Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Pharmacology ; 108(2): 188-198, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36724743

RESUMO

INTRODUCTION: Multiple sclerosis (MS) is the most common chronic inflammatory, demyelinating disease of the central nervous system. Dimethyl fumarate (DMF) and monomethyl fumarate (MMF) belong to the disease-modifying drugs in treatment of MS. There is evidence that astrocytes and microglia are involved in MS pathology, but few studies are available about MMF and DMF effects on astrocytes and microglia. The aim of this study was to investigate the effects of MMF and DMF on microglial activation and morphology as well as potential effects on glial viability, Cx43, and AQP4 expressions in different set-ups of an in vitro astrocyte-microglia co-culture model of inflammation. METHODS: Primary rat glial co-cultures of astrocytes containing 5% (M5, mimicking "physiological" conditions) or 30% (M30, mimicking "pathological, inflammatory" conditions) of microglia were treated with different concentrations of MMF (0.1, 0.5, and 2 µg/mL) or DMF (1.5, 5, and 15 µM) for 24 h. Viability, proliferation, and cytotoxicity of glial cells were examined using MTT assay. Immunocytochemistry was performed to analyze the microglial phenotypes. Connexin 43 (Cx43) and aquaporin 4 (AQP4) expressions were quantified by immunoblot analysis. RESULTS: Treatment with different concentrations of MMF or DMF for 24 h did not change the glial cell viability in M5 and M30 co-cultures. Microglial phenotypes were not altered by DMF under physiological M5 conditions, but treatment with higher concentration of DMF (15 µM) induced microglial activation under inflammatory M30 conditions. Incubation with different concentrations of MMF had no effects on microglial phenotypes. The Cx43 expression in M5 and M30 co-cultures was not changed significantly by immunoblot analysis after incubation with different concentrations of DMF or MMF for 24 h. The AQP4 expression was significantly increased in M5 co-cultures after incubation with 5 µm DMF. Under the other conditions, AQP4 expression was not affected by DMF or MMF. DISCUSSION: In different set-ups of the astrocyte-microglia co-culture model of inflammation, MMF has not shown significant effects. DMF had only limited effects on microglia phenotypes and AQP4 expression. In summary, mechanisms of action of fumarates probably do not involve direct effects on microglia phenotypes as well as Cx43 and AQP4 expression.


Assuntos
Fumarato de Dimetilo , Microglia , Ratos , Animais , Fumarato de Dimetilo/metabolismo , Fumarato de Dimetilo/farmacologia , Microglia/metabolismo , Astrócitos , Conexina 43/metabolismo , Conexina 43/farmacologia , Técnicas de Cocultura , Inflamação/metabolismo
2.
Int J Neuropsychopharmacol ; 25(3): 185-196, 2022 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-34791253

RESUMO

BACKGROUND: Astrocytes and microglia are involved in the pathophysiology of epilepsy and bipolar disorder with a link to inflammation. We aimed to investigate the effects of the antiepileptic and mood-stabilizing drugs lamotrigine (LTG) and topiramate (TPM) on glial viability, microglial activation, cytokine release, and expression of gap-junctional protein connexin 43 (Cx43) in different set-ups of an in vitro astrocyte-microglia co-culture model of inflammation. METHODS: Primary rat co-cultures of astrocytes containing 5% (M5, representing "physiological" conditions) or 30% (M30, representing "pathological, inflammatory" conditions) of microglia were treated with different concentrations of LTG and TPM for 24 hours. An 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was performed to measure the glial cell viability. The microglial activation state was analyzed by immunocytochemistry. The pro-inflammatory tumor necrosis factor-α (TNF-α) and anti-inflammatory transforming growth factor-ß1 (TGF-ß1) cytokine levels were measured by enzyme-linked immunosorbent assay. The astroglial Cx43 expression was quantified by western blot. RESULTS: A significant reduction of the glial cell viability after incubation with LTG or TPM was observed in a concentration-dependent manner under all conditions. LTG caused no significant alterations of the microglial phenotypes. Under pathological conditions, TPM led to a significant concentration-dependent reduction of microglial activation. This correlated with increased astroglial Cx43 expression. TNF-α levels were not affected by LTG and TPM. Treatment with higher concentrations of LTG, but not with TPM, led to a significant increase in TGF-ß1 levels in M5 and M30 co-cultures. CONCLUSIONS: Despite the possible glial toxicity of LTG and TPM, both drugs reduced inflammatory activity, suggesting potential positive effects on the neuroinflammatory components of the pathogenesis of epilepsy and bipolar disorder.


Assuntos
Anticonvulsivantes , Epilepsia , Animais , Anticonvulsivantes/farmacologia , Anticonvulsivantes/uso terapêutico , Astrócitos/metabolismo , Técnicas de Cocultura , Conexina 43/metabolismo , Citocinas/metabolismo , Inflamação/metabolismo , Lamotrigina/metabolismo , Lamotrigina/farmacologia , Lamotrigina/uso terapêutico , Microglia , Ratos , Topiramato/farmacologia , Topiramato/uso terapêutico , Fator de Necrose Tumoral alfa
3.
BMC Neurosci ; 22(1): 21, 2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33765917

RESUMO

BACKGROUND: Hepatic encephalopathy (HE) is a neurological complication resulting from acute or chronic liver disease. Hyperammonemia leading to astrocyte swelling and cerebral edema in combination with neuroinflammation including microglia activation, mainly contribute to the pathogenesis of HE. However, little is known about microglia and their inflammatory response, as well as their influence on astrocytic channels and astrocyte swelling under hyperammonemia. OBJECTIVE: To investigate the effects of ammonia on the microglial activation and morphology in different set-ups of an in vitro astrocyte-microglia co-culture model. Further, potential effects on glial viability, connexin 43 (Cx43) and aquaporin 4 (AQP4) expression were tested. METHODS: Primary rat glial co-cultures of astrocytes containing 5% (M5, representing "physiological" conditions) or 30% (M30, representing "pathological" conditions) of microglia were incubated with 3 mM, 5 mM, 10 mM and 20 mM ammonium chloride (NH4Cl) for 6 h and 24 h in order to mimic the conditions of HE. An MTT assay was performed to measure the viability, proliferation and cytotoxicity of cells. The microglial phenotypes were analyzed by immunocytochemistry. The expression of Cx43 and AQP4 were quantified by immunoblot analysis. RESULTS: A significant reduction of glial viability was observed in M30 co-cultures after incubation with 20 mM NH4Cl for 6 h, whereas in M5 co-cultures the viability remained unchanged. Microglial activation was detected by immunocytochemistry after incubation with 3 mM, 5 mM and 10 mM NH4Cl for 6 h and 24 h in M5 as well as in M30 co-cultures. The Cx43 expression was slightly increased in M30 co-cultures after 6 h incubation with 5 mM NH4Cl. Also, the AQP4 expression was slightly increased only in M5 co-cultures treated with 10 mM NH4Cl for 6 h. Under the other conditions, Cx43 and AQP4 expression was not affected by NH4Cl. CONCLUSIONS: The novel aspect of our study was the significant microglial activation and decrease of viability after NH4Cl incubation in different set-ups of an in vitro astrocyte-microglia co-culture model, contributing to better understanding of pathophysiological mechanisms of HE. Hyperammonemia led to limited effects on Cx43 and AQP4 expression, the relevance of these minimal changes should be viewed with caution.


Assuntos
Cloreto de Amônio/toxicidade , Aquaporina 4/metabolismo , Conexina 43/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Animais , Astrócitos/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Encefalopatia Hepática/metabolismo , Ratos
5.
Epilepsia ; 55(1): 184-92, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24299259

RESUMO

PURPOSE: The contribution of glial cells, mainly astrocytes and microglia, to the pathophysiology of epilepsy is increasingly appreciated. Glia play a pivotal role in the initiation and maintenance of the central nervous system (CNS) immune response and neuronal metabolic and trophic supply. Recent clinical and experimental evidence suggests a direct relationship between epileptic activity and CNS inflammation, which is characterized by accumulation, activation, and proliferation of microglia and astrocytes. Concomitant glia-mediated mechanisms of action of several antiepileptic drugs (AEDs) have been proposed. However, their direct effects on glial cells have been rarely investigated. We aimed to investigate the effect of commonly used AEDs on glial viability, the gap junctional network, the microglial activation, and cytokine expression in an in vitro astroglia/microglia co-culture model. METHODS: Primary astrocytic cultures were prepared from brains of postnatal (P0-P2) Wistar rats and co-cultured with a physiologic amount of 5%, as well as 30% microglia in order to mimic inflammatory conditions. Co-cultures were treated with valproic acid (VPA), carbamazepine (CBZ), phenytoin (PHE), and gabapentin (GBT). Viability and proliferation were measured using the tetrazolium (MTT) assay. The microglial activation state was determined by immunocytochemical labeling. The astroglial connexin 43 (Cx43) expression was measured by Western blot analysis. The transforming growth factor-ß1 (TGF-ß1) and tumor necrosis factor-α (TNF-α) cytokine levels were measured by the quantitative sandwich enzyme immunosorbent assay (ELISA). KEY FINDINGS: Astrocytes, co-cultured with 5% microglia (M5 co-cultures), showed a dose-dependent, significant reduction in glial viability after incubation with PHE and CBZ. Furthermore, VPA led to highly significant microglial activation at all doses examined. The antiinflammatory cytokine TGF-ß1 release was induced by high doses of GBT and PHE. Astrocytes co-cultured with 30% microglia (M30 co-cultures) revealed a dose-dependent significant reduction in glial viability after incubation with PHE, accompanied by increased TGF-ß1 and TNF-α levels. However, CBZ significantly reduced the amount of activated microglial cells and increased the total number of inactivated microglia. Finally, CBZ resulted in reduced viability at all doses examined. SIGNIFICANCE: CNS inflammation is characterized by a disturbance of glial cell functions. Strong microglial activation, a typical hallmark of inflammation, was induced by VPA in M5 and continued in M30 co-cultures. With regard to the direct relation between CNS inflammation and seizures, VPA seems to be unsuitable for reducing inflammatory conditions. The reverse effect was achieved after CBZ. We noticed significant microglial inactivation, after incubation of the M30 co-cultures. In conclusion, we suggest that AEDs with antiinflammatory glial features are beneficial for seizures caused by persistent brain inflammation.


Assuntos
Anticonvulsivantes/farmacologia , Astrócitos/fisiologia , Epilepsia/etiologia , Inflamação/fisiopatologia , Microglia/fisiologia , Neuroglia/fisiologia , Aminas/farmacologia , Aminas/uso terapêutico , Animais , Anticonvulsivantes/uso terapêutico , Astrócitos/efeitos dos fármacos , Western Blotting , Carbamazepina/farmacologia , Carbamazepina/uso terapêutico , Células Cultivadas , Técnicas de Cocultura , Conexina 43/biossíntese , Ácidos Cicloexanocarboxílicos/farmacologia , Ácidos Cicloexanocarboxílicos/uso terapêutico , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Epilepsia/tratamento farmacológico , Epilepsia/fisiopatologia , Gabapentina , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/fisiologia , Inflamação/tratamento farmacológico , Microglia/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Fenitoína/farmacologia , Fenitoína/uso terapêutico , Ratos , Ratos Wistar , Fator de Crescimento Transformador beta1/biossíntese , Ácido Valproico/farmacologia , Ácido Valproico/uso terapêutico , Ácido gama-Aminobutírico/farmacologia , Ácido gama-Aminobutírico/uso terapêutico
6.
Neuroimmunomodulation ; 21(4): 189-94, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24504116

RESUMO

BACKGROUND: Glutamate and its specific ionotropic receptors, including N-methyl-D-aspartate (NMDA) and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors, are supposed to play an important role in neurodegeneration as well as neuronal regeneration. Although autoantibodies (aab) to glutamate receptors (GluR) have been identified in several neurologic diseases, including paraneoplastic encephalitis and Rasmussen's encephalitis (RE) with an increasing prevalence, the presence and role of anti-GluR aab in multiple sclerosis (MS) have not been studied yet. OBJECTIVES AND METHODS: In this study, we tested the serum samples of 56 subjects, including patients with relapsing-remitting MS (n = 25), patients with RE (n = 8), and healthy donors (HD; n = 23), for anti-GluR aab by immunoblot analysis of a panel of recombinantly expressed GluR proteins, including GluN1, GluN2C, GluA3, GluK2, and GluD2. RESULTS: aab were mainly found directed against GluN1 and, except for one aab positive to GluK2 in 1 MS patient and 2 HD controls positive for GluA3, no other anti-GluR aab were detected. In the sera of RE patients, no anti-GluR aab were found. In patients with MS, 8 of the 25 sera (32%) tested positive for GluN1. Compared to the HD (6/23; 26%), this difference was not statistically significant (p = 0.28). CONCLUSIONS: Our study showed that if anti-GluR aab were detectable in HD and MS patients, they were mainly directed against GluN1 (in particular to oligomeric protein complexes) and were not found in RE. Those antibodies may have low titers and low affinities and might be considered an immune epiphenomenon. Hence, further studies will have to clarify their potential role as a surrogate marker for the extent of neuronal destruction or regeneration, respectively.


Assuntos
Autoanticorpos/sangue , Encefalite/imunologia , Esclerose Múltipla Recidivante-Remitente/imunologia , Receptores de Glutamato/imunologia , Adulto , Autoantígenos/imunologia , Encefalite/sangue , Feminino , Humanos , Immunoblotting , Masculino , Esclerose Múltipla Recidivante-Remitente/sangue
7.
Epilepsia Open ; 8(2): 692-701, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37078803

RESUMO

The γ-aminobutyric acid (GABA) is the main inhibitory transmitter in the central nervous system and GABA receptors mediate the inhibitory synaptic transmission. GABA binding to neuronal GABAA R leads to a rapid hyperpolarization and a higher excitation threshold due to an increase in membrane Cl- permeability. The synaptic GABAA R is mostly composed of two α(1-3), two ß, and one γ subunit with the most abundant configuration α1ß2γ2. Recently, antibodies (Abs) against α1, ß3, and γ2 subunits of GABAA R were detected in a severe form of autoimmune encephalitis with refractory seizures, status epilepticus, and multifocal brain lesions, affecting gray and white matter. Experimental studies confirmed multiple mechanisms and direct functional effects of GABAA R Abs on neurons with decreased GABAergic synaptic transmission and increased neuronal excitability. The expression of GABAA R on astrocytes is well established. However, extensive studies about the effects of autoimmune GABAA R Abs on astrocytic GABAA R are missing. We hypothesize that GABAA R Abs may lead additionally to blocking astrocytic GABAA Rs with impaired Ca2+ homeostasis/spreading, astrocytic Cl- imbalance, dysfunction of astrocyte-mediated gliotransmission (e.g., decreased adenosine levels) and accumulation of excitatory neurotransmission, all this contributing to seizures, variable clinical/MRI presentations, and severity. The most abundant expressed GABAA R subunits in rodent astrocytes are α1, α2, ß1, ß3, and γ1 localized in both white and gray matter. Data about GABAA R subunits in human astrocytes are even more limited, comprising α2, ß1, and γ1. Overlapping binding of GABAA R Abs to neuronal and astroglial receptors is still possible. In vitro and in vivo animal models can be helpful to test the effects of GABAA R Abs on glia. This is from an epileptological point of view relevant because of the increasing evidence, confirming the glial involvement in the pathogenesis of epilepsy. Taken together, autoimmune disorders are complex and multiple mechanisms including glia could contribute to the pathogenesis of GABAA R encephalitis with seizures.


Assuntos
Doenças Autoimunes do Sistema Nervoso , Encefalite , Animais , Humanos , Receptores de GABA-A/metabolismo , Astrócitos/metabolismo , Convulsões , Ácido gama-Aminobutírico/fisiologia , Proteínas de Transporte , Anticorpos/metabolismo
8.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 3253-3267, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37231170

RESUMO

Due to the role of astrocytes and microglia in the pathophysiology of epilepsy and limited studies of antiseizure medication (ASM) effects on glial cells, we studied tiagabine (TGB) and zonisamide (ZNS) in an astrocyte-microglia co-culture model of inflammation. Different concentrations of ZNS (10, 20, 40, 100 µg/ml) or TGB (1, 10, 20, 50 µg/ml) were added to primary rat astrocytes co-cultures with 5-10% (M5, physiological conditions) or 30-40% (M30, pathological inflammatory conditions) microglia for 24 h, aiming to study glial viability, microglial activation, connexin 43 (Cx43) expression and gap-junctional coupling. ZNS led to the reduction of glial viability by only 100 µg/ml under physiological conditions. By contrast, TGB revealed toxic effects with a significant, concentration-dependent reduction of glial viability under physiological and pathological conditions. After the incubation of M30 co-cultures with 20 µg/ml TGB, the microglial activation was significantly decreased and resting microglia slightly increased, suggesting possible anti-inflammatory features of TGB under inflammatory conditions. Otherwise, ZNS caused no significant changes of microglial phenotypes. The gap-junctional coupling was significantly decreased after the incubation of M5 co-cultures with 20 and 50 µg/ml TGB, which can be related to its anti-epileptic activity under noninflammatory conditions. A significant decrease of Cx43 expression and cell-cell coupling was found after the incubation of M30 co-cultures with 10 µg/ml ZNS, suggesting additional anti-seizure effects of ZNS with the disruption of glial gap-junctional communication under inflammatory conditions. TGB and ZNS differentially regulated the glial properties. Developing novel ASMs targeting glial cells may have future potential as an "add-on" therapy to classical ASMs targeting neurons.


Assuntos
Astrócitos , Microglia , Ratos , Animais , Técnicas de Cocultura , Tiagabina/metabolismo , Tiagabina/farmacologia , Conexina 43/metabolismo , Zonisamida/farmacologia , Zonisamida/metabolismo , Comunicação Celular , Neuroglia/metabolismo , Inflamação/patologia
9.
Brain Sci ; 13(3)2023 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-36979303

RESUMO

Depression may occur in patients with multiple sclerosis, especially during interferon-ß (IFN-ß) treatment, and therapy with antidepressants may be necessary. Interactions of IFN-ß with antidepressants concerning glia-mediated inflammation have not yet been studied. Primary rat co-cultures of astrocytes containing 5% (M5, consistent with "physiological" conditions) or 30% (M30, consistent with "pathological, inflammatory" conditions) of microglia were incubated with 10 ng/mL amitriptyline or doxepin for 2 h, or with 2000 U/mL IFN-ß for 22 h. To investigate the effects of antidepressants on IFN-ß treatment, amitriptyline or doxepin was added to IFN-ß pre-treated co-cultures. An MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay was performed to measure the glial cell viability, immunocytochemistry was performed to evaluate the microglial activation state, and ELISA was performed to measure pro-inflammatory TNF-α and IL-6 cytokine concentrations. Incubation of inflammatory astrocyte-microglia co-cultures with amitriptyline, doxepin or IFN-ß alone, or co-incubation of IFN-ß pre-treated co-cultures with both antidepressants, significantly reduced the extent of inflammation, with the inhibition of microglial activation. TNF-α and IL-6 levels were not affected. Accordingly, the two antidepressants did not interfere with the anti-inflammatory effect of IFN-ß on astrocytes and microglia. Furthermore, no cytotoxic effects on glial cells were observed. This is the first in vitro study offering novel perspectives in IFN-ß treatment and accompanying depression regarding glia.

10.
Eur J Pharmacol ; 915: 174696, 2022 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-34902360

RESUMO

PURPOSE: Understanding the effects of antiepileptic drugs on glial cells and glia-mediated inflammation is a new approach to future treatment of epilepsy. Little is known about direct effects of the antiepileptic drug lacosamide (LCM) on glial cells. Therefore, we aimed to study the LCM effects on glial viability, microglial activation, expression of gap-junctional (GJ) protein Cx43 as well as intercellular communication in an in vitro astrocyte-microglia co-culture model of inflammation. METHODS: Primary rat astrocytes co-cultures containing 5% (M5, "physiological" conditions) or 30% (M30, "pathological inflammatory" conditions) of microglia were treated with different concentrations of LCM [5, 15, 30, and 90 µg/ml] for 24 h. Glial cell viability was measured by MTT assay. Immunocytochemistry was performed to analyze the microglial activation state. Western blot analysis was used to quantify the astroglial Cx43 expression. The GJ cell communication was studied via Scrape Loading. RESULTS: A concentration-dependent incubation with LCM did not affect the glial cell viability both under physiological and pathological conditions. LCM induced a significant concentration-dependent decrease of activated microglia with parallel increase of ramified microglia under pathological inflammatory conditions. This correlated with an increase in astroglial Cx43 expression. Nevertheless, the functional coupling via GJs was significantly reduced after incubation with LCM. CONCLUSION: LCM has not shown effects on the glial cell viability. The reduced GJ coupling by LCM could be related to its anti-epileptic activity. The anti-inflammatory glial features of LCM with inhibition of microglial activation under inflammatory conditions support beneficial role in epilepsy associated with neuroinflammation.


Assuntos
Astrócitos
11.
Front Cell Neurosci ; 16: 995861, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36406753

RESUMO

Implications of glia in the pathophysiology of epilepsy raise the question of how these cells besides neurons are responsive to antiseizure medications (ASMs). Understanding ASM effects on glia and glia-mediated inflammation may help to explore astrocytes and microglia as potential targets for alternative anti-epileptogenic therapies. The aim of this study was to investigate the effects of the new generation ASM brivaracetam (BRV) in an astrocyte-microglia co-culture model of inflammation. Primary rat astrocytes co-cultures containing 5%-10% (M5, "physiological" conditions) or 30%-40% (M30, "pathological inflammatory" conditions) of microglia were treated with different concentrations of BRV (0.5, 2, 10, and 20 µg/ml) for 24 h. Glial cell viability was measured by MTT assay. Microglial activation states were analyzed by immunocytochemistry and astroglial connexin 43 (Cx43) expression by Western blot analysis and immunocytochemistry. Gap-junctional coupling was studied via Scrape Loading. Incubation with high, overdose concentration (20 µg/ml) of BRV significantly reduced the glial cell viability under physiological conditions (p < 0.01: **). Treatment with BRV in therapeutic concentrations (0.5 and 2 µg/ml) reduced the resting microglia (p < 0.05: *) and increased the microglial activation under inflammatory conditions (p < 0.01: **). Astroglial Cx43 expression was not affected. The gap-junctional coupling significantly increased only by 0.5 µg/ml BRV under physiological conditions (p < 0.05: *). Our findings suggest mild pro-inflammatory, in vitro features of BRV with regard to microglia morphology. BRV showed no effects on Cx43 expression and only limited effects on gap-junctional coupling. Reduction of glial viability by overdose BRV indicates possible toxic effects.

12.
J Neurooncol ; 103(3): 479-89, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21107646

RESUMO

Similar to astrocytes, glioma cells form a well-coupled syncytium via gap junctions. This can be influenced, for example, by activated microglia, the main inflammatory cell population within the central nervous system (CNS). Under pathological conditions such as neoplastic cell growth, microglia number and activation state are enhanced. The aim of the present study is to analyze the influence of dexamethasone (DEX) on cellular and molecular properties in glial coculture models consisting of astroglia and microglia and human and rat glioma cell lines. Primary rat glial cocultures of astrocytes containing 5% (M5, representing "physiological" conditions) or 30% (M30, representing "pathological" conditions) microglia as well as rat and human glioma cell lines (F98, C6, U87) were incubated with DEX for 24 h. DEX-treated M30 cocultures showed significant increased gap junctional intercellular communication (GJIC). DEX treatment of glioma cells resulted in depolarization of the membrane resting potential (MRP) and a significant reduction of GJIC. Furthermore, DEX reduced the amount of activated microglia in M30 cocultures. DEX had no significant effects on the tested variables in the M5 coculture. DEX differentially regulates functional membrane properties of glioma cells and astrocytes in primary glial cocultures, which might resemble steroid effects in glioma cells and adjacent glial components in vivo.


Assuntos
Antineoplásicos Hormonais/farmacologia , Astrócitos/fisiologia , Neoplasias Encefálicas/fisiopatologia , Dexametasona/farmacologia , Glioma/fisiopatologia , Potenciais da Membrana/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Antineoplásicos Hormonais/uso terapêutico , Astrócitos/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura/métodos , Conexina 43/metabolismo , Dexametasona/uso terapêutico , Relação Dose-Resposta a Droga , Interações Medicamentosas , Ectodisplasinas/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/patologia , Antagonistas de Hormônios/farmacologia , Humanos , Potenciais da Membrana/fisiologia , Mifepristona/farmacologia , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Estatísticas não Paramétricas
13.
Front Cell Neurosci ; 15: 805755, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975415

RESUMO

Astrocytes and microglia are the main cell population besides neurons in the central nervous system (CNS). Astrocytes support the neuronal network via maintenance of transmitter and ion homeostasis. They are part of the tripartite synapse, composed of pre- and postsynaptic neurons and perisynaptic astrocytic processes as a functional unit. There is an increasing evidence that astroglia are involved in the pathophysiology of CNS disorders such as epilepsy, autoimmune CNS diseases or neuropsychiatric disorders, especially with regard to glia-mediated inflammation. In addition to astrocytes, investigations on microglial cells, the main immune cells of the CNS, offer a whole network approach leading to better understanding of non-neuronal cells and their pathological role in CNS diseases and treatment. An in vitro astrocyte-microglia co-culture model of inflammation was developed by Faustmann et al. (2003), which allows to study the endogenous inflammatory reaction and the cytokine expression under drugs in a differentiated manner. Commonly used antiepileptic drugs (e.g., levetiracetam, valproic acid, carbamazepine, phenytoin, and gabapentin), immunomodulatory drugs (e.g., dexamethasone and interferon-beta), hormones and psychotropic drugs (e.g., venlafaxine) were already investigated, contributing to better understanding mechanisms of actions of CNS drugs and their pro- or anti-inflammatory properties concerning glial cells. Furthermore, the effects of drugs on glial cell viability, proliferation and astrocytic network were demonstrated. The in vitro astrocyte-microglia co-culture model of inflammation proved to be suitable as unique in vitro model for pharmacological investigations on astrocytes and microglia with future potential (e.g., cancer drugs, antidementia drugs, and toxicologic studies).

14.
Acta Neuropathol ; 119(6): 771-8, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20309567

RESUMO

The anterior cingulate cortex (ACC) represents a phylogenetically ancient region of the mammalian brain that has undergone recent adaptive changes in humans. It contains a large spindle-shaped cell type, referred to as von Economo neuron (VEN) that has been shown to be involved in the pathophysiology of various neuropsychiatric disorders. Schizophrenia is a group of disorders that is, in part, characterised by a disruption of neuronal migration in early ontogeny and presumably secondary degeneration after the first psychotic episode in some patients. Accordingly, we tested the hypothesis that the density of VENs is reduced in a neurodevelopmental subtype of schizophrenia, which we defined by an early onset of the disorder. The density of VENs was estimated in layer Vb of Brodmann's area 24 in 20 subjects diagnosed with schizophrenia. The results were compared with 19 specimens from patients with bipolar disorder as a clinical control and 22 non-psychiatric samples. The density of VENs did not differ between the three groups. However, the VEN density in the right ACC correlated with the age at onset, and inversely with the duration of the illness in schizophrenia, but not in bipolar disorder. Thus, patients with early onset schizophrenia (and longer duration of illness) had a reduced VEN density. Age, sex, postmortem interval, brain weight, and cortical thickness had no significant impact on the results. These findings suggest that VENs in the ACC are involved in neurodevelopmental and perhaps neurodegenerative processes specific to schizophrenia.


Assuntos
Giro do Cíngulo/patologia , Neurônios/patologia , Esquizofrenia/patologia , Adulto , Fatores Etários , Idade de Início , Transtorno Bipolar/patologia , Encéfalo/patologia , Estudos de Casos e Controles , Contagem de Células , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Tamanho do Órgão , Fatores Sexuais , Fatores de Tempo , Adulto Jovem
15.
Med Hypotheses ; 139: 109612, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32085980

RESUMO

The N-methyl-d-aspartate receptor (NMDAR) encephalitis is the most common form of autoimmune encephalitis. Antibodies against the GluN1 subunit of the NMDAR showed in primary cultures of rat hippocampal neurons and in a mouse model pathogenic effects including cross-linking and internalization of the target receptors (NMDAR). Several studies demonstrated that not only neurons, but also astrocytes express functional NMDA receptors including GluN1 subunit. It is conceivable that the pathogenic antibodies against the NMDAR causing the anti-NMDAR encephalitis affect not only the neuronal receptors, but also the NMDAR on astrocytes. We hypothesize that antibodies against NMDAR can lead to cross-linking and internalization of the target receptors in astrocytes similar to neurons with disruption of the calcium release within the astrocytes and consequently blocking release of inhibitory gliotransmitters. Further, we assume influence on expression of aquaporin 4 channels and gap-junctional communication due to modification of the astrocytic NMDAR. The disruption of these interactions and dysbalance could result in impairment of CNS homeostasis and co-determine the severity of clinical disease manisfestation and recovery.


Assuntos
Encefalite Antirreceptor de N-Metil-D-Aspartato , Doença de Hashimoto , Animais , Astrócitos , Células Cultivadas , Camundongos , Ratos , Receptores de N-Metil-D-Aspartato
16.
Epilepsy Behav ; 14(1): 66-70, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18762279

RESUMO

OBJECTIVE: The goal of this study was to examine the influence of seizure freedom on executive function in outpatients with generalized epilepsy and extrafrontal partial epilepsy. Recent investigations of cognitive function in epilepsy have revealed executive deficits in persons with focal temporal as well as generalized epilepsies. Additional studies have suggested an influence of seizure freedom on cognitive function. METHODS: Thirty-five consecutive outpatients were divided into seizure free 3 months (n=17). The neuropsychological tests administered included: verbal fluency tasks, the Cognitive Estimation Test, the Hayling and Brixton Test, and the Behavioural Assessment of the Dysexecutive Syndrome (BADS) battery. Both patient groups were compared with matched healthy controls (n=16). RESULTS: The extensive testing revealed significant differences between patients with shorter seizure-free periods and healthy controls with respect to overall errors and phonemic verbal fluency, response suppression, and BADS overall profile scores. Subjects seizure free >3 months exhibited a trend toward impairment in the phonemic fluency task only. CONCLUSIONS: The results suggest that deficits in executive function were present in patients with extrafrontal partial epilepsy and generalized epilepsy, indicating the potential influence of epileptic activity on the ability to focus on relevant information and switch attention to other relevant information, to plan tasks and subtasks, and to check on and encode working memory content. The results also suggest that those deficits may be more pronounced in patients with relatively short seizure-free periods.


Assuntos
Epilepsias Parciais/psicologia , Epilepsia Generalizada/psicologia , Desempenho Psicomotor/fisiologia , Convulsões/psicologia , Adulto , Cognição/fisiologia , Interpretação Estatística de Dados , Feminino , Humanos , Masculino , Testes Neuropsicológicos , Comportamento Verbal/fisiologia
18.
J Neurosci Res ; 86(8): 1781-8, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18335543

RESUMO

There is accumulating evidence that epileptic activity is accompanied by inflammatory processes. In the present study, we evaluated the effect of levetiracetam (Keppra), an anticonvulsant drug with decisive antiepileptic features, with regard to its putative antiinflammatory potential. We previously established an in vitro cell culture model to mimic inflammatory conditions: Primary astrocytic cultures of newborn rats were cocultured with 30% (M30) microglial cells. Alternatively, cocultures containing 5% microglia (M5) were incubated with the proinflammatory mediator, the cytokine interleukin-1beta (IL-1beta), and lipopolysaccharide (LPS), a potent bacterial activator of the immune system. For the M30 cocultures, we observed reduced expression of connexin 43 (Cx43), the predominant gap junction protein. Impaired functional dye coupling and depolarized membrane resting potential (MRP) were monitored in M30 cocultures as well as in M5 cocultures treated with IL-1beta and LPS. We could show that the Cx43 expression, the coupling property, and the membrane resting potential on which we focused our inflammatory coculture model were normalized to noninflammatory level under treatment with levetiracetam (Keppra). Cumulatively, our results provide evidence for antiinflammatory properties of levetiracetam in seizure treatment.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Anticonvulsivantes/farmacologia , Astrócitos/efeitos dos fármacos , Piracetam/análogos & derivados , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Astrócitos/patologia , Células Cultivadas , Técnicas de Cocultura , Levetiracetam , Piracetam/farmacologia , Ratos , Ratos Wistar
19.
Behav Brain Res ; 291: 325-333, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26048428

RESUMO

While it is well known that the left hemisphere is more efficient than the right in most tasks involving perception of speech stimuli, the neurophysiological pathways leading to these lateralised performance differences are as yet rather unclear. In particular, the question whether language lateralisation depends on semantic processing or is already evident in early perceptual stimulus processing has not been answered unequivocally. In the present study, we therefore recorded event-related potentials (ERPs) during tachistoscopic presentation of horizontally or vertically presented verbal stimuli in the left (LVF) and the right visual field (RVF). Participants were asked to indicate, whether the presented stimulus was a word or a non-word. On the behavioural level, participants showed stronger hemispheric asymmetries for horizontal, than for vertical stimulus presentation. In addition, ERP asymmetries were also modulated by stimulus presentation format, as the electrode by visual field interactions for P1 and N1 were stronger after vertical, than after horizontal stimulus presentation. Moreover, sLORETA revealed that ERP left-right asymmetries were mainly driven by the extrastriate cortex and reading-associated areas in the parietal cortex. Taken together, the present study shows electrophysiological support for the assumption that language lateralisation during speech perception arises from a left dominance for the processing of early perceptual stimulus aspects.


Assuntos
Lateralidade Funcional/fisiologia , Reconhecimento Visual de Modelos/fisiologia , Leitura , Percepção da Fala/fisiologia , Córtex Visual/fisiologia , Mapeamento Encefálico , Eletroencefalografia , Potenciais Evocados , Feminino , Humanos , Testes de Linguagem , Masculino , Testes Neuropsicológicos , Lobo Parietal/fisiologia , Campos Visuais , Adulto Jovem
20.
Neurosci Lett ; 330(3): 293-5, 2002 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-12270649

RESUMO

In the pathogenesis of multiple sclerosis (MS) genetic factors are known to influence autoreactive T-cell-actions like proliferation and chemotaxis across the blood-brain barrier via chemokine receptors (CCR) and G-protein coupled activating mechanisms. For the first time, we studied the frequencies of a recently described C825T polymorphism in the G-protein encoding gene for the beta3 subunit (GNB3) together with frequencies of a 32-base-pair deletion in the CCR5 gene (delta32 CCR5) in patients with MS (n = 253: relapsing-remitting (RR), n = 124 and chronic progressive course, n = 129). Apart from a trend to a reduced frequency of delta32 CCR5 and increased GNB3 825T polymorphism in primary chronic progressive patients, numbers did not reach statistical significance in any group of MS. These results could not support differences in the genetic background of MS based on that CCR5 mutation or the described GNB3 polymorphism.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP/genética , Esclerose Múltipla/genética , Receptores CCR5/genética , Adolescente , Adulto , Idoso , DNA , Progressão da Doença , Feminino , Predisposição Genética para Doença , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Polimorfismo Genético
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA