Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Brain ; 146(3): 977-990, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35348614

RESUMO

Autoimmune neurological syndromes (AINS) with autoantibodies against the 65 kDa isoform of the glutamic acid decarboxylase (GAD65) present with limbic encephalitis, including temporal lobe seizures or epilepsy, cerebellitis with ataxia, and stiff-person-syndrome or overlap forms. Anti-GAD65 autoantibodies are also detected in autoimmune diabetes mellitus, which has a strong genetic susceptibility conferred by human leukocyte antigen (HLA) and non-HLA genomic regions. We investigated the genetic predisposition in patients with anti-GAD65 AINS. We performed a genome-wide association study (GWAS) and an association analysis of the HLA region in a large German cohort of 1214 individuals. These included 167 patients with anti-GAD65 AINS, recruited by the German Network for Research on Autoimmune Encephalitis (GENERATE), and 1047 individuals without neurological or endocrine disease as population-based controls. Predictions of protein expression changes based on GWAS findings were further explored and validated in the CSF proteome of a virtually independent cohort of 10 patients with GAD65-AINS and 10 controls. Our GWAS identified 16 genome-wide significant (P < 5 × 10-8) loci for the susceptibility to anti-GAD65 AINS. The top variant, rs2535288 [P = 4.42 × 10-16, odds ratio (OR) = 0.26, 95% confidence interval (CI) = 0.187-0.358], localized to an intergenic segment in the middle of the HLA class I region. The great majority of variants in these loci (>90%) mapped to non-coding regions of the genome. Over 40% of the variants have known regulatory functions on the expression of 48 genes in disease relevant cells and tissues, mainly CD4+ T cells and the cerebral cortex. The annotation of epigenomic marks suggested specificity for neural and immune cells. A network analysis of the implicated protein-coding genes highlighted the role of protein kinase C beta (PRKCB) and identified an enrichment of numerous biological pathways participating in immunity and neural function. Analysis of the classical HLA alleles and haplotypes showed no genome-wide significant associations. The strongest associations were found for the DQA1*03:01-DQB1*03:02-DRB1*04:01HLA haplotype (P = 4.39 × 10-4, OR = 2.5, 95%CI = 1.499-4.157) and DRB1*04:01 allele (P = 8.3 × 10-5, OR = 2.4, 95%CI = 1.548-3.682) identified in our cohort. As predicted, the CSF proteome showed differential levels of five proteins (HLA-A/B, C4A, ATG4D and NEO1) of expression quantitative trait loci genes from our GWAS in the CSF proteome of anti-GAD65 AINS. These findings suggest a strong genetic predisposition with direct functional implications for immunity and neural function in anti-GAD65 AINS, mainly conferred by genomic regions outside the classical HLA alleles.


Assuntos
Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos , Predisposição Genética para Doença/genética , Proteoma/genética , Antígenos de Histocompatibilidade Classe II , Antígenos HLA , Haplótipos , Alelos , Autoanticorpos , Cadeias HLA-DRB1/genética
2.
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
3.
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
4.
Eur J Neurol ; 29(5): 1303-1310, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34288284

RESUMO

BACKGROUND AND PURPOSE: Consensus criteria for autoimmune limbic encephalitis (ALE) allow for a diagnosis even without neuronal antibodies (Abs), but it remains unclear which clinical features should prompt neuronal Ab screening in temporal lobe epilepsy patients. The aim of the study was to investigate whether patients with temporal lobe seizures associated with additional symptoms or signs of limbic involvement may harbor neuronal Abs, and which clinical features should prompt neuronal Ab screening in these patients. METHODS: We identified 47 patients from a tertiary epilepsy center with mediotemporal lobe seizures and additional features suggestive of limbic involvement, including either memory deficits, psychiatric symptoms, mediotemporal magnetic resonance imaging (MRI) hyperintensities or inflammatory cerebrospinal fluid (CSF). Neuronal Ab testing was carried out at two independent reference laboratories (Bielefeld-Bethel, Germany, and Barcelona, Spain). All brain MRI scans were assessed by two reviewers independently. RESULTS: Temporal lobe seizures were accompanied by memory deficits in 35/46 (76%), psychiatric symptoms in 27/42 (64%), and both in 19/42 patients (45%). Limbic T2/fluid-attenuated inversion recovery signal hyperintensities were found in 26/46 patients (57%; unilateral: n = 22, bilateral: n = 4). Standard CSF studies were abnormal in 2/37 patients (5%). Neuronal Abs were confirmed in serum and/or CSF in 8/47 patients (17%) and were directed against neuronal cell-surface targets (leucine-rich glioma inactivated protein 1: n = 1, contactin-associated protein-2: n = 1, undetermined target: n = 3) or glutamic acid decarboxylase in its 65-kD isoform (n = 3, all with high titers). Compared to Ab-negative patients, those who harbored neuronal Abs were more likely to have uni- or bilateral mediotemporal MRI changes (8/8, 100% vs. 18/38, 47%; p = 0.01, Fisher's exact test). CONCLUSIONS: In patients with temporal lobe seizures and additional limbic signs, 17% had neuronal Abs affirming ALE diagnosis. Mediotemporal MRI changes were found in all Ab-positive cases and had a positive likelihood ratio of 2.11 (95% confidence interval 1.51-2.95).


Assuntos
Epilepsia do Lobo Temporal , Encefalite Límbica , Autoanticorpos , Doenças Autoimunes , Epilepsia do Lobo Temporal/complicações , Epilepsia do Lobo Temporal/diagnóstico por imagem , Humanos , Encefalite Límbica/diagnóstico , Encefalite Límbica/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Transtornos da Memória/complicações , Convulsões/complicações , Convulsões/etiologia , Lobo Temporal/diagnóstico por imagem
5.
Int J Mol Sci ; 23(7)2022 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-35409067

RESUMO

Autoimmune encephalitis associated with antibodies (Abs) against α1, ß3, and γ2 subunits of γ-aminobutyric acid receptor A (GABAAR) represents a severe form of encephalitis with refractory seizures and status epilepticus. Reduction in inhibitory GABAergic synaptic activity is linked to dysfunction of neuronal networks, hyperexcitability, and seizures. The aim in this study was to investigate the direct pathogenic effect of a recombinant GABAAR autoantibody (rAb-IP2), derived from the cerebrospinal fluid (CSF) of a patient with autoimmune GABAAR encephalitis, on hippocampal CA1 and CA3 networks. Acute brain slices from C57BL/6 mice were incubated with rAb-IP2. The spontaneous synaptic GABAergic transmission was measured using electrophysiological recordings in voltage-clamp mode. The GABAAR autoantibody rAb-IP2 reduced inhibitory postsynaptic signaling in the hippocampal CA1 pyramidal neurons with regard to the number of spontaneous inhibitory postsynaptic currents (sIPSCs) but did not affect their amplitude. In the hippocampal CA3 network, decreased number and amplitude of sIPSCs were detected, leading to decreased GABAergic synaptic transmission. Immunohistochemical staining confirmed the rAb-IP2 bound to hippocampal tissue. These findings suggest that GABAAR autoantibodies exert direct functional effects on both hippocampal CA1 and CA3 pyramidal neurons and play a crucial role in seizure generation in GABAAR autoimmune encephalitis.


Assuntos
Encefalite , Receptores de GABA-A , Animais , Autoanticorpos/metabolismo , Encefalite/metabolismo , Doença de Hashimoto , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Células Piramidais/metabolismo , Receptores de GABA-A/metabolismo , Convulsões/metabolismo , Transmissão Sináptica , Ácido gama-Aminobutírico/metabolismo
6.
J Neuroinflammation ; 18(1): 260, 2021 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-34749759

RESUMO

Autoimmune encephalitis (AE) is an inflammatory brain disease which is frequently associated with antibodies (Abs) against cell-surface, synaptic or intracellular neuronal proteins. There is increasing evidence that dendritic cells (DCs) are implicated as key modulators in keeping the balance between immune response and tolerance in the CNS. Migratory features of DCs to and from the brain are linked to initiating and maintaining of neuroinflammation. Genetic polymorphisms together with other triggers such as systemic or cerebral viral infection, or systemic malignancies could contribute to the dysbalance of "regulatory" and "encephalitogenic" DCs with subsequent dysregulated T and B cell reactions in AE. Novel in vivo models with implantation of mature DCs containing neuronal antigens could help to study the pathogenesis and perhaps to understand the origin of AE. Investigations of DCs in human blood, lymphoid tissues, CSF, and brain parenchyma of patients with AE are necessary to deepen our knowledge about the complex interactions between DCs, T and B cells during neuroinflammation in AE. This can support developing new therapy strategies.


Assuntos
Doenças Autoimunes/imunologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Encefalite/imunologia , Doenças Neuroinflamatórias/imunologia , Animais , Doenças Autoimunes/patologia , Encefalite/patologia , Humanos , Doenças Neuroinflamatórias/patologia
7.
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
8.
BMC Neurol ; 21(1): 386, 2021 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-34615476

RESUMO

BACKGROUND: Posterior reversible encephalopathy syndrome (PRES) is clinical-neuroradiologically defined and potentially reversible, so there are limited data about histopathological findings. We aimed to describe the clinical and paraclinical features of patients with PRES with regard to its reversibility. METHODS: This retrospective case series encompasses 15 PRES cases out of 1300 evaluated patients from a single German center between January 1, 2010, and June 15, 2020. PRES was established according to the diagnostic criteria as proposed by the Berlin PRES Study 2012. One of the cases studied was subject to brain autopsy. RESULTS: From the 15 patients studied (median age 53 years, range 17-73; 11 female), 67 % presented with epileptic seizures, 40 % suffered from encephalopathy with reduced consciousness and 53 % developed delirium, while 47 % had headache and visual disturbances. Subcortical brain MRI abnormalities related to PRES were observed in all patients. One patient developed spinal ischemia and another Guillain-Barré syndrome in addition to PRES. Hypertensive blood pressure was the main underlying/trigger condition in all patients. Clinical symptoms and MRI changes were not reversible in 42 %, even progressive in 3 out of these 5 patients. Median time from symptom onset to diagnosis in these non-reversible cases was 7 days (range 0-13), while the median delay in diagnosis in the reversible group was 1 day (range 0-3). Cerebellar/brain stem involvement and status epilepticus were more frequently in patients with non-reversible disease course. Mortality due to PRES occurred in 13 % of these patients. Neuropathological examination of the brain of a 57-year-old female patient revealed major leukencephalopathic changes, fibrinoid necrosis of endothelial cells and fresh petechial hemorrhages in accordance with PRES. CONCLUSIONS: Our case series demonstrates that PRES was not reversible in 42 % of the studied patients. Delay in diagnosis seems to contribute to limited reversibility and poor outcome.


Assuntos
Encefalopatias , Síndrome da Leucoencefalopatia Posterior , Adolescente , Adulto , Idoso , Células Endoteliais , Feminino , Humanos , Imageamento por Ressonância Magnética , Pessoa de Meia-Idade , Síndrome da Leucoencefalopatia Posterior/diagnóstico por imagem , Estudos Retrospectivos , Adulto Jovem
9.
J Neurooncol ; 131(3): 469-476, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27848138

RESUMO

Gap junctions (GJs) in astrocytes and glioma cells are important channels for cell-to-cell communication that contribute to homo- and heterocellular coupling. According to recent studies, heterocellular gap-junctional communication (H-GJC) between glioma cells and their surrounding environment enhances glioma progression. Therefore, we developed a new in vitro model to examine H-GJC between glioma cells, astrocytes and microglia. Consequently, F98 rat glioma cells were double-labeled with GJ-impermeable (CM-DiI) and GJ-permeable dye (calcein AM) and were seeded on unlabeled astrocyte-microglia co-cultures. Dual whole cell voltage clamp recordings were carried out on selected cell pairs to characterize the functional properties of H-GJC in vitro. The expression of four types of connexins (Cxs), including Cx32, Cx36, Cx43 and Cx45, and microglial phenotypes were analyzed by immunocytochemistry. The H-GJC between glioma cells and astrocytes/microglia increased after a longer incubation period with a higher number of glioma cells. We provided evidence for the direct GJ coupling of microglia and glioma cells under native in vitro conditions. In addition, we exploited this model to evaluate H-GJC after incubation with levetiracetam (LEV) and/or dexamethasone (DEX). Previous in vitro studies suggest that LEV and DEX are frequently used to control seizure and edema in glioma. Our findings showed that LEV and/or DEX decrease the number of heterocellular coupled cells significantly. In conclusion, our newly developed model demonstrated H-GJC between glioma cells and both astrocytes and microglia. The reduced H-GJC by LEV and DEX suggests a potential effect of both drugs on glioma progression.


Assuntos
Antineoplásicos/farmacologia , Comunicação Celular/efeitos dos fármacos , Dexametasona/farmacologia , Junções Comunicantes/efeitos dos fármacos , Glioma/fisiopatologia , Neuroglia/fisiologia , Piracetam/análogos & derivados , Animais , Antineoplásicos/uso terapêutico , Astrócitos/fisiologia , Linhagem Celular Tumoral , Conexina 43/metabolismo , Conexinas/metabolismo , Dexametasona/uso terapêutico , Glioma/tratamento farmacológico , Técnicas In Vitro , Levetiracetam , Microglia/fisiologia , Neuroglia/efeitos dos fármacos , Piracetam/farmacologia , Piracetam/uso terapêutico , Ratos , Células Tumorais Cultivadas , Proteína beta-1 de Junções Comunicantes , Proteína delta-2 de Junções Comunicantes
11.
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
12.
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
13.
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.

15.
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
16.
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.

17.
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).

18.
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
20.
Seizure ; 51: 6-8, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28750305

RESUMO

PURPOSE: Survivors of viral encephalitis can develop refractory epilepsy and hippocampal sclerosis. Both the initial infectious insult and the secondary effects of recurrent seizures have been implicated in chronic disease progression. Recently, post-infectious autoimmunity, involved in acute relapses, has also been proposed as a pathomechanism for chronic disease progression. Our case series suggests a potential role of antibodies against the N-methyl-d-aspartate receptor (NMDAR) in chronic inflammatory disease beyond acute manifestations. METHODS: Retrospective chart review of four patients with epilepsy, hippocampal sclerosis following viral encephalitis and NMDAR-antibodies in CSF. RESULTS: The four patients were female, developed hippocampal sclerosis (in 3/4 in a step-wise progression) after Herpes simplex or Varicella zoster virus encephalitis and harboured immunoglobulin G antibodies against the NMDAR in their CSF. Two patients were treated with short-term immunosuppression but did not benefit. CONCLUSION: This case series presents the first tentative evidence in support of chronic autoimmune inflammation driving disease progression after viral encephalitis beyond the known acute immune-mediated relapses. The anecdotal nature of the data does not, however, permit conclusive judgement on causality. Should our findings be replicated in larger cohorts, the treatment of post-infectious epilepsy could potentially be expanded to include immunosuppressive strategies in antibody-positive cases.


Assuntos
Autoanticorpos/imunologia , Encefalite por Herpes Simples/imunologia , Encefalite por Varicela Zoster/imunologia , Epilepsia/imunologia , Receptores de N-Metil-D-Aspartato/imunologia , Adolescente , Adulto , Autoantígenos/imunologia , Criança , Encefalite por Herpes Simples/complicações , Encefalite por Varicela Zoster/complicações , Feminino , Hipocampo/patologia , Humanos , Pessoa de Meia-Idade , Estudos Retrospectivos , Esclerose/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA