Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 11(1): 5692, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33173030

RESUMO

The control of water-intake behavior is critical for life because an excessive water intake induces pathological conditions, such as hyponatremia or water intoxication. However, the brain mechanisms controlling water intake currently remain unclear. We previously reported that thirst-driving neurons (water neurons) in the subfornical organ (SFO) are cholecystokinin (CCK)-dependently suppressed by GABAergic interneurons under Na-depleted conditions. We herein show that CCK-producing excitatory neurons in the SFO stimulate the activity of GABAergic interneurons via CCK-B receptors. Fluorescence-microscopic Ca2+ imaging demonstrates two distinct subpopulations in CCK-positive neurons in the SFO, which are persistently activated under hyponatremic conditions or transiently activated in response to water drinking, respectively. Optical and chemogenetic silencings of the respective types of CCK-positive neurons both significantly increase water intake under water-repleted conditions. The present study thus reveals CCK-mediated neural mechanisms in the central nervous system for the control of water-intake behaviors.


Assuntos
Comportamento de Ingestão de Líquido/fisiologia , Neurônios/fisiologia , Órgão Subfornical/citologia , Animais , Masculino , Camundongos , Microscopia de Fluorescência/métodos , Sódio/metabolismo , Órgão Subfornical/fisiologia
2.
Nature ; 588(7836): 112-117, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33057193

RESUMO

Fluid intake is an essential innate behaviour that is mainly caused by two distinct types of thirst1-3. Increased blood osmolality induces osmotic thirst that drives animals to consume pure water. Conversely, the loss of body fluid induces hypovolaemic thirst, in which animals seek both water and minerals (salts) to recover blood volume. Circumventricular organs in the lamina terminalis are critical sites for sensing both types of thirst-inducing stimulus4-6. However, how different thirst modalities are encoded in the brain remains unknown. Here we employed stimulus-to-cell-type mapping using single-cell RNA sequencing to identify the cellular substrates that underlie distinct types of thirst. These studies revealed diverse types of excitatory and inhibitory neuron in each circumventricular organ structure. We show that unique combinations of these neuron types are activated under osmotic and hypovolaemic stresses. These results elucidate the cellular logic that underlies distinct thirst modalities. Furthermore, optogenetic gain of function in thirst-modality-specific cell types recapitulated water-specific and non-specific fluid appetite caused by the two distinct dipsogenic stimuli. Together, these results show that thirst is a multimodal physiological state, and that different thirst states are mediated by specific neuron types in the mammalian brain.


Assuntos
Neurônios/classificação , Neurônios/fisiologia , Sede/fisiologia , Animais , Sequência de Bases , Ingestão de Líquidos/fisiologia , Feminino , Hipovolemia/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Organum Vasculosum/citologia , Organum Vasculosum/fisiologia , Pressão Osmótica , Análise de Célula Única , Órgão Subfornical/citologia , Órgão Subfornical/fisiologia , Privação de Água
3.
Neuron ; 103(2): 242-249.e4, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31153646

RESUMO

For thirsty animals, fluid intake provides both satiation and pleasure of drinking. How the brain processes these factors is currently unknown. Here, we identified neural circuits underlying thirst satiation and examined their contribution to reward signals. We show that thirst-driving neurons receive temporally distinct satiation signals by liquid-gulping-induced oropharyngeal stimuli and gut osmolality sensing. We demonstrate that individual thirst satiation signals are mediated by anatomically distinct inhibitory neural circuits in the lamina terminalis. Moreover, we used an ultrafast dopamine (DA) sensor to examine whether thirst satiation itself stimulates the reward-related circuits. Interestingly, spontaneous drinking behavior but not thirst drive reduction triggered DA release. Importantly, chemogenetic stimulation of thirst satiation neurons did not activate DA neurons under water-restricted conditions. Together, this study dissected the thirst satiation circuit, the activity of which is functionally separable from reward-related brain activity.


Assuntos
Neurônios GABAérgicos/fisiologia , Rede Nervosa/fisiologia , Vias Neurais/fisiologia , Saciação/fisiologia , Estômago/inervação , Órgão Subfornical/citologia , Animais , Cálcio/metabolismo , Dopamina/metabolismo , Ingestão de Líquidos/fisiologia , Feminino , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo I/metabolismo , Optogenética , Concentração Osmolar , Fragmentos de Peptídeos/metabolismo , Estimulação Física
4.
J Neuroendocrinol ; 30(12): e12654, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30365188

RESUMO

The subfornical organ (SFO) lacks the normal blood-brain barrier and senses the concentrations of many different circulating signals, including glucose and angiotensin II (ANG II). ANG II has recently been implicated in the control of food intake and body weight gain. The present study assessed whether single SFO neurones sense changes in glucose and ANG II, and also whether changes in glucose concentration alter the responsiveness of these neurones to ANG II. SFO neurones dissociated from male Sprague-Dawley rats (100-175 g) were used. We first examined whether glucose concentration modulates AT1 receptor expression. Similar AT1a mRNA expression levels were found at glucose concentrations of 1, 5 and 10 mmol L-1 in dissociated SFO neurones. Glucose responsiveness of SFO neurones was assessed using perforated current-clamp recordings and switching between 5 and 10 mmol L-1 glucose artificial cerebrospinal fluid to classify single neurones as nonresponsive (nGS), glucose-excited (GE) or glucose-inhibited (GI). In total, 26.7% of the SFO neurones were GI (n = 24 of 90), 21.1% were GE (n = 19 of 90) and 52.2% were nGS (n = 47 of 90). Once classified, the effects of 10 nmol L-1 ANG II on the excitability of these neurones were tested, with 52% of GE (n = 10 of 19), 71% of GI (n = 17 of 24) and 43% of nGS (n = 20 of 47) neurones being ANG II sensitive. Finally, we tested whether acute changes in glucose concentration modified the response to ANG II and showed that some neurones (4/17) only respond to ANG II at 10 mmol L-1 glucose. Our data demonstrate that the same SFO neurone can sense glucose and ANG II and that acute changes in glucose concentration may change ANG II responsiveness.


Assuntos
Angiotensina II/farmacologia , Glucose/metabolismo , Glucose/farmacologia , Órgão Subfornical/efeitos dos fármacos , Órgão Subfornical/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Interações Medicamentosas , Masculino , Potenciais da Membrana/efeitos dos fármacos , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Ratos , Receptor Tipo 1 de Angiotensina/biossíntese , Órgão Subfornical/citologia
5.
J Neurophysiol ; 120(5): 2269-2281, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30089060

RESUMO

Subfornical organ (SFO) neurons exhibit heterogeneity in current expression and spiking behavior, where the two major spiking phenotypes appear as tonic and burst firing. Insight into the mechanisms behind this heterogeneity is critical for understanding how the SFO, a sensory circumventricular organ, integrates and selectively influences physiological function. To integrate efficient methods for studying this heterogeneity, we built a single-compartment, Hodgkin-Huxley-type model of an SFO neuron that is parameterized by SFO-specific in vitro patch-clamp data. The model accounts for the membrane potential distribution and spike train variability of both tonic and burst firing SFO neurons. Analysis of model dynamics confirms that a persistent Na+ and Ca2+ currents are required for burst initiation and maintenance and suggests that a slow-activating K+ current may be responsible for burst termination in SFO neurons. Additionally, the model suggests that heterogeneity in current expression and subsequent influence on spike afterpotential underlie the behavioral differences between tonic and burst firing SFO neurons. Future use of this model in coordination with single neuron patch-clamp electrophysiology provides a platform for explaining and predicting the response of SFO neurons to various combinations of circulating signals, thus elucidating the mechanisms underlying physiological signal integration within the SFO. NEW & NOTEWORTHY Our understanding of how the subfornical organ (SFO) selectively influences autonomic nervous system function remains incomplete but theoretically results from the electrical responses of SFO neurons to physiologically important signals. We have built a computational model of SFO neurons, derived from and supported by experimental data, which explains how SFO neurons produce different electrical patterns. The model provides an efficient system to theoretically and experimentally explore how changes in the essential features of SFO neurons affect their electrical activity.


Assuntos
Potenciais de Ação , Canais de Cálcio/metabolismo , Modelos Neurológicos , Neurônios/fisiologia , Canais de Sódio/metabolismo , Órgão Subfornical/fisiologia , Animais , Células Cultivadas , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Órgão Subfornical/citologia , Órgão Subfornical/metabolismo
6.
Am J Physiol Regul Integr Comp Physiol ; 315(3): R425-R433, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29668324

RESUMO

Inflammation is thought to play a fundamental role in the pathophysiology of hypertension and heart failure, although the mechanisms for this remain unclear. Proinflammatory cytokines, such as tumor necrosis factor-α (TNF-α), influence the subfornical organ (SFO) to modulate sympathetic activity and blood pressure. The pressor effects of TNF-α in the SFO are partially mediated by angiotensin II (ANG II) receptor type 1 (AT1R), and TNF-α is known to potentiate ANG II-induced hypertension. However, the cellular mechanism of the interaction between TNF-α and ANG II/AT1R signaling remains unknown. In the present study, we performed Ca2+ imaging on dissociated SFO neurons in vitro from male Sprague-Dawley rats to determine whether TNF-α modulates ANG II-induced increases in intracellular Ca2+ in SFO neurons. We first established that a proportion of SFO neurons respond to ANG II, an effect that required AT1R signaling and extracellular Ca2+. We then tested the hypothesis that TNF-α may modulate the effects of ANG II on SFO neurons by examining the effects of TNF-α treatment on the ANG II-induced rise in intracellular Ca2+. We discovered that TNF-α potentiated the ANG II-induced rise in intracellular Ca2+, an effect that was dependent on the duration of TNF-α treatment. Finally, we determined that this potentiation of ANG II-induced Ca2+ activity relied on tetrodotoxin-sensitive voltage-gated Na+ (vgNa+) channels. These data suggest that the potentiation of ANG II/AT1R activity by TNF-α in SFO neurons results from the previously demonstrated ability of this cytokine to modulate the activation threshold of vgNa+ currents.


Assuntos
Angiotensina II/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Órgão Subfornical/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Células Cultivadas , Sinergismo Farmacológico , Masculino , Potenciais da Membrana , Neurônios/metabolismo , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/agonistas , Receptor Tipo 1 de Angiotensina/metabolismo , Órgão Subfornical/citologia , Órgão Subfornical/metabolismo , Fatores de Tempo , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Canais de Sódio Disparados por Voltagem/metabolismo
7.
Nature ; 555(7695): 204-209, 2018 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-29489747

RESUMO

Neural circuits for appetites are regulated by both homeostatic perturbations and ingestive behaviour. However, the circuit organization that integrates these internal and external stimuli is unclear. Here we show in mice that excitatory neural populations in the lamina terminalis form a hierarchical circuit architecture to regulate thirst. Among them, nitric oxide synthase-expressing neurons in the median preoptic nucleus (MnPO) are essential for the integration of signals from the thirst-driving neurons of the subfornical organ (SFO). Conversely, a distinct inhibitory circuit, involving MnPO GABAergic neurons that express glucagon-like peptide 1 receptor (GLP1R), is activated immediately upon drinking and monosynaptically inhibits SFO thirst neurons. These responses are induced by the ingestion of fluids but not solids, and are time-locked to the onset and offset of drinking. Furthermore, loss-of-function manipulations of GLP1R-expressing MnPO neurons lead to a polydipsic, overdrinking phenotype. These neurons therefore facilitate rapid satiety of thirst by monitoring real-time fluid ingestion. Our study reveals dynamic thirst circuits that integrate the homeostatic-instinctive requirement for fluids and the consequent drinking behaviour to maintain internal water balance.


Assuntos
Ingestão de Líquidos/fisiologia , Vias Neurais , Área Pré-Óptica/citologia , Área Pré-Óptica/fisiologia , Órgão Subfornical/citologia , Órgão Subfornical/fisiologia , Sede/fisiologia , Animais , Apetite/fisiologia , Feminino , Neurônios GABAérgicos/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Homeostase , Instinto , Masculino , Camundongos , Óxido Nítrico Sintase/metabolismo , Resposta de Saciedade/fisiologia , Equilíbrio Hidroeletrolítico
8.
J Neurophysiol ; 118(3): 1532-1541, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28637815

RESUMO

Tumor necrosis factor-α (TNF-α) is a proinflammatory cytokine implicated in cardiovascular and autonomic regulation via actions in the central nervous system. TNF-α-/- mice do not develop angiotensin II (ANG II)-induced hypertension, and administration of TNF-α into the bloodstream of rats increases blood pressure and sympathetic tone. Recent studies have shown that lesion of the subfornical organ (SFO) attenuates the hypertensive and autonomic effects of TNF-α, while direct administration of TNF-α into the SFO increases blood pressure, suggesting the SFO to be a key site for the actions of TNF-α. Therefore, we used patch-clamp techniques to examine both acute and long-term effects of TNF-α on the excitability of Sprague-Dawley rat SFO neurons. It was observed that acute bath application of TNF-α depolarized SFO neurons and subsequently increased action potential firing rate. Furthermore, the magnitude of depolarization and the proportion of depolarized SFO neurons were concentration dependent. Interestingly, following 24-h incubation with TNF-α, the basal firing rate of the SFO neurons was increased and the rheobase was decreased, suggesting that TNF-α elevates SFO neuron excitability. This effect was likely mediated by the transient sodium current, as TNF-α increased the magnitude of the current and lowered its threshold of activation. In contrast, TNF-α did not appear to modulate either the delayed rectifier potassium current or the transient potassium current. These data suggest that acute and long-term TNF-α exposure elevates SFO neuron activity, providing a basis for TNF-α hypertensive and sympathetic effects.NEW & NOTEWORTHY Considerable recent evidence has suggested important links between inflammation and the pathological mechanisms underlying hypertension. The present study describes cellular mechanisms through which acute and long-term exposure of tumor necrosis factor-α (TNF-α) influences the activity of subfornical organ neurons by modulating the voltage-gated transient Na+ current. This provides critical new information regarding the specific pathological mechanisms through which inflammation and TNF-α in particular may result in the development of hypertension.


Assuntos
Potenciais de Ação , Neurônios/efeitos dos fármacos , Órgão Subfornical/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Células Cultivadas , Masculino , Neurônios/metabolismo , Neurônios/fisiologia , Canais de Potássio/metabolismo , Ratos , Ratos Sprague-Dawley , Canais de Sódio/metabolismo , Órgão Subfornical/citologia , Órgão Subfornical/fisiologia
9.
Exp Brain Res ; 235(4): 1053-1062, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28070623

RESUMO

It is known that the median preoptic nucleus (POMe) sends dense projections to the subfornical organ (SFO). However, the functional significance of these projections have not been well discussed. In this electron microscopic study, we investigated the presence of synapses between POMe-derived axon terminals and SFO neurons that project to the paraventricular hypothalamic nucleus (PVN). After injection of a retrograde tracer, wheat germ agglutinin-conjugated horseradish peroxidase-colloidal gold complex, into the PVN, many labeled neurons were found in the SFO. In contrast, after injection of an anterograde tracer, biotinylated dextran amine, in the POMe, abundant labeled axon varicosities were observed in the SFO. Using electron microscopy, synapses were identified between retrogradely labeled dendrites and cell bodies, and anterogradely labeled axon terminals, indicating that POMe neurons innervate SFO neurons projecting to the PVN. The possibility that POMe neurons play multiple roles in the neuronal circuit responsible for vasopressin release and/or cardiovascular regulation is also discussed.


Assuntos
Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/citologia , Área Pré-Óptica/citologia , Órgão Subfornical/citologia , Sinapses/fisiologia , Animais , Biotina/análogos & derivados , Biotina/metabolismo , Dextranos/metabolismo , Coloide de Ouro/metabolismo , Masculino , Microinjeções , Microscopia Imunoeletrônica , Vias Neurais/fisiologia , Neurônios/metabolismo , Neurônios/ultraestrutura , Ratos , Ratos Sprague-Dawley , Sinapses/ultraestrutura , Aglutininas do Germe de Trigo/metabolismo
10.
Nature ; 537(7622): 680-684, 2016 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-27487211

RESUMO

Thirst motivates animals to drink in order to maintain fluid balance. Thirst has conventionally been viewed as a homeostatic response to changes in blood volume or tonicity. However, most drinking behaviour is regulated too rapidly to be controlled by blood composition directly, and instead seems to anticipate homeostatic imbalances before they arise. How this is achieved remains unknown. Here we reveal an unexpected role for the subfornical organ (SFO) in the anticipatory regulation of thirst in mice. By monitoring deep-brain calcium dynamics, we show that thirst-promoting SFO neurons respond to inputs from the oral cavity during eating and drinking and then integrate these inputs with information about the composition of the blood. This integration allows SFO neurons to predict how ongoing food and water consumption will alter fluid balance in the future and then to adjust behaviour pre-emptively. Complementary optogenetic manipulations show that this anticipatory modulation is necessary for drinking in several contexts. These findings provide a neural mechanism to explain longstanding behavioural observations, including the prevalence of drinking during meals, the rapid satiation of thirst, and the fact that oral cooling is thirst-quenching.


Assuntos
Ingestão de Líquidos/fisiologia , Ingestão de Alimentos/fisiologia , Homeostase , Neurônios/fisiologia , Órgão Subfornical/citologia , Sede/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia , Animais , Sangue , Cálcio/metabolismo , Retroalimentação Fisiológica , Feminino , Masculino , Camundongos , Boca/inervação , Boca/fisiologia , Vias Neurais , Optogenética , Órgão Subfornical/fisiologia , Fatores de Tempo
11.
J Neurophysiol ; 115(6): 3123-9, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27030736

RESUMO

The subfornical organ (SFO) plays a pivotal role in body fluid homeostasis through its ability to integrate neurohumoral signals and subsequently alter behavior, neuroendocrine function, and autonomic outflow. The purpose of the present study was to evaluate whether selective activation of SFO neurons using virally mediated expression of Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) stimulated thirst and salt appetite. Male C57BL/6 mice (12-15 wk) received an injection of rAAV2-CaMKII-HA-hM3D(Gq)-IRES-mCitrine targeted at the SFO. Two weeks later, acute injection of clozapine N-oxide (CNO) produced dose-dependent increases in water intake of mice with DREADD expression in the SFO. CNO also stimulated the ingestion of 0.3 M NaCl. Acute injection of CNO significantly increased the number of Fos-positive nuclei in the SFO of mice with robust DREADD expression. Furthermore, in vivo single-unit recordings demonstrate that CNO significantly increases the discharge frequency of both ANG II- and NaCl-responsive neurons. In vitro current-clamp recordings confirm that bath application of CNO produces a significant membrane depolarization and increase in action potential frequency. In a final set of experiments, chronic administration of CNO approximately doubled 24-h water intake without an effect on salt appetite. These findings demonstrate that DREADD-induced activation of SFO neurons stimulates thirst and that DREADDs are a useful tool to acutely or chronically manipulate neuronal circuits influencing body fluid homeostasis.


Assuntos
Apetite/efeitos dos fármacos , Drogas Desenhadas/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Cloreto de Sódio/metabolismo , Órgão Subfornical/efeitos dos fármacos , Sede/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Angiotensina II/farmacologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Clozapina/análogos & derivados , Clozapina/farmacologia , Ingestão de Líquidos/efeitos dos fármacos , Ingestão de Líquidos/genética , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Proteínas Oncogênicas v-fos/metabolismo , Receptores Acoplados a Proteínas G/genética , Solução Salina Hipertônica/administração & dosagem , Órgão Subfornical/citologia
12.
J Neurophysiol ; 114(3): 1641-51, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26180118

RESUMO

Hydrogen sulfide (H2S) is an endogenously found gasotransmitter that has been implicated in a variety of beneficial physiological functions. This study was performed to investigate the cellular mechanisms underlying actions of H2S previously observed in subfornical organ (SFO), where H2S acts to regulate blood pressure through a depolarization of the membrane and an overall increase in the excitability of SFO neurons. We used whole cell patch-clamp electrophysiology in the voltage-clamp configuration to analyze the effect of 1 mM NaHS, an H2S donor, on voltage-gated potassium, sodium, and calcium currents. We observed no effect of NaHS on potassium currents; however, both voltage-gated sodium currents (persistent and transient) and the N-type calcium current had a depolarized activation curve and an enhanced peak-induced current in response to a series of voltage-step and ramp protocols run in the control and NaHS conditions. These effects were not responsible for the previously observed depolarization of the membrane potential, as depolarizing effects of H2S were still observed following block of these conductances with tetrodotoxin (5 µM) and ω-conotoxin-GVIA (100 nM). Our studies are the first to investigate the effect of H2S on a variety of voltage-gated conductances in a single brain area, and although they do not explain mechanisms underlying the depolarizing actions of H2S on SFO neurons, they provide evidence of potential mechanisms through which this gasotransmitter influences the excitability of neurons in this important brain area as a consequence of the modulation of multiple ion channels.


Assuntos
Sulfeto de Hidrogênio/metabolismo , Neurônios/metabolismo , Sódio/metabolismo , Órgão Subfornical/metabolismo , Canais de Sódio Disparados por Voltagem/metabolismo , Potenciais de Ação , Animais , Cálcio/metabolismo , Células Cultivadas , Masculino , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Órgão Subfornical/citologia , Órgão Subfornical/fisiologia , Sulfetos/farmacologia
13.
Physiol Behav ; 147: 291-9, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25911266

RESUMO

The brainstem plays an important role in controlling sodium and water homeostasis. It is a major regulatory site for autonomic and motor functions. Moreover, it integrates cerebrospinal fluid (CSF) signals with neuronal and hormonal signals. Evidence suggests that the CSF-contacting nucleus (CSF-CN) transmits and integrates CSF signals, but, the definitive role of CSF-CN in sodium homeostasis is poorly understood. In this study, we used c-Fos as a marker of neuronal activity and causing colocalization of Nax channel and 5-HT. This proved that CSF-CN played a role in sensing the increase of CSF sodium level. Then, we determined the role of the CSF-contacting nucleus in increasing the sodium appetite of rats. So, we performed targeted lesion of the CSF-contacting nucleus in the brainstem using the cholera toxin subunit B-saporin (CB-SAP), a cytotoxin coupled to cholera toxin subunit B. The lesion of the CSF-CN showed decreased and degenerative neurons, while sodium appetite have increased and Fos immunocytochemistry detected neuronal activity in the lateral parabrachial nucleus (LPBN), but not in the subfornical organ (SFO) and organum vasculosum of the lamina terminalis (OVLT). These results indicate that the CSF-CN plays an important role in sensing CSF sodium level and satiating sodium appetite by influencing the LPBN but not SFO and OVLT. The Nax channel and 5-HT might be the molecular mechanisms through which contribute to sodium homeostasis.


Assuntos
Apetite/fisiologia , Tronco Encefálico/metabolismo , Líquido Cefalorraquidiano , Neurônios/fisiologia , Sódio na Dieta/administração & dosagem , Órgão Subfornical/fisiologia , Animais , Apetite/efeitos dos fármacos , Tronco Encefálico/citologia , Tronco Encefálico/efeitos dos fármacos , Toxina da Cólera/farmacocinética , Ingestão de Líquidos , Furosemida/farmacologia , Peroxidase do Rábano Silvestre/metabolismo , Injeções Intraventriculares , Masculino , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Inativadoras de Ribossomos Tipo 1/farmacocinética , Solução Salina Hipertônica/farmacologia , Saporinas , Serotonina/metabolismo , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Órgão Subfornical/citologia , Canais de Sódio Disparados por Voltagem/metabolismo
14.
J Histochem Cytochem ; 63(5): 367-83, 2015 05.
Artigo em Inglês | MEDLINE | ID: mdl-25673286

RESUMO

The subfornical organ (SFO) is a circumventricular organ with a chemosensitive function, and its vessels have no blood-brain barrier. Our study investigated the glial and vascular components in the SFO to determine whether their distributions indicate subdivisions, how to characterize the vessels and how to demarcate the SFO. To this end, we investigated glial markers (GFAP, glutamine synthetase, S100) and other markers, including vimentin and nestin (immature glia), laminin (basal lamina), ß-dystroglycan (glio-vascular connections), and aquaporin 4 (glial water channels). We determined that the 'shell' of the SFO was marked by immunoreactivity for S100, GFAP and aquaporin 4. Nestin immunoreactivity was characteristic of the 'core'. Vimentin was almost evenly distributed. Glutamine synthetase immunoreactivity occurred in the shell but its expression was sparse. Vessels in the core were decorated with laminin but showed a discontinuous expression of aquaporin 4. Vimentin and GFAP staining was usually in separate glial elements, which may be related to their functional differences. Similar to other vessels in the brain, ß-dystroglycan was detected along the shell vessels but laminin was not. The gradual disappearance of the laminin immunopositivity was attributed to the gradual disappearance of the perivascular space. Thus, our findings suggest that the shell and core glio-vascular structures are adapted to different sensory functions: osmoperception and the perception of circulating peptides, respectively.


Assuntos
Distroglicanas/metabolismo , Neuroglia/citologia , Órgão Subfornical/irrigação sanguínea , Órgão Subfornical/citologia , Animais , Aquaporina 4/metabolismo , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Glutamato-Amônia Ligase/metabolismo , Laminina/metabolismo , Masculino , Nestina/metabolismo , Neuroglia/metabolismo , Ratos Wistar , Proteínas S100/metabolismo , Órgão Subfornical/metabolismo , Vimentina/metabolismo
15.
Nature ; 520(7547): 349-52, 2015 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-25624099

RESUMO

Thirst is the basic instinct to drink water. Previously, it was shown that neurons in several circumventricular organs of the hypothalamus are activated by thirst-inducing conditions. Here we identify two distinct, genetically separable neural populations in the subfornical organ that trigger or suppress thirst. We show that optogenetic activation of subfornical organ excitatory neurons, marked by the expression of the transcription factor ETV-1, evokes intense drinking behaviour, and does so even in fully water-satiated animals. The light-induced response is highly specific for water, immediate and strictly locked to the laser stimulus. In contrast, activation of a second population of subfornical organ neurons, marked by expression of the vesicular GABA transporter VGAT, drastically suppresses drinking, even in water-craving thirsty animals. These results reveal an innate brain circuit that can turn an animal's water-drinking behaviour on and off, and probably functions as a centre for thirst control in the mammalian brain.


Assuntos
Comportamento de Ingestão de Líquido/fisiologia , Órgão Subfornical/citologia , Órgão Subfornical/fisiologia , Sede/fisiologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Desidratação/fisiopatologia , Ingestão de Líquidos , Água Potável , Lasers , Camundongos , Optogenética , Resposta de Saciedade , Fatores de Transcrição/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
16.
J Neuroendocrinol ; 26(4): 237-46, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24612143

RESUMO

Nesfatin-1, a centrally acting anorexigenic peptide, is produced in several brain areas involved in metabolic processes and has been implicated in the control of ingestive behaviours and cardiovascular functions. The present study aimed to determine whether the subfornical organ (SFO), a central nervous system (CNS) site that has been extensively implicated in the regulation of appetite and thirst, may represent a potential site for central actions of nesfatin-1. We first used the reverse transcriptase-polymerase chain reaction and were able to confirm the presence of mRNA for the nucleobindin-2 gene in the SFO. We then used whole-cell patch clamp recordings to investigate the influence of nesfatin-1 on the membrane potential of dissociated SFO neurones. A total of 80.3% (49 of 61) of neurones tested showed a response to nesfatin-1 (100 nm, 10 nm and 1 nm). Of these, 47.5% depolarised, with a mean depolarisation of 8.2 ± 0.9 mV (n = 29) and 32.8% hyperpolarised with a mean hyperpolarisation of -8.9 ± 1.2 mV (n = 20). Peak magnitudes were seen at a concentration of 1 nm nesfatin-1, whereas no effect was observed at 100 pm nesftain-1 (n = 3). Furthermore, voltage clamp ramp and step protocols revealed a nesfatin-1 induced activation of the delayed rectifier potassium conductance, IK . Pharmacological blockade of this conductance greatly reduced the magnitude and occurrence of the observed hyperpolarisations. The present study thus demonstrates that nesfatin-1 has the ability to influence the membrane potential of SFO neurones, and thus identifies the SFO as a potential site at which nesfatin-1 may act to regulate ingestive behaviour and cardiovascular control.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Órgão Subfornical/citologia , Animais , Masculino , Neurônios/fisiologia , Nucleobindinas , Canais de Potássio/fisiologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Am J Physiol Regul Integr Comp Physiol ; 306(5): R363-73, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24430886

RESUMO

The subfornical organ (SFO) is an important sensory circumventricular organ implicated in the regulation of fluid homeostasis and energy balance. We investigated whether the SFO is activated by the hormone cholecystokinin (CCK). CCK1 and CCK2 receptors were identified in the SFO by RT-PCR. Dissociated SFO neurons that responded to CCK (40/77), were mostly depolarized (9.2 ± 0.9 mV, 30/77), but some were hyperpolarized (-7.3 ± 1.1 mV, 10/77). We next examined the responses of SFO neurons in vivo to CCK (16 µg/kg ip), in the presence and absence of CCK1 or CCK2 receptor antagonists (devazepide; 600 µg/kg and L-365,260; 100 µg/kg, respectively), using the functional activation markers c-Fos and phosphorylated extracellular signal-related kinase (p-ERK). The nucleus of the solitary tract (NTS) served as a control for CCK-induced activity. There was a significant increase in c-Fos expression in the NTS (259.2 ± 20.8 neurons) compared with vehicle (47.5 ± 2.5). Similarly, in the SFO, c-Fos was expressed in 40.5 ± 10.6 neurons in CCK-treated compared with 6.6 ± 2.7 in vehicle-treated rats (P < 0.01). Devazepide significantly reduced the effects of CCK in the NTS but not in SFO. L-365,260 blocked the effects of CCK in both brain regions. CCK increased the number of p-ERK neurons in NTS (27.0 ± 4.0) as well as SFO (18.0 ± 4.0), compared with vehicle (8.0 ± 2.6 and 4.3 ± 0.6, respectively; P < 0.05). Both devazepide and L-365,260 reduced CCK-induced p-ERK in NTS, but only L-365,260 reduced it in the SFO. In conclusion, the SFO represents a novel brain region at which circulating CCK may act via CCK2 receptors to influence central autonomic control.


Assuntos
Colecistocinina/farmacologia , Fragmentos de Peptídeos/farmacologia , Órgão Subfornical/efeitos dos fármacos , Animais , Benzodiazepinonas/farmacologia , Devazepida/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica/fisiologia , Genes fos/genética , Genes fos/fisiologia , Antagonistas de Hormônios/farmacologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Compostos de Fenilureia/farmacologia , RNA/genética , RNA/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores da Colecistocinina/antagonistas & inibidores , Receptores da Colecistocinina/genética , Receptores da Colecistocinina/metabolismo , Órgão Subfornical/citologia , Órgão Subfornical/fisiologia
18.
Cell Biochem Funct ; 32(1): 51-61, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23629811

RESUMO

The blood-brain barrier (BBB) is a barrier that prevents free access of blood-derived substances to the brain through the tight junctions and maintains a specialized brain environment. Circumventricular organs (CVOs) lack the typical BBB. The fenestrated vasculature of the sensory CVOs, including the organum vasculosum of the lamina terminalis (OVLT), subfornical organ (SFO) and area postrema (AP), allows parenchyma cells to sense a variety of blood-derived information, including osmotic ones. In the present study, we utilized immunohistochemistry to examine changes in the expression of NG2 and platelet-derived growth factor receptor beta (PDGFRB) in the OVLT, SFO and AP of adult mice during chronic osmotic stimulation. The expression of NG2 and PDGFRB was remarkably prominent in pericytes, although these angiogenesis-associated proteins are highly expressed at pericytes of developing immature vasculature. The chronic salt loading prominently increased the expression of NG2 in the OVLT and SFO and that of PDGFRB in the OVLT, SFO and AP. The vascular permeability of low-molecular-mass tracer fluorescein isothiocyanate was increased significantly by chronic salt loading in the OVLT and SFO but not AP. In conclusion, the present study demonstrates changes in pericyte expression of NG2 and PDGFRB and vascular permeability in the sensory CVOs by chronic osmotic stimulation, indicating active participation of the vascular system in osmotic homeostasis.


Assuntos
Antígenos/metabolismo , Área Postrema/metabolismo , Permeabilidade Capilar , Hipotálamo/metabolismo , Pericitos/metabolismo , Proteoglicanas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Órgão Subfornical/metabolismo , Animais , Antígenos/genética , Área Postrema/irrigação sanguínea , Área Postrema/citologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Hipotálamo/irrigação sanguínea , Hipotálamo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Osmorregulação , Pericitos/citologia , Proteoglicanas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Cloreto de Sódio/farmacologia , Órgão Subfornical/irrigação sanguínea , Órgão Subfornical/citologia
19.
Am J Physiol Regul Integr Comp Physiol ; 305(10): R1141-52, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24049115

RESUMO

The sensory circumventricular organs (CVOs) are specialized collections of neurons and glia that lie in the midline of the third and fourth ventricles of the brain, lack a blood-brain barrier, and function as chemosensors, sampling both the cerebrospinal fluid and plasma. These structures, which include the organum vasculosum of the lamina terminalis (OVLT), subfornical organ (SFO), and area postrema (AP), are sensitive to changes in sodium concentration but the cellular mechanisms involved remain unknown. Epithelial sodium channel (ENaC)-expressing neurons of the CVOs may be involved in this process. Here we demonstrate with immunohistochemical and in situ hybridization methods that ENaC-expressing neurons are densely concentrated in the sensory CVOs. These neurons become c-Fos activated, a marker for neuronal activity, after various manipulations of peripheral levels of sodium including systemic injections with hypertonic saline, dietary sodium deprivation, and sodium repletion after prolonged sodium deprivation. The increases seen c-Fos activity in the CVOs were correlated with parallel increases in plasma sodium levels. Since ENaCs play a central role in sodium reabsorption in kidney and other epithelia, we present a hypothesis here suggesting that these channels may also serve a related function in the CVOs. ENaCs could be a significant factor in modulating CVO neuronal activity by controlling the magnitude of sodium permeability in neurons. Hence, some of the same circulating hormones controlling ENaC expression in kidney, such as angiotensin II and atrial natriuretic peptide, may coordinate ENaC expression in sensory CVO neurons and could potentially orchestrate sodium appetite, osmoregulation, and vasomotor sympathetic drive.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Hipotálamo/citologia , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Sódio/farmacologia , Órgão Subfornical/citologia , Animais , Área Postrema/citologia , Canais Epiteliais de Sódio/genética , Feminino , Imuno-Histoquímica , Hibridização In Situ , Masculino , Proteínas Proto-Oncogênicas c-fos/genética , Ratos
20.
Behav Brain Res ; 250: 343-50, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23714076

RESUMO

Nesfatin-1 (Nes-1), an 82-amino acid protein cleaved from nucleobindin-2, has been suggested to play a role in ingestive behaviors. Intracerebroventricular (icv) injections of Nes-1 reduce water intake, although the sites of action for this effect are not known. Two series of experiments were done to identify potential sites of action of Nes-1 in drinking behavior. In the first series, icv injections of Nes-1 were made in urethane-anesthetized rats to investigate the distribution of neurons containing Fos-like immunoreactivity (Fos-ir) within the forebrain. Circumventricular organs, including subfornical organ (SFO), were found to contain neurons expressing Fos-ir. Additionally, several hypothalamic, thalamic and limbic nuclei also contained Fos-labeled neurons. As SFO is a pivotal central site in the regulation of water intake, a second series of experiments was done to investigate the role of direct injections of Nes-1 into SFO on water intake in conscious, freely moving rats. Nes-1 (2pmol) injections into SFO induced an increase in water intake compared to vehicle injections. However, when food was made available for ingestion after the Nes-1 injection, the dipsogenic effects of Nes-1 were attenuated. Additionally, the drinking response to Nes-1 was found to be more potent than that observed after injections of ANG II into SFO. Neither simultaneous injections ANG II nor the ANG II type-1 receptor blocker losartan affected the Nes-1 dipsogenic response. Taken together, these results suggest that Nes-1 is a potent dipsogenic agent in SFO, and that Nes-1 may act independently of the SFO angiotensinergic system to elicit the dipsogenic effect.


Assuntos
Proteínas de Ligação ao Cálcio/farmacologia , Proteínas de Ligação a DNA/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Proteínas do Tecido Nervoso/farmacologia , Prosencéfalo/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Órgão Subfornical/efeitos dos fármacos , Análise de Variância , Angiotensina II/farmacologia , Animais , Injeções Intraventriculares/métodos , Masculino , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Neurônios/metabolismo , Nucleobindinas , Prosencéfalo/citologia , Proteínas Proto-Oncogênicas c-fos/genética , Ratos , Ratos Sprague-Dawley , Órgão Subfornical/citologia , Fatores de Tempo , Vigília
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...