Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 229
Filtrar
1.
Biochem Biophys Res Commun ; 465(1): 145-51, 2015 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-26239662

RESUMO

The balance of cation-chloride co-transporters, particularly KCC2 and NKCC1, is critical for GABAergic inhibitory signaling. However, KCC2/NKCC1 balance is disrupted in many neurodegenerative diseases. Moreover, correlations between chronic stress, KCC2 and NKCC1 in the hippocampus remain poorly understood. Despite the fact that emotional disorders in humans are far more prevalent in women, there have been relatively few studies about female subjects. Here we investigated behaviors and expression patterns of KCC2 and NKCC1 in the hippocampi of female mice under chronic stress. Repeated stress (RS) was induced in experimental mice by repeated forced water administration. Then, expression patterns of GABAergic signaling molecules were identified by immunohistochemical analysis and performance was assessed using several behavioral tests. The results of semi-quantitative analysis showed that RS decreased KCC2 expression and increased NKCC1 expression in membranes of granular and pyramidal cells in the hippocampus. The novel object recognition (NOR) test and sociability test revealed that RS induced cognitive and sociability deficits, whereas RS increased the time spent in the open arms of the elevated plus maze test and induced attention deficits in other tests. In summary, RS induced alterations in membrane KCC2/NKCC1 balance in the hippocampus of female mice, which may contribute to GABAergic disinhibition associated with cognitional, sociability and attention deficits.


Assuntos
Comportamento Exploratório , Hipocampo/metabolismo , Comportamento Social , Membro 2 da Família 12 de Carreador de Soluto/genética , Estresse Psicológico/genética , Simportadores/genética , Animais , Atenção , Membrana Celular/metabolismo , Cognição , Feminino , Regulação da Expressão Gênica , Hipocampo/fisiopatologia , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Células Piramidais/metabolismo , Células Piramidais/fisiopatologia , Transdução de Sinais , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Estresse Psicológico/etiologia , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Natação , Simportadores/metabolismo , Água/administração & dosagem , Cotransportadores de K e Cl-
2.
Neuroscience ; 288: 145-55, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-25555930

RESUMO

Apart from therapeutic discovery, the study of mild traumatic brain injury (mTBI) has been focused on two challenges: why do a majority of individuals recover with little concern, while a considerable proportion suffer with persistent and often debilitating symptomology; and, how do mild injuries significantly increase risk for an early-onset neurodegeneration? Owing to a lack of observable damage following mTBI, this study was designed to determine if there were changes in neuronal morphology, synaptic connectivity, and epigenetic patterning that could contribute to the manifestation of persistent neurological dysfunction. Prefrontal cortex tissue from male and female rats was used for Golgi-Cox analysis along with the profiling of changes in gene expression (BDNF, DNMT1, FGF2, IGF1, Nogo-A, OXYR, and TERT) and telomere length (TL), following a single mTBI or sham injury in the juvenile period. Golgi-Cox analysis of dendritic branch order, dendritic length, and spine density demonstrate that an early mTBI increases complexity of pyramidal neurons in the mPFC. Furthermore, there are also substantial changes in the expression levels of the seven genes of interest and TL following a single mild injury in this brain region. The results from the neuroanatomical measures and changes in gene expression indicate that the mTBI disrupts normal pruning processes that are typically underway at this point in development. In addition, there are significant interactions between the social environment and epigenetic processes that work in concert to perpetuate neurological dysfunction.


Assuntos
Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Córtex Pré-Frontal/patologia , Córtex Pré-Frontal/fisiopatologia , Animais , Dendritos/patologia , Dendritos/fisiologia , Modelos Animais de Doenças , Epigênese Genética/fisiologia , Feminino , Expressão Gênica/fisiologia , Perfilação da Expressão Gênica , Masculino , Córtex Pré-Frontal/crescimento & desenvolvimento , Células Piramidais/patologia , Células Piramidais/fisiopatologia , Ratos , Sinapses/patologia , Sinapses/fisiologia , Telômero/metabolismo , Telômero/patologia
3.
Hippocampus ; 25(2): 159-68, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25209930

RESUMO

Rett syndrome (RTT) is a neurodevelopment disorder associated with intellectual disabilities and caused by loss-of-function mutations in the gene encoding the transcriptional regulator Methyl-CpG-binding Protein-2 (MeCP2). Neuronal dysfunction and changes in cortical excitability occur in RTT individuals and Mecp2-deficient mice, including hippocampal network hyperactivity and higher frequency of spontaneous multiunit spikes in the CA3 cell body layer. Here, we describe impaired synaptic inhibition and an excitation/inhibition (E/I) imbalance in area CA3 of acute slices from symptomatic Mecp2 knockout male mice (referred to as Mecp2(-/y) ). The amplitude of TTX-resistant miniature inhibitory postsynaptic currents (mIPSC) was smaller in CA3 pyramidal neurons of Mecp2(-/y) slices than in wildtype controls, while the amplitude of miniature excitatory postsynaptic currents (mEPSC) was significantly larger in Mecp2(-/y) neurons. Consistently, quantitative confocal immunohistochemistry revealed significantly lower intensity of the alpha-1 subunit of GABAA Rs in the CA3 cell body layer of Mecp2(-/y) mice, while GluA1 puncta intensities were significantly higher in the CA3 dendritic layers of Mecp2(-/y) mice. In addition, the input/output (I/O) relationship of evoked IPSCs had a shallower slope in CA3 pyramidal neurons Mecp2(-/y) neurons. Consistent with the absence of neuronal degeneration in RTT and MeCP2-based mouse models, the density of parvalbumin- and somatostatin-expressing interneurons in area CA3 was not affected in Mecp2(-/y) mice. Furthermore, the intrinsic membrane properties of several interneuron subtypes in area CA3 were not affected by Mecp2 loss. However, mEPSCs are smaller and less frequent in CA3 fast-spiking basket cells of Mecp2(-/y) mice, suggesting an impaired glutamatergic drive in this interneuron population. These results demonstrate that a loss-of-function mutation in Mecp2 causes impaired E/I balance onto CA3 pyramidal neurons, leading to a hyperactive hippocampal network, likely contributing to limbic seizures in Mecp2(-/y) mice and RTT individuals.


Assuntos
Região CA3 Hipocampal/fisiopatologia , Proteína 2 de Ligação a Metil-CpG/deficiência , Proteína 2 de Ligação a Metil-CpG/fisiologia , Inibição Neural/fisiologia , Células Piramidais/fisiopatologia , Sinapses/fisiologia , Animais , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores , Imuno-Histoquímica , Potenciais Pós-Sinápticos Inibidores , Interneurônios/fisiologia , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos Knockout , Microscopia Confocal , Potenciais Pós-Sinápticos em Miniatura , Técnicas de Patch-Clamp , Receptores de AMPA/metabolismo , Receptores de GABA-A/metabolismo , Síndrome de Rett , Técnicas de Cultura de Tecidos
4.
CNS Neurosci Ther ; 21(2): 152-63, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25438677

RESUMO

AIMS: Rasmussen encephalitis (RE) is a rare but devastating condition, mainly in children, characterized by sustained brain inflammation, atrophy of one cerebral hemisphere, epilepsy, and progressive cognitive deterioration. The etiology of RE-induced seizures associated with the inflammatory process remains unknown. METHODS: Cortical tissue samples from children undergoing surgical resections for the treatment of RE (n = 16) and non-RE (n = 12) were compared using electrophysiological, morphological, and immunohistochemical techniques to examine neuronal properties and the relationship with microglial activation using the specific microglia/macrophage calcium-binding protein, IBA1 in conjunction with connexins and pannexin expression. RESULTS: Compared with non-RE cases, pyramidal neurons from RE cases displayed increased cell capacitance and reduced input resistance. However, neuronal somatic areas were not increased in size. Instead, intracellular injection of biocytin led to increased dye coupling between neurons from RE cases. By Western blot, expression of IBA1 and pannexin was increased while connexin 32 was decreased in RE cases compared with non-RE cases. IBA1 immunostaining overlapped with pannexin and connexin 36 in RE cases. CONCLUSIONS: In RE, these results support the notion that a possible mechanism for cellular hyperexcitability may be related to increased intercellular coupling from pannexin linked to increased microglial activation. Such findings suggest that a possible antiseizure treatment for RE may involve the use of gap junction blockers.


Assuntos
Córtex Cerebral/patologia , Encefalite/patologia , Células Piramidais/fisiologia , Células Piramidais/fisiopatologia , 4-Aminopiridina/farmacologia , Adolescente , Biofísica , Proteínas de Ligação ao Cálcio , Criança , Estudos de Coortes , Conexinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Humanos , Técnicas In Vitro , Lisina/análogos & derivados , Imageamento por Ressonância Magnética , Masculino , Mefloquina/farmacologia , Potenciais da Membrana/fisiologia , Proteínas dos Microfilamentos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia
5.
Epilepsy Res ; 109: 13-27, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25524838

RESUMO

The most commonly reported precipitating factor for seizures is stress. However, the underlying mechanisms whereby stress triggers seizures are not yet fully understood. Here we demonstrate a potential mechanism underlying changes in neuronal excitability in the hippocampus following chronic stress, involving a shift in the reversal potential for GABA (EGABA) associated with a dephosphorylation of the potassium chloride co-transporter, KCC2. Mice subjected to chronic restraint stress (30min/day for 14 consecutive days) exhibit an increase in serum corticosterone levels which is associated with increased susceptibility to seizures induced with kainic acid (20mg/kg). Following chronic stress, but not acute stress, we observe a dephosphorylation of KCC2 residue S940, which regulates KCC2 cell surface expression and function, in the hippocampus. To determine the impact of alterations in KCC2 expression following chronic stress, we performed gramicidin perforated patch recordings to measure changes in EGABA and neuronal excitability of principal hippocampal neurons. We observe a depolarizing shift in EGABA in hippocampal CA1 pyramidal neurons after chronic stress. In addition, there is an increase in the intrinsic excitability of CA1 pyramidal neurons, evident by a shift in the input-output curve which could be reversed with the NKCC1 inhibitor, bumetanide. These data uncover a potential mechanism involving chronic stress-induced plasticity in chloride homeostasis which may contribute to stress-induced seizure susceptibility.


Assuntos
Hipocampo/fisiopatologia , Convulsões/fisiopatologia , Estresse Psicológico/fisiopatologia , Ácido gama-Aminobutírico/metabolismo , Doença Aguda , Animais , Bumetanida/farmacologia , Doença Crônica , Corticosterona/sangue , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Ácido Caínico , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos Endogâmicos C57BL , Fosforilação , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiopatologia , Restrição Física , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Simportadores/metabolismo , Cotransportadores de K e Cl-
6.
Neuroscience ; 286: 325-37, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25498224

RESUMO

Norepinephrine acting via ß-adrenergic receptors (ß-ARs) plays an important role in hippocampal plasticity including the subiculum which is the principal target of CA1 pyramidal cells and which controls information transfer from the hippocampus to other brain regions including the neighboring presubiculum and the entorhinal cortex (EC). Subicular pyramidal cells are classified as regular- (RS) and burst-spiking (BS) cells. Activation of ß-ARs at CA1-subiculum synapses induces long-term potentiation (LTP) in burst- but not in RS cells (Wójtowicz et al., 2010). To elucidate seizure-associated disturbances in the norepinephrine-dependent modulation of hippocampal output, we investigated the functional consequences of the ß-AR-dependent synaptic plasticity at CA1-subiculum synapses for the transfer of hippocampal output to the parahippocampal region in the pilocarpine model of temporal lobe epilepsy. Using single-cell and multi-channel field recordings in slices, we studied ß-AR-mediated changes in the functional connectivity between CA1, the subiculum and its target-structures. We confirm that application of the ß-adrenergic agonist isoproterenol induces LTP in subicular BS- but not RS cells. Due to the distinct spatial distribution of RS- and BS cells in the proximo-to-distal axis of the subiculum, in field recordings, LTP was significantly stronger in the distal than in the proximal subiculum. In pilocarpine-treated animals, ß-AR-mediated LTP was strongly reduced in the distal subiculum. The attenuated LTP was associated with a disturbed polysynaptic transmission from the CA1, via the subiculum to the presubiculum, but with a preserved transmission to the medial EC. Our findings suggest that synaptic plasticity may influence target-related information flow and that such regulation is disturbed in pilocarpine-treated epileptic rats.


Assuntos
Epilepsia do Lobo Temporal/fisiopatologia , Hipocampo/fisiopatologia , Células Piramidais/fisiopatologia , Receptores Adrenérgicos beta/fisiologia , Potenciais de Ação/efeitos dos fármacos , Agonistas Adrenérgicos beta/farmacologia , Animais , Modelos Animais de Doenças , Estimulação Elétrica , Epilepsia do Lobo Temporal/induzido quimicamente , Hipocampo/efeitos dos fármacos , Isoproterenol/farmacologia , Potenciação de Longa Duração , Masculino , Giro Para-Hipocampal/efeitos dos fármacos , Giro Para-Hipocampal/fisiopatologia , Pilocarpina , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Wistar
7.
Behav Brain Res ; 281: 149-55, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25541037

RESUMO

This study aims to determine whether low-frequency repetitive transcranial magnetic stimulation (rTMS) protects pyramidal cells from apoptosis and promotes hippocampal synaptic plasticity in a vascular dementia (VaD) rat model. Following establishment of a VaD rat model using two-vessel occlusion (2VO), learning and memory were evaluated via the Morris Water Maze (MWM), hippocampal CA1 neuron ultrastructure was examined via electron microscopy, and hippocampal synaptic plasticity was assessed by long-term potentiation (LTP). Western blot was used to detect the expression of N-methyl-d-aspartic acid receptor 1 (NMDAR1), Bcl-2, and Bax. Compared with VaD group, rats treated with low-frequency rTMS had reduced-escape latencies, increased swimming time in the target quadrant (P<0.05), and significantly less synaptic structure damage. LTP at hippocampal CA3-CA1 synapses was enhanced (P<0.05). Low-frequency rTMS significantly up-regulated NMDAR1 and Bcl-2 expression and down-regulated Bax expression. Low-frequency rTMS improves learning and memory, protects the synapse, and increases synaptic plasticity in VaD model rats. Increased Bcl-2 expression and reduced Bax expression may be a novel protective mechanism of low-frequency rTMS treatment for VaD.


Assuntos
Apoptose , Demência Vascular/terapia , Hipocampo/patologia , Memória , Plasticidade Neuronal , Estimulação Magnética Transcraniana/métodos , Animais , Comportamento Animal , Demência Vascular/patologia , Demência Vascular/fisiopatologia , Demência Vascular/psicologia , Modelos Animais de Doenças , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Potenciação de Longa Duração , Masculino , Aprendizagem em Labirinto , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células Piramidais/metabolismo , Células Piramidais/fisiopatologia , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Resultado do Tratamento , Proteína X Associada a bcl-2/metabolismo
8.
Int J Neuropsychopharmacol ; 18(4)2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25522394

RESUMO

BACKGROUND: Olfactory bulbectomized rats generally manifest many of the neurochemical, physiological, and behavioral features of major depressive disorder in humans. Another interesting feature of this model is that it responds to chronic but not acute antidepressant treatments, including selective serotonin reuptake inhibitors. The purpose of the present study was first to characterize the firing activity of dorsal raphe serotonin neurons in olfactory bulbectomized rats and then examine the effects of 2 antidepressants, bupropion and paroxetine. METHODS: Olfactory bulbectomy was performed by aspirating olfactory bulbs in anesthetized rats. Vehicle and drugs were delivered for 2 and 14 days via subcutaneously implanted minipumps. In vivo electrophysiological recordings were carried out in male anesthetized Sprague-Dawley rats. RESULTS: Following ablation of olfactory bulbs, the firing rate of serotonin neurons was decreased by 36%, leaving those of norepinephrine and dopamine neurons unchanged. In olfactory bulbectomized rats, bupropion (30 mg/kg/d) restored the firing rate of serotonin neurons to the control level following 2- and 14-day administration and also induced an increase in the tonic activation of serotonin(1A) receptors; paroxetine (10 mg/kg/d) did not result in a return to normal of the attenuated firing of serotonin neurons in olfactory bulbectomized rats. In the hippocampus, although at a higher dose of WAY 100635 than that required in bupropion-treated animals, paroxetine administration also resulted in an increase in the tonic activation of serotonin(1A) receptors. CONCLUSIONS: The present results indicate that unlike paroxetine, bupropion administration normalized serotonin neuronal activity and increased tonic activation of the serotonin(1A) receptors in hippocampus.


Assuntos
Antidepressivos de Segunda Geração/farmacologia , Bupropiona/farmacologia , Núcleo Dorsal da Rafe/efeitos dos fármacos , Bulbo Olfatório/fisiopatologia , Paroxetina/farmacologia , Neurônios Serotoninérgicos/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Região CA3 Hipocampal/efeitos dos fármacos , Região CA3 Hipocampal/fisiopatologia , Transtorno Depressivo Maior , Modelos Animais de Doenças , Núcleo Dorsal da Rafe/fisiopatologia , Locus Cerúleo/efeitos dos fármacos , Locus Cerúleo/fisiopatologia , Masculino , Piperazinas/farmacologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiopatologia , Piridinas/farmacologia , Ratos Sprague-Dawley , Receptor 5-HT1A de Serotonina/metabolismo , Neurônios Serotoninérgicos/fisiologia , Antagonistas da Serotonina/farmacologia , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiologia
9.
J Neurosci ; 34(46): 15208-22, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25392490

RESUMO

Epileptic seizures are characterized by periods of hypersynchronous, hyperexcitability within brain networks. Most seizures involve two stages: an initial tonic phase, followed by a longer clonic phase that is characterized by rhythmic bouts of synchronized network activity called afterdischarges (ADs). Here we investigate the cellular and network mechanisms underlying hippocampal ADs in an effort to understand how they maintain seizure activity. Using in vitro hippocampal slice models from rats and mice, we performed electrophysiological recordings from CA3 pyramidal neurons to monitor network activity and changes in GABAergic signaling during epileptiform activity. First, we show that the highest synchrony occurs during clonic ADs, consistent with the idea that specific circuit dynamics underlie this phase of the epileptiform activity. We then show that ADs require intact GABAergic synaptic transmission, which becomes excitatory as a result of a transient collapse in the chloride (Cl(-)) reversal potential. The depolarizing effects of GABA are strongest at the soma of pyramidal neurons, which implicates somatic-targeting interneurons in AD activity. To test this, we used optogenetic techniques to selectively control the activity of somatic-targeting parvalbumin-expressing (PV(+)) interneurons. Channelrhodopsin-2-mediated activation of PV(+) interneurons during the clonic phase generated excitatory GABAergic responses in pyramidal neurons, which were sufficient to elicit and entrain synchronous AD activity across the network. Finally, archaerhodopsin-mediated selective silencing of PV(+) interneurons reduced the occurrence of ADs during the clonic phase. Therefore, we propose that activity-dependent Cl(-) accumulation subverts the actions of PV(+) interneurons to perpetuate rather than terminate pathological network hyperexcitability during the clonic phase of seizures.


Assuntos
Região CA3 Hipocampal/fisiopatologia , Neurônios GABAérgicos/fisiologia , Interneurônios/fisiologia , Parvalbuminas/metabolismo , Células Piramidais/fisiopatologia , Convulsões/fisiopatologia , Potenciais de Ação/fisiologia , Animais , Camundongos , Modelos Neurológicos , Optogenética , Ratos
10.
Neuroscience ; 280: 243-53, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25242645

RESUMO

Previous work has shown a reduction of apical dendritic length and spine density in neurons from the CA1 hippocampus subfield of spontaneously hypertensive rats (SHRs). These abnormalities are prevented by treatment for 2 weeks with 17ß-estradiol. In view of the fact that diabetes and hypertension are comorbid diseases, we have now studied the effect of Streptozotocin-induced diabetes on the dendritic tree and spines of CA1 hippocampus neurons, and also compared the regulation of these parameters by 17ß-estradiol in diabetic and normoglycemic SHR. Twenty-week-old male SHR received i.v. 40-mg/kg Streptozotocin or vehicle and studied 1 month afterward. A group of normoglycemic and hyperglycemic SHR also received s.c. a single 17ß-estradiol pellet or vehicle for 2weeks. Hippocampus sections were impregnated with silver nitrate following a modified Golgi's method and the arbor of CA1 pyramidal neurons analyzed by Sholl's method. 17ß-Estradiol treatment of normoglycemic SHR reversed the reduced length of apical dendrites, the low spine density and additionally decreased blood pressure (BP). Diabetic SHR showed increased length of apical and basal dendrites but reduced spine density compared to normoglycemic SHR. Diabetes also decreased BP of SHR. Treatment with 17ß-estradiol of diabetic SHR enhanced dendritic length, increased dendritic spine density and further decreased BP. Thus, changes of cytoarchitecture of CA1 neurons due to 17ß-estradiol treatment of normoglycemic SHR persisted after diabetes induction. A decrease of BP may also contribute to the central effects of 17ß-estradiol in SHR diabetic rats.


Assuntos
Região CA1 Hipocampal/efeitos dos fármacos , Dendritos/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Estradiol/farmacologia , Fármacos Neuroprotetores/farmacologia , Células Piramidais/efeitos dos fármacos , Animais , Pressão Sanguínea/efeitos dos fármacos , Região CA1 Hipocampal/patologia , Região CA1 Hipocampal/fisiopatologia , Dendritos/patologia , Dendritos/fisiologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/fisiopatologia , Hipertensão/tratamento farmacológico , Hipertensão/patologia , Hipertensão/fisiopatologia , Masculino , Fotomicrografia , Células Piramidais/patologia , Células Piramidais/fisiopatologia , Ratos Endogâmicos SHR
11.
Neuroscience ; 280: 181-92, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25241070

RESUMO

The mechanisms underlying antiepileptic or antiepileptogenic effects of repeated transcranial magnetic stimulation (rTMS) are poorly understood. In this study, we investigated the effect of rTMS applied during rapid amygdala kindling on some electrophysiological properties of hippocampal CA1 pyramidal neurons. Male Wistar rats were kindled by daily electrical stimulation of the basolateral amygdala in a semi-rapid manner (12 stimulations/day) until they achieved stage-5 seizure. One group (kindled+rTMS (KrTMS)) of animals received rTMS (1Hz for 4min) 5min after termination of daily kindling stimulations. Twenty four hours following the last kindling stimulation electrophysiological properties of hippocampal CA1 pyramidal neurons were investigated using whole-cell patch-clamp technique. Amygdala kindling significantly depolarized the resting membrane potential and increased the input resistance, spontaneous firing activity, number of evoked spikes and half-width of the first evoked spike. Kindling also decreased the first-spike latency and amplitude significantly. Application of rTMS during kindling somehow prevented the development of seizures and protected CA1 pyramidal neurons of hippocampus against deleterious effect of kindling on both passive and active neuronal electrophysiological properties. Interestingly, application of rTMS alone enhanced the excitability of CA1 pyramidal neurons significantly. Based on the results of our study, it may be suggested that rTMS exerts its anticonvulsant effect, in part, through preventing the amygdala kindling-induced changes in electrophysiological properties of hippocampal CA1 pyramidal neurons. It seems that rTMS exerts protective effects on the neural circuits involved in spreading the seizures from the focus to other parts of the brain.


Assuntos
Região CA1 Hipocampal/fisiopatologia , Excitação Neurológica/fisiologia , Células Piramidais/fisiopatologia , Convulsões/fisiopatologia , Convulsões/terapia , Estimulação Magnética Transcraniana/métodos , Animais , Complexo Nuclear Basolateral da Amígdala/fisiopatologia , Modelos Animais de Doenças , Impedância Elétrica , Neuroestimuladores Implantáveis , Masculino , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Distribuição Aleatória , Ratos Wistar , Técnicas de Cultura de Tecidos
12.
Eur J Neurosci ; 40(10): 3538-47, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25209309

RESUMO

Major depressive disorder (MDD) is one of the leading causes of morbidity worldwide. Several antidepressants have been widely prescribed to treat patients with MDD. However, neuronal changes in brain function remain poorly understood. Based on the standard chronic mild stress (CMS) model of depression in mice, we investigated the neuronal mechanisms of the classic antidepressant, fluoxetine, and a new compound (termed YY-23 in this study) derived from furostanol saponin. The results showed that both fluoxetine and YY-23 normalized CMS-induced depressive-like behaviors. YY-23 caused antidepressant-like behaviors with a faster action than fluoxetine. In terms of in vivo neuronal activities, a CMS-induced decrease in spontaneous firing in burst of medial prefrontal cortex pyramidal neurons rather than ventral tegmental area (VTA) was reversed by the chronic administration of fluoxetine and YY-23. We also found that CMS-induced deficits in the expression of prefrontal brain-derived neurotrophic factor (BDNF) were also restored by chronically administering YY-23 and fluoxetine. In addition, chronic administration of fluoxetine rather than YY-23 resulted in an improvement of antidepressive-like behavior and a change of burst firing of VTA in control-housed animals, indicating that the pharmacological effects of YY-23 were specific to CMS-treated animals. Together, these data suggest that the burst-firing patterns of pyramidal cells may be a neural biomarker of depressive-like mice and antidepressant action. Furthermore, synaptic transmission and BDNF may contribute to the rapid antidepressant-like effects on depression.


Assuntos
Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/fisiopatologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiopatologia , Potenciais de Ação/efeitos dos fármacos , Animais , Antidepressivos/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Doença Crônica , Modelos Animais de Doenças , Fluoxetina/farmacologia , Masculino , Camundongos , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiopatologia , Distribuição Aleatória , Estresse Psicológico , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiopatologia
13.
Neuroscience ; 280: 275-81, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25194787

RESUMO

Lipoprotein lipase (LPL) is expressed at high levels in hippocampal neurons, although its function is unclear. We previously reported that LPL-deficient mice have learning and memory impairment and fewer synaptic vesicles in hippocampal neurons, but properties of synaptic activity in LPL-deficient neurons remain unexplored. In this study, we found reduced frequency of miniature excitatory postsynaptic currents (mEPSCs) and readily releasable pool (RRP) size in LPL-deficient neurons, which led to presynaptic dysfunction and plasticity impairment without altering postsynaptic activity. We demonstrated that synaptic vesicle recycling, which is known to play an important role in maintaining the RRP size in active synapses, is impaired in LPL-deficient neurons. Moreover, lipid assay revealed deficient docosahexaenoic acid (DHA) and arachidonic acid (AA) in the hippocampus of LPL-deficient mice; exogenous DHA or AA supplement partially restored synaptic vesicle recycling capability. These results suggest that impaired synaptic vesicle recycling results from deficient DHA and AA and contributes to the presynaptic dysfunction and plasticity impairment in LPL-deficient neurons.


Assuntos
Hipocampo/fisiopatologia , Lipase Lipoproteica/deficiência , Terminações Pré-Sinápticas/enzimologia , Vesículas Sinápticas/enzimologia , Animais , Ácido Araquidônico/administração & dosagem , Ácido Araquidônico/metabolismo , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácidos Docosa-Hexaenoicos/metabolismo , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Cromatografia Gasosa-Espectrometria de Massas , Hipocampo/efeitos dos fármacos , Hiperlipoproteinemia Tipo I/tratamento farmacológico , Hiperlipoproteinemia Tipo I/fisiopatologia , Lipase Lipoproteica/genética , Masculino , Camundongos Endogâmicos C57BL , Potenciais Pós-Sinápticos em Miniatura/efeitos dos fármacos , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiopatologia , Vesículas Sinápticas/efeitos dos fármacos , Técnicas de Cultura de Tecidos
14.
Brain Res ; 1582: 220-6, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25086204

RESUMO

OBJECTIVE: mild traumatic brain injury (mTBI) can produce lasting memory deficits even in the absence of cell loss. We investigated changes in hippocampal firing patterns during exploration and during a novel object recognition (NOR) task. METHODS: six male Sprague-Dawley rats were subjected to mTBI via fluid percussion injury and were compared with sham-operated rats. Microelectrodes were implanted into CA1 and CA3 and multiple units were recorded from the pyramidal cell layer. Spontaneous "burst" characteristics were analyzed and temporal firing patterns were correlated with object encounters to establish object-specific firing patterns. RESULTS: mTBI was associated with significantly less hippocampal bursting (p<0.05) with a trend toward longer bursts and lower interburst spike frequency. mTBI was also associated with no preference for a novel object at 12h (p<0.05). During the NOR task, a subset of pyramidal cells were identified which consistently demonstrated a transiently increased firing rate upon encounter of a specific object ("object-specific" cell). Across both groups, there was a significant (p<0.05) correlation between preference for object novelty and the difference between the total number of novel object-specific cells and familiar object-specific cells. The proportion of object-specific cells that responded to the unexpected (novel) object compared to those responding to the familiar object was significantly smaller in rats that had been exposed to mTBI (p<0.05). CONCLUSION: memory deficits after mTBI are associated with decreased intrinsic burst activity and impaired context-specific firing patterns in the hippocampus during object exploration.


Assuntos
Lesões Encefálicas/fisiopatologia , Região CA1 Hipocampal/fisiopatologia , Região CA3 Hipocampal/fisiopatologia , Transtornos da Memória/fisiopatologia , Células Piramidais/fisiopatologia , Reconhecimento Psicológico/fisiologia , Potenciais de Ação/fisiologia , Animais , Lesões Encefálicas/complicações , Modelos Animais de Doenças , Eletrodos Implantados , Masculino , Transtornos da Memória/etiologia , Testes Neuropsicológicos , Distribuição Aleatória , Ratos Sprague-Dawley
15.
J Neurophysiol ; 112(7): 1703-13, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24990567

RESUMO

A variety of major developmental cortical malformations are closely associated with clinically intractable epilepsy. Pathophysiological aspects of one such disorder, human polymicrogyria, can be modeled by making neocortical freeze lesions (FL) in neonatal rodents, resulting in the formation of microgyri. Previous studies showed enhanced excitatory and inhibitory synaptic transmission and connectivity in cortical layer V pyramidal neurons in the paramicrogyral cortex. In young adult transgenic mice that express green fluorescent protein (GFP) specifically in parvalbumin positive fast-spiking (FS) interneurons, we used laser scanning photostimulation (LSPS) of caged glutamate to map excitatory and inhibitory synaptic connectivity onto FS interneurons in layer V of paramicrogyral cortex in control and FL groups. The proportion of uncaging sites from which excitatory postsynaptic currents (EPSCs) could be evoked (hotspot ratio) increased slightly but significantly in FS cells of the FL vs. control cortex, while the mean amplitude of LSPS-evoked EPSCs at hotspots did not change. In contrast, the hotspot ratio of inhibitory postsynaptic currents (IPSCs) was significantly decreased in FS neurons of the FL cortex. These alterations in synaptic inputs onto FS interneurons may result in an enhanced inhibitory output. We conclude that alterations in synaptic connectivity to cortical layer V FS interneurons do not contribute to hyperexcitability of the FL model. Instead, the enhanced inhibitory output from these neurons may partially offset an earlier demonstrated increase in synaptic excitation of pyramidal cells and thereby maintain a relative balance between excitation and inhibition in the affected cortical circuitry.


Assuntos
Neurônios GABAérgicos/fisiologia , Interneurônios/fisiologia , Polimicrogiria/fisiopatologia , Potenciais Sinápticos , Animais , Modelos Animais de Doenças , Congelamento , Camundongos , Camundongos Transgênicos , Células Piramidais/fisiopatologia , Sinapses/fisiologia
16.
Hippocampus ; 24(12): 1421-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24976385

RESUMO

Anorexia nervosa (AN) is a psychiatric illness characterized by restricted eating and irrational fears of gaining weight. There is no accepted pharmacological treatment for AN, and AN has the highest mortality rate among psychiatric illnesses. Anorexia nervosa most commonly affects females during adolescence, suggesting an effect of sex and hormones on vulnerability to the disease. Activity-based anorexia (ABA) is a rodent model of AN that shares symptoms with AN, including over-exercise, elevation of stress hormones, and genetic links to anxiety traits. We previously reported that ABA in adolescent female rats results in increased apical dendritic branching in CA1 pyramidal cells of the ventral hippocampus at postnatal day 44 (P44). To examine the long-term effects of adolescent ABA (P44) in female rats, we compared the apical branching in the ventral hippocampal CA1 after recovery from ABA (P51) and after a relapse of ABA (P55) with age-matched controls. To examine the age-dependence of the hippocampal plasticity, we examined the effect of ABA during adulthood (P67). We found that while ABA at P44 resulted in increased branching of ventral hippocampal pyramidal cells, relapse of ABA at P55 resulted in decreased branching. ABA induced during adulthood did not have an effect on dendritic branching, suggesting an age-dependence of the vulnerability to structural plasticity. Cells from control animals were found to exhibit a dramatic increase in branching, more than doubling from P44 to P51, followed by pruning from P51 to P55. The proportion of mature spines on dendrites from the P44-ABA animals is similar to that on dendrites from P55-CON animals. These results suggest that the experience of ABA may cause precocious anatomical development of the ventral hippocampus. Importantly, we found that adolescence is a period of continued development of the hippocampus, and increased vulnerability to mental disorders during adolescence may be due to insults during this developmentally critical period.


Assuntos
Anorexia/fisiopatologia , Região CA1 Hipocampal/crescimento & desenvolvimento , Região CA1 Hipocampal/fisiopatologia , Atividade Motora/fisiologia , Células Piramidais/crescimento & desenvolvimento , Células Piramidais/fisiopatologia , Animais , Anorexia/patologia , Peso Corporal , Região CA1 Hipocampal/patologia , Dendritos/patologia , Dendritos/fisiologia , Modelos Animais de Doenças , Feminino , Plasticidade Neuronal/fisiologia , Células Piramidais/patologia , Ratos Sprague-Dawley , Recidiva
17.
Mol Psychiatry ; 19(10): 1133-42, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24934177

RESUMO

Intellectual disabilities (IDs) and autism spectrum disorders link to human APC inactivating gene mutations. However, little is known about adenomatous polyposis coli's (APC's) role in the mammalian brain. This study is the first direct test of the impact of APC loss on central synapses, cognition and behavior. Using our newly generated APC conditional knock-out (cKO) mouse, we show that deletion of this single gene in forebrain neurons leads to a multisyndromic neurodevelopmental disorder. APC cKO mice, compared with wild-type littermates, exhibit learning and memory impairments, and autistic-like behaviors (increased repetitive behaviors, reduced social interest). To begin to elucidate neuronal changes caused by APC loss, we focused on the hippocampus, a key brain region for cognitive function. APC cKO mice display increased synaptic spine density, and altered synaptic function (increased frequency of miniature excitatory synaptic currents, modestly enhanced long-term potentiation). In addition, we found excessive ß-catenin levels and associated changes in canonical Wnt target gene expression and N-cadherin synaptic adhesion complexes, including reduced levels of presenilin1. Our findings identify some novel functional and molecular changes not observed previously in other genetic mutant mouse models of co-morbid cognitive and autistic-like disabilities. This work thereby has important implications for potential therapeutic targets and the impact of their modulation. We provide new insights into molecular perturbations and cell types that are relevant to human ID and autism. In addition, our data elucidate a novel role for APC in the mammalian brain as a hub that links to and regulates synaptic adhesion and signal transduction pathways critical for normal cognition and behavior.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Transtorno Autístico/fisiopatologia , Transtornos Cognitivos/fisiopatologia , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Transtorno Autístico/patologia , Caderinas/metabolismo , Transtornos Cognitivos/patologia , Espinhas Dendríticas/patologia , Espinhas Dendríticas/fisiologia , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Potenciação de Longa Duração/fisiologia , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos Knockout , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Células Piramidais/patologia , Células Piramidais/fisiopatologia , Comportamento Social , Memória Espacial/fisiologia , Comportamento Estereotipado/fisiologia , Sinapses/patologia , Sinapses/fisiologia , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo
18.
Neurobiol Dis ; 69: 225-34, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24907493

RESUMO

Familial hemiplegic migraine type 1 (FHM1), a monogenic subtype of migraine with aura, is caused by gain-of-function mutations in CaV2.1 (P/Q-type) calcium channels. In FHM1 knockin mice, excitatory neurotransmission at cortical pyramidal cell synapses is enhanced, but inhibitory neurotransmission at connected pairs of fast-spiking (FS) interneurons and pyramidal cells is unaltered, despite being initiated by CaV2.1 channels. The mechanism underlying the unaltered GABA release at cortical FS interneuron synapses remains unknown. Here, we show that the FHM1 R192Q mutation does not affect inhibitory transmission at autapses of cortical FS and other types of multipolar interneurons in microculture from R192Q knockin mice, and investigate the underlying mechanism. Lowering the extracellular [Ca(2+)] did not reveal gain-of-function of evoked transmission neither in control nor after prolongation of the action potential (AP) with tetraethylammonium, indicating unaltered AP-evoked presynaptic calcium influx at inhibitory autapses in FHM1 KI mice. Neither saturation of the presynaptic calcium sensor nor short duration of the AP can explain the unaltered inhibitory transmission in the mutant mice. Recordings of the P/Q-type calcium current in multipolar interneurons in microculture revealed that the current density and the gating properties of the CaV2.1 channels expressed in these interneurons are barely affected by the FHM1 mutation, in contrast with the enhanced current density and left-shifted activation gating of mutant CaV2.1 channels in cortical pyramidal cells. Our findings suggest that expression of specific CaV2.1 channels differentially sensitive to modulation by FHM1 mutations in inhibitory and excitatory cortical neurons underlies the gain-of-function of excitatory but unaltered inhibitory synaptic transmission and the likely consequent dysregulation of the cortical excitatory-inhibitory balance in FHM1.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Canais de Cálcio/metabolismo , Córtex Cerebral/fisiopatologia , Enxaqueca com Aura/fisiopatologia , Inibição Neural/fisiologia , Transmissão Sináptica/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Cálcio/metabolismo , Canais de Cálcio/genética , Canais de Cálcio Tipo N/genética , Técnicas de Cultura de Células , Córtex Cerebral/efeitos dos fármacos , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Interneurônios/efeitos dos fármacos , Interneurônios/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Enxaqueca com Aura/genética , Mutação , Inibição Neural/efeitos dos fármacos , Terminações Pré-Sinápticas/fisiologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiopatologia , Transmissão Sináptica/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo
19.
Eur. j. psychiatry ; 28(2): 114-123, abr.-jun. 2014. ilus, tab
Artigo em Inglês | IBECS | ID: ibc-125149

RESUMO

Background and Objectives: Previously we reported a significant reduction in the numerical density of oligodendrocytes and oligodendrocyte clusters in the inferior parietal lobule (IPL) in schizophrenia that was associated with lack of insight. We also found a significant decrease in the number of perineuronal oligodendrocytes (PnOl) in the prefrontal cortex in schizophrenia and therefore we hypothesized that there may also be a deficit of PnOl in the IPL in schizophrenia and that it could be associated with poor insight. Methods: We estimated the number of PnOl adjacent to pyramidal neurons in layer 3 of BA39 and BA40 in Nissl stained sections from 24 males with schizophrenia and 24 normal male controls from the Stanley Parietal Collection. The schizophrenia group was divided into three subgroups based on level of insight: poor, fair or good.Results: We found a significant deficit of PnOl in layer 3 of BA39 and BA40 in the schizophrenia group as compared to the control group (p<0.01). In the control group but not in the schizophrenia group in BA39 the number of PnOl was significantly higher in the left hemisphere compared to the right hemisphere. In schizophrenia, in BA39 the number of PnOl was decreased in the subgroup with poor insight vs. controls. In BA40 the subgroups with both poor and fair insight were decreased vs. controls (p<0.01). In BA40 the subjects with fair insight also differed from those with good insight (p<0.01). Conclusions: The reduction of PnOl in the IPL in schizophrenia is associated with impaired insight and lack of hemispheric asymmetry (AU)


No disponible


Assuntos
Humanos , Masculino , Oligodendroglia , Lobo Parietal/fisiopatologia , Esquizofrenia/fisiopatologia , Células Piramidais/fisiopatologia , Estudos de Casos e Controles , Tamanho do Órgão , Cérebro/anatomia & histologia
20.
Neurobiol Dis ; 69: 117-23, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24874546

RESUMO

Rare de novo mutations of sodium channels are thought to be an important cause of sporadic epilepsy. The well established role of de novo mutations of sodium channel SCN1A in Dravet Syndrome supports this view, but the etiology of many cases of epileptic encephalopathy remains unknown. We sought to identify the genetic cause in a patient with early onset epileptic encephalopathy by whole exome sequencing of genomic DNA. The heterozygous mutation c. 2003C>T in SCN8A, the gene encoding sodium channel Nav1.6, was detected in the patient but was not present in either parent. The resulting missense substitution, p.Thr767Ile, alters an evolutionarily conserved residue in the first transmembrane segment of channel domain II. The electrophysiological effects of this mutation were assessed in neuronal cells transfected with mutant or wildtype cDNA. The mutation causes enhanced channel activation, with a 10mV depolarizing shift in voltage dependence of activation as well as increased ramp current. In addition, pyramidal hippocampal neurons expressing the mutant channel exhibit increased spontaneous firing with PDS-like complexes as well as increased frequency of evoked action potentials. The identification of this new gain-of-function mutation of Nav1.6 supports the inclusion of SCN8A as a causative gene in infantile epilepsy, demonstrates a novel mechanism for hyperactivity of Nav1.6, and further expands the role of de novo mutations in severe epilepsy.


Assuntos
Epilepsia/genética , Mutação de Sentido Incorreto , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Neurônios/fisiologia , Potenciais de Ação/fisiologia , Idade de Início , Animais , Análise Mutacional de DNA , Epilepsia/fisiopatologia , Células HEK293 , Hipocampo/fisiopatologia , Humanos , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Células Piramidais/fisiopatologia , Ratos , Ratos Sprague-Dawley , Homologia de Sequência de Aminoácidos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...