Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 224
Filtrar
1.
EMBO Rep ; 24(12): e57176, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-37870400

RESUMO

Chronic stress induces depression and insulin resistance, between which there is a bidirectional relationship. However, the mechanisms underlying this comorbidity remain unclear. White adipose tissue (WAT), innervated by sympathetic nerves, serves as a central node in the interorgan crosstalk through adipokines. Abnormal secretion of adipokines is involved in mood disorders and metabolic morbidities. We describe here a brain-sympathetic nerve-adipose circuit originating in the hypothalamic paraventricular nucleus (PVN) with a role in depression and insulin resistance induced by chronic stress. PVN neurons are labelled after inoculation of pseudorabies virus (PRV) into WAT and are activated under restraint stress. Chemogenetic manipulations suggest a role for the PVN in depression and insulin resistance. Chronic stress increases the sympathetic innervation of WAT and downregulates several antidepressant and insulin-sensitizing adipokines, including leptin, adiponectin, Angptl4 and Sfrp5. Chronic activation of the PVN has similar effects. ß-adrenergic receptors translate sympathetic tone into an adipose response, inducing downregulation of those adipokines and depressive-like behaviours and insulin resistance. We finally show that AP-1 has a role in the regulation of adipokine expression under chronic stress.


Assuntos
Resistência à Insulina , Núcleo Hipotalâmico Paraventricular , Ratos , Animais , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos Sprague-Dawley , Depressão , Obesidade/metabolismo , Adipocinas/metabolismo , Adipocinas/farmacologia
2.
Biol Reprod ; 110(2): 339-354, 2024 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-37971364

RESUMO

Entering pregnancy with a history of adversity, including adverse childhood experiences and racial discrimination stress, is a predictor of negative maternal and fetal health outcomes. Little is known about the biological mechanisms by which preconception adverse experiences are stored and impact future offspring health outcomes. In our maternal preconception stress (MPS) model, female mice underwent chronic stress from postnatal days 28-70 and were mated 2 weeks post-stress. Maternal preconception stress dams blunted the pregnancy-induced shift in the circulating extracellular vesicle proteome and reduced glucose tolerance at mid-gestation, suggesting a shift in pregnancy adaptation. To investigate MPS effects at the maternal:fetal interface, we probed the mid-gestation placental, uterine, and fetal brain tissue transcriptome. Male and female placentas differentially regulated expression of genes involved in growth and metabolic signaling in response to gestation in an MPS dam. We also report novel offspring sex- and MPS-specific responses in the uterine tissue apposing these placentas. In the fetal compartment, MPS female offspring reduced expression of neurodevelopmental genes. Using a ribosome-tagging transgenic approach we detected a dramatic increase in genes involved in chromatin regulation in a PVN-enriched neuronal population in females at PN21. While MPS had an additive effect on high-fat-diet (HFD)-induced weight gain in male offspring, both MPS and HFD were necessary to induce significant weight gain in female offspring. These data highlight the preconception period as a determinant of maternal health in pregnancy and provides novel insights into mechanisms by which maternal stress history impacts offspring developmental programming.


Assuntos
Placenta , Aumento de Peso , Humanos , Gravidez , Camundongos , Feminino , Masculino , Animais , Placenta/metabolismo , Feto/metabolismo , Transdução de Sinais , Dieta Hiperlipídica/efeitos adversos
3.
J Neurosci ; 42(14): 2885-2904, 2022 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-35197315

RESUMO

Oxytocin (OXT) neurons in paraventricular nucleus of hypothalamus (PVN) are involved in modulating multiple functions, including social, maternal, feeding, and emotional related behaviors. PVN OXT neurons are canonically classified into magnocellular (Magno) and parvocellular (Parvo) subtypes. However, morpho-electric properties and the diversity of PVN OXT neurons are not well investigated. In this study, we profiled the morpho-electric properties of PVN OXT neurons by combining transgenic mice, electrophysiological recording, morphologic reconstruction, and unsupervised clustering analyses. Total 224 PVN OXT neurons from 23 mice were recorded and used for analyses in this study, and 29 morpho-electric parameters were measured. Magno and Parvo OXT neurons have prominent differences in their morpho-electric features, and PVN OXT neurons in male and female mice share similar neuronal properties. Some morpho-electric features of PVN OXT neurons, especially Magno neurons, exhibit significant diverse changes along the rostral-caudal axis. Furthermore, we find that PVN OXT neurons are classified into at least six subtypes based on their morpho-electric properties via unsupervised clustering. Only one Magno-Parvo mixed subtype in posterior PVN subregion, but not the other five subtypes, showed significant neuronal activity change in different feeding conditions. Our study supports the diversity of PVN OXT neurons and subtle neuron classification will promote excavating the functions of oxytocinergic system.SIGNIFICANCE STATEMENT Oxytocin (OXT) is well known for its function in labor induction, but it also plays multiple roles in social, feeding, and emotional behaviors via modulating different brain regions. Paraventricular nucleus of hypothalamus (PVN) OXT neurons are traditionally classified into magnocellular and parvocellular. However, functional and single-cell transcriptomic studies indicate that OXT neurons should be further classified. Here, we thoroughly investigated the morpho-electric properties and spatial distribution of PVN OXT neurons, and find that OXT neurons have at least six subtypes based on their morpho-electric features. Among these six subtypes, only one magnocellular-parvocellular mixed subtype, which are distributed in the posterior PVN subregion, change their activities with different feeding states. Our study uncovers the diversity of PVN OXT neurons and suggests the necessary of subtle neuronal classification.


Assuntos
Neurônios , Ocitocina , Núcleo Hipotalâmico Paraventricular , Animais , Feminino , Hipotálamo , Masculino , Camundongos , Neurônios/fisiologia , Ocitocina/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Ratos , Ratos Wistar
4.
Synapse ; 77(2): e22259, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36271777

RESUMO

Angiotensinergic, GABAergic, and glutamatergic neurons are present in the parvocellular region of the paraventricular nucleus (PVNp). It has been shown that microinjection of AngII into the PVNp increases arterial pressure (AP) and heart rate (HR). The presence of synapses between the angiotensinergic, GABAergic, and glutamatergic neurons has been shown in the PVNp. In this study, we investigated the possible interaction between these three systems of the PVNp for control of AP and HR. All drugs were bilaterally (100 nl/side) microinjected into the PVNp of urethane-anesthetized rats, and AP and HR were recorded continuously. Microinjection of AngII into the PVNp produced pressor and tachycardia responses. Pretreatment of PVNp with AP5 or CNQX, glutamatergic NMDA and AMPA receptors antagonists, attenuated the responses to AngII. Pretreatment of PVNp with bicuculline greatly attenuated the pressor and tachycardia responses to AngII. In conclusion, this study provides the first evidence that pressor and tachycardia responses to microinjection of AngII into the PVNp are partly mediated by both NMDA and non-NMDA receptors of glutamate. Activation of glutamatergic neurons by AngII stimulates the sympathoexcitatory neurons. We also showed that the responses to AngII were strongly mediated by GABAA receptors, probably through activation of GABAergic neurons, which in turn inhibit sympathoinhibitory neurons.


Assuntos
Núcleo Hipotalâmico Paraventricular , Taquicardia , Ratos , Animais , Pressão Sanguínea/fisiologia , Frequência Cardíaca/fisiologia , Neurônios GABAérgicos
5.
Appetite ; 188: 106618, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37257508

RESUMO

Melanocortin and neuropeptide-Y (NPY) are both involved in feeding and energy regulation, and they have opposite effects in the paraventricular nucleus of the hypothalamus (PVN). The present study examined an interaction between melanocortin in the nucleus of the solitary tract (NTS) and NPY in the PVN. Male Sprague-Dawley rats were implanted with cannulae in the injection sites of interest. In Experiment 1, subjects received either the melanocortin 3/4-receptor (MC3/4) antagonist SHU9119 (0, 10, 50 and 100 pmol/0.5 µl) or the MC3/4 agonist MTII (0, 10, 50, 100 and 200 pmol/0.5 µl) into the NTS. Food intake was measured at 1, 2, 4, 6 and 24-h post-injection. Administration of SHU9119 into the NTS significantly and dose-dependently increased food intake at 1, 2, 4, 6 and 6-24-h, and administration of MTII into the NTS significantly and dose-dependently decreased 24-h free feeding. In Experiment 2, subjects received the MC3/4 agonist MTII (0, 10, 50, 100 and 200 pmol/0.5 µl) into the NTS just prior to NPY (0 and 1µg/0.5 µl) in the PVN. PVN injection of NPY stimulated feeding, and administration of MTII (50, 100 and 200 pmol) into the NTS significantly and dose-dependently decreased NPY-induced feeding at 2, 4, 6 and 6-24-h. These data suggest that there could be a neuronal association between melanocortin in the NTS and NPY in the PVN, and that the melanocortin system in the NTS has an antagonistic effect on NPY-induced feeding in the PVN.


Assuntos
Neuropeptídeo Y , Núcleo Solitário , Humanos , Ratos , Animais , Masculino , Neuropeptídeo Y/farmacologia , Ratos Sprague-Dawley , Núcleo Hipotalâmico Paraventricular/fisiologia , Melanocortinas/farmacologia , Ingestão de Alimentos/fisiologia
6.
Int J Mol Sci ; 24(9)2023 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-37175953

RESUMO

1,5-Anhydro-D-fructose (1,5-AF) is a bioactive monosaccharide that is produced by the glycogenolysis in mammalians and is metabolized to 1,5-anhydro-D-glucitol (1,5-AG). 1,5-AG is used as a marker of glycemic control in diabetes patients. 1,5-AF has a variety of physiological activities, but its effects on energy metabolism, including feeding behavior, are unclarified. The present study examined whether 1,5-AF possesses the effect of satiety. Peroral administration of 1,5-AF, and not of 1,5-AG, suppressed daily food intake. Intracerebroventricular (ICV) administration of 1,5-AF also suppressed feeding. To investigate the neurons targeted by 1,5-AF, we investigated c-Fos expression in the hypothalamus and brain stem. ICV injection of 1,5-AF significantly increased c-Fos positive oxytocin neurons and mRNA expression of oxytocin in the paraventricular nucleus (PVN). Moreover, 1,5-AF increased cytosolic Ca2+ concentration of oxytocin neurons in the PVN. Furthermore, the satiety effect of 1,5-AF was abolished in oxytocin knockout mice. These findings reveal that 1,5-AF activates PVN oxytocin neurons to suppress feeding, indicating its potential as the energy storage monitoring messenger to the hypothalamus for integrative regulation of energy metabolism.


Assuntos
Ocitocina , Núcleo Hipotalâmico Paraventricular , Camundongos , Animais , Núcleo Hipotalâmico Paraventricular/metabolismo , Ocitocina/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Mamíferos/metabolismo
7.
Nutr Neurosci ; 25(5): 1105-1114, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-33151127

RESUMO

The orexin peptides promote hedonic intake and other reward behaviors through different brain sites. The opioid dynorphin peptides are co-released with orexin peptides but block their effects on reward in the ventral tegmental area (VTA). We previously showed that in the paraventricular hypothalamic nucleus (PVN), dynorphin and not orexin peptides enhance hedonic intake, suggesting they have brain-site-specific effects. Obesity alters the expression of orexin and dynorphin receptors, but whether their expression across different brain sites is important to hedonic intake is unclear. We hypothesized that hedonic intake is regulated by orexin and dynorphin peptides in PVN and that hedonic intake in obesity correlates with expression of their receptors. Here we show that in mice, injection of DYN-A1-13 (an opioid dynorphin peptide) in the PVN enhanced hedonic intake, whereas in the VTA, injection of OXA (orexin-A, an orexin peptide) enhanced hedonic intake. In PVN, OXA blunted the increase in hedonic intake caused by DYN-A1-13. In PVN, injection of norBNI (opioid receptor antagonist) reduced hedonic intake but a subsequent OXA injection failed to increase hedonic intake, suggesting that OXA activity in PVN is not influenced by endogenous opioid activity. In the PVN, DYN-A1-13 increased the intake of the less-preferred food in a two-food choice task. In obese mice fed a cafeteria diet, orexin 1 receptor mRNA across brain sites involved in hedonic intake correlated with fat preference but not caloric intake. Together, these data support that orexin and dynorphin peptides regulate hedonic intake in an opposing manner with brain-site-specific effects.


Assuntos
Dinorfinas , Núcleo Hipotalâmico Paraventricular , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Encéfalo/metabolismo , Dinorfinas/metabolismo , Dinorfinas/farmacologia , Camundongos , Obesidade/metabolismo , Orexinas/metabolismo
8.
Molecules ; 27(3)2022 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-35164012

RESUMO

5-Hydroxymethyl-2'-deoxycytidine (5hmdC) phosphoramidite and triphosphate are important building blocks in 5hmdC-containing DNA synthesis for epigenetic studies. However, efficient and practical methods for the synthesis of these compounds are still limited. The current research provides an intensively improved synthetic method that enables the preparation of commercially available cyanoethyl-protected 5hmdC phosphoramidite with an overall yield of 39% on 5 g scale. On the basis of facile and efficient accesses to cyanoethyl protected-5hmdU and 5hmdC intermediates, two efficient synthetic routes for 5hmdC triphosphate were also developed.

9.
Physiol Genomics ; 53(1): 12-21, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33252993

RESUMO

Hypertension is a global health burden. The hypothalamic paraventricular nucleus (PVN) is an essential component of the neuronal network that regulates sodium homeostasis and blood pressure (BP). Previously, we have shown PVN-specific G protein-coupled receptor-coupled Gαi2 subunit proteins are essential to counter the development of salt-sensitive hypertension by mediating the sympathoinhibitory and natriuretic responses to increased dietary sodium intake to maintain sodium homeostasis and normotension. However, the cellular localization and identity of PVN Gαi2-expressing neurons are currently unknown. In this study using in situ hybridization, we determined the neuroanatomical characterization of Gαi2-expressing PVN neurons in 3-mo-old male and female Sprague-Dawley rats. We observed that Gαi2-expressing neurons containing Gnai2 mRNA are highly localized in the parvocellular region of the hypothalamic PVN. At level 2 of the hypothalamic PVN, Gnai2 mRNA colocalized with ∼ 85% of GABA-expressing neurons and ∼28% of glutamatergic neurons. Additionally, within level 2 Gnai2 mRNA colocalized with ∼75% of corticotrophin-releasing hormone PVN neurons. Gnai2 neurons had lower colocalization with tyrosine hydroxylase (∼33%)-, oxytocin (∼6%)-, and arginine vasopressin (∼10%)-expressing parvocellular neurons in level 2 PVN. Colocalization was similar among male and female rats. The high colocalization of Gnai2 mRNA with GABAergic neurons, in conjunction with our previous findings that PVN Gαi2 proteins mediate sympathoinhibition, suggests that Gαi2 proteins potentially modulate GABAergic signaling to impact sympathetic outflow and BP.


Assuntos
Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/anatomia & histologia , Núcleo Hipotalâmico Paraventricular/citologia , Animais , Hormônio Liberador da Corticotropina/metabolismo , Feminino , Neurônios GABAérgicos/metabolismo , Ácido Glutâmico/metabolismo , Masculino , Ocitocina/metabolismo , Ratos Sprague-Dawley , Tirosina 3-Mono-Oxigenase/metabolismo
10.
J Physiol ; 599(14): 3531-3547, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34053068

RESUMO

KEY POINTS: Purinergic and glutamatergic signalling pathways play a key role in regulating the activity of hypothalamic magnocellular neurosecretory neurons (MNNs). However, the precise cellular mechanisms by which ATP and glutamate act in concert to regulate osmotically driven MNN neuronal excitability remains unknown. Here, we report that ATP acts on purinergic P2 receptors in MNNs to potentiate in a Ca2+ -dependent manner extrasynaptic NMDAR function. The P2-NMDAR coupling is engaged in response to an acute hyperosmotic stimulation, contributing to osmotically driven firing activity in MNNs. These results help us to better understand the precise mechanisms contributing to the osmotic regulation of firing activity and hormone release from MNNs. ABSTRACT: The firing activity of hypothalamic magnocellular neurosecretory neurons (MNNs) located in the paraventricular nucleus (PVN) and supraoptic nucleus (SON) is coordinated by the combined, fine-tuned action of intrinsic membrane properties, synaptic and extrasynaptic signalling. Among these, purinergic and glutamatergic signalling pathways have been shown to play a key role regulating the activity of MNNs. However, the precise cellular mechanisms by which ATP and glutamate act in concert to regulate osmotically driven MNN neuronal excitability remains unknown. Whole-cell patch-clamp recordings obtained from MNNs showed that ATP (100 µM) induced an increase in firing rate, an effect that was blocked by either 4-[[4-formyl-5-hydroxy-6-methyl-3-[(phosphonooxy)methyl]2-pyridinyl]azo]1,3-benzenedisulfonic acid tetrasodium salt (PPADS) (10 µM) or kynurenic acid (1 mm). While ATP did not affect the frequency or magnitude of glutamatergic excitatory postsynaptic currents (EPSCs), it induced an inward shift in the holding current that was prevented by PPADS or kynurenic acid treatment, suggesting that ATP enhances a tonic extrasynaptic glutamatergic excitatory current. We observed that ATP-potentiated glutamatergic receptor-mediated currents were evoked by focal application of L-glu (1 mm) and NMDA (50 µM), but not AMPA (50 µM). ATP potentiation of NMDA-evoked currents was blocked by PPADS (10 µM) and by chelation of intracellular Ca2+ with BAPTA (10 mm). Finally, we report that a hyperosmotic stimulus (mannitol 1%, +55 mOsm/kgH2 O) potentiated NMDA-evoked currents and increased MNN firing activity, effects that were blocked by PPADS. Taken together, our data support a functional excitatory coupling between P2 and extrasynaptic NMDA receptors in MNNs, which is engaged in response to an acute hyperosmotic stimulus.


Assuntos
Ácido Glutâmico , Receptores de N-Metil-D-Aspartato , Receptores Purinérgicos P2 , Animais , Neurônios/metabolismo , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Purinérgicos P2/metabolismo , Núcleo Supraóptico/metabolismo
11.
Int J Mol Sci ; 22(21)2021 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-34769243

RESUMO

Adipocytokine chemerin is a biologically active molecule secreted from adipose tissue. Chemerin elicits a variety of functions via chemokine-like receptor 1 (CMKLR1). The cardiovascular center in brain that regulates blood pressure (BP) is involved in pathophysiology of systemic hypertension. Thus, we explored the roles of brain chemerin/CMKLR1 on regulation of BP in spontaneously hypertensive rats (SHR). For this aim, we examined effects of intracerebroventricular (i.c.v.) injection of CMKLR1 small interfering (si)RNA on both systemic BP as measured by tail cuff system and protein expression in paraventricular nucleus (PVN) of SHR as determined by Western blotting. We also examined both central and peripheral protein expression of chemerin by Western blotting. Systolic BP of SHR but not normotensive Wistar Kyoto rats (WKY) was decreased by CMKLR1 siRNA. The decrease of BP by CMKLR1 siRNA persisted for 3 days. Protein expression of CMKLR1 in PVN of SHR tended to be increased compared with WKY, which was suppressed by CMKLR1 siRNA. Protein expression of chemerin in brain, peripheral plasma, and adipose tissue was not different between WKY and SHR. In summary, we for the first time revealed that the increased protein expression of CMKLR1 in PVN is at least partly responsible for systemic hypertension in SHR.


Assuntos
Regulação da Expressão Gênica , Hipertensão/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores de Quimiocinas/biossíntese , Animais , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY
12.
Int J Mol Sci ; 22(16)2021 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-34445795

RESUMO

Stress adaptation is of utmost importance for the maintenance of homeostasis and, therefore, of life itself. The prevalence of stress-related disorders is increasing, emphasizing the importance of exploratory research on stress adaptation. Two major regulatory pathways exist: the hypothalamic-pituitary-adrenocortical axis and the sympathetic adrenomedullary axis. They act in unison, ensured by the enormous bidirectional connection between their centers, the paraventricular nucleus of the hypothalamus (PVN), and the brainstem monoaminergic cell groups, respectively. PVN and especially their corticotropin-releasing hormone (CRH) producing neurons are considered to be the centrum of stress regulation. However, the brainstem seems to be equally important. Therefore, we aimed to summarize the present knowledge on the role of classical neurotransmitters of the brainstem (GABA, glutamate as well as serotonin, noradrenaline, adrenaline, and dopamine) in stress adaptation. Neuropeptides, including CRH, might be co-localized in the brainstem nuclei. Here we focused on CRH as its role in stress regulation is well-known and widely accepted and other CRH neurons scattered along the brain may also complement the function of the PVN. Although CRH-positive cells are present on some parts of the brainstem, sometimes even in comparable amounts as in the PVN, not much is known about their contribution to stress adaptation. Based on the role of the Barrington's nucleus in micturition and the inferior olivary complex in the regulation of fine motoric-as the main CRH-containing brainstem areas-we might assume that these areas regulate stress-induced urination and locomotion, respectively. Further studies are necessary for the field.


Assuntos
Adaptação Fisiológica/fisiologia , Tronco Encefálico/metabolismo , Tronco Encefálico/fisiologia , Hormônio Liberador da Corticotropina/metabolismo , Estresse Fisiológico/fisiologia , Animais , Humanos , Neurônios/metabolismo , Neurônios/fisiologia
13.
J Neurosci ; 39(14): 2649-2663, 2019 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-30683681

RESUMO

The anterior lateral bed nucleus of the stria terminalis (alBST) expresses glucagon-like peptide-1 receptors (GLP1Rs) and receives input from caudal brainstem GLP1 neurons. GLP1 administered centrally reduces food intake and increases anxiety-like behavior and plasma corticosterone (cort) levels in rats, whereas central GLP1R antagonism has opposite effects. Anxiogenic threats and other stressors robustly activate c-fos expression in both GLP1-producing neurons and also in neurons within alBST subregions expressing GLP1R. To examine the functional role of GLP1R signaling within the alBST, adult male Sprague Dawley rats received bilateral alBST-targeted injections of an adeno-associated virus (AAV) vector expressing short hairpin RNA (shRNA) to knock down the translation of GLP1R mRNA (GLP1R-KD rats), or similar injections of a control AAV (CTRL rats). In situ hybridization revealed that GLP1R mRNA is expressed in a subset of GABAergic alBST neurons, and quantitative real-time PCR confirmed that GLP1R-KD rats displayed a significant 60% reduction in translatable GLP1R mRNA. Compared with CTRL rats, GLP1R-KD rats gained more body weight over time and displayed less anxiety-like behavior, including a loss of light-enhanced acoustic startle and less stress-induced hypophagia. Conversely, while baseline plasma cort levels were similar in GLP1R-KD and CTRL rats, GLP1R-KD rats displayed a prolonged stress-induced elevation of plasma cort levels. GLP1R-KD and CTRL rats displayed similar home cage food intake and a similar hypophagic response to systemic Exendin-4, a GLP1R agonist that crosses the blood-brain barrier. We conclude that GLP1R expressed within the alBST contributes to multiple behavioral responses to anxiogenic threats, yet also serves to limit the plasma cort response to acute stress.SIGNIFICANCE STATEMENT Anxiety is an affective and physiological state that supports threat avoidance. Identifying the neural bases of anxiety-like behaviors in animal models is essential for understanding mechanisms that contribute to normative and pathological anxiety in humans. In rats, anxiety/avoidance behaviors can be elicited or enhanced by visceral or cognitive threats that increase glucagon-like peptide-1 (GLP1) signaling from the caudal brainstem to the hypothalamus and limbic forebrain. Data reported here support a role for limbic GLP1 receptor signaling to enhance anxiety-like behavior and to attenuate stress-induced elevations in plasma cort levels in rats. Improved understanding of central GLP1 neural pathways that impact emotional responses to stress could expand potential therapeutic options for anxiety and other stress-related disorders in humans.


Assuntos
Ansiedade/metabolismo , Regulação do Apetite/fisiologia , Corticosterona/sangue , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , RNA Mensageiro/metabolismo , Núcleos Septais/metabolismo , Estresse Psicológico/sangue , Animais , Ansiedade/prevenção & controle , Ansiedade/psicologia , Regulação do Apetite/efeitos dos fármacos , Biomarcadores/sangue , Receptor do Peptídeo Semelhante ao Glucagon 1/antagonistas & inibidores , Receptor do Peptídeo Semelhante ao Glucagon 1/genética , Masculino , Biossíntese de Proteínas/efeitos dos fármacos , Biossíntese de Proteínas/fisiologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , RNA Interferente Pequeno/administração & dosagem , Ratos , Ratos Sprague-Dawley , Reflexo de Sobressalto/efeitos dos fármacos , Reflexo de Sobressalto/fisiologia , Estresse Psicológico/genética , Estresse Psicológico/psicologia
14.
Stress ; 23(4): 457-465, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32093522

RESUMO

The chronic variable stress (CVS) paradigm is frequently used to model the changes in hypothalamic pituitary adrenal (HPA) axis function characteristic of many stress-related diseases. However, male C57BL/6 mice are typically resistant to CVS's effects, making it difficult to determine how chronic stress exposure may alter acute HPA function and regulation in these mice. As social support in rodents can profoundly influence physiological and behavioral processes, including the HPA axis, we sought to characterize the effects of CVS exposure on basal and acute stress-induced HPA axis function in pair- and single-housed adult male mice. Despite all subjects exhibiting decreased body weight gain after six weeks of CVS, the corticosterone response to a novel, acute restraint stressor was enhanced by CVS exclusively in single-housed males. CVS also significantly increased arginine vasopressin (AVP) mRNA in the hypothalamic paraventricular nucleus (PVN) in single-housed males only. Moreover, in single-, but not pair-housed mice, CVS attenuated decreases in circulating OT found following acute restraint. Only the effect of CVS to elevate PVN corticotropin releasing hormone (CRH) mRNA levels after an acute stressor was restricted to pair-housed mice. Collectively, our findings suggest that social isolation reveals effects of CVS on the HPA axis in male C57BL/6 mice.


Assuntos
Sistema Hipotálamo-Hipofisário , Sistema Hipófise-Suprarrenal , Animais , Corticosterona , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Hipotalâmico Paraventricular/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Isolamento Social , Estresse Psicológico
15.
Horm Behav ; 120: 104685, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31935400

RESUMO

Social relationships among spouses, family members, and friends are known to affect physical and mental health. In particular, long-lasting bonds between socio-sexual partners have profound effects on cognitive, social, emotional, and physical well-being. We have previously reported that pair bonding in monogamous prairie voles (Microtus ochrogaster) is prevented by a single prolonged stress (SPS) paradigm, which causes behavioral and endocrine symptoms resembling post-traumatic stress disorder (PTSD) patients in rats (Arai et al., 2016). Since fear memory function is crucial for anxiety-related disorders such as PTSD, we investigated the effects of pair bonding on fear learning in prairie voles. We applied an SPS paradigm to male prairie voles after the cohabitation with a male (cage-mate group) or female (pair-bonded group). The cage-mate group, but not the pair-bonded group, showed enhanced fear response in a contextual fear conditioning test following the SPS treatment. Immunohistochemical analyses revealed that cFos-positive cells in the central amygdala were increased in the pair-bonded group after the contextual fear conditioning test and that oxytocin immunoreactivity in the paraventricular nucleus of the hypothalamus was significantly higher in the pair-bonded group than the cage-mate group. This pair-bonding dependent blunting of fear memory response was confirmed by a passive avoidance test, another fear-based learning test. Interestingly, intracerebroventricular injection of an oxytocin receptor antagonist 30 min before the passive avoidance test blocked the blunting effect of pair bonding on fear learning. Thus, pair bonding between socio-sexual partners results in social buffering in the absence of the partner, blunting fear learning, which may be mediated by oxytocin signaling.


Assuntos
Condicionamento Psicológico/efeitos dos fármacos , Medo/efeitos dos fármacos , Aprendizagem/efeitos dos fármacos , Ornipressina/análogos & derivados , Ligação do Par , Receptores de Ocitocina/antagonistas & inibidores , Animais , Ansiedade/etiologia , Ansiedade/patologia , Arvicolinae/fisiologia , Aprendizagem da Esquiva/efeitos dos fármacos , Feminino , Infusões Intraventriculares , Masculino , Ornipressina/administração & dosagem , Ornipressina/farmacologia , Ocitocina/fisiologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Transtornos de Estresse Pós-Traumáticos/patologia , Transtornos de Estresse Pós-Traumáticos/psicologia
16.
Horm Behav ; 126: 104822, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32730760

RESUMO

Oxytocin is important for postnatal developmental experiences for mothers, infants, and transactions between them. Oxytocin is also implicated in adult affiliative behaviors, including social buffering of stress. There is evidence for connections between early life experience and adult oxytocin system functioning, but effects of early experience on behavioral, endocrine, and neurophysiological outcomes related to adult social buffering are not well explored. We use a limited bedding and nesting (LBN) material paradigm as an environmental disruption of early experiences and assessed central oxytocin systems in brain regions related to hypothalamic-pituitary-adrenal (HPA) axis regulation (paraventricular nucleus of the hypothalamus, amygdala, hippocampus). We also assessed developmentally-appropriate social behaviors and HPA reactivity during social buffering testing in adulthood. LBN litters had larger huddles and more pups visible compared to control litters during the first two weeks of life. LBN also altered the developmental trajectory of oxytocin-expressing cells and oxytocin receptor cells, with increases in oxytocin receptor cells at P15 in LBN pups. By adulthood, LBN females had more and LBN males had fewer oxytocin and oxytocin receptor cells in these areas compared to sex-matched controls. Adult LBN females, but not LBN males, had behavioral changes during social interaction and social buffering testing. The sex-specific effects of early experience on central oxytocin systems and social behavior may contribute to female resilience to early life adversity.


Assuntos
Sistema Hipotálamo-Hipofisário , Comportamento Materno/fisiologia , Ocitocina/metabolismo , Sistema Hipófise-Suprarrenal , Receptores de Ocitocina/metabolismo , Animais , Animais Recém-Nascidos , Comportamento Animal/fisiologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Feminino , Sistema Hipotálamo-Hipofisário/crescimento & desenvolvimento , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Comportamento de Nidação/fisiologia , Ocitocina/farmacologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Sistema Hipófise-Suprarrenal/crescimento & desenvolvimento , Sistema Hipófise-Suprarrenal/metabolismo , Ratos , Ratos Long-Evans , Caracteres Sexuais , Comportamento Social , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Estresse Psicológico/prevenção & controle
17.
J Cell Mol Med ; 23(1): 112-125, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30353660

RESUMO

Malignant ventricular arrhythmias (VAs) following myocardial infarction (MI) is a lethal complication resulting from sympathetic nerve hyperactivity. Numerous evidence have shown that inflammation within the paraventricular nucleus (PVN) participates in sympathetic hyperactivity. Our aim was to explore the role of Macrophage-inducible C-type lectin (Mincle) within the PVN in augmenting sympathetic activity following MI,and whether NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome/IL-1ß axis is involved in this activity. MI was induced by coronary artery ligation. Mincle expression localized in microglia within the PVN was markedly increased at 24 hours post-MI together with sympathetic hyperactivity, as indicated by measurement of the renal sympathetic nerve activity (RSNA) and norepinephrine (NE) concentration. Mincle-specific siRNA was administrated locally to the PVN, which consequently decreased microglial activation and sympathetic nerve activity. The MI rats exhibited a higher arrhythmia score after programmed electric stimulation than that treated with Mincle siRNA, suggesting that the inhibition of Mincle attenuated foetal ventricular arrhythmias post-MI. The underlying mechanism of Mincle in sympathetic hyperactivity was investigated in lipopolysaccharide (LPS)-primed naïve rats. Recombinant Sin3A-associated protein 130kD (rSAP130), an endogenous ligand for Mincle, induced high levels of NLRP3 and mature IL-1ß protein. PVN-targeted injection of NLRP3 siRNA or IL-1ß antagonist gevokizumab attenuated sympathetic hyperactivity. Together, the data indicated that the knockdown of Mincle in microglia within the PVN prevents VAs by attenuating sympathetic hyperactivity and ventricular susceptibility, in part by inhibiting its downstream NLRP3/IL-1ß axis following MI. Therapeutic interventions targeting Mincle signalling pathway could constitute a novel approach for preventing infarction injury.


Assuntos
Lectinas Tipo C/metabolismo , Microglia/metabolismo , Infarto do Miocárdio/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores Imunológicos/metabolismo , Sistema Nervoso Simpático/metabolismo , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Coração/fisiopatologia , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lectinas Tipo C/genética , Masculino , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Norepinefrina/metabolismo , Interferência de RNA , Ratos Sprague-Dawley , Receptores Imunológicos/genética , Sistema Nervoso Simpático/fisiopatologia
18.
J Neurophysiol ; 121(2): 602-608, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30565964

RESUMO

Activation of corticotropin-releasing hormone (CRH) type 2 receptors (CRHR2) in the nucleus of the solitary tract (NTS) contributes to the development of hypertension, but the source of CRH inputs to the NTS that increases blood pressure remains unknown. This study tested the hypothesis that activation of CRH-containing projections from the paraventricular nucleus of the hypothalamus (PVN) to the NTS increase blood pressure. We expressed channelrhodopsin 2 (ChR2), a light-sensitive ion channel, into CRH-containing neurons in the PVN. This was achieved by injecting Cre-inducible virus expressing ChR2 into the PVN of CRH-Cre mice. CRH-Cre mice are genetically modified mice expressing Cre recombinase only in neurons producing CRH. We found that optogenetic stimulation of CRH-containing somas in the PVN or CRH-containing fibers in the NTS originating from the PVN significantly increased blood pressure and heart rate. Microinjection of K-41498 (CRHR2 antagonist) into the NTS attenuated the pressor and tachycardiac responses induced by optogenetic stimulation of CRH-containing somas in the PVN. In vitro loose-patch recordings revealed that optogenetic stimulation of CRH-containing fibers in the NTS originating from the PVN significantly increased the discharge frequency of NTS neurons. This effect was attenuated by pretreatment of K-41498 and was abolished by pretreatment of kynurenic acid (nonselective glutamate receptor antagonist). These results suggest that activation of PVN-NTS CRH-containing projections increases blood pressure and heart rate. The cardiovascular responses may be mediated at least in part by the corelease of CRH and glutamate from NTS CRH-containing axons originating from the PVN. NEW & NOTEWORTHY Optogenetic stimulation of paraventricular nucleus of the hypothalamus (PVN) corticotropin-releasing hormone (CRH)-containing somas or nucleus of the solitary tract (NTS) CRH-containing fibers originating from the PVN increased blood pressure and heart rate. Corelease of CRH and glutamate from NTS CRH-containing axons originating from the PVN may contribute to the pressor and tachycardiac responses elicited by optogenetic stimulation of PVN CRH-containing somas.


Assuntos
Pressão Sanguínea , Hormônio Liberador da Corticotropina/metabolismo , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Núcleo Solitário/fisiologia , Proteínas de Anfíbios/farmacologia , Animais , Channelrhodopsins/antagonistas & inibidores , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Frequência Cardíaca , Ácido Cinurênico/farmacologia , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Optogenética , Núcleo Hipotalâmico Paraventricular/citologia , Hormônios Peptídicos/farmacologia , Núcleo Solitário/citologia
19.
Biol Reprod ; 101(1): 4-25, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30848786

RESUMO

Gasotransmitters are endogenous small gaseous messengers exemplified by nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S or sulfide). Gasotransmitters are implicated in myriad physiologic functions including many aspects of reproduction. Our objective was to comprehensively review basic mechanisms and functions of gasotransmitters during pregnancy from conception to uterine involution and highlight future research opportunities. We searched PubMed and Web of Science databases using combinations of keywords nitric oxide, carbon monoxide, sulfide, placenta, uterus, labor, and pregnancy. We included English language publications on human and animal studies from any date through August 2018 and retained basic and translational articles with relevant original findings. All gasotransmitters activate cGMP signaling. NO and sulfide also covalently modify target protein cysteines. Protein kinases and ion channels transduce gasotransmitter signals, and co-expressed gasotransmitters can be synergistic or antagonistic depending on cell type. Gasotransmitters influence tubal transit, placentation, cervical remodeling, and myometrial contractility. NO, CO, and sulfide dilate resistance vessels, suppress inflammation, and relax myometrium to promote uterine quiescence and normal placentation. Cervical remodeling and rupture of fetal membranes coincide with enhanced oxidation and altered gasotransmitter metabolism. Mechanisms mediating cellular and organismal changes in pregnancy due to gasotransmitters are largely unknown. Altered gasotransmitter signaling has been reported for preeclampsia, intrauterine growth restriction, premature rupture of membranes, and preterm labor. However, in most cases specific molecular changes are not yet characterized. Nonclassical signaling pathways and the crosstalk among gasotransmitters are emerging investigation topics.


Assuntos
Fertilização/fisiologia , Gasotransmissores/fisiologia , Parto/fisiologia , Animais , Monóxido de Carbono , Colo do Útero/fisiologia , Feminino , Humanos , Sulfeto de Hidrogênio , Miométrio/fisiologia , Óxido Nítrico , Circulação Placentária/fisiologia , Placentação/fisiologia , Gravidez , Transdução de Sinais/fisiologia , Útero/fisiologia
20.
Cell Tissue Res ; 375(1): 85-91, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29911261

RESUMO

Oxytocin (OT) has drawn the attention of researchers since 1930. Since then, many aspects of oxytocin have been uncovered, such as reproductive functions, dampening anxiety, enhancing socioemotional behavior, or regulating genomic effects on a cellular level. Here, we want to focus on the interaction between the OT system and the stress/corticotropin-releasing factor (CRF)-system of the brain. Depending on the nature of the stressor, OT is released simultaneously or directly after the stress from the neurohypophysis into the periphery and/or via somato-dendritic release in stress-sensitive brain areas. This stress-induced OT release might serve to modulate or dampen the stress response; however, the functional relevance is not yet fully understood. In this review, we will describe the effects of OT and discuss the interplay between OT and CRF on a cellular, physiological, and behavioral level.


Assuntos
Ansiedade/fisiopatologia , Comportamento/fisiologia , Hormônio Liberador da Corticotropina/metabolismo , Ocitocina/metabolismo , Estresse Psicológico/fisiopatologia , Hormônio Adrenocorticotrópico/metabolismo , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA