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










Base de datos
Intervalo de año de publicación
2.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 3253-3267, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37231170

RESUMEN

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.


Asunto(s)
Astrocitos , Microglía , Ratas , Animales , Técnicas de Cocultivo , Tiagabina/metabolismo , Tiagabina/farmacología , Conexina 43/metabolismo , Zonisamida/farmacología , Zonisamida/metabolismo , Comunicación Celular , Neuroglía/metabolismo , Inflamación/patología
3.
Epilepsia Open ; 8(2): 692-701, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37078803

RESUMEN

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.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso , Encefalitis , Animales , Humanos , Receptores de GABA-A/metabolismo , Astrocitos/metabolismo , Convulsiones , Ácido gamma-Aminobutírico/fisiología , Proteínas Portadoras , Anticuerpos/metabolismo
4.
Brain Sci ; 13(3)2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36979303

RESUMEN

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.

5.
Pharmacology ; 108(2): 188-198, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36724743

RESUMEN

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.


Asunto(s)
Dimetilfumarato , Microglía , Ratas , Animales , Dimetilfumarato/metabolismo , Dimetilfumarato/farmacología , Microglía/metabolismo , Astrocitos , Conexina 43/metabolismo , Conexina 43/farmacología , Técnicas de Cocultivo , Inflamación/metabolismo
6.
Front Cell Neurosci ; 16: 995861, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36406753

RESUMEN

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.

7.
Int J Neuropsychopharmacol ; 25(3): 185-196, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-34791253

RESUMEN

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.


Asunto(s)
Anticonvulsivantes , Epilepsia , Animales , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Astrocitos/metabolismo , Técnicas de Cocultivo , Conexina 43/metabolismo , Citocinas/metabolismo , Inflamación/metabolismo , Lamotrigina/metabolismo , Lamotrigina/farmacología , Lamotrigina/uso terapéutico , Microglía , Ratas , Topiramato/farmacología , Topiramato/uso terapéutico , Factor de Necrosis Tumoral alfa
8.
Eur J Pharmacol ; 915: 174696, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34902360

RESUMEN

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.


Asunto(s)
Astrocitos
10.
BMC Neurosci ; 22(1): 21, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33765917

RESUMEN

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.


Asunto(s)
Cloruro de Amonio/toxicidad , Acuaporina 4/metabolismo , Conexina 43/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Animales , Astrocitos/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Encefalopatía Hepática/metabolismo , Ratas
11.
Front Cell Neurosci ; 15: 805755, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34975415

RESUMEN

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).

12.
Med Hypotheses ; 139: 109612, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32085980

RESUMEN

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.


Asunto(s)
Encefalitis Antirreceptor N-Metil-D-Aspartato , Enfermedad de Hashimoto , Animales , Astrocitos , Células Cultivadas , Ratones , Ratas , Receptores de N-Metil-D-Aspartato
13.
Behav Brain Res ; 291: 325-333, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26048428

RESUMEN

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.


Asunto(s)
Lateralidad Funcional/fisiología , Reconocimiento Visual de Modelos/fisiología , Lectura , Percepción del Habla/fisiología , Corteza Visual/fisiología , Mapeo Encefálico , Electroencefalografía , Potenciales Evocados , Femenino , Humanos , Pruebas del Lenguaje , Masculino , Pruebas Neuropsicológicas , Lóbulo Parietal/fisiología , Campos Visuales , Adulto Joven
14.
Front Physiol ; 5: 186, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24904426

RESUMEN

Gap junctions (GJs) are hemichannels on cell membrane. Once they are intercellulary connected to the neighboring cells, they build a functional syncytium which allows rapid transfer of ions and molecules between cells. This characteristic makes GJs a potential modulator in proliferation, migration, and development of the cells. So far, several types of GJs are recognized on different brain cells as well as in glioma. Astrocytes, as one of the major cells that maintain neuronal homeostasis, express different types of GJs that let them communicate with neurons, oligodendrocytes, and endothelial cells of the blood brain barrier; however, the main GJ in astrocytes is connexin 43. There are different cerebral diseases in which astrocyte GJs might play a role. Several drugs have been reported to modulate gap junctional communication in the brain which can consequently have beneficial or detrimental effects on the course of treatment in certain diseases. However, the exact cellular mechanism behind those pharmaceutical efficacies on GJs is not well-understood. Accordingly, how specific drugs would affect GJs and what some consequent specific brain diseases would be are the interests of the authors of this chapter. We would focus on pharmaceutical effects on GJs on astrocytes in specific diseases where GJs could possibly play a role including: (1) migraine and a novel therapy for migraine with aura, (2) neuroautoimmune diseases and immunomodulatory drugs in the treatment of demyelinating diseases of the central nervous system such as multiple sclerosis, (3) glioma and antineoplastic and anti-inflammatory agents that are used in treating brain tumors, and (4) epilepsy and anticonvulsants that are widely used for seizures therapy. All of the above-mentioned therapeutic categories can possibly affect GJs expression of astrocytes and the role is discussed in the upcoming chapter.

15.
Neuroimmunomodulation ; 21(4): 189-94, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24504116

RESUMEN

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.


Asunto(s)
Autoanticuerpos/sangre , Encefalitis/inmunología , Esclerosis Múltiple Recurrente-Remitente/inmunología , Receptores de Glutamato/inmunología , Adulto , Autoantígenos/inmunología , Encefalitis/sangre , Femenino , Humanos , Immunoblotting , Masculino , Esclerosis Múltiple Recurrente-Remitente/sangre
16.
Epilepsia ; 55(1): 184-92, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24299259

RESUMEN

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.


Asunto(s)
Anticonvulsivantes/farmacología , Astrocitos/fisiología , Epilepsia/etiología , Inflamación/fisiopatología , Microglía/fisiología , Neuroglía/fisiología , Aminas/farmacología , Aminas/uso terapéutico , Animales , Anticonvulsivantes/uso terapéutico , Astrocitos/efectos de los fármacos , Western Blotting , Carbamazepina/farmacología , Carbamazepina/uso terapéutico , Células Cultivadas , Técnicas de Cocultivo , Conexina 43/biosíntesis , Ácidos Ciclohexanocarboxílicos/farmacología , Ácidos Ciclohexanocarboxílicos/uso terapéutico , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Epilepsia/tratamiento farmacológico , Epilepsia/fisiopatología , Gabapentina , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/fisiología , Inflamación/tratamiento farmacológico , Microglía/efectos de los fármacos , Neuroglía/efectos de los fármacos , Fenitoína/farmacología , Fenitoína/uso terapéutico , Ratas , Ratas Wistar , Factor de Crecimiento Transformador beta1/biosíntesis , Ácido Valproico/farmacología , Ácido Valproico/uso terapéutico , Ácido gamma-Aminobutírico/farmacología , Ácido gamma-Aminobutírico/uso terapéutico
17.
Int Immunopharmacol ; 14(1): 9-12, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22691576

RESUMEN

Levetiracetam (LEV) is an established anticonvulsant with numerous mechanisms of action. Apart from its anti-epileptic effects, recent experimental studies suggest anti-inflammatory properties via modulation of interleukin (IL)-1ß and transforming-growth-factor (TGF)-ß1. However, its anti-inflammatory properties have not yet been examined in an autoimmune inflammatory disease of the central nervous system (CNS). We investigated LEV anti-inflammatory properties in experimental autoimmune encephalomyelitis, an established mouse model of multiple sclerosis. FACS analyses, ELISA, histology and rt-PCR experiments were done to explore potential anti-inflammatory effects. In line with prior studies, we demonstrate that LEV modulates both the relative gene expression and secretion of IL-1ß and TGF-1ß. However, these changes were not sufficient to alter the disease course or histological parameters. Additionally, LEV showed no effects on the absolute number of different immune cell subsets. In summary, LEV showed only minor anti-inflammatory effects not sufficient to ameliorate disease course in an autoimmune inflammatory disease of CNS.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Esclerosis Múltiple/tratamiento farmacológico , Neuroglía/efectos de los fármacos , Piracetam/análogos & derivados , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Inmunomodulación , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Levetiracetam , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Glicoproteína Mielina-Oligodendrócito/inmunología , Neuroglía/inmunología , Fragmentos de Péptidos/inmunología , Piracetam/administración & dosificación , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
18.
PLoS One ; 6(6): e20936, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21731632

RESUMEN

Suicide is the most important incident in psychiatric disorders. Psychological pain and empathy to pain involves a neural network that involves the anterior cingulate cortex (ACC) and the anterior insula (AI). At the neuronal level, little is known about how complex emotions such as shame, guilt, self-derogation and social isolation, all of which feature suicidal behavior, are represented in the brain. Based on the observation that the ACC and the AI contain a large spindle-shaped cell type, referred to as von Economo neuron (VEN), which has dramatically increased in density during human evolution, and on growing evidence that VENs play a role in the pathophysiology of various neuropsychiatric disorders, including autism, psychosis and dementia, we examined the density of VENs in the ACC of suicide victims. The density of VENs was determined using cresyl violet-stained sections of the ACC of 39 individuals with psychosis (20 cases with schizophrenia, 19 with bipolar disorder). Nine subjects had died from suicide. Twenty specimen were available from the right, 19 from the left ACC. The density of VENs was significantly greater in the ACC of suicide victims with psychotic disorders compared with psychotic individuals who died from other causes. This effect was restricted to the right ACC. VEN density in the ACC seems to be increased in suicide victims with psychosis. This finding may support the assumption that VEN have a special role in emotion processing and self-evaluation, including negative self-appraisal.


Asunto(s)
Encéfalo/patología , Neuronas/patología , Trastornos Psicóticos/patología , Suicidio , Adulto , Trastorno Bipolar/patología , Demografía , Femenino , Giro del Cíngulo/patología , Humanos , Masculino , Persona de Mediana Edad , Esquizofrenia/patología
19.
Br J Pharmacol ; 162(2): 491-507, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20955362

RESUMEN

BACKGROUND AND PURPOSE: The involvement of astrocytes as immune-competent players in inflammation and the pathogenesis of epilepsy and seizure-induced brain damage has recently been recognized. In clinical trials and practice, levetiracetam (LEV) has proven to be an effective antiepileptic drug (AED) in various forms of epileptic seizures, when applied as mono- or added therapy. Little is known about the mechanism(s) of action of LEV. Evidence so far suggests a mode of action different from that of classical AEDs. We have shown that LEV restored functional gap junction coupling and basic membrane properties in an astrocytic inflammatory model in vitro. EXPERIMENTAL APPROACH: Here, we used neonatal rat astrocytes co-cultured with high proportions (30%) of activated microglia or treated with the pro-inflammatory cytokine interleukin-1ß to provoke inflammatory responses. Effects of LEV (50 µg·mL⁻¹) on electrophysiological properties of astrocytes (by whole cell patch clamp) and on secretion of TGFß1 (by (ELISA)) were studied in these co-cultures. KEY RESULTS: LEV restored impaired astrocyte membrane resting potentials via modification of inward and outward rectifier currents, and promoted TGFß1 expression in inflammatory and control co-cultures. Furthermore, LEV and TGFß1 exhibited similar facilitating effects on the generation of astrocyte voltage-gated currents in inflammatory co-cultures and the effects of LEV were prevented by antibody to TGFß1. CONCLUSIONS AND IMPLICATIONS: Our data suggest that LEV is likely to reduce the harmful spread of excitation elicited by seizure events within the astro-glial functional syncytium, with stabilizing consequences for neuronal-glial interactions.


Asunto(s)
Antiinflamatorios/farmacología , Anticonvulsivantes/farmacología , Astrocitos/efectos de los fármacos , Fenómenos Electrofisiológicos/efectos de los fármacos , Piracetam/análogos & derivados , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Astrocitos/metabolismo , Técnicas de Cocultivo , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Levetiracetam , Potenciales de la Membrana/efectos de los fármacos , Microglía/metabolismo , Técnicas de Placa-Clamp , Piracetam/farmacología , Ratas , Ratas Wistar , Factor de Crecimiento Transformador beta1/farmacología
20.
J Neurooncol ; 103(3): 479-89, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21107646

RESUMEN

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.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Astrocitos/fisiología , Neoplasias Encefálicas/fisiopatología , Dexametasona/farmacología , Glioma/fisiopatología , Potenciales de la Membrana/efectos de los fármacos , Animales , Animales Recién Nacidos , Antineoplásicos Hormonales/uso terapéutico , Astrocitos/efectos de los fármacos , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo/métodos , Conexina 43/metabolismo , Dexametasona/uso terapéutico , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Ectodisplasinas/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/patología , Antagonistas de Hormonas/farmacología , Humanos , Potenciales de la Membrana/fisiología , Mifepristona/farmacología , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Estadísticas no Paramétricas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA