Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neurobiol Learn Mem ; 162: 15-22, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31047996

RESUMO

Reciprocal connections between the mediodorsal thalamic nucleus (MD) and the prefrontal cortex (PFC) are important for memory processes. Since the co-abuse of nicotine and ethanol affects memory formation, this study investigated the effect of nitric oxide inhibition in the MD on memory retrieval induced by co-administration of nicotine and ethanol. Subsequently, western blot analysis was used to evaluate how this change would alter the PFC pCREB/CREB signaling pathway. Male Wistar rats were bilaterally cannulated into the MD and the memory retrieval was measured by passive avoidance task. Intraperitoneal (i.p.) administration of ethanol (1 g/kg, i.p) 30 min before the test impaired memory retrieval and caused ethanol-induced amnesia. Subcutaneous (s.c.) administration of nicotine (0.05-0.2 mg/kg, s.c.) prevented ethanol-induced amnesia and improved memory retrieval. Intra-MD microinjection of a nitric oxide synthase (NOS) inhibitor, L-NAME (0.5-1 µg/rat) inhibited the improving effect of nicotine (0.2 mg/kg, s.c.) on ethanol-induced amnesia, while intra-MD microinjection of a precursor of nitric oxide, l-arginine (0.25-1 µg/rat), potentiated such effect. Noteworthy, intra-MD microinjection of the same doses of L-NAME or l-arginine by itself had no effect on memory retrieval. Furthermore, intra-MD microinjection of L-NAME (0.05, 0.1 and 0.3 µg/rat) reversed the l-arginine improving effect on nicotine response. Successful memory retrieval significantly increased the p-CREB/CREB ratio in the PFC tissue. Ethanol-induced amnesia, however, decreased this ratio in the PFC while the co-administration of nicotine and ethanol increased the PFC CREB signaling. Interestingly, the inhibitory effect of L-NAME and the potentiating effect of l-arginine on nicotine response were associated with the decrease and increase of the PFC p-CREB/CREB ratio respectively. It can be concluded that MD-PFC connections are involved in the combined effects of nicotine and ethanol on memory retrieval. The mediodorsal thalamic NO system possibly mediated this interaction via the pCREB/CREB signaling pathways in the PFC.


Assuntos
Etanol/farmacologia , Núcleo Mediodorsal do Tálamo/efeitos dos fármacos , Rememoração Mental/efeitos dos fármacos , Nicotina/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Córtex Pré-Frontal/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Masculino , Núcleo Mediodorsal do Tálamo/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Agonistas Nicotínicos/farmacologia , Óxido Nítrico/metabolismo , Fosforilação/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Wistar
2.
Exp Brain Res ; 237(6): 1397-1407, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30887077

RESUMO

A cellular degeneration of two thalamic nuclei belonging to the "limbic thalamus", i.e., the anteroventral (AV) and mediodorsal (MD) nuclei, has been shown in patients suffering from Fatal Familial Insomnia (FFI), a lethal prion disease characterized by autonomic activation and severe insomnia. To better assess the physiological role of these nuclei in autonomic and sleep regulation, c-Fos expression was measured in rats during a prolonged exposure to low ambient temperature (Ta, - 10 °C) and in the first hours of the subsequent recovery period at normal laboratory Ta (25 °C). Under this protocol, the thermoregulatory and autonomic activation led to a tonic increase in waking and to a reciprocal depression in sleep occurrence, which was more evident for REM sleep. These effects were followed by a clear REM sleep rebound and by a rebound of Delta power during non-REM sleep in the following recovery period. In the anterior thalamic nuclei, c-Fos expression was (1) larger during the activity rather than the rest period in the baseline; (2) clamped at a level in-between the normal daily variation during cold exposure; (3) not significantly affected during the recovery period in comparison to the time-matched baseline. No significant changes were observed in either the MD or the paraventricular thalamic nucleus, which is also part of the limbic thalamus. The observed changes in the activity of the anterior thalamic nuclei appear, therefore, to be more specifically related to behavioral activation than to autonomic or sleep regulation.


Assuntos
Núcleos Anteriores do Tálamo/metabolismo , Sistema Nervoso Autônomo/fisiologia , Regulação da Temperatura Corporal/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fases do Sono/fisiologia , Vigília/fisiologia , Animais , Eletroencefalografia , Masculino , Núcleo Mediodorsal do Tálamo/metabolismo , Núcleos da Linha Média do Tálamo/metabolismo , Ratos , Ratos Sprague-Dawley , Sono REM/fisiologia , Sono de Ondas Lentas/fisiologia
3.
Pharmacol Res Perspect ; 7(1): e00457, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30784207

RESUMO

Deficiencies in N-methyl-d-aspartate (NMDA)/glutamate receptor (NMDAR) signaling have been considered central to the cognitive impairments of schizophrenia; however, an NMDAR antagonist memantine (MEM) improves cognitive impairments of Alzheimer's disease and schizophrenia. These mechanisms of paradoxical clinical effects of NMDAR antagonists remain unclear. To explore the mechanisms by which MK801 and MEM affect thalamocortical transmission, we determined interactions between local administrations of MK801, MEM, system xc- (Sxc), and metabotropic glutamate receptors (mGluRs) on extracellular glutamate and GABA levels in the mediodorsal thalamic nucleus (MDTN) and medial prefrontal cortex (mPFC) using dual-probe microdialysis with ultra-high-pressure liquid chromatography. Effects of MK801 and MEM on Sxc activity were also determined using primary cultured astrocytes. Sxc activity was enhanced by MEM, but was unaffected by MK801. MK801 enhanced thalamocortical glutamatergic transmission by GABAergic disinhibition in the MDTN. In the MDTN and the mPFC, MEM weakly increased glutamate release by activating Sxc, whereas MEM inhibited thalamocortical glutamatergic transmission. Paradoxical effects of MEM were induced following secondary activation of inhibitory II-mGluR and III-mGluR by exporting glutamate from astroglial Sxc. The present results suggest that the effects of therapeutically relevant concentrations of MEM on thalamocortical glutamatergic transmission are predominantly caused by activation of Sxc rather than inhibition of NMDAR. These demonstrations suggest that the combination between reduced NMDAR and activated Sxc contribute to the neuroprotective effects of MEM. Furthermore, activation of Sxc may compensate for the cognitive impairments that are induced by hyperactivation of thalamocortical glutamatergic transmission following activation of Sxc/II-mGluR in the MDTN and Sxc/II-mGluR/III-mGluR in the mPFC.


Assuntos
Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Memantina/farmacologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Células Cultivadas , Cromatografia Líquida de Alta Pressão/métodos , Ácido Glutâmico/metabolismo , Masculino , Núcleo Mediodorsal do Tálamo/metabolismo , Microdiálise/métodos , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Tálamo/metabolismo
4.
Brain Struct Funct ; 222(6): 2527-2545, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28150086

RESUMO

Early postnatal damage to the mediodorsal thalamus (MD) produces deficits in cognition and behavior believed to be associated with early prefrontal cortical maldevelopment. We assessed the role of MD afferents during development on the morphological and functional maturation of the prefrontal cortex (PFC) and the basolateral amygdala (BLA). Sprague-Dawley rat pups (n = 56) received a bilateral electrolytic lesion of the MD or a MD Sham lesion on postnatal day 4. 7 weeks later, all rats were tested in anxiety-related and cognitive paradigms using the elevated plus maze and novel object recognition tests. Following behavioral testing (P70), rats were killed and the baseline expression of C-Fos protein and the number of GABAergic neurons were evaluated in the PFC and the BLA. The dendritic morphology and spine density in the PFC using Golgi-Cox staining was also evaluated. Adult rats with early postnatal bilateral MD damage exhibited disrupted recognition memory and increased anxiety-like behaviors. The lesion also caused a significant diminution of C-Fos immunolabeling and an increase of the number of GABAergic neurons in the PFC. In the BLA, the number of GABAergic neurons was significantly reduced, associated with an increase in C-Fos immunolabeling. Furthermore, in the PFC the lesion induced a significant reduction in dendritic branching and spine density. Our data are consistent with the hypothesis that the MD plays a role in the development of the PFC and, therefore, may be a good animal model to investigate cognitive symptoms associated with schizophrenia.


Assuntos
Complexo Nuclear Basolateral da Amígdala/fisiopatologia , Comportamento Animal , Núcleo Mediodorsal do Tálamo/fisiopatologia , Córtex Pré-Frontal/fisiopatologia , Esquizofrenia/fisiopatologia , Animais , Animais Recém-Nascidos , Ansiedade/patologia , Ansiedade/fisiopatologia , Ansiedade/psicologia , Complexo Nuclear Basolateral da Amígdala/metabolismo , Complexo Nuclear Basolateral da Amígdala/patologia , Cognição , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Estimulação Elétrica , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/patologia , Aprendizagem em Labirinto , Núcleo Mediodorsal do Tálamo/metabolismo , Núcleo Mediodorsal do Tálamo/patologia , Vias Neurais/metabolismo , Vias Neurais/patologia , Vias Neurais/fisiopatologia , Neurônios Aferentes/metabolismo , Neurônios Aferentes/patologia , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Sprague-Dawley , Reconhecimento Psicológico , Esquizofrenia/metabolismo , Esquizofrenia/patologia , Psicologia do Esquizofrênico , Ácido gama-Aminobutírico/metabolismo
5.
Aging Cell ; 14(6): 1045-54, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26268661

RESUMO

Mice in which the genes for growth hormone (GH) or GH receptor (GHR(-/-) ) are disrupted from conception are dwarfs, possess low levels of IGF-1 and insulin, have low rates of cancer and diabetes, and are extremely long-lived. Median longevity is also increased in mice with deletion of hypothalamic GH-releasing hormone (GHRH), which leads to isolated GH deficiency. The remarkable extension of longevity in hypopituitary Ames dwarf mice can be reversed by a 6-week course of GH injections started at the age of 2 weeks. Here, we demonstrate that mutations that interfere with GH production or response, in the Snell dwarf, Ames dwarf, or GHR(-/-) mice lead to reduced formation of both orexigenic agouti-related peptide (AgRP) and anorexigenic proopiomelanocortin (POMC) projections to the main hypothalamic projection areas: the arcuate nucleus (ARH), paraventricular nucleus (PVH), and dorsomedial nucleus (DMH). These mutations also reduce hypothalamic inflammation in 18-month-old mice. GH injections, between 2 and 8 weeks of age, reversed both effects in Ames dwarf mice. Disruption of GHR specifically in liver (LiGHRKO), a mutation that reduces circulating IGF-1 but does not lead to lifespan extension, had no effect on hypothalamic projections or inflammation, suggesting an effect of GH, rather than peripheral IGF-1, on hypothalamic development. Hypothalamic leptin signaling, as monitored by induction of pStat3, is not impaired by GHR deficiency. Together, these results suggest that early-life disruption of GH signaling produces long-term hypothalamic changes that may contribute to the longevity of GH-deficient and GH-resistant mice.


Assuntos
Proteínas de Transporte/genética , Nanismo Hipofisário/genética , Hormônio do Crescimento/farmacologia , Fator de Crescimento Insulin-Like I/metabolismo , Insulina/metabolismo , Longevidade/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Nanismo Hipofisário/metabolismo , Hormônio do Crescimento/genética , Inflamação/patologia , Leptina/metabolismo , Fígado/metabolismo , Masculino , Núcleo Mediodorsal do Tálamo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Núcleo Hipotalâmico Paraventricular/metabolismo , Pró-Opiomelanocortina/metabolismo , Fator de Transcrição STAT3/biossíntese
6.
J Chem Neuroanat ; 52: 20-4, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23660497

RESUMO

This study explores the regions activated by deep brain stimulation of the mediodorsal thalamic nucleus through examination of immediate early genes as markers of neuronal activation. Stimulation was delivered unilaterally with constant current 100 µs duration pulses at a frequency of 130 Hz delivered at an amplitude of 200 µA for 3h. Brains were removed, sectioned and radio-labelled for the IEGs zif-268 and c-fos. In anaesthetised rats, deep brain stimulation of mediodorsal thalamic nucleus produced robust increases in the expression of zif-268 but not c-fos localised to regions that are reciprocally connected with the mediodorsal thalamic nucleus, including the prelimbic and orbitofrontal cortices, and the premotor cortex indicating an increase in synaptic activity in these regions. These findings map those brain regions that are persistently, rather than transiently, activated by high frequency electrical stimulation of the mediodorsal thalamic nucleus by a putatively antidromic mechanism which may be relevant to neuropsychiatric disorders such as schizophrenia in which thalamocortical systems are disrupted and in which DBS protocols are being considered.


Assuntos
Estimulação Encefálica Profunda/métodos , Proteína 1 de Resposta de Crescimento Precoce/biossíntese , Lobo Frontal/metabolismo , Genes fos/fisiologia , Núcleo Mediodorsal do Tálamo/metabolismo , Animais , Masculino , Vias Neurais/metabolismo , Ratos
7.
Proc Natl Acad Sci U S A ; 108(11): 4459-64, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21368141

RESUMO

Recent findings suggest that extrasynaptic δ-subunit-containing GABA(A) receptors are sensitive to low-to-moderate concentrations of alcohol, raising the possibility that these receptors mediate the reinforcing effects of alcohol after consumption of one or a few drinks. We used the technique of viral-mediated RNAi to reduce expression of the GABA(A) receptor δ-subunit in adult rats in localized regions of the nucleus accumbens (NAc) to test the hypothesis that δ-subunit-containing GABA(A) receptors in the NAc are necessary for oral alcohol consumption. We found that knockdown of the δ-subunit in the medial shell region of the NAc, but not in the ventral or lateral shell or in the core, reduced alcohol intake. In contrast, δ-subunit knockdown in the medial shell did not affect intake of a 2% sucrose solution, suggesting that the effects of GABA(A) receptor δ-subunit reduction are specific to alcohol. These results provide strong evidence that extrasynaptic δ-subunit-containing GABA(A) receptors in the medial shell of the NAc are critical for the reinforcing effects of oral ethanol.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Núcleo Mediodorsal do Tálamo/metabolismo , Núcleo Accumbens/metabolismo , Receptores de GABA-A/metabolismo , Sinapses/metabolismo , Adenoviridae/genética , Administração Oral , Animais , Técnicas de Silenciamento de Genes , Masculino , Núcleo Mediodorsal do Tálamo/patologia , Núcleo Accumbens/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Long-Evans , Receptores de GABA-A/genética , Reprodutibilidade dos Testes , Sacarose/metabolismo
9.
J Biol Rhythms ; 25(6): 421-31, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21135158

RESUMO

Internal synchrony among external cycles and internal oscillators allows adaptation of physiology to cyclic demands for homeostasis. Night work and shift work lead to a disrupted phase relationship between external time cues and internal rhythms, also losing internal coherence among oscillations. This process results in internal desynchrony (ID) in which behavioral, hormonal, and metabolic variables cycle out of phase. It is still not clear whether ID originates at a peripheral or at a central level. In order to determine the possible role of hypothalamic oscillators in ID, we explored with a rat model of "night work" daily rhythms of activity and clock gene expression in the hypothalamus. This study provides evidence that wakefulness and activity during the normal resting phase lead to a shift in the diurnal rhythms of c-Fos and induce a rhythm of PER1 in the arcuate and dorsomedial nucleus of the hypothalamus, both associated with metabolism and regulation of the sleep/wake cycle. Moreover, the number of orexin (ORX)-positive neurons and c-Fos in the perifornical area increased during the working period, suggesting a relevant switch of activity in this brain region induced by the scheduled activity; however, the colocalization of c-Fos in ORX-positive cells was not increased. In contrast, the suprachiasmatic nucleus and the paraventricular nucleus remained locked to the light/dark cycle, resulting in ID in the hypothalamus. Present data suggest that ID occurs already at the level of the first output projections from the SCN, relaying nuclei that transmit temporal signals to other brain areas and to the periphery.


Assuntos
Ritmo Circadiano/fisiologia , Hipotálamo/metabolismo , Proteínas Circadianas Period/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Análise de Variância , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Ingestão de Alimentos/fisiologia , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Núcleo Mediodorsal do Tálamo/metabolismo , Microscopia Confocal , Modelos Animais , Atividade Motora/fisiologia , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Orexinas , Fotoperíodo , Ratos , Ratos Wistar , Núcleo Supraquiasmático/metabolismo
10.
Dev Psychobiol ; 48(5): 360-7, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16770763

RESUMO

Otsuka Long Evans Tokushima Fatty (OLETF) rats have a deletion in the gene encoding the cholecystokinin-1 (CCK1) receptor. This deletion prevents protein expression, making the OLETF rat a CCK1 receptor knockout model. Consistent with the absence of CCK1 receptors, OLETF rats do not reduce their food intake in response to exogenously administered CCK and consume larger than normal meals. This deficit in within-meal feedback signaling is evident in liquid as well as solid meals. Neonatal OLETF rats show similar differences in independent ingestion tests. Intake is higher and is reflected in greater licking behavior. Neonatal OLETF rats also have diminished latencies to consume and higher initial ingestion rats. Adult OLETF rats are hyperphagic and obese. Although arcuate nucleus peptide gene expression is apparently normal in OLETF rats, when obesity is prevented through pair-feeding to amounts consumed by control Long Evans Tokushima Otsuka (LETO) rats, dorsomedial hypothalamic NPY mRNA expression is significantly elevated in OLETF rats. NPY overexpression is also evident in preobese, juvenile OLETF rats suggesting a causal role for this overexpression in the hyperphagia and obesity. Running wheel exercise normalizes food intake and body weight in OLETF rats. When access to exercise is provided at a time when OLETF rats are obese, the effects are limited to the period of exercise. When running wheel access is available to younger, preobese OLETF rats, exercise results in long lasting reductions in food intake and body weight and improved glucose regulation. These lasting metabolic effects of exercise may be secondary to an exercise induced reduction in DMH NPY mRNA expression.


Assuntos
Colecistocinina/administração & dosagem , Hiperfagia/genética , Obesidade/genética , Receptores da Colecistocinina/genética , Animais , Colecistocinina/farmacocinética , Ingestão de Energia , Comportamento Alimentar , Deleção de Genes , Hiperfagia/metabolismo , Núcleo Mediodorsal do Tálamo/metabolismo , Obesidade/metabolismo , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos OLETF , Receptores da Colecistocinina/metabolismo
11.
Am J Physiol Regul Integr Comp Physiol ; 289(2): R359-R366, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15879054

RESUMO

Orexin A is produced in caudal lateral, posterior, perifornical, and dorsomedial hypothalamic areas. Orexin A in the rostro-dorsal lateral hypothalamic area (rLHa) stimulates feeding and activates several feeding-regulatory brain areas. We hypothesized that aging diminishes feeding and c-Fos-immunoreactivity (c-Fos-ir; marker of neuronal activation) response to orexin A. Young (3 mo), middle-aged (12 mo), and old (24 mo) male Fischer 344 rLHa-cannulated rats were injected with orexin A (0.5, 1, and 2 nmol). Food intake was measured at 1, 2, and 4 h. c-Fos-ir in hypothalamic, limbic, and hindbrain regions was measured in two additional sets of rLHa-orexin A injected rats. In a separate study, orexin A effects on feeding and c-Fos-ir were measured in 6-mo-old rats. Orexin A significantly elevated feeding in rats aged 3, 6, and 12 mo in the 0-1 and 1-2- h time intervals, whereas in old rats this was significant in the 1-2 h time interval only. At 1 h, 6-8 (of 14) brain areas showed elevated c-Fos-ir in response to orexin A in 3- and 6-mo-old rats, but 24-mo-old rats exhibited attenuated or absent c-Fos-ir response in all brain regions except the hypothalamic paraventricular nucleus (PVN) and rostral nucleus of the solitary tract (rNTS). Orexin A did not elevate c-Fos-ir in 3-mo-old rats at 2 h after injection, whereas the PVN and mediodorsal thalamic nucleus (MD) showed elevated c-Fos-ir at 2 h in 24-mo-old rats. These data suggest that delayed and diminished feeding responses in old animals may be due to ineffective neural signaling and implicate the orexin A network as one feeding system affected by aging.


Assuntos
Envelhecimento/fisiologia , Encéfalo/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Neuropeptídeos/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Encéfalo/citologia , Imuno-Histoquímica , Masculino , Núcleo Mediodorsal do Tálamo/metabolismo , Neurônios/metabolismo , Orexinas , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Ratos Endogâmicos F344 , Fatores de Tempo
12.
J Neurosci ; 21(10): 3593-9, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11331388

RESUMO

Status epilepticus (StE) in immature rats causes long-term functional impairment. Whether this is associated with structural alterations remains controversial. The present study was designed to test the hypothesis that StE at an early age results in neuronal loss. StE was induced with lithium-pilocarpine in 12-d-old rats, and the presence of neuronal damage was investigated in the brain from 12 hr up to 1 week later using silver and Fluoro-Jade B staining techniques. Analysis of the sections indicated consistent neuronal damage in the central and lateral segments of the mediodorsal nucleus of the thalamus, which was confirmed using adjacent cresyl violet-stained preparations. The mechanism of thalamic damage (necrosis vs apoptosis) was investigated further using TUNEL, immunohistochemistry for caspase-3 and cytochrome c, and electron microscopy. Activated microglia were detected using OX-42 immunohistochemistry. The presence of silver and Fluoro-Jade B-positive degenerating neurons in the mediodorsal thalamic nucleus was associated with the appearance of OX-42-immunopositive activated microglia but not with the expression of markers of programmed cell death, caspase-3, or cytochrome c. Electron microscopy revealed necrosis of the ultrastructure of damaged neurons, providing further evidence that the mechanism of StE-induced damage in the mediodorsal thalamic nucleus at postnatal day 12 is necrosis rather than apoptosis. Finally, these data together with previously described functions of the medial and lateral segments of the mediodorsal thalamic nucleus suggest that some functions, such as adaptation to novelty, might become compromised after StE early in development.


Assuntos
Antígenos CD , Antígenos de Neoplasias , Proteínas Aviárias , Proteínas Sanguíneas , Núcleo Mediodorsal do Tálamo/patologia , Estado Epiléptico/patologia , Animais , Antígenos de Superfície/metabolismo , Apoptose , Basigina , Caspase 3 , Caspases/metabolismo , Grupo dos Citocromos c/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Cloreto de Lítio , Masculino , Núcleo Mediodorsal do Tálamo/metabolismo , Glicoproteínas de Membrana/metabolismo , Microglia/metabolismo , Microglia/patologia , Microscopia Eletrônica , Necrose , Neurônios/metabolismo , Neurônios/patologia , Pilocarpina , Ratos , Ratos Wistar , Estado Epiléptico/induzido quimicamente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA