Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Aging Clin Exp Res ; 33(6): 1713-1716, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31732960

RESUMO

New onset epilepsy and seizures are common neurological disorders in aged people, second only to stroke and dementia. They are frequently related to other pathological conditions including stroke, trauma, tumors and neurological diseases whereas in about one-third of cases the origin is unknown. Besides the origin, the cellular and molecular events that suddenly trigger seizures are poorly defined. Using an acute model of seizure generation that better resembles new onset seizures, we studied GABAergic interneurons and astrocytes during seizure generation. We found that seizures are preceded by a GABAergic rhythmic hyperactivity that synchronizes pyramidal neurons by inducing a rebound spiking that favors seizures' onset. Furthermore, the intense activity in GABAergic interneurons evokes Ca2+ elevations in astrocytes that, by releasing glutamate, further excite neuronal network. Elucidating the cellular and molecular events that generate seizures may reveal new targets for treatment of new onset seizures and epilepsy.


Assuntos
Epilepsia , Convulsões , Idoso , Humanos , Interneurônios , Neurônios
2.
Glia ; 66(10): 2188-2199, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30144319

RESUMO

The gliotransmitter glutamate in different brain regions modulates neuronal excitability and synaptic transmission through a variety of mechanisms. Among the hallmarks of astrocytic glutamate release are the slow depolarizing inward currents (SICs) in neurons mediated by N-methyl-d-aspartate receptor activation. Different stimuli that evoke Ca2+ elevations in astrocytes induce neuronal SICs suggesting a Ca2+ -dependent exocytotic glutamate release mechanism of SIC generation. To gain new insights into this mechanism, we investigated the relationship between astrocytic Ca2+ elevations and neuronal SICs in mouse hippocampal slice preparations. Here we provide evidence that SICs, occurring either spontaneously or following a hypotonic challenge, are unchanged in the virtual absence of Ca2+ signal changes at astrocytic soma and processes, including spatially restricted Ca2+ microdomains. SICs are also unchanged in the presence of Bafilomycin A1 that after prolonged slice incubation depletes glutamate from astrocytic vesicles. We also found that hemichannels and TREK family channels-that recent studies proposed to mediate astrocytic glutamate release - are not involved in SIC generation. SICs are reduced by the volume-sensitive anion channel antagonists diisothiocyanatostilbene-2,2'-disulfonic acid (DIDS), quinine and fluoxetine, suggesting a possible contribution of these channels in SIC generation. Direct measurements of astrocytic glutamate release further confirm that hypotonicity-evoked gliotransmission is impaired following DIDS, quinine and fluoxetine while the exocytotic release of glutamate-that is proposed to mediate synaptic transmission modulation by astrocytes-remains unaffected. In conclusion, our data provide evidence that the release of glutamate generating SICs occurs independently on exocytotic Ca2+ -dependent glutamate release mechanism.


Assuntos
Astrócitos/metabolismo , Ácido Glutâmico/metabolismo , Potenciais da Membrana/fisiologia , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica/fisiologia , Animais , Astrócitos/efeitos dos fármacos , Cálcio/metabolismo , Cátions Bivalentes/metabolismo , Fármacos do Sistema Nervoso Central/farmacologia , Exocitose/fisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Canais de Potássio de Domínios Poros em Tandem/genética , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Técnicas de Cultura de Tecidos
3.
Glia ; 65(4): 569-580, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28130845

RESUMO

Astrocytes play crucial roles in brain homeostasis and are emerging as regulatory elements of neuronal and synaptic physiology by responding to neurotransmitters with Ca2+ elevations and releasing gliotransmitters that activate neuronal receptors. Aging involves neuronal and astrocytic alterations, being considered risk factor for neurodegenerative diseases. Most evidence of the astrocyte-neuron signaling is derived from studies with young animals; however, the features of astrocyte-neuron signaling in adult and aging brain remain largely unknown. We have investigated the existence and properties of astrocyte-neuron signaling in physiologically and pathologically aging mouse hippocampal and cortical slices at different lifetime points (0.5 to 20 month-old animals). We found that astrocytes preserved their ability to express spontaneous and neurotransmitter-dependent intracellular Ca2+ signals from juvenile to aging brains. Likewise, resting levels of gliotransmission, assessed by neuronal NMDAR activation by glutamate released from astrocytes, were largely preserved with similar properties in all tested age groups, but DHPG-induced gliotransmission was reduced in aged mice. In contrast, gliotransmission was enhanced in the APP/PS1 mouse model of Alzheimer's disease, indicating a dysregulation of astrocyte-neuron signaling in pathological conditions. Disruption of the astrocytic IP3 R2 mediated-signaling, which is required for neurotransmitter-induced astrocyte Ca2+ signals and gliotransmission, boosted the progression of amyloid plaque deposits and synaptic plasticity impairments in APP/PS1 mice at early stages of the disease. Therefore, astrocyte-neuron interaction is a fundamental signaling, largely conserved in the adult and aging brain of healthy animals, but it is altered in Alzheimer's disease, suggesting that dysfunctions of astrocyte Ca2+ physiology may contribute to this neurodegenerative disease. GLIA 2017 GLIA 2017;65:569-580.


Assuntos
Envelhecimento , Astrócitos/fisiologia , Encéfalo/citologia , Comunicação Celular/fisiologia , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Acetilcolina/farmacologia , Trifosfato de Adenosina/farmacologia , Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/genética , Animais , Astrócitos/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Cálcio/metabolismo , Comunicação Celular/efeitos dos fármacos , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Metoxi-Hidroxifenilglicol/análogos & derivados , Metoxi-Hidroxifenilglicol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Presenilina-1/deficiência , Presenilina-1/genética , Transdução de Sinais/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Potenciais Sinápticos/efeitos dos fármacos , Potenciais Sinápticos/genética
4.
Cereb Cortex ; 25(10): 3699-712, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25260706

RESUMO

Endocannabinoids (eCBs) play key roles in brain function, acting as modulatory signals in synaptic transmission and plasticity. They are recognized as retrograde messengers that mediate long-term synaptic depression (LTD), but their ability to induce long-term potentiation (LTP) is poorly known. We show that eCBs induce the long-term enhancement of transmitter release at single hippocampal synapses through stimulation of astrocytes when coincident with postsynaptic activity. This LTP requires the coordinated activity of the 3 elements of the tripartite synapse: 1) eCB-evoked astrocyte calcium signal that stimulates glutamate release; 2) postsynaptic nitric oxide production; and 3) activation of protein kinase C and presynaptic group I metabotropic glutamate receptors, whose location at presynaptic sites was confirmed by immunoelectron microscopy. Hence, while eCBs act as retrograde signals to depress homoneuronal synapses, they serve as lateral messengers to induce LTP in distant heteroneuronal synapses through stimulation of astrocytes. Therefore, eCBs can trigger LTP through stimulation of astrocyte-neuron signaling, revealing novel cellular mechanisms of eCB effects on synaptic plasticity.


Assuntos
Astrócitos/fisiologia , Endocanabinoides/metabolismo , Hipocampo/fisiologia , Potenciação de Longa Duração , Células Piramidais/fisiologia , Animais , Astrócitos/metabolismo , Cálcio/metabolismo , Potenciais Pós-Sinápticos Excitadores , Hipocampo/metabolismo , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Proteína Quinase C/metabolismo , Células Piramidais/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Transdução de Sinais , Sinapses/metabolismo
5.
PLoS Biol ; 10(2): e1001259, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22347811

RESUMO

Long-term potentiation (LTP) of synaptic transmission represents the cellular basis of learning and memory. Astrocytes have been shown to regulate synaptic transmission and plasticity. However, their involvement in specific physiological processes that induce LTP in vivo remains unknown. Here we show that in vivo cholinergic activity evoked by sensory stimulation or electrical stimulation of the septal nucleus increases Ca²âº in hippocampal astrocytes and induces LTP of CA3-CA1 synapses, which requires cholinergic muscarinic (mAChR) and metabotropic glutamate receptor (mGluR) activation. Stimulation of cholinergic pathways in hippocampal slices evokes astrocyte Ca²âº elevations, postsynaptic depolarizations of CA1 pyramidal neurons, and LTP of transmitter release at single CA3-CA1 synapses. Like in vivo, these effects are mediated by mAChRs, and this cholinergic-induced LTP (c-LTP) also involves mGluR activation. Astrocyte Ca²âº elevations and LTP are absent in IP3R2 knock-out mice. Downregulating astrocyte Ca²âº signal by loading astrocytes with BAPTA or GDPßS also prevents LTP, which is restored by simultaneous astrocyte Ca²âº uncaging and postsynaptic depolarization. Therefore, cholinergic-induced LTP requires astrocyte Ca²âº elevations, which stimulate astrocyte glutamate release that activates mGluRs. The cholinergic-induced LTP results from the temporal coincidence of the postsynaptic activity and the astrocyte Ca²âº signal simultaneously evoked by cholinergic activity. Therefore, the astrocyte Ca²âº signal is necessary for cholinergic-induced synaptic plasticity, indicating that astrocytes are directly involved in brain storage information.


Assuntos
Astrócitos/fisiologia , Neurônios Colinérgicos/fisiologia , Potenciação de Longa Duração , Sinapses/fisiologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Atropina/farmacologia , Sinalização do Cálcio , Neurônios Colinérgicos/efeitos dos fármacos , Neurônios Colinérgicos/metabolismo , Ácido Glutâmico/metabolismo , Glicina/análogos & derivados , Glicina/farmacologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Antagonistas Muscarínicos/farmacologia , Ratos , Ratos Wistar , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Receptores de Glutamato Metabotrópico/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Transmissão Sináptica/efeitos dos fármacos
6.
PLoS Biol ; 8(4): e1000352, 2010 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-20405049

RESUMO

Seizures in focal epilepsies are sustained by a highly synchronous neuronal discharge that arises at restricted brain sites and subsequently spreads to large portions of the brain. Despite intense experimental research in this field, the earlier cellular events that initiate and sustain a focal seizure are still not well defined. Their identification is central to understand the pathophysiology of focal epilepsies and to develop new pharmacological therapies for drug-resistant forms of epilepsy. The prominent involvement of astrocytes in ictogenesis was recently proposed. We test here whether a cooperation between astrocytes and neurons is a prerequisite to support ictal (seizure-like) and interictal epileptiform events. Simultaneous patch-clamp recording and Ca2+ imaging techniques were performed in a new in vitro model of focal seizures induced by local applications of N-methyl-D-aspartic acid (NMDA) in rat entorhinal cortex slices. We found that a Ca2+ elevation in astrocytes correlates with both the initial development and the maintenance of a focal, seizure-like discharge. A delayed astrocyte activation during ictal discharges was also observed in other models (including the whole in vitro isolated guinea pig brain) in which the site of generation of seizure activity cannot be precisely monitored. In contrast, interictal discharges were not associated with Ca2+ changes in astrocytes. Selective inhibition or stimulation of astrocyte Ca2+ signalling blocked or enhanced, respectively, ictal discharges, but did not affect interictal discharge generation. Our data reveal that neurons engage astrocytes in a recurrent excitatory loop (possibly involving gliotransmission) that promotes seizure ignition and sustains the ictal discharge. This neuron-astrocyte interaction may represent a novel target to develop effective therapeutic strategies to control seizures.


Assuntos
Potenciais de Ação/fisiologia , Astrócitos/fisiologia , Convulsões/fisiopatologia , 4-Aminopiridina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/metabolismo , Quelantes/metabolismo , Ácido Egtázico/análogos & derivados , Ácido Egtázico/metabolismo , Córtex Entorrinal/citologia , Córtex Entorrinal/fisiopatologia , Agonistas de Aminoácidos Excitatórios/metabolismo , Ácido Glutâmico/metabolismo , Cobaias , Camundongos , Camundongos Transgênicos , N-Metilaspartato/metabolismo , Neurônios/fisiologia , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/metabolismo , Ratos , Ratos Wistar
7.
Neuron ; 111(7): 920-921, 2023 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-37023713

RESUMO

In this issue of Neuron, Yang et al.1 highlight a hitherto unknown action of cocaine in VTA circuitry. They found that chronic cocaine use increased tonic inhibition selectively onto GABA neurons through Swell1 channel-dependent GABA release from astrocytes, leading to disinhibition-mediated hyperactivity in DA neurons and addictive behavior.


Assuntos
Transtornos Relacionados ao Uso de Cocaína , Cocaína , Humanos , Astrócitos , Área Tegmentar Ventral/fisiologia , Cocaína/farmacologia , Neurônios GABAérgicos , Neurônios Dopaminérgicos/fisiologia
8.
Cells ; 11(9)2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35563706

RESUMO

At glutamatergic synapses, astrocytes respond to the neurotransmitter glutamate with intracellular Ca2+ elevations and the release of gliotransmitters that modulate synaptic transmission. While the functional interactions between neurons and astrocytes have been intensively studied at glutamatergic synapses, the role of astrocytes at GABAergic synapses has been less investigated. In the present study, we combine optogenetics with 2-photon Ca2+ imaging experiments and patch-clamp recording techniques to investigate the signaling between Somatostatin (SST)-releasing GABAergic interneurons and astrocytes in brain slice preparations from the visual cortex (VCx). We found that an intense stimulation of SST interneurons evokes Ca2+ elevations in astrocytes that fundamentally depend on GABAB receptor (GABABR) activation, and that this astrocyte response is modulated by the neuropeptide somatostatin. After episodes of SST interneuron hyperactivity, we also observed a long-lasting reduction of the inhibitory postsynaptic current (IPSC) amplitude onto pyramidal neurons (PNs). This reduction of inhibitory tone (i.e., disinhibition) is counterbalanced by the activation of astrocytes that upregulate SST interneuron-evoked IPSC amplitude by releasing ATP that, after conversion to adenosine, activates A1Rs. Our results describe a hitherto unidentified modulatory mechanism of inhibitory transmission to VCx layer II/III PNs that involves the functional recruitment of astrocytes by SST interneuron signaling.


Assuntos
Astrócitos , Córtex Visual , Astrócitos/metabolismo , Interneurônios/metabolismo , Somatostatina/metabolismo , Ácido gama-Aminobutírico
9.
Front Cell Neurosci ; 16: 919493, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35936501

RESUMO

Dravet Syndrome (DS) is a rare autosomic encephalopathy with epilepsy linked to Nav1.1 channel mutations and defective GABAergic signaling. Effective therapies for this syndrome are lacking, urging a better comprehension of the mechanisms involved. In a recognized mouse model of DS, we studied GABA tonic current, a form of inhibition largely neglected in DS, in brain slices from developing mice before spontaneous seizures are reported. In neurons from the temporal cortex (TeCx) and CA1 region, GABA tonic current was reduced in DS mice compared to controls, while in the entorhinal cortex (ECx) it was not affected. In this region however allopregnanonole potentiation of GABA tonic current was reduced in DS mice, suggesting altered extrasynaptic GABAA subunits. Using THIP as a selective agonist, we found reduced δ subunit mediated tonic currents in ECx of DS mice. Unexpectedly in the dentate gyrus (DG), a region with high δ subunit expression, THIP-evoked currents in DS mice were larger than in controls. An immunofluorescence study confirmed that δ subunit expression was reduced in ECx and increased in DG of DS mice. Finally, considering the importance of neuroinflammation in epilepsy and neurodevelopmental disorders, we evaluated classical markers of glia activation. Our results show that DS mice have increased Iba1 reactivity and GFAP expression in both ECx and DG, compared to controls. Altogether we report that before spontaneous seizures, DS mice develop significant alterations of GABA tonic currents and glial cell activation. Understanding all the mechanisms involved in these alterations during disease maturation and progression may unveil new therapeutic targets.

11.
Nat Neurosci ; 25(12): 1639-1650, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36396976

RESUMO

The plasticity of glutamatergic transmission in the ventral tegmental area (VTA) represents a fundamental mechanism in the modulation of dopamine neuron burst firing and phasic dopamine release at target regions. These processes encode basic behavioral responses, including locomotor activity, learning and motivated behaviors. Here we describe a hitherto unidentified mechanism of long-term synaptic plasticity in mouse VTA. We found that the burst firing in individual dopamine neurons induces a long-lasting potentiation of excitatory synapses on adjacent dopamine neurons that crucially depends on Ca2+ elevations in astrocytes, mediated by endocannabinoid CB1 and dopamine D2 receptors co-localized at the same astrocytic process, and activation of pre-synaptic metabotropic glutamate receptors. Consistent with these findings, selective in vivo activation of astrocytes increases the burst firing of dopamine neurons in the VTA and induces locomotor hyperactivity. Astrocytes play, therefore, a key role in the modulation of VTA dopamine neuron functional activity.


Assuntos
Neurônios Dopaminérgicos , Área Tegmentar Ventral , Animais , Camundongos , Astrócitos , Dopamina , Receptores de Dopamina D2
12.
Front Cell Neurosci ; 15: 673433, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34163329

RESUMO

The glial cells astrocytes have long been recognized as important neuron-supporting elements in brain development, homeostasis, and metabolism. After the discovery that the reciprocal communication between astrocytes and neurons is a fundamental mechanism in the modulation of neuronal synaptic communication, over the last two decades astrocytes became a hot topic in neuroscience research. Crucial to their functional interactions with neurons are the cytosolic Ca2+ elevations that mediate gliotransmission. Large attention has been posed to the so-called Ca2+microdomains, dynamic Ca2+ changes spatially restricted to fine astrocytic processes including perisynaptic astrocytic processes (PAPs). With presynaptic terminals and postsynaptic neuronal membranes, PAPs compose the tripartite synapse. The distinct spatial-temporal features and functional roles of astrocyte microdomain Ca2+ activity remain poorly defined. However, thanks to the development of genetically encoded Ca2+ indicators (GECIs), advanced microscopy techniques, and innovative analytical approaches, Ca2+ transients in astrocyte microdomains were recently studied in unprecedented detail. These events have been observed to occur much more frequently (∼50-100-fold) and dynamically than somatic Ca2+ elevations with mechanisms that likely involve both IP3-dependent and -independent pathways. Further progress aimed to clarify the complex, dynamic machinery responsible for astrocytic Ca2+ activity at microdomains is a crucial step in our understanding of the astrocyte role in brain function and may also reveal astrocytes as novel therapeutic targets for different brain diseases. Here, we review the most recent studies that improve our mechanistic understanding of the essential features of astrocyte Ca2+ microdomains.

13.
Epilepsia ; 51(8): 1493-502, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20067512

RESUMO

PURPOSE: Despite intensive studies, our understanding of the cellular and molecular mechanisms underlying epileptogenesis remains largely unsatisfactory. Our defective knowledge derives in part from the lack of adequate experimental models of the distinct phases that characterize the epileptic event, that is, initiation, propagation, and cessation. The aim of our study is the development of a new brain slice model in which a focal seizure can be repetitively evoked at a precise and predictable site. METHODS: Epileptiform activities were studied by fast Ca²(+) imaging coupled with simultaneous single and double patch-clamp or extracellular recordings from neurons of entorhinal cortex (EC) slices from Wistar rats and C57BL6J mice at postnatal days 13-17. RESULTS: In the presence of 4-aminopyridine (4-AP) and low Mg²(+) , activation of layer V-VI neurons by local N-methyl-d-aspartate (NMDA) applications evolved into an ictal discharge (ID) that propagated to the entire EC. NMDA-evoked IDs were similar to spontaneous events. IDs with similar pattern and duration could be repetitively triggered from the same site by successive NMDA stimulations. The high ID reproducibility is an important feature of the model that allowed testing of the effects of currently used antiepileptic drugs (AEDs) on initiation, propagation, and cessation of focal ictal events. CONCLUSIONS: By offering the unique opportunity to repetitively evoke an ID from the same restricted site, this model represents a powerful approach to study the cellular and molecular events at the basis of initiation, propagation, and cessation of focal seizures.


Assuntos
Potenciais de Ação/fisiologia , Córtex Entorrinal/citologia , Córtex Entorrinal/fisiologia , Neurônios/fisiologia , 4-Aminopiridina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Anticonvulsivantes/farmacologia , Cálcio/metabolismo , Estimulação Elétrica/métodos , Córtex Entorrinal/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp/métodos , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Wistar
14.
Nat Neurosci ; 23(1): 47-60, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31844317

RESUMO

The prefrontal cortex (PFC) is implicated in processing of the affective state of others through non-verbal communication. This social cognitive function is thought to rely on an intact cortical neuronal excitatory and inhibitory balance. Here combining in vivo electrophysiology with a behavioral task for affective state discrimination in mice, we show a differential activation of medial PFC (mPFC) neurons during social exploration that depends on the affective state of the conspecific. Optogenetic manipulations revealed a double dissociation between the role of interneurons in social cognition. Specifically, inhibition of mPFC somatostatin (SOM+), but not of parvalbumin (PV+) interneurons, abolishes affective state discrimination. Accordingly, synchronized activation of mPFC SOM+ interneurons selectively induces social discrimination. As visualized by in vivo single-cell microendoscopic Ca2+ imaging, an increased synchronous activity of mPFC SOM+ interneurons, guiding inhibition of pyramidal neurons, is associated with affective state discrimination. Our findings provide new insights into the neurobiological mechanisms of affective state discrimination.


Assuntos
Afeto/fisiologia , Interneurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Comportamento Social , Animais , Masculino , Camundongos , Somatostatina/metabolismo
15.
Methods Mol Biol ; 1925: 173-182, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30674027

RESUMO

In brain networks, neurons are constantly involved in a dynamic interaction with the other cell populations and, particularly, with astrocytes, the most abundant glial cells in the brain. Astrocytes respond to neurotransmitters with Ca2+ elevations which represent a key event in the modulation of local brain circuits played by these glial cells. Due to technical limitations, the study of Ca2+ signal dynamics in astrocytes has focused for decades almost exclusively on somatic and perisomatic regions. Accordingly, Ca2+ signal in astrocytic fine protrusions, which are in close contact with the synapse, has been poorly investigated. Over the last years, the diffusion of novel tools such as the viral vector gene delivery of genetically encoded Ca2+ indicators (GECI), the optogenetics, and multiphoton laser scanning microscopy has boosted significantly our capability to study astrocytic Ca2+ signals in the different subcellular compartments. Here we report a protocol that combines these techniques to study astrocyte Ca2+ signaling in response to somatostatin (SST)-expressing interneurons, one of the main classes of GABAergic inhibitory interneurons.


Assuntos
Astrócitos/metabolismo , Cálcio/metabolismo , Interneurônios/metabolismo , Microscopia Confocal/métodos , Optogenética/métodos , Animais , Astrócitos/citologia , Astrócitos/ultraestrutura , Cálcio/análise , Sinalização do Cálcio , Cátions Bivalentes/análise , Cátions Bivalentes/metabolismo , Desenho de Equipamento , Interneurônios/citologia , Interneurônios/ultraestrutura , Camundongos , Microscopia Confocal/instrumentação , Optogenética/instrumentação
16.
Methods Mol Biol ; 1925: C1, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31175658

RESUMO

Figures 1 and 2 were inadvertently switched during the production and this has been corrected so the figures appear in the proper order.

17.
iScience ; 16: 340-355, 2019 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-31203189

RESUMO

Genetically Encoded Ca2+ Indicators (GECIs) are extensively used to study organelle Ca2+ homeostasis, although some available probes are still plagued by a number of problems, e.g., low fluorescence intensity, partial mistargeting, and pH sensitivity. Furthermore, in the most commonly used mitochondrial Förster Resonance Energy Transfer based-GECIs, the donor protein ECFP is characterized by a double exponential lifetime that complicates the fluorescence lifetime analysis. We have modified the cytosolic and mitochondria-targeted Cameleon GECIs by (1) substituting the donor ECFP with mCerulean3, a brighter and more stable fluorescent protein with a single exponential lifetime; (2) extensively modifying the constructs to improve targeting efficiency and fluorescence changes caused by Ca2+ binding; and (3) inserting the cDNAs into adeno-associated viral vectors for in vivo expression. The probes have been thoroughly characterized in situ by fluorescence microscopy and Fluorescence Lifetime Imaging Microscopy, and examples of their ex vivo and in vivo applications are described.

18.
Nat Commun ; 9(1): 82, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29311610

RESUMO

The signaling diversity of GABAergic interneurons to post-synaptic neurons is crucial to generate the functional heterogeneity that characterizes brain circuits. Whether this diversity applies to other brain cells, such as the glial cells astrocytes, remains unexplored. Using optogenetics and two-photon functional imaging in the adult mouse neocortex, we here reveal that parvalbumin- and somatostatin-expressing interneurons, two key interneuron classes in the brain, differentially signal to astrocytes inducing weak and robust GABAB receptor-mediated Ca2+ elevations, respectively. Furthermore, the astrocyte response depresses upon parvalbumin interneuron repetitive stimulations and potentiates upon somatostatin interneuron repetitive stimulations, revealing a distinguished astrocyte plasticity. Remarkably, the potentiated response crucially depends on the neuropeptide somatostatin, released by somatostatin interneurons, which activates somatostatin receptors at astrocytic processes. Our study unveils, in the living brain, a hitherto unidentified signaling specificity between interneuron subtypes and astrocytes opening a new perspective into the role of astrocytes as non-neuronal components of inhibitory circuits.


Assuntos
Astrócitos/metabolismo , Interneurônios/metabolismo , Transdução de Sinais , Córtex Somatossensorial/metabolismo , Somatostatina/metabolismo , Animais , Cálcio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neocórtex/citologia , Neocórtex/metabolismo , Plasticidade Neuronal , Optogenética , Parvalbuminas/metabolismo , Técnicas de Patch-Clamp , Receptores de GABA-B/metabolismo
20.
J Neurosci ; 26(36): 9312-22, 2006 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-16957087

RESUMO

The release of glutamate from astrocytes activates synchronous slow inward currents (SICs) in hippocampal pyramidal neurons, which are mediated by the NMDA receptor and represent a nonsynaptic mechanism to promote the synchronization of neuronal activity. Two recent studies demonstrate that SICs generate neuronal paroxysmal depolarizations resembling those typical of interictal epileptiform activity and proposed that there could be an astrocytic basis of epilepsy (Kang et al., 2005; Tian et al., 2005). We tested this hypothesis using two in vitro models of epileptiform activity in hippocampal slices. Removal of extracellular Mg2+ and application of picrotoxin or perfusion with 0.5 mM Mg2+ and 8.5 mM K+-containing saline result mainly in neuronal ictal- and interictal-like epileptiform activity, respectively. Although both models trigger epileptiform activity, astrocytic Ca2+ oscillations were increased only after slice perfusion with 0 mM Mg2+ and picrotoxin. The activation of astrocytic Ca2+ signaling correlates with an increased frequency of SICs, and, when paired neurons were within 100 microm of one another with synchronous neuronal Ca2+ elevations, the generation of synchronous neuronal depolarizations and action potential discharges. TTX blocked both ictal- and interictal-like epileptiform activity without affecting SICs or SIC-mediated neuronal synchronization. In contrast, NMDA receptor antagonists, which block SICs, did not prevent the generation of either ictal- or interictal-like events. Based on this clear-cut pharmacology, our data demonstrate that nonsynaptic glutamate release from astrocytes is not necessary for the generation of epileptiform activity in vitro, although we cannot exclude the possibility that it may modulate the strength of the ictal (seizure)-like event.


Assuntos
Potenciais de Ação/fisiologia , Astrócitos/fisiologia , Relógios Biológicos/fisiologia , Epilepsia/fisiopatologia , Ácido Glutâmico/metabolismo , Células Piramidais/fisiologia , Animais , Comunicação Celular/fisiologia , Células Cultivadas , Hipocampo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA