Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Physiol Behav ; 194: 199-204, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29775631

RESUMO

In mammals, anorexia accompanying infection is thought to be mediated via cytokines including interleukins, interferons (IFNs), and tumor necrosis factor (TNF). However, there is a lack of related knowledge on birds. Therefore, the purpose of the present study was to determine if cytokines are associated with reduced food intake in chicks (Gallus gallus). Specifically, we evaluated the effects of TNF-like cytokine 1A (TL1A), a member of the TNF family, interferon-α (IFN-α), and interferon-γ (IFN-γ) on food intake. Additionally, the effect of lipopolysaccharide (LPS) and polyinosinic:polycytidylic acid (poly I:C) on cytokine mRNA expression in the diencephalon and spleen was also measured. Intracerebroventricular (ICV) injection of 0.05 or 0.5 µg TL1A, IFN-α, and IFN-γ had no effect on food intake. However, when 1.0 µg each of these factors was evaluated, TL1A significantly decreased food intake at 180 and 240 min after the injection, but IFN-α and IFN-γ had no effect. When chicks received intraperitoneal (IP) injections of 100 µg LPS or 400 µg poly I:C, their food intake was reduced. Diencephalic mRNA expression of TL1A was significantly decreased following IP injection of LPS or poly I:C. Additionally, diencephalic mRNA expression of IFN-γ mRNA was significantly increased by IP injection of LPS but decreased by IP injection of poly I:C. For the spleen, IP injection of LPS and poly I:C both significantly increased TL1A and IFN-γ mRNA expression. In sum, we have provided evidence that central TL1A but not IFN-α or IFN-γ are related to reduction of food intake in chicks, but the role of these cytokines for mediating anorexia associated with infections may differ from mammals.


Assuntos
Anorexia/imunologia , Ingestão de Alimentos/imunologia , Interferon-alfa/metabolismo , Interferon gama/metabolismo , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Anorexia/etiologia , Galinhas , Diencéfalo/efeitos dos fármacos , Diencéfalo/imunologia , Ingestão de Alimentos/efeitos dos fármacos , Comportamento de Doença/fisiologia , Interferon-alfa/administração & dosagem , Interferon gama/administração & dosagem , Lipopolissacarídeos , Masculino , Poli I-C , RNA Mensageiro/metabolismo , Baço/efeitos dos fármacos , Baço/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/administração & dosagem
2.
Physiol Behav ; 164(Pt A): 268-76, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27317836

RESUMO

Recently, it was found that the avian central vasotocin receptor (V1aR) is associated with the regulation of food intake. To identify V1aR-containing brain structures regulating food intake, a selective V1aR antagonist SR-49059 that induced food intake was administrated intracerebroventricularly in male chickens followed by detection of brain structures using FOS immunoreactivity. Particularly, the hypothalamic core region of the paraventricular nucleus, lateral hypothalamic area, dorsomedial hypothalamic nucleus, a subnucleus of the central extended amygdalar complex [dorsolateral bed nucleus of the stria terminalis], medial septal nucleus and caudal brainstem [nucleus of the solitary tract] showed significantly increased FOS-ir cells. On the other hand, the supraoptic nucleus of the preoptic area and the nucleus of the hippocampal commissure of the septum showed suppressed FOS immunoreactivity in the V1aR antagonist treatment group. Further investigation revealed that neuronal activity of arginine vasotocin (AVT-ir) magnocellular neurons in the supraoptic nucleus, preoptic periventricular nucleus, paraventricular nucleus and ventral periventricular hypothalamic nucleus and most likely corticotropin releasing hormone (CRH-ir) neurons in the nucleus of the hippocampal commissure were reduced following the antagonist treatment. Dual immunofluorescence labeling results showed that perikarya of AVT-ir magnocellular neurons in the preoptic area and hypothalamus were colabeled with V1aR. Within the nucleus of the hippocampal commissure, CRH-ir neurons were shown in close contact with V1aR-ir glial cells. Results of the present study suggest that the V1aR plays a role in the regulation of food intake by modulating neurons that synthesize and release anorectic neuropeptides in the avian brain.


Assuntos
Regulação do Apetite/fisiologia , Proteínas Aviárias/metabolismo , Diencéfalo/metabolismo , Ingestão de Alimentos/fisiologia , Receptores de Vasopressinas/metabolismo , Septo do Cérebro/metabolismo , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Regulação do Apetite/efeitos dos fármacos , Comportamento Apetitivo/efeitos dos fármacos , Comportamento Apetitivo/fisiologia , Proteínas Aviárias/antagonistas & inibidores , Fármacos do Sistema Nervoso Central/administração & dosagem , Galinhas , Diencéfalo/citologia , Diencéfalo/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Indóis/farmacologia , Masculino , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeo Y/administração & dosagem , Proteínas Proto-Oncogênicas c-fos/metabolismo , Pirrolidinas/farmacologia , Distribuição Aleatória , Septo do Cérebro/citologia , Septo do Cérebro/efeitos dos fármacos
3.
Dev Dyn ; 245(6): 667-77, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27005305

RESUMO

BACKGROUND: Antioxidants such as the green tea polyphenol epigallocatechin gallate (EGCG) are neuroprotective under many conditions in mature nervous systems; however, their impact has rarely been explored in developing nervous systems, in which a critical step is the formation of connections between neurons. Axons emerge from newly formed neurons and are led by a dynamic structure found at their tip called a growth cone. Here we explore the impact of EGCG on the development of retinal ganglion cell (RGC) axons, which connect the eye to the brain. RESULTS: EGCG acts directly on RGC axons to increase the number of growth cone filopodia, fingerlike projections that respond to extrinsic signals, in vitro and in vivo. Furthermore, EGCG exposure leads to a dramatic defect in the guided growth of RGC axons where the axons fail to make a key turn in the mid-diencephalon required to reach their target. Intriguingly, at guidance points where RGCs do not show a change in direction, EGCG has no influence on RGC axon behavior. CONCLUSIONS: We propose that EGCG stabilizes filopodia and prevents normal filopodial dynamics required for axons to change their direction of outgrowth at guidance decision points. Developmental Dynamics 245:667-677, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Axônios/efeitos dos fármacos , Axônios/metabolismo , Catequina/análogos & derivados , Cones de Crescimento/efeitos dos fármacos , Pseudópodes/efeitos dos fármacos , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/efeitos dos fármacos , Animais , Orientação de Axônios/efeitos dos fármacos , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Catequina/farmacologia , Gonadotropina Coriônica/farmacologia , Diencéfalo/citologia , Diencéfalo/efeitos dos fármacos , Embrião não Mamífero/citologia , Embrião não Mamífero/efeitos dos fármacos , Feminino , Humanos , Hibridização In Situ , Neurogênese/efeitos dos fármacos , Retina/citologia , Retina/efeitos dos fármacos , Xenopus
4.
Mol Cell Endocrinol ; 420: 75-84, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26586207

RESUMO

In the frog Pelophylax esculentus, the endocannabinoid anandamide (AEA) modulates Gonadotropin Releasing Hormone (GnRH) system in vitro and down-regulates steroidogenic enzymes in vivo. Thus, male frogs were injected with AEA ± SR141716A, a cannabinoid receptor 1 (CB1) antagonist, to evaluate possible effects on GnRH and Kiss1/Gpr54 systems, gonadotropin receptors and steroid levels. In frog diencephalons, AEA negatively affected both GnRH and Kiss1/Gpr54 systems. In testis, AEA induced the expression of gonadotropin receptors, cb1, gnrh2 and gnrhr3 meanwhile reducing gnrhr2 mRNA and Kiss1/Gpr54 proteins. Furthermore, aromatase (Cyp19) expression increased in parallel to testosterone decrease and estradiol increase. In vitro treatment of testis with AEA revealed direct effects on Cyp19 and induced the expression of the AEA-degrading enzyme Faah. Lastly, AEA effects on Faah were counteracted by the antiestrogen ICI182780, indicating estradiol mediated effect. In conclusion, for the first time we show in a vertebrate that AEA regulates testicular activity through kisspeptin system.


Assuntos
Ácidos Araquidônicos/farmacologia , Endocanabinoides/farmacologia , Kisspeptinas/metabolismo , Alcamidas Poli-Insaturadas/farmacologia , Rana esculenta/metabolismo , Testículo/metabolismo , Amidoidrolases/metabolismo , Animais , Aromatase/metabolismo , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Estradiol/metabolismo , Masculino , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptores da Gonadotropina/metabolismo , Rimonabanto , Testosterona/metabolismo
5.
Pharmacol Res ; 103: 328-39, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26657418

RESUMO

Histone deacetylases (HDACs) are key epigenetic enzymes and emerging drug targets in cancer and neurodegeneration. Pan-HDAC inhibitors provided neuroprotection in Parkinson's Disease (PD) models, however, the HDAC isoforms with highest neuroprotective potential remain unknown. Zebrafish larvae (powerful pharmacological testing tools bridging cellular and in vivo studies) have thus far been used in PD modelling with limited phenotypic characterization. Here we characterize the behavioural and metabolic phenotypes of a zebrafish PD model induced with MPP(+), assess the feasibility of targeting zebrafish HDAC1 and HDAC6 isoforms, and test the in vivo effects of their selective inhibitors MS-275 and tubastatin A, respectively. MPP(+) induced a concentration-dependent decrease in metabolic activity and sensorimotor reflexes, and induced locomotor impairments rescuable by the dopaminergic agonist apomorphine. Zebrafish HDAC1 and HDAC6 isoforms show high sequence identity with mammalian homologues at the deacetylase active sites, and pharmacological inhibition increased acetylation of their respective histone and tubulin targets. MS-275 and tubastatin rescued the MPP(+)-induced decrease in diencephalic tyrosine hydroxylase immunofluorescence and in whole-larvae metabolic activity, without modifying mitochondrial complex activity or biogenesis. MS-275 or tubastatin alone modulated spontaneous locomotion. When combined with MPP(+), however, neither MS-275 nor tubastatin rescued locomotor impairments, although tubastatin did ameliorate the head-reflex impairment. This study demonstrates the feasibility of pharmacologically targeting the zebrafish HDAC1 and HDAC6 isoforms, and indicates that their inhibition can rescue cellular metabolism in a PD model. Absence of improvement in locomotion, however, suggests that monotherapy with either HDAC1 or HDAC6 inhibitors is unlikely to provide strong benefits in PD. This study highlights parameters dependent on the integrity of zebrafish neuronal circuits as a valuable complement to cell-based studies. Also, the demonstrated feasibility of pharmacologically targeting HDAC1 and HDAC6 in this organism paves the way for future studies investigating HDAC inhibitors in other diseases modelled in zebrafish.


