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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Physiol ; 600(10): 2401-2427, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35365894

RESUMO

Slow oscillations, the hallmark of non-REM sleep, and their cellular counterpart, Up and Down states (UDSs), are considered a signature of cortical dynamics that reflect the intrinsic network organization. Although previous studies have explored the role of inhibition in regulating UDSs, little is known about whether this role changes with maturation. This is surprising since both slow oscillations and UDSs exhibit significant age-dependent alterations. To elucidate the developmental impact of GABAB and GABAA receptors on UDS activity, we conducted simultaneous local field potentials and intracellular recordings ex vivo, in brain slices of young and adult male mice, using selective blockers, CGP55845 and a non-saturating concentration of gabazine, respectively. Blockade of both GABAB and GABAA signalling showed age-differentiated functions. CGP55845 caused an increase in Down state duration in young animals, but a decrease in adults. Gabazine evoked spike and wave discharges in both ages; however, while young networks became completely epileptic, adults maintained the ability to generate UDSs. Furthermore, voltage clamp recordings of miniature inhibitory postsynaptic currents revealed that gabazine selectively blocks phasic currents, particularly involving postsynaptic mechanisms. The latter exhibit clear maturational changes, suggesting a different subunit composition of GABAA receptors in young vs. adult animals. Indeed, subsequent local field potential recordings under diazepam (nanomolar or micromolar concentrations) revealed that mechanisms engaging the drug's classical binding site, mediated by α1-subunit-containing GABAA receptors, make a bigger contribution to Up state initiation in young networks compared to adults. Taken together, these findings help clarify the mechanisms that underlie the maturation of cortical network activity and enhance our understanding regarding the emergence of neurodevelopmental disorders. KEY POINTS: Slow oscillations, the EEG hallmark of non-REM sleep, and their cellular counterpart, Up and Down states (UDSs), are considered the default activity of the cerebral cortex and reflect the underlying neural connectivity. GABAB - and GABAA -receptor-mediated inhibition play a major role in regulating UDS activity. Although slow oscillations and UDSs exhibit significant alterations as a function of age, it is unknown how developmental changes in inhibition contribute to the developmental profile of this activity. In this study, we reveal for the first time age-dependent effects of GABAB and GABAA signalling on UDSs. We also document the differential subunit composition of postsynaptic GABAA receptors in young and adult animals, highlighting the α1-subunit as a major component of the age-differentiated regulation of UDSs. These findings help clarify the mechanisms that underlie the maturation of cortical network activity, and enhance our understanding regarding the emergence of neurodevelopmental disorders.


Assuntos
Potenciais Pós-Sinápticos Inibidores , Receptores de GABA-A , Animais , Córtex Cerebral/fisiologia , Diazepam/farmacologia , Potenciais Pós-Sinápticos Inibidores/fisiologia , Masculino , Camundongos , Receptores de GABA-A/metabolismo , Receptores de GABA-B/metabolismo , Ácido gama-Aminobutírico
2.
Med Mol Morphol ; 55(3): 174-186, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35461467

RESUMO

Cleidocranial dysplasia (CCD) is a hereditary disorder associated with skeletal dysplasia and dental abnormalities. CCD arises from heterozygous loss of function mutations in the Runt-related transcription factor 2 (RUNX2) gene. Osteoporosis is often observed in CCD patients and conventional vitamin D supplementation is recommended. However, sufficient evidences have not been presented yet. This study investigated the role of RUNX2 in osteoblastic differentiation and sought to identify potential target genes for the treatment of osteoporosis associated with CCD, using induced pluripotent stem cell (iPSC) technology. We successfully established Runx2-/-, Runx2+/- and wild-type miPSCs from litter-matched mice and found poor Vdr expression in Runx2-/-cells. Significant down-regulation of osteoblastic differentiation in Runx2-/- miPSCs was observed. Gene expression array revealed unexpected results such as remarkable increase of Rankl expression and decrease of Vdr in Runx2-/- cells. Insufficient response to vitamin D in Runx2-/- cells was also observed. Our results suggest that RUNX2 functions as a regulator of Rankl and Vdr and thereby controls bone density. These findings also suggest that conventional vitamin D supplementation may not be as effective as previously expected, in the treatment of osteoporosis associated with CCD, and that inhibiting RANKL function might be worth considering as an alternative treatment strategy.


Assuntos
Displasia Cleidocraniana , Subunidade alfa 1 de Fator de Ligação ao Core , Células-Tronco Pluripotentes Induzidas , Osteoporose , Vitamina D , Animais , Diferenciação Celular , Displasia Cleidocraniana/genética , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Camundongos , Camundongos Knockout , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Osteoporose/genética , Vitamina D/farmacologia
3.
J Neurophysiol ; 123(1): 178-190, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31693436

RESUMO

In this study we explored whether learning leads to enduring changes in inhibitory synaptic transmission in lateral amygdala (LA). We revealed that olfactory discrimination (OD) learning in rats led to a long-lasting increase in postsynaptic GABAA channel-mediated miniature inhibitory postsynaptic currents (mIPSCs) in LA. Olfactory fear conditioning, but not auditory fear conditioning, also led to enduring enhancement in GABAA-mediated mIPSCs. Auditory fear conditioning, but not olfactory fear conditioning or OD learning, induced an enduring reduction in the frequency but not the current of mIPSC events. We found that p21-activated kinase (PAK) activity is needed to maintain OD and olfactory fear conditioning learning-induced enduring enhancement of mIPSCs. Further analysis revealed that OD led to an increase in GABAA channel conductance whereas olfactory fear conditioning increased the number of GABAA channels. These alterations in GABAA channels conductance and level are controlled by PAK activity. Our study shows that the learning-induced increase in postsynaptic inhibitory transmission in LA is specific to the sensory modality. However, the mechanism that mediates the increase in inhibitory transmission, namely the increase in the conductance or in the level of GABAA channel, is learning specific.NEW & NOTEWORTHY Here we studied whether learning leads to long-lasting alterations in inhibitory synaptic transmission in lateral amygdala (LA). We revealed that learning led to enduring changes in inhibitory synaptic transmission in LA that are affected by the sensory modality (auditory or olfaction) used during learning. However, the mechanism that mediated the changes in inhibitory transmission (alterations in GABAA channel level or conductance) depended on the type of learning. These long-lasting alterations are maintained by p21-activated kinase.


Assuntos
Percepção Auditiva/fisiologia , Complexo Nuclear Basolateral da Amígdala/fisiologia , Condicionamento Clássico/fisiologia , Aprendizagem por Discriminação/fisiologia , Potenciais Pós-Sinápticos Inibidores/fisiologia , Percepção Olfatória/fisiologia , Receptores de GABA-A/metabolismo , Quinases Ativadas por p21/metabolismo , Animais , Comportamento Animal/fisiologia , Medo/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley
4.
Proc Natl Acad Sci U S A ; 110(48): 19603-8, 2013 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-24218551

RESUMO

ErbB4 signaling in the central nervous system is implicated in neuropsychiatric disorders and epilepsy. In cortical tissue, ErbB4 associates with excitatory synapses located on inhibitory interneurons. However, biochemical and histological data described herein demonstrate that the vast majority of ErbB4 is extrasynaptic and detergent-soluble. To explore the function of this receptor population, we used unbiased proteomics, in combination with electrophysiological, biochemical, and cell biological techniques, to identify a clinically relevant ErbB4-interacting protein, the GABAA receptor α1 subunit (GABAR α1). We show that ErbB4 and GABAR α1 are robustly coexpressed in hippocampal interneurons, and that ErbB4-null mice have diminished cortical GABAR α1 expression. Moreover, we characterize a Neuregulin-mediated ErbB4 signaling modality, independent of receptor tyrosine kinase activity, that couples ErbB4 to decreased postsynaptic GABAR currents on inhibitory interneurons. Consistent with an evolving understanding of GABAR trafficking, this pathway requires both clathrin-mediated endocytosis and protein kinase C to reduce GABAR inhibitory currents, surface GABAR α1 expression, and colocalization with the inhibitory postsynaptic protein gephyrin. Our results reveal a function of ErbB4, independent of its tyrosine kinase activity, that modulates postsynaptic inhibitory control of hippocampal interneurons and may provide a novel pharmacological target in the treatment of neuropsychiatric disorders and epilepsy.


Assuntos
Receptores ErbB/metabolismo , Hipocampo/citologia , Interneurônios/metabolismo , Neurregulinas/metabolismo , Receptores de GABA-A/metabolismo , Transdução de Sinais/fisiologia , Sinapses/metabolismo , Animais , Hipocampo/metabolismo , Imuno-Histoquímica , Imunoprecipitação , Espectrometria de Massas , Microscopia Confocal , Técnicas de Patch-Clamp , Proteômica , Ratos , Ratos Sprague-Dawley , Receptor ErbB-4
5.
Brain Behav Immun ; 33: 102-11, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23770090

RESUMO

Anxiety-like responses to stress are accompanied by elevation of brain cytokine-mRNAs. Because cytokines microinjected into central-amygdala (CeA) substitute for stress in a behavioral paradigm, the possibility was raised that cytokines increased by stress influence behavior by affecting CeA-neural activity. Previously, cytokines increased firing-rate of CeA-neurons comparable to that induced by corticotropin-releasing factor (CRF). In this investigation, tumor-necrosis-factor-α (TNFα) increased amplitude, but not frequency of mEPSCs from CeA-neurons. Additionally, TNFα decreased the threshold for triggering action potentials from CeA-neurons without altering membrane-properties during current-clamp recording. Glutamate-receptor-antagonist blockade of mEPSCs and the TNFα-induced reduction in firing threshold implicated glutamate in these changes. A phosphatidyl-inositol-3-kinase-antagonist prevented the TNFα-induced increased in amplitude of mEPSCs, documenting a TNFα intracellular influence. Additionally, TNFα increased frequency, but not amplitude of mIPSCs. CRF-receptor-antagonists were found to prevent the TNFα-induced increase in mIPSC-frequency, without altering the TNFα-induced amplitude increase in mEPSCs or the reduced threshold for action-potentials by TNFα. To clarify how TNFα was increasing CRF-release in the presence of tetrodotoxin, the possibility tested was whether preventing glial-activation would prevent this elevated mIPSC-frequency blocked by CRF-receptor antagonists. Minocycline, which blocks glial activation, prevented the TNFα-induced increase in mIPSC-frequency - a finding consistent with glia contributing to the CRF-involvement in this TNFα action. To fully understand the means by which a CRF1-receptor-antagonist and minocycline prevent TNFα from increasing mIPSC-frequency will require further clarification. Nonetheless, these data provide convincing evidence that release of TNFα by stress could alter neural activity of CeA-neurons by influencing GABA-and glutamate function.


Assuntos
Tonsila do Cerebelo/imunologia , Potenciais Pós-Sinápticos Excitadores/imunologia , Células-Tronco Pluripotentes Induzidas/imunologia , Inibição Neural/imunologia , Neurônios/imunologia , Estresse Fisiológico/imunologia , Transmissão Sináptica/imunologia , Fator de Necrose Tumoral alfa/fisiologia , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/metabolismo , Animais , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo
6.
Neurosci Lett ; 756: 135950, 2021 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-33979698

RESUMO

The mechanisms of general anaesthetics such as propofol have drawn substantial attention. The effects of propofol on inhibitory postsynaptic currents are not exactly the same in different brain nuclei. Recent studies revealed that the paraventricular thalamic nucleus (PVT) is a critical nucleus modulating wakefulness. However, the effects of propofol on PVT neurons and the mechanisms underlying such effects remain unknown. Here, we performed the whole-cell recording of the PVT neurons in acute brain slices and bath application of propofol. We found that propofol hyperpolarized the membrane potentials of the PVT neurons and suppressed the action potentials induced by step-current injection. Propofol did not affect the spontaneous inhibitory postsynaptic currents (sIPSCs) amplitude or frequency, but prolonged the sIPSCs half-width. Besides, propofol increased miniature inhibitory synaptic currents (mIPSCs) frequency and half-width. Furthermore, propofol could induce GABAA receptors-mediated tonic inhibitory currents dose-dependently. Thus, our results demonstrate that propofol hyperpolarizes PVT neurons by modulating inhibitory currents via GABAA receptors in mice.


Assuntos
Anestésicos Intravenosos/farmacologia , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Núcleos da Linha Média do Tálamo/efeitos dos fármacos , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Propofol/farmacologia , Animais , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Técnicas de Patch-Clamp
7.
CNS Neurosci Ther ; 27(10): 1157-1172, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34111331

RESUMO

AIM: This study aimed to investigate the regulation of pain hypersensitivity induced by the spinal synaptic transmission mechanisms underlying interleukin (IL)-10 and glucagon-like peptide 1 receptor (GLP-1R) agonist exenatide-induced pain anti-hypersensitivity in neuropathic rats through spinal nerve ligations. METHODS: Neuropathic pain model was established by spinal nerve ligation of L5/L6 and verified by electrophysiological recording and immunofluorescence staining. Microglial expression of ß-endorphin through autocrine IL-10- and exenatide-induced inhibition of glutamatergic transmission were performed by behavioral tests coupled with whole-cell recording of miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs) through application of endogenous and exogenous IL-10 and ß-endorphin. RESULTS: Intrathecal injections of IL-10, exenatide, and the µ-opioid receptor (MOR) agonists ß-endorphin and DAMGO inhibited thermal hyperalgesia and mechanical allodynia in neuropathic rats. Whole-cell recordings of bath application of exenatide, IL-10, and ß-endorphin showed similarly suppressed enhanced frequency and amplitude of the mEPSCs in the spinal dorsal horn neurons of laminae II, but did not reduce the frequency and amplitude of mIPSCs in neuropathic rats. The inhibitory effects of IL-10 and exenatide on pain hypersensitive behaviors and spinal synaptic plasticity were totally blocked by pretreatment of IL-10 antibody, ß-endorphin antiserum, and MOR antagonist CTAP. In addition, the microglial metabolic inhibitor minocycline blocked the inhibitory effects of IL-10 and exenatide but not ß-endorphin on spinal synaptic plasticity. CONCLUSION: This suggests that spinal microglial expression of ß-endorphin mediates IL-10- and exenatide-induced inhibition of glutamatergic transmission and pain hypersensitivity via presynaptic and postsynaptic MORs in spinal dorsal horn.


Assuntos
Exenatida/farmacologia , Interleucina-10 , Microglia , Neuralgia/fisiopatologia , Plasticidade Neuronal/efeitos dos fármacos , Nervos Espinhais/fisiopatologia , beta-Endorfina/fisiologia , Analgésicos Opioides/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Potenciais Pós-Sinápticos Excitadores , Ácido Glutâmico , Injeções Espinhais , Interleucina-10/metabolismo , Interleucina-10/farmacologia , Neuralgia/psicologia , Técnicas de Patch-Clamp , Ratos , Receptores Opioides mu/agonistas , Transdução de Sinais , Transmissão Sináptica , beta-Endorfina/farmacologia
8.
Neuropharmacology ; 187: 108492, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33582153

RESUMO

In this work, modulation by orexin-A of the release of glutamate and GABA from bipolar and amacrine cells respectively was studied by examining the effects of the neuropeptide on miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs) of rat retinal ganglion cells (GCs). Using RNAscope in situ hybridization in combination with immunohistochemistry, we showed positive signals for orexin receptor-1 (OX1R) mRNA in the bipolar cell terminals and those for orexin receptor-2 (OX2R) mRNA in the amacrine cell terminals. With whole-cell patch-clamp recordings in rat retinal slices, we demonstrated that application of orexin-A reduced the interevent interval of mEPSCs of GCs through OX1R. However, it increased the interevent interval of mIPSCs, mediated by GABAA receptors, through OX2R. Furthermore, orexin-A-induced reduction of mEPSC interevent interval was abolished by the application of PI-PLC inhibitors or PKC inhibitors. In contrast, orexin-A-induced increase of GABAergic mIPSC interevent interval was mimicked by 8-Br-cAMP or an adenylyl cyclase activator, but was eliminated by PKA antagonists. Finally, application of nimodipine, an L-type Ca2+ channel blocker, increased both mEPSC and mIPSC interevent interval, and co-application of orexin-A no longer changed the mEPSCs and mIPSCs. We conclude that orexin-A increases presynaptic glutamate release onto GCs by activating L-type Ca2+ channels in bipolar cells, a process that is mediated by an OX1R/PI-PLC/PKC signaling pathway. However, orexin-A decreases presynaptic GABA release onto GCs by inhibiting L-type Ca2+ channels in amacrine cells, a process that is mediated by an OX2R/cAMP-PKA signaling pathway.


Assuntos
Células Amácrinas/metabolismo , Potenciais Pós-Sinápticos Excitadores/genética , Potenciais Pós-Sinápticos Inibidores/genética , Receptores de Orexina/genética , Orexinas/metabolismo , Células Bipolares da Retina/metabolismo , Células Ganglionares da Retina/metabolismo , Transmissão Sináptica/genética , Células Amácrinas/efeitos dos fármacos , Animais , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Ácido Glutâmico/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Receptores de Orexina/metabolismo , Orexinas/farmacologia , Técnicas de Patch-Clamp , Fosfoinositídeo Fosfolipase C/antagonistas & inibidores , Fosfoinositídeo Fosfolipase C/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Ratos , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Células Bipolares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo
9.
Brain Res ; 1712: 82-92, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30735639

RESUMO

Aerobic exercise lowers blood pressure in patients with hypertension, but the underlying mechanisms remain incompletely understood. The hypothalamic paraventricular nucleus (PVN) plays a key role in the control of sympathetic outflow and cardiovascular tone. We examined whether chronic aerobic exercise altered synaptic transmission and reactive oxygen species (ROS) production in the PVN. In the present study, spontaneously hypertensive rats (SHRs) were subjected to exercise training for 8 weeks, five times per week, with Wistar Kyoto (WKY) rats as the cohort control. Miniature excitatory and inhibitory postsynaptic currents (mEPSCs and mIPSCs) were recorded from the PVN in ex vivo hypothalamic slice preparations obtained after the last training, and biomarkers of oxidative stress and physical indexes were observed. The mean frequency and amplitude, as well as the rise time and the decay time constant of mIPSCs, significantly decreased in 20-wk-old SHRs compared to WKY 20-wk-old controls. In contrast to mIPSCs, only the mean mEPSC frequency was higher, and there were no other changes in mEPSCs in comparison to the control group. SHRs exhibited higher ROS, 8-OHdG, and MDA; and lower SOD1, SOD2, CAT, Ogg1, and SOD and CAT activity in the PVN. These SHRs also had a significant increase in heart rate, blood pressure and sympathetic nerve activity, and higher levels of norepinephrine (NE). Exercise training ameliorated all these abnormalities, resulting in an increase in the mean frequency, amplitude and kinetics of mIPSCs, accompanied by a decrease in the mean frequency of mEPSCs in the PVN. This study demonstrates that moderate intensity, high frequency exercise training induces a selective enhancement of inhibitory synaptic transmission in the PVN, which may dampen sympathetic activity and reduce blood pressure in hypertension. These changes may be due to antioxidant-related adaptations in the PVNs of SHRs.


Assuntos
Núcleo Hipotalâmico Paraventricular/metabolismo , Condicionamento Físico Animal/fisiologia , Transmissão Sináptica/fisiologia , Animais , Pressão Sanguínea , Potenciais Pós-Sinápticos Excitadores/fisiologia , Frequência Cardíaca , Hipertensão/fisiopatologia , Hipotálamo/metabolismo , Masculino , Neurônios/metabolismo , Condicionamento Físico Animal/métodos , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Espécies Reativas de Oxigênio/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sistema Nervoso Simpático/metabolismo
10.
In Vitro Cell Dev Biol Anim ; 55(2): 130-137, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30659476

RESUMO

Recent studies have successfully generated tooth-like structure by mimicking the reciprocal interaction between dental epithelial and mesenchymal cells in tooth organogenesis. However, clinical applications of these methods are limited primarily due to the lack of appropriate sources for dental epithelial cells. Induced pluripotent stem cells (iPSCs) are attractive as a source for dental epithelial cells due to their unique characteristics. In this study, we examined the effect of neurotrophin-4 (NT-4) on the differentiation of mouse iPSCs (miPSCs) into dental epithelial cells. Our results showed that the addition of NT-4 during the formation of embryoid body significantly triggered the upregulation of epithelial markers such as p63 and CK14, suggesting that NT-4 provides an inductive condition for the differentiation of miPSCs into epithelial cells. Expansion of the NT-4-treated cells under serum-free culture conditions improves the formation of cells with cobblestone-like morphology and significantly downregulated the expression of pluripotent and ectodermal markers. Phenotypic analysis revealed that a dental epithelial surface marker, CD49f, was highly expressed on these cells. Formation of miPSCs-derived dental epithelial-like cells was further confirmed by the expression of ameloblast-specific markers. These results suggest that the addition of NT-4 during the formation of embryoid body together with the serum-free culture condition promoted the differentiation of miPSCs into dental epithelial-like cells.


Assuntos
Diferenciação Celular , Células Epiteliais/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Soro/metabolismo , Dente/citologia , Animais , Biomarcadores/metabolismo , Células Cultivadas , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Fenótipo
11.
Neurosci Lett ; 709: 134377, 2019 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-31352043

RESUMO

The song control system in the brain of songbirds is important for the production and acquisition of song and exhibits some of the largest neural sex differences observed in vertebrates. The robust nucleus of the arcopallium (RA) is a premotor nucleus, playing a key role in controlling singing. RA projection neurons (PNs) receives denser synapse inputs including excitatory in males than in females. However, the inhibitory synaptic transmission in the RA has not been reported. In the present study, using whole-cell voltage-clamp recording, spontaneous inhibitory postsynaptic currents (sIPSCs) and miniature inhibitory postsynaptic currents (mIPSCs) of the males and females were recorded. The average frequency and amplitude of sIPSCs/mIPSCs in males were higher than females. These results demonstrate the sexually dimorphic of the inhibitory synaptic transmission in the RA PNs and the RA PNs in males receive more inhibitory synaptic transmission. These findings contribute to further illuminate the neural mechanisms under the sexually dimorphism song production of adult zebra finches.


Assuntos
Encéfalo/fisiologia , Potenciais Pós-Sinápticos Inibidores/fisiologia , Inibição Neural/fisiologia , Caracteres Sexuais , Transmissão Sináptica/fisiologia , Vocalização Animal/fisiologia , Animais , Feminino , Tentilhões , Masculino , Aves Canoras
12.
Curr Biol ; 29(15): 2455-2464.e5, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31327715

RESUMO

The retrieval of odor-related memories shapes animal behavior. The anterior piriform cortex (aPC) is the largest part of the olfactory cortex, and it plays important roles in olfactory processing and memory. However, it is still unclear whether specific cellular mechanisms in the aPC control olfactory memory, depending on the appetitive or aversive nature of the stimuli involved. Cannabinoid-type 1 (CB1) receptors are present in the aPC (aPC-CB1), but their potential impact on olfactory memory was never explored. Here, we used a combination of behavioral, genetic, anatomical, and electrophysiological approaches to characterize the functions of aPC-CB1 receptors in the regulation of appetitive and aversive olfactory memory. Pharmacological blockade or genetic deletion of aPC-CB1 receptors specifically impaired the retrieval of conditioned odor preference (COP). Interestingly, expression of conditioned odor aversion (COA) was unaffected by local CB1 receptor blockade, indicating that the role of aPC endocannabinoid signaling is selective for retrieval of appetitive memory. Anatomical investigations revealed that CB1 receptors are highly expressed on aPC GABAergic interneurons, and ex vivo electrophysiological recordings showed that their pharmacological activation reduces miniature inhibitory post-synaptic currents (mIPSCs) onto aPC semilunar (SL), but not pyramidal principal neurons. COP retrieval, but not COA, was associated with a specific CB1-receptor-dependent decrease of mIPSCs in SL cells. Altogether, these data indicate that aPC-CB1 receptor-dependent mechanisms physiologically control the retrieval of olfactory memory, depending on odor valence and engaging modulation of local inhibitory transmission.


Assuntos
Memória , Percepção Olfatória , Córtex Piriforme/fisiologia , Receptor CB1 de Canabinoide/genética , Olfato , Animais , Masculino , Camundongos , Odorantes , Receptor CB1 de Canabinoide/metabolismo
13.
Mol Neurobiol ; 55(5): 4267-4279, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28623616

RESUMO

Differentiation of pluripotent stem cells (PSCs) to neural lineages has gathered huge attention in both basic research and regenerative medicine. The major hurdle lies in the efficiency of differentiation and identification of small molecules that facilitate neurogenesis would partly circumvent this limitation. The small molecule Cyclosporine A (CsA), a commonly used immunosuppressive drug, has been shown to enhance in vivo neurogenesis. To extend the information to in vitro neurogenesis, we examined the effect of CsA on neural differentiation of PSCs. We found CsA to increase the expression of neural progenitor genes during early neural differentiation. Gene silencing approach revealed CsA-mediated neural induction to be dependent on blocking the Ca2+-activated phosphatase calcineurin (Cn) signaling. Similar observation with FK506, an independent inhibitor of Cn, further strengthened the necessity of blocking Cn for enhanced neurogenesis. Surprisingly, mechanistic insight revealed Cn-inhibition dependent upregulation of IL-6 protein to be necessary for CsA-mediated neurogenesis. Together, these findings provide a comprehensive understanding of the role of CsA in neurogenesis, thus suggesting a method for obtaining large numbers of neural progenitors from PSCs for possible transplantation.


Assuntos
Ciclosporina/farmacologia , Interleucina-6/metabolismo , Neurônios/citologia , Células-Tronco Pluripotentes/metabolismo , Animais , Calcineurina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Corpos Embrioides/citologia , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/metabolismo , Fatores de Transcrição NFATC/metabolismo , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Transdução de Sinais
14.
Neuropharmacology ; 138: 170-181, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29894770

RESUMO

miRNA-137 is an extremely abundant miRNA in the central nervous system and is thought to be closely related to synaptic plasticity. Here, we report a previously unrecognized role of miRNA-137 in epilepsy. The expression of miRNA-137 was decreased both in patients with temporal lobe epilepsy (TLE) and in two different mouse models of epilepsy. Overexpression of miRNA-137 induced by an intrahippocampal injection of a specific agomir prolonged the latency to spontaneous recurrent seizures (SRSs) and reduced seizure severity in a mouse model of pilocarpine-induced epilepsy. Elevated levels of miRNA-137 also prolonged the latency to full kindling and reduced the seizure severity in a mouse model of pentylenetetrazol (PTZ)-kindled epilepsy. Suppression of miRNA-137 levels decreased the latency to the first SRS or the latency to full kindling and increased the seizure severity in both epileptic mouse models. Whole-cell patch-clamp recordings showed that overexpression of miRNA-137 reduced the excitability of pyramidal neurons in the hippocampal CA3a region in a Mg2+-free-induced brain slice model of epileptiform activity. This effect may have been achieved by the regulation of the frequency of miniature inhibitory postsynaptic currents (mIPSCs) and presynaptic inhibitory neurotransmitter release. These results suggest that elevated levels of miRNA-137 may exert an antiepileptic effect via a presynaptic neurotransmission mechanism. These data may provide a new potential target and therapeutic strategy for treating epilepsy in the future.


Assuntos
Encéfalo/metabolismo , Epilepsia/metabolismo , MicroRNAs/metabolismo , Convulsões/metabolismo , Adolescente , Adulto , Animais , Modelos Animais de Doenças , Epilepsia/terapia , Feminino , Humanos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Deficiência de Magnésio , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Pentilenotetrazol , Terminações Pré-Sinápticas/metabolismo , Células Piramidais/metabolismo , Distribuição Aleatória , Convulsões/terapia , Técnicas de Cultura de Tecidos , Adulto Jovem
15.
Front Cell Neurosci ; 12: 46, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29541022

RESUMO

Patients with autism spectrum disorder (ASD) display abnormalities in neuronal development, synaptic function and neural circuits. The imbalance of excitatory and inhibitory (E/I) synaptic transmission has been proposed to cause the main behavioral characteristics of ASD. Repetitive transcranial magnetic stimulation (rTMS) can directly or indirectly induce excitability and synaptic plasticity changes in the brain noninvasively. However, whether rTMS can ameliorate autistic-like behaviors in animal model via regulating the balance of E/I synaptic transmission is unknown. By using our recent reported animal model with autistic-like behaviors induced by neonatal isolation (postnatal days 1-9), we found that low-frequency rTMS (LF-rTMS, 1 Hz) treatment for 2 weeks effectively alleviated the acquired autistic-like symptoms, as reflected by an increase in social interaction and decrease in self-grooming, anxiety- and depressive-like behaviors in young adult rats compared to those in untreated animals. Furthermore, the amelioration in autistic-like behavior was accompanied by a restoration of the balance between E/I activity, especially at the level of synaptic transmission and receptors in synaptosomes. These findings indicated that LF-rTMS may alleviate the symptoms of ASD-like behaviors caused by neonatal isolation through regulating the synaptic GABA transmission, suggesting that LF-rTMS may be a potential therapeutic technique to treat ASD.

16.
Brain Struct Funct ; 221(1): 301-16, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25273281

RESUMO

In the inner retina, ganglion cells (RGCs) integrate and process excitatory signal from bipolar cells (BCs) and inhibitory signal from amacrine cells (ACs). Using multiple labeling immunohistochemistry, we first revealed the expression of the cannabinoid CB1 receptor (CB1R) at the terminals of ACs and BCs in rat retina. By patch-clamp techniques, we then showed how the activation of this receptor dichotomously regulated miniature inhibitory postsynaptic currents (mIPSCs), mediated by GABAA receptors and glycine receptors, and miniature excitatory postsynaptic currents (mEPSCs), mediated by AMPA receptors, of RGCs in rat retinal slices. WIN55212-2 (WIN), a CB1R agonist, reduced the mIPSC frequency due to an inhibition of L-type Ca(2+) channels no matter whether AMPA receptors were blocked. In contrast, WIN reduced the mEPSC frequency by suppressing T-type Ca(2+) channels only when inhibitory inputs to RGCs were present, which could be in part due to less T-type Ca(2+) channels of cone BCs, presynaptic to RGCs, being in an inactivation state under such condition. This unique feature of CB1R-mediated retrograde regulation provides a novel mechanism for modulating excitatory synaptic transmission in the inner retina. Moreover, depolarization of RGCs suppressed mIPSCs of these cells, an effect that was eliminated by the CB1R antagonist SR141716, suggesting that endocannabinoid is indeed released from RGCs.


Assuntos
Células Amácrinas/metabolismo , Potenciais Pós-Sinápticos Excitadores , Potenciais Pós-Sinápticos Inibidores , Receptor CB1 de Canabinoide/metabolismo , Receptor CB1 de Canabinoide/fisiologia , Células Bipolares da Retina/metabolismo , Células Ganglionares da Retina/fisiologia , Animais , Benzoxazinas/farmacologia , Canais de Cálcio Tipo L/fisiologia , Canais de Cálcio Tipo T/fisiologia , Masculino , Potenciais Pós-Sinápticos em Miniatura , Morfolinas/farmacologia , Naftalenos/farmacologia , Piperidinas/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptores de AMPA/fisiologia , Receptores de GABA-A/fisiologia , Rimonabanto , Transdução de Sinais
17.
Neurosci Lett ; 592: 64-9, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25711796

RESUMO

Previous studies have indicated that mu-opioid receptors in the ventrolateral orbital cortex (VLO) are involved in antinociception in tail flick tests and GABAergic neurons or terminals express mu-opioid receptors in the VLO. The current study examined the effect of selective mu-opioid receptor agonist DAMGO on the GABAergic miniature inhibitory postsynaptic currents (mIPSCs) in the VLO in rats using the whole-cell patch clamp. The results demonstrated that 5 µM DAMGO application into the rat VLO slices significantly reduced the GABAergic mIPSCs frequency, without any effect on its amplitude, and this effect of DAMGO was reversed by pretreatment with selective mu-opioid receptor antagonist 1 µM CTOP. Importantly, application of CTOP alone into the VLO slices did not produce any effect on the frequency and amplitude of GABAergic mIPSCs. These results indicate a presynaptic effect of mu-opioid receptor activation on the GABAergic neurons in the VLO. The current data suggests that a presynaptic inhibition of the GABA release may contribute to the mu-opioid receptor mediated effects in the VLO and provides novel electrophysiological evidence for the underlying mechanisms of mu-opioid receptors in the VLO.


Assuntos
Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos em Miniatura/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Receptores Opioides mu/agonistas , Ácido gama-Aminobutírico/fisiologia , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Técnicas In Vitro , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Córtex Pré-Frontal/fisiologia , Ratos Sprague-Dawley , Receptores Opioides mu/antagonistas & inibidores , Somatostatina/análogos & derivados , Somatostatina/farmacologia
18.
Neuroscience ; 258: 62-73, 2014 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-24231737

RESUMO

Dorsal raphe nucleus (DRN) serotonin (5-HT) neurons play an important role in feeding, mood control and stress responses. One important feature of their activity across the sleep-wake cycle is their reduced firing during rapid-eye-movement (REM) sleep which stands in stark contrast to the wake/REM-on discharge pattern of brainstem cholinergic neurons. A prominent model of REM sleep control posits a reciprocal interaction between these cell groups. 5-HT inhibits cholinergic neurons, and activation of nicotinic receptors can excite DRN 5-HT neurons but the cholinergic effect on inhibitory inputs is incompletely understood. Here, in vitro, in DRN brain slices prepared from GAD67-GFP knock-in mice, a brief (3 min) bath application of carbachol (50 µM) increased the frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) in GFP-negative, putative 5-HT neurons but did not affect miniature (tetrodotoxin-insensitive) IPSCs. Carbachol had no direct postsynaptic effect. Thus, carbachol likely increases the activity of local GABAergic neurons which synapse on 5-HT neurons. Removal of dorsal regions of the slice including the ventrolateral periaqueductal gray (vlPAG) region where GABAergic neurons projecting to the DRN have been identified, abolished the effect of carbachol on sIPSCs whereas the removal of ventral regions containing the oral region of the pontine reticular nucleus (PnO) did not. In addition, carbachol directly excited GFP-positive, GABAergic vlPAG neurons. Antagonism of both muscarinic and nicotinic receptors completely abolished the effects of carbachol. We suggest cholinergic neurons inhibit DRN 5-HT neurons when acetylcholine levels are lower i.e. during quiet wakefulness and the beginning of REM sleep periods, in part via excitation of muscarinic and nicotinic receptors located on local vlPAG and DRN GABAergic neurons. Higher firing rates or burst firing of cholinergic neurons associated with attentive wakefulness or phasic REM sleep periods leads to excitation of 5-HT neurons via the activation of nicotinic receptors located postsynaptically and presynaptically on excitatory afferents.


Assuntos
Carbacol/farmacologia , Agonistas Colinérgicos/farmacologia , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios Serotoninérgicos/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Animais , Feminino , Neurônios GABAérgicos/fisiologia , Técnicas de Introdução de Genes , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos , Antagonistas Muscarínicos/farmacologia , Antagonistas Nicotínicos/farmacologia , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Substância Cinzenta Periaquedutal/fisiologia , Núcleos da Rafe/efeitos dos fármacos , Núcleos da Rafe/fisiologia , Receptores Muscarínicos/metabolismo , Receptores Nicotínicos/metabolismo , Neurônios Serotoninérgicos/fisiologia , Sinapses/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
19.
Neuroscience ; 253: 316-29, 2013 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-24025868

RESUMO

Presynaptic functions of the mammalian central neurons are regulated by a network of protein interactions. Synaptic vesicle recycling in and neurotransmitter release from the presynaptic nerve terminals are altered when a glutamate-deleting mutation is present in the torsinA protein (ΔE-torsinA). This mutation is linked with a hereditary form of the movement disorder dystonia known as DYT1 dystonia. Although torsinA expression is prevalent throughout the central nervous system, its subcellular localization - in particular with respect to presynaptic nerve terminals - remains unclear. This information would be useful in narrowing down possible models for how wild-type torsinA affects presynaptic function, as well as the nature of the presynaptic dysfunction that arises in the context of ΔE-torsinA mutation. Here we report on an analysis of the presynaptic localization of torsinA in cultured neurons obtained from a knock-in mouse model of DYT1 dystonia. Primary cultures of neurons were established from heterozygous and homozygous ΔE-torsinA knock-in mice, as well as from their wild-type littermates. Neurons were obtained from the striatum, cerebral cortex and hippocampus of these mice, and were subjected to immunocytochemistry. This analysis revealed the expression of both proteins in the somata and dendrites. However, neither the nerve terminals nor axonal shafts were immunoreactive. These results were confirmed by fluorogram-based quantitation. Our findings indicate that neither the wild-type nor the ΔE-torsinA mutant protein is present at substantial levels in the presynaptic structures of cultured neurons. Thus, the effects of torsinA, in wild-type and mutant forms, appear to influence presynaptic function indirectly, without residing in presynaptic structures.


Assuntos
Hipocampo/citologia , Chaperonas Moleculares/metabolismo , Terminações Nervosas/metabolismo , Neurônios/citologia , Análise de Variância , Animais , Cloreto de Cádmio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Imunofluorescência , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/efeitos dos fármacos , Potássio/farmacologia , ômega-Agatoxina IVA/farmacologia , ômega-Conotoxina GVIA/farmacologia
20.
Front Cell Neurosci ; 5: 2, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21503156

RESUMO

In monoamine-releasing terminals, neurotransmitter transporters - in addition to terminating synaptic transmission by clearing released transmitters from the extracellular space - are the primary mechanism for replenishing transmitter stores and thus regulate presynaptic homeostasis. Here, we analyze whether GAT-1, the main plasma membrane GABA transporter, plays a similar role in GABAergic terminals. Re-examination of existing literature and recent data gathered in our laboratory show that GABA homeostasis in GABAergic terminals is dominated by the activity of the GABA synthesizing enzyme and that GAT-1-mediated GABA transport contributes to cytosolic GABA levels. However, analysis of GAT-1 KO, besides demonstrating the effects of reduced clearance, reveals the existence of changes compatible with an impaired presynaptic function, as miniature IPSCs frequency is reduced by one-third and glutamic acid decarboxylases and phosphate-activated glutaminase levels are significantly up-regulated. Although the changes observed are less robust than those reported in mice with impaired dopamine, noradrenaline, and serotonin plasma membrane transporters, they suggest that in GABAergic terminals GAT-1 impacts on presynaptic GABA homeostasis, and may contribute to the activity-dependent regulation of inhibitory efficacy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA