Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 175: 105929, 2022 12.
Article in English | MEDLINE | ID: mdl-36410634

ABSTRACT

Sharp wave-ripples (SWRs) are hippocampal oscillations associated with memory consolidation. The subiculum, as the hippocampal output structure, ensures that hippocampal memory representations are transferred correctly to the consolidating neocortical regions. Because patients with temporal lobe epilepsy often develop memory deficits, we hypothesized that epileptic networks may disrupt subicular SWRs. We therefore investigated the impact of experimentally induced status epilepticus (SE) on subicular SWRs and contributing pyramidal neurons using electrophysiological recordings in mouse hippocampal slices. Subicular SWRs expressed hyperexcitable features post-SE, including increased ripple and unit activity. While regular firing neurons normally remain silent during SWRs, selective disinhibition recruited more regular firing neurons for action potential generation during SWRs post-SE. By contrast, burster neurons generated fewer action potential bursts during SWRs post-SE. Furthermore, altered timing of postsynaptic and action potentials suggested distorted neuronal recruitment during SWRs. Distorted subicular SWRs may therefore impair information processing and memory consolidation in epilepsy.


Subject(s)
Hippocampus , Status Epilepticus , Mice , Animals , Hippocampus/physiology , Neurons/physiology , Action Potentials/physiology , Pyramidal Cells/physiology
2.
J Neuroimmunol ; 368: 577881, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35537331

ABSTRACT

Immunological priming by type II interferon (IFN-γ) is crucial for evoking neurotoxic phenotypes of microglia (tissue-resident macrophages). We report that serial exposure of hippocampal slice cultures to IFN-γ and lipopolysaccharide (Toll-like receptor 4 ligand) induces high release of IL-6, TNF-α and nitric oxide, concomitant loss of electrical network activity (neuronal gamma oscillations) and neurodegeneration. Notably, these effects are still present after 3 days of IFN-γ removal but neither mimicked by IFN-α nor attenuated by anti-inflammatory cytokine, IL-10. Our findings might be relevant for brain diseases featuring elevated IFN-γ levels, such as viral and bacterial infections, multiple sclerosis and Alzheimer's disease.


Subject(s)
Interferon-gamma , Microglia , Hippocampus/metabolism , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Interleukin-10 , Lipopolysaccharides/pharmacology , Microglia/metabolism , Neurons/metabolism , Nitric Oxide , Tumor Necrosis Factor-alpha/metabolism
3.
Brain Behav Immun ; 96: 80-91, 2021 08.
Article in English | MEDLINE | ID: mdl-34015428

ABSTRACT

Recognition of pathogen- or damage-associated molecular patterns (PAMPs, DAMPs) by innate Toll-like receptors (TLRs) is central to the activation of microglia (brain macrophages) in many CNS diseases. Notably, TLR-mediated microglial activation is complex and modulated by additional exogenous and endogenous immunological signals. The impact of different microglial reactive phenotypes on electrical activity and neurotransmission is widely unknown, however. We explored the effects of TLR ligands on microglia and neuronal network function in rat organotypic hippocampal slice cultures (in situ), i.e., postnatal cortical tissue lacking adaptive immunity. Single exposure of slice cultures to TLR2 or TLR3 ligands [PGN, poly(I:C)] for 2-3 days induced moderate microglial activation featuring IL-6 and TNF-α release and only mild alterations of fast neuronal gamma band oscillations (30-70 Hz) that are fundamental to higher cognitive functions, such as perception, memory and behavior. Paired exposure to TLR3/TLR2 or TLR3/TLR4 ligands (LPS) induced nitric oxide (NO) release, enhanced TNF-α release, and associated with advanced network dysfunction, including slowing to the beta frequency band (12-30 Hz) and neural bursts (hyperexcitability). Paired exposure to a TLR ligand and the leukocyte cytokine IFN-γ enhanced NO release and associated with severe network dysfunction, albeit sensitive parvalbumin- and somatostatin-positive inhibitory interneurons were preserved. Notably, the neuronal disturbance was prevented by either microglial depletion or pharmacological inhibition of oxidant-producing enzymes, inducible NO synthase (iNOS) and NADPH oxidase. In conclusion, TLR-activated microglia can induce different levels of neuronal network dysfunction, in which severe dysfunction is mainly caused by reactive oxygen and nitrogen species rather than proinflammatory cytokines. Our findings provide a mechanistic insight into microglial activation and functional neuronal network impairment, with relevance to neuroinflammation and neurodegeneration observed in, e.g., meningoencephalitis, multiple sclerosis and Alzheimer's disease.