Assuntos
Benzamidas/farmacologia , Histona Desacetilase 1/antagonistas & inibidores , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Indóis/farmacologia , Doença de Parkinson Secundária , Piridinas/farmacologia , Proteínas de Peixe-Zebra/antagonistas & inibidores , 1-Metil-4-fenilpiridínio , Animais , Comportamento Animal/efeitos dos fármacos , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Modelos Animais de Doenças , Histona Desacetilase 1/genética , Desacetilase 6 de Histona , Histona Desacetilases/genética , Larva , Locomoção/efeitos dos fármacos , Neurotoxinas , Oxazinas/metabolismo , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/tratamento farmacológico , Doença de Parkinson Secundária/metabolismo , Doença de Parkinson Secundária/fisiopatologia , RNA Mensageiro/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Xantenos/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
6.
Gen Comp Endocrinol ; 195: 9-20, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24505600

RESUMO

The first studies that identified leptin and its receptor (LepR) in mammals were based on mutant animals that displayed dramatic changes in body-weight and regulation of energy homeostasis. Subsequent studies have shown that a deficiency of leptin or LepR in homoeothermic mammals results in hyperphagia, obesity, infertility and a number of other abnormalities. The physiological roles of leptin-mediated signaling in ectothermic teleosts are still being explored. Here, we produced medaka with homozygous LepR gene mutation using the targeting induced local lesions in a genome method. This knockout mutant had a point mutation of cysteine for stop codon at the 357th amino acid just before the leptin-binding domain. The evidence for loss of function of leptin-mediated signaling in the mutant is based on a lack of response to feeding in the expression of key appetite-related neuropeptides in the diencephalon. The mutant lepr−/− medaka expressed constant up-regulated levels of mRNA for the orexigenic neuropeptide Ya and agouti-related protein and a suppressed level of anorexigenic proopiomelanocortin 1 in the diencephalon independent of feeding, which suggests that the mutant did not possess functional LepR. Phenotypes of the LepR-mutant medaka were analyzed in order to understand the effects on food intake, growth, and fat accumulation in the tissues. The food intake of the mutant medaka was higher in post-juveniles and adult stages than that of wild-type (WT) fish. The hyperphagia led to a high growth rate at the post-juvenile stage, but did not to significant alterations in final adult body size. There was no additional deposition of fat in the liver and muscle in the post-juvenile and adult mutants, or in the blood plasma in the adult mutant. However, adult LepR mutants possessed large deposits of visceral fat, unlike in the WT fish, in which there were none. Our analysis confirms that LepR in medaka exert a powerful influence on the control on food intake. Further analyses using the mutant will contribute to a better understanding of the role of leptin in fish. This is the first study to produce fish with leptin receptor deficiency.


Assuntos
Animais Geneticamente Modificados/crescimento & desenvolvimento , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Técnicas de Inativação de Genes , Gordura Intra-Abdominal/efeitos dos fármacos , Neuropeptídeos/farmacologia , Receptores para Leptina/fisiologia , Proteína Relacionada com Agouti/metabolismo , Animais , Animais Geneticamente Modificados/metabolismo , Apetite/efeitos dos fármacos , Apetite/fisiologia , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Hiperfagia/genética , Hiperfagia/patologia , Leptina/metabolismo , Mutação/genética , Obesidade/metabolismo , Oryzias/genética , Oryzias/crescimento & desenvolvimento , Oryzias/metabolismo , Regulação para Cima
7.
Gen Comp Endocrinol ; 199: 86-93, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24472706

RESUMO

In sheep as in rat, it has been highly suggested that neuronal histamine (HA) participates to the estradiol (E2)-induced GnRH and LH surges, through H1 receptor. With the aim of determining if E2 could act directly on HA neurons, we examined here whether HA neurons express estrogen receptor alpha (ERα) in the ewe diencephalon during the breeding season. We first produced a specific polyclonal antibody directed against recombinant ovine histidine decarboxylase (oHDC), the HA synthesizing enzyme. Using both this anti-oHDC antibody and an anti-ERα monoclonal antibody in double label immunohistochemistry, we showed that HA neurons do not express ERα in diencephalon of ewes with different hormonal status. This result diverges from those obtained in rat, in which around three quarters of HA neurons express ERα in their nucleus. This discrepancy between these two mammal species may reflect difference in their neuronal network.


Assuntos
Cruzamento , Diencéfalo/metabolismo , Receptor alfa de Estrogênio/metabolismo , Histamina/metabolismo , Neurônios/metabolismo , Estações do Ano , Ovinos/metabolismo , Animais , Anticorpos/metabolismo , Especificidade de Anticorpos/imunologia , Diencéfalo/citologia , Diencéfalo/efeitos dos fármacos , Diencéfalo/enzimologia , Eletroforese em Gel de Poliacrilamida , Estradiol/farmacologia , Feminino , Histidina Descarboxilase/imunologia , Imuno-Histoquímica , Masculino , Neurônios/efeitos dos fármacos , Ratos , Proteínas Recombinantes/isolamento & purificação
8.
J Steroid Biochem Mol Biol ; 141: 37-43, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24434281

RESUMO

As one of the third generation of aromatase inhibitors, letrozole is a favored drug for the treatment of hormone receptor-positive breast cancer with some adverse effects on the nervous system, but the knowledge is limited and the results are controversial, the mechanism underlying its central action is also unclear. Accumulated evidences have demonstrated that estrogens derived from androgens by aromatase play profound roles in the brain through their receptors, which needs coactivator for the transcription regulation, among which steroid receptor coactivator-1 (SRC-1) has been shown to be multifunctional potentials in the brain, but whether it is regulated by letrozole is currently unknown. In this study, we examined letrozole regulation on SRC-1 expression in adult mice brain using immunohistochemistry. The results showed that letrozole induced dramatic decrease of SRC-1 in the medial septal, hippocampus, medial habenular nucleus, arcuate hypothalamic nucleus and superior colliculus (p<0.01). Significant decrease was detected in the dorsal lateral septal nucleus, bed nucleus of stria terminalis, ventral taenia tecta, dorsomedial and ventromedial hypothalamic nuclei, dorsomedial periaqueductal gray, superior paraolivary nucleus and pontine nucleus (p<0.05). In the hippocampus, levels of estradiol content, androgen receptor, estrogen receptor α and ß also decreased significantly after letrozole injection. The above results demonstrated letrozole downregulation of SRC-1 in specific regions that are primarily related to learning and memory, cognition and mood, neuroendocrine as well as information integration, indicating that SRC-1 may be one important downstream central target of letrozole. Furthermore, these potential central adverse effects of letrozole should be taken into serious considerations.


Assuntos
Inibidores da Aromatase/farmacologia , Regulação para Baixo/efeitos dos fármacos , Hipocampo/metabolismo , Nitrilas/farmacologia , Coativador 1 de Receptor Nuclear/metabolismo , Triazóis/farmacologia , Animais , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/metabolismo , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Hipocampo/efeitos dos fármacos , Letrozol , Masculino , Memória/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Córtex Motor/efeitos dos fármacos , Córtex Motor/metabolismo , Sistemas Neurossecretores/efeitos dos fármacos , Sistemas Neurossecretores/metabolismo , Coativador 1 de Receptor Nuclear/genética , Especificidade de Órgãos , Receptores Androgênicos/metabolismo , Telencéfalo/efeitos dos fármacos , Telencéfalo/metabolismo
9.
Neuroscience ; 260: 106-19, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24361177

RESUMO

Brain injury due to neonatal hypoxia-ischemia (HI) is more homogenously severe in male than in female mice. Because, necrostatin-1 (nec-1) prevents injury progression only in male mice, we hypothesized that changes in brain-derived neurotrophic factor (BDNF) signaling after HI and nec-1 are also sex-specific providing differential conditions to promote recovery of those more severely injured. The increased aromatization of testosterone in male mice during early development and the link between 17-ß-estradiol (E2) levels and BDNF transcription substantiate this hypothesis. Hence, we aimed to investigate if sexual differences in BDNF signaling existed in forebrain and diencephalon after HI and HI/nec-1 and their correlation with estrogen receptors (ER). C57B6 mice (p7) received nec-1 (0.1µl [8µM]) or vehicle (veh) intracerebroventricularly after HI. At 24h after HI, BDNF levels increased in both sexes in forebrain without evidence of tropomyosin-receptor-kinase B (TrkB) activation. At 96h after HI, BDNF levels in forebrain decreased below those seen in control mice of both sexes. Additionally, only in female mice, truncated TrkB (Tc.TrkB) and p75 neurotrophic receptor (p75ntr) levels increased in forebrain and diencephalon. In both, forebrain and diencephalon, nec-1 treatment increased BDNF levels and TrkB activation in male mice while, nec-1 prevented Tc.TrkB and p75ntr increases in female mice. While E2 levels were unchanged by HI or HI/nec-1 in either sex or treatment, ERα:ERß ratios were increased in diencephalon of nec-1-treated male mice and directly correlated with BDNF levels. Neonatal HI produces sex-specific signaling changes in the BDNF system, that are differentially modulated by nec-1. The regional differences in BDNF levels may be a consequence of injury severity after HI, but sexual differences in response to nec-1 after HI may represent a differential thalamo-cortical preservation or alternatively off-target regional effect of nec-1. The biological significance of ERα predominance and its correlation with BDNF levels is still unclear.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Diencéfalo/metabolismo , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Imidazóis/uso terapêutico , Indóis/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Prosencéfalo/metabolismo , Animais , Animais Recém-Nascidos , Diencéfalo/efeitos dos fármacos , Estradiol/análise , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Parvalbuminas/metabolismo , Prosencéfalo/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Fatores Sexuais
10.
Endocrinology ; 154(6): 2114-28, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23554453

RESUMO

The sulfated neurosteroids pregnenolone sulfate (Δ(5)PS) and dehydroepiandrosterone sulfate (DHEAS) are known to play a role in the control of reproductive behavior. In the frog Pelophylax ridibundus, the enzyme hydroxysteroid sulfotransferase (HST), responsible for the biosynthesis of Δ(5)PS and DHEAS, is expressed in the magnocellular nucleus and the anterior preoptic area, two hypothalamic regions that are richly innervated by GnRH1-containing fibers. This observation suggests that GnRH1 may regulate the formation of sulfated neurosteroids to control sexual activity. Double labeling of frog brain slices with HST and GnRH1 antibodies revealed that GnRH1-immunoreactive fibers are located in close vicinity of HST-positive neurons. The cDNAs encoding 3 GnRH receptors (designated riGnRHR-1, -2, and -3) were cloned from the frog brain. RT-PCR analyses revealed that riGnRHR-1 is strongly expressed in the hypothalamus and the pituitary whereas riGnRHR-2 and -3 are primarily expressed in the brain. In situ hybridization histochemistry indicated that GnRHR-1 and GnRHR-3 mRNAs are particularly abundant in preoptic area and magnocellular nucleus whereas the concentration of GnRHR-2 mRNA in these 2 nuclei is much lower. Pulse-chase experiments using tritiated Δ(5)P and DHEA as steroid precursors, and 3'-phosphoadenosine 5'-phosphosulfate as a sulfonate moiety donor, showed that GnRH1 stimulates, in a dose-dependent manner, the biosynthesis of Δ(5)PS and DHEAS in frog diencephalic explants. Because Δ(5)PS and DHEAS, like GnRH, stimulate sexual activity, our data strongly suggest that some of the behavioral effects of GnRH could be mediated via the modulation of sulfated neurosteroid production.


Assuntos
Sulfato de Desidroepiandrosterona/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Pregnenolona/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Perfilação da Expressão Gênica , Hormônio Liberador de Gonadotropina/farmacologia , Hibridização In Situ , Masculino , Microscopia Confocal , Dados de Sequência Molecular , Neurônios/metabolismo , Hipófise/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ranidae , Receptores LHRH/genética , Receptores LHRH/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Sulfotransferases/metabolismo
11.
Age (Dordr) ; 35(3): 821-37, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22648398

RESUMO

Estrogens are not only critical for sexual differentiation it is well-known for the role of 17ß-estradiol (E2) in the adult brain modulating memory, learning, mood and acts as a neuroprotector. E2 exerts its actions through two classical receptors: estrogen receptor alpha (ERα) and estrogen receptor beta (ERß). The distribution of both receptors changes from one brain area to another, E2 being able to modulate their expression. Among the classical features of aging in humans, we find cognitive impairment, dementia, memory loss, etc. As estrogen levels change with age, especially in females, it is important to know the effects of low E2 levels on ERα distribution; results from previous studies are controversial regarding this issue. In the present work, we have studied the effects of long-term E2 depletion as well as the ones of E2 treatment on ERα brain distribution of ovariectomized rats along aging in the diencephalon and in the telencephalon. We have found that ovariectomy causes downregulation and affects subcellular localization of ERα expression during aging, meanwhile prolonged estrogen treatment produces upregulation and overexpression of the receptor levels. Our results support the idea of the region-specific neuroprotection mechanisms mediated by estradiol.


Assuntos
Envelhecimento/metabolismo , Química Encefálica/fisiologia , Encéfalo/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Terapia de Reposição Hormonal , Espaço Intracelular/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Córtex Cerebral/química , Córtex Cerebral/citologia , Densitometria , Diencéfalo/química , Diencéfalo/citologia , Diencéfalo/efeitos dos fármacos , Estrogênios/farmacologia , Feminino , Imuno-Histoquímica , Espaço Intracelular/efeitos dos fármacos , Ratos , Ratos Wistar , Telencéfalo/química , Telencéfalo/citologia , Telencéfalo/efeitos dos fármacos
12.
J Biomed Sci ; 19: 51, 2012 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-22613782

RESUMO

BACKGROUND: Higher aluminum (Al) content in infant formula and its effects on neonatal brain development are a cause for concern. This study aimed to evaluate the distribution and concentration of Al in neonatal rat brain following Al treatment, and oxidative stress in brain tissues induced by Al overload. METHODS: Postnatal day 3 (PND 3) rat pups (n =46) received intraperitoneal injection of aluminum chloride (AlCl3), at dosages of 0, 7, and 35 mg/kg body wt (control, low Al (LA), and high Al (HA), respectively), over 14 d. RESULTS: Aluminum concentrations were significantly higher in the hippocampus (751.0 ± 225.8 ng/g v.s. 294.9 ± 180.8 ng/g; p < 0.05), diencephalon (79.6 ± 20.7 ng/g v.s. 20.4 ± 9.6 ng/g; p < 0.05), and cerebellum (144.8 ± 36.2 ng/g v.s. 83.1 ± 15.2 ng/g; p < 0.05) in the HA group compared to the control. The hippocampus, diencephalon, cerebellum, and brain stem of HA animals displayed significantly higher levels of lipid peroxidative products (TBARS) than the same regions in the controls. However, the average superoxide dismutase (SOD) activities in the cerebral cortex, hippocampus, cerebellum, and brain stem were lower in the HA group compared to the control. The HA animals demonstrated increased catalase activity in the diencephalon, and increased glutathione peroxidase (GPx) activity in the cerebral cortex, hippocampus, cerebellum, and brain stem, compared to controls. CONCLUSION: Aluminum overload increases oxidative stress (H2O2) in the hippocampus, diencephalon, cerebellum, and brain stem in neonatal rats.


Assuntos
Alumínio/administração & dosagem , Desenvolvimento Embrionário/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Alumínio/metabolismo , Animais , Mapeamento Encefálico , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/metabolismo , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Ratos , Distribuição Tecidual
13.
J Pharmacol Exp Ther ; 342(2): 245-54, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22532626

RESUMO

Chronic nicotine produces up-regulation of α4ß2* nicotinic acetylcholine receptors (nAChRs) (* denotes that an additional subunit may be part of the receptor). However, the extent of up-regulation to persistent ligand exposure varies across brain regions. The aim of this work was to study the cellular distribution and function of nAChRs after chronic nicotine treatment in primary cultures of mouse brain neurons. Initially, high-affinity [(125)I]epibatidine binding to cell membrane homogenates from primary neuronal cultures obtained from diencephalon and hippocampus of C57BL/6J mouse embryos (embryonic days 16-18) was measured. An increase in α4ß2*-nAChR binding sites was observed in hippocampus, but not in diencephalon, after 24 h of treatment with 1 µM nicotine. However, a nicotine dose-dependent up-regulation of approximately 3.5- and 0.4-fold in hippocampus and diencephalon, respectively, was found after 96 h of nicotine treatment. A significant fraction of total [(125)I]epibatidine binding sites in both hippocampus (45%) and diencephalon (65%) was located on the cell surface. Chronic nicotine (96 h) up-regulated both intracellular and surface binding in both brain regions without changing the proportion of those binding sites compared with control neurons. The increase in surface binding was not accompanied by an increase in nicotine-stimulated Ca(2+) influx, suggesting persistent desensitization or inactivation of receptors at the plasma membrane occurred. Given the differences observed between hippocampus and diencephalon neurons exposed to nicotine, multiple mechanisms may play a role in the regulation of nAChR expression and function.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Nicotina/farmacologia , Piridinas/metabolismo , Receptores Nicotínicos/metabolismo , Alquilação/efeitos dos fármacos , Animais , Sítios de Ligação , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Cálcio/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Radioisótopos do Iodo/análise , Camundongos , Camundongos Endogâmicos C57BL , Piridinas/farmacologia , Regulação para Cima/efeitos dos fármacos
14.
J Comp Neurol ; 520(10): 2143-62, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22173890

RESUMO

Kisspeptin is essential for reproductive functions in humans. As a model for the human we have used the female guinea pig, which has a long ovulatory cycle similar to that of primates. Initially, we cloned a guinea pig kisspeptin cDNA sequence and subsequently explored the distribution and 17ß-estradiol (E2) regulation of kisspeptin mRNA (Kiss1) and protein (kisspeptin) by using in situ hybridization, real-time PCR and immunocytochemistry. In ovariectomized females, Kiss1 neurons were scattered throughout the preoptic periventricular areas (PV), but the vast majority of Kiss1 neurons were localized in the arcuate nucleus (Arc). An E2 treatment that first inhibits (negative feedback) and then augments (positive feedback) serum luteinizing hormone (LH) increased Kiss1 mRNA density and number of cells expressing Kiss1 in the PV at both time points. Within the Arc, Kiss1 mRNA density was reduced at both time points. Quantitative real-time PCR confirmed the in situ hybridization results during positive feedback. E2 reduced the number of immunoreactive kisspeptin cells in the PV at both time points, perhaps an indication of increased release. Within the Arc, the kisspeptin immunoreactivity was decreased during negative feedback but increased during positive feedback. Therefore, it appears that in guinea pig both the PV and the Arc kisspeptin neurons act cooperatively to excite gonadotropin-releasing hormone (GnRH) neurons during positive feedback. We conclude that E2 regulation of negative and positive feedback may reflect a complex interaction of the kisspeptin circuitry, and both the PV and the Arc respond to hormone signals to encode excitation of GnRH neurons during the ovulatory cycle.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Expressão Gênica/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Kisspeptinas/metabolismo , Análise de Variância , Animais , Clonagem Molecular , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Cobaias , Hipotálamo/anatomia & histologia , Hipotálamo/metabolismo , Kisspeptinas/genética , Ovariectomia , RNA Mensageiro/metabolismo
15.
J Neuroendocrinol ; 22(2): 110-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20002963

RESUMO

Kisspeptin (Kiss) is a key regulator of reproductive function in both prepubertal and adult mammals. Its expression appears to vary throughout the year in seasonal species. We aimed to determine the impact of a change of photoperiod on the size of Kiss neuronal populations found in the preoptic area (POA) and arcuate nucleus (ARC) of the ewe brain. Using immunocytochemistry, we first examined the proportion of neurones expressing Kiss, using HuC/D as a neuronal marker, at different time-points after transition from long days (LD; 16 : 8 h light/dark cycle) to short days (SD; 8 : 16 h light/dark cycle). Luteinising hormone (LH) secretion was measured in ovariectomised oestradiol replaced ewes from the month preceding the transition to SD until the sacrifice of the animals at days 0, 45 and 112 from this photoperiodic transition. High LH levels were only observed in animals killed at day 112. The number of Kiss neurones/mm(2) doubled in the caudal ARC at day 112. The percentage of neurones showing Kiss immunoreactivity increased significantly in both the POA and ARC in the day 112 group. In a second experiment, ewes kept in LD received an i.c.v. injection of colchicine 20 h before sacrifice. Colchicine treatment increased the number and the percentage of neurones with Kiss in both the POA and caudal ARC. The data obtained suggest that the increase in Kiss neurones detected in the POA and caudal ARC after transition to SD stemmed from an increase in Kiss synthesis. This up-regulation of Kiss content under the shorter day condition appears to be a late event within the cascade activated by a longer secretion of melatonin, which is a critical factor in switching gonadotrophin-releasing hormone secretion to a breeding season profile.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Neurônios/metabolismo , Fotoperíodo , Área Pré-Óptica/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Contagem de Células , Fármacos do Sistema Nervoso Central/farmacologia , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/fisiologia , Colchicina/farmacologia , Escuridão , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Estradiol/metabolismo , Feminino , Imuno-Histoquímica , Hormônio Luteinizante/sangue , Hormônio Luteinizante/metabolismo , Neurônios/efeitos dos fármacos , Ovariectomia , Área Pré-Óptica/efeitos dos fármacos , Ovinos , Fatores de Tempo
16.
J Toxicol Sci ; 34(3): 315-25, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19483385

RESUMO

Methylmercury (MeHg) is an environmental pollutant known to cause neurobehavioral defects and is especially toxic to the developing brain. With recent studies showing that fetal exposure to low-dose MeHg causes developmental abnormalities, it is therefore important to find ways to combat its effects as well as to clarify the mechanism(s) underlying MeHg toxicity. In the present study, the effects of MeHg on cultured neural progenitor cells (NPC) derived from mouse embryonic brain were investigated. We first confirmed the vulnerability of embryonic NPC to MeHg toxicity, NPC from the telencephalon were more sensitive to MeHg compared to those from the diencephalon. Buthionine sulfoximine (BSO) which is known to inhibit glutathione synthesis accelerated MeHg toxicity. Furthermore, antioxidants such as N-acetyl cysteine and alpha-tocopherol dramatically rescued the NPC from MeHg's toxic effects. Interestingly, a 12 hr delay in the addition of either antioxidant was still able to prevent the cells from undergoing cell death. Although it is now difficult to avoid MeHg exposure from our environment and contaminated foods, taking anti-oxidants from foods or supplements may prevent or diminish the toxicological effects of MeHg.


Assuntos
Antioxidantes/farmacologia , Poluentes Ambientais/toxicidade , Compostos de Metilmercúrio/toxicidade , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Butionina Sulfoximina/farmacologia , Contagem de Células , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Diencéfalo/efeitos dos fármacos , Diencéfalo/embriologia , Diencéfalo/patologia , Interações Medicamentosas , Glutationa/antagonistas & inibidores , Glutationa/biossíntese , Camundongos , Camundongos Endogâmicos ICR , Neurônios/metabolismo , Neurônios/patologia , Células-Tronco/metabolismo , Células-Tronco/patologia , Telencéfalo/efeitos dos fármacos , Telencéfalo/embriologia , Telencéfalo/patologia
17.
J Neuroendocrinol ; 20(9): 1078-88, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18638025

RESUMO

Gonadotrophin-inhibitory hormone (GnIH), a hypothalamic RFamide, has been found to inhibit gonadotrophin secretion from the anterior pituitary gland originally in birds and, subsequently, in mammalian species. The gene encoding a transmembrane receptor for GnIH (GnIHR) was recently identified in the brain, pituitary gland and gonads of song bird, chicken and Japanese quail. The objectives of the present study are to characterise the expression of GnIHR mRNA and protein in the chicken pituitary gland, and to determine whether sexual maturation and gonadal steroids influence pituitary GnIHR mRNA abundance. GnIHR mRNA quantity was found to be significantly higher in diencephalon compared to either anterior pituitary gland or ovaries. GnIHR mRNA quantity was significantly higher in the pituitaries of sexually immature chickens relative to sexually mature chickens. Oestradiol or a combination of oestradiol and progesterone treatment caused a significant decrease in pituitary GnIHR mRNA quantity relative to vehicle controls. GnIHR-immunoreactive (ir) cells were identified in the chicken pituitary gland cephalic and caudal lobes. Furthermore, GnIHR-ir cells were found to be colocalised with luteinising hormone (LH)beta mRNA-, or follicle-stimulating hormone (FSH)beta mRNA-containing cells. GnIH treatment significantly decreased LH release from anterior pituitary gland slices collected from sexually immature, but not from sexually mature chickens. Taken together, GnIHR gene expression is possibly down regulated in response to a surge in circulating oestradiol and progesterone levels as the chicken undergoes sexual maturation to allow gonadotrophin secretion. Furthermore, GnIHR protein expressed in FSHbeta or LHbeta mRNA-containing cells is likely to mediate the inhibitory effect of GnIH on LH and FSH secretion.


Assuntos
Proteínas Aviárias/genética , Galinhas/genética , Hormônios Esteroides Gonadais/farmacologia , Hipófise/metabolismo , Receptores de Superfície Celular/genética , Maturidade Sexual/fisiologia , Animais , Proteínas Aviárias/metabolismo , Galinhas/metabolismo , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Estradiol/farmacologia , Feminino , Hormônio Foliculoestimulante/genética , Hormônio Foliculoestimulante/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hormônio Luteinizante/genética , Hormônio Luteinizante/metabolismo , Ovário/metabolismo , Hipófise/efeitos dos fármacos , Progesterona/farmacologia , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/metabolismo , Maturidade Sexual/genética , Distribuição Tecidual
18.
J Neurosci ; 28(53): 14492-9, 2008 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-19118184

RESUMO

Goldfish (Carassius auratus) use reproductive hormones as endocrine signals to synchronize sexual behavior with gamete maturation and as exogenous signals (pheromones) to mediate spawning interactions between conspecifics. We examined the differential effects of two hormonal pheromones, prostaglandin F(2alpha) (PGF(2alpha)) and 17alpha,20beta-dihydroxy-4-pregnen-3-one (17,20beta-P) on neurogenesis, neurotransmission, and neuronal activities, and on plasma androstenedione (AD) levels. Exposure to waterborne PGF(2alpha) induced a multitude of changes in male goldfish brain. Histological examination indicated an increase in the number of dividing cells in male diencephalon (p < 0.05, Kruskal-Wallis test). Real-time quantitative PCR tests showed elevated levels of transcripts for the salmon gonadotropin-releasing hormone (GnRH) in the male telencephalon and cerebellum (p < 0.005, one-way ANOVA) and for ChAT (choline acetyltransferase) in the male vagal lobe and the brainstem underneath the vagal lobe (p < 0.05, one-way ANOVA). Therefore, PGF(2alpha) seemed to modulate male brain plasticity that coincided with behavioral changes during spawning season. Exposure to waterborne 17,20beta-P, however, increased circulatory levels of immunoreactive AD in males and the transcripts of androgen receptor and cGnRH-II (chicken-II GnRH) in the female cerebellum (p < 0.05, one-way ANOVA). PGF(2alpha) and 17,20beta-P thereby seemed to act through distinct pathways to elicit different responses in the neuroendocrine system. This is the first finding that links a specific pheromone molecule (PGF(2alpha)) to neurogenesis in a vertebrate animal.


Assuntos
Dinoprosta/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/metabolismo , Hidroxiprogesteronas/farmacologia , Neurogênese/efeitos dos fármacos , Análise de Variância , Androstenodiona/sangue , Animais , Química Encefálica/efeitos dos fármacos , Colina O-Acetiltransferase/genética , Colina O-Acetiltransferase/metabolismo , Diencéfalo/citologia , Diencéfalo/efeitos dos fármacos , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Carpa Dourada/fisiologia , Hormônio Liberador de Gonadotropina/genética , Masculino , Neurotransmissores/genética , Neurotransmissores/metabolismo , RNA Mensageiro/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo
19.
J Neuropathol Exp Neurol ; 66(5): 383-8, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17483695

RESUMO

Restless legs syndrome (RLS) is a common neurologic condition involving iron and dopamine systems. We sought to create an animal model consistent with RLS based on current understanding of human pathology. We performed bilateral 6-hydroxydopamine (6-OHDA) lesioning in the A11 nucleus of C57BL/6 mice and deprived a subset of mice from dietary iron to observe whether these manipulations can increase motor activity. Iron levels in serum, brain, and especially spinal cord were significantly decreased after iron deprivation. Interestingly, 6-OHDA lesioning appeared to further reduce CNS iron stores. Pathologic examination demonstrated a 94% reduction in A11 tyrosine hydroxylase staining cells in mice injected with 6-OHDA but minimal effects on other areas. Locomotor activities were significantly increased in both the mice that were iron deprived and the A11-lesioned mice compared with controls. The combination of iron deprivation and A11 lesions further significantly augmented activity. Additionally, the mice in the combined iron-deprived and lesioned group were more aggressive. The increased activity in A11-lesioned mice with or without iron deprivation was normalized after treatment with the D2/D3 agonist ropinirole, as is seen in human RLS but was worsened by the D1 agonist SKF38393. This model could be consistent with human RLS, attention deficit hyperactivity disorder, or akathisia.


Assuntos
Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Diencéfalo/patologia , Deficiências de Ferro , Locomoção/fisiologia , Animais , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/metabolismo , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Modelos Animais de Doenças , Agonistas de Dopamina/farmacologia , Indóis/farmacologia , Ferro/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxidopamina , Síndrome das Pernas Inquietas , Medula Espinal/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
20.
Brain Res Bull ; 65(3): 267-73, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15811591

RESUMO

The xenoestrogen bisphenol A, a contaminant used in the manufacturing of polymers for many consumer products, has been shown to mimic estrogenic actions. This xenoestrogen regulates secretion and expression of pituitary lactotrophs plus morphological and structural features of estrogen target tissues in rodents. Recently, ecological hazards produced by bisphenol A have drawn interests towards the effects of this environmental chemical on neurobiological functions of aquatic vertebrates of which little is known. In this study, the effects of bisphenol A on the distribution of the biologically more active somatostatin receptor subtypes in diencephalic regions of the teleost fish Coris julis were assessed using nonpeptide agonists (L-779, 976 and L-817, 818) that are highly selective for subtype(2) and subtype(5), respectively. Bisphenol A proved to be responsible for highly significant increased binding levels of subtype(2) in hypothalamic areas, while markedly decreased levels of subtype(5) were found in these diencephalic areas, as well as in the medial preglomerular nucleus. The extensive distribution of somatostatin receptor subtype(2) and subtype(5) in the teleost diencephalic areas suggests that, like in mammals, this receptor system may not only be involved in enhanced hypophysiotropic neurohormonal functions but might also promote neuroplasticity events.


Assuntos
Diencéfalo/efeitos dos fármacos , Estrogênios não Esteroides/farmacologia , Fenóis/farmacologia , Receptores de Somatostatina/fisiologia , Animais , Autorradiografia/métodos , Compostos Benzidrílicos , Ligação Competitiva/efeitos dos fármacos , Diencéfalo/anatomia & histologia , Diencéfalo/metabolismo , Relação Dose-Resposta a Droga , Peixes , Receptores de Somatostatina/agonistas , Somatostatina/agonistas , Somatostatina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA