Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 234
Filtrar
1.
Neurochem Int ; 161: 105420, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36170907

RESUMO

Sex differences in the sensitivity to hypertension and inflammatory processes are well characterized but insufficiently understood. In male mice, tumor necrosis factor alpha (TNFα) in the hypothalamic paraventricular nucleus (PVN) contributes to hypertension following slow-pressor angiotensin II (AngII) infusion. However, the role of PVN TNFα in the response to AngII in female mice is unknown. Using a combination of in situ hybridization, high-resolution electron microscopic immunohistochemistry, spatial-temporal gene silencing, and dihydroethidium microfluorography we investigated the influence of AngII on both blood pressure and PVN TNFα signaling in female mice. We found that chronic (14-day) infusion of AngII in female mice did not impact blood pressure, TNFα levels, the expression of the TNFα type 1 receptor (TNFR1), or the subcellular distribution of TNFR1 in the PVN. However, it was shown that blockade of estrogen receptor ß (ERß), a major hypothalamic estrogen receptor, was accompanied by both elevated PVN TNFα and hypertension following AngII. Further, AngII hypertension following ERß blockade was attenuated by inhibiting PVN TNFα signaling by local TNFR1 silencing. It was also shown that ERß blockade in isolated PVN-spinal cord projection neurons (i.e. sympathoexcitatory) heightened TNFα-induced production of NADPH oxidase (NOX2)-mediated reactive oxygen species, molecules that may play a key role in mediating the effect of TNFα in hypertension. These results indicate that ERß contributes to the reduced sensitivity of female mice to hypothalamic inflammatory cytokine signaling and hypertension in response to AngII.


Assuntos
Hipertensão , Núcleo Hipotalâmico Paraventricular , Camundongos , Feminino , Masculino , Animais , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/patologia , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Angiotensina II/efeitos adversos , Angiotensina II/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/efeitos adversos , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Neurônios/metabolismo , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Pressão Sanguínea
2.
J Ethnopharmacol ; 276: 114218, 2021 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-34029638

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Kamikihito (KKT) is a Kampo medicine that is prescribed in Japan for the treatment of anemia, insomnia and mental anxiety in Japan. However, its precise mechanism of action remains unclear. AIM OF THE STUDY: This study aimed to evaluate the possible antistress effect of KKT in rats with acute stress and the contribution of oxytocin to the process. MATERIALS AND METHODS: Acute immobilization stress (AIS; for 90 min) was used to assess the effect of KKT on acute stress. Male Wistar rats were orally treated with KKT. Parameters of stress were evaluated, and concentrations of oxytocin in plasma and cerebrospinal fluid (CSF) were measured. RESULTS: AIS-induced defecation and fecal weight were significantly decreased because of treatment with KKT. The plasma levels of stress-related hormones following AIS were investigated. The pre-administration of KKT significantly increased adrenocorticotrophic hormone (ACTH) and corticosterone (CORT) levels following AIS. Conversely, there was no significant change in the plasma oxytocin level. Microdialysis and hydrophilic interaction liquid chromatography-tandem mass spectrometry (HILIC-MS/MS) were used to monitor the oxytocin secretion in CSF. Oxytocin level increased during AIS following the treatment of KKT. At 30 min after AIS, the level remained higher than before AIS. Furthermore, using an open field test, the locomotion (exploratory behavior) immediately after AIS was examined. The total traveled distance decreased after AIS; however, the decrease was significantly inhibited by the treatment of KKT. However, the effect of KKT was obstructed by the pre-administration of the oxytocin receptor antagonist. CONCLUSIONS: These results suggest that KKT has antistress activity and increased oxytocin secretion may be a mechanism underlying this phenomenon.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Ocitocina/líquido cefalorraquidiano , Estresse Fisiológico/efeitos dos fármacos , Administração Oral , Hormônio Adrenocorticotrópico/sangue , Animais , Comportamento Animal/efeitos dos fármacos , Corticosterona/sangue , Defecação/efeitos dos fármacos , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/administração & dosagem , Locomoção/efeitos dos fármacos , Masculino , Medicina Kampo/métodos , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Ratos Wistar , Restrição Física/efeitos adversos
3.
J Comp Neurol ; 529(9): 2283-2310, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33341960

RESUMO

Within the hypothalamic paraventricular nucleus (PVN), estrogen receptor (ER) ß and other gonadal hormone receptors play a role in central cardiovascular processes. However, the influence of sex and age on the cellular and subcellular relationships of ERß with ERα, G-protein ER (GPER1), as well as progestin and androgen receptors (PR and AR) in the PVN is uncertain. In young (2- to 3-month-old) females and males, ERß-enhanced green fluorescent protein (EGFP) containing neurons were approximately four times greater than ERα-labeled and PR-labeled nuclei in the PVN. In subdivisions of the PVN, young females, compared to males, had: (1) more ERß-EGFP neurons in neuroendocrine rostral regions; (2) fewer ERα-labeled nuclei in neuroendocrine and autonomic projecting medial subregions; and (3) more ERα-labeled nuclei in an autonomic projecting caudal region. In contrast, young males, compared to females, had approximately 20 times more AR-labeled nuclei, which often colocalized with ERß-EGFP in neuroendocrine (approximately 70%) and autonomic (approximately 50%) projecting subregions. Ultrastructurally, in soma and dendrites, PVN ERß-EGFP colocalized primarily with extranuclear AR (approximately 85% soma) and GPER1 (approximately 70% soma). Aged (12- to 24-month-old) males had more ERß-EGFP neurons in a rostral neuroendocrine subregion compared to aged females and females with accelerated ovarian failure (AOF) and in a caudal autonomic subregion compared to post-AOF females. Late-aged (18- to 24-month-old) females compared to early-aged (12- to 14-month-old) females and AOF females had fewer AR-labeled nuclei in neuroendrocrine and autonomic projecting subregions. These findings indicate that gonadal steroids may directly and indirectly influence PVN neurons via nuclear and extranuclear gonadal hormone receptors in a sex-specific manner.


Assuntos
Receptor beta de Estrogênio/biossíntese , Hormônios Esteroides Gonadais/biossíntese , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Caracteres Sexuais , Fatores Etários , Animais , Receptor beta de Estrogênio/análise , Receptor beta de Estrogênio/ultraestrutura , Feminino , Hormônios Esteroides Gonadais/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/ultraestrutura , Núcleo Hipotalâmico Paraventricular/química , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Receptores Androgênicos/análise , Receptores Androgênicos/biossíntese , Receptores Androgênicos/ultraestrutura , Receptores Acoplados a Proteínas G/análise , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/ultraestrutura
4.
Neuroendocrinology ; 104(3): 239-256, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27078860

RESUMO

Hypertension in male and aging female rodents is associated with glutamate-dependent plasticity in the hypothalamus, but existing models have failed to capture distinct transitional menopausal phases that could have a significant impact on the synaptic plasticity and emergent hypertension. In rodents, accelerated ovarian failure (AOF) induced by systemic injection of 4-vinylcyclohexane diepoxide mimics the estrogen fluctuations seen in human menopause including the perimenopause transition (peri-AOF) and postmenopause (post-AOF). Thus, we used the mouse AOF model to determine the impact of slow-pressor angiotensin II (AngII) administration on blood pressure and on the subcellular distribution of obligatory N-methyl-D-aspartate (NMDA) receptor GluN1 subunits in the paraventricular hypothalamic nucleus (PVN), a key estrogen-responsive cardiovascular regulatory area. Estrogen-sensitive neuronal profiles were identified in mice expressing enhanced green fluorescent protein under the promoter for estrogen receptor (ER) ß, a major ER in the PVN. Slow-pressor AngII increased arterial blood pressure in mice at peri- and post-AOF time points. In control oil-injected (nonhypertensive) mice, AngII decreased the total number of GluN1 in ERß-containing PVN dendrites. In contrast, AngII resulted in a reapportionment of GluN1 from the cytoplasm to the plasma membrane of ERß-containing PVN dendrites in peri-AOF mice. Moreover, in post-AOF mice, AngII increased total GluN1, dendritic size and radical production in ERß-containing neurons. These results indicate that unique patterns of hypothalamic glutamate receptor plasticity and dendritic structure accompany the elevated blood pressure in peri- and post-AOF time points. Our findings suggest the possibility that distinct neurobiological processes are associated with the increased blood pressure during perimenopausal and postmenopausal periods.


Assuntos
Hipertensão , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Doenças Ovarianas/etiologia , Núcleo Hipotalâmico Paraventricular/patologia , Receptores de Estrogênio/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Angiotensina II/toxicidade , Animais , Pressão Sanguínea/efeitos dos fármacos , Cicloexenos/toxicidade , Modelos Animais de Doenças , Ciclo Estral/efeitos dos fármacos , Ciclo Estral/genética , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipertensão/induzido quimicamente , Hipertensão/complicações , Hipertensão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Imunoeletrônica , Neurônios/ultraestrutura , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Espécies Reativas de Oxigênio/metabolismo , Receptores de Estrogênio/genética , Compostos de Vinila/toxicidade
5.
Front Neural Circuits ; 10: 13, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27065810

RESUMO

Water-homeostasis is a fundamental physiological process for terrestrial life. In vertebrates, thirst drives water intake, but the neuronal circuits that connect the physiology of water regulation with emotional context are poorly understood. Vasopressin (VP) is a prominent messenger in this circuit, as well as L-glutamate. We have investigated the role of a VP circuit and interaction between thirst and motivational behaviors evoked by life-threatening stimuli in rats. We demonstrate a direct pathway from hypothalamic paraventricular VP-expressing, glutamatergic magnocellular neurons to the medial division of lateral habenula (LHbM), a region containing GABAergic neurons. In vivo recording and juxtacellular labeling revealed that GABAergic neurons in the LHbM had locally branching axons, and received VP-positive axon terminal contacts on their dendrites. Water deprivation significantly reduced freezing and immobility behaviors evoked by innate fear and behavioral despair, respectively, accompanied by decreased Fos expression in the lateral habenula. Our results reveal a novel VP-expressing hypothalamus to the LHbM circuit that is likely to evoke GABA-mediated inhibition in the LHbM, which promotes escape behavior during stress coping.


Assuntos
Ácido Glutâmico/metabolismo , Habenula/fisiologia , Transdução de Sinais/fisiologia , Estresse Psicológico/fisiopatologia , Sede/fisiologia , Vasopressinas/metabolismo , Animais , Gatos , Colchicina/farmacologia , Modelos Animais de Doenças , Medo/psicologia , Glutamato Descarboxilase/metabolismo , Habenula/citologia , Habenula/efeitos dos fármacos , Habenula/ultraestrutura , Masculino , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Proteínas Oncogênicas v-fos/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Estresse Psicológico/patologia , Sinapses/metabolismo , Sede/efeitos dos fármacos , Moduladores de Tubulina/farmacologia , Privação de Água/fisiologia , Ácido gama-Aminobutírico/metabolismo
6.
J Neurosci ; 35(26): 9558-67, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26134639

RESUMO

Hypertension induced by angiotensin II (Ang II) is associated with glutamate-dependent dysregulation of the hypothalamic paraventricular nucleus (PVN). Many forms of glutamate-dependent plasticity are mediated by NMDA receptor GluN1 subunit expression and the distribution of functional receptor to the plasma membrane of dendrites. Here, we use a combined ultrastructural and functional analysis to examine the relationship between PVN NMDA receptors and the blood pressure increase induced by chronic infusion of a low dose of Ang II. We report that the increase in blood pressure produced by a 2 week administration of a subpressor dose of Ang II results in an elevation in plasma membrane GluN1 in dendrites of PVN neurons in adult male mice. The functional implications of these observations are further demonstrated by the finding that GluN1 deletion in PVN neurons attenuated the Ang II-induced increases in blood pressure. These results indicate that NMDA receptor plasticity in PVN neurons significantly contributes to the elevated blood pressure mediated by Ang II.


Assuntos
Angiotensina II/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/genética , Proteínas do Tecido Nervoso/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Análise de Variância , Animais , Lateralidade Funcional , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Imunoeletrônica , N-Metilaspartato/farmacologia , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Neurônios/ultraestrutura , Óxido Nítrico Sintase Tipo I/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Pletismografia , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Vasoconstritores
7.
Am J Alzheimers Dis Other Demen ; 30(5): 478-87, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25380804

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disorder, characterized by irreversible decline of mental faculties, emotional and behavioral changes, loss of motor skills, and dysfunction of autonomic nervous system and disruption of circadian rhythms (CRs). We attempted to describe the morphological findings of the hypothalamus in early cases of AD, focusing our study mostly on the suprachiasmatic nucleus (SCN), the supraoptic nucleus (SON), and the paraventricular nucleus (PVN). Samples were processed for electron microscopy and silver impregnation techniques. The hypothalamic nuclei demonstrated a substantial decrease in the neuronal population, which was particularly prominent in the SCN. Marked abbreviation of dendritic arborization, in association with spinal pathology, was also seen. The SON and PVN demonstrated a substantial number of dystrophic axons and abnormal spines. Alzheimer's pathology, such as deposits of amyloid-ß peptide and neurofibrillary degeneration, was minimal. Electron microscopy revealed mitochondrial alterations in the cell body and the dendritic branches. The morphological alterations of the hypothalamic nuclei in early cases of AD may be related to the gradual alteration of CRs and the instability of autonomic regulation.


Assuntos
Doença de Alzheimer/patologia , Neurônios/ultraestrutura , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Núcleo Supraquiasmático/ultraestrutura , Núcleo Supraóptico/ultraestrutura , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Espinhas Dendríticas/ultraestrutura , Feminino , Complexo de Golgi/ultraestrutura , Humanos , Hipotálamo/ultraestrutura , Masculino , Microscopia Eletrônica , Pessoa de Meia-Idade , Mitocôndrias/ultraestrutura , Coloração pela Prata
8.
Brain Struct Funct ; 220(3): 1511-28, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24633808

RESUMO

Human ciliopathies are genetic disorders caused by mutations in genes responsible for the formation and function of primary cilia. Some are associated with hyperphagia and obesity (e.g., Bardet-Biedl Syndrome, Alström Syndrome), but the mechanisms underlying these problems are not fully understood. The human gene ANKRD26 is located on 10p12, a locus that is associated with some forms of hereditary obesity. Previously, we reported that disruption of this gene causes hyperphagia, obesity and gigantism in mice. In the present study, we looked for the mechanisms that induce hyperphagia in the Ankrd26-/- mice and found defects in primary cilia in regions of the central nervous system that control appetite and energy homeostasis.


Assuntos
Proteínas de Ligação a DNA/genética , Hiperfagia/genética , Hiperfagia/patologia , Neurônios/ultraestrutura , Obesidade/genética , Obesidade/patologia , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Fatores de Transcrição/genética , Acetilcolina/sangue , Animais , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Cílios/metabolismo , Cílios/ultraestrutura , Corticosterona/sangue , Proteínas de Ligação a DNA/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Receptores para Leptina/metabolismo , Estresse Fisiológico , Fatores de Transcrição/metabolismo
9.
Am J Physiol Regul Integr Comp Physiol ; 304(12): R1096-106, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23576605

RESUMO

Adaptive changes in glutamatergic signaling within the hypothalamic paraventricular nucleus (PVN) may play a role in the neurohumoral dysfunction underlying the hypertension induced by "slow-pressor" ANG II infusion. We hypothesized that these adaptive changes alter production of gp91phox NADPH oxidase (NOX)-derived reactive oxygen species (ROS) or nitric oxide (NO), resulting in enhanced glutamatergic signaling in the PVN. Electron microscopic immunolabeling showed colocalization of NOX2 and N-methyl-D-aspartate receptor (NMDAR) NR1 subunits in PVN dendrites, an effect enhanced (+48%, P < 0.05 vs. saline) in mice receiving ANG II (600 ng·kg⁻¹·min⁻¹ sc). Isolated PVN cells or spinally projecting PVN neurons from ANG II-infused mice had increased levels of ROS at baseline (+40 ± 5% and +57.6 ± 7.7%, P < 0.01 vs. saline) and after NMDA (+24 ± 7% and +17 ± 5.5%, P < 0.01 and P < 0.05 vs. saline). In contrast, ANG II infusion suppressed NO production in PVN cells at baseline (-29.1 ± 5.2%, P < 0.05 vs. saline) and after NMDA (-18.9 ± 2%, P < 0.01 vs. saline), an effect counteracted by NOX inhibition. In whole cell recording of unlabeled and spinally labeled PVN neurons in slices, NMDA induced a larger inward current in ANG II than in saline groups (+79 ± 24% and +82.9 ± 6.6%, P < 0.01 vs. saline), which was reversed by the ROS scavenger MnTBAP and the NO donor S-nitroso-N-acetylpenicillamine (P > 0.05 vs. control). These findings suggest that slow-pressor ANG II increases the association of NR1 with NOX2 in dendrites of PVN neurons, resulting in enhanced NOX-derived ROS and reduced NO during glutamatergic activity. The resulting enhancement of NMDAR activity may contribute to the neurohumoral dysfunction underlying the development of slow-pressor ANG II hypertension.


Assuntos
Angiotensina II/efeitos adversos , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Glicoproteínas de Membrana/metabolismo , N-Metilaspartato/metabolismo , NADPH Oxidases/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Superóxidos/metabolismo , Angiotensina II/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Cálcio/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Modelos Animais , NADPH Oxidase 2 , Neurônios/citologia , Neurônios/metabolismo , Neurônios/ultraestrutura , Óxido Nítrico/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Técnicas de Patch-Clamp , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
10.
Nat Commun ; 4: 1526, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23443554

RESUMO

Brain-derived neurotrophic factor (BDNF) suppresses food intake by acting on neurons in the hypothalamus. Here we show that BDNF-producing haematopoietic cells control appetite and energy balance by migrating to the hypothalamic paraventricular nucleus. These haematopoietic-derived paraventricular nucleus cells produce microglial markers and make direct contacts with neurons in response to feeding status. Mice with congenital BDNF deficiency, specifically in haematopoietic cells, develop hyperphagia, obesity and insulin resistance. These abnormalities are ameliorated by bone marrow transplantation with wild-type bone marrow cells. Furthermore, when injected into the third ventricle, wild-type bone marrow mononuclear cells home to the paraventricular nucleus and reverse the hyperphagia of BDNF-deficient mice. Our results suggest a novel mechanism of feeding control based on the production of BDNF by haematopoietic cells and highlight a potential new therapeutic route for the treatment of obesity.


Assuntos
Apetite , Movimento Celular , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Hipotálamo/metabolismo , Animais , Apetite/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Fator Neurotrófico Derivado do Encéfalo/administração & dosagem , Fator Neurotrófico Derivado do Encéfalo/deficiência , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Movimento Celular/efeitos dos fármacos , Comportamento de Ingestão de Líquido/efeitos dos fármacos , Jejum/metabolismo , Comportamento Alimentar/efeitos dos fármacos , Deleção de Genes , Células-Tronco Hematopoéticas/efeitos dos fármacos , Hiperfagia/complicações , Hiperfagia/patologia , Hiperfagia/fisiopatologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/patologia , Hipotálamo/ultraestrutura , Injeções Intraventriculares , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Obesidade/complicações , Obesidade/patologia , Obesidade/fisiopatologia , Especificidade de Órgãos/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/patologia , Núcleo Hipotalâmico Paraventricular/ultraestrutura
11.
Neuro Endocrinol Lett ; 32(6): 763-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22286787

RESUMO

OBJECTIVES: The purpose of this study was to identify and characterize Na+-dependent, high affinity glutamate transporter (GLUT) activity in the hypothalamic paraventricular nucleus (PVN) and to compare GLUT activity in PVN of euhydrated versus water-deprived rats. METHODS: Sprague-Dawley rats were deprived of water for two days before sacrifice. Control rats received water ad libitum. After sacrifice, PVN and cerebrum were removed and synaptosomes were prepared using standard techniques. Glutamate uptake was measured using [3H]-glutamate as substrate, physiological buffer, approximately 100 µg of synaptosomal tissue per assay and a Brandel cell harvester. RESULTS: Glutamate uptake was saturable in PVN synaptosomes from euhydrated, control rats with a Vmax of 541 ± 22 pmol/min-mg protein (SEM) and Km of 17.6 ± 3.8 µM (SEM). In contrast, Vmax of glutamate uptake was 808 ± 58 pmol/min-mg protein in PVN of rats deprived of water for 2 days. This was significantly higher than controls (p<0.001). Km was 21.2 ± 7.3 µM and not significantly different from controls (NS). CONCLUSIONS: Our results suggest that water deprivation of rats results in significantly higher synaptosomal glutamate uptake in PVN. Although the exact mechanism is unknown, increased transcription of the GLUT gene and/or increased cell surface expression of GLUT may contribute to the observed increase of glutamate uptake in dehydrated rats. Increased glutamate uptake may serve to restrict dehydration-induced activation of PVN efferent pathways specifically involved in release of neurohypophysial hormones and activation of sympathetic outflow that operate to maintain body fluid balance and cardiovascular function.


Assuntos
Desidratação/fisiopatologia , Ácido Glutâmico/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Sódio/metabolismo , Sinaptossomos/metabolismo , Privação de Água/fisiologia , Animais , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Equilíbrio Hidroeletrolítico
12.
J Neurosci ; 30(36): 12103-12, 2010 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-20826673

RESUMO

Chronic intermittent hypoxia (CIH) is a concomitant of sleep apnea that produces a slowly developing chemosensory-dependent blood pressure elevation ascribed in part to NMDA receptor-dependent plasticity and reduced nitric oxide (NO) signaling in the carotid body. The hypothalamic paraventricular nucleus (PVN) is responsive to hypoxic stress and also contains neurons that express NMDA receptors and neuronal nitric oxide synthase (nNOS). We tested the hypothesis that extended (35 d) CIH results in a decrease in the surface/synaptic availability of the essential NMDA NR1 subunit in nNOS-containing neurons and NMDA-induced NO production in the PVN of mice. As compared with controls, the 35 d CIH-exposed mice showed a significant increase in blood pressure and an increased density of NR1 immunogold particles located in the cytoplasm of nNOS-containing dendrites. Neither of these between-group differences was seen after 14 d, even though there was already a reduction in the NR1 plasmalemmal density at this time point. Patch-clamp recording of PVN neurons in slices showed a significant reduction in NMDA currents after either 14 or 35 d exposure to CIH compared with sham controls. In contrast, NO production, as measured by the NO-sensitive fluorescent dye 4-amino-5-methylamino-2',7'-difluorofluorescein, was suppressed only in the 35 d CIH group. We conclude that CIH produces a reduction in the surface/synaptic targeting of NR1 in nNOS neurons and decreases NMDA receptor-mediated currents in the PVN before the emergence of hypertension, the development of which may be enabled by suppression of NO signaling in this brain region.


Assuntos
Hipóxia/patologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Óxido Nítrico/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Arginina/farmacologia , Gasometria/métodos , Pressão Sanguínea/fisiologia , Óxidos N-Cíclicos/farmacologia , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Sequestradores de Radicais Livres/farmacologia , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Hipóxia/fisiopatologia , Imidazóis/farmacologia , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão/métodos , N-Metilaspartato/farmacologia , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo I/ultraestrutura , Núcleo Hipotalâmico Paraventricular/patologia , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Receptores de N-Metil-D-Aspartato/ultraestrutura , S-Nitroso-N-Acetilpenicilamina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Vasopressinas/metabolismo
13.
J Neurosci ; 30(2): 703-13, 2010 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-20071535

RESUMO

Increased sensory input from maternal care attenuates neuroendocrine and behavioral responses to stress long term and results in a lifelong phenotype of resilience to depression and improved cognitive function. Whereas the mechanisms of this clinically important effect remain unclear, the early, persistent suppression of the expression of the stress neurohormone corticotropin-releasing hormone (CRH) in hypothalamic neurons has been implicated as a key aspect of this experience-induced neuroplasticity. Here, we tested whether the innervation of hypothalamic CRH neurons of rat pups that received augmented maternal care was altered in a manner that might promote the suppression of CRH expression and studied the cellular mechanisms underlying this suppression. We found that the number of excitatory synapses and the frequency of miniature excitatory synaptic currents onto CRH neurons were reduced in "care-augmented" rats compared with controls, as were the levels of the glutamate vesicular transporter vGlut2. In contrast, analogous parameters of inhibitory synapses were unchanged. Levels of the transcriptional repressor neuron-restrictive silencer factor (NRSF), which negatively regulates Crh gene transcription, were markedly elevated in care-augmented rats, and chromatin immunoprecipitation demonstrated that this repressor was bound to a cognate element (neuron-restrictive silencing element) on the Crh gene. Whereas the reduced excitatory innervation of CRH-expressing neurons dissipated by adulthood, increased NRSF levels and repression of CRH expression persisted, suggesting that augmented early-life experience reprograms Crh gene expression via mechanisms involving transcriptional repression by NRSF.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Privação Materna , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Estresse Psicológico/patologia , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Imunoprecipitação da Cromatina/métodos , Hormônio Liberador da Corticotropina/genética , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Masculino , Microscopia Eletrônica de Transmissão/métodos , Neurônios/ultraestrutura , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Técnicas de Patch-Clamp , Estimulação Física , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Estresse Psicológico/metabolismo , Potenciais Sinápticos/efeitos dos fármacos , Potenciais Sinápticos/fisiologia , Tetrodotoxina/farmacologia , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
14.
J Chem Neuroanat ; 37(4): 229-33, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19481007

RESUMO

Previous studies revealed that oxytocin release is increased by various forms of stress. Hypertonic saline injection, immobilization, and several other stressors elevated the blood level of oxytocin in rats. However, the mechanism of the stress-induced oxytocin release in human is not elucidated yet. Although numerous studies indicate that catecholamines play a pivotal role in modulating the release of oxytocin, there is a lack of data regarding the morphological substrate of this phenomenon. In order to reveal putative juxtapositions between tyrosine hydroxylase-immunoreactive (TH-IR) catecholaminergic and the oxytocinergic systems in the human hypothalamus, we utilized double-label immunohistochemistry in the present study. Numerous TH-IR axon varicosities abutted on oxytocin-IR neurons in the supraoptic and paraventricular nuclei, forming synapse-like associations. Close examination of these juxtapositions with high magnification failed to reveal any gaps between the contacting elements. In summary, the intimate associations between the TH-IR and oxytocin-IR elements may be functional synapses and may represent the morphological substrate of stress-influenced oxytocin release. The finding that several oxytocin-IR perikarya did not receive apparent TH innervation suggests that additional mechanisms may play significant roles in the oxytocin modulation by stressors.


Assuntos
Catecolaminas/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Hipotálamo/metabolismo , Vias Neurais/metabolismo , Neurossecreção/fisiologia , Ocitocina/metabolismo , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/análise , Biomarcadores/metabolismo , Mapeamento Encefálico , Feminino , Humanos , Sistema Hipotálamo-Hipofisário/ultraestrutura , Hipotálamo/ultraestrutura , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Vias Neurais/ultraestrutura , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Núcleo Supraóptico/metabolismo , Núcleo Supraóptico/ultraestrutura , Transmissão Sináptica/fisiologia , Tirosina 3-Mono-Oxigenase/análise , Tirosina 3-Mono-Oxigenase/metabolismo
15.
Neurosci Res ; 59(4): 390-8, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17897744

RESUMO

The organization of projections from the central amygdaloid nucleus (CeA) to the paraventricuilar hypothalamic nucleus (PVH) has been studied in order to understand the anatomical substrates of amygdaloid modulation of endocrine and autonomic functions, and a hypothesis that the bed nucleus of the stria terminalis (BST) may act as a relay site between the CeA and PVH has been proposed. Using anterograde and retrograde tract-tracing methods, in the rat, we first indicated that neurons in the parastrial nucleus (PS), where projection fibers from the central amygdaloid nucleus (CeA) terminated, sent their axons to the paraventricular hypothalamic nucleus (PVH). We further demonstrated that the CeA terminals formed symmetrical synaptic contacts with somata and dendrites of the PVH-projecting PS neurons, and that the PS received CeA fibers predominantly from the lateral part and sent large numbers of projection fibers to almost all the subdivisions of the PVH. Using anterograde tracing combined with the postembedding immunogold method, we finally revealed that nearly all the CeA terminals in the PS were immunoreactive for gamma-aminobutyric acid. The present data suggest that output signals from the CeA are transmitted disynaptically to the PVH neurons via the PS neurons and modulate PVH neuron activity by way of disinhibition.


Assuntos
Tonsila do Cerebelo/ultraestrutura , Vias Neurais/ultraestrutura , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Núcleos Septais/ultraestrutura , Sinapses/ultraestrutura , Tonsila do Cerebelo/fisiologia , Animais , Transporte Axonal/fisiologia , Biotina/análogos & derivados , Mapeamento Encefálico , Toxina da Cólera , Dendritos/fisiologia , Dendritos/ultraestrutura , Dextranos , Masculino , Microscopia Imunoeletrônica , Inibição Neural/fisiologia , Vias Neurais/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Terminações Pré-Sinápticas/fisiologia , Terminações Pré-Sinápticas/ultraestrutura , Ratos , Ratos Wistar , Núcleos Septais/fisiologia , Sinapses/fisiologia , Ácido gama-Aminobutírico/metabolismo
16.
Endocrinology ; 148(12): 5842-50, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17872373

RESUMO

The regulatory actions of estrogens on magnocellular oxytocin (OT) neurons of the paraventricular and supraoptic nuclei are well documented. Although the expression and distribution of nuclear estrogen receptor-beta, but not estrogen receptor-alpha, in the OT neuron has been described, the nuclear receptors may not explain all aspects of estrogen function in the hypothalamic OT neuron. Recently a G protein-coupled receptor (GPR) for estrogens, GPR30, has been identified as a membrane-localized estrogen receptor in several cancer cell lines. In this study, we therefore investigated the expression and localization of GPR30 in magnocellular OT neurons to understand the mode of rapid estrogen actions within these neurons. Here we show that, in the paraventricular nucleus and supraoptic nucleus, GPR30 is expressed in magnocellular OT neurons at both mRNA and protein levels but is not expressed in vasopressin neurons. Specific markers for intracellular organelles and immunoelectron microscopy revealed that GPR30 was localized mainly in the Golgi apparatus of the neurons but could not be detected at the cell surface. In addition, the expression of GPR30 is also detected in the neurohypophysis. These results suggest that GPR30 may serve primarily as a nongenomic transducer of estrogen actions in the hypothalamo-neurohypophyseal system.


Assuntos
Neurônios/metabolismo , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Núcleo Supraóptico/metabolismo , Animais , Western Blotting , Imuno-Histoquímica , Hibridização In Situ , Microscopia Imunoeletrônica , Neurônios/ultraestrutura , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Núcleo Supraóptico/citologia , Núcleo Supraóptico/ultraestrutura , Vasopressinas/metabolismo
17.
J Neurosci ; 26(46): 11893-902, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17108163

RESUMO

Cranial visceral afferents activate central pathways that mediate systemic homeostatic processes. Afferent information arrives in the brainstem nucleus of the solitary tract (NTS) and is relayed to other CNS sites for integration into autonomic responses and complex behaviors. Little is known about the organization or nature of processing within NTS. We injected fluorescent retrograde tracers into two nuclei to identify neurons that project to sites involved in autonomic regulation: the caudal ventrolateral medulla (CVLM) or paraventricular nucleus of the hypothalamus (PVN). We found distinct differences in synaptic connections and performance in the afferent path through NTS to these neurons. Anatomical studies using confocal and electron microscopy found prominent, primary afferent synapses directly on somata and dendrites of CVLM-projecting NTS neurons identifying them as second-order neurons. In brainstem slices, afferent activation evoked large, constant latency EPSCs in CVLM-projecting NTS neurons that were consistent with the precise timing and rare failures of monosynaptic contacts on second-order neurons. In contrast, most PVN-projecting NTS neurons lacked direct afferent input and responded to afferent stimuli with highly variable, intermittently failing synaptic responses, indicating polysynaptic pathways to higher-order neurons. The afferent-evoked EPSCs in most PVN-projecting NTS neurons were smaller and unreliable but also often included multiple, convergent polysynaptic responses not observed in CVLM-projecting neurons. A few PVN-projecting NTS neurons had monosynaptic EPSC characteristics. Together, we found that cranial visceral afferent pathways are structured distinctly within NTS depending on the projection target. Such, intra-NTS pathway architecture will substantially impact performance of autonomic or neuroendocrine reflex arcs.


Assuntos
Nervos Cranianos/fisiologia , Bulbo/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Núcleo Solitário/fisiologia , Sinapses/fisiologia , Fibras Aferentes Viscerais/fisiologia , Potenciais de Ação/fisiologia , Animais , Vias Autônomas/fisiologia , Vias Autônomas/ultraestrutura , Nervos Cranianos/ultraestrutura , Potenciais Pós-Sinápticos Excitadores/fisiologia , Corantes Fluorescentes , Masculino , Bulbo/anatomia & histologia , Bulbo/ultraestrutura , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Vias Neurais/fisiologia , Vias Neurais/ultraestrutura , Técnicas de Cultura de Órgãos , Núcleo Hipotalâmico Paraventricular/anatomia & histologia , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Formação Reticular/anatomia & histologia , Formação Reticular/fisiologia , Formação Reticular/ultraestrutura , Núcleo Solitário/anatomia & histologia , Núcleo Solitário/ultraestrutura , Sinapses/ultraestrutura , Transmissão Sináptica/fisiologia , Fibras Aferentes Viscerais/ultraestrutura
18.
Neurosci Behav Physiol ; 36(8): 847-50, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16964462

RESUMO

Structures of the pancreas and magnocellular (supraoptic and paraventricular) nuclei of the hypothalamus of adult male rats were studied in conditions of acute pancreatitis at the light and electron microscopic levels. Histo-and organotypic changes in the parenchymatous and stromal elements of the pancreas were analyzed simultaneously with cytological assessment of the state of the nonapeptidergic neurosecretory centers of the hypothalamus. Blockade of the release of hypothalamic nonapeptides was found to occur at the level of axovasal complexes, and this aggravated the outcome of destructive and necrobiotic changes in the pathologically altered organ.


Assuntos
Neurônios/patologia , Pancreatite/patologia , Núcleo Hipotalâmico Paraventricular/patologia , Núcleo Supraóptico/patologia , Doença Aguda , Animais , Bile/química , Contagem de Células , Coledocolitíase/complicações , Modelos Animais de Doenças , Humanos , Masculino , Microscopia Eletrônica de Transmissão/métodos , Neurônios/ultraestrutura , Pancreatite/induzido quimicamente , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Ratos , Núcleo Supraóptico/ultraestrutura , Fatores de Tempo
19.
Neurosci Behav Physiol ; 36(5): 463-6, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16645758

RESUMO

Light and electron microscopic methods were used to analyze changes in secretory neurons in the supraoptic (SON) and paraventricular (PVN) nuclei in the hypothalamus in 100 adult male rats at time points from the first minutes to 180 days after 48 hours of full-time exposure to bright light. At the early time points after exposure, the cellular formulae of the SON and PVN shifted towards functionally active neurons with minimal quantities of secretory granules, large nuclei and nucleoli, low RNA contents, small numbers of rough endoplasmic reticulum cisterns, vacuoles, and lysosomes in the perikarya. The number of cells depositing secretion was greater than in controls at 24 h in the SON and PVN and at 10 days in the SON. Normalization of the cellular formula and the structural organization of the protein-synthesizing apparatus of PVN neurons occurred at 10-30 days, with normalization in the SON at 30-180 days. These data provide evidence that the range of plasticity of neurons in the PVN on exposure to full-time bright light was more significant than that in the SON.


Assuntos
Plasticidade Neuronal/fisiologia , Neurônios/ultraestrutura , Sistemas Neurossecretores/ultraestrutura , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Núcleo Supraóptico/ultraestrutura , Adaptação Fisiológica , Animais , Grânulos Citoplasmáticos/ultraestrutura , Retículo Endoplasmático/ultraestrutura , Complexo de Golgi/ultraestrutura , Luz , Masculino , Plasticidade Neuronal/efeitos da radiação , Neurônios/efeitos da radiação , Sistemas Neurossecretores/fisiologia , Sistemas Neurossecretores/efeitos da radiação , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos da radiação , Fotoperíodo , Ratos , Núcleo Supraóptico/metabolismo , Núcleo Supraóptico/efeitos da radiação , Vasopressinas/metabolismo , Vasopressinas/efeitos da radiação
20.
Radiats Biol Radioecol ; 45(5): 599-604, 2005.
Artigo em Russo | MEDLINE | ID: mdl-16304776

RESUMO

The structural changes of neurons of the rat hypothalamic supraoptic (SON) and of paraventricular (PVN) nucleus after 48 h of bright light exposure, of 5 Gy whole-body X-irradiation and of their combination subjected to the analysis by means of light-optic and of electron microscopy for the estimation of radimodificated effect of light exposure lasted 24 h a day and plasticity of neuroendocrine transducers interacted with the optic sensory system. The structural changes of neurons of the SON after combined action are less considerable and more prolonged in comparison with the PVN that loas defermined by their direct connection with the optic sensory system via the retinohypothalamic tract.


Assuntos
Luz , Sistemas Neurossecretores/efeitos dos fármacos , Sistemas Neurossecretores/efeitos da radiação , Raios X , Animais , Hipotálamo/efeitos dos fármacos , Hipotálamo/efeitos da radiação , Hipotálamo/ultraestrutura , Masculino , Microscopia Eletrônica , Sistemas Neurossecretores/ultraestrutura , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos da radiação , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Ratos , Irradiação Corporal Total
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...