Subject(s)
Microglia , Toll-Like Receptor 2 , Animals , Cells, Cultured , Macrophages , Neurons , Rats , Toll-Like Receptor 3
4.
J Neuroinflammation ; 17(1): 235, 2020 Aug 11.
Article in English | MEDLINE | ID: mdl-32782006

ABSTRACT

BACKGROUND: The granulocyte-macrophage colony-stimulating factor (GM-CSF) (or CSF-2) is involved in myeloid cell growth and differentiation, and, possibly, a major mediator of inflammation in body tissues. The role of GM-CSF in the activation of microglia (CNS resident macrophages) and the consequent impacts on neuronal survival, excitability, and synaptic transmission are widely unknown, however. Here, we focused on electrical neuronal network rhythms in the gamma frequency band (30-70 Hz). Gamma oscillations are fundamental to higher brain functions, such as perception, attention, and memory, and they are exquisitely sensitive to metabolic and oxidative stress. METHODS: We explored the effects of chronic GM-CSF exposure (72 h) on microglia in male rat organotypic hippocampal slice cultures (in situ), i.e., postnatal cortex tissue lacking leukocyte invasion (adaptive immunity). We applied extracellular electrophysiological recordings of local field potential, immunohistochemistry, design-based stereology, biochemical analysis, and pharmacological ablation of microglia. RESULTS: GM-CSF triggered substantial proliferation of microglia (microgliosis). By contrast, the release of proinflammatory cytokines (IL-6, TNF-α) and nitric oxide, the hippocampal cytoarchitecture as well as the morphology of parvalbumin-positive inhibitory interneurons were unaffected. Notably, GM-CSF induced concentration-dependent, long-lasting disturbances of gamma oscillations, such as slowing (beta frequency band) and neural burst firing (hyperexcitability), which were not mimicked by the T lymphocyte cytokine IL-17. These disturbances were attenuated by depletion of the microglial cell population with liposome-encapsulated clodronate. In contrast to priming with the cytokine IFN-γ (type II interferon), GM-CSF did not cause inflammatory neurodegeneration when paired with the TLR4 ligand LPS. CONCLUSIONS: GM-CSF has a unique role in the activation of microglia, including the potential to induce neuronal network dysfunction. These immunomodulatory properties might contribute to cognitive impairment and/or epileptic seizure development in disease featuring elevated GM-CSF levels, blood-brain barrier leakage, and/or T cell infiltration.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hippocampus/drug effects , Interneurons/drug effects , Microglia/drug effects , Animals , Cell Proliferation/drug effects , Hippocampus/metabolism , Interleukin-6/metabolism , Interneurons/metabolism , Male , Microglia/metabolism , Nitric Oxide/metabolism , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
5.
J Neurosci Res ; 98(10): 1953-1967, 2020 10.
Article in English | MEDLINE | ID: mdl-32638411

ABSTRACT

Microglial cells (resident macrophages) feature rapid activation in CNS disease and can acquire multiple phenotypes exerting neuroprotection or neurotoxicity. The functional impact of surveying ("resting") microglia on neural excitability and neurotransmission in physiology is widely unknown, however. We addressed this issue in male rat hippocampal slice cultures (in situ) by pharmacological microglial ablation within days and by characterizing neuronal gamma-band oscillations (30-70 Hz) that are highly sensitive to neuromodulators and disturbances in ion and energy regulation. Gamma oscillations support action potential timing and synaptic plasticity, associate with higher brain functions like perception and memory, and require precise communication between excitatory pyramidal cells and inhibitory (GABAergic) interneurons. The slice cultures featured well-preserved hippocampal cytoarchitecture and parvalbumin-positive interneuron networks, microglia with ramified morphology, and low basal levels of IL-6, TNF-α, and nitric oxide (NO). Stimulation of slice cultures with the pro-inflammatory cytokine IFN-γ or bacterial LPS serving as positive controls for microglial reactivity induced MHC-II expression and increased cytokine and NO release. Chronic exposure of slice cultures to liposome-encapsulated clodronate reduced the microglial cell population by about 96%, whereas neuronal structures, astrocyte GFAP expression, and basal levels of cytokines and NO were unchanged. Notably, the properties of gamma oscillations reflecting frequency, number and synchronization of synapse activity were regular after microglial depletion. Also, electrical stimulus-induced transients of the extracellular potassium concentration ([K+ ]o ) reflecting cellular K+ efflux, clearance and buffering were unchanged. This suggests that nonreactive microglia are dispensable for neuronal homeostasis and neuromodulation underlying network signaling and rhythm generation in cortical tissue.


Subject(s)
Gamma Rhythm/physiology , Hippocampus/physiology , Microglia/physiology , Neurons/physiology , Potassium/physiology , Animals , Animals, Newborn , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Hippocampus/cytology , Male , Organ Culture Techniques , Rats , Rats, Wistar
6.
iScience ; 23(7): 101316, 2020 Jul 24.
Article in English | MEDLINE | ID: mdl-32653807

ABSTRACT

Lactate shuttled from blood, astrocytes, and/or oligodendrocytes may serve as the major glucose alternative in brain energy metabolism. However, its effectiveness in fueling neuronal information processing underlying complex cortex functions like perception and memory is unclear. We show that sole lactate disturbs electrical gamma and theta-gamma oscillations in hippocampal networks by either attenuation or neural bursts. Bursting is suppressed by elevating the glucose fraction in substrate supply. By contrast, lactate does not affect electrical sharp wave-ripple activity featuring lower energy use. Lactate increases the oxygen consumption during the network states, reflecting enhanced oxidative ATP synthesis in mitochondria. Finally, lactate attenuates synaptic transmission in excitatory pyramidal cells and fast-spiking, inhibitory interneurons by reduced neurotransmitter release from presynaptic terminals, whereas action potential generation in the axon is regular. In conclusion, sole lactate is less effective and potentially harmful during gamma-band rhythms by omitting obligatory ATP delivery through fast glycolysis at the synapse.

7.
Hippocampus ; 30(10): 1044-1057, 2020 10.
Article in English | MEDLINE | ID: mdl-32412680

ABSTRACT

The rodent hippocampus expresses a variety of neuronal network oscillations depending on the behavioral state of the animal. Locomotion and active exploration are accompanied by theta-nested gamma oscillations while resting states and slow-wave sleep are dominated by intermittent sharp wave-ripple complexes. It is believed that gamma rhythms create a framework for efficient acquisition of information whereas sharp wave-ripples are thought to be involved in consolidation and retrieval of memory. While not strictly mutually exclusive, one of the two patterns usually dominates in a given behavioral state. Here we explore how different input patterns induce either of the two network states, using an optogenetic stimulation approach in hippocampal brain slices of mice. We report that the pattern of the evoked oscillation depends strongly on the initial synchrony of activation of excitatory cells within CA3. Short, synchronous activation favors the emergence of sharp wave-ripple complexes while persistent but less synchronous activity-as typical for sensory input during exploratory behavior-supports the generation of gamma oscillations. This dichotomy is reflected by different degrees of synchrony of excitatory and inhibitory synaptic currents within these two states. Importantly, the induction of these two fundamental network patterns does not depend on the presence of any neuromodulatory transmitter like acetylcholine, but is merely based on a different synchrony in the initial activation pattern.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Gamma Rhythm/physiology , Hippocampus/physiology , Inhibitory Postsynaptic Potentials/physiology , Nerve Net/physiology , Animals , Hippocampus/chemistry , Male , Mice , Mice, Inbred C57BL , Microelectrodes , Nerve Net/chemistry , Optogenetics/methods , Organ Culture Techniques
8.
J Cereb Blood Flow Metab ; 40(12): 2401-2415, 2020 12.
Article in English | MEDLINE | ID: mdl-31842665

ABSTRACT

Disturbances of cognitive functions occur rapidly during acute metabolic stress. However, the underlying mechanisms are not fully understood. Cortical gamma oscillations (30-100 Hz) emerging from precise synaptic transmission between excitatory principal neurons and inhibitory interneurons, such as fast-spiking GABAergic basket cells, are associated with higher brain functions, like sensory perception, selective attention and memory formation. We investigated the alterations of cholinergic gamma oscillations at the level of neuronal ensembles in the CA3 region of rat hippocampal slice cultures. We combined electrophysiology, calcium imaging (CamKII.GCaMP6f) and mild metabolic stress that was induced by rotenone, a lipophilic and highly selective inhibitor of complex I in the respiratory chain of mitochondria. The detected pyramidal cell ensembles showing repetitive patterns of activity were highly sensitive to mild metabolic stress. Whereas such synchronised multicellular activity diminished, the overall activity of individual pyramidal cells was unaffected. Additionally, mild metabolic stress had no effect on the rate of action potential generation in fast-spiking neural units. However, the partial disinhibition of slow-spiking neural units suggests that disturbances of ensemble formation likely result from alterations in synaptic inhibition. Our study bridges disturbances on the (multi-)cellular and network level to putative cognitive impairment on the system level.


Subject(s)
Cognitive Dysfunction/metabolism , Gamma Rhythm/physiology , Hippocampus/metabolism , Pyramidal Cells/drug effects , Stress, Physiological/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cognitive Dysfunction/physiopathology , Electrophysiology/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Gamma Rhythm/drug effects , Hippocampus/drug effects , Hippocampus/physiopathology , Interneurons/classification , Interneurons/drug effects , Interneurons/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Rats , Rats, Wistar , Rotenone/administration & dosage , Rotenone/pharmacology , Stress, Physiological/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Uncoupling Agents/administration & dosage , Uncoupling Agents/pharmacology
9.
Proc Natl Acad Sci U S A ; 116(10): 4637-4642, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30782788

ABSTRACT

Type II IFN (IFN-γ) is a proinflammatory T lymphocyte cytokine that serves in priming of microglia-resident CNS macrophages-during the complex microglial activation process under pathological conditions. Priming generally permits an exaggerated microglial response to a secondary inflammatory stimulus. The impact of primed microglia on physiological neuronal function in intact cortical tissue (in situ) is widely unknown, however. We explored the effects of chronic IFN-γ exposure on microglia in hippocampal slice cultures, i.e., postnatal parenchyma lacking leukocyte infiltration (adaptive immunity). We focused on fast neuronal network waves in the gamma-band (30-70 Hz). Such gamma oscillations are fundamental to higher brain functions, such as perception, attention, and memory, and are exquisitely sensitive to metabolic and oxidative stress. IFN-γ induced substantial morphological changes and cell population expansion in microglia as well as moderate up-regulation of activation markers, MHC-II, CD86, IL-6, and inducible nitric oxide synthase (iNOS), but not TNF-α. Cytoarchitecture and morphology of pyramidal neurons and parvalbumin-positive inhibitory interneurons were well-preserved. Notably, gamma oscillations showed a specific decline in frequency of up to 8 Hz, which was not mimicked by IFN-α or IL-17 exposure. The rhythm disturbance was caused by moderate microglial nitric oxide (NO) release demonstrated by pharmacological microglia depletion and iNOS inhibition. In conclusion, IFN-γ priming induces substantial proliferation and moderate activation of microglia that is capable of slowing neural information processing. This mechanism might contribute to cognitive impairment in chronic brain disease featuring elevated IFN-γ levels, blood-brain barrier leakage, and/or T cell infiltration, well before neurodegeneration occurs.


Subject(s)
Interferon-gamma/metabolism , Microglia/metabolism , Neurons/cytology , Animals , Cell Proliferation , Hippocampus/chemistry , Hippocampus/cytology , Hippocampus/metabolism , Microglia/chemistry , Microglia/cytology , Neuronal Plasticity , Neurons/chemistry , Neurons/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Wistar
10.
PLoS One ; 14(1): e0209228, 2019.
Article in English | MEDLINE | ID: mdl-30645585

ABSTRACT

Several lines of evidence imply changes in inhibitory interneuron connectivity and subsequent alterations in oscillatory network activities in the pathogenesis of Alzheimer's Disease (AD). Recently, we provided evidence for an increased immunoreactivity of both the postsynaptic scaffold protein gephyrin and the GABAA receptor γ2-subunit in the hippocampus of young (1 and 3 months of age), APPPS1 mice. These mice represent a well-established model of cerebral amyloidosis, which is a hallmark of human AD. In this study, we demonstrate a robust increase of parvalbumin immunoreactivity and accentuated projections of parvalbumin positive (PV+) interneurons, which target perisomatic regions of pyramidal cells within the hippocampal subregions CA1 and CA3 of 3-month-old APPPS1 mice. Colocalisation studies confirmed a significant increase in the density of PV+ projections labeled with antibodies against a presynaptic (vesicular GABA transporter) and a postsynaptic marker (gephyrin) of inhibitory synapses within the pyramidal cell layer of CA1 and CA3. As perisomatic inhibition by PV+-interneurons is crucial for the generation of hippocampal network oscillations involved in spatial processing, learning and memory formation we investigated the impact of the putative enhanced perisomatic inhibition on two types of fast neuronal network oscillations in acute hippocampal slices: 1. spontaneously occurring sharp wave-ripple complexes (SPW-R), and 2. cholinergic γ-oscillations. Interestingly, both network patterns were generally preserved in APPPS1 mice similar to WT mice. However, the comparison of simultaneous CA3 and CA1 recordings revealed that the incidence and amplitude of SPW-Rs were significantly lower in CA1 vs CA3 in APPPS1 slices, whereas the power of γ-oscillations was significantly higher in CA3 vs CA1 in WT-slices indicating an impaired communication between the CA3 and CA1 network activities in APPPS1 mice. Taken together, our data demonstrate an increased GABAergic synaptic output of PV+ interneurons impinging on pyramidal cells of CA1 and CA3, which might limit the coordinated cross-talk between these two hippocampal areas in young APPPS1 mice and mediate long-term changes in synaptic inhibition during progression of amyloidosis.


Subject(s)
Alzheimer Disease/metabolism , Amyloidosis/metabolism , Hippocampus/metabolism , Action Potentials , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloidosis/genetics , Amyloidosis/pathology , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/pathology , Disease Models, Animal , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Gamma Rhythm , Hippocampus/pathology , Humans , In Vitro Techniques , Interneurons/metabolism , Interneurons/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/metabolism , Nerve Net/pathology , Parvalbumins/metabolism , Presenilin-1/genetics , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Synapses/metabolism
11.
Pflugers Arch ; 470(9): 1377-1389, 2018 09.
Article in English | MEDLINE | ID: mdl-29808353

ABSTRACT

Gamma oscillations (30-100 Hz) represent a physiological fast brain rhythm that occurs in many cortex areas in awake mammals, including humans. They associate with sensory perception, voluntary movement, and memory formation and require precise synaptic transmission between excitatory glutamatergic neurons and inhibitory GABAergic interneurons such as parvalbumin-positive basket cells. Notably, gamma oscillations are exquisitely sensitive to shortage in glucose and oxygen supply (metabolic stress), with devastating consequences for higher cognitive functions. Herein, we explored the robustness of gamma oscillations against changes in the availability of alternative energy substrates and amino acids, which is partially regulated by glial cells such as astrocytes. We used organotypic slice cultures of the rat hippocampus expressing acetylcholine-induced persistent gamma oscillations under normoxic recording conditions (20% oxygen fraction). Our main findings are (1) partial substitution of glucose with pyruvate and the ketone body ß-hydroxybutyrate increases the frequency of gamma oscillations, even at different stages of neuronal tissue development. (2) Supplementation with the astrocytic neurotransmitter precursor glutamine has no effect on the properties of gamma oscillations. (3) Supplementation with glycine increases power, frequency, and inner coherence of gamma oscillations in a dose-dependent manner. (4) During these treatments switches to other frequency bands or pathological network states such as neural burst firing or synchronized epileptic activity are absent. Our study indicates that cholinergic gamma oscillations show general robustness against these changes in nutrient and amino acid composition of the cerebrospinal fluid; however, modulation of their properties may impact on cortical information processing under physiological and pathophysiological conditions.


Subject(s)
Neurons/metabolism , Amino Acids/metabolism , Animals , Astrocytes/metabolism , Hippocampus/metabolism , Interneurons/metabolism , Rats , Rats, Wistar , Synaptic Transmission/physiology
12.
Front Mol Neurosci ; 9: 47, 2016.
Article in English | MEDLINE | ID: mdl-27378847

ABSTRACT

Fast neuronal network oscillations in the gamma frequency band (30-100 Hz) occur in various cortex regions, require timed synaptic excitation and inhibition with glutamate and GABA, respectively, and are associated with higher brain functions such as sensory perception, attentional selection and memory formation. However, little is known about energy and ion homeostasis during the gamma oscillation. Recent studies addressed this topic in slices of the rodent hippocampus using cholinergic and glutamatergic receptor models of gamma oscillations (GAM). Methods with high spatial and temporal resolution were applied in vitro, such as electrophysiological recordings of local field potential (LFP) and extracellular potassium concentration ([K(+)]o), live-cell fluorescence imaging of nicotinamide adenine dinucleotide (phosphate) and flavin adenine dinucleotide [NAD(P)H and FAD, respectively] (cellular redox state), and monitoring of the interstitial partial oxygen pressure (pO2) in depth profiles with microsensor electrodes, including mathematical modeling. The main findings are: (i) GAM are associated with high oxygen consumption rate and significant changes in the cellular redox state, indicating rapid adaptations in glycolysis and oxidative phosphorylation; (ii) GAM are accompanied by fluctuating elevations in [K(+)]o of less than 0.5 mmol/L from baseline, likely reflecting effective K(+)-uptake mechanisms of neuron and astrocyte compartments; and (iii) GAM are exquisitely sensitive to metabolic stress induced by lowering oxygen availability or by pharmacological inhibition of the mitochondrial respiratory chain. These findings reflect precise cellular adaptations to maintain adenosine-5'-triphosphate (ATP), ion and neurotransmitter homeostasis and thus neural excitability and synaptic signaling during GAM. Conversely, the exquisite sensitivity of GAM to metabolic stress might significantly contribute the exceptional vulnerability of higher brain functions in brain disease.

13.
Epilepsia ; 57(5): 746-56, 2016 05.
Article in English | MEDLINE | ID: mdl-27087530

ABSTRACT

OBJECTIVE: The need for alternative pharmacologic strategies in treatment of epilepsies is pressing for about 30% of patients with epilepsy who do not experience satisfactory seizure control with present treatments. In temporal lobe epilepsy (TLE) even up to 80% of patients are pharmacoresistant, and surgical resection of the ictogenic tissue is only possible for a minority of TLE patients. In this study we investigate purinergic modulation of drug-resistant seizure-like events (SLEs) in human temporal cortex slices. METHODS: Layer V/VI field potentials from a total of 77 neocortical slices from 17 pharmacoresistant patients were recorded to monitor SLEs induced by application of 8 mM [K(+) ] and 50 µm bicuculline. RESULTS: Activating A1 receptors with a specific agonist completely suppressed SLEs in 73% of human temporal cortex slices. In the remaining slices, incidence of SLEs was markedly reduced. Because a subportion of slices can be pharmacosensitive, we tested effects of an A1 agonist, in slices insensitive to a high dose of carbamazepine (50 µm). Also in these cases the A1 agonist was equally efficient. Moreover, ATP and adenosine blocked or modulated SLEs, an effect mediated not by P2 receptors but rather by adenosine A1 receptors. SIGNIFICANCE: Selective activation of A1 receptors mediates a strong anticonvulsant action in human neocortical slices from pharmacoresistant patients. We propose that our human slice model of seizure-like activity is a feasible option for future studies investigating new antiepileptic drug (AED) candidates.


Subject(s)
Drug Resistant Epilepsy/pathology , Neocortex/drug effects , Neocortex/metabolism , Receptors, Purinergic P1/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine Triphosphate/pharmacology , Adult , Bicuculline/analogs & derivatives , Bicuculline/pharmacology , Carbamazepine/adverse effects , Carbamazepine/pharmacology , Drug Resistant Epilepsy/drug therapy , Electric Stimulation , Evoked Potentials/drug effects , Female , Humans , In Vitro Techniques , Male , Middle Aged , Potassium/pharmacology , Purinergic Agents/pharmacology , Time Factors , Young Adult
14.
Neurobiol Dis ; 91: 155-65, 2016 07.
Article in English | MEDLINE | ID: mdl-26972679

ABSTRACT

Epileptogenesis following insults to the brain may be triggered by a dysfunctional blood-brain barrier (BBB) associated with albumin extravasation and activation of astrocytes. Using ex vivo recordings from the BBB-disrupted hippocampus after neocortical photothrombotic stroke, we previously demonstrated abnormal activity-dependent accumulation of extracellular potassium with facilitated generation of seizure like events and spreading depolarizations. Similar changes could be observed after intracerebroventricular (icv) application of albumin. We hypothesized that alterations in extracellular potassium and glutamate homeostasis might lead to alterations in synaptic interactions. We therefore assessed the effects of icv albumin on homo- and heterosynaptic plasticity in hippocampal CA1, 24h after a single injection or 7days after continuous infusion of icv albumin. We demonstrate alterations in both homo- and heterosynaptic plasticity compared to control conditions in ex vivo slice studies. Albumin-treated tissue reveals (1) reduced long-term depression following low-frequency stimulation; (2) increased long-term potentiation of population spikes in response to 20Hz stimulation; (3) potentiated responses to Schaffer collateral stimulation following high-frequency stimulation of the direct cortical input and low-frequency stimulation of alveus and finally, (4) TGFß receptor II (TGFßR-II) involvement in albumin-induced homosynaptic plasticity changes. We conclude that albumin-induced network hyperexcitability is associated with abnormal homo- and heterosynaptic plasticity that could partly be reversed by interference with TGFßR-II-mediated signaling and therefore it might be an important factor in the process of epileptogenesis.


Subject(s)
Albumins/pharmacology , Blood-Brain Barrier/drug effects , CA1 Region, Hippocampal/drug effects , Long-Term Potentiation/drug effects , Neuronal Plasticity/drug effects , Albumins/administration & dosage , Animals , Astrocytes/drug effects , Blood-Brain Barrier/physiopathology , CA1 Region, Hippocampal/cytology , Injections, Intraventricular , Long-Term Potentiation/physiology , Male , Neuronal Plasticity/physiology , Neurons/drug effects , Neurons/physiology , Rats, Wistar
15.
Neuropharmacology ; 101: 188-203, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26409781

ABSTRACT

Sharp wave ripples (SPW-Rs) are thought to play an important role in memory consolidation. By rapid replay of previously stored information during slow wave sleep and consummatory behavior, they result from the formation of neural ensembles during a learning period. Serotonin (5-HT), suggested to be able to modify SPW-Rs, can affect many neurons simultaneously by volume transmission and alter network functions in an orchestrated fashion. In acute slices from dorsal hippocampus, SPW-Rs can be induced by repeated high frequency stimulation that induces long-lasting LTP. We used this model to study SPW-R appearance and modulation by 5-HT. Although stimulation in presence of 5-HT permitted LTP induction, SPW-Rs were "masked"--but appeared after 5-HT wash-out. This SPW-R masking was dose dependent with 100 nM 5-HT being sufficient--if the 5-HT re-uptake inhibitor citalopram was present. Fenfluramine, a serotonin releaser, could also mask SPW-Rs. Masking was due to 5-HT1A and 5-HT2A/C receptor activation. Neither membrane potential nor membrane conductance changes in pyramidal cells caused SPW-R blockade since both remained unaffected by combining 5-HT and citalopram. Moreover, 10 and 30 µM 5-HT mediated SPW-R masking preceded neuronal hyperpolarization and involved reduced presynaptic transmitter release. 5-HT, as well as a 5-HT1A agonist, augmented paired pulse facilitation and affected the coefficient of variance. Spontaneous SPW-Rs in mice hippocampal slices were also masked by 5-HT and fenfluramine. While neuronal ensembles can acquire long lasting LTP during higher 5-HT levels, lower 5-HT levels enable neural ensembles to replay previously stored information and thereby permit memory consolidation memory.


Subject(s)
Hippocampus/drug effects , Long-Term Potentiation/drug effects , Nerve Net/drug effects , Serotonin Agents/pharmacology , Serotonin/pharmacology , Animals , Biophysics , Citalopram/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation , Hippocampus/physiology , In Vitro Techniques , Piperazines/pharmacology , Presynaptic Terminals/drug effects , Rats , Rats, Wistar
16.
Front Neurol ; 6: 30, 2015.
Article in English | MEDLINE | ID: mdl-25741317

ABSTRACT

Drug resistant patients undergoing epilepsy surgery have a good chance to become sensitive to anticonvulsant medication, suggesting that the resected brain tissue is responsible for drug resistance. Here, we address the question whether P-glycoprotein (Pgp) and multidrug resistance-associated proteins (MRPs) expressed in the resected tissue contribute to drug resistance in vitro. Effects of anti-epileptic drugs [carbamazepine (CBZ), sodium valproate, phenytoin] and two unspecific inhibitors of Pgp and MRPs [verapamil (VPM) and probenecid (PBN)] on seizure-like events (SLEs) induced in slices from 35 hippocampal and 35 temporal cortex specimens of altogether 51 patients (161 slices) were studied. Although in slice preparations the blood brain barrier is not functional, we found that SLEs predominantly persisted in the presence of anticonvulsant drugs (90%) and also in the presence of VPM and PBN (86%). Following subsequent co-administration of anti-epileptic drugs and drug transport inhibitors, SLEs continued in 63% of 143 slices. Drug sensitivity in slices was recognized either as transition to recurrent epileptiform transients (30%) or as suppression (7%), particularly by perfusion with CBZ in PBN containing solutions (43, 9%). Summarizing responses to co-administration from more than one slice per patient revealed that suppression of seizure-like activity in all slices was only observed in 7% of patients. Patients whose tissue was completely or partially sensitive (65%) presented with higher seizure frequencies than those with resistant tissue (35%). However, corresponding subgroups of patients do not differ with respect to expression rates of drug transporters. Our results imply that parenchymal MRPs and Pgp are not responsible for drug resistance in resected tissue.

17.
Neurosci Lett ; 574: 15-20, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24796812

ABSTRACT

Sharp wave-ripple complexes (SPW-R) are observed in vivo during resting immobility, consummatory behavior and during slow wave sleep, and they have been proposed to support memory consolidation. It has been suggested that GABAergic cells play important roles in controlling incidence of sharp waves and of ripple frequency. We report here that the GABAB agonist baclofen reversibly suppresses SPW-R activity in rat hippocampal slices, presumably affecting the strength of neuronal coupling in the associative network of area CA3. The effect is specific as the GABAB receptor antagonist CGP55846 prevents this effect; however, CGP55846 application had no major effect on incidence of SPW-R. Interestingly, repetitive stimulation in the presence of baclofen is able to induce SPW-R activity, which only appears after washout of baclofen. Our findings suggest that GABA levels through activation of GABAB receptors may be involved in the transition from theta-gamma to SPW-R working mode in the hippocampus.


Subject(s)
Hippocampus/physiology , Receptors, GABA-B/metabolism , Animals , Electric Stimulation , Excitatory Postsynaptic Potentials , GABA-B Receptor Agonists/pharmacology , GABA-B Receptor Antagonists/pharmacology , Hippocampus/drug effects , In Vitro Techniques , Male , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...