse
|
59
|
Gabbi P, Ribeiro LR, Jessié Martins G, Cardoso AS, Haupental F, Rodrigues FS, Machado AK, Sperotto Brum J, Medeiros Frescura Duarte MM, Schetinger MRC, da Cruz IBM, Flávia Furian A, Oliveira MS, Dos Santos ARS, Royes LFF, Fighera MR, de Freitas ML. Methylmalonate Induces Inflammatory and Apoptotic Potential: A Link to Glial Activation and Neurological Dysfunction. J Neuropathol Exp Neurol 2017; 76:160-178. [PMID: 28395089 DOI: 10.1093/jnen/nlw121] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Methylmalonic acid (MMA) accumulates in tissues in methylmalonic acidemia, a heterogeneous group of inherited childhood diseases characterized by neurological dysfunction, oxidative stress and neuroinflammation; it is associated with degeneration of striatal neurons and cerebral cortical atrophy. It is presently unknown, however, whether transient exposure to MMA in the neonatal period is sufficient to trigger inflammatory and apoptotic processes that lead to brain structural damage. Here, newborn mice were given a single intracerebroventricular dose of MMA at 12 hours after birth. Maze testing of 21- and 40-day-old mice showed that MMA-injected animals exhibited deficit in the working memory test but not in the reference test. MMA-injected mice showed increased levels of the reactive oxygen species marker 2',7'-dichlorofluorescein diacetate, tumor necrosis factor, interleukin-1β, caspases 1, 3, and 8, and increased acetylcholinesterase activity in the cortex, hippocampus and striatum. This was associated with increased astrocyte and microglial immunoreactivity in all brain regions. These findings suggest that transient exposure to MMA may alter the redox state and cause neuroinflammatory/apoptotic processes and glial activation during critical periods of brain development. Similar processes may underlie brain dysfunction and cognitive impairment in patients with methylmalonic acidemia.
Collapse
Affiliation(s)
- Patricia Gabbi
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM.,Centro, de Departamento Neuropsiquiatria; de Ciências da Saúde, UFSM
| | - Leandro Rodrigo Ribeiro
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM
| | | | - Alexandra Seide Cardoso
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM.,Centro, de Departamento Neuropsiquiatria; de Ciências da Saúde, UFSM
| | - Fernanda Haupental
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM.,Centro, de Departamento Neuropsiquiatria; de Ciências da Saúde, UFSM
| | - Fernanda Silva Rodrigues
- Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM.,Centro, de Departamento Neuropsiquiatria; de Ciências da Saúde, UFSM.,de Programa Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica
| | - Alencar Kolinski Machado
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde
| | | | | | | | | | - Ana Flávia Furian
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde
| | - Mauro Schneider Oliveira
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde
| | - Adair Roberto Soares Dos Santos
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,de Programa Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica.,Universidade Federal de Santa Catarina, Centro, de Programa Pós-graduação em Neurociências, de Ciências Biológicas
| | - Luiz Fernando Freire Royes
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM.,de Programa Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica
| | - Michele Rechia Fighera
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM.,Centro, de Departamento Neuropsiquiatria; de Ciências da Saúde, UFSM.,de Programa Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica
| | | |
Collapse
|
60
|
Lana D, Ugolini F, Melani A, Nosi D, Pedata F, Giovannini MG. The neuron-astrocyte-microglia triad in CA3 after chronic cerebral hypoperfusion in the rat: Protective effect of dipyridamole. Exp Gerontol 2017; 96:46-62. [PMID: 28606482 DOI: 10.1016/j.exger.2017.06.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/07/2017] [Accepted: 06/08/2017] [Indexed: 12/01/2022]
Abstract
We investigated the quantitative and morphofunctional alterations of neuron-astrocyte-microglia triads in CA3 hippocampus, in comparison to CA1, after 2 Vessel Occlusion (2VO) and the protective effect of dipyridamole. We evaluated 3 experimental groups: sham-operated rats (sham, n=15), 2VO-operated rats treated with vehicle (2VO-vehicle, n=15), and 2VO-operated rats treated with dipyridamole from day 0 to day 7 (2VO-dipyridamole, n=15), 90days after 2VO. We analyzed Stratum Pyramidalis (SP), Stratum Lucidum (SL) and Stratum Radiatum (SR) of CA3. 1) ectopic neurons increased in SL and SR of 2VO-vehicle, and 2VO-dipyridamole rats; 2) apoptotic neurons increased in SP of 2VO-vehicle rats and dipyridamole reverted this effect; 3) astrocytes increased in SP, SL and SR of 2VO-vehicle and 2VO-dipyridamole rats; 4) TNF-α expression increased in astrocytes, blocked by dipyridamole, and in dendrites in SR of 2VO-vehicle rats; 5) total microglia increased in SL and SR of 2VO-vehicle and 2VO-dipyridamole rats; 6) triads increased in SR of 2VO-vehicle rats and dipyridamole reverted this effect. Microglia cooperated with astrocytes to phagocytosis of apoptotic neurons and debris, and engulfed ectopic non-fragmented neurons in SL of 2VO-vehicle and 2VO-dipyridamole rats, through a new mechanism called phagoptosis. CA3 showed a better adaptive capacity than CA1 to the ischemic insult, possibly due to the different behaviour of astrocytes and microglial cells. Dipyridamole had neuroprotective effects.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Filippo Ugolini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Alessia Melani
- Department of NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, 50139 Firenze, Italy.
| | - Felicita Pedata
- Department of NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| |
Collapse
|
61
|
Adamsky A, Goshen I. Astrocytes in Memory Function: Pioneering Findings and Future Directions. Neuroscience 2017; 370:14-26. [PMID: 28571720 DOI: 10.1016/j.neuroscience.2017.05.033] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/05/2017] [Accepted: 05/19/2017] [Indexed: 12/29/2022]
Abstract
Astrocytes have been generally believed to perform mainly homeostatic and supportive functions for neurons in the central nervous system. Recently, a growing body of evidence suggests previously unrecognized and surprising functions for astrocytes, including regulation of synaptic formation, transmission and plasticity, all of which are considered as the infrastructure for information processing and memory formation and stabilization. This review discusses the involvement of astrocytes in memory functions and the possible mechanisms that may underlie it. We review the important breakthroughs obtained in this field, as well as some of the controversies that arose from the past difficulty to manipulate these cells in a cell type-specific and non-invasive manner. Finally, we present new research avenues based on the advanced tools becoming available in recent years: optogenetics and chemogenetics, and the potential ways in which these tools may further illuminate the role of astrocytes in memory processes.
Collapse
Affiliation(s)
- Adar Adamsky
- Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University, Givat Ram, Jerusalem 91904, Israel
| | - Inbal Goshen
- Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University, Givat Ram, Jerusalem 91904, Israel.
| |
Collapse
|
62
|
Grings M, Moura AP, Parmeggiani B, Pletsch JT, Cardoso GMF, August PM, Matté C, Wyse ATS, Wajner M, Leipnitz G. Bezafibrate prevents mitochondrial dysfunction, antioxidant system disturbance, glial reactivity and neuronal damage induced by sulfite administration in striatum of rats: Implications for a possible therapeutic strategy for sulfite oxidase deficiency. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2135-2148. [PMID: 28529047 DOI: 10.1016/j.bbadis.2017.05.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 04/30/2017] [Accepted: 05/17/2017] [Indexed: 12/13/2022]
Abstract
Sulfite accumulates in tissues of patients affected by sulfite oxidase (SO) deficiency, a neurometabolic disease characterized by seizures and progressive encephalopathy, often resulting in early death. We investigated the effects of sulfite on mitochondrial function, antioxidant system, glial reactivity and neuronal damage in rat striatum, as well as the potential protective effects of bezafibrate on sulfite-induced toxicity. Thirty-day-old rats were intrastriatally administered with sulfite (2μmol) or NaCl (2μmol; control) and euthanized 30min after injection for evaluation of biochemical parameters and western blotting, or 7days after injection for analysis of glial reactivity and neuronal damage. Treatment with bezafibrate (30 or 100mg/kg/day) was performed by gavage during 7days before (pre-treatment) or after sulfite administration. Sulfite decreased creatine kinase and citrate synthase activities, mitochondrial mass, and PGC-1α nuclear content whereas bezafibrate pre-treatment prevented these alterations. Sulfite also diminished cytochrome c oxidase (COX) IV-1 content, glutathione levels and the activities of glutathione peroxidase (GPx), glutathione reductase (GR), glutathione S-transferase (GST) and glucose-6-phosphate dehydrogenase (G6PDH). On the other hand, catalase activity was increased by sulfite. Bezafibrate pre-treatment prevented the reduction of GPx, GR, GST and G6PDH activities. Finally, sulfite induced glial reactivity and neuronal damage, which were prevented by bezafibrate when administered before or after sulfite administration. Our findings provide strong evidence that sulfite induces neurotoxicity that leads to glial reactivity and neuronal damage. Since bezafibrate exerts neuroprotective effects against sulfite toxicity, it may be an attractive agent for the development of novel therapeutic strategies for SO-deficient patients.
Collapse
Affiliation(s)
- Mateus Grings
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Alana Pimentel Moura
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Belisa Parmeggiani
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Julia Tauana Pletsch
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Gabriela Miranda Fernandez Cardoso
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Pauline Maciel August
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Cristiane Matté
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Angela T S Wyse
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Moacir Wajner
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil; Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, CEP 90035-903 Porto Alegre, RS, Brazil
| | - Guilhian Leipnitz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil.
| |
Collapse
|
63
|
Li N, Zhang X, Dong H, Hu Y, Qian Y. Bidirectional relationship of mast cells-neurovascular unit communication in neuroinflammation and its involvement in POCD. Behav Brain Res 2017; 322:60-69. [PMID: 28082194 DOI: 10.1016/j.bbr.2017.01.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 12/31/2016] [Accepted: 01/02/2017] [Indexed: 11/26/2022]
Abstract
Postoperative cognitive dysfunction (POCD) has been hypothesized to be mediated by surgery-induced neuroinflammation, which is also a key element in the pathobiology of neurodegenerative diseases, stroke, and neuropsychiatric disorders. There is extensive communication between the immune system and the central nervous system (CNS). Inflammation resulting from activation of the innate immune system cells in the periphery can impact central nervous system behaviors, such as cognitive performance. Mast cells (MCs), as the"first responders" in the CNS, can initiate, amplify, and prolong other immune and nervous responses upon activation. In addition, MCs and their secreted mediators modulate inflammatory processes in multiple CNS pathologies and can thereby either contribute to neurological damage or confer neuroprotection. Neuroinflammation has been considered to be linked to neurovascular dysfunction in several neurological disorders. This review will provide a brief overview of the bidirectional relationship of MCs-neurovascular unit communication in neuroinflammation and its involvement in POCD, providing a new and unique therapeutic target for the adjuvant treatment of POCD.
Collapse
Affiliation(s)
- Nana Li
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| | - Xiang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| | - Hongquan Dong
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| | - Youli Hu
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| | - Yanning Qian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, PR China.
| |
Collapse
|
64
|
Verkhratsky A, Zorec R, Rodriguez JJ, Parpura V. Neuroglia: Functional Paralysis and Reactivity in Alzheimer’s Disease and Other Neurodegenerative Pathologies. ADVANCES IN NEUROBIOLOGY 2017; 15:427-449. [DOI: 10.1007/978-3-319-57193-5_17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
65
|
Cognitive dysfunction correlates with elevated serum S100B concentration in drug-free acutely relapsed patients with schizophrenia. Psychiatry Res 2017; 247:6-11. [PMID: 27863321 DOI: 10.1016/j.psychres.2016.09.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 09/04/2016] [Accepted: 09/20/2016] [Indexed: 02/07/2023]
Abstract
S100B, a biomarker of glial dysfunction and blood-brain barrier (BBB) disruption, has been proposed to be involved in the pathophysiology of schizophrenia. In the present study, we aimed at exploring the association of serum S100B levels with cognitive deficits using MATRICS Consensus Cognitive Battery (MCCB) in schizophrenia, by excluding the impact of antipsychotics. Sixty-two unmedicated patients with schizophrenia during their acute phases were divided into a drug-naïve group (n=34) and a drug-free group (n=28). S100B serum concentrations were measured and MCCB was administered to all of the patients. Forty healthy controls donated their blood samples for S100B assessment. The results indicated that serum S100B was significantly elevated in the drug-naive/free acute-stage schizophrenic patients when compared to the healthy controls. In the drug-free group, the serum S100B level was an independent contributor to the global cognitive dysfunctions, particularly for the speed of processing, attention/vigilance, visual learning and reasoning/problem solving subscores. Nevertheless, no significant associations between S100B and MCCB composite score or any cognitive domain subscore were observed in the drug-naïve group. These findings support the hypothesis that glial dysfunction and associated marker protein S100B may contribute to the pathophysiologic development of neurocognitive deficits in the relapsed individuals with schizophrenia.
Collapse
|
66
|
Selvaraj P, Xiao L, Lee C, Murthy SRK, Cawley NX, Lane M, Merchenthaler I, Ahn S, Loh YP. Neurotrophic Factor-α1: A Key Wnt-β-Catenin Dependent Anti-Proliferation Factor and ERK-Sox9 Activated Inducer of Embryonic Neural Stem Cell Differentiation to Astrocytes in Neurodevelopment. Stem Cells 2016; 35:557-571. [PMID: 27709799 DOI: 10.1002/stem.2511] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 08/08/2016] [Accepted: 09/06/2016] [Indexed: 12/31/2022]
Abstract
Embryonic neurodevelopment involves inhibition of proliferation of multipotent neural stem cells (NSCs) followed by differentiation into neurons, astrocytes and oligodendrocytes to form the brain. We have identified a new neurotrophic factor, NF-α1, which inhibits proliferation and promotes differentiation of NSC/progenitors derived from E13.5 mouse cortex. Inhibition of proliferation of these cells was mediated through negatively regulating the Wnt pathway and decreasing β-catenin. NF-α1 induced differentiation of NSCs to astrocytes by enhancing Glial Fibrillary Acidic Protein (GFAP) expression through activating the ERK1/2-Sox9 signaling pathway. Cultured E13.5 cortical stem cells from NF-α1-knockout mice showed decreased astrocyte numbers compared to wild-type mice, which was rescued by treatment with NF-α1. In vivo, immunocytochemistry of brain sections and Western blot analysis of neocortex of mice showed a gradual increase of NF-α1 expression from E14.5 to P1 and a surge of GFAP expression at P1, the time of increase in astrogenesis. Importantly, NF-α1-Knockout mice showed ∼49% fewer GFAP positive astrocytes in the neocortex compared to WT mice at P1. Thus, NF-α1 is critical for regulating antiproliferation and cell fate determination, through differentiating embryonic stem cells to GFAP-positive astrocytes for normal neurodevelopment. Stem Cells 2017;35:557-571.
Collapse
Affiliation(s)
| | - Lan Xiao
- Section on Cellular Neurobiology, Bethesda, Maryland, USA
| | - Cheol Lee
- Unit on Developmental Neurogenetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Niamh X Cawley
- Section on Cellular Neurobiology, Bethesda, Maryland, USA
| | - Malcolm Lane
- Department of Epidemiology and Public Health and Anatomy and Neurobiology, University of Maryland, Baltimore, Maryland, USA
| | - Istvan Merchenthaler
- Department of Epidemiology and Public Health and Anatomy and Neurobiology, University of Maryland, Baltimore, Maryland, USA
| | - Sohyun Ahn
- Unit on Developmental Neurogenetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Y Peng Loh
- Section on Cellular Neurobiology, Bethesda, Maryland, USA
| |
Collapse
|
67
|
Sun Q, Liao Y, Wang T, Wang G, Zhao F, Jin Y. Alteration in mitochondrial function and glutamate metabolism affected by 2-chloroethanol in primary cultured astrocytes. Toxicol In Vitro 2016; 37:50-60. [PMID: 27596522 DOI: 10.1016/j.tiv.2016.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 08/08/2016] [Accepted: 09/01/2016] [Indexed: 02/08/2023]
Abstract
The aim of this study was to explore the mechanisms that contribute to 1,2-dichloroethane (1,2-DCE) induced brain edema by focusing on alteration of mitochondrial function and glutamate metabolism in primary cultured astrocytes induced by 2-chloroethanol (2-CE), a metabolite of 1,2-DCE in vivo. The cells were exposed to different levels of 2-CE in the media for 24h. Mitochondrial function was evaluated by its membrane potential and intracellular contents of ATP, lactic acid and reactive oxygen species (ROS). Glutamate metabolism was indicated by expression of glutamine synthase (GS), glutamate-aspartate transporter (GLAST) and glutamate transporter-1 (GLT-1) at both protein and gene levels. Compared to the control group, exposure to 2-CE could cause a dose dependent damage in astrocytes, indicated by decreased cell viability and morphological changes, and supported by decreased levels of nonprotein sulfhydryl (NPSH) and inhibited activities of Na+/K+-ATPase and Ca2+-ATPase in the cells. The present study also revealed both mitochondrial function and glutamate metabolism in astrocytes were significantly disturbed by 2-CE. Of which, mitochondrial function was much vulnerable to the effects of 2-CE. In conclusion, our findings suggested that mitochondrial dysfunction and glutamate metabolism disorder could contribute to 2-CE-induced cytotoxicity in astrocytes, which might be related to 1,2-DCE-induced brain edema.
Collapse
Affiliation(s)
- Qi Sun
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yingjun Liao
- Department of Physiology, China Medical University, People's Republic of China
| | - Tong Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Gaoyang Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Fenghong Zhao
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yaping Jin
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
68
|
Boscia F, Begum G, Pignataro G, Sirabella R, Cuomo O, Casamassa A, Sun D, Annunziato L. Glial Na(+) -dependent ion transporters in pathophysiological conditions. Glia 2016; 64:1677-97. [PMID: 27458821 DOI: 10.1002/glia.23030] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/22/2016] [Accepted: 06/29/2016] [Indexed: 12/12/2022]
Abstract
Sodium dynamics are essential for regulating functional processes in glial cells. Indeed, glial Na(+) signaling influences and regulates important glial activities, and plays a role in neuron-glia interaction under physiological conditions or in response to injury of the central nervous system (CNS). Emerging studies indicate that Na(+) pumps and Na(+) -dependent ion transporters in astrocytes, microglia, and oligodendrocytes regulate Na(+) homeostasis and play a fundamental role in modulating glial activities in neurological diseases. In this review, we first briefly introduced the emerging roles of each glial cell type in the pathophysiology of cerebral ischemia, Alzheimer's disease, epilepsy, Parkinson's disease, Amyotrophic Lateral Sclerosis, and myelin diseases. Then, we discussed the current knowledge on the main roles played by the different glial Na(+) -dependent ion transporters, including Na(+) /K(+) ATPase, Na(+) /Ca(2+) exchangers, Na(+) /H(+) exchangers, Na(+) -K(+) -Cl(-) cotransporters, and Na(+) - HCO3- cotransporter in the pathophysiology of the diverse CNS diseases. We highlighted their contributions in cell survival, synaptic pathology, gliotransmission, pH homeostasis, and their role in glial activation, migration, gliosis, inflammation, and tissue repair processes. Therefore, this review summarizes the foundation work for targeting Na(+) -dependent ion transporters in glia as a novel strategy to control important glial activities associated with Na(+) dynamics in different neurological disorders. GLIA 2016;64:1677-1697.
Collapse
Affiliation(s)
- Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Gulnaz Begum
- Department of Neurology, University of Pittsburgh Medical School
| | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Rossana Sirabella
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Antonella Casamassa
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh Medical School.,Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, Pennsylvania, 15213
| | - Lucio Annunziato
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| |
Collapse
|
69
|
Lana D, Iovino L, Nosi D, Wenk GL, Giovannini MG. The neuron-astrocyte-microglia triad involvement in neuroinflammaging mechanisms in the CA3 hippocampus of memory-impaired aged rats. Exp Gerontol 2016; 83:71-88. [PMID: 27466072 DOI: 10.1016/j.exger.2016.07.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/23/2016] [Accepted: 07/20/2016] [Indexed: 01/08/2023]
Abstract
We examined the effects of inflammaging on memory encoding, and qualitative and quantitative modifications on proinflammatory proteins, apoptosis, neurodegeneration and morphological changes of neuron-astrocyte-microglia triads in CA3 Stratum Pyramidale (SP), Stratum Lucidum (SL) and Stratum Radiatum (SR) of young (3months) and aged rats (20months). Aged rats showed short-term memory impairments in the inhibitory avoidance task, increased expression of iNOS and activation of p38MAPK in SP, increase of apoptotic neurons in SP and of ectopic neurons in SL, and decrease of CA3 pyramidal neurons. The number of astrocytes and their branches length decreased in the three CA3 subregions of aged rats, with morphological signs of clasmatodendrosis. Total and activated microglia increased in the three CA3 subregions of aged rats. In aged rats CA3, astrocytes surrounded ectopic degenerating neurons forming "micro scars" around them. Astrocyte branches infiltrated the neuronal cell body, and, together with activated microglia formed "triads". In the triads, significantly more numerous in CA3 SL and SR of aged rats, astrocytes and microglia cooperated in fragmentation and phagocytosis of ectopic neurons. Inflammaging-induced modifications of astrocytes and microglia in CA3 of aged rats may help clearing neuronal debris derived from low-grade inflammation and apoptosis. These events might be common mechanisms underlying many neurodegenerative processes. The frequency to which they appear might depend upon, or might be the cause of, the burden and severity of neurodegeneration. Targeting the triads may represent a therapeutic strategy which may control inflammatory processes and spread of further cellular damage to neighboring cells.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Ludovica Iovino
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, Viale Morgagni 63 and Section of Anatomy and Histology, Largo Brambilla 3, University of Florence, 50134 Firenze, Italy.
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, Viale Morgagni 63 and Section of Anatomy and Histology, Largo Brambilla 3, University of Florence, 50134 Firenze, Italy.
| | - Gary L Wenk
- Department of Psychology, The Ohio State University, OH, USA..
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| |
Collapse
|
70
|
Skatchkov SN, Antonov SM, Eaton MJ. Glia and glial polyamines. Role in brain function in health and disease. BIOCHEMISTRY MOSCOW SUPPLEMENT SERIES A-MEMBRANE AND CELL BIOLOGY 2016. [DOI: 10.1134/s1990747816010116] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
71
|
Grygorowicz T, Wełniak-Kamińska M, Strużyńska L. Early P2X7R-related astrogliosis in autoimmune encephalomyelitis. Mol Cell Neurosci 2016; 74:1-9. [PMID: 26921791 DOI: 10.1016/j.mcn.2016.02.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 01/22/2016] [Accepted: 02/22/2016] [Indexed: 01/16/2023] Open
Abstract
Astrocytes are the main cells responsible for maintenance of brain homeostasis. Undisturbed action and signaling with other cells are crucial for proper functioning of the central nervous system (CNS). Dysfunctional astrocytes may determine the degree of neuronal injury and are associated with several brain pathologies, among which are multiple sclerosis (MS) and the animal model of this disease which is known as experimental autoimmune encephalomyelitis (EAE). One of the many functions of astrocytes is their response to CNS damage when they undergo reactive gliosis. Our data reveal that activation of astrocytes occurs in forebrains of immunized rats at a very early stage of EAE, well before the symptomatic phase of the disease. We have noted enhanced expression of GFAP and S100β starting from day 4 post-immunization. Temporal coincidence between the expression of astrocyte activation markers and the expression of connexin 43 and purinergic P2X7 receptor (P2X7R) was also observed. Administration of Brilliant blue G, an antagonist of P2X7R, significantly decreases astrogliosis as confirmed by immunohistochemical analysis and observation of decreased levels of GFAP and S100β. The condition of the treated animals was improved and the neurological symptoms of the disease were alleviated. With the knowledge that cerebral astroglia represent the main source of ATP and glutamate which are potentially neurotoxic substances released through P2X7R and connexin hemichannels, we suggest that astroglia may be involved in pathogenesis of MS/EAE at a very early stage through the purinergic/glutamatergic mechanisms.
Collapse
Affiliation(s)
- Tomasz Grygorowicz
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland
| | - Marlena Wełniak-Kamińska
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland
| | - Lidia Strużyńska
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland.
| |
Collapse
|
72
|
Harada K, Kamiya T, Tsuboi T. Gliotransmitter Release from Astrocytes: Functional, Developmental, and Pathological Implications in the Brain. Front Neurosci 2016; 9:499. [PMID: 26793048 PMCID: PMC4709856 DOI: 10.3389/fnins.2015.00499] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 12/15/2015] [Indexed: 12/20/2022] Open
Abstract
Astrocytes comprise a large population of cells in the brain and are important partners to neighboring neurons, vascular cells, and other glial cells. Astrocytes not only form a scaffold for other cells, but also extend foot processes around the capillaries to maintain the blood–brain barrier. Thus, environmental chemicals that exist in the blood stream could have potentially harmful effects on the physiological function of astrocytes. Although astrocytes are not electrically excitable, they have been shown to function as active participants in the development of neural circuits and synaptic activity. Astrocytes respond to neurotransmitters and contribute to synaptic information processing by releasing chemical transmitters called “gliotransmitters.” State-of-the-art optical imaging techniques enable us to clarify how neurotransmitters elicit the release of various gliotransmitters, including glutamate, D-serine, and ATP. Moreover, recent studies have demonstrated that the disruption of gliotransmission results in neuronal dysfunction and abnormal behaviors in animal models. In this review, we focus on the latest technical approaches to clarify the molecular mechanisms of gliotransmitter exocytosis, and discuss the possibility that exposure to environmental chemicals could alter gliotransmission and cause neurodevelopmental disorders.
Collapse
Affiliation(s)
- Kazuki Harada
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Tokyo, Japan
| | - Taichi Kamiya
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Tokyo, Japan
| | - Takashi Tsuboi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Tokyo, Japan
| |
Collapse
|
73
|
Astrocyte Dysfunction in Developmental Neurometabolic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:227-243. [PMID: 27714692 DOI: 10.1007/978-3-319-40764-7_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Astrocytes play crucial roles in maintaining brain homeostasis and in orchestrating neural development, all through tightly coordinated steps that cooperate to maintain the balance needed for normal development. Here, we review the alterations in astrocyte functions that contribute to a variety of developmental neurometabolic disorders and provide additional data on the predominant role of astrocyte dysfunction in the neurometabolic neurodegenerative disease glutaric acidemia type I. Finally, we describe some of the therapeutical approaches directed to neurometabolic diseases and discuss if astrocytes can be possible therapeutic targets for treating these disorders.
Collapse
|
74
|
Daniele S, Zappelli E, Martini C. Trazodone regulates neurotrophic/growth factors, mitogen-activated protein kinases and lactate release in human primary astrocytes. J Neuroinflammation 2015; 12:225. [PMID: 26627476 PMCID: PMC4666178 DOI: 10.1186/s12974-015-0446-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 11/25/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND In the central nervous system, glial cells provide metabolic and trophic support to neurons and respond to protracted stress and insults by up-regulating inflammatory processes. Reactive astrocytes and microglia are associated with the pathophysiology of neuronal injury, neurodegenerative diseases and major depression, in both animal models and human brains. Several studies have reported clear anti-inflammatory effects of anti-depressant treatment on astrocytes, especially in models of neurological disorders. Trazodone (TDZ) is a triazolopyridine derivative that is structurally unrelated to other major classes of antidepressants. Although the molecular mechanisms of TDZ in neurons have been investigated, it is unclear whether astrocytes are also a TDZ target. METHODS The effects of TDZ on human astrocytes were investigated in physiological conditions and following inflammatory insult with lipopolysaccharide (LPS) and tumour necrosis factor-α (TNF-α). Astrocytes were assessed for their responses to pro-inflammatory mediators and cytokines, and the receptors and signalling pathways involved in TDZ-mediated effects were evaluated. RESULTS TDZ had no effect on cell proliferation, but it decreased pro-inflammatory mediator release and modulated trophic and transcription factor mRNA expression. Following TDZ treatment, the AKT pathway was activated, whereas extracellular signal-regulated kinase and c-Jun NH2-terminal kinase were inhibited. Most importantly, a 72-h TDZ pre-treatment before inflammatory insult completely reversed the anti-proliferative effects induced by LPS-TNF-α. The expression or the activity of inflammatory mediators, including interleukin-6, c-Jun NH2-terminal kinase and nuclear factor κB, were also reduced. Furthermore, TDZ affected astrocyte metabolic support to neurons by counteracting the inflammation-mediated lactate decrease. Finally, TDZ protected neuronal-like cells against neurotoxicity mediated by activated astrocytes. These effects mainly involved an activation of 5-HT1A and an antagonism at 5-HT2A/C serotonin receptors. Fluoxetine, used in parallel, showed similar final effects nevertheless it activates different receptors/intracellular pathways. CONCLUSIONS Altogether, our results demonstrated that TDZ directly acts on astrocytes by regulating intracellular signalling pathways and increasing specific astrocyte-derived neurotrophic factor expression and lactate release. TDZ may contribute to neuronal support by normalizing trophic and metabolic support during neuroinflammation, which is associated with neurological diseases, including major depression.
Collapse
Affiliation(s)
- Simona Daniele
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, Pisa, 56126, PI, Italy.
| | - Elisa Zappelli
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, Pisa, 56126, PI, Italy.
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, Pisa, 56126, PI, Italy.
| |
Collapse
|
75
|
Vieira BDM, Radford RA, Chung RS, Guillemin GJ, Pountney DL. Neuroinflammation in Multiple System Atrophy: Response to and Cause of α-Synuclein Aggregation. Front Cell Neurosci 2015; 9:437. [PMID: 26778958 PMCID: PMC4700780 DOI: 10.3389/fncel.2015.00437] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 10/19/2015] [Indexed: 11/13/2022] Open
Abstract
Multiple system atrophy (MSA) is a progressive neurodegenerative disease presenting with combinations of autonomic dysfunction, parkinsonism, cerebellar ataxia and/or pyramidal signs. Oligodendroglial cytoplasmic inclusions (GCIs) rich in α-synuclein (α-syn) constitute the disease hallmark, accompanied by neuronal loss and activation of glial cells which indicate neuroinflammation. Recent studies demonstrate that α-syn may be released from degenerating neurons to mediate formation of abnormal inclusion bodies and to induce neuroinflammation which, interestingly, might also favor the formation of intracellular α-syn aggregates as a consequence of cytokine release and the shift to a pro-inflammatory environment. Here, we critically review the relationships between α-syn and astrocytic and microglial activation in MSA to explore the potential of therapeutics which target neuroinflammation.
Collapse
Affiliation(s)
| | - Rowan A Radford
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Roger S Chung
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Gilles J Guillemin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Dean L Pountney
- Menzies Health Institute Queensland, Griffith University Gold Coast, QLD, Australia
| |
Collapse
|
76
|
Mohn TC, Koob AO. Adult Astrogenesis and the Etiology of Cortical Neurodegeneration. J Exp Neurosci 2015; 9:25-34. [PMID: 26568684 PMCID: PMC4634839 DOI: 10.4137/jen.s25520] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/06/2015] [Accepted: 10/08/2015] [Indexed: 01/09/2023] Open
Abstract
As more evidence points to a clear role for astrocytes in synaptic processing, synaptogenesis and cognition, continuing research on astrocytic function could lead to strategies for neurodegenerative disease prevention. Reactive astrogliosis results in astrocyte proliferation early in injury and disease states and is considered neuroprotective, indicating a role for astrocytes in disease etiology. This review describes the different types of human cortical astrocytes and the current evidence regarding adult cortical astrogenesis in injury and degenerative disease. A role for disrupted astrogenesis as a cause of cortical degeneration, with a focus on the tauopathies and synucleinopathies, will also be considered.
Collapse
Affiliation(s)
- Tal C. Mohn
- Biology Department, University of Wisconsin—River Falls, River Falls, Wisconsin, USA
| | - Andrew O. Koob
- Biology Department, University of Wisconsin—River Falls, River Falls, Wisconsin, USA
| |
Collapse
|
77
|
Olivera-Bravo S, Barbeito L. A role of astrocytes in mediating postnatal neurodegeneration in Glutaric acidemia-type 1. FEBS Lett 2015; 589:3492-7. [DOI: 10.1016/j.febslet.2015.09.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/02/2015] [Accepted: 09/08/2015] [Indexed: 01/12/2023]
|
78
|
Lim SY, Mah W. Abnormal Astrocytosis in the Basal Ganglia Pathway of Git1(-/-) Mice. Mol Cells 2015; 38:540-7. [PMID: 25997734 PMCID: PMC4469912 DOI: 10.14348/molcells.2015.0041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 03/02/2015] [Accepted: 03/02/2015] [Indexed: 11/27/2022] Open
Abstract
Attention deficit/hyperactivity disorder (ADHD) is one of the most common neurodevelopmental disorders, affecting approximately 5% of children. However, the neural mechanisms underlying its development and treatment are yet to be elucidated. In this study, we report that an ADHD mouse model, which harbors a deletion in the Git1 locus, exhibits severe astrocytosis in the globus pallidus (GP) and thalamic reticular nucleus (TRN), which send modulatory GABAergic inputs to the thalamus. A moderate level of astrocytosis was displayed in other regions of the basal ganglia pathway, including the ventrobasal thalamus and cortex, but not in other brain regions, such as the caudate putamen, basolateral amygdala, and hippocampal CA1. This basal ganglia circuit-selective astrocytosis was detected in both in adult (2-3 months old) and juvenile (4 weeks old) Git1(-/-) mice, suggesting a developmental origin. Astrocytes play an active role in the developing synaptic circuit; therefore, we performed an immunohistochemical analysis of synaptic markers. We detected increased and decreased levels of GABA and parvalbumin (PV), respectively, in the GP. This suggests that astrocytosis may alter synaptic transmission in the basal ganglia. Intriguingly, increased GABA expression colocalized with the astrocyte marker, GFAP, indicative of an astrocytic origin. Collectively, these results suggest that defects in basal ganglia circuitry, leading to impaired inhibitory modulation of the thalamus, are neural correlates for the ADHD-associated behavioral manifestations in Git1(-/-) mice.
Collapse
Affiliation(s)
- Soo-Yeon Lim
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412,
Korea
| | - Won Mah
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412,
Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| |
Collapse
|
79
|
Zhao W, Jiang B, Hu H, Zhang S, Lv S, Yuan J, Qian Y, Zou Y, Li X, Jiang H, Liu F, Shao C, Gong Y. Lack of CUL4B leads to increased abundance of GFAP-positive cells that is mediated by PTGDS in mouse brain. Hum Mol Genet 2015; 24:4686-97. [PMID: 26025376 DOI: 10.1093/hmg/ddv200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 05/26/2015] [Indexed: 01/05/2023] Open
Abstract
Astrocytes are the most abundant cell type in the mammalian brain and are important for the functions of the central nervous system. Glial fibrillary acidic protein (GFAP) is regarded as a hallmark of mature astrocytes, though some GFPA-positive cells may act as neural stem cells. Missense heterozygous mutations in GFAP cause Alexander disease that manifests leukodystrophy and intellectual disability. Here, we show that CUL4B, a scaffold protein that assembles E3 ubiquitin ligase, represses the expression of GFAP in neural progenitor cells (NPCs) during brain development. Lack of Cul4b in NPCs in cultures led to increased generation of astrocytes, marked by GFAP and S100β. The GFAP+ cells were also found to be more abundant in the brains of nervous system-specific Cul4b knockout mice in vivo. Moreover, we demonstrated that the increased generation of GFAP+ cells from Cul4b-null NPCs was mediated by an upregulation of prostaglandin D2 synthase PTGDS. We showed that the increased GFAP expression can be attenuated by pharmacological inhibition of the PTGDS enzymatic activity or by shRNA-mediated knockdown of Ptgds. Importantly, exogenously added PTGDS could promote the generation of GFAP+ cells from wild-type NPCs. We further observed that Ptgds is targeted and repressed by the CUL4B/PRC2 complex. Together, our results demonstrate CUL4B as a negative regulator of GFAP expression during neural development.
Collapse
Affiliation(s)
- Wei Zhao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Baichun Jiang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Huili Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Shuqian Zhang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Shuaishuai Lv
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Jupeng Yuan
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Yanyan Qian
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Xi Li
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Hong Jiang
- Institute of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong 250012, China and
| | - Fang Liu
- Department of Neuroscience, Centre for Addiction and Mental Health, Toronto, Ontario, Canada M5T 1R8
| | - Changshun Shao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China,
| | - Yaoqin Gong
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China,
| |
Collapse
|
80
|
Kong H, Yin F, He F, Omran A, Li L, Wu T, Wang Y, Peng J. The Effect of miR-132, miR-146a, and miR-155 on MRP8/TLR4-Induced Astrocyte-Related Inflammation. J Mol Neurosci 2015; 57:28-37. [PMID: 25957996 DOI: 10.1007/s12031-015-0574-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 04/28/2015] [Indexed: 01/20/2023]
Abstract
Astrocyte activation, associated with the release of pro-inflammatory cytokines interleukin 1-β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor alpha (TNF-α), is a hallmark of multiple brain diseases, including mesial temporal lobe epilepsy. In recent years, several microRNAs have emerged as important controllers of Toll-like receptor (TLR) signaling. In this study, we investigated the effect of miR-132, miR-146a, and miR-155 on myeloid-related protein-8 (MRP8) induced astrocyte-related inflammation. Using quantitative polymerase chain reaction (qPCR) and western blot, we found clear upregulation of TLR4 and downstream inflammatory cytokines, along with dysregulation of miR-132, miR-146a, and miR-155 in in vitro astrocytes after exposing them to different concentrations of MRP8. In addition, we focused on the effect of miR-132 on astrocyte-related inflammation induced by MRP8 via lentiviral infection then evaluated the expression of its possible target genes: acetylcholinesterase (AChE) and interleukin-1 receptor-associated kinase (IRAK4). Our results show that miR-132 is a negative feedback regulator of IL-1β and IL-6, but not TNF-α, by targeting IRAK4. Together, our findings demonstrate the novel role of TLR4-related microRNAs, especially miR-132, in the regulation of MRP8-induced astrocyte activation and highlight the importance of miR-132 in the modulation of innate immune response induced by endogenous ligands in neurological diseases.
Collapse
Affiliation(s)
- Huimin Kong
- Department of Pediatrics, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, China
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Abstract
The most accredited (and fashionable) hypothesis of the pathogenesis of Alzheimer Disease (AD) sees accumulation of β-amyloid protein in the brain (in both soluble and insoluble forms) as a leading mechanism of neurotoxicity. How β-amyloid triggers the neurodegenerative disorder is at present unclear, but growing evidence suggests that a deregulation of Ca(2+) homeostasis and deficient Ca(2+) signalling may represent a fundamental pathogenic factor. Given that symptoms of AD are most likely linked to synaptic dysfunction (at the early stages) followed by neuronal loss (at later and terminal phases of the disease), the effects of β-amyloid have been mainly studied in neurones. Yet, it must be acknowledged that neuroglial cells, including astrocytes, contribute to pathological progression of most (if not all) neurological diseases. Here, we review the literature pertaining to changes in Ca(2+) signalling in astrocytes exposed to exogenous β-amyloid or in astrocytes from transgenic Alzheimer disease animals models, characterized by endogenous β-amyloidosis. Accumulated experimental data indicate deregulation of Ca(2+) homeostasis and signalling in astrocytes in AD, which should be given full pathogenetic consideration. Further studies are warranted to comprehend the role of deficient astroglial Ca(2+) signalling in the disease progression.
Collapse
|
82
|
Biesmans S, Acton PD, Cotto C, Langlois X, Ver Donck L, Bouwknecht JA, Aelvoet SA, Hellings N, Meert TF, Nuydens R. Effect of stress and peripheral immune activation on astrocyte activation in transgenic bioluminescent Gfap-luc mice. Glia 2015; 63:1126-37. [PMID: 25690758 DOI: 10.1002/glia.22804] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 01/22/2015] [Indexed: 12/13/2022]
Abstract
Neuroinflammation and the accompanying activation of glial cells is an important feature of many neurodegenerative conditions. It is known that factors such as peripheral infections and stress can influence immune processes in the brain. However, the effect of these stressors on astrocyte activation in vivo remains elusive. In this study, transgenic Gfap-luc mice expressing the luciferase gene under the transcriptional control of the glial fibrillary acidic protein promoter were used to quantify the kinetics of in vivo astrocyte activation following immune challenges relevant to clinical inflammation. It was found that astrocytes respond rapidly to peripheral immune activation elicited by either bacterial lipopolysaccharide (LPS) or the viral mimetic polyinosinic:polycytidylic acid (poly(I:C)). By measuring bioluminescence and 18-kDa translocator protein radioligand binding in the same animal it was observed that LPS induces both astrocyte as well as microglial activation at 6 h post-administration. Furthermore, the astrocyte response decreased upon repeated systemic LPS injections, indicating development of tolerance to the LPS challenge. Finally, restraining Gfap-luc mice for 1 h daily on 5 consecutive days did not affect brain bioluminescence, thereby indicating that sub-chronic stress does not influence astrocyte activation under unchallenged conditions. However, stressed animals showed a reduced response to a subsequent systemic LPS injection, suggesting that the immune system is compromised in these animals. Here, we demonstrate that Gfap-luc mice can be used to study astrocyte activation in response to stimuli relevant for clinical inflammation and that this approach may provide a more complete characterization of existing and novel models of neuroinflammation
Collapse
Affiliation(s)
- Steven Biesmans
- Biomedical Research Institute, Hasselt University, Agoralaan C Building, Diepenbeek, Belgium; Neurosciences, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Cross-talk between neurons and astrocytes in response to bilirubin: adverse secondary impacts. Neurotox Res 2015; 26:1-15. [PMID: 24122290 DOI: 10.1007/s12640-013-9427-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 09/23/2013] [Accepted: 09/24/2013] [Indexed: 12/25/2022]
Abstract
Previous studies using monotypic nerve cell cultures have shown that bilirubin-induced neurological dysfunction (BIND) involves apoptosis and necrosis-like cell death, following neuritic atrophy and astrocyte activation,and that glycoursodeoxycholic acid (GUDCA) has therapeutic efficacy against BIND. Cross-talk between neurons and astrocytes may protect or aggravate neurotoxicity by unconjugated bilirubin (UCB). In a previous work we have shown that bidirectional signaling during astrocyte-neuron recognition attenuates neuronal damage by UCB. Here, we investigated whether the establishment of neuron-astrocyte homeostasis prior to cell exposure to UCB was instead associated with a lower resistance of neurons to UCB toxicity, and if the pro-survival properties of GUDCA were replicated in that experimental model. We have introduced a 24 h adaptation period for neuron-glia communication prior to the 48 h treatment with UCB. In such conditions, UCB induced glial activation, which aggravated neuronal damage, comprising increased apoptosis,cell demise and neuritic atrophy, which were completely prevented in the presence of GUDCA. Neuronal multidrug resistance-associated protein 1 expression and tumor necrosis factor-a secretion, although unchanged by UCB, increased in the presence of astrocytes. The rise in S100B and nitric oxide in the co-cultures medium may have contributed to UCB neurotoxicity. Since the levels of these diffusible molecules did not change by GUDCA we may assume that they are not directly involved in its beneficial effects. Data indicate that astrocytes, in an indirect neuron-astrocyte co-culture model and after homeostatic setting regulation of the system, are critically influencing neurodegeneration by UCB, and support GUDCA for the prevention of BIND.
Collapse
|
84
|
Prion infection of mouse brain reveals multiple new upregulated genes involved in neuroinflammation or signal transduction. J Virol 2014; 89:2388-404. [PMID: 25505076 DOI: 10.1128/jvi.02952-14] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
UNLABELLED Gliosis is often a preclinical pathological finding in neurodegenerative diseases, including prion diseases, but the mechanisms facilitating gliosis and neuronal damage in these diseases are not understood. To expand our knowledge of the neuroinflammatory response in prion diseases, we assessed the expression of key genes and proteins involved in the inflammatory response and signal transduction in mouse brain at various times after scrapie infection. In brains of scrapie-infected mice at pre- and postclinical stages, we identified 15 previously unreported differentially expressed genes related to inflammation or activation of the STAT signal transduction pathway. Levels for the majority of differentially expressed genes increased with time postinfection. In quantitative immunoblotting experiments of STAT proteins, STAT1α, phosphorylated-STAT1α (pSTAT1α), and pSTAT3 were increased between 94 and 131 days postinfection (p.i.) in brains of mice infected with strain 22L. Furthermore, a select group of STAT-associated genes was increased preclinically during scrapie infection, suggesting early activation of the STAT signal transduction pathway. Comparison of inflammatory markers between mice infected with scrapie strains 22L and RML indicated that the inflammatory responses and gene expression profiles in the brains were strikingly similar, even though these scrapie strains infect different brain regions. The endogenous interleukin-1 receptor antagonist (IL-1Ra), an inflammatory marker, was newly identified as increasing preclinically in our model and therefore might influence scrapie pathogenesis in vivo. However, in IL-1Ra-deficient or overexpressor transgenic mice inoculated with scrapie, neither loss nor overexpression of IL-1Ra demonstrated any observable effect on gliosis, protease-resistant prion protein (PrPres) formation, disease tempo, pathology, or expression of the inflammatory genes analyzed. IMPORTANCE Prion infection leads to PrPres deposition, gliosis, and neuroinflammation in the central nervous system before signs of clinical illness. Using a scrapie mouse model of prion disease to assess various time points postinoculation, we identified 15 unreported genes that were increased in the brains of scrapie-infected mice and were associated with inflammation and/or JAK-STAT activation. Comparison of mice infected with two scrapie strains (22L and RML), which have dissimilar neuropathologies, indicated that the inflammatory responses and gene expression profiles in the brains were similar. Genes that increased prior to clinical signs might be involved in controlling scrapie infection or in facilitating damage to host tissues. We tested the possible role of the endogenous IL-1Ra, which was increased at 70 days p.i. In scrapie-infected mice deficient in or overexpressing IL-1Ra, there was no observable effect on gliosis, PrPres formation, disease tempo, pathology, or expression of inflammatory genes analyzed.
Collapse
|
85
|
Abstract
This review focuses on the roles of glia and polyamines (PAs) in brain function and dysfunction, highlighting how PAs are one of the principal differences between glia and neurons. The novel role of PAs, such as putrescine, spermidine, and spermine and their precursors and derivatives, is discussed. However, PAs have not yet been a focus of much glial research. They affect many neuronal and glial receptors, channels, and transporters. They are therefore key elements in the development of many diseases and syndromes, thus forming the rationale for PA-focused and glia-focused therapy for these conditions.
Collapse
Affiliation(s)
- Serguei N Skatchkov
- Department of Biochemistry, School of Medicine, Universidad, Central del Caribe, PO Box 60-327, Bayamón, PR 00960-6032, USA; Department of Physiology, School of Medicine, Universidad, Central del Caribe, PO Box 60-327, Bayamón, PR 00960-6032, USA.
| | - Michel A Woodbury-Fariña
- Department of Psychiatry, University of Puerto Rico School of Medicine, 307 Calle Eleonor Roosevelt, San Juan, PR 00918-2720, USA
| | - Misty Eaton
- Department of Biochemistry, School of Medicine, Universidad, Central del Caribe, PO Box 60-327, Bayamón, PR 00960-6032, USA
| |
Collapse
|
86
|
Lana D, Melani A, Pugliese AM, Cipriani S, Nosi D, Pedata F, Giovannini MG. The neuron-astrocyte-microglia triad in a rat model of chronic cerebral hypoperfusion: protective effect of dipyridamole. Front Aging Neurosci 2014; 6:322. [PMID: 25505884 PMCID: PMC4245920 DOI: 10.3389/fnagi.2014.00322] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 11/04/2014] [Indexed: 11/13/2022] Open
Abstract
Chronic cerebral hypoperfusion during aging may cause progressive neurodegeneration as ischemic conditions persist. Proper functioning of the interplay between neurons and glia is fundamental for the functional organization of the brain. The aim of our research was to study the pathophysiological mechanisms, and particularly the derangement of the interplay between neurons and astrocytes-microglia with the formation of "triads," in a model of chronic cerebral hypoperfusion induced by the two-vessel occlusion (2VO) in adult Wistar rats (n = 15). The protective effect of dipyridamole given during the early phases after 2VO (4 mg/kg/day i.v., the first 7 days after 2VO) was verified (n = 15). Sham-operated rats (n = 15) were used as controls. Immunofluorescent triple staining of neurons (NeuN), astrocytes (GFAP), and microglia (IBA1) was performed 90 days after 2VO. We found significantly higher amount of "ectopic" neurons, neuronal debris and apoptotic neurons in CA1 Str. Radiatum and Str. Pyramidale of 2VO rats. In CA1 Str. Radiatum of 2VO rats the amount of astrocytes (cells/mm(2)) did not increase. In some instances several astrocytes surrounded ectopic neurons and formed a "micro scar" around them. Astrocyte branches could infiltrate the cell body of ectopic neurons, and, together with activated microglia cells formed the "triads." In the triad, significantly more numerous in CA1 Str. Radiatum of 2VO than in sham rats, astrocytes and microglia cooperated in the phagocytosis of ectopic neurons. These events might be common mechanisms underlying many neurodegenerative processes. The frequency to which they appear might depend upon, or might be the cause of, the burden and severity of neurodegeneration. Dypiridamole significantly reverted all the above described events. The protective effect of chronic administration of dipyridamole might be a consequence of its vasodilatory, antioxidant and anti-inflammatory role during the early phases after 2VO.
Collapse
Affiliation(s)
- Daniele Lana
- Section of Pharmacology and Clinical Oncology, Department of Health Sciences, University of Florence Florence, Italy
| | - Alessia Melani
- Section of Pharmacology and Toxicology, Department of NEUROFARBA, University of Florence Florence, Italy
| | - Anna Maria Pugliese
- Section of Pharmacology and Toxicology, Department of NEUROFARBA, University of Florence Florence, Italy
| | | | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence Florence, Italy
| | - Felicita Pedata
- Section of Pharmacology and Toxicology, Department of NEUROFARBA, University of Florence Florence, Italy
| | - Maria Grazia Giovannini
- Section of Pharmacology and Clinical Oncology, Department of Health Sciences, University of Florence Florence, Italy
| |
Collapse
|
87
|
Peng L, Parpura V, Verkhratsky A. EDITORIAL Neuroglia as a Central Element of Neurological Diseases: An Underappreciated Target for Therapeutic Intervention. Curr Neuropharmacol 2014; 12:303-7. [PMID: 25342938 PMCID: PMC4207070 DOI: 10.2174/1570159x12999140829152550] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neuroglia of the central nervous system (CNS), represented by cells of neural (astrocytes, oligodendrocytes and NG2 glial cells) and myeloid (microglia) origins are fundamental for homeostasis of the nervous tissue. Astrocytes are critical for the development of the CNS, they are indispensable for synaptogenesis, and they define structural organisation of the nervous tissue, as well as the generation and maintenance of CNS-blood and cerebrospinal fluid-blood barriers. Astroglial cells control homeostasis of ions and neurotransmitters and provide neurones with metabolic support. Oligodendrocytes, through the process of myelination, as well as by homoeostatic support of axons provide for interneuronal connectivity. The NG2 cells receive direct synaptic inputs, and might be important elements of adult remyelination. Microglial cells, which originate from foetal macrophages invading the brain early in embryogenesis, shape the synaptic connections through removing of redundant synapses and phagocyting apoptotic neurones. Neuroglia also form the defensive system of the CNS through complex and context-specific programmes of activation, known as reactive gliosis. Many neurological diseases are associated with neurogliopathologies represented by asthenic and atrophic changes in glial cells that, through the loss or diminution of their homeostatic and defensive functions, assist evolution of pathology. Conceptually, neurological and psychiatric disorders can be regarded as failures of neuroglial homeostatic/
defensive responses, and, hence, glia represent a (much underappreciated) target for therapeutic intervention.
Collapse
Affiliation(s)
- Liang Peng
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, P. R. China
| | - Vladimir Parpura
- Department of Neurobiology, Center for Glial Biology in Medicine, Atomic Force Microscopy & Nanotechnology Laboratories, Civitan International Research Center, Evelyn F. McKnight Brain Institute, University of Alabama, Birmingham, AL 35294, USA ; Department of Biotechnology, University or Rijeka, 51000 Rijeka, Croatia
| | - Alexei Verkhratsky
- Faculty of Life Science, The University of Manchester, Manchester, UK ; Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain ; University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| |
Collapse
|
88
|
Tao C, Zhang X. Development of astrocytes in the vertebrate eye. Dev Dyn 2014; 243:1501-10. [PMID: 25236977 DOI: 10.1002/dvdy.24190] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 08/22/2014] [Accepted: 09/12/2014] [Indexed: 02/04/2023] Open
Abstract
Astrocytes represent the earliest glial population in the embryonic optic nerve, contributing critically to retinal angiogenesis and formation of brain-retinal-barrier. Despite of many developmental and clinical implications of astrocytes, answers to some of the most fundamental questions of this unique type of glial cells remain elusive. This review provides an overview of the current knowledge about the origination, proliferation, and differentiation of astrocytes, their journey from the optic nerve toward the neuroretina, and their involvement in physiological and pathological development of the visual system.
Collapse
Affiliation(s)
- Chenqi Tao
- Stark Neuroscience Institute, Indiana University School of Medicine, Indianapolis, Indiana; Departments of Ophthalmology, Pathology, and Cell Biology, Columbia University, New York, New York
| | | |
Collapse
|
89
|
Methamphetamine alters the normal progression by inducing cell cycle arrest in astrocytes. PLoS One 2014; 9:e109603. [PMID: 25290377 PMCID: PMC4188627 DOI: 10.1371/journal.pone.0109603] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/11/2014] [Indexed: 12/20/2022] Open
Abstract
Methamphetamine (MA) is a potent psychostimulant with a high addictive capacity, which induces many deleterious effects on the brain. Chronic MA abuse leads to cognitive dysfunction and motor impairment. MA affects many cells in the brain, but the effects on astrocytes of repeated MA exposure is not well understood. In this report, we used Gene chip array to analyze the changes in the gene expression profile of primary human astrocytes treated with MA for 3 days. Range of genes were found to be differentially regulated, with a large number of genes significantly downregulated, including NEK2, TTK, TOP2A, and CCNE2. Gene ontology and pathway analysis showed a highly significant clustering of genes involved in cell cycle progression and DNA replication. Further pathway analysis showed that the genes downregulated by multiple MA treatment were critical for G2/M phase progression and G1/S transition. Cell cycle analysis of SVG astrocytes showed a significant reduction in the percentage of cell in the G2/M phase with a concomitant increase in G1 percentage. This was consistent with the gene array and validation data, which showed that repeated MA treatment downregulated the genes associated with cell cycle regulation. This is a novel finding, which explains the effect of MA treatment on astrocytes and has clear implication in neuroinflammation among the drug abusers.
Collapse
|
90
|
Abstract
Exclusively neuron-centric approaches to neuropathological mechanisms have not resulted in major new breakthroughs in the prevention and therapy of neurodegenerative diseases. In the present paper, we review the role of glia in neurodegeneration in an attempt to identify novel targets that could be used to develop much-needed strategies for the containment and cure of neurodegenerative disorders. We discuss this in the context of glial roles in the homoeostasis and defence of the brain. We consider the mounting evidence supporting a change away from the perception of reactive glial responses merely as secondary detrimental processes that exacerbate the course of neurological disorders, in favour of an emerging contemporary view of glial pathological responses as complex and multistaged defensive processes that also have the potential for dysfunction.
Collapse
|
91
|
Lucchinetti CF, Guo Y, Popescu BFG, Fujihara K, Itoyama Y, Misu T. The pathology of an autoimmune astrocytopathy: lessons learned from neuromyelitis optica. Brain Pathol 2014; 24:83-97. [PMID: 24345222 DOI: 10.1111/bpa.12099] [Citation(s) in RCA: 286] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 12/13/2022] Open
Abstract
Neuromyelitis optica (NMO) is a disabling autoimmune astrocytopathy characterized by typically severe and recurrent attacks of optic neuritis and longitudinally extensive myelitis. Until recently, NMO was considered an acute aggressive variant of multiple sclerosis (MS), despite the fact that early studies postulated that NMO and MS may be two distinct diseases with a common clinical picture. With the discovery of a highly specific serum autoantibody (NMO-IgG), Lennon and colleagues provided the first unequivocal evidence distinguishing NMO from MS and other central nervous system (CNS) inflammatory demyelinating disorders. The target antigen of NMO-IgG was confirmed to be aquaporin-4 (AQP4), the most abundant water channel protein in the CNS, mainly expressed on astrocytic foot processes at the blood-brain barrier, subpial and subependymal regions. Pathological studies demonstrated that astrocytes were selectively targeted in NMO as evidenced by the extensive loss of immunoreactivities for the astrocytic proteins, AQP4 and glial fibrillary acidic protein (GFAP), as well as perivascular deposition of immunoglobulins and activation of complement even within lesions with a relative preservation of myelin. In support of these pathological findings, GFAP levels in the cerebrospinal fluid (CSF) during acute NMO exacerbations were found to be remarkably elevated in contrast to MS where CSF-GFAP levels did not substantially differ from controls. Additionally, recent experimental studies showed that AQP4 antibody is pathogenic, resulting in selective astrocyte destruction and dysfunction in vitro, ex vivo and in vivo. These findings strongly suggest that NMO is an autoimmune astrocytopathy where damage to astrocytes exceeds both myelin and neuronal damage. This chapter will review recent neuropathological studies that have provided novel insights into the pathogenic mechanisms, cellular targets, as well as the spectrum of tissue damage in NMO.
Collapse
|
92
|
Transendothelial Transport and Its Role in Therapeutics. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:309404. [PMID: 27355037 PMCID: PMC4897564 DOI: 10.1155/2014/309404] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/13/2014] [Accepted: 06/18/2014] [Indexed: 12/17/2022]
Abstract
Present review paper highlights role of BBB in endothelial transport of various substances into the brain. More specifically, permeability functions of BBB in transendothelial transport of various substances such as metabolic fuels, ethanol, amino acids, proteins, peptides, lipids, vitamins, neurotransmitters, monocarbxylic acids, gases, water, and minerals in the peripheral circulation and into the brain have been widely explained. In addition, roles of various receptors, ATP powered pumps, channels, and transporters in transport of vital molecules in maintenance of homeostasis and normal body functions have been described in detail. Major role of integral membrane proteins, carriers, or transporters in drug transport is highlighted. Both diffusion and carrier mediated transport mechanisms which facilitate molecular trafficking through transcellular route to maintain influx and outflux of important nutrients and metabolic substances are elucidated. Present review paper aims to emphasize role of important transport systems with their recent advancements in CNS protection mainly for providing a rapid clinical aid to patients. This review also suggests requirement of new well-designed therapeutic strategies mainly potential techniques, appropriate drug formulations, and new transport systems for quick, easy, and safe delivery of drugs across blood brain barrier to save the life of tumor and virus infected patients.
Collapse
|
93
|
Verkhratsky A, Marutle A, Rodríguez-Arellano JJ, Nordberg A. Glial Asthenia and Functional Paralysis: A New Perspective on Neurodegeneration and Alzheimer's Disease. Neuroscientist 2014; 21:552-568. [PMID: 25125026 DOI: 10.1177/1073858414547132] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Neuroglia are represented by several population of cells heterogeneous in structure and function that provide for the homeostasis of the brain and the spinal cord. Neuroglial cells are also central for neuroprotection and defence of the central nervous system against exo- and endogenous insults. At the early stages of neurodegenerative diseases including Alzheimer's disease neuroglial cells become asthenic and lose some of their homeostatic, neuroprotective, and defensive capabilities. Astroglial reactivity, for example, correlates with preservation of cognitive function in patients with mild cognitive impairment and prodromal Alzheimer's disease. Here, we overview the experimental data indicating glial paralysis in neurodegeneration and argue that loss of glial function is fundamental for defining the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Manchester, UK Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Amelia Marutle
- Karolinska Institutet, Dept NVS, Center for Alzheimer Research, Translational Alzheimer Neurobiology, Novum, Huddinge, Sweden
| | - J J Rodríguez-Arellano
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain
| | - Agneta Nordberg
- Karolinska Institutet, Dept NVS, Center for Alzheimer Research, Translational Alzheimer Neurobiology, Novum, Huddinge, Sweden Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
94
|
Karki P, Smith K, Johnson J, Aschner M, Lee E. Role of transcription factor yin yang 1 in manganese-induced reduction of astrocytic glutamate transporters: Putative mechanism for manganese-induced neurotoxicity. Neurochem Int 2014; 88:53-9. [PMID: 25128239 DOI: 10.1016/j.neuint.2014.08.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 07/31/2014] [Accepted: 08/05/2014] [Indexed: 10/24/2022]
Abstract
Astrocytes are the most abundant non-neuronal glial cells in the brain. Once relegated to a mere supportive role for neurons, contemporary dogmas ascribe multiple active roles for these cells in central nervous system (CNS) function, including maintenance of optimal glutamate levels in synapses. Regulation of glutamate levels in the synaptic cleft is crucial for preventing excitotoxic neuronal injury. Glutamate levels are regulated predominantly by two astrocytic glutamate transporters, glutamate transporter 1 (GLT-1) and glutamate aspartate transporter (GLAST). Indeed, the dysregulation of these transporters has been linked to several neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD) and Parkinson's disease (PD), as well as manganism, which is caused by overexposure to the trace metal, manganese (Mn). Although Mn is an essential trace element, its excessive accumulation in the brain as a result of chronic occupational or environmental exposures induces a neurological disorder referred to as manganism, which shares common pathological features with Parkinsonism. Mn decreases the expression and function of both GLAST and GLT-1. Astrocytes are commonly targeted by Mn, and thus reduction in astrocytic glutamate transporter function represents a critical mechanism of Mn-induced neurotoxicity. In this review, we will discuss the role of astrocytic glutamate transporters in neurodegenerative diseases and Mn-induced neurotoxicity.
Collapse
Affiliation(s)
- Pratap Karki
- Department of Physiology, Meharry Medical College, Nashville, TN 37208, United States
| | - Keisha Smith
- Department of Physiology, Meharry Medical College, Nashville, TN 37208, United States
| | - James Johnson
- Department of Physiology, Meharry Medical College, Nashville, TN 37208, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Eunsook Lee
- Department of Physiology, Meharry Medical College, Nashville, TN 37208, United States.
| |
Collapse
|
95
|
Smith KM, Maragnoli ME, Phull PM, Tran KM, Choubey L, Vaccarino FM. Fgfr1 inactivation in the mouse telencephalon results in impaired maturation of interneurons expressing parvalbumin. PLoS One 2014; 9:e103696. [PMID: 25116473 PMCID: PMC4130531 DOI: 10.1371/journal.pone.0103696] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 07/04/2014] [Indexed: 11/18/2022] Open
Abstract
Fibroblast growth factors (Fgfs) and their receptors (Fgfr) are expressed in the developing and adult CNS. Previous studies demonstrated a decrease in cortical interneurons and locomotor hyperactivity in mice with a conditional Fgfr1 deletion generated in radial glial cells during midneurogenesis (Fgfr1f/f;hGfapCre+). Here, we report earlier and more extensive inactivation of Fgfr1 in neuroepithelial cells of the CNS (Fgfr1f/f;NesCre+). Similar to findings in Fgfr1f/f;hGfapCre+ mice, parvalbumin positive (PV+) cortical interneurons are also decreased in the neocortex of Fgfr1f/f;NesCre+ mice when compared to control littermates (Fgfr1f/f). Fgfr1f/f;NesCre+ embryos do not differ from controls in the initial specification of GABAergic cells in the ganglionic eminence (GE) as assessed by in situ hybridization for Dlx2, Mash1 and Nkx2. Equal numbers of GABAergic neuron precursors genetically labeled with green fluorescent protein (GFP) were observed at P0 in Fgfr1f/f;hGfapCre+;Gad1-GFP mutant mice. However, fewer GFP+ and GFP+/PV+ interneurons were observed in these mutants at adulthood, indicating that a decrease in cortical interneuron markers is occurring postnatally. Fgfr1 is expressed in cortical astrocytes in the postnatal brain. To test whether the astrocytes of mice lacking Fgfr1 are less capable of supporting interneurons, we co-cultured wild type Gad1-GFP+ interneuron precursors isolated from the medial GE (MGE) with astrocytes from Fgfr1f/f control or Fgfr1f/f;hGfapCre+ mice. Interneurons grown on Fgfr1 deficient astrocytes had small soma size and fewer neurites per cell, but no differences in cell survival. Decreased soma size of Gad67 immunopositive interneurons was also observed in the cortex of adult Fgfr1f/f;NesCre+ mice. Our data indicate that astrocytes from Fgfr1 mutants are impaired in supporting the maturation of cortical GABAergic neurons in the postnatal period. This model may elucidate potential mechanisms of impaired PV interneuron maturation relevant to neuropsychiatric disorders that develop in childhood and adolescence.
Collapse
Affiliation(s)
- Karen Müller Smith
- Child Study Center, Yale University, New Haven, Connecticut, United States of America
| | | | - Pooja M. Phull
- Child Study Center, Yale University, New Haven, Connecticut, United States of America
| | - Kathy May Tran
- Child Study Center, Yale University, New Haven, Connecticut, United States of America
| | - Lisha Choubey
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana, United States of America
| | - Flora M. Vaccarino
- Child Study Center, Yale University, New Haven, Connecticut, United States of America
- Department of Neurobiology, Yale University, New Haven, Connecticut, United States of America
- Kavli Institute for Neuroscience, Yale University, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
96
|
Verkhratsky A, Nedergaard M, Hertz L. Why are astrocytes important? Neurochem Res 2014; 40:389-401. [PMID: 25113122 DOI: 10.1007/s11064-014-1403-2] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/22/2014] [Accepted: 07/26/2014] [Indexed: 12/27/2022]
Abstract
Astrocytes, which populate the grey and white mater of the brain and the spinal cord are highly heterogeneous in their morphology and function. These cells are primarily responsible for homeostasis of the central nervous system (CNS). Most central synapses are surrounded by exceedingly thin astroglial perisynaptic processes, which act as "astroglial cradle" critical for genesis, maturation and maintenance of synaptic connectivity. The perisynaptic glial processes are densely packed with numerous transporters, which provide for homeostasis of ions and neurotransmitters in the synaptic cleft, for local metabolic support and for release of astroglial derived scavengers of reactive oxygen species. Through perivascular processes astrocytes contribute to blood-brain barrier and form "glymphatic" drainage system of the CNS. Furthermore astrocytes are indispensible for glutamatergic and γ-aminobutyrate-ergic synaptic transmission being the supplier of neurotransmitters precursor glutamine via an astrocytic/neuronal cycle. Pathogenesis of many neurological disorders, including neuropsychiatric and neurodegenerative diseases is defined by loss of homeostatic function (astroglial asthenia) or remodelling of astroglial homoeostatic capabilities. Astroglial cells further contribute to neuropathologies through mounting complex defensive programme generally known as reactive astrogliosis.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK,
| | | | | |
Collapse
|
97
|
Verkhratsky A, Rodríguez JJ, Parpura V. Neuroglia in ageing and disease. Cell Tissue Res 2014; 357:493-503. [PMID: 24652503 DOI: 10.1007/s00441-014-1814-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 01/14/2014] [Indexed: 11/28/2022]
Abstract
The proper operation of the mammalian brain requires dynamic interactions between neurones and glial cells. Various types of glial cells are susceptible to morpho-functional changes in a variety of brain pathological states, including toxicity, neurodevelopmental, neurodegenerative and psychiatric disorders. Morphological modifications include a change in the glial cell size and shape; the latter is evident by changes of the appearance and number of peripheral processes. The most blatant morphological change is associated with the alteration of the sheer number of neuroglia cells in the brain. Functionally, glial cells can undergo various metabolic and biochemical changes, the majority of which reflect upon homeostasis of neurotransmitters, in particular that of glutamate, as well as on defence mechanisms provided by neuroglia. Not only glial cells exhibit changes associated with the pathology of the brain but they also change with brain aging.
Collapse
|
98
|
Jin X, Chen Z, Liu X, Liang B, Zhang H, Zhang Z. The expression of endothelial barrier antigen (EBA) and S100B in the rat parietal cortex following brain irradiation. Brain Res 2014; 1558:84-9. [PMID: 24569094 DOI: 10.1016/j.brainres.2014.02.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/21/2014] [Accepted: 02/17/2014] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To visualize the dynamic expression of endothelial barrier antigen (EBA) and S100B in the rat parietal cortex at the acute phase of radiation-induced brain injury using computed tomography (CT). METHODS A rat model of brain injury was established by CT scanning. The expression of EBA and S100B in the parietal cortex was analyzed at different time points by immunohistochemistry (IHC) and western blotting. RESULTS Significantly increased EBA expression was detected in the animals in the control group compared with the animals receiving CT radiation, which exhibited significantly reduced EBA levels within the vessel walls (F=33.29, p<0.05), particularly at day 3 after radiation. Both immunohistochemical staining and western blot analysis indicated that the positive expression levels of S100B among radiation groups were increased compared with the control group (IHC, F=28.05, p<0.05; WB, F=175.3, p<0.05). The expression of S100B peaked at day 3 (IHC, 102718±8710; WB, 2320±0.129), and subsequently decreased. CONCLUSION CT radiation can induce altered EBA and S100B protein expression. Decreased EBA expression levels indicated that the integrity of the blood-brain barrier (BBB) was affected by radiation. The destruction of the BBB and the expression of S100B might play important roles in the incidence and repair of the early radiation-induced brain injury, and radiation represents a cause of mental disorders.
Collapse
Affiliation(s)
- Xuelong Jin
- Department of Physiology & Pathophysiology, Tianjin Medical University, Tianjin 300070, China.
| | - Zequn Chen
- Department of Physiology & Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Xingju Liu
- Department of Physiology & Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Bin Liang
- Department of Physiology & Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Hong Zhang
- Department of Radiology, Tianjin Chest Hospital, Tianjin, China
| | - Zhiwen Zhang
- Department of Neurosurgery, First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
99
|
Laureys G, Gerlo S, Spooren A, Demol F, De Keyser J, Aerts JL. β₂-adrenergic agonists modulate TNF-α induced astrocytic inflammatory gene expression and brain inflammatory cell populations. J Neuroinflammation 2014; 11:21. [PMID: 24479486 PMCID: PMC3942172 DOI: 10.1186/1742-2094-11-21] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 01/15/2014] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The NF-κB signaling pathway orchestrates many of the intricate aspects of neuroinflammation. Astrocytic β₂-adrenergic receptors have emerged as potential regulators in central nervous system inflammation and are potential targets for pharmacological modulation. The aim of this study was to elucidate the crosstalk between astrocytic β₂-adrenergic receptors and the TNF-α induced inflammatory gene program. METHODS Proinflammatory conditions were generated by the administration of TNF-α. Genes that are susceptible to astrocytic crosstalk between β₂-adrenergic receptors (stimulated by clenbuterol) and TNF-α were identified by qPCR-macroarray-based gene expression analysis in a human 1321 N1 astrocytoma cell line. Transcriptional patterns of the identified genes in vitro were validated by RT-PCR on the 1321 N1 cell line as well as on primary rat astrocytes. In vivo expression patterns were examined by intracerebroventricular administration of clenbuterol and/or TNF-α in rats. To examine the impact on the inflammatory cell content of the brain we performed extensive FACS analysis of rat brain immune cells after intracerebroventricular clenbuterol and/or TNF-α administration. RESULTS Parallel transcriptional patterns in vivo and in vitro confirmed the relevance of astrocytic β₂-adrenergic receptors as modulators of brain inflammatory responses. Importantly, we observed pronounced effects of β2-adrenergic receptor agonists and TNF-α on IL-6, CXCL2, CXCL3, VCAM1, and ICAM1 expression, suggesting a role in inflammatory brain cell homeostasis. Extensive FACS-analysis of inflammatory cell content in the brain demonstrated that clenbuterol/TNF-α co-administration skewed the T cell population towards a double negative phenotype and induced a shift in the myeloid brain cell population towards a neutrophilic predominance. CONCLUSIONS Our results show that astrocytic β₂-adrenergic receptors are potent regulators of astrocytic TNF-α-activated genes in vitro and in vivo, and ultimately modulate the molecular network involved in the homeostasis of inflammatory cells in the central nervous system. Astrocytic β₂-adrenergic receptors and their downstream signaling pathway may serve as potential targets to modulate neuroinflammatory responses.
Collapse
Affiliation(s)
- Guy Laureys
- Department of Neurology, University Hospital Brussels, Center for Neurosciences, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Sarah Gerlo
- VIB Department of Medical Protein Research, Ghent University Department of Biochemistry (Faculty of Medicine and Health Sciences), Albert Baertsoenkaai 3, 9000 Ghent, Belgium
| | - Anneleen Spooren
- Department of Physiology, Laboratory of Eukaryotic Gene Expression and Signal Transduction, Ghent University, Proeftuinstraat 86, 9000 Ghent, Belgium
| | - Frauke Demol
- Department of Neurology, University Hospital Brussels, Center for Neurosciences, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Jacques De Keyser
- Department of Neurology, University Hospital Brussels, Center for Neurosciences, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
- Department of Neurology, University Medical Center Groningen, RUG, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Joeri L Aerts
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| |
Collapse
|
100
|
Uranova NA, Vikhreva OV, Rakhmanova VI, Orlovskaia DD. [Reactivity of perineuronal astrocytes in the prefrontal cortex in schizophrenia: an ultrastructural morphometric study]. Zh Nevrol Psikhiatr Im S S Korsakova 2014; 114:65-72. [PMID: 25726783 DOI: 10.17116/jnevro201411412165-72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Previously the ultrastructural alterations of astrocytes have been reported in schizophrenia. Reduced dendritic arborization of the neurons in layer 5 of the prefrontal cortex has been found in schizophrenia. Authors hypothesized that the abnormalities in perineuronal astrocytes (PA) might contribute to these neuronal changes. It was aimed to study the ultrastructure of PA in the prefrontal cortex in schizophrenia. MATERIAL AND METHODS Postmortem electron microscopic morphometric study of PA was performed in layer 5, area 10 of the prefrontal cortex in 39 cases of schizophrenia and 37 controls. RESULTS No significant group differences were found in areas of cell, nucleus, cytoplasm, volume fraction (Vv) of lipofuscin granules and areal density of PA. However, in the subgroup of women with schizophrenia, the areal density of PA was significantly lower and the area of PA was significantly higher as compared to the subgroup of healthy women (-52%, p<0,01; +32%, p<0.05 respectively) and to the subgroup of men with schizophrenia (-56%, p<0,01; +23%, p<0,05 respectively). The area of PA nucleus was negatively correlated with the duration of disease (r= -0.37, p=0.02) and positively with the age of disease onset (ADO) (r=0,47, p<0,01). Areas of PA and of PA nucleus were significantly lower in early ADO (<21 y.o.) as compared to the adult ADO (>21 y.o.) (-24%, p<0.05). Vv of lypofuscin granules was correlated with the age in control group (r=0.52, p=0.001), but not in schizophrenia group (r=0.13, p=0.4). CONCLUSION Significant differences in PA reactivity in the prefrontal cortex in the schizophrenia are associated with gender and age at onset of the disease.
Collapse
Affiliation(s)
- N A Uranova
- FGBU 'Nauchnyĭ tsentr psikhicheskogo zdorov'ia' RAMN, Moskva
| | - O V Vikhreva
- FGBU 'Nauchnyĭ tsentr psikhicheskogo zdorov'ia' RAMN, Moskva
| | - V I Rakhmanova
- FGBU 'Nauchnyĭ tsentr psikhicheskogo zdorov'ia' RAMN, Moskva
| | - D D Orlovskaia
- FGBU 'Nauchnyĭ tsentr psikhicheskogo zdorov'ia' RAMN, Moskva
| |
Collapse
|