Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 193
Filtrar
1.
Eur J Neurosci ; 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39235324

RESUMO

The paraventricular nucleus of the hypothalamus (PVN) regulates physiological and behavioural responses evoked by stressful stimuli, but the local neurochemical and signalling mechanisms involved are not completely understood. The soluble guanylate cyclase (sGC) within the PVN is implicated in autonomic and cardiovascular control in rodents under resting conditions. However, the involvement of PVN sGC-mediated signalling in stress responses is unknown. Therefore, we investigated the role of sGC within the PVN in cardiovascular, autonomic, neuroendocrine, and local neuronal responses to acute restraint stress in rats. Bilateral microinjection of the selective sGC inhibitor ODQ (1 nmol/100 nl) into the PVN reduced both the increased arterial pressure and the drop in cutaneous tail temperature evoked by restraint stress, while the tachycardia was enhanced. Intra-PVN injection of ODQ did not alter the number of Fos-immunoreactive neurons in either the dorsal cap parvocellular (PaDC), ventromedial (PaV), medial parvocellular (PaMP), or lateral magnocelllular (PaLM) portions of the PVN following acute restraint stress. Local microinjection of ODQ into the PVN did not affect the restraint-induced increases in plasma corticosterone concentration. Taken together, these findings suggest that sGC-mediated signalling in the PVN plays a key role in acute stress-induced pressor responses and sympathetically mediated cutaneous vasoconstriction, whereas the tachycardiac response is inhibited. Absence of an effect of ODQ on corticosterone and PVN neuronal activation in and the PaV and PaMP suggests that PVN sGC is not involved in restraint-evoked hypothalamus-pituitary-adrenal (HPA) axis activation and further indicates that autonomic and neuroendocrine responses are dissociable at the level of the PVN.

2.
Mol Psychiatry ; 28(11): 4729-4741, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37644175

RESUMO

Psychological loss is a common experience that erodes well-being and negatively impacts quality of life. The molecular underpinnings of loss are poorly understood. Here, we investigate the mechanisms of loss using an environmental enrichment removal (ER) paradigm in male rats. The basolateral amygdala (BLA) was identified as a region of interest, demonstrating differential Fos responsivity to ER and having an established role in stress processing and adaptation. A comprehensive multi-omics investigation of the BLA, spanning multiple cohorts, platforms, and analyses, revealed alterations in microglia and the extracellular matrix (ECM). Follow-up studies indicated that ER decreased microglia size, complexity, and phagocytosis, suggesting reduced immune surveillance. Loss also substantially increased ECM coverage, specifically targeting perineuronal nets surrounding parvalbumin interneurons, suggesting decreased plasticity and increased inhibition within the BLA following loss. Behavioral analyses suggest that these molecular effects are linked to impaired BLA salience evaluation, leading to a mismatch between stimulus and reaction intensity. These loss-like behaviors could be rescued by depleting BLA ECM during the removal period, helping us understand the mechanisms underlying loss and revealing novel molecular targets to ameliorate its impact.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Ratos , Animais , Masculino , Complexo Nuclear Basolateral da Amígdala/fisiologia , Neurobiologia , Qualidade de Vida , Interneurônios , Matriz Extracelular
3.
Stress ; 27(1): 2361238, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38962839

RESUMO

Chronic stress leads to hypofunction of the medial prefrontal cortex (mPFC), mechanisms of which remain to be determined. Enhanced activation of GABAergic of parvalbumin (PV) expressing interneurons (INs) is thought to play a role in stress-induced prefrontal inhibition. In this study, we tested whether chemogenetic inhibition of mPFC PV INs after chronic stress can rescue chronic stress-related behavioral and physiological phenotypes. Mice underwent 2 weeks of chronic variable stress (CVS) followed by a battery of behavioral tests known to be affected by chronic stress exposure, e.g. an open field (OF), novel object recognition (NOR), tail suspension test (TST), sucrose preference test (SPT), and light dark (LD) box. Inhibitory DREADDs were actuated by 3 mg/kg CNO administered 30 min prior to each behavioral test. CVS caused hyperactivity in the OF, reduced sucrose preference in the SPT (indicative of enhanced anhedonia), and increased anxiety-like behavior in the LD box. Inhibition of PV IN after stress mitigated these effects. In addition, CVS also resulted in reduced thymus weight and body weight loss, which were also mitigated by PV IN inhibition. Our results indicate that chronic stress leads to plastic changes in PV INs that may be mitigated by chemogenetic inhibition. Our findings implicate cortical GABAergic INs as a therapeutic target in stress-related diseases.


Assuntos
Comportamento Animal , Interneurônios , Parvalbuminas , Córtex Pré-Frontal , Estresse Psicológico , Animais , Córtex Pré-Frontal/metabolismo , Parvalbuminas/metabolismo , Masculino , Interneurônios/metabolismo , Camundongos , Estresse Psicológico/fisiopatologia , Ansiedade , Camundongos Endogâmicos C57BL
4.
Neurobiol Dis ; 178: 106014, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36702319

RESUMO

Status epilepticus (SE) is a life-threatening medical emergency with significant morbidity and mortality. SE is associated with a robust and sustained increase in serum glucocorticoids, reaching concentrations sufficient to activate the dense population of glucocorticoid receptors (GRs) expressed among hippocampal excitatory neurons. Glucocorticoid exposure can increase hippocampal neuron excitability; however, whether activation of hippocampal GRs during SE exacerbates seizure severity remains unknown. To test this, a viral strategy was used to delete GRs from a subset of hippocampal excitatory neurons in adult male and female mice, producing hippocampal GR knockdown mice. Two weeks after GR knockdown, mice were challenged with the convulsant drug pilocarpine to induce SE. GR knockdown had opposing effects on early vs late seizure behaviors, with sex influencing responses. For both male and female mice, the onset of mild behavioral seizures was accelerated by GR knockdown. In contrast, GR knockdown delayed the onset of more severe convulsive seizures and death in male mice. Concordantly, GR knockdown also blunted the SE-induced rise in serum corticosterone in male mice. GR knockdown did not alter survival times or serum corticosterone in females. To assess whether loss of GR affected susceptibility to SE-induced cell death, within-animal analyses were conducted comparing local GR knockdown rates to local cell loss. GR knockdown did not affect the degree of localized neuronal loss, suggesting cell-intrinsic GR signaling neither protects nor sensitizes neurons to acute SE-induced death. Overall, the findings reveal that hippocampal GRs exert an anti-convulsant role in both males and females in the early stages of SE, followed by a switch to a pro-convulsive role for males only. Findings reveal an unexpected complexity in the interaction between hippocampal GR activation and the progression of SE.


Assuntos
Receptores de Glucocorticoides , Estado Epiléptico , Camundongos , Masculino , Feminino , Animais , Receptores de Glucocorticoides/metabolismo , Corticosterona , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/metabolismo , Hipocampo/metabolismo , Convulsões/induzido quimicamente , Convulsões/metabolismo , Glucocorticoides/metabolismo , Pilocarpina/toxicidade , Convulsivantes
5.
Mol Psychiatry ; 27(10): 4023-4034, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35754044

RESUMO

In psychiatric disorders, mismatches between disease states and therapeutic strategies are highly pronounced, largely because of unanswered questions regarding specific vulnerabilities of different cell types and therapeutic responses. Which cellular events (housekeeping or salient) are most affected? Which cell types succumb first to challenges, and which exhibit the strongest response to drugs? Are these events coordinated between cell types? How does disease and drug effect this coordination? To address these questions, we analyzed single-nucleus-RNAseq (sn-RNAseq) data from the human anterior cingulate cortex-a region involved in many psychiatric disorders. Density index, a metric for quantifying similarities and dissimilarities across functional profiles, was employed to identify common or salient functional themes across cell types. Cell-specific signatures were integrated with existing disease and drug-specific signatures to determine cell-type-specific vulnerabilities, druggabilities, and responsiveness. Clustering of functional profiles revealed cell types jointly participating in these events. SST and VIP interneurons were found to be most vulnerable, whereas pyramidal neurons were least. Overall, the disease state is superficial layer-centric, influences cell-specific salient themes, strongly impacts disinhibitory neurons, and influences astrocyte interaction with a subset of deep-layer pyramidal neurons. In absence of disease, drugs profiles largely recapitulate disease profiles, offering a possible explanation for drug side effects. However, in presence of disease, drug activities, are deep layer-centric and involve activating a distinct subset of deep-layer pyramidal neurons to circumvent the disease state's disinhibitory circuit malfunction. These findings demonstrate a novel application of sn-RNAseq data to explain drug and disease action at a systems level.


Assuntos
Giro do Cíngulo , Interneurônios , Humanos , Interneurônios/metabolismo , Neurônios/metabolismo , Células Piramidais/fisiologia
6.
Mol Psychiatry ; 26(9): 4853-4863, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33504954

RESUMO

The common molecular mechanisms underlying psychiatric disorders are not well understood. Prior attempts to assess the pathological mechanisms responsible for psychiatric disorders have been limited by biased selection of comparable disorders, datasets/cohort availability, and challenges with data normalization. Here, using DisGeNET, a gene-disease associations database, we sought to expand such investigations in terms of number and types of diseases. In a top-down manner, we analyzed an unbiased cluster of 36 psychiatric disorders and comorbid conditions at biological pathway, cell-type, drug-target, and chromosome levels and deployed density index, a novel metric to quantify similarities (close to 1) and dissimilarities (close to 0) between these disorders at each level. At pathway level, we show that cognition and neurotransmission drive the similarity and are involved across all disorders, whereas immune-system and signal-response coupling (cell surface receptors, signal transduction, gene expression, and metabolic process) drives the dissimilarity and are involved with specific disorders. The analysis at the drug-target level supports the involvement of neurotransmission-related changes across these disorders. At cell-type level, dendrite-targeting interneurons, across all layers, are most involved. Finally, by matching the clustering pattern at each level of analysis, we showed that the similarity between the disorders is influenced most at the chromosomal level and to some extent at the cellular level. Together, these findings provide first insights into distinct cellular and molecular pathologies, druggable mechanisms associated with several psychiatric disorders and comorbid conditions and demonstrate that similarities between these disorders originate at the chromosome level and disperse in a bottom-up manner at cellular and pathway levels.


Assuntos
Transtornos Mentais , Análise por Conglomerados , Cognição , Estudos de Coortes , Expressão Gênica , Humanos , Transtornos Mentais/genética
7.
Stress ; 24(2): 196-205, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33726625

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) is an excitatory neuromodulatory peptide strongly implicated in nervous stress processing. Human polymorphism of the primary PACAP receptor (PAC1) is linked to psychiatric disorders, including posttraumatic stress disorder (PTSD). Prefrontal cortex PACAP signaling is associated with processing of traumatic stress and fear learning, suggesting a potential role in PTSD-related deficits. We used RNAscope to define the cellular location of PACAP and PAC1 in the infralimbic cortex (IL). Subsequent experiments used a pharmacological approach to assess the impact of IL PACAP infusion on behavioral and physiological stress response and fear memory. Adult male Sprague-Dawley rats were bilaterally microinjected with PACAP (1 ug) or vehicle into the IL, 30 minutes prior to forced swim test (FST). Blood was sampled at 15, 30, 60, and 120 minutes for analysis of hypothalamic pituitary adrenal (HPA) axis reactivity. Five days after, animals were tested in a 3-day passive avoidance paradigm with subsequent testing of fear retention two weeks later. We observed that PACAP is highly expressed in putative pyramidal neurons (identified by VGlut1 expression), while PAC1 is enriched in interneurons (identified by GAD). Pretreatment with PACAP increased active coping style in the FST, despite higher levels of ACTH and corticosterone. The treatment was also sufficient to cause an increase in anxiety-like behavior in a dark/light crossover test and enhanced retention of passive avoidance. Our data suggest that IL PACAP plays a role in driving stress responses and in processing of fear memories, likely mediated by inhibition of cortical drive.


Assuntos
Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Estresse Psicológico , Animais , Masculino , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley
8.
Stress ; 23(6): 617-632, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33345670

RESUMO

Regulation of stress reactivity is a fundamental priority of all organisms. Stress responses are critical for survival, yet can also cause physical and psychological damage. This review provides a synopsis of brain mechanisms designed to control physiological responses to stress, focusing primarily on glucocorticoid secretion via the hypothalamo-pituitary-adrenocortical (HPA) axis. The literature provides strong support for multi-faceted control of HPA axis responses, involving both direct and indirect actions at paraventricular nucleus (PVN) corticotropin releasing hormone neurons driving the secretory cascade. The PVN is directly excited by afferents from brainstem and hypothalamic circuits, likely relaying information on homeostatic challenge. Amygdala subnuclei drive HPA axis responses indirectly via disinhibition, mediated by GABAergic relays onto PVN-projecting neurons in the hypothalamus and bed nucleus of the stria terminalis (BST). Inhibition of stressor-evoked HPA axis responses is mediated by an elaborate network of glucocorticoid receptor (GR)-containing circuits, providing a distributed negative feedback signal that inhibits PVN neurons. Prefrontal and hippocampal neurons play a major role in HPA axis inhibition, again mediated by hypothalamic and BST GABAergic relays to the PVN. The complexity of the regulatory process suggests that information on stressors is integrated across functional disparate brain circuits prior to accessing the PVN, with regions such as the BST in prime position to relay contextual information provided by these sources into appropriate HPA activation. Dysregulation of the HPA in disease is likely a product of inappropriate checks and balances between excitatory and inhibitory inputs ultimately impacting PVN output.


Assuntos
Sistema Hipotálamo-Hipofisário , Sistema Hipófise-Suprarrenal , Encéfalo , Hormônio Liberador da Corticotropina , Retroalimentação , Núcleo Hipotalâmico Paraventricular , Estresse Fisiológico , Estresse Psicológico
9.
Front Neuroendocrinol ; 48: 50-57, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29042142

RESUMO

Glucocorticoids act via multiple mechanisms to mobilize energy for maintenance and restoration of homeostasis. In adipose tissue, glucocorticoids can promote lipolysis and facilitate adipocyte differentiation/growth, serving both energy-mobilizing and restorative processes during negative energy balance. Recent data suggest that adipose-dependent feedback may also be involved in regulation of stress responses. Adipocyte glucocorticoid receptor (GR) deletion causes increased HPA axis stress reactivity, due to a loss of negative feedback signals into the CNS. The fat-to-brain signal may be mediated by neuronal mechanisms, release of adipokines or increased lipolysis. The ability of adipose GRs to inhibit psychogenic as well as metabolic stress responses suggests that (1) feedback regulation of the HPA axis occurs across multiple bodily compartments, and (2) fat tissue integrates psychogenic stress signals. These studies support a link between stress biology and energy metabolism, a connection that has clear relevance for numerous disease states and their comorbidities.


Assuntos
Adipócitos/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Receptores de Glucocorticoides/metabolismo , Estresse Fisiológico/fisiologia , Estresse Psicológico/metabolismo , Animais , Humanos
10.
J Neurosci ; 37(1): 184-193, 2017 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-28053040

RESUMO

Organismal stress initiates a tightly orchestrated set of responses involving complex physiological and neurocognitive systems. Here, we present evidence for glucagon-like peptide 1 (GLP-1)-mediated paraventricular hypothalamic circuit coordinating the global stress response. The GLP-1 receptor (Glp1r) in mice was knocked down in neurons expressing single-minded 1, a transcription factor abundantly expressed in the paraventricular nucleus (PVN) of the hypothalamus. Mice with single-minded 1-mediated Glp1r knockdown had reduced hypothalamic-pituitary-adrenal axis responses to both acute and chronic stress and were protected against weight loss associated with chronic stress. In addition, regional Glp1r knockdown attenuated stress-induced cardiovascular responses accompanied by decreased sympathetic drive to the heart. Finally, Glp1r knockdown reduced anxiety-like behavior, implicating PVN GLP-1 signaling in behavioral stress reactivity. Collectively, these findings support a circuit whereby brainstem GLP-1 activates PVN signaling to mount an appropriate whole-organism response to stress. These results raise the possibility that dysfunction of this system may contribute to stress-related pathologies, and thereby provide a novel target for intervention. SIGNIFICANCE STATEMENT: Dysfunctional stress responses are linked to a number of somatic and psychiatric diseases, emphasizing the importance of precise neuronal control of effector pathways. Pharmacological evidence suggests a role for glucagon-like peptide-1 (GLP-1) in modulating stress responses. Using a targeted knockdown of the GLP-1 receptor in the single-minded 1 neurons, we show dependence of paraventricular nucleus GLP-1 signaling in the coordination of neuroendocrine, autonomic, and behavioral responses to acute and chronic stress. To our knowledge, this is the first direct demonstration of an obligate brainstem-to-hypothalamus circuit orchestrating general stress excitation across multiple effector systems. These findings provide novel information regarding signaling pathways coordinating central control of whole-body stress reactivity.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Histona-Lisina N-Metiltransferase/genética , Proteínas Repressoras/genética , Transdução de Sinais/genética , Estresse Psicológico/fisiopatologia , Doença Aguda , Animais , Ansiedade/etiologia , Ansiedade/genética , Ansiedade/psicologia , Comportamento Animal , Doença Crônica , Ingestão de Alimentos , Receptor do Peptídeo Semelhante ao Glucagon 1/genética , Frequência Cardíaca/genética , Sistema Hipotálamo-Hipofisário/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Núcleo Hipotalâmico Paraventricular , Sistema Hipófise-Suprarrenal/fisiopatologia , Estresse Psicológico/psicologia , Natação/psicologia
11.
Cell Mol Neurobiol ; 38(1): 25-35, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28895001

RESUMO

Hindbrain neurons in the nucleus of the solitary tract (NTS) are critical for regulation of hypothalamo-pituitary-adrenocortical (HPA) responses to stress. It is well known that noradrenergic (as well as adrenergic) neurons in the NTS send direct projections to hypophysiotropic corticotropin-releasing hormone (CRH) neurons and control activation of HPA axis responses to acute systemic (but not psychogenic) stressors. Norepinephrine (NE) signaling via alpha1 receptors is primarily excitatory, working either directly on CRH neurons or through presynaptic activation of glutamate release. However, there is also evidence for NE inhibition of CRH neurons (possibly via beta receptors), an effect that may occur at higher levels of stimulation, suggesting that NE effects on the HPA axis may be context-dependent. Lesions of ascending NE inputs to the paraventricular nucleus attenuate stress-induced ACTH but not corticosterone release after chronic stress, indicating reduction in central HPA drive and increased adrenal sensitivity. Non-catecholaminergic NTS glucagon-like peptide 1/glutamate neurons play a broader role in stress regulation, being important in HPA activation to both systemic and psychogenic stressors as well as HPA axis sensitization under conditions of chronic stress. Overall, the data highlight the importance of the NTS as a key regulatory node for coordination of acute and chronic stress.


Assuntos
Córtex Suprarrenal/metabolismo , Corpos Aórticos/metabolismo , Sistema Hipotálamo-Hipofisário/fisiologia , Sistema Hipófise-Suprarrenal/fisiologia , Núcleo Solitário/metabolismo , Estresse Psicológico/metabolismo , Córtex Suprarrenal/efeitos dos fármacos , Animais , Corpos Aórticos/efeitos dos fármacos , Corticosterona/metabolismo , Corticosterona/farmacologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Ácido Glutâmico/metabolismo , Ácido Glutâmico/farmacologia , Humanos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Núcleo Solitário/efeitos dos fármacos , Estresse Psicológico/psicologia
12.
Stress ; 21(5): 464-473, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29166811

RESUMO

The late adolescent period is characterized by marked neurodevelopmental and endocrine fluctuations in the transition to early adulthood. Adolescents are highly responsive to the external environment, which enhances their ability to adapt and recover from challenges when given nurturing influences, but also makes them vulnerable to aberrant development when exposed to prolonged adverse situations. Female rats are particularly sensitive to the effects of chronic stress in adolescence, which manifests as passive coping strategies and blunted hypothalamo-pituitary adrenocortical (HPA) stress responses in adulthood. We sought to intervene by exposing adolescent rats to environmental enrichment (EE) immediately prior to and during chronic stress, hypothesizing that EE would minimize or prevent the long-term effects of stress that emerge in adult females. To test this, we exposed male and female rats to EE on postnatal days (PND) 33-60 and implemented chronic variable stress (CVS) on PND 40-60. CVS consisted of twice-daily unpredictable stressors. Experimental groups included: CVS/unenriched, unstressed/EE, CVS/EE and unstressed/unenriched (n = 10 of each sex/group). In adulthood, we measured behavior in the open field test and forced swim test (FST) and collected blood samples following the FST. We found that environmental enrichment given during the adolescent period prevented the chronic stress-induced transition to passive coping in the FST and reversed decreases in peak adrenocortical responsiveness observed in adult females. Adolescent enrichment had little to no effect on males or unstressed females tested in adulthood, indicating that beneficial effects are specific to females that were exposed to chronic stress.


Assuntos
Sistema Hipotálamo-Hipofisário/fisiopatologia , Sistema Hipófise-Suprarrenal/fisiopatologia , Estresse Fisiológico/fisiologia , Estresse Psicológico/fisiopatologia , Animais , Comportamento Animal/fisiologia , Meio Ambiente , Feminino , Abrigo para Animais , Masculino , Atividade Motora/fisiologia , Ratos , Ratos Sprague-Dawley , Fatores Sexuais
13.
Brain Behav Immun ; 57: 263-270, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27177449

RESUMO

Previous work from our group has shown that chronic homotypic stress (repeated restraint - RR) increases microglial morphological activation in the prefrontal cortex (PFC), while chronic heterotypic stress (chronic variable stress - CVS) produces no such effect. Therefore, we hypothesized that stressor modality would also determine the susceptibility of the PFC to a subsequent inflammatory stimulus (low dose lipopolysaccharide (LPS)). We found that RR, but not CVS, increased Iba-1 soma size in the PFC after LPS injection, consistent with microglial activation. In contrast, CVS decreased gene expression of proinflammatory cytokines and Iba-1 in the PFC under baseline conditions, which were not further affected by LPS. Thus, RR appears to promote microglial responses to LPS, whereas CVS is largely immunosuppressive. The results suggest that neuroimmune changes caused by CVS may to some extent protect the PFC from subsequent inflammatory stimuli. These data suggest that modality and/or intensity of stressful experiences will be a major determinant of central inflammation and its effect on prefrontal cortex-mediated functions.


Assuntos
Citocinas/metabolismo , Lipopolissacarídeos/farmacologia , Microglia/imunologia , Córtex Pré-Frontal/imunologia , Estresse Psicológico/imunologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Lipopolissacarídeos/administração & dosagem , Masculino , Proteínas dos Microfilamentos/metabolismo , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Estresse Psicológico/classificação , Estresse Psicológico/metabolismo
14.
Front Neuroendocrinol ; 35(2): 180-196, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24361584

RESUMO

Environmental stimuli that signal real or potential threats to homeostasis lead to glucocorticoid secretion by the hypothalamic-pituitary-adrenocortical (HPA) axis. Glucocorticoids promote energy redistribution and are critical for survival and adaptation. This adaptation requires the integration of multiple systems and engages key limbic-neuroendocrine circuits. Consequently, glucocorticoids have profound effects on synaptic physiology, circuit regulation of stress responsiveness, and, ultimately, behavior. While glucocorticoids initiate adaptive processes that generate energy for coping, prolonged or inappropriate glucocorticoid secretion becomes deleterious. Inappropriate processing of stressful information may lead to energetic drive that does not match environmental demand, resulting in risk factors for pathology. Thus, dysregulation of the HPA axis may promote stress-related illnesses (e.g. depression, PTSD). This review summarizes the latest developments in central glucocorticoid actions on synaptic, neuroendocrine, and behavioral regulation. Additionally, these findings will be discussed in terms of the energetic integration of stress and the importance of context-specific regulation of glucocorticoids.


Assuntos
Comportamento/fisiologia , Encéfalo/fisiologia , Hormônio Liberador da Corticotropina/fisiologia , Glucocorticoides/fisiologia , Estresse Fisiológico/fisiologia , Sinapses/fisiologia , Animais , Humanos
15.
16.
Stress ; 18(2): 221-32, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25765732

RESUMO

Chronic stress causes hypothalamo-pituitary-adrenal (HPA) axis hyperactivity and cardiovascular dyshomeostasis. Noradrenergic (NA) neurons in the nucleus of the solitary tract (NTS) are considered to play a role in these changes. In this study, we tested the hypothesis that NTS NA A2 neurons are required for cardiovascular and HPA axis responses to both acute and chronic stress. Adult male rats received bilateral microinjection into the NTS of 6-hydroxydopamine (6-OHDA) to lesion A2 neurons [cardiovascular study, n = 5; HPA study, n = 5] or vehicle [cardiovascular study, n = 6; HPA study, n = 4]. Rats were exposed to acute restraint stress followed by 14 d of chronic variable stress (CVS). On the last day of testing, rats were placed in a novel elevated plus maze (EPM) to test post-CVS stress responses. Lesions of NTS A2 neurons reduced the tachycardic response to acute restraint, confirming that A2 neurons promote sympathetic activation following acute stress. In addition, CVS increased the ratio of low-frequency to high-frequency power for heart rate variability, indicative of sympathovagal imbalance, and this effect was significantly attenuated by 6-OHDA lesion. Lesions of NTS A2 neurons reduced acute restraint-induced corticosterone secretion, but did not affect the corticosterone response to the EPM, indicating that A2 neurons promote acute HPA axis responses, but are not involved in CVS-mediated HPA axis sensitization. Collectively, these data indicate that A2 neurons promote both cardiovascular and HPA axis responses to acute stress. Moreover, A2 catecholaminergic neurons may contribute to the potentially deleterious enhancement of sympathetic drive following chronic stress.


Assuntos
Neurônios Adrenérgicos/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Oxidopamina/farmacologia , Núcleo Solitário/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos , Simpatolíticos/farmacologia , Neurônios Adrenérgicos/fisiologia , Animais , Sistema Cardiovascular , Corticosterona/metabolismo , Frequência Cardíaca/fisiologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/metabolismo , Ratos , Ratos Sprague-Dawley , Núcleo Solitário/citologia , Núcleo Solitário/fisiologia , Estresse Fisiológico/fisiologia , Estresse Psicológico
17.
J Vis ; 15(6): 3, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26047359

RESUMO

Studies of covert spatial attention have largely used motion, orientation, and contrast stimuli as these features are fundamental components of vision. The feature dimension of color is also fundamental to visual perception, particularly for catarrhine primates, and yet very little is known about the effects of spatial attention on color perception. Here we present results using novel dynamic color stimuli in both discrimination and color-change detection tasks. We find that our stimuli yield comparable discrimination thresholds to those obtained with static stimuli. Further, we find that an informative spatial cue improves performance and speeds response time in a color-change detection task compared with an uncued condition, similar to what has been demonstrated for motion, orientation, and contrast stimuli. Our results demonstrate the use of dynamic color stimuli for an established psychophysical task and show that color stimuli are well suited to the study of spatial attention.


Assuntos
Percepção de Cores/fisiologia , Sinais (Psicologia) , Processamento Espacial/fisiologia , Adulto , Atenção/fisiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Psicofísica , Tempo de Reação
18.
J Neurosci ; 33(11): 4825-33, 2013 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-23486953

RESUMO

Obesity is associated with increased levels of angiotensin-II (Ang-II), which activates angiotensin type 1a receptors (AT1a) to influence cardiovascular function and energy homeostasis. To test the hypothesis that specific AT1a within the brain control these processes, we used the Cre/lox system to delete AT1a from the paraventricular nucleus of the hypothalamus (PVN) of mice. PVN AT1a deletion did not affect body mass or adiposity when mice were maintained on standard chow. However, maintenance on a high-fat diet revealed a gene by environment interaction whereby mice lacking AT1a in the PVN had increased food intake and decreased energy expenditure that augmented body mass and adiposity relative to controls. Despite this increased adiposity, PVN AT1a deletion reduced systolic blood pressure, suggesting that this receptor population mediates the positive correlation between adiposity and blood pressure. Gene expression studies revealed that PVN AT1a deletion decreased hypothalamic expression of corticotrophin-releasing hormone and oxytocin, neuropeptides known to control food intake and sympathetic nervous system activity. Whole-cell patch-clamp recordings confirmed that PVN AT1a deletion eliminates responsiveness of PVN parvocellular neurons to Ang-II, and suggest that Ang-II responsiveness is increased in obese wild-type mice. Central inflammation is associated with metabolic and cardiovascular disorders and PVN AT1a deletion reduced indices of hypothalamic inflammation. Collectively, these studies demonstrate that PVN AT1a regulate energy balance during environmental challenges that promote metabolic and cardiovascular pathologies. The implication is that the elevated Ang-II that accompanies obesity serves as a negative feedback signal that activates PVN neurons to alleviate weight gain.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Obesidade , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptor Tipo 1 de Angiotensina/deficiência , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Glândulas Suprarrenais/metabolismo , Análise de Variância , Angiotensina II/farmacologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Composição Corporal/genética , Peso Corporal/genética , Encéfalo/patologia , Calorimetria , Sistema Cardiovascular/fisiopatologia , Modelos Animais de Doenças , Ingestão de Líquidos/genética , Ingestão de Alimentos/genética , Metabolismo Energético/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Técnicas In Vitro , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/prevenção & controle , Técnicas de Patch-Clamp , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Cloreto de Sódio/metabolismo , Telemetria
19.
J Neurophysiol ; 111(11): 2343-54, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24623511

RESUMO

In the natural environment, humans make saccades almost continuously. In many eye movement experiments, however, observers are required to fixate for unnaturally long periods of time. The resulting long and monotonous experimental sessions can become especially problematic when collecting data in a clinical setting, where time can be scarce and subjects easily fatigued. With this in mind, we tested whether the well-studied motor learning process of saccade adaptation could be induced with a dramatically shortened intertrial interval. Observers made saccades to targets that stepped left or right either ∼250 ms or ∼1,600 ms after the saccade landed. In experiment I, we tested baseline saccade parameters to four different target amplitudes (5°, 10°, 15°, and 20°) in the two timing settings. In experiments II and III, we adapted 10° saccades via 2° intrasaccadic steps either backwards or forwards, respectively. Seven subjects performed eight separate adaptation sessions (2 intertrial timings × 2 adaptation direction × 2 session trial lengths). Adaptation proceeded remarkably similarly in both timing conditions across the multiple sessions. In the faster-paced sessions, robust adaptation was achieved in under 2 min, demonstrating the efficacy of our approach to streamlining saccade adaptation experiments. Although saccade amplitudes were similar between conditions, the faster-paced condition unexpectedly resulted in significantly higher peak velocities in all subjects. This surprising finding demonstrates that the stereotyped "main sequence" relationship between saccade amplitude and peak velocity is not as fixed as originally thought.


Assuntos
Adaptação Fisiológica/fisiologia , Percepção de Movimento/fisiologia , Destreza Motora/fisiologia , Plasticidade Neuronal/fisiologia , Estimulação Luminosa/métodos , Movimentos Sacádicos/fisiologia , Análise e Desempenho de Tarefas , Feminino , Humanos , Masculino , Adulto Jovem
20.
Eur J Neurosci ; 39(11): 1903-11, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24766138

RESUMO

Chronic variable stress (CVS) exposure modifies the paraventricular nucleus of the hypothalamus (PVN) in a manner consistent with enhanced central drive of the hypothalamo-pituitary-adrenocortical (HPA) axis. As previous reports suggest that post-stress enhancement of norepinephrine (NE) action contributes to chronic stress regulation at the level of the PVN, we hypothesised that PVN-projecting NE neurons were necessary for the stress facilitatory effects of CVS. Following intra-PVN injection of saporin toxin conjugated to a dopamine beta-hydroxylase (DBH) antibody (DSAP), in rats PVN DBH immunoreactivity was almost completely eliminated, but immunoreactive afferents to other key regions involved in stress integration were spared (e.g. DBH fiber densities were unaffected in the central nucleus of the amygdala). Reductions in DBH-positive fiber density were associated with reduced numbers of DBH-immunoreactive neurons in the nucleus of the solitary tract and locus coeruleus. Following 2 weeks of CVS, DSAP injection did not alter stress-induced adrenal hypertrophy or attenuation of body weight gain, indicating that PVN-projecting NE [and epinephrine (E)] neurons are not essential for these physiological effects of chronic stress. In response to acute restraint stress, PVN-targeted DSAP injection attenuated peak adrenocorticotrophic hormone (ACTH) and corticosterone in controls, but only attenuated peak ACTH in CVS animals, suggesting that enhanced adrenal sensitivity compensated for reduced excitatory drive of the PVN. Our data suggest that PVN-projecting NE/E neurons contribute to the generation of acute stress responses, and are required for HPA axis drive (ACTH release) during chronic stress. However, loss of NE/E drive at the PVN appears to be buffered by compensation at the level of the adrenal.


Assuntos
Fibras Adrenérgicas/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Estresse Psicológico/fisiopatologia , Hormônio Adrenocorticotrópico/sangue , Animais , Corticosterona/sangue , Neurônios Dopaminérgicos/fisiologia , Sistema Hipotálamo-Hipofisário/fisiopatologia , Masculino , Núcleo Hipotalâmico Paraventricular/citologia , Sistema Hipófise-Suprarrenal/fisiopatologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa