Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 13765, 2021 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-34215817

RESUMO

The aim of this study was to evaluate the effects of deep brain stimulation of the anterior nucleus of the thalamus (ANT-DBS) on systemic inflammatory responses in patients with drug-resistant epilepsy (DRE). Twenty-two Finnish patients with ANT-DBS implantation were enrolled in this pilot study. Changes in plasma interleukin-6 (IL-6) and interleukin-10 (IL-10) levels were examined using generalized estimating equation models at seven time points (before DBS surgery and 1, 2, 3, 6, 9 and 12 months after implantation). In the whole group, the IL-6/IL-10 ratio decreased significantly over time following ANT-DBS, while the decrease in IL-6 levels and increase in IL-10 levels were not significant. In the responder and nonresponder groups, IL-6 levels remained unchanged during the follow-up. Responders had significantly lower pre-DBS IL-10 levels before the ANT-DBS treatment than nonresponders, but the levels significantly increased over time after the treatment. In addition, responders had a higher pre-DBS IL-6/IL-10 ratio than nonresponders, and the ratio decreased for both groups after treatment, but the decrease did not reach the level of statistical significance. The rate of decrease in the ratio per month tended to be higher in responders than in nonresponders. These results may highlight the anti-inflammatory properties of ANT-DBS treatment associated with its therapeutic effectiveness in patients with DRE. Additional studies are essential to evaluate the potential of the proinflammatory cytokine IL-6, the anti-inflammatory cytokine IL-10, and their ratio as biomarkers to evaluate the therapeutic response to DBS treatment, which could facilitate treatment optimization.


Assuntos
Estimulação Encefálica Profunda , Epilepsia Resistente a Medicamentos/terapia , Interleucina-10/sangue , Interleucina-6/sangue , Adulto , Idoso , Núcleos Anteriores do Tálamo/imunologia , Núcleos Anteriores do Tálamo/metabolismo , Núcleos Anteriores do Tálamo/efeitos da radiação , Citocinas/sangue , Epilepsia Resistente a Medicamentos/sangue , Epilepsia Resistente a Medicamentos/imunologia , Epilepsia Resistente a Medicamentos/fisiopatologia , Estimulação Elétrica , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Estimulação do Nervo Vago/métodos , Adulto Jovem
2.
Cereb Cortex ; 31(4): 2169-2186, 2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33251536

RESUMO

In a changing environment, organisms need to decide when to select items that resemble previously rewarded stimuli and when it is best to switch to other stimulus types. Here, we used chemogenetic techniques to provide causal evidence that activity in the rodent anterior cingulate cortex and its efferents to the anterior thalamic nuclei modulate the ability to attend to reliable predictors of important outcomes. Rats completed an attentional set-shifting paradigm that first measures the ability to master serial discriminations involving a constant stimulus dimension that reliably predicts reinforcement (intradimensional-shift), followed by the ability to shift attention to a previously irrelevant class of stimuli when reinforcement contingencies change (extradimensional-shift). Chemogenetic disruption of the anterior cingulate cortex (Experiment 1) as well as selective disruption of anterior cingulate efferents to the anterior thalamic nuclei (Experiment 2) impaired intradimensional learning but facilitated 2 sets of extradimensional-shifts. This pattern of results signals the loss of a corticothalamic system for cognitive control that preferentially processes stimuli resembling those previously associated with reward. Previous studies highlight a separate medial prefrontal system that promotes the converse pattern, that is, switching to hitherto inconsistent predictors of reward when contingencies change. Competition between these 2 systems regulates cognitive flexibility and choice.


Assuntos
Núcleos Anteriores do Tálamo/metabolismo , Atenção/fisiologia , Giro do Cíngulo/metabolismo , Optogenética/métodos , Recompensa , Adenoviridae/metabolismo , Animais , Núcleos Anteriores do Tálamo/química , Núcleos Anteriores do Tálamo/efeitos dos fármacos , Atenção/efeitos dos fármacos , Aprendizagem por Discriminação/efeitos dos fármacos , Aprendizagem por Discriminação/fisiologia , Giro do Cíngulo/química , Giro do Cíngulo/efeitos dos fármacos , Injeções Intraventriculares , Masculino , Vias Neurais/química , Vias Neurais/efeitos dos fármacos , Vias Neurais/metabolismo , Piperazinas/administração & dosagem , Piperazinas/análise , Piperazinas/metabolismo , Ratos
3.
Brain Res ; 1718: 176-185, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31071305

RESUMO

Non-competitive N-methyl-d-aspartate receptor (NMDA-R) antagonists have been suggested to evoke psychotomimetic-like behaviors by selectively targeting GABAergic elements in cortical and thalamic circuits. In previous studies, we had reported the involvement of the reticular and anterior thalamic nuclei (ATN) in the MK-801-evoked hyperactivity and other motor alterations. Consistent with the possibility that these responses were mediated by thalamic disinhibition, we examined the participation of cortical and hippocampal areas innervated by ATN in the responses elicited by the systemic administration of MK-801 (0.2 mg/kg) and compared them to the effects produced by the microinjection of a subconvulsive dose of bicuculline (GABAA receptor antagonist) in the ATN. We used the expression of Fos related antigen 2 (Fra-2) as a neuronal activity marker in the ATN and its projection areas such as hippocampus (HPC), retrosplenial cortex (RS), entorhinal cortex (EC) and medial prefrontal cortex (mPFC). Dorsal (caudate-putamen, CPu) and ventral striatum (nucleus accumbens, core and shell, NAc,co and NAc,sh) were also studied. Behavioral and brain activation results suggest a partial overlap after the effect of MK-801 administration and ATN disinhibition. MK-801 and ATN disinhibition increases locomotor activity and disorganized movements, while ATN disinhibition also reduces rearing behavior. A significant increase in Fra-2 immunoreactivity (Fra-2-IR) in the ATN, mPFC (prelimbic area, PrL) and NAc,sh was observed after MK-801, while a different pattern of Fra-2-IR was detected following ATN disinhibition (e.g., increase in DG and NAc,sh, and decrease in PrL cortex). Overall, our data may contribute to the understanding of dysfunctional neural circuits involved in schizophrenia.


Assuntos
Núcleos Anteriores do Tálamo/efeitos dos fármacos , Maleato de Dizocilpina/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Núcleos Anteriores do Tálamo/metabolismo , Modelos Animais de Doenças , Antagonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Receptores de GABA-A , Giro do Cíngulo/metabolismo , Hipocampo/metabolismo , Masculino , Neurônios/metabolismo , Núcleo Accumbens/metabolismo , Córtex Pré-Frontal/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Esquizofrenia/metabolismo
4.
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
5.
Brain Res Bull ; 147: 133-139, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30658130

RESUMO

BACKGROUND: Deep brain stimulation (DBS) refers to the delivery of electric current to specific deep brain structures through implanted electrodes. Recently approved for use in United States, DBS to the anterior nucleus of thalamus (ANT) is a safe and effective alternative treatment for medically refractory seizures. Despite the anti-seizure effects of ANT DBS, preclinical and clinical studies have failed to demonstrate it actions at a whole brain level. OBJECTIVE: Here, we used a magnetic resonance imaging (MRI)-based approach in healthy adult rats to investigate the effects of ANT DBS through the circuit of Papez, which has central role in the generation and propagation of limbic seizures, in temporal lobe epilepsy (TLE). METHODS: After ANT electrode implantation and recovery, ANT DBS and SHAM (sham animals had electrodes implanted but were not stimulated) rats received one single injection of the contrast enhancer, manganese chloride (60 mg/kg, ip). Twelve hours after, rats underwent the baseline scan using the MEMRI (Manganese-Enhanced Magnetic Resonance Imaging) technique. We used the same MEMRI and parvalbumin sequence to follow the DBS delivered during 1 h (130 Hz and 200 µA). Perfusion was followed by subsequent c-Fos and parvalbumin immunostaining of brain sections. RESULTS: Acute unilateral ANT DBS significantly reduced the overall manganese uptake and consequently, the MEMRI contrast in the circuit of Papez. Additionally, c-Fos expression was bilaterally increased in the cingulate cortex and posterior hypothalamus, areas directly connected to ANT, as well as in amygdala and subiculum, within the limbic circuitry. CONCLUSION: Our data indicate that MEMRI can be used to detect whole-brain responses to DBS, as the high frequency stimulation parameters used here caused a significant reduction of cell activity in the circuit of Papez that might help to explain the antiepileptic effects of ANT DBS.


Assuntos
Núcleos Anteriores do Tálamo/metabolismo , Convulsões/terapia , Tonsila do Cerebelo/metabolismo , Animais , Núcleo Celular/metabolismo , Estimulação Encefálica Profunda/métodos , Eletrodos Implantados , Epilepsia/metabolismo , Epilepsia/terapia , Epilepsia do Lobo Temporal/terapia , Hipocampo/metabolismo , Sistema Límbico , Imageamento por Ressonância Magnética/métodos , Masculino , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Convulsões/metabolismo , Tálamo/metabolismo
6.
Brain Struct Funct ; 221(4): 1955-70, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-25725627

RESUMO

Lesions to the anterior thalamic nuclei (ATN) in rats produce robust spatial memory deficits that reflect their influence as part of an extended hippocampal system. Recovery of spatial working memory after ATN lesions was examined using a 30-day administration of the neurotrophin cerebrolysin and/or an enriched housing environment. As expected, ATN lesions in standard-housed rats given saline produced severely impaired reinforced spatial alternation when compared to standard-housed rats with sham lesions. Both cerebrolysin and enrichment substantially improved this working memory deficit, including accuracy on trials that required attention to distal cues for successful performance. The combination of cerebrolysin and enrichment was more effective than either treatment alone when the delay between successive runs in a trial was increased to 40 s. Compared to the intact rats, ATN lesions in standard-housed groups produced substantial reduction in c-Fos expression in the retrosplenial cortex, which remained low after cerebrolysin and enrichment treatments. Evidence that multiple treatment strategies restore some memory functions in the current lesion model reinforces the prospect for treatments in human diencephalic amnesia.


Assuntos
Aminoácidos/administração & dosagem , Núcleos Anteriores do Tálamo/fisiologia , Meio Ambiente , Memória de Curto Prazo/fisiologia , Nootrópicos/administração & dosagem , Recuperação de Função Fisiológica , Memória Espacial/fisiologia , Animais , Núcleos Anteriores do Tálamo/efeitos dos fármacos , Núcleos Anteriores do Tálamo/metabolismo , Encéfalo/metabolismo , Feminino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Memória de Curto Prazo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Memória Espacial/efeitos dos fármacos
7.
Neurosci Biobehav Rev ; 54: 131-44, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25195980

RESUMO

Lesions of the rodent anterior thalamic nuclei cause severe deficits to multiple spatial learning tasks. Possible explanations for these effects are examined, with particular reference to T-maze alternation. Anterior thalamic lesions not only impair allocentric place learning but also disrupt other spatial processes, including direction learning, path integration, and relative length discriminations, as well as aspects of nonspatial learning, e.g., temporal discriminations. Working memory tasks, such as T-maze alternation, appear particularly sensitive as they combine an array of these spatial and nonspatial demands. This sensitivity partly reflects the different functions supported by individual anterior thalamic nuclei, though it is argued that anterior thalamic lesion effects also arise from covert pathology in sites distal to the thalamus, most critically in the retrosplenial cortex and hippocampus. This two-level account, involving both local and distal lesion effects, explains the range and severity of the spatial deficits following anterior thalamic lesions. These findings highlight how the anterior thalamic nuclei form a key component in a series of interdependent systems that support multiple spatial functions.


Assuntos
Núcleos Anteriores do Tálamo/fisiologia , Aprendizagem Espacial/fisiologia , Memória Espacial/fisiologia , Animais , Núcleos Anteriores do Tálamo/metabolismo , Núcleos Anteriores do Tálamo/patologia , Córtex Cerebral/fisiologia , Hipocampo/fisiologia , Humanos , Aprendizagem em Labirinto/fisiologia , Camundongos , Rede Nervosa/patologia , Rede Nervosa/fisiologia , Vias Neurais/patologia , Vias Neurais/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos
8.
Endocrinology ; 154(10): 3836-46, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24025225

RESUMO

The principal nucleus of the bed nucleus of the stria terminalis (BNSTp) and anteroventral periventricular nucleus of the hypothalamus (AVPV) are sexually dimorphic, hormone-sensitive forebrain regions. Here we report a profound sex difference in estrogen receptor-α (ERα) immunoreactivity (IR) in the BNSTp, with robust ERα IR in females and the near absence of labeling in males. This sex difference is due to the suppression of ERα IR by testicular hormones in adulthood: it was not present at birth and was not altered by neonatal treatment of females with estradiol; gonadectomy of adult males increased ERα IR to that of females, whereas gonadectomy of adult females had no effect. Treating gonadally intact males with an aromatase inhibitor partially feminized ERα IR in the BNSTp, suggesting that testicular suppression required aromatization. By contrast, in AVPV we found a modest sex difference in ERα IR that was relatively insensitive to steroid manipulations in adulthood. ERα IR in AVPV was, however, masculinized in females treated with estradiol at birth, suggesting that the sex difference is due to organizational effects of estrogens. The difference in ERα IR in the BNSTp of males and females appears to be at least in part due to greater expression of mRNA of the ERα gene (Esr1) in females. The sex difference in message is smaller than the difference in immunoreactivity, however, suggesting that posttranscriptional mechanisms also contribute to the pronounced suppression of ERα IR and presumably to functions mediated by ERα in the male BNSTp.


Assuntos
Núcleos Anteriores do Tálamo/metabolismo , Receptor alfa de Estrogênio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Núcleos Septais/metabolismo , Androgênios/farmacologia , Animais , Animais Recém-Nascidos , Núcleos Anteriores do Tálamo/citologia , Núcleos Anteriores do Tálamo/efeitos dos fármacos , Núcleos Anteriores do Tálamo/crescimento & desenvolvimento , Inibidores da Aromatase/farmacologia , Receptor alfa de Estrogênio/biossíntese , Receptor alfa de Estrogênio/genética , Estrogênios/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/efeitos dos fármacos , Orquiectomia/efeitos adversos , Especificidade de Órgãos , Ovariectomia/efeitos adversos , RNA Mensageiro/metabolismo , Núcleos Septais/citologia , Núcleos Septais/efeitos dos fármacos , Núcleos Septais/crescimento & desenvolvimento , Caracteres Sexuais
9.
Endocrinology ; 154(9): 3273-83, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23825121

RESUMO

Kisspeptin, encoded by Kiss1, stimulates reproduction. In rodents, one Kiss1 population resides in the hypothalamic anterior ventral periventricular nucleus and neighboring rostral periventricular nucleus (AVPV/PeN). AVPV/PeN Kiss1 neurons are sexually dimorphic (greater in females), yet the mechanisms regulating their development and sexual differentiation remain poorly understood. Neonatal estradiol (E2) normally defeminizes AVPV/PeN kisspeptin neurons, but emerging evidence suggests that developmental E2 may also influence feminization of kisspeptin, although exactly when in development this process occurs is unknown. In addition, the obligatory role of GnRH signaling in governing sexual differentiation of Kiss1 or other sexually dimorphic traits remains untested. Here, we assessed whether AVPV/PeN Kiss1 expression is permanently impaired in adult hpg (no GnRH or E2) or C57BL6 mice under different E2 removal or replacement paradigms. We determined that 1) despite lacking GnRH signaling in development, marked sexual differentiation of Kiss1 still occurs in hpg mice; 2) adult hpg females, who lack lifetime GnRH and E2 exposure, have reduced AVPV/PeN Kiss1 expression compared to wild-type females, even after chronic adulthood E2 treatment; 3) E2 exposure to hpg females during the pubertal period does not rescue their submaximal adult Kiss1 levels; and 4) in C57BL6 females, removal of ovarian E2 before the pubertal or juvenile periods does not impair feminization and maximal adult AVPV/PeN Kiss1 expression nor the ability to generate LH surges, indicating that puberty is not a critical period for Kiss1 development. Thus, sexual differentiation still occurs without GnRH, but GnRH or downstream E2 signaling is needed sometime before juvenile development for complete feminization and maximal Kiss1 expression in adult females.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Neurônios/metabolismo , Diferenciação Sexual , Transdução de Sinais , Núcleos Talâmicos/metabolismo , Regulação para Cima , Animais , Núcleos Anteriores do Tálamo/citologia , Núcleos Anteriores do Tálamo/efeitos dos fármacos , Núcleos Anteriores do Tálamo/crescimento & desenvolvimento , Núcleos Anteriores do Tálamo/metabolismo , Estradiol/farmacologia , Estradiol/uso terapêutico , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Terapia de Reposição de Estrogênios , Estrogênios/farmacologia , Estrogênios/uso terapêutico , Feminino , Hormônio Liberador de Gonadotropina/genética , Hipogonadismo/tratamento farmacológico , Hipogonadismo/metabolismo , Hipogonadismo/patologia , Núcleos Intralaminares do Tálamo/citologia , Núcleos Intralaminares do Tálamo/efeitos dos fármacos , Núcleos Intralaminares do Tálamo/crescimento & desenvolvimento , Núcleos Intralaminares do Tálamo/metabolismo , Kisspeptinas/biossíntese , Kisspeptinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Ovariectomia/efeitos adversos , Diferenciação Sexual/efeitos dos fármacos , Desenvolvimento Sexual/efeitos dos fármacos , Núcleos Talâmicos/citologia , Núcleos Talâmicos/efeitos dos fármacos , Núcleos Talâmicos/crescimento & desenvolvimento , Regulação para Cima/efeitos dos fármacos
10.
Peptides ; 45: 85-90, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23651990

RESUMO

Kiss1 mRNA and its corresponding peptide products, kisspeptins, are expressed in two restricted brain areas of rodents, the anteroventral periventricular nucleus (AVPV) and the arcuate nucleus (ARC). The concentration of mature kisspeptins may not directly correlate with Kiss1 mRNA levels, because mRNA translation and/or posttranslational modification, degradation, transportation and release of kisspeptins could be regulated independently of gene expression, and there may thus be differences in kisspeptin expression even in species with similar Kiss1 mRNA profiles. We measured and compared kisspeptin-immunoreactivity in both nuclei and both sexes of rats and mice and quantified kisspeptin-immunoreactive nerve fibers. We also determined Kiss1 mRNA levels and measured kisspeptin-immunoreactivity in colchicine pretreated rats. Overall, we find higher levels of kisspeptin-immunoreactivity in the mouse compared to the rat, independently of brain region and gender. In the female mouse AVPV high numbers of kisspeptin-immunoreactive neurons were present, while in the rat, the female AVPV displays a similar number of kisspeptin-immunoreactive neurons compared to the level of Kiss1 mRNA expressing cells, only after axonal transport inhibition. Interestingly, the density of kisspeptin innervation in the anterior periventricular area was higher in female compared to male in both species. Species differences in the ARC were evident, with the mouse ARC containing dense fibers, while the rat ARC contains clearly discernable cells. In addition, we show a marked sex difference in the ARC, with higher kisspeptin levels in females. These findings show that the translation of Kiss1 mRNA and/or the degradation/transportation/release of kisspeptins are different in mice and rats.


Assuntos
Kisspeptinas/genética , RNA Mensageiro/genética , Animais , Núcleos Anteriores do Tálamo/citologia , Núcleos Anteriores do Tálamo/efeitos dos fármacos , Núcleos Anteriores do Tálamo/metabolismo , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Colchicina/farmacologia , Feminino , Regulação da Expressão Gênica , Imuno-Histoquímica , Kisspeptinas/metabolismo , Masculino , Camundongos , Fibras Nervosas/efeitos dos fármacos , Fibras Nervosas/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Biossíntese de Proteínas , Transporte Proteico , RNA Mensageiro/metabolismo , Ratos , Fatores Sexuais , Especificidade da Espécie
11.
Adv Exp Med Biol ; 784: 275-95, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23550011

RESUMO

Kisspeptin cells appear to be the "missing link," bridging the divide between levels of gonadal steroids and feedback control of gonadotropin-releasing hormone (GnRH) secretion. Kisspeptin neurons are important in the generation of both sex steroid negative and estrogen positive feedback signals to GnRH neurons, the former being involved in the tonic regulation of GnRH secretion in males and females and the latter governing the preovulatory GnRH/luteinizing hormone (LH) surge in females. In rodents, kisspeptin-producing cells populate the anteroventral periventricular nucleus (AVPV) and the arcuate nucleus (ARC), and estrogen regulation of kisspeptin has been extensively studied in these regions. Kisspeptin cells in the ARC appear to receive and forward signals applicable to negative feedback regulation of GnRH. In the female rodent AVPV, kisspeptin cells are important for positive feedback regulation of GnRH and the preovulatory LH surge. In sheep and primates, a rostral population of kisspeptin cells is located in the dorsolateral preoptic area (POA) as well as the ARC. Initial studies showed kisspeptin cells in the latter were involved in both the positive and negative feedback regulation of GnRH. Interestingly, further studies now suggest that kisspeptin cells in the ovine POA may also play an important role in generating estrogen positive feedback. This chapter discusses the current consensus knowledge regarding the interaction between sex steroids and kisspeptin neurons in mammals.


Assuntos
Núcleos Anteriores do Tálamo/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Hormônios Esteroides Gonadais/metabolismo , Kisspeptinas/metabolismo , Área Pré-Óptica/metabolismo , Transdução de Sinais/fisiologia , Animais , Núcleos Anteriores do Tálamo/citologia , Núcleo Arqueado do Hipotálamo/citologia , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Hormônio Luteinizante/metabolismo , Masculino , Neurônios/citologia , Neurônios/metabolismo , Área Pré-Óptica/citologia , Ovinos
12.
Adv Exp Med Biol ; 784: 221-52, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23550009

RESUMO

The neuropeptide kisspeptin, encoded by the Kiss1 gene, is required for mammalian puberty and fertility. Examining the development of the kisspeptin system contributes to our understanding of pubertal progression and adult reproduction and sheds light on possible mechanisms underlying the development of reproductive disorders, such as precocious puberty or hypogonadotropic hypogonadism. Recent work, primarily in rodent models, has begun to study the development of kisspeptin neurons and their regulation by sex steroids and other factors at early life stages. In the brain, kisspeptin is predominantly expressed in two areas of the hypothalamus, the anteroventral periventricular nucleus and neighboring periventricular nucleus (pre-optic area in some species) and the arcuate nucleus. Kisspeptin neurons in these two hypothalamic regions are differentially regulated by testosterone and estradiol, both in development and in adulthood, and also display differences in their degree of sexual dimorphism. In this chapter, we discuss what is currently known and not known about the ontogeny, maturation, and sexual differentiation of kisspeptin neurons, as well as their regulation by sex steroids and other factors during development.


Assuntos
Núcleos Anteriores do Tálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Kisspeptinas/metabolismo , Caracteres Sexuais , Adulto , Animais , Núcleos Anteriores do Tálamo/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Estradiol/metabolismo , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Hipogonadismo/metabolismo , Masculino , Puberdade/fisiologia , Puberdade Precoce/metabolismo , Reprodução/fisiologia , Testosterona/metabolismo
13.
Endocrinology ; 154(1): 363-74, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23150494

RESUMO

The role of norepinephrine (NE) in regulation of LH is still controversial. We investigated the role played by NE in the positive feedback of estradiol and progesterone. Ovarian-steroid control over NE release in the preoptic area (POA) was determined using microdialysis. Compared with ovariectomized (OVX) rats, estradiol-treated OVX (OVX+E) rats displayed lower release of NE in the morning but increased release coincident with the afternoon surge of LH. OVX rats treated with estradiol and progesterone (OVX+EP) exhibited markedly greater NE release than OVX+E rats, and amplification of the LH surge. The effect of NE on LH secretion was confirmed using reverse microdialysis. The LH surge and c-Fos expression in anteroventral periventricular nucleus neurons were significantly increased in OVX+E rats dialyzed with 100 nm NE in the POA. After Fluoro-Gold injection in the POA, c-Fos expression in Fluoro-Gold/tyrosine hydroxylase-immunoreactive neurons increased during the afternoon in the A2 of both OVX+E and OVX+EP rats, in the locus coeruleus (LC) of OVX+EP rats, but was unchanged in the A1. The selective lesion of LC terminals, by intracerebroventricular N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine, reduced the surge of LH in OVX+EP but not in OVX+E rats. Thus, estradiol and progesterone activate A2 and LC neurons, respectively, and this is associated with the increased release of NE in the POA and the magnitude of the LH surge. NE stimulates LH secretion, at least in part, through activation of anteroventral periventricular neurons. These findings contribute to elucidation of the role played by NE during the positive feedback of ovarian steroids.


Assuntos
Núcleos Anteriores do Tálamo/efeitos dos fármacos , Núcleos Anteriores do Tálamo/metabolismo , Hormônio Luteinizante/metabolismo , Norepinefrina/farmacologia , Animais , Cromatografia Líquida de Alta Pressão , Estradiol/farmacologia , Feminino , Imuno-Histoquímica , Microdiálise , Ovariectomia , Progesterona/farmacologia , Radioimunoensaio , Ratos , Ratos Wistar
14.
Proc Natl Acad Sci U S A ; 109(20): E1294-301, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22505735

RESUMO

This study aims to determine the epigenetic mechanism regulating Kiss1 gene expression in the anteroventral periventricular nucleus (AVPV) to understand the mechanism underlying estrogen-positive feedback action on gonadotropin-releasing hormone/gonadotropin surge. We investigated estrogen regulation of the epigenetic status of the mouse AVPV Kiss1 gene locus in comparison with the arcuate nucleus (ARC), in which Kiss1 expression is down-regulated by estrogen. Histone of AVPV Kiss1 promoter region was highly acetylated, and estrogen receptor α was highly recruited at the region by estrogen. In contrast, the histone of ARC Kiss1 promoter region was deacetylated by estrogen. Inhibition of histone deacetylation up-regulated in vitro Kiss1 expression in a hypothalamic non-Kiss1-expressing cell line. Gene conformation analysis indicated that estrogen induced formation of a chromatin loop between Kiss1 promoter and the 3' intergenic region, suggesting that the intergenic region serves to enhance estrogen-dependent Kiss1 expression in the AVPV. This notion was proved, because transgenic reporter mice with a complete Kiss1 locus sequence showed kisspeptin neuron-specific GFP expression in both the AVPV and ARC, but the deletion of the 3' region resulted in greatly reduced GFP expression only in the AVPV. Taken together, these results demonstrate that estrogen induces recruitment of estrogen receptor α and histone acetylation in the Kiss1 promoter region of the AVPV and consequently enhances chromatin loop formation of Kiss1 promoter and Kiss1 gene enhancer, resulting in an increase in AVPV-specific Kiss1 gene expression. These results indicate that epigenetic regulation of the Kiss1 gene is involved in estrogen-positive feedback to generate the gonadotropin-releasing hormone/gonadotropin surge.


Assuntos
Núcleos Anteriores do Tálamo/metabolismo , Epigênese Genética/fisiologia , Estrogênios/metabolismo , Retroalimentação Fisiológica/fisiologia , Regulação da Expressão Gênica/fisiologia , Kisspeptinas/metabolismo , Acetilação , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Sequência de Bases , DNA Intergênico/metabolismo , Epigênese Genética/genética , Hormônio Liberador de Gonadotropina/metabolismo , Proteínas de Fluorescência Verde/genética , Histonas/metabolismo , Kisspeptinas/genética , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Análise de Sequência de DNA
15.
Endocrinology ; 153(6): 2839-50, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22454148

RESUMO

Ovulation in mammals is gated by a master circadian clock in the suprachiasmatic nucleus (SCN). GnRH neurons represent the converging pathway through which the brain triggers ovulation, but precisely how the SCN times GnRH neurons is unknown. We tested the hypothesis that neurons expressing kisspeptin, a neuropeptide coded by the Kiss1 gene and necessary for the activation of GnRH cells during ovulation, represent a relay station for circadian information that times ovulation. We first show that the circadian increase of Kiss1 expression, as well as the activation of GnRH cells, relies on intact ipsilateral neural input from the SCN. Second, by desynchronizing the dorsomedial (dm) and ventrolateral (vl) subregions of the SCN, we show that a clock residing in the dmSCN acts independently of the light-dark cycle, and the vlSCN, to time Kiss1 expression in the anteroventral periventricular nucleus of the hypothalamus and that this rhythm is always in phase with the LH surge. In addition, we show that although the timing of the LH surge is governed by the dmSCN, its amplitude likely depends on the phase coherence between the vlSCN and dmSCN. Our results suggest that whereas dmSCN neuronal oscillators are sufficient to time the LH surge through input to kisspeptin cells in the anteroventral periventricular nucleus of the hypothalamus, the phase coherence among dmSCN, vlSCN, and extra-SCN oscillators is critical for shaping it. They also suggest that female reproductive disorders associated with nocturnal shift work could emerge from the desynchronization between subregional oscillators within the master circadian clock.


Assuntos
Ritmo Circadiano/fisiologia , Expressão Gênica , Kisspeptinas/genética , Hormônio Luteinizante/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Núcleos Anteriores do Tálamo/citologia , Núcleos Anteriores do Tálamo/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Kisspeptinas/metabolismo , Masculino , Neurônios/metabolismo , Ovariectomia , Fotoperíodo , Ratos , Ratos Wistar , Núcleo Supraquiasmático/citologia
16.
Biol Reprod ; 86(4): 119, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22278983

RESUMO

Fibroblast growth factor (FGF) signaling is essential for the development of the gonadotropin-releasing hormone (GnRH) system. Mice harboring deficiencies in Fgf8 or Fgf receptor 1 (Fgfr1) suffer a significant loss of GnRH neurons, but their reproductive phenotypes have not been examined. This study examined if female mice hypomorphic for Fgf8, Fgfr1, or both (compound hypomorphs) exhibited altered parameters of pubertal onset, estrous cyclicity, and fertility. Further, we examined the number of kisspeptin (KP)-immunoreactive (ir) neurons in the anteroventral periventricular/periventricular nuclei (AVPV/PeV) of these mice to assess if changes in the KP system, which stimulates the GnRH system, could contribute to the reproductive phenotypes. Single hypomorphs (Fgfr1(+/-) or Fgf8(+/-)) had normal timing for vaginal opening (VO) but delayed first estrus. However, after achieving the first estrus, they underwent normal expression of estrous cycles. In contrast, the compound hypomorphs underwent early VO and normal first estrus, but had disorganized estrous cycles that subsequently reduced their fertility. KP immunohistochemistry on Postnatal Day 15, 30, and 60 transgenic female mice revealed that female compound hypomorphs had significantly more KP-ir neurons in the AVPV/PeV compared to their wild-type littermates, suggesting increased KP-ir neurons may drive early VO but could not maintain the cyclic changes in GnRH neuronal activity required for female fertility. Overall, these data suggest that Fgf signaling deficiencies differentially alter the parameters of female pubertal onset and cyclicity. Further, these deficiencies led to changes in the AVPV/PeV KP-ir neurons that may have contributed to the accelerated VO in the compound hypomorphs.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Infertilidade Feminina/metabolismo , Kisspeptinas/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Reprodução/fisiologia , Animais , Núcleos Anteriores do Tálamo/metabolismo , Comunicação Celular/fisiologia , Ciclo Estral/metabolismo , Feminino , Fator 8 de Crescimento de Fibroblasto/genética , Hormônio Liberador de Gonadotropina/metabolismo , Camundongos , Camundongos Transgênicos , Núcleos da Linha Média do Tálamo/metabolismo , Neurônios/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Maturidade Sexual/fisiologia , Transdução de Sinais/fisiologia
17.
Neuroscience ; 169(3): 1255-67, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20570608

RESUMO

There is growing evidence that lesions of the anterior thalamic nuclei cause long-lasting intrinsic changes to retrosplenial cortex, with the potential to alter its functional properties. The present study had two goals. The first was to identify the pattern of changes in eight markers, as measured by in-situ hydridisation, in the granular retrosplenial cortex (area Rgb) following anterior thalamic lesions. The second was to use retrograde trans-neuronal tracing methods to identify the potential repercussions of intrinsic changes within granular retrosplenial cortex. In Experiment 1, adult rats received unilateral lesions of the anterior thalamic nuclei and were perfused 4 weeks later. Of the eight markers, four (c-fos, zif268, 5ht2rc, kcnab2) showed a very similar pattern of change, with decreased levels in superficial retrosplenial cortex (lamina II) in the ipsilateral hemisphere but little or no change in deeper layers (lamina V). A fifth marker (cox6b) showed a shift in activity levels in the opposite direction to the previous four markers. Three other markers (cox6a1, CD74, ncs-1) did not appear to change activity levels after surgery. The predominant pattern of change, a decrease in superficial cortical activity, points to potential alterations in plasticity and metabolism. In Experiment 2, wheat germ agglutin (WGA) was injected into the anterior thalamic nuclei in rats given different survival times, sometimes in combination with the retrograde, fluorescent tracer, Fast Blue. Dense aggregations of retrogradely labeled cells were always found in lamina VI of granular retrosplenial cortex, but additional labeled cells in lamina II were only found: (1) in WGA cases, that is never after Fast Blue injections, and (2) after longer WGA survival times (3 days). These layer II Rgb cells are likely to have been trans-neuronally labeled, revealing a pathway from lamina II of Rgb to those deeper retrosplenial cells that project directly to the anterior thalamic nuclei.


Assuntos
Núcleos Anteriores do Tálamo/efeitos dos fármacos , Córtex Cerebral/metabolismo , Giro do Cíngulo/metabolismo , N-Metilaspartato/toxicidade , Animais , Núcleos Anteriores do Tálamo/metabolismo , Núcleos Anteriores do Tálamo/patologia , Biomarcadores/metabolismo , Contagem de Células , Hibridização In Situ , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Ratos
18.
Endocrinology ; 151(5): 2223-32, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20211970

RESUMO

Neurons in the rodent anteroventral periventricular nucleus (AVPV) play a key role in integrating circadian and gonadal steroid hormone information in the control of fertility. In particular, estradiol-sensitive kisspeptin neurons located in the AVPV, and adjacent structures [together termed the rostral periventricular area of the third ventricle (RP3V)], are critical for puberty onset and the preovulatory LH surge. The present study aimed to establish the morphological and electrical firing characteristics of RP3V neurons, including kisspeptin neurons, in the adult female mouse. Cell-attached electrical recordings, followed by juxtacellular dye filling, of 129 RP3V neurons in the acute brain slice preparation revealed these cells to exhibit multipolar (53%), bipolar (43%), or unipolar (4%) dendritic morphologies along with silent (16%), irregular (41%), bursting (25%), or tonic (34%) firing patterns. Postrecording immunocytochemistry identified 17 of 100 filled RP3V cells as being kisspeptin neurons, all of which exhibited complex multipolar dendritic trees and significantly (P < 0.05) higher bursting or high tonic firing rates compared with nonkisspeptin neurons. The firing pattern of RP3V neurons fluctuated across the estrous cycle with a significant (P < 0.05) switch from irregular to tonic firing patterns found on proestrus. A similar nonsignificant trend was found for kisspeptin neurons. All RP3V neurons responded to gamma-aminobutyric acid and glutamate, about 10% to RFamide-related peptide-3, about 5% to vasopressin, 0% to vasoactive intestinal peptide, and 0% to kisspeptin. These studies provide a morphological and electrical description of AVPV/RP3V neurons and demonstrate their cycle-dependent firing patterns along with an unexpected lack of acute response to the circadian neuropeptides.


Assuntos
Dendritos/fisiologia , Neurônios/fisiologia , Terceiro Ventrículo/citologia , Proteínas Supressoras de Tumor/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Núcleos Anteriores do Tálamo/citologia , Núcleos Anteriores do Tálamo/metabolismo , Relação Dose-Resposta a Droga , Ciclo Estral/fisiologia , Feminino , GABAérgicos/farmacologia , Glutamatos/farmacologia , Hipotálamo Anterior/citologia , Hipotálamo Anterior/metabolismo , Kisspeptinas , Camundongos , Camundongos Endogâmicos C57BL , Núcleos da Linha Média do Tálamo/citologia , Núcleos da Linha Média do Tálamo/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Neuropeptídeos/farmacologia , Terceiro Ventrículo/metabolismo , Peptídeo Intestinal Vasoativo/farmacologia , Vasopressinas/farmacologia , Ácido gama-Aminobutírico/farmacologia
19.
Int J Dev Neurosci ; 27(6): 567-74, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19563881

RESUMO

There is increasing evidence that early adverse experience contributes to the development of stress susceptibility, and increases the onset of stress-related psychiatric disorders in stressful environments in adulthood. This study addressed whether or not prolonged maternal separation, a well-established model of early stress, affects adult limbic areas related to the regulation of the hypothalamic-pituitary-adrenal axis in exposure to chronic variable stress in adulthood. Rats were subjected to daily maternal separation for 4.5h during postnatal days 1-21. As adults, the animals were exposed to a variable chronic stress paradigm of 24 days. Persistent changes were assessed in glucocorticoid receptor density and Fos activity in the anterodorsal thalamic nuclei, mammillary nuclei and retrosplenial cortex. Immunohistochemical analysis revealed that adult maternally separated animals had increased levels of c-Fos immunoreactivity in the anterodorsal thalamic nuclei as well as in the mammillary nuclei compared to normal non-maternally separated animals. Chronic variable stress in maternally separated and non-maternally separated animals diminished glucocorticoid receptor density in the anterodorsal thalamic nuclei but not in the rest of the nuclei analyzed. These results indicate that c-Fos immunoreactivity as well as glucocorticoid receptor expression in the anterodorsal thalamic nuclei and mammillary nuclei exhibit long-term alterations in adult rats following repeated maternal separation and subsequent stress exposure. Recognition of these adaptations helps to define the brain regions and neural circuitry associated with persistent alterations induced by early life environment and the development of stress-associated disorders.


Assuntos
Encéfalo/fisiopatologia , Sistema Límbico/fisiopatologia , Privação Materna , Transtornos Neurocognitivos/fisiopatologia , Receptores de Glucocorticoides/metabolismo , Estresse Psicológico/fisiopatologia , Adaptação Fisiológica/fisiologia , Animais , Animais Recém-Nascidos , Núcleos Anteriores do Tálamo/metabolismo , Núcleos Anteriores do Tálamo/fisiopatologia , Biomarcadores/análise , Biomarcadores/metabolismo , Encéfalo/metabolismo , Doença Crônica , Modelos Animais de Doenças , Feminino , Giro do Cíngulo/metabolismo , Giro do Cíngulo/fisiopatologia , Imuno-Histoquímica , Sistema Límbico/metabolismo , Corpos Mamilares/metabolismo , Corpos Mamilares/fisiopatologia , Vias Neurais/metabolismo , Vias Neurais/fisiopatologia , Transtornos Neurocognitivos/etiologia , Transtornos Neurocognitivos/metabolismo , Proteínas Proto-Oncogênicas c-fos/análise , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Estresse Psicológico/complicações , Estresse Psicológico/metabolismo
20.
J Neurosci ; 29(29): 9390-5, 2009 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-19625529

RESUMO

Kisspeptin is a product of the Kiss1 gene and is expressed in the forebrain. Neurons that express Kiss1 play a crucial role in the regulation of pituitary luteinizing hormone secretion and reproduction. These neurons are the direct targets for the action of estradiol-17beta (E(2)), which acts via the estrogen receptor alpha isoform (ER alpha) to regulate Kiss1 expression. In the arcuate nucleus (Arc), where the dynorphin gene (Dyn) is expressed in Kiss1 neurons, E(2) inhibits the expression of Kiss1 mRNA. However, E(2) induces the expression of Kiss1 in the anteroventral periventricular nucleus (AVPV). The mechanism for differential regulation of Kiss1 in the Arc and AVPV by E(2) is unknown. ER alpha signals through multiple pathways, which can be categorized as either classical, involving the estrogen response element (ERE), or nonclassical, involving ERE-independent mechanisms. To elucidate the molecular basis for the action of E(2) on Kiss1 and Dyn expression, we studied the effects of E(2) on Kiss1 and Dyn mRNAs in the brains of mice bearing targeted alterations in the ER alpha signaling pathways. We found that stimulation of Kiss1 expression by E(2) in the AVPV and inhibition of Dyn in the Arc required an ERE-dependent pathway, whereas the inhibition of Kiss1 expression by E(2) in the Arc involved ERE-independent mechanisms. Thus, distinct ER alpha signaling pathways can differentially regulate the expression of identical genes across different brain regions, and E(2) can act within the same neuron through divergent ER alpha signaling pathways to regulate different neurotransmitter genes.


Assuntos
Encéfalo/efeitos dos fármacos , Dinorfinas/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Proteínas/metabolismo , Animais , Núcleos Anteriores do Tálamo/efeitos dos fármacos , Núcleos Anteriores do Tálamo/metabolismo , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Encéfalo/metabolismo , Dinorfinas/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Introdução de Genes , Kisspeptinas , Hormônio Luteinizante/sangue , Camundongos , Camundongos Transgênicos , Núcleos da Linha Média do Tálamo/efeitos dos fármacos , Núcleos da Linha Média do Tálamo/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas/genética , RNA Mensageiro/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA