Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 19522, 2023 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-37945644

RESUMO

Myelination is a process tightly regulated by a variety of neurotrophic factors. Here, we show-by analyzing two transgenic mouse lines, one overexpressing EPO selectively in the brain Tg21(PDGFB-rhEPO) and another with targeted removal of EPO receptors (EPORs) from oligodendrocyte progenitor cells (OPC)s (Sox10-cre;EpoRfx/fx mice)-a key function for EPO in regulating developmental brain myelination. Overexpression of EPO resulted in faster postnatal brain growth and myelination, an increased number of myelinating oligodendrocytes, faster axonal myelin ensheathment, and improved motor coordination. Conversely, targeted ablation of EPORs from OPCs reduced the number of mature oligodendrocytes and impaired motor coordination during the second postnatal week. Furthermore, we found that EPORs are transiently expressed in the subventricular zone (SVZ) during the second postnatal week and EPO increases the postnatal expression of essential oligodendrocyte pro-differentiation and pro-maturation (Nkx6.2 and Myrf) transcripts, and the Nfatc2/calcineurin pathway. In contrast, ablation of EPORs from OPCs inactivated the Erk1/2 pathway and reduced the postnatal expression of the transcripts. Our results reveal developmental time windows in which EPO therapies could be highly effective for stimulating oligodendrocyte maturation and myelination.


Assuntos
Eritropoetina , Oligodendroglia , Camundongos , Animais , Oligodendroglia/metabolismo , Bainha de Mielina/metabolismo , Encéfalo/metabolismo , Camundongos Transgênicos , Eritropoetina/genética , Eritropoetina/farmacologia , Eritropoetina/metabolismo , Diferenciação Celular/fisiologia
2.
eNeuro ; 8(1)2021.
Artigo em Inglês | MEDLINE | ID: mdl-33495244

RESUMO

Several neurodevelopmental disabilities are strongly associated with alterations in GABAergic transmission, and therapies to stimulate its normal development are lacking. Erythropoietin (EPO) is clinically used in neonatology to mitigate acute brain injury, and to stimulate neuronal maturation. Yet it remains unclear whether EPO can stimulate maturation of the GABAergic system. Here, with the use of a transgenic mouse line that constitutively overexpresses neuronal EPO (Tg21), we show that EPO stimulates postnatal GABAergic maturation in the hippocampus. We show an increase in hippocampal GABA-immunoreactive neurons, and postnatal elevation of interneurons expressing parvalbumin (PV), somatostatin (SST), and neuropeptide Y (NPY). Analysis of perineuronal net (PNN) formation and innervation of glutamatergic terminals onto PV+ cells, shows to be enhanced early in postnatal development. Additionally, an increase in GABAAergic synapse density and IPSCs in CA1 pyramidal cells from Tg21 mice is observed. Detection of EPO receptor (EPOR) mRNA was observed to be restricted to glutamatergic pyramidal cells and increased in Tg21 mice at postnatal day (P)7, along with reduced apoptosis. Our findings show that EPO can stimulate postnatal GABAergic maturation in the hippocampus, by increasing neuronal survival, modulating critical plasticity periods, and increasing synaptic transmission. Our data supports EPO's clinical use to balance GABAergic dysfunction.


Assuntos
Eritropoetina , Parvalbuminas , Animais , Neurônios GABAérgicos/metabolismo , Hipocampo/metabolismo , Interneurônios/metabolismo , Camundongos , Camundongos Transgênicos , Parvalbuminas/metabolismo
3.
Eur J Neurosci ; 52(5): 3353-3374, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32599671

RESUMO

Lack of dopamine (DA) in the striatum and the consequential dysregulation of thalamocortical circuits are major causes of motor impairments in Parkinson's disease. The striatum receives multiple cortical and subcortical afferents. Its role in movement control and motor skills learning is regulated by DA from the nigrostriatal pathway. In Parkinson's disease, DA loss affects striatal network activity and induces a functional imbalance of its output pathways, impairing thalamocortical function. Striatal projection neurons are GABAergic and form two functionally antagonistic pathways: the direct pathway, originating from DA receptor type 1-expressing medium spiny neurons (D1 R-MSN), and the indirect pathway, from D2 R-MSN. Here, we investigated whether DA depletion in mouse striatum also affects GABAergic function. We recorded GABAergic miniature IPSCs (mIPSC) and tonic inhibition from D1 R- and D2 R-MSN and used immunohistochemical labeling to study GABAA R function and subcellular distribution in DA-depleted and control mice. We observed slower decay kinetics and increased tonic inhibition in D1 R-MSN, while D2 R-MSN had increased mIPSC frequency after DA depletion. Perisomatic synapses containing the GABAA R subunits α1 or α2 were not affected, but there was a strong decrease in non-synaptic GABAA Rs containing these subunits, suggesting altered receptor trafficking. To broaden these findings, we also investigated GABAA Rs in GABAergic and cholinergic interneurons and found cell type-specific alterations in receptor distribution, likely reflecting changes in connectivity. Our results reveal that chronic DA depletion alters striatal GABAergic transmission, thereby affecting cellular and circuit activity. These alterations either result from pathological changes or represent a compensatory mechanism to counteract imbalance of output pathways.


Assuntos
Corpo Estriado , Dopamina , Animais , Corpo Estriado/metabolismo , Neurônios Dopaminérgicos/metabolismo , Camundongos , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo
4.
J Neurophysiol ; 123(4): 1496-1503, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32159408

RESUMO

As the main input nucleus of the basal ganglia, the striatum plays a central role in planning, control, and execution of movement and motor skill learning. More than 90% of striatal neurons, so-called medium spiny neurons (MSN), are GABAergic projection neurons, innervating primarily the substantia nigra pars reticulata or the globus pallidus internus. The remaining neurons are GABAergic and cholinergic interneurons, synchronizing and controlling striatal output by reciprocal connections with MSN. Besides prominent local cholinergic influence, striatal function is globally regulated by dopamine (DA) from the nigrostriatal pathway. Little is known about whether DA depletion, as occurs in Parkinson's disease, affects the activity of striatal interneurons. Here we focused on neuropeptide Y (NPY)-expressing interneurons, which are among the major subgroups of GABAergic interneurons in the striatum. We investigated the effects of striatal DA depletion on GABAergic transmission in NPY interneurons by electrophysiologically recording GABAergic spontaneous (s) and miniature (m) inhibitory postsynaptic currents (IPSCs) in identified NPY interneurons in slices from 6-hydroxydopamine (6-OHDA)- and vehicle-injected transgenic NPY-humanized Renilla green fluorescent protein (hrGFP) mice with the whole cell patch-clamp technique. We report a significant increase in sIPSC and mIPSC frequency as well as the occurrence of giant synaptic and burst sIPSCs in the 6-OHDA group, suggesting changes in GABAergic circuit activity and synaptic transmission. IPSC kinetics remained unchanged, pointing to mainly presynaptic changes in GABAergic transmission. These results show that chronic DA depletion following 6-OHDA injection causes activity-dependent and -independent increase of synaptic GABAergic inhibition onto striatal NPY interneurons, confirming their involvement in the functional impairments of the DA-depleted striatum.NEW & NOTEWORTHY Neuropeptide Y (NPY) interneurons regulate the function of striatal projection neurons and are upregulated upon dopamine depletion in the striatum. Here we investigated how dopamine depletion affects NPY circuits and show electrophysiologically that it leads to the occurrence of giant synaptic and burst GABAergic spontaneous inhibitory postsynaptic currents (IPSCs) and to an activity-independent increase in GABAergic miniature IPSC frequency in NPY neurons. We suggest that degeneration of dopaminergic terminals in the striatum causes functional changes in striatal GABAergic function.


Assuntos
Corpo Estriado/fisiologia , Dopamina/metabolismo , Neurônios GABAérgicos/fisiologia , Potenciais Pós-Sinápticos Inibidores/fisiologia , Interneurônios/fisiologia , Neuropeptídeo Y/metabolismo , Transmissão Sináptica/fisiologia , Adrenérgicos/farmacologia , Animais , Corpo Estriado/metabolismo , Neurônios GABAérgicos/metabolismo , Interneurônios/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxidopamina/farmacologia , Técnicas de Patch-Clamp
5.
Sci Rep ; 10(1): 3760, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-32111960

RESUMO

Epilepsy is a widespread neurological disease characterized by abnormal neuronal activity resulting in recurrent seizures. There is mounting evidence that a circadian system disruption, involving clock genes and their downstream transcriptional regulators, is associated with epilepsy. In this study, we characterized the hippocampal expression of clock genes and PAR bZIP transcription factors (TFs) in a mouse model of temporal lobe epilepsy induced by intrahippocampal injection of kainic acid (KA). The expression of PAR bZIP TFs was significantly altered following KA injection as well as in other rodent models of acquired epilepsy. Although the PAR bZIP TFs are regulated by proinflammatory cytokines in peripheral tissues, we discovered that the regulation of their expression is inflammation-independent in hippocampal tissue and rather mediated by clock genes and hyperexcitability. Furthermore, we report that hepatic leukemia factor (Hlf), a member of PAR bZIP TFs family, is invariably downregulated in animal models of acquired epilepsy, regulates neuronal activity in vitro and its overexpression in dentate gyrus neurons in vivo leads to altered expression of genes associated with seizures and epilepsy. Overall, our study provides further evidence of PAR bZIP TFs involvement in epileptogenesis and points to Hlf as the key player.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Giro Denteado/metabolismo , Epilepsia/metabolismo , Regulação da Expressão Gênica , Animais , Giro Denteado/patologia , Modelos Animais de Doenças , Epilepsia/induzido quimicamente , Ácido Caínico/efeitos adversos , Ácido Caínico/farmacologia , Masculino , Camundongos
6.
J Comp Neurol ; 527(12): 2030-2046, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-30773633

RESUMO

The striatum is the main input nucleus of the basal ganglia, mediating motor and cognitive functions. Striatal projection neurons are GABAergic medium spiny neurons (MSN), expressing either the dopamine receptor type 1 (D1 -R MSN) and forming the direct, movement-promoting pathway, or dopamine receptor type 2 (D2 -R MSN), forming the indirect movement-suppressing pathway. Locally, activity and synchronization of MSN are modulated by several subtypes of GABAergic and cholinergic interneurons. Overall, GABAergic circuits in the striatum remain poorly characterized, and little is known about the intrastriatal connectivity of interneurons and the distribution of GABAA receptor (GABAA R) subtypes, distinguished by their subunit composition, in striatal synapses. Here, by using immunofluorescence in mouse tissue, we investigated the distribution of GABAA Rs containing the α1 , α2 , or α3 subunit in perisomatic synapses of striatal MSN and interneurons, as well as the innervation pattern of D1 R- and D2 R-MSN soma and axonal initial segment (AIS) by GABAergic and cholinergic interneurons. Our results show that perisomatic GABAergic synapses of D1 R- and D2 R-MSN contain the GABAA R α1 and/or α2 subunits, but not the α3 subunit; D2 R-MSN have significantly more α1 -GABAA Rs on their soma than D1 R-MSN. Further, interneurons have few perisomatic synapses containing α2 -GABAA Rs, whereas α3 -GABAA Rs (along with the α1 -GABAA Rs) are abundant in perisomatic synapses of CCK+ , NPY+ /SOM+ , and vAChT+ interneurons. Each MSN and interneuron population analyzed received a distinct pattern of GABAergic and cholinergic innervation, complementing this postsynaptic heterogeneity. In conclusion, intra-striatal GABAergic circuits are distinguished by cell-type specific innervation patterns, differential expression and postsynaptic targeting of GABAA R subtypes.


Assuntos
Corpo Estriado/citologia , Corpo Estriado/metabolismo , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais/citologia , Vias Neurais/metabolismo , Receptores de GABA-A/análise , Receptores de GABA-A/metabolismo
7.
J Neurosci ; 39(12): 2238-2250, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30655357

RESUMO

Gastrin-releasing peptide (GRP) is a spinal itch transmitter expressed by a small population of dorsal horn interneurons (GRP neurons). The contribution of these neurons to spinal itch relay is still only incompletely understood, and their potential contribution to pain-related behaviors remains controversial. Here, we have addressed this question in a series of experiments performed in GRP::cre and GRP::eGFP transgenic male mice. We combined behavioral tests with neuronal circuit tracing, morphology, chemogenetics, optogenetics, and electrophysiology to obtain a more comprehensive picture. We found that GRP neurons form a rather homogeneous population of central cell-like excitatory neurons located in lamina II of the superficial dorsal horn. Multicolor high-resolution confocal microscopy and optogenetic experiments demonstrated that GRP neurons receive direct input from MrgprA3-positive pruritoceptors. Anterograde HSV-based neuronal tracing initiated from GRP neurons revealed ascending polysynaptic projections to distinct areas and nuclei in the brainstem, midbrain, thalamus, and the somatosensory cortex. Spinally restricted ablation of GRP neurons reduced itch-related behaviors to different pruritogens, whereas their chemogenetic excitation elicited itch-like behaviors and facilitated responses to several pruritogens. By contrast, responses to painful stimuli remained unaltered. These data confirm a critical role of dorsal horn GRP neurons in spinal itch transmission but do not support a role in pain.SIGNIFICANCE STATEMENT Dorsal horn gastrin-releasing peptide neurons serve a well-established function in the spinal transmission of pruritic (itch) signals. A potential role in the transmission of nociceptive (pain) signals has remained controversial. Our results provide further support for a critical role of dorsal horn gastrin-releasing peptide neurons in itch circuits, but we failed to find evidence supporting a role in pain.


Assuntos
Peptídeo Liberador de Gastrina/fisiologia , Nociceptividade/fisiologia , Dor/fisiopatologia , Células do Corno Posterior/fisiologia , Prurido/fisiopatologia , Animais , Modelos Animais de Doenças , Peptídeo Liberador de Gastrina/metabolismo , Interneurônios/metabolismo , Interneurônios/patologia , Interneurônios/fisiologia , Masculino , Camundongos Transgênicos , Dor/complicações , Dor/patologia , Células do Corno Posterior/metabolismo , Células do Corno Posterior/patologia , Prurido/complicações , Prurido/patologia
8.
Nat Commun ; 9(1): 3130, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30087324

RESUMO

Fast inhibitory synaptic transmission is mediated by γ-aminobutyric acid type A receptors (GABAARs) that are enriched at functionally diverse synapses via mechanisms that remain unclear. Using isothermal titration calorimetry and complementary methods we demonstrate an exclusive low micromolar binding of collybistin to the α2-subunit of GABAARs. To explore the biological relevance of collybistin-α2-subunit selectivity, we generate mice with a mutation in the α2-subunit-collybistin binding region (Gabra2-1). The mutation results in loss of a distinct subset of inhibitory synapses and decreased amplitude of inhibitory synaptic currents. Gabra2-1 mice have a striking phenotype characterized by increased susceptibility to seizures and early mortality. Surviving Gabra2-1 mice show anxiety and elevations in electroencephalogram δ power, which are ameliorated by treatment with the α2/α3-selective positive modulator, AZD7325. Taken together, our results demonstrate an α2-subunit selective binding of collybistin, which plays a key role in patterned brain activity, particularly during development.


Assuntos
Receptores de GABA-A/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Convulsões/tratamento farmacológico , Convulsões/mortalidade , Animais , Encéfalo/metabolismo , Eletroencefalografia , Células HEK293 , Compostos Heterocíclicos com 2 Anéis/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Peptídeos/química , Fenótipo , Ligação Proteica , Domínios Proteicos , Receptores de GABA-A/genética , Sinapses/metabolismo , Transmissão Sináptica
9.
J Neurochem ; 147(4): 477-494, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30142695

RESUMO

In dissociated neuronal cultures the absence of spatial and temporal cues causes the emergence of mismatched synapses, where post-synaptic proteins of GABAergic synapses are in part apposed to glutamatergic pre-synaptic terminals and vice versa. This mismatch offers an opportunity to study the mechanisms that regulate correct apposition of pre- and post-synaptic elements. We report here that the IQ motif and Sec7 domain-containing protein 3 (IQSEC3; BRAG3; synArfGEF) specifically regulates the mislocalization of GABAergic post-synaptic density (PSD) proteins. Over-expression of IQSEC3 constructs harboring mutations that ablate Sec7 domain or IQ motif function revealed that IQSEC3 catalytic activity is involved in the control of apposition between the GABAergic PSD and glutamatergic terminals. Neurons co-expressing eGFP-gephyrin with IQSEC3 Sec7 mutant displayed a drastically increased fraction of mismatched eGFP-gephyrin clusters compared to other IQSEC3 constructs. Along with eGFP-gephyrin, endogenous GABAA receptor cluster mismatching was increased by IQSEC3 Sec7 mutant over-expression. Conversely, GFP-PSD-95 clusters were unaffected by over-expression of any IQSEC3 construct. The GABAergic PSD mismatch phenotype was recapitulated by Arf6 dominant-negative mutant over-expression, suggesting that Arf6 activation by IQSEC3 is an essential step in this pathway. In addition, we provide biochemical evidence to confirm gephyrin/IQSEC3 interaction near the IQSEC3 IQ motif, which in turn binds calmodulin at low Ca2+ concentrations. Taken together, our findings identify a post-synaptic protein which specifically regulates correct apposition of the GABAergic PSD to pre-synaptic terminals.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Neurotransmissores/metabolismo , Sinapses/metabolismo , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/metabolismo , Animais , Cálcio/metabolismo , Calmodulina/metabolismo , Catálise , Feminino , Fatores de Troca do Nucleotídeo Guanina/genética , Hipocampo/citologia , Processamento de Imagem Assistida por Computador , Densidade Pós-Sináptica , Gravidez , Terminações Pré-Sinápticas/metabolismo , Cultura Primária de Células , Ratos , Ratos Wistar , Ácido gama-Aminobutírico/fisiologia
10.
Eur J Neurosci ; 47(12): 1534-1562, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29862588

RESUMO

Aberrant epileptic activity is detectable at early disease stages in Alzheimer's disease (AD) patients and in AD mouse models. Here, we investigated in young ArcticAß mice whether AD-like pathology renders neuronal networks more susceptible to the development of acquired epilepsy induced by unilateral intrahippocampal injection of kainic acid (IHK). In this temporal lobe epilepsy model, IHK induces a status epilepticus followed after two weeks by spontaneous recurrent seizures (SRS). ArcticAß mice exhibited more severe status epilepticus and early onset of SRS. This hyperexcitable phenotype was characterized in CA1 neurons by decreased synaptic strength, increased kainic acid-induced LTP and reduced frequency of spontaneous inhibitory currents. However, no difference in neurodegeneration, neuroinflammation, axonal reorganization or adult neurogenesis was observed in ArcticAß mice compared to wild-type littermates following IHK-induced epileptogenesis. Neuropeptide Y (NPY) expression was reduced at baseline and its IHK-induced elevation in mossy fibres and granule cells was attenuated. However, although this alteration might underlie premature seizure onset, neutralization of soluble Aß species by intracerebroventricular Aß-specific antibody application mitigated the hyperexcitable phenotype of ArcticAß mice and prevented early SRS onset. Therefore, the development of seizures at early stages of AD is mediated primarily by Aß species causing widespread changes in synaptic function.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Epilepsia do Lobo Temporal , Hipocampo , Plasticidade Neuronal , Convulsões , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Animais , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/fisiopatologia , Giro Denteado/metabolismo , Giro Denteado/fisiopatologia , Modelos Animais de Doenças , Eletroencefalografia , Epilepsia do Lobo Temporal/metabolismo , Epilepsia do Lobo Temporal/fisiopatologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Ácido Caínico/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Plasticidade Neuronal/fisiologia , Convulsões/metabolismo , Convulsões/fisiopatologia , Estado Epiléptico/metabolismo , Estado Epiléptico/fisiopatologia
11.
Neuron ; 96(4): 709-711, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29144966

RESUMO

Multiple cell-adhesion molecules contribute to synapse formation by mediating trans-synaptic interactions with presynaptic signaling molecules. In this issue of Neuron, Li et al. (2017) report cooperativity between Neuroligin2 and Slitrk3, exerting distinct effects on GABAergic synapse formation in immature and mature neurons.


Assuntos
Moléculas de Adesão Celular Neuronais , Sinapses , Proteínas do Tecido Nervoso , Neurogênese
12.
PLoS Genet ; 13(10): e1007073, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29069083

RESUMO

In developing brain neuronal migration, dendrite outgrowth and dendritic spine outgrowth are controlled by Cdc42, a small GTPase of the Rho family, and its activators. Cdc42 function in promoting actin polymerization is crucial for glutamatergic synapse regulation. Here, we focus on GABAergic synapse-specific activator of Cdc42, collybistin (CB) and examine functional differences between its splice isoforms CB1 and CB2. We report that CB1 and CB2 differentially regulate GABAergic synapse formation in vitro along proximal-distal axis and adult-born neuron maturation in vivo. The functional specialization between CB1 and CB2 isoforms arises from their differential protein half-life, in turn regulated by ubiquitin conjugation of the unique CB1 C-terminus. We report that CB1 and CB2 negatively regulate Cdc42; however, Cdc42 activation is dependent on CB interaction with gephyrin. During hippocampal adult neurogenesis CB1 regulates neuronal migration, while CB2 is essential for dendrite outgrowth. Finally, using mice lacking Gabra2 subunit, we show that CB1 function is downstream of GABAARs, and we can rescue adult neurogenesis deficit observed in Gabra2 KO. Overall, our results uncover previously unexpected role for CB isoforms downstream of α2-containing GABAARs during neuron maturation in a Cdc42 dependent mechanism.


Assuntos
Neurônios/fisiologia , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Sinapses/fisiologia , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Movimento Celular , Regulação da Expressão Gênica , Células HEK293 , Hipocampo/citologia , Hipocampo/fisiologia , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurogênese , Neurônios/citologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de GABA-A/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo
13.
Brain Struct Funct ; 222(9): 4149-4161, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28643105

RESUMO

Perisomatic GABAergic synapses onto hippocampal pyramidal cells arise from two populations of basket cells with different neurochemical and functional properties. The presence of the dystrophin-glycoprotein complex in their postsynaptic density (PSD) distinguishes perisomatic synapses from GABAergic synapses on dendrites and the axon-initial segment. Targeted deletion of neuroligin 2 (NL2), a transmembrane protein interacting with presynaptic neurexin, has been reported to disrupt postsynaptic clustering of GABAA receptors (GABAAR) and their anchoring protein, gephyrin, at perisomatic synapses. In contrast, targeted deletion of Gabra2 disrupts perisomatic clustering of gephyrin, but not of α1-GABAAR, NL2, or dystrophin/dystroglycan. Unexpectedly, conditional deletion of Dag1, encoding dystroglycan, selectively prevents the formation of perisomatic GABAergic synapses from basket cells expressing cholecystokinin. Collectively, these observations suggest that multiple mechanisms regulate formation and molecular composition of the GABAergic PSD at perisomatic synapses. Here, we further explored this issue by investigating the effect of targeted deletion of Gabra1 and NL2 on the dystrophin-glycoprotein complex and on perisomatic synapse formation, using immunofluorescence analysis with a battery of GABAergic pre- and postsynaptic markers. We show that the absence of α1-GABAAR increases GABAergic synapses containing the α2 subunit, without affecting the clustering of dystrophin and NL2; in contrast, the absence of NL2 produces highly variable effects postsynaptically, not restricted to perisomatic synapses and being more severe for the GABAAR subunits and gephyrin than dystrophin. Altogether, the results confirm the importance of NL2 as organizer of the GABAergic PSD and unravel distinct roles for α1- and α2-GABAARs in the formation of GABAergic circuits in close interaction with the dystrophin-glycoprotein complex.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Neurônios GABAérgicos/fisiologia , Regulação da Expressão Gênica/genética , Hipocampo/citologia , Proteínas do Tecido Nervoso/metabolismo , Receptores de GABA-A/metabolismo , Sinapses/fisiologia , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Animais , Moléculas de Adesão Celular Neuronais/genética , Distrofina/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Parvalbuminas/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Receptores de GABA-A/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Ácido gama-Aminobutírico/metabolismo
15.
Exp Neurol ; 287(Pt 1): 44-53, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27780732

RESUMO

GABAergic disinhibition has been suggested to play a critical role in the pathophysiology of several basal ganglia disorders, including dystonia, a common movement disorder. Previous studies have shown a deficit of striatal GABAergic interneurons (IN) in the dtsz mutant hamster, one of the few phenotypic animal models of dystonia. However, mechanisms underlying this deficit are largely unknown. In the present study, we investigated the migration and maturation of striatal IN during postnatal development (18days of age) and at age of highest severity of dystonia (33days of age) in this hamster model. In line with previous findings, the density of GAD67-positive IN and the level of parvalbumin mRNA, a marker for fast spiking GABAergic IN, were lower in the dtsz mutant than in control hamsters. However, an unaltered density of Nkx2.1 labeled cells and Nkx2.1 mRNA level suggested that the migration of GABAergic IN into the striatum was not retarded. Therefore, different factors that indicate maturation of GABAergic IN were determined. While mRNA of the KCC2 cation/chloride transporters and the cytosolic carboanhydrase VII, used as markers for the so called GABA switch, as well as BDNF were unaltered, we found a reduced number of IN expressing the alpha1 subunit of the GABAA-receptor (37.5%) in dtsz hamsters at an age of 33days, but not after spontaneous remission of dystonia at an age of 90days. Since IN shift expression from alpha2 to alpha1 subunits during postnatal maturation, this result together with a decreased parvalbumin mRNA expression suggest a delayed maturation of striatal GABAergic IN in this animal model, which might underlie abnormal neuronal activity and striatal plasticity.


Assuntos
Corpo Estriado/crescimento & desenvolvimento , Corpo Estriado/patologia , Distonia/patologia , Neurônios GABAérgicos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Cricetinae , Modelos Animais de Doenças , Distonia/genética , Feminino , Neurônios GABAérgicos/enzimologia , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Glutamato Descarboxilase/metabolismo , Masculino , Mesocricetus/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Parvalbuminas/genética , Parvalbuminas/metabolismo , Fenótipo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Simportadores/genética , Simportadores/metabolismo , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Cotransportadores de K e Cl-
16.
Nat Commun ; 7: 13365, 2016 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-27819299

RESUMO

GABAA receptors (GABAARs) mediate the majority of fast inhibitory neurotransmission in the brain via synergistic association with the postsynaptic scaffolding protein gephyrin and its interaction partners. However, unlike their counterparts at glutamatergic synapses, gephyrin and its binding partners lack canonical protein interaction motifs; hence, the molecular basis for gephyrin scaffolding has remained unclear. In this study, we identify and characterize two new posttranslational modifications of gephyrin, SUMOylation and acetylation. We demonstrate that crosstalk between SUMOylation, acetylation and phosphorylation pathways regulates gephyrin scaffolding. Pharmacological intervention of SUMO pathway or transgenic expression of SUMOylation-deficient gephyrin variants rescued gephyrin clustering in CA1 or neocortical neurons of Gabra2-null mice, which otherwise lack gephyrin clusters, indicating that gephyrin SUMO modification is an essential determinant for scaffolding at GABAergic synapses. Together, our results demonstrate that concerted modifications on a protein scaffold by evolutionarily conserved yet functionally diverse signalling pathways facilitate GABAergic transmission.


Assuntos
Proteínas de Transporte/fisiologia , Neurônios GABAérgicos/fisiologia , Proteínas de Membrana/fisiologia , Transdução de Sinais/fisiologia , Transmissão Sináptica/fisiologia , Acetilação , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/fisiologia , Proteínas de Transporte/metabolismo , Feminino , Flavonas/farmacologia , Células HEK293 , Humanos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Neocórtex/citologia , Neocórtex/metabolismo , Fosforilação , Cultura Primária de Células , Ratos , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sumoilação/efeitos dos fármacos , Sinapses/fisiologia
17.
J Neurosci ; 36(40): 10296-10313, 2016 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-27707967

RESUMO

Distinct types of GABAergic interneurons target different subcellular domains of pyramidal cells, thereby shaping pyramidal cell activity patterns. Whether the presynaptic heterogeneity of GABAergic innervation is mirrored by specific postsynaptic factors is largely unexplored. Here we show that dystroglycan, a protein responsible for the majority of congenital muscular dystrophies when dysfunctional, has a function at postsynaptic sites restricted to a subset of GABAergic interneurons. Conditional deletion of Dag1, encoding dystroglycan, in pyramidal cells caused loss of CCK-positive basket cell terminals in hippocampus and neocortex. PV-positive basket cell terminals were unaffected in mutant mice, demonstrating interneuron subtype-specific function of dystroglycan. Loss of dystroglycan in pyramidal cells had little influence on clustering of other GABAergic postsynaptic proteins and of glutamatergic synaptic proteins. CCK-positive terminals were not established at P21 in the absence of dystroglycan and were markedly reduced when dystroglycan was ablated in adult mice, suggesting a role for dystroglycan in both formation and maintenance of CCK-positive terminals. The necessity of neuronal dystroglycan for functional innervation by CCK-positive basket cell axon terminals was confirmed by reduced frequency of inhibitory events in pyramidal cells of dystroglycan-deficient mice and further corroborated by the inefficiency of carbachol to increase IPSC frequency in these cells. Finally, neurexin binding seems dispensable for dystroglycan function because knock-in mice expressing binding-deficient T190M dystroglycan displayed normal CCK-positive terminals. Together, we describe a novel function of dystroglycan in interneuron subtype-specific trans-synaptic signaling, revealing correlation of presynaptic and postsynaptic molecular diversity. SIGNIFICANCE STATEMENT: Dystroglycan, an extracellular and transmembrane protein of the dystrophin-glycoprotein complex, is at the center of molecular studies of muscular dystrophies. Although its synaptic distribution in cortical brain regions is long established, function of dystroglycan in the synapse remained obscure. Using mice that selectively lack neuronal dystroglycan, we provide evidence that a subset of GABAergic interneurons requires dystroglycan for formation and maintenance of axonal terminals on pyramidal cells. As such, dystroglycan is the first postsynaptic GABAergic protein for which an interneuron terminal-specific function could be shown. Our findings also offer a new perspective on the mechanisms that lead to intellectual disability in muscular dystrophies without associated brain malformations.


Assuntos
Colecistocinina/metabolismo , Distroglicanas/fisiologia , Terminações Pré-Sinápticas/fisiologia , Células Piramidais/fisiologia , Animais , Proteínas de Ligação ao Cálcio , Carbacol/farmacologia , Distroglicanas/genética , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Técnicas de Introdução de Genes , Interneurônios/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Agonistas Muscarínicos/farmacologia , Moléculas de Adesão de Célula Nervosa/metabolismo , Sistema Nervoso Parassimpático/efeitos dos fármacos , Sistema Nervoso Parassimpático/fisiologia , Ácido gama-Aminobutírico/fisiologia
18.
Eur J Neurosci ; 44(5): 2258-71, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27364953

RESUMO

Alterations of neuronal activity due to changes in GABAA receptors (GABAA R) mediating tonic inhibition influence different hippocampal functions. Gabra5-null mice and α5 subunit((H105R)) knock-in mice exhibit signs of hippocampal dysfunction, but are capable of improved performance in several learning and memory tasks. Accordingly, alleviating abnormal GABAergic tonic inhibition in the hippocampal formation by selective α5-GABAA R modulators represents a possible therapeutic approach for several intellectual deficit disorders. Adult neurogenesis in the dentate gyrus is an important facet of hippocampal plasticity; it is regulated by tonic GABAergic transmission, as shown by deficits in proliferation, migration and dendritic development of adult-born neurons in Gabra4-null mice. Here, we investigated the contribution of α5-GABAA Rs to granule cell development, using retroviral vectors expressing eGFP for labeling precursor cells in the subgranular zone. Global α5-GABAA R knockout (α5-KO) mice showed no alterations in migration and morphological development of eGFP-positive granule cells. However, upregulation of α1 subunit-immunoreactivity was observed in the hippocampal formation and cerebral cortex. In contrast, partial gene inactivation in α5-heterozygous (α5-het) mice, as well as single-cell deletion of Gabra5 in newborn granule cells from α5-floxed mice, caused severe alterations of migration and dendrite development. In α5-het mice, retrovirally mediated overexpression of Cdk5 resulted in normal migration and dendritic branching, suggesting that Cdk5 cooperates with α5-GABAA Rs to regulate neuronal development. These results show that minor imbalance of α5-GABAA R-mediated transmission may have major consequences for neuronal plasticity; and call for caution upon chronic therapeutic use of negative allosteric modulators acting at these receptors.


Assuntos
Giro Denteado/citologia , Neurogênese , Neurônios/citologia , Receptores de GABA-A/metabolismo , Animais , Movimento Celular , Quinase 5 Dependente de Ciclina/genética , Quinase 5 Dependente de Ciclina/metabolismo , Espinhas Dendríticas/metabolismo , Giro Denteado/embriologia , Giro Denteado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Receptores de GABA-A/genética
19.
Elife ; 5: e14120, 2016 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-26971710

RESUMO

Recent findings indicate a high level of specialization at the level of microcircuits and cell populations within brain structures with regards to the control of fear and anxiety. The hippocampus, however, has been treated as a unitary structure in anxiety and fear research despite mounting evidence that different hippocampal subregions have specialized roles in other cognitive domains. Using novel cell-type- and region-specific conditional knockouts of the GABAA receptor α2 subunit, we demonstrate that inhibition of the principal neurons of the dentate gyrus or CA3 via α2-containing GABAA receptors (α2GABAARs) is required to suppress anxiety, while the inhibition of CA1 pyramidal neurons is required to suppress fear responses. We further show that the diazepam-modulation of hippocampal theta activity shows certain parallels with our behavioral findings, suggesting a possible mechanism for the observed behavioral effects. Thus, our findings demonstrate a double dissociation in the regulation of anxiety versus fear by hippocampal microcircuitry.


Assuntos
Ansiedade , Mapeamento Encefálico , Medo , Hipocampo/fisiologia , Animais , Camundongos , Rede Nervosa , Receptores de GABA-A/genética
20.
Neuropharmacology ; 105: 594-606, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26930443

RESUMO

Excessive stimulation of NMDA receptors with glutamate or other potent agonists such as NMDA leads to excitotoxicity and neural injury. In this study, we aimed to provide insight into an animal model of brain excitotoxic damage; single unilateral infusion of NMDA at mild dose into the hippocampal formation. NMDA infusion induced chronic, focal neurodegeneration in the proximity of the injection site. The lesion was accompanied by severe and progressive neuroinflammation and affected preferentially principal neurons while sparing GABAergic interneurons. Furthermore, the unilateral lesion did not cause significant impairment of spatial learning abilities. Finally, GluN1 and GluN2B subunits of NMDA receptor were significantly upregulated up to 3 days after the NMDA infusion, while GABAA α5 subunit was downregulated at 30 days after the lesion. Taken together, a single infusion of NMDA into the hippocampal formation represents an animal model of excitotoxicity-induced chronic neurodegeneration of principal neurons accompanied by severe neuroinflammation and subunit specific changes in NMDA and GABAA receptors.


Assuntos
Hipocampo/metabolismo , N-Metilaspartato/toxicidade , Doenças Neurodegenerativas/metabolismo , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Modelos Animais de Doenças , Lateralidade Funcional , Hipocampo/diagnóstico por imagem , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , N-Metilaspartato/administração & dosagem , Degeneração Neural/diagnóstico por imagem , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Doenças Neurodegenerativas/diagnóstico por imagem , Doenças Neurodegenerativas/patologia , Neuroimunomodulação/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Ratos Long-Evans
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...