Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 187(8): 1874-1888.e14, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38518773

RESUMO

Infections of the lung cause observable sickness thought to be secondary to inflammation. Signs of sickness are crucial to alert others via behavioral-immune responses to limit contact with contagious individuals. Gram-negative bacteria produce exopolysaccharide (EPS) that provides microbial protection; however, the impact of EPS on sickness remains uncertain. Using genome-engineered Pseudomonas aeruginosa (P. aeruginosa) strains, we compared EPS-producers versus non-producers and a virulent Escherichia coli (E. coli) lung infection model in male and female mice. EPS-negative P. aeruginosa and virulent E. coli infection caused severe sickness, behavioral alterations, inflammation, and hypothermia mediated by TLR4 detection of the exposed lipopolysaccharide (LPS) in lung TRPV1+ sensory neurons. However, inflammation did not account for sickness. Stimulation of lung nociceptors induced acute stress responses in the paraventricular hypothalamic nuclei by activating corticotropin-releasing hormone neurons responsible for sickness behavior and hypothermia. Thus, EPS-producing biofilm pathogens evade initiating a lung-brain sensory neuronal response that results in sickness.


Assuntos
Infecções por Escherichia coli , Escherichia coli , Pulmão , Polissacarídeos Bacterianos , Infecções por Pseudomonas , Pseudomonas aeruginosa , Animais , Feminino , Masculino , Camundongos , Biofilmes , Escherichia coli/fisiologia , Hipotermia/metabolismo , Hipotermia/patologia , Inflamação/metabolismo , Inflamação/patologia , Pulmão/microbiologia , Pulmão/patologia , Pneumonia/microbiologia , Pneumonia/patologia , Pseudomonas aeruginosa/fisiologia , Células Receptoras Sensoriais , Polissacarídeos Bacterianos/metabolismo , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Infecções por Pseudomonas/metabolismo , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Nociceptores/metabolismo
2.
Physiol Rev ; 103(2): 1667-1691, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36395349

RESUMO

The corticotropin-releasing hormone cells in the paraventricular nucleus of the hypothalamus (CRHPVN) control the slow endocrine response to stress. The synapses on these cells are exquisitely sensitive to acute stress, leveraging local signals to leave a lasting imprint on this system. Additionally, recent work indicates that these cells also play key roles in the control of distinct stress and survival behaviors. Here we review these observations and provide a perspective on the role of CRHPVN neurons as integrative and malleable hubs for behavioral, physiological, and endocrine responses to stress.


Assuntos
Hormônio Liberador da Corticotropina , Núcleo Hipotalâmico Paraventricular , Humanos , Hormônio Liberador da Corticotropina/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Neurônios/fisiologia , Sinapses/metabolismo , Estresse Fisiológico
3.
Nat Immunol ; 23(8): 1137-1139, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35864244
4.
Proc Natl Acad Sci U S A ; 121(21): e2313207121, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38753512

RESUMO

Arginine vasopressin (AVP) neurons of the hypothalamic paraventricular region (AVPPVN) mediate sex-biased social behaviors across most species, including mammals. In mice, neural sex differences are thought to be established during a critical window around birth ( embryonic (E) day 18 to postnatal (P) day 2) whereby circulating testosterone from the fetal testis is converted to estrogen in sex-dimorphic brain regions. Here, we found that AVPPVN neurons are sexually dimorphic by E15.5, prior to this critical window, and that gestational bisphenol A (BPA) exposure permanently masculinized female AVPPVN neuronal numbers, projections, and electrophysiological properties, causing them to display male-like phenotypes into adulthood. Moreover, we showed that nearly twice as many neurons that became AVP+ by P0 were born at E11 in males and BPA-exposed females compared to control females, suggesting that AVPPVN neuronal masculinization occurs between E11 and P0. We further narrowed this sensitive period to around the timing of neurogenesis by demonstrating that exogenous estrogen exposure from E14.5 to E15.5 masculinized female AVPPVN neuronal numbers, whereas a pan-estrogen receptor antagonist exposed from E13.5 to E15.5 blocked masculinization of males. Finally, we showed that restricting BPA exposure to E7.5-E15.5 caused adult females to display increased social dominance over control females, consistent with an acquisition of male-like behaviors. Our study reveals an E11.5 to E15.5 window of estrogen sensitivity impacting AVPPVN sex differentiation, which is impacted by prenatal BPA exposure.


Assuntos
Compostos Benzidrílicos , Neurônios , Fenóis , Diferenciação Sexual , Animais , Compostos Benzidrílicos/toxicidade , Fenóis/toxicidade , Feminino , Masculino , Camundongos , Diferenciação Sexual/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Gravidez , Hipotálamo/metabolismo , Hipotálamo/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Arginina Vasopressina/metabolismo , Vasopressinas/metabolismo , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Camundongos Endogâmicos C57BL , Estrogênios/metabolismo , Estrogênios/farmacologia
5.
J Neurosci ; 43(45): 7657-7667, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37833068

RESUMO

Worldwide, alcohol use and abuse are a leading risk of mortality, causing 5.3% of all deaths (World Health Organization, 2022). The endocrine stress system, initiated by the peripheral release of corticotropin releasing hormone (CRH) from primarily glutamatergic neurons in the paraventricular nucleus of the hypothalamus (PVN), is profoundly linked with alcohol use, abuse, and relapse (Blaine and Sinha, 2017). These PVN CRH-releasing (PVNCRH) neurons are essential for peripheral and central stress responses (Rasiah et al., 2023), but little is known about how alcohol affects these neurons. Here, we show that two-bottle choice alcohol consumption blunts the endocrine-mediated corticosterone response to stress during acute withdrawal in female mice. Conversely, using slice electrophysiology, we demonstrate that acute withdrawal engenders a hyperexcitable phenotype of PVNCRH neurons in females that is accompanied by increased glutamatergic transmission in both male and female mice. GABAergic synaptic transmission was unaffected by alcohol history. We then tested whether chemogenetic inhibition of PVNCRH neurons would restore stress response in female mice with a history of alcohol drinking in the looming disk test, which mimics an approaching predator threat. Accordingly, inhibition of PVNCRH neurons reduced active escape in hM4Di alcohol history mice only. This study indicates that stress-responsive PVNCRH neurons in females are particularly affected by a history of alcohol consumption. Interestingly, women have indicated an increase in heavy alcohol use to cope with stress (Rodriguez et al., 2020), perhaps pointing to a potential underlying mechanism in alcohol-mediated changes to PVNCRH neurons that alter stress response.SIGNIFICANCE STATEMENT Paraventricular nucleus of the hypothalamus neurons that release corticotropin releasing hormone (PVNCRH) are vital for stress response. These neurons have been understudied in relation to alcohol and withdrawal despite profound relations between stress, alcohol use disorders (AUD), and relapse. In this study, we use a variety of techniques to show that acute withdrawal from a history of alcohol impacts peripheral stress response, PVNCRH neurons, and behavior. Specifically, PVNCRH are in a hyperactive state during withdrawal, which drives an increase in active stress coping behaviors in female mice only. Understanding how alcohol use and withdrawal affects stress responding PVNCRH neurons may contribute to finding new potential targets for the treatment of alcohol use disorder.


Assuntos
Alcoolismo , Hormônio Liberador da Corticotropina , Humanos , Feminino , Masculino , Camundongos , Animais , Hormônio Liberador da Corticotropina/metabolismo , Hormônio Adrenocorticotrópico , Hormônios Liberadores de Hormônios Hipofisários , Hipotálamo/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Neurônios/fisiologia , Consumo de Bebidas Alcoólicas , Recidiva
6.
J Physiol ; 601(15): 3151-3171, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36223200

RESUMO

Electrophysiological recordings can provide detailed information of single neurons' dynamical features and shed light on their response to stimuli. Unfortunately, rapidly modelling electrophysiological data for inferring network-level behaviours remains challenging. Here, we investigate how modelled single neuron dynamics leads to network-level responses in the paraventricular nucleus of the hypothalamus (PVN), a critical nucleus for the mammalian stress response. Recordings of corticotropin releasing hormone neurons from the PVN (CRHPVN ) were performed using whole-cell current-clamp. These, neurons, which initiate the endocrine response to stress, were rapidly and automatically fit to a modified adaptive exponential integrate-and-fire model (AdEx) with particle swarm optimization (PSO). All CRHPVN neurons were accurately fit by the AdEx model with PSO. Multiple sets of parameters were found that reliably reproduced current-clamp traces for any single neuron. Despite multiple solutions, the dynamical features of the models such as the rheobase, fixed points, and bifurcations, were shown to be stable across fits. We found that CRHPVN neurons can be divided into two subtypes according to their bifurcation at the onset of firing: CRHPVN -integrators and CRHPVN -resonators. The existence of CRHPVN -resonators was then directly confirmed in a follow-up patch-clamp hyperpolarization protocol which readily induced post-inhibitory rebound spiking in 33% of patched neurons. We constructed networks of CRHPVN model neurons to investigate the network level responses of CRHPVN neurons. We found that CRHPVN -resonators maintain baseline firing in networks even when all inputs are inhibitory. The dynamics of a small subset of CRHPVN neurons may be critical to maintaining a baseline firing tone in the PVN. KEY POINTS: Corticotropin-releasing hormone neurons (CRHPVN ) in the paraventricular nucleus of the hypothalamus act as the final neural controllers of the stress response. We developed a computational modelling platform that uses particle swarm optimization to rapidly and accurately fit biophysical neuron models to patched CRHPVN neurons. A model was fitted to each patched neuron without the use of dynamic clamping, or other procedures requiring sophisticated inputs and fitting algorithms. Any neuron undergoing standard current clamp step protocols for a few minutes can be fitted by this procedure The dynamical analysis of the modelled neurons shows that CRHPVN neurons come in two specific 'flavours': CRHPVN -resonators and CRHPVN -integrators. We directly confirmed the existence of these two classes of CRHPVN neurons in subsequent experiments. Network simulations show that CRHPVN -resonators are critical to retaining the baseline firing rate of the entire network of CRHPVN neurons as these cells can fire rebound spikes and bursts in the presence of strong inhibitory synaptic input.


Assuntos
Hormônio Liberador da Corticotropina , Núcleo Hipotalâmico Paraventricular , Hormônio Liberador da Corticotropina/metabolismo , Hipotálamo/metabolismo , Neurônios/fisiologia
7.
Alcohol Clin Exp Res ; 46(8): 1616-1629, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35797227

RESUMO

BACKGROUND: Alcohol withdrawal is a key component of severe alcohol use disorder. Animal models of alcohol withdrawal tend to focus on traditional anxiety/stress tests. While these have been essential to advancing our understanding of the biology of alcohol withdrawal, abrupt cessation of drinking following heavy alcohol consumption can also trigger withdrawal-related affective states that impact responses to a variety of life events and stressors. To this end, we show that behaviors in a variety of tasks that differ in task demand and intensity are altered during withdrawal in male and female mice after voluntary alcohol access. METHODS: Male and female miceunderwent six weeks of intermittent two-bottle choice alcohol exposure followed by behavioral tests. The tests included-Home cage: low-stress baseline environment to measure spontaneous natural behaviors; Open field: anxiety-inducing bright novel environment; Looming disc: arena with a protective hut where mice are exposed to a series of discs that mimic an overhead advancing predator, and Robogator-simulated predator task: forced foraging behavioral choice in the presence of an advancing robot predator that "attacks" when mice are near a food pellet in a large open arena. RESULTS: A history of alcohol exposure impacted behaviors in these tasks in a sex-dependent manner. In the home cage, alcohol induced reductions in digging and heightened stress coping through an increase in grooming time. In males, increased rearing yielded greater vigilance/exploration in a familiar environment. The open-field test revealed an anxiety phenotype in both male and female mice exposed to alcohol. Male mice showed no behavioral alterations to the looming disc task, while females exposed to alcohol showed greater escape responses than water controls, indicative of active stress-response behaviors. In males, the Robogator task revealed a hesitant/avoidant phenotype in alcohol-exposed mice under greater task demands. CONCLUSIONS: Few drugs show robust evidence of efficacy in clinical trials for alcohol withdrawal. Understanding how withdrawal alters a variety of behaviors in both males and females that are linked to stress coping can increase our understanding of alcohol misuse and aid in developing better medications for treating individuals with AUD.


Assuntos
Alcoolismo , Síndrome de Abstinência a Substâncias , Consumo de Bebidas Alcoólicas/psicologia , Alcoolismo/psicologia , Animais , Ansiedade , Etanol/farmacologia , Feminino , Masculino , Camundongos , Síndrome de Abstinência a Substâncias/psicologia
8.
J Neurosci ; 40(46): 8842-8852, 2020 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-33051356

RESUMO

In many species, social networks provide benefit for both the individual and the collective. In addition to transmitting information to others, social networks provide an emotional buffer for distressed individuals. Our understanding about the cellular mechanisms that contribute to buffering is poor. Stress has consequences for the entire organism, including a robust change in synaptic plasticity at glutamate synapses onto corticotropin-releasing hormone (CRH) neurons in the paraventricular nucleus of the hypothalamus (PVN). In females, however, this stress-induced metaplasticity is buffered by the presence of a naive partner. This buffering may be because of discrete behavioral interactions, signals in the context in which the interaction occurs (i.e., olfactory cues), or it may be influenced by local signaling events in the PVN. Here, we show that local vasopressin (VP) signaling in PVN buffers the short-term potentiation (STP) at glutamate synapses after stress. This social buffering of metaplasticity, which requires the presence of another individual, was prevented by pharmacological inhibition of the VP 1a receptor (V1aR) in female mice. Exogenous VP mimicked the effects of social buffering and reduced STP in CRHPVN neurons from females but not males. These findings implicate VP as a potential mediator of social buffering in female mice.SIGNIFICANCE STATEMENT In many organisms, including rodents and humans, social groups are beneficial to overall health and well-being. Moreover, it is through these social interactions that the harmful effects of stress can be mitigated, a phenomenon known as social buffering. In the present study, we describe a critical role for the neuropeptide vasopressin (VP) in social buffering of synaptic metaplasticity in stress-responsive corticotropin-releasing hormone (CRH) neurons in female mice. These effects of VP do not extend to social buffering of stress behaviors, suggesting this is a very precise and local form of sex-specific neuropeptide signaling.


Assuntos
Estresse Psicológico/fisiopatologia , Sinapses , Vasopressinas , Animais , Hormônio Liberador da Corticotropina , Sinais (Psicologia) , Feminino , Asseio Animal , Técnicas In Vitro , Masculino , Camundongos , Plasticidade Neuronal , Neurônios , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Receptores de N-Metil-D-Aspartato , Receptores de Vasopressinas/efeitos dos fármacos , Caracteres Sexuais , Olfato , Meio Social , Estresse Psicológico/psicologia
9.
Nat Rev Neurosci ; 16(7): 377-88, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26087679

RESUMO

Stress necessitates an immediate engagement of multiple neural and endocrine systems. However, exposure to a single stressor causes adaptive changes that modify responses to subsequent stressors. Recent studies examining synapses onto neuroendocrine cells in the paraventricular nucleus of the hypothalamus demonstrate that stressful experiences leave indelible marks that alter the ability of these synapses to undergo plasticity. These adaptations include a unique form of metaplasticity at glutamatergic synapses, bidirectional changes in endocannabinoid signalling and bidirectional changes in strength at GABAergic synapses that rely on distinct temporal windows following stress. This rich repertoire of plasticity is likely to represent an important building block for dynamic, experience-dependent modulation of neuroendocrine stress adaptation.


Assuntos
Hipotálamo/metabolismo , Plasticidade Neuronal/fisiologia , Estresse Psicológico/metabolismo , Sinapses/metabolismo , Animais , Humanos , Rede Nervosa/metabolismo , Estresse Psicológico/psicologia
10.
J Neurosci ; 38(49): 10444-10453, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-30355625

RESUMO

GABA synapses in the brain undergo depolarization-induced suppression of inhibition (DSI) that requires activation of presynaptic cannabinoid type 1 receptors (CB1Rs). The brevity of DSI, lasting ∼1 min in most brain regions, has been ascribed to the transient production of 2-arachidonoylglycerol (2-AG). Here, we propose that the duration of DSI is controlled by heterologous interactions between presynaptic mGluRs and CB1Rs. By examining GABA synapses on parvocellular corticotropin-releasing hormone-expressing neurons in the paraventricular nucleus of the hypothalamus (PVN) of male and female mice, we show that DSI decays quickly in experimental conditions in which both GABA and glutamate are released from adjacent nerve terminals. Pharmacological inhibition of group I mGluRs prolongs DSI, whereas prior activation of mGluRs inhibits DSI, collectively suggesting that group I mGluRs quench presynaptic CB1R signaling. When photostimulation of genetically identified terminals is used to release only GABA, CB1R-dependent DSI persists for many minutes. Under the same conditions, activation of group I mGluRs reestablishes classical, transient DSI. The long-lasting DSI observed when GABA synapses are independently recruited functionally uncouples inhibitory input to PVN neurons. These observations suggest that heterologous interactions between mGluRs and CB1Rs control the temporal window of DSI at GABA synapses, providing evidence for a powerful new way to affect functional circuit connectivity in the brain.SIGNIFICANCE STATEMENT Postsynaptic depolarization liberates endocannabinoids, resulting in a rapid and transient decrease in release probability at GABA synapses. We discovered that mGluRs control the duration of depolarization-induced suppression of inhibition (DSI), most likely through heterologous desensitization of cannabinoid type 1 receptors by presynaptic mGluR5 By shortening the duration of DSI, mGluRs control the temporal window for retrograde signaling at GABA synapses. Physiological or pathological changes that affect glutamate spillover may profoundly affect network excitability by shifting the duration of cannabinoid inhibition at GABA synapses.


Assuntos
Endocanabinoides/fisiologia , Potenciais Pós-Sinápticos Inibidores/fisiologia , Receptor CB1 de Canabinoide/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Receptores Pré-Sinápticos/metabolismo , Animais , Endocanabinoides/farmacologia , Feminino , Ácido Glutâmico/metabolismo , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Masculino , Metoxi-Hidroxifenilglicol/análogos & derivados , Metoxi-Hidroxifenilglicol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Optogenética/métodos , Receptor CB1 de Canabinoide/química , Receptores de Glutamato Metabotrópico/química , Receptores Pré-Sinápticos/química , Ácido gama-Aminobutírico/metabolismo
11.
J Neurosci ; 38(40): 8515-8525, 2018 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-30108130

RESUMO

Whether synapses in appetite-regulatory brain regions undergo long-term changes in strength in response to satiety peptides is poorly understood. Here we show that following bursts of afferent activity, the neuromodulator and satiety peptide cholecystokinin (CCK) shifts the plasticity of GABA synapses in the dorsomedial nucleus of the hypothalamus of male Sprague Dawley rats from long-term depression to long-term potentiation (LTP). This LTP requires the activation of both type 2 CCK receptors and group 5 metabotropic glutamate receptors, resulting in a rise in astrocytic intracellular calcium and subsequent ATP release. ATP then acts on presynaptic P2X receptors to trigger a prolonged increase in GABA release. Our observations demonstrate a novel form of CCK-mediated plasticity that requires astrocytic ATP release, and could serve as a mechanism for appetite regulation.SIGNIFICANCE STATEMENT Satiety peptides, like cholecystokinin, play an important role in the central regulation of appetite, but their effect on synaptic plasticity is not well understood. The current data provide novel evidence that cholecystokinin shifts the plasticity from long-term depression to long-term potentiation at GABA synapses in the rat dorsomedial nucleus of the hypothalamus. We also demonstrate that this plasticity requires the concerted action of cholecystokinin and glutamate on astrocytes, triggering the release of the gliotransmitter ATP, which subsequently increases GABA release from neighboring inhibitory terminals. This research reveals a novel neuropeptide-induced switch in the direction of synaptic plasticity that requires astrocytes, and could represent a new mechanism by which cholecystokinin regulates appetite.


Assuntos
Trifosfato de Adenosina/metabolismo , Astrócitos/fisiologia , Colecistocinina/fisiologia , Núcleo Hipotalâmico Dorsomedial/fisiologia , Potenciação de Longa Duração , Depressão Sináptica de Longo Prazo , Ácido gama-Aminobutírico/fisiologia , Animais , Masculino , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5/fisiologia , Receptores da Colecistocinina/fisiologia , Receptores Purinérgicos P2X/fisiologia , Transmissão Sináptica
12.
Glia ; 67(10): 1806-1820, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30889320

RESUMO

An organism's response to stress requires activation of multiple brain regions. This can have long-lasting effects on synaptic transmission and plasticity that likely provide adaptive benefits. Recent evidence implicates not only neurones, but also glial cells in the regulation of the central response to stress. Intense, repeated or uncontrolled stress has been implicated in the emergence of multiple neuropsychiatric conditions. Human studies have consistently observed glial dysfunction in mood and stress disorders such as major depression. Interestingly animal models of stress have recapitulated glial abnormalities that are comparable to the human condition, validating the use of rodent models for the study of stress disorders. In this review we will focus upon one family of glia, the astrocytes, and describe the evidence to date that links astrocytes to possible stress-related disorders.


Assuntos
Astrócitos/patologia , Astrócitos/fisiologia , Estresse Psicológico/patologia , Estresse Psicológico/fisiopatologia , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , Humanos
13.
J Physiol ; 596(10): 1919-1929, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29419884

RESUMO

KEY POINTS: GABA transporter (GAT) blockade recruits extrasynaptic GABAA receptors (GABAA Rs) and amplifies constitutive presynaptic GABAB R activity. Extrasynaptic GABAA Rs contribute to a tonic current. Corticosteroids increase the tonic current mediated by extrasynaptic GABAA Rs. ABSTRACT: Corticotropin-releasing hormone (CRH) neurons in the paraventricular nucleus of the hypothalamus (PVN) are integratory hubs that regulate the endocrine response to stress. GABA inputs provide a basal inhibitory tone that constrains this system and circulating glucocorticoids (CORT) are important feedback controllers of CRH output. Surprisingly little is known about the direct effects of CORT on GABA synapses in PVN. Here we used whole-cell patch clamp recordings from CRH neurons in mouse hypothalamic brain slices to examine the effects of CORT on synaptic and extrasynaptic GABA signalling. We show that GABA transporters (GATs) limit constitutive activation of presynaptic GABAB receptors and ensure high release probability at GABA synapses. GATs in combination with GABAB receptors also curtail extrasynaptic GABAA R signalling. CORT has no effect on synaptic GABA signalling, but increases extrasynaptic GABA tone through upregulation of postsynaptic GABAA receptors. These data show that efficient GABA clearance and autoinhibition control the balance between synaptic (phasic) and extrasynaptic (tonic) inhibition in PVN CRH neurons. This balance is shifted towards increased extrasynaptic inhibition by CORT.


Assuntos
Corticosterona/farmacologia , Hormônio Liberador da Corticotropina/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/fisiologia , Potenciais Pós-Sinápticos Inibidores , Inibição Neural , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Hormônio Adrenocorticotrópico/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Neurônios/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Receptores de GABA-A/metabolismo , Receptores de GABA-B/metabolismo , Sinapses/fisiologia
14.
J Neurosci ; 35(38): 13160-70, 2015 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-26400945

RESUMO

Somatodendritically released peptides alter synaptic function through a variety of mechanisms, including autocrine actions that liberate retrograde transmitters. Cholecystokinin (CCK) is a neuropeptide expressed in neurons in the dorsomedial hypothalamic nucleus (DMH), a region implicated in satiety and stress. There are clear demonstrations that exogenous CCK modulates food intake and neuropeptide expression in the DMH, but there is no information on how endogenous CCK alters synaptic properties. Here, we provide the first report of somatodendritic release of CCK in the brain in male Sprague Dawley rats. CCK is released from DMH neurons in response to repeated postsynaptic depolarizations, and acts in an autocrine fashion on CCK2 receptors to enhance postsynaptic NMDA receptor function and liberate the retrograde transmitter, nitric oxide (NO). NO subsequently acts presynaptically to enhance GABA release through a soluble guanylate cyclase-mediated pathway. These data provide the first demonstration of synaptic actions of somatodendritically released CCK in the hypothalamus and reveal a new form of retrograde plasticity, depolarization-induced potentiation of inhibition. Significance statement: Somatodendritic signaling using endocannabinoids or nitric oxide to alter the efficacy of afferent transmission is well established. Despite early convincing evidence for somatodendritic release of neurohypophysial peptides in the hypothalamus, there is only limited evidence for this mode of release for other peptides. Here, we provide the first evidence for somatodendritic release of the satiety peptide cholecystokinin (CCK) in the brain. We also reveal a new form of synaptic plasticity in which postsynaptic depolarization results in enhancement of inhibition through the somatodendritic release of CCK.


Assuntos
Colecistocinina/metabolismo , Núcleo Hipotalâmico Dorsomedial/citologia , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo , Animais , Animais Recém-Nascidos , Colecistocinina/farmacologia , GABAérgicos/farmacologia , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacologia , Técnicas In Vitro , Masculino , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Proglumida/análogos & derivados , Proglumida/farmacologia , Quinazolinonas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor de Colecistocinina B/antagonistas & inibidores , Receptor de Colecistocinina B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína 25 Associada a Sinaptossoma/antagonistas & inibidores , Proteína 25 Associada a Sinaptossoma/metabolismo , Tionucleotídeos/farmacologia , Ácido gama-Aminobutírico/farmacologia
15.
J Neurosci ; 35(13): 5144-55, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25834041

RESUMO

The Na-K-2Cl cotransporter 2 (NKCC2) was thought to be kidney specific. Here we show expression in the brain hypothalamo-neurohypophyseal system (HNS), wherein upregulation follows osmotic stress. The HNS controls osmotic stability through the synthesis and release of the neuropeptide hormone, arginine vasopressin (AVP). AVP travels through the bloodstream to the kidney, where it promotes water conservation. Knockdown of HNS NKCC2 elicited profound effects on fluid balance following ingestion of a high-salt solution-rats produced significantly more urine, concomitant with increases in fluid intake and plasma osmolality. Since NKCC2 is the molecular target of the loop diuretics bumetanide and furosemide, we asked about their effects on HNS function following disturbed water balance. Dehydration-evoked GABA-mediated excitation of AVP neurons was reversed by bumetanide, and furosemide blocked AVP release, both in vivo and in hypothalamic explants. Thus, NKCC2-dependent brain mechanisms that regulate osmotic stability are disrupted by loop diuretics in rats.


Assuntos
Sistema Hipotálamo-Hipofisário/metabolismo , Osmorregulação/fisiologia , Neuro-Hipófise/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Animais , Arginina Vasopressina/sangue , Arginina Vasopressina/efeitos dos fármacos , Bumetanida/farmacologia , Desidratação/fisiopatologia , Furosemida/farmacologia , Expressão Gênica/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/citologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Masculino , Núcleos da Linha Média do Tálamo/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Quiasma Óptico/fisiologia , Neuro-Hipófise/citologia , Neuro-Hipófise/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Membro 1 da Família 12 de Carreador de Soluto/biossíntese , Equilíbrio Hidroeletrolítico/efeitos dos fármacos , Equilíbrio Hidroeletrolítico/fisiologia
16.
J Physiol ; 594(4): 1005-15, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26662615

RESUMO

KEY POINTS: Many excitatory synapses in the brain release glutamate with both synchronous and asynchronous components. Immediately following an action potential, neurons display a reduced excitability due to the post-spike afterhyperpolarization (AHP). This gives rise to a relative refractory period. When an action potential is evoked by glutamate synaptic input possessing asynchronous release, the delayed glutamate release events act to depolarize the neuron during the AHP and overcome the relative refractory period. These results demonstrate a new role for asynchronous release in regulating post-spike excitability and the relative refractory period in central neurons. ABSTRACT: Post-spike afterhyperpolarizations (AHPs) functionally inhibit neuronal excitability for tens to hundreds of milliseconds following each action potential. This imposes a relative refractory period during which synaptic excitation is less effective at evoking spikes. Here we asked whether some synapses have mechanisms in place that allow them to overcome the AHP and drive spiking in target cells during this period of reduced excitability. We examined glutamate synapses onto oxytocin and vasopressin neurons in the paraventricular nucleus of the hypothalamus. These synapses can display pronounced asynchronous glutamate release following a single presynaptic spike, with the time course of release being similar to that of the post-spike AHP. To test whether asynchronous release is more effective at overcoming the relative refractory period, we evoked a single action potential with either a brief synchronous depolarization or an asynchronous potential and then assessed excitability at multiple time points following the spike. Neurons receiving asynchronous depolarizing synaptic inputs had a shorter relative refractory period than those receiving synchronous depolarizations. Our data demonstrate that synapses releasing glutamate in an asynchronous and delayed manner are ideally adapted to counter the AHP. By effectively overcoming the relative refractory period, the kinetics of excitatory synaptic input can play an important role in controlling post-spike excitability.


Assuntos
Potenciais Pós-Sinápticos Excitadores , Ácido Glutâmico/metabolismo , Terminações Pré-Sinápticas/metabolismo , Período Refratário Eletrofisiológico , Animais , Exocitose , Masculino , Neurônios/metabolismo , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiologia , Terminações Pré-Sinápticas/fisiologia , Ratos , Ratos Sprague-Dawley
17.
Bioessays ; 36(6): 561-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24648386

RESUMO

We recently described a novel form of stress-associated bidirectional plasticity at GABA synapses onto hypothalamic parvocellular neuroendocrine cells (PNCs), the apex of the hypothalamus-pituitary-adrenal axis. This plasticity may contribute to neuroendocrine adaptation. However, this GABA synapse plasticity likely does not translate into a simple more and less of inhibition because the ionic driving force for Cl(-) , the primary charge carrier for GABAA receptors, is dynamic. Specifically, stress impairs a Cl(-) extrusion mechanism in PNCs. This not only renders the steady-state GABA response less hyperpolarizing but also makes PNCs susceptible to the activity-dependent accumulation of Cl(-) . Accordingly, GABA synapse plasticity impacts both the robustness of GABA voltage response and dynamic Cl(-) loading, imposing nonlinear influences on PNC excitability during circuit activities. This theoretical consideration predicts roles for GABA transmission far more versatile than canonical inhibition. We propose potential impacts of GABA synapse plasticity on the experience-dependent fine-tuning of neuroendocrine stress responses.


Assuntos
Sistemas Neurossecretores/fisiologia , Estresse Fisiológico , Sinapses/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Cloretos/metabolismo , Humanos , Plasticidade Neuronal
18.
J Neurosci ; 34(18): 6177-81, 2014 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-24790188

RESUMO

Alterations in synaptic endocannabinoid signaling are a widespread neurobiological consequence of many in vivo experiences, including stress. Here, we report that stressor salience is critical for bidirectionally modifying presynaptic CB-1 receptor (CB1R) function at hypothalamic GABA synapses controlling the neuroendocrine stress axis in male rats. While repetitive, predictable stressor exposure impairs presynaptic CB1R function, these changes are rapidly reversed upon exposure to a high salience experience such as novel stress or by manipulations that enhance neural activity levels in vivo or in vitro. Together these data demonstrate that experience salience, through alterations in afferent synaptic activity, induces rapid changes in endocannabinoid signaling.


Assuntos
Endocanabinoides/metabolismo , Hipotálamo/patologia , Transdução de Sinais/fisiologia , Estresse Psicológico/patologia , Sinapses/fisiologia , Analgésicos/farmacologia , Animais , Animais Recém-Nascidos , Benzoxazinas/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Eletrochoque/efeitos adversos , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Masculino , Morfolinas/farmacologia , Naftalenos/farmacologia , Odorantes , Piperidinas/farmacologia , Cloreto de Potássio/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Estresse Psicológico/etiologia , Natação/psicologia , Sinapses/efeitos dos fármacos
19.
J Exp Biol ; 217(Pt 1): 102-8, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24353209

RESUMO

Any unanticipated threat to survival triggers an immediate sequence of events in the brain that culminate in a coordinated neural, endocrine and behavioural response. There is increasing evidence that stress itself modifies neural circuits. In other words, neural stress circuits learn from stress. This self-teaching is surprising as one might expect these essential circuits to be hard-wired. Our recent findings, however, indicate that repeated homotypic stress in rats causes functional changes in neural circuitry in the hypothalamus. In particular, we focus on signalling via endocannabinoids and describe plasticity in this system that impacts fast retrograde signalling at synapses on to the stress command neurons in the brain. Interestingly, this plasticity appears to be limited to early adolescence, hinting at unique modes of control of neural circuits by stress during different developmental stages.


Assuntos
Endocanabinoides/metabolismo , Neurônios GABAérgicos/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Plasticidade Neuronal/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Animais , Moduladores de Receptores de Canabinoides , Hormônio Liberador da Corticotropina , Neurotransmissores , Ratos , Receptores de Canabinoides/fisiologia , Transdução de Sinais , Estresse Fisiológico , Estresse Psicológico , Ácido gama-Aminobutírico
20.
Neurophotonics ; 11(Suppl 1): S11511, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38799809

RESUMO

Significance: Motion artifacts in the signals recorded during optical fiber-based measurements can lead to misinterpretation of data. In this work, we address this problem during in-vivo rodent experiments and develop a motion artifacts correction (MAC) algorithm for single-fiber system (SFS) hemodynamics measurements from the brains of rodents. Aim: (i) To distinguish the effect of motion artifacts in the SFS signals. (ii) Develop a MAC algorithm by combining information from the experiments and simulations and validate it. Approach: Monte-Carlo (MC) simulations were performed across 450 to 790 nm to identify wavelengths where the reflectance is least sensitive to blood absorption-based changes. This wavelength region is then used to develop a quantitative metric to measure motion artifacts, termed the dissimilarity metric (DM). We used MC simulations to mimic artifacts seen during experiments. Further, we developed a mathematical model describing light intensity at various optical interfaces. Finally, an MAC algorithm was formulated and validated using simulation and experimental data. Results: We found that the 670 to 680 nm wavelength region is relatively less sensitive to blood absorption. The standard deviation of DM (σDM) can measure the relative magnitude of motion artifacts in the SFS signals. The artifacts cause rapid shifts in the reflectance data that can be modeled as transmission changes in the optical lightpath. The changes observed during the experiment were found to be in agreement to those obtained from MC simulations. The mathematical model developed to model transmission changes to represent motion artifacts was extended to an MAC algorithm. The MAC algorithm was validated using simulations and experimental data. Conclusions: We distinguished motion artifacts from SFS signals during in vivo hemodynamic monitoring experiments. From simulation and experimental data, we showed that motion artifacts can be modeled as transmission changes. The developed MAC algorithm was shown to minimize artifactual variations in both simulation and experimental data.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA