Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
2.
Int J Mol Sci ; 23(15)2022 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-35897767

RESUMEN

Endogenous anticonvulsant mechanisms represent a reliable and currently underdeveloped strategy against recurrent seizures and may recall novel original therapeutics. Here, we investigated whether the intensification of the astroglial Glu-GABA exchange mechanism by application of the GABA precursor putrescine (PUT) may be effective against convulsive and non-convulsive seizures. We explored the potential of PUT to inhibit spontaneous spike-and-wave discharges (SWDs) in WAG/Rij rats, a genetic model of absence epilepsy. Significant shortening of SWDs in response to intraperitoneally applied PUT has been observed, which could be antagonized by blocking GAT-2/3-mediated astrocytic GABA release with the specific inhibitor SNAP-5114. Direct application of exogenous GABA also reduced SWD duration, suggesting that PUT-triggered astroglial GABA release through GAT-2/3 may be a critical step in limiting seizure duration. PUT application also dose-dependently shortened seizure-like events (SLEs) in the low-[Mg2+] in vitro model of temporal lobe epilepsy. SNAP-5114 reversed the antiepileptic effect of PUT in the in vitro model as well, further confirming that PUT reduces seizure duration by triggering glial GABA release. In accordance, we observed that PUT specifically reduces the frequency of excitatory synaptic potentials, suggesting that it specifically acts at excitatory synapses. We also identified that PUT specifically eliminated the tonic depolarization-induced desynchronization of SLEs. Since PUT is an important source of glial GABA and we previously showed significant GABA release, it is suggested that the astroglial Glu-GABA exchange mechanism plays a key role in limiting ictal discharges, potentially opening up novel pathways to control seizure propagation and generalization.


Asunto(s)
Electroencefalografía , Putrescina , Animales , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Modelos Animales de Enfermedad , Ratas , Convulsiones , Ácido gamma-Aminobutírico
3.
Cell Commun Signal ; 16(1): 80, 2018 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-30419921

RESUMEN

Following publication of the original article [1], the authors reported an error in Table 3. The correct version of Table 3 is shown below:The publishers apologise for this error. The original article [1] has been corrected.

4.
Cell Commun Signal ; 16(1): 71, 2018 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-30348177

RESUMEN

Copper-containing enzymes perform fundamental functions by activating dioxygen (O2) and therefore allowing chemical energy-transfer for aerobic metabolism. The copper-dependence of O2 transport, metabolism and production of signalling molecules are supported by molecular systems that regulate and preserve tightly-bound static and weakly-bound dynamic cellular copper pools. Disruption of the reducing intracellular environment, characterized by glutathione shortage and ambient Cu(II) abundance drives oxidative stress and interferes with the bidirectional, copper-dependent communication between neurons and astrocytes, eventually leading to various brain disease forms. A deeper understanding of of the regulatory effects of copper on neuro-glia coupling via polyamine metabolism may reveal novel copper signalling functions and new directions for therapeutic intervention in brain disorders associated with aberrant copper metabolism.


Asunto(s)
Cobre/metabolismo , Transducción de Señal , Animales , Homeostasis , Humanos , Oxidación-Reducción , Sinapsis/metabolismo
5.
Glia ; 64(10): 1655-66, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26566753

RESUMEN

The transmembrane Na(+) concentration gradient is an important source of energy required not only to enable the generation of action potentials in excitable cells, but also for various transmembrane transporters both in excitable and non-excitable cells, like astrocytes. One of the vital functions of astrocytes in the central nervous system (CNS) is to regulate neurotransmitter concentrations in the extracellular space. Most neurotransmitters in the CNS are removed from the extracellular space by Na(+) -dependent neurotransmitter transporters (NeuTs) expressed both in neurons and astrocytes. Neuronal NeuTs control mainly phasic synaptic transmission, i.e., synaptically induced transient postsynaptic potentials, while astrocytic NeuTs contribute to the termination of phasic neurotransmission and modulate the tonic tone, i.e., the long-lasting activation of extrasynaptic receptors by neurotransmitter that has diffused out of the synaptic cleft. Consequently, local intracellular Na(+) ([Na(+) ]i ) transients occurring in astrocytes, for example via the activation of ionotropic neurotransmitter receptors, can affect the driving force for neurotransmitter uptake, in turn modulating the spatio-temporal profiles of neurotransmitter levels in the extracellular space. As some NeuTs are close to thermodynamic equilibrium under resting conditions, an increase in astrocytic [Na(+) ]i can stimulate the direct release of neurotransmitter via NeuT reversal. In this review we discuss the role of astrocytic [Na(+) ]i changes in the regulation of uptake/release of neurotransmitters. It is emphasized that an activation of one neurotransmitter system, including either its ionotropic receptor or Na(+) -coupled co-transporter, can strongly influence, or even reverse, other Na(+) -dependent NeuTs, with potentially significant consequences for neuronal communication. GLIA 2016;64:1655-1666.


Asunto(s)
Astrocitos/fisiología , Transducción de Señal/fisiología , Sodio/metabolismo , Transmisión Sináptica/fisiología , Animales
6.
Bioorg Med Chem Lett ; 26(2): 417-423, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26706177

RESUMEN

Supply of major metabolites such as γ-aminobutyric acid (GABA), ß-alanine and taurine is an essential instrument that shapes signalling, proper cell functioning and survival in the brain and peripheral organs. This background motivates the synthesis of novel classes of compounds regulating their selective transport through various fluid-organ barriers via the low-affinity γ-aminobutyric acid (GABA) transporter subtype 2 (GAT2). Natural and synthetic spirocyclic compounds or therapeutics with a range of structures and biological activity are increasingly recognised in this regard. Based on pre-validated GABA transport activity, straightforward and efficient synthesis method was developed to provide an azaspiro[4.5]decane scaffold, holding a variety of charge, substituent and 3D constrain of spirocyclic amine. Investigation of the azaspiro[4.5]decane scaffold in cell lines expressing the four GABA transporter subtypes led to the discovery of a subclass of a GAT2-selective compounds with acyl-substituted azaspiro[4.5]decane core.


Asunto(s)
Alcanos/química , Alcanos/farmacología , Compuestos Aza/química , Compuestos Aza/farmacología , Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Compuestos de Espiro/química , Compuestos de Espiro/farmacología , Acilación , Alcanos/síntesis química , Animales , Compuestos Aza/síntesis química , Humanos , Compuestos de Espiro/síntesis química , Ácido gamma-Aminobutírico/metabolismo
7.
Langmuir ; 31(28): 7815-25, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26099064

RESUMEN

Polyamidoamine (PAMAM) dendrimers are hyperbranched, nanosized polymers with promising biomedical applications as nanocarriers in targeted drug delivery and gene therapy. For the development of safe dendrimer-based biomedical applications it is necessary to gain an understanding of the detailed mechanism of the interactions of both cationic and anionic dendrimers with cell membranes. To characterize dendrimer-membrane interactions we applied solid-supported lipid bilayers as biomembrane models and utilized infrared-visible sum-frequency vibrational spectroscopy to independently probe the interactions of cationic G5-NH2 and anionic G4.5-COONa dendrimers with the two leaflets of the lipid bilayers. Interaction with both dendrimers led to changes in the interfacial water structure and charge density as evidenced by the changes in the OH band intensities in the sum-frequency spectra of the bilayers. Interaction with the G5-NH2 dendrimer also led to a unique inversion of the sign of the OH-stretch amplitudes, in addition to a decrease in their absolute values. We suggest that the positively charged amino groups on the G5-NH2 dendrimer surface bind to the negatively charged bilayer, while uncompensated positive charges not involved in the binding cause a reversal of the electric field and thus an opposite orientation of the interfacial water molecules. More subtle but nonetheless significant changes were seen in the relative magnitudes of the CH amplitudes. The methyl antisymmetric to symmetric stretch amplitude ratios are altered, implying changes in the tilt angles of the phospholipid alkyl chains. The conformational order of the phospholipid alkyl chains of both leaflets is also influenced by the G5-NH2 dendrimer while G4.5-COONa has no effect on the alkyl chain conformation.


Asunto(s)
Dendrímeros/química , Membrana Dobles de Lípidos/química , Vibración , Modelos Moleculares , Conformación Molecular , Espectrofotometría Infrarroja
8.
Biochim Biophys Acta ; 1828(8): 1873-80, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23597947

RESUMEN

Polyamidoamine (PAMAM) dendrimers are highly charged hyperbranched protein-like polymers that are known to interact with cell membranes. In order to disclose the mechanisms of dendrimer-membrane interaction, we monitored the effect of PAMAM generation five (G5) dendrimer on the membrane permeability of living neuronal cells followed by exploring the underlying structural changes with infrared-visible sum frequency vibrational spectroscopy (SVFS), small angle X-ray scattering (SAXS) and transmission electron microscopy (TEM). G5 dendrimers were demonstrated to irreversibly increase the membrane permeability of neurons that could be blocked in low-[Na(+)], but not in low-[Ca(2+)] media suggesting the formation of specific Na(+) permeable channels. SFVS measurements on silica supported DPPG-DPPC bilayers suggested G5-specific trans-polarization of the membrane. SAXS data and freeze-fracture TEM imaging of self-organized DPPC vesicle systems demonstrated disruption of DPPC vesicle layers by G5 through polar interactions between G5 terminal amino groups and the anionic head groups of DPPC. We propose a nanoscale mechanism by which G5 incorporates into the membrane through multiple polar interactions that disrupt proximate membrane bilayer and shape a unique hydrophilic Na(+) ion permeable channel around the dendrimer. In addition, we tested whether these artificial Na(+) channels can be exploited as antibiotic tools. We showed that G5 quickly arrest the growth of resistant bacterial strains below 10µg/ml concentration, while they show no detrimental effect on red blood cell viability, offering the chance for the development of new generation anti-resistant antibiotics.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Membrana Celular/metabolismo , Dendrímeros/metabolismo , Hipocampo/metabolismo , Poliaminas/metabolismo , Canales de Sodio/metabolismo , Sodio/metabolismo , Animales , Membrana Celular/química , Supervivencia Celular , Células Cultivadas , Dendrímeros/química , Eritrocitos/metabolismo , Escherichia coli/metabolismo , Hipocampo/citología , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Masculino , Microscopía Electrónica de Transmisión , Neuronas/citología , Neuronas/metabolismo , Técnicas de Placa-Clamp , Poliaminas/química , Ratas , Ratas Wistar , Sodio/química , Análisis Espectral
9.
J Nanobiotechnology ; 11: 9, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23556550

RESUMEN

BACKGROUND: The potential nanocarrier polyamidoamine (PAMAM) generation 5 (G5-NH(2)) dendrimer has been shown to evoke lasting neuronal depolarization and cell death in a concentration-dependent manner. In this study we explored the early progression of G5-NH(2) action in brain tissue on neuronal and astroglial cells. RESULTS: In order to describe early mechanisms of G5-NH(2) dendrimer action in brain tissue we assessed G5-NH(2) trafficking, free intracellular Ca(2+) and mitochondrial membrane potential (Ψ(MITO)) changes in the rat hippocampal slice by microfluorimetry. With the help of fluorescent dye conjugated G5-NH(2), we observed predominant appearance of the dendrimer in the plasma membrane of pyramidal neurons and glial cells within 30 min. Under this condition, G5-NH(2) evoked robust intracellular Ca(2+) enhancements and Ψ(MITO) depolarization both in pyramidal neurons and astroglial cells. Intracellular Ca(2+) enhancements clearly preceded Ψ(MITO) depolarization in astroglial cells. Comparing activation dynamics, neurons and glia showed prevalence of lasting and transient Ψ(MITO) depolarization, respectively. Transient as opposed to lasting Ψ(MITO) changes to short-term G5-NH(2) application suggested better survival of astroglia, as observed in the CA3 stratum radiatum area. We also showed that direct effect of G5-NH(2) on astroglial Ψ(MITO) was significantly enhanced by neuron-astroglia interaction, subsequent to G5-NH(2) evoked neuronal activation. CONCLUSION: These findings indicate that the interaction of the PAMAM dendrimer with the plasma membrane leads to robust activation of neurons and astroglial cells, leading to mitochondrial depolarization. Distinguishable dynamics of mitochondrial depolarization in neurons and astroglia suggest that the enhanced mitochondrial depolarization followed by impaired oxidative metabolism of neurons may be the primary basis of neurotoxicity.


Asunto(s)
Dendrímeros/toxicidad , Hipocampo/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Poliaminas/toxicidad , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/patología , Señalización del Calcio , Membrana Celular/química , Supervivencia Celular/efectos de los fármacos , Dendrímeros/química , Colorantes Fluorescentes/análisis , Colorantes Fluorescentes/metabolismo , Hipocampo/metabolismo , Masculino , Mitocondrias/patología , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/patología , Poliaminas/química , Ratas , Ratas Wistar
10.
BMC Biol ; 10: 26, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22420899

RESUMEN

BACKGROUND: Glutamate and γ-aminobutyric acid (GABA) transporters play important roles in balancing excitatory and inhibitory signals in the brain. Increasing evidence suggest that they may act concertedly to regulate extracellular levels of the neurotransmitters. RESULTS: Here we present evidence that glutamate uptake-induced release of GABA from astrocytes has a direct impact on the excitability of pyramidal neurons in the hippocampus. We demonstrate that GABA, synthesized from the polyamine putrescine, is released from astrocytes by the reverse action of glial GABA transporter (GAT) subtypes GAT-2 or GAT-3. GABA release can be prevented by blocking glutamate uptake with the non-transportable inhibitor DHK, confirming that it is the glutamate transporter activity that triggers the reversal of GABA transporters, conceivably by elevating the intracellular Na+ concentration in astrocytes. The released GABA significantly contributes to the tonic inhibition of neurons in a network activity-dependent manner. Blockade of the Glu/GABA exchange mechanism increases the duration of seizure-like events in the low-[Mg2+] in vitro model of epilepsy. Under in vivo conditions the increased GABA release modulates the power of gamma range oscillation in the CA1 region, suggesting that the Glu/GABA exchange mechanism is also functioning in the intact hippocampus under physiological conditions. CONCLUSIONS: The results suggest the existence of a novel molecular mechanism by which astrocytes transform glutamatergic excitation into GABAergic inhibition providing an adjustable, in situ negative feedback on the excitability of neurons.


Asunto(s)
Astrocitos/metabolismo , Epilepsia/fisiopatología , Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Ácido Glutámico/metabolismo , Hipocampo/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Astrocitos/citología , Modelos Animales de Enfermedad , Epilepsia/metabolismo , Retroalimentación Fisiológica , Glutamato Descarboxilasa/metabolismo , Hipocampo/fisiopatología , Humanos , Inhibición Neural , Ratas , Ratas Wistar , Transmisión Sináptica
11.
BMC Neurosci ; 12: 96, 2011 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-21967230

RESUMEN

BACKGROUND: Accumulating evidence suggests that glial signalling is activated by different brain functions. However, knowledge regarding molecular mechanisms of activation or their relation to neuronal activity is limited. The purpose of the present study is to identify the characteristics of ATP-evoked glial signalling in the brain reward area, the nucleus accumbens (NAc), and thereby to explore the action of citric acid cycle intermediate succinate (SUC). RESULTS: We described the burst-like propagation of Ca2+ transients evoked by ATP in acute NAc slices from rat brain. Co-localization of the ATP-evoked Ca2+ signalling with immunoreactivities of the astroglia-specific gap junction forming channel protein connexin43 (Cx43) and the glial fibrillary acidic protein (GFAP) indicated that the responsive cells were a subpopulation of Cx43 and GFAP immunoreactive astrocytes. The ATP-evoked Ca2+ transients were present under the blockade of neuronal activity, but were inhibited by Ca2+ store depletion and antagonism of the G protein coupled purinergic P2Y1 receptor subtype-specific antagonist MRS2179. Similarly, Ca2+ transients evoked by the P2Y1 receptor subtype-specific agonist 2-(Methylthio)adenosine 5'-diphosphate were also blocked by MRS2179. These characteristics implied that intercellular Ca2+ signalling originated from the release of Ca2+ from internal stores, triggered by the activation of P2Y1 receptors. Inhibition by the gap junction blockers carbenoxolone and flufenamic acid and by an antibody raised against the gating-associated segment of Cx43 suggested that intercellular Ca2+ signalling proceeded through gap junctions. We demonstrated for the first time that extracellular SUC also evoked Ca2+ transients (EC50 = 50-60 µM) in about 15% of the ATP-responsive NAc astrocytes. By contrast to glial cells, electrophysiologically identified NAc neurons surrounded by ATP-responsive astrocytes were not activated simultaneously. CONCLUSIONS: We concluded, therefore, that ATP- and SUC-sensitive Ca2+ transients appear to represent a signalling layer independent of NAc neurons. This previously unrecognised glial action of SUC, a major cellular energy metabolite, may play a role in linking metabolism to Ca2+ signalling in astrocytic networks under physiological and pathological conditions such as exercise and metabolic diseases.


Asunto(s)
Adenosina Trifosfato/fisiología , Astrocitos/metabolismo , Señalización del Calcio/fisiología , Neuronas/fisiología , Núcleo Accumbens/fisiología , Ácido Succínico/farmacología , Animales , Astrocitos/citología , Ciclo del Ácido Cítrico/fisiología , Masculino , Neuronas/citología , Núcleo Accumbens/citología , Núcleo Accumbens/metabolismo , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar
12.
Front Cell Neurosci ; 15: 617989, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33732110

RESUMEN

Fluctuations of cytosolic Ca2+ concentration in astrocytes are regarded as a critical non-neuronal signal to regulate neuronal functions. Although such fluctuations can be evoked by neuronal activity, rhythmic astrocytic Ca2+ oscillations may also spontaneously arise. Experimental studies hint that these spontaneous astrocytic Ca2+ oscillations may lie behind different kinds of emerging neuronal synchronized activities, like epileptogenic bursts or slow-wave rhythms. Despite the potential importance of spontaneous Ca2+ oscillations in astrocytes, the mechanism by which they develop is poorly understood. Using simple 3D synapse models and kinetic data of astrocytic Glu transporters (EAATs) and the Na+/Ca2+ exchanger (NCX), we have previously shown that NCX activity alone can generate markedly stable, spontaneous Ca2+ oscillation in the astrocytic leaflet microdomain. Here, we extend that model by incorporating experimentally determined real 3D geometries of 208 excitatory synapses reconstructed from publicly available ultra-resolution electron microscopy datasets. Our simulations predict that the surface/volume ratio (SVR) of peri-synaptic astrocytic processes prominently dictates whether NCX-mediated spontaneous Ca2+ oscillations emerge. We also show that increased levels of intracellular astrocytic Na+ concentration facilitate the appearance of Ca2+ fluctuations. These results further support the principal role of the dynamical reshaping of astrocyte processes in the generation of intrinsic Ca2+ oscillations and their spreading over larger astrocytic compartments.

13.
Biomolecules ; 12(1)2021 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-35053197

RESUMEN

Connexin (Cx) proteins establish intercellular gap junction channels (Cx GJCs) through coupling of two apposed hexameric Cx hemichannels (Cx HCs, connexons). Pre- and post-GJ interfaces consist of extracellular EL1 and EL2 loops, each with three conserved cysteines. Previously, we reported that known peptide inhibitors, mimicking a variety of Cx43 sequences, appear non-selective when binding to homomeric Cx43 vs. Cx36 GJC homology model subtypes. In pursuit of finding potentially Cx subtype-specific inhibitors of connexon-connexon coupling, we aimed at to understand better how the GJ interface is formed. Here we report on the discovery of Cx GJC subtype-specific protein stabilization centers (SCs) featuring GJ interface architecture. First, the Cx43 GJC homology model, embedded in two opposed membrane bilayers, has been devised. Next, we endorsed the fluctuation dynamics of SCs of the interface domain of Cx43 GJC by applying standard molecular dynamics under open and closed cystine disulfide bond (CS-SC) preconditions. The simulations confirmed the major role of the unique trans-GJ SC pattern comprising conserved (55N, 56T) and non-conserved (57Q) residues of the apposed EL1 loops in the stabilization of the GJC complex. Importantly, clusters of SC patterns residing close to the GJ interface domain appear to orient the interface formation via the numerous SCs between EL1 and EL2. These include central 54CS-S198C or 61CS-S192C contacts with residues 53R, 54C, 55N, 197D, 199F or 64V, 191P, respectively. In addition, we revealed that GJC interface formation is favoured when the psi dihedral angle of the nearby 193P residue is stable around 180° and the interface SCs disappear when this angle moves to the 0° to -45° range. The potential of the association of non-conserved residues with SC motifs in connexon-connexon coupling makes the development of Cx subtype-specific inhibitors viable.


Asunto(s)
Conexinas , Uniones Comunicantes , Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Canales Iónicos/metabolismo , Simulación de Dinámica Molecular
14.
Biomolecules ; 11(4)2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33921742

RESUMEN

Astrocytes serve essential roles in human brain function and diseases. Growing evidence indicates that astrocytes are central players of the feedback modulation of excitatory Glu signalling during epileptiform activity via Glu-GABA exchange. The underlying mechanism results in the increase of tonic inhibition by reverse operation of the astroglial GABA transporter, induced by Glu-Na+ symport. GABA, released from astrocytes, is synthesized from the polyamine (PA) putrescine and this process involves copper amino oxidase. Through this pathway, putrescine can be considered as an important source of inhibitory signaling that counterbalances epileptic discharges. Putrescine, however, is also a precursor for spermine that is known to enhance gap junction channel communication and, consequently, supports long-range Ca2+ signaling and contributes to spreading of excitatory activity through the astrocytic syncytium. Recently, we presented the possibility of neuron-glia redox coupling through copper (Cu+/Cu2+) signaling and oxidative putrescine catabolism. In the current work, we explore whether the Cu+/Cu2+ homeostasis is involved in astrocytic control on neuronal excitability by regulating PA catabolism. We provide supporting experimental data underlying this hypothesis. We show that the blockade of copper transporter (CTR1) by AgNO3 (3.6 µM) prevents GABA transporter-mediated tonic inhibitory currents, indicating causal relationship between copper (Cu+/Cu2+) uptake and the catabolism of putrescine to GABA in astrocytes. In addition, we show that MnCl2 (20 µM), an inhibitor of the divalent metal transporter DMT1, also prevents the astrocytic Glu-GABA exchange. Furthermore, we observed that facilitation of copper uptake by added CuCl2 (2 µM) boosts tonic inhibitory currents. These findings corroborate the hypothesis that modulation of neuron-glia coupling by copper uptake drives putrescine → GABA transformation, which leads to subsequent Glu-GABA exchange and tonic inhibition. Findings may in turn highlight the potential role of copper signaling in fine-tuning the activity of the tripartite synapse.


Asunto(s)
Astrocitos/metabolismo , Cobre/metabolismo , Neuronas/fisiología , Putrescina/metabolismo , Animales , Células Cultivadas , Proteínas Transportadoras de Cobre/antagonistas & inhibidores , Proteínas Transportadoras de Cobre/metabolismo , Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Potenciales Postsinápticos Inhibidores , Ratones , Neuronas/metabolismo , Ácido gamma-Aminobutírico/metabolismo
15.
Front Cell Neurosci ; 15: 787319, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069115

RESUMEN

Accumulating evidence indicate that astrocytes are essential players of the excitatory and inhibitory signaling during normal and epileptiform activity via uptake and release of gliotransmitters, ions, and other substances. Polyamines can be regarded as gliotransmitters since they are almost exclusively stored in astrocytes and can be released by various mechanisms. The polyamine putrescine (PUT) is utilized to synthesize GABA, which can also be released from astrocytes and provide tonic inhibition on neurons. The polyamine spermine (SPM), synthesized form PUT through spermidine (SPD), is known to unblock astrocytic Cx43 gap junction channels and therefore facilitate astrocytic synchronization. In addition, SPM released from astrocytes may also modulate neuronal NMDA, AMPA, and kainate receptors. As a consequence, astrocytic polyamines possess the capability to significantly modulate epileptiform activity. In this study, we investigated different steps in polyamine metabolism and coupled GABA release to assess their potential to control seizure generation and maintenance in two different epilepsy models: the low-[Mg2+] model of temporal lobe epilepsy in vitro and in the WAG/Rij rat model of absence epilepsy in vivo. We show that SPM is a gliotransmitter that is released from astrocytes and significantly contributes to network excitation. Importantly, we found that inhibition of SPD synthesis completely prevented seizure generation in WAG/Rij rats. We hypothesize that this antiepileptic effect is attributed to the subsequent enhancement of PUT to GABA conversion in astrocytes, leading to GABA release through GAT-2/3 transporters. This interpretation is supported by the observation that antiepileptic potential of the Food and Drug Administration (FDA)-approved drug levetiracetam can be diminished by specifically blocking astrocytic GAT-2/3 with SNAP-5114, suggesting that levetiracetam exerts its effect by increasing surface expression of GAT-2/3. Our findings conclusively suggest that the major pathway through which astrocytic polyamines contribute to epileptiform activity is the production of GABA. Modulation of astrocytic polyamine levels, therefore, may serve for a more effective antiepileptic drug development in the future.

16.
J Neurosci ; 29(26): 8565-77, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19571147

RESUMEN

Nitric oxide (NO) modulates synaptic transmission, and its level is elevated during epileptic activity in animal models of epilepsy. However, the role of NO for development and maintenance of epileptic activity is controversial. We studied this aspect in rat organotypic hippocampal slice cultures and acute hippocampal-entorhinal cortex slices from wild-type and neuronal NO synthase (nNOS) knock-out mice combining electrophysiological and fluorescence imaging techniques. Slice cultures contained nNOS-positive neurons and an elaborated network of nNOS-positive fibers. Lowering of extracellular Mg(2+) concentration led to development of epileptiform activity and increased NO formation as revealed by NO-selective probes, 4-amino-5-methylamino-2',7'-difluorofluorescein and 1,2-diaminoanthraquinone sulfate. NO deprivation by NOS inhibitors and NO scavengers caused depression of both EPSCs and IPSCs and prevented initiation of seizure-like events (SLEs) in 75% of slice cultures and 100% of hippocampal-entorhinal cortex slices. This effect was independent of the guanylyl cyclase/cGMP pathway. Suppression of SLE initiation in acute slices from mice was achieved by both the broad-spectrum NOS inhibitor N-methyl-L-arginine acetate and the nNOS-selective inhibitor 7-nitroindazole, whereas inhibition of inducible NOS by aminoguanidine was ineffective, suggesting that nNOS activity was crucial for SLE initiation. Additional evidence was obtained from knock-out animals because SLEs developed in a significantly lower percentage of slices from nNOS(-/-) mice and showed different characteristics, such as prolongation of onset latency and higher variability of SLE intervals. We conclude that enhancement of synaptic transmission by NO under epileptic conditions represents a positive feedback mechanism for the initiation of seizure-like events.


Asunto(s)
Corteza Entorrinal/fisiopatología , Hipocampo/fisiopatología , Óxido Nítrico/metabolismo , Convulsiones/patología , Análisis de Varianza , Animales , Animales Recién Nacidos , Antraquinonas/metabolismo , Estimulación Eléctrica/métodos , Inhibidores Enzimáticos/farmacología , Potenciales Evocados/efectos de los fármacos , Potenciales Evocados/fisiología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Magnesio/metabolismo , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico Sintasa de Tipo I/metabolismo , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Potasio/farmacología , Convulsiones/genética , Convulsiones/metabolismo , Transducción de Señal/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/genética
17.
Cell Calcium ; 86: 102137, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31838438

RESUMEN

The synergy between synaptic Glu release and astrocytic Glu-Na+ symport is essential to the signalling function of the tripartite synapse. Here we used kinetic data of astrocytic Glu transporters (EAAT) and the Na+/Ca2+ exchanger (NCX) to simulate Glu release, Glu uptake and subsequent Na+ and Ca2+ dynamics in the astrocytic leaflet microdomain following single release event. Model simulations show that Glu-Na+ symport differently affect intracellular [Na+] in synapses with different extent of astrocytic coverage. Surprisingly, NCX activity alone has been shown to generate markedly stable, spontaneous Ca2+ oscillation in the astrocytic leaflet. These on-going oscillations appear when NCX operates either in the forward or reverse direction. We conjecture that intrinsic NCX activity may play a prominent role in the generation of astrocytic Ca2+ oscillations.


Asunto(s)
Astrocitos/metabolismo , Señalización del Calcio , Calcio/metabolismo , Microdominios de Membrana/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Animales , Simulación por Computador , Humanos
18.
J Neurophysiol ; 102(4): 2538-53, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19675286

RESUMEN

Here we address how dynamics of glutamatergic and GABAergic synaptic input to CA3 pyramidal cells contribute to spontaneous emergence and evolution of recurrent seizure-like events (SLEs) in juvenile (P10-13) rat hippocampal slices bathed in low-[Mg(2+)] artificial cerebrospinal fluid. In field potential recordings from the CA3 pyramidal layer, a short epoch of high-frequency oscillation (HFO; 400-800 Hz) was observed during the first 10 ms of SLE onset. GABAergic synaptic input currents to CA3 pyramidal cells were synchronized and coincided with HFO, whereas the glutamatergic input lagged by approximately 10 ms. If the intracellular [Cl(-)] remained unperturbed (cell-attached recordings) or was set high with whole cell electrode solution, CA3 pyramidal cell firing peaked with HFO and GABAergic input. By contrast, with low intracellular [Cl(-)], spikes of CA3 pyramidal cells lagged behind HFO and GABAergic input. This temporal arrangement of HFO, synaptic input sequence, synchrony of GABAergic currents, and pyramidal cell firing emerged gradually with preictal discharges until the SLE onset. Blockade of GABA(A) receptor-mediated currents by picrotoxin reduced the inter-SLE interval and the number of preictal discharges and did not block recurrent SLEs. Our data suggest that dynamic changes of the functional properties of GABAergic input contribute to ictogenesis and GABAergic and glutamatergic inputs are both excitatory at the instant of SLE onset. At the SLE onset GABAergic input contributes to synchronization and recruitment of pyramidal cells. We conjecture that this network state is reached by an activity-dependent shift in GABA reversal potential during the preictal phase.


Asunto(s)
Región CA3 Hipocampal/fisiopatología , Periodicidad , Células Piramidales/fisiopatología , Convulsiones/fisiopatología , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/metabolismo , Envejecimiento , Animales , Región CA3 Hipocampal/efectos de los fármacos , Cloruros/metabolismo , Antagonistas del GABA/farmacología , Ácido Glutámico/metabolismo , Técnicas In Vitro , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/fisiología , Deficiencia de Magnesio/tratamiento farmacológico , Deficiencia de Magnesio/fisiopatología , Masculino , Picrotoxina/farmacología , Células Piramidales/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de GABA-A/metabolismo , Convulsiones/tratamiento farmacológico , Sinapsis/efectos de los fármacos , Sinapsis/fisiología , Transmisión Sináptica/efectos de los fármacos , Factores de Tiempo
19.
Biochem Biophys Res Commun ; 385(2): 210-4, 2009 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-19450549

RESUMEN

Crystal structures of transmembrane transport proteins belonging to the important families of neurotransmitter-sodium symporters reveal how they transport neurotransmitters across membranes. Substrate-induced structural conformations of gated neurotransmitter-sodium symporters have been in the focus of research, however, a key question concerning the mechanism of Na(+) ion coupling remained unanswered. Homology models of human glial transporter subtypes of the major inhibitory neurotransmitter gamma-aminobutyric acid were built. In accordance with selectivity data for subtype 2 vs. 3, docking and molecular dynamics calculations suggest similar orthosteric substrate (inhibitor) conformations and binding crevices but distinguishable allosteric Zn(2+) ion binding motifs. Considering the occluded conformational states of glial human gamma-aminobutyric acid transporter subtypes, we found major semi-extended and minor ring-like conformations of zwitterionic gamma-aminobutyric acid in complex with Na(+) ion. The existence of the minor ring-like conformation of gamma-aminobutyric acid in complex with Na(+) ion may be attributed to the strengthening of the intramolecular H-bond by the electrostatic effect of Na(+) ion. Coupling substrate uptake into cells with the thermodynamically favorable Na(+) ion movement through substrate-Na(+) ion complex formation may be a mechanistic principle featuring transmembrane neurotransmitter-sodium symporter proteins.


Asunto(s)
Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Sodio/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Regulación Alostérica , Secuencias de Aminoácidos , Cristalografía , Proteínas Transportadoras de GABA en la Membrana Plasmática/química , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Sodio/química , Zinc/química , Zinc/metabolismo , Ácido gamma-Aminobutírico/química
20.
Neuropharmacology ; 161: 107629, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31103619

RESUMEN

Glutamatergic transmission composed of the arriving of action potential at the axon terminal, fast vesicular Glu release, postsynaptic Glu receptor activation, astrocytic Glu clearance and Glu→Gln shuttle is an abundantly investigated phenomenon. Despite its essential role, however, much less is known about the consequences of the mechanistic connotations of Glu:Na+ symport. Due to the coupled Na+ transport, Glu uptake results in significantly elevated intracellular astrocytic [Na+] that markedly alters the driving force of other Na+-coupled astrocytic transporters. The resulting GABA and Gln release by reverse transport through the respective GAT-3 and SNAT3 transporters help to re-establish the physiological Na+ homeostasis without ATP dissipation and consequently leads to enhanced tonic inhibition and replenishment of axonal glutamate pool. Here, we place this emerging astrocytic adjustment of synaptic excitability into the centre of future perspectives. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.


Asunto(s)
Adaptación Fisiológica/fisiología , Astrocitos/fisiología , Retroalimentación Fisiológica/fisiología , Proteínas de Transporte de Glutamato en la Membrana Plasmática/fisiología , Glutamina/metabolismo , Sinapsis/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Astrocitos/metabolismo , Humanos , Transmisión Sináptica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA