Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
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.
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
3.
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
4.
J Exp Biol ; 220(Pt 20): 3742-3750, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28819054

RESUMO

Seasonal reproductive cycles of most birds are regulated by photoperiod via neuroendocrine control. Gonadotropin-inhibitory hormone (GnIH) in the hypothalamus has been reported to act as neuroendocrine integrator of photoperiodic cues. In this study, both captive and field investigations were carried out to understand the effects of photoperiod and seasonality on GnIH expression in subtropical tree sparrows. Monthly observations of GnIH mRNA and peptide expression in wild birds over a year revealed a significant increase in GnIH mRNA level and number of GnIH-ir neurons during the non-breeding season when compared with their expression in the breeding season. GnIH-ir neurons were found primarily in the paraventricular nucleus (PVN) with their fibers projecting into the median eminence and some other areas of the brain. In an 8 month-long experiment, birds exposed to short days had higher GnIH expression compared with birds exposed to long days regardless of sampling month. Long-day birds with regressed testes had similar GnIH levels to short-day birds. Though the number of GnIH peptide-expressing neurons ran almost parallel to the levels of GnIH mRNA, they were inversely related to gonadal size in both sexes under natural and artificial photoperiodic conditions. These results clearly indicate an inhibitory role of GnIH in photoperiodic regulation of seasonal reproduction in the tree sparrow.


Assuntos
Proteínas Aviárias/genética , Hormônios Hipotalâmicos/genética , Fotoperíodo , Reprodução , Pardais/fisiologia , Animais , Proteínas Aviárias/metabolismo , Feminino , Hormônios Hipotalâmicos/metabolismo , Masculino , Estações do Ano
5.
Cell Rep ; 43(7): 114380, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38935503

RESUMO

Circadian rhythms are internal biological rhythms driving temporal tissue-specific, metabolic programs. Loss of the circadian transcription factor BMAL1 in the paraventricular nucleus (PVN) of the hypothalamus reveals its importance in metabolic rhythms, but its functions in individual PVN cells are poorly understood. Here, loss of BMAL1 in the PVN results in arrhythmicity of processes controlling energy balance and alters peripheral diurnal gene expression. BMAL1 chromatin immunoprecipitation sequencing (ChIP-seq) and single-nucleus RNA sequencing (snRNA-seq) reveal its temporal regulation of target genes, including oxytocin (OXT), and restoring circulating OXT peaks in BMAL1-PVN knockout (KO) mice rescues absent activity rhythms. While glutamatergic neurons undergo day/night changes in expression of genes involved in cell morphogenesis, astrocytes and oligodendrocytes show gene expression changes in cytoskeletal organization and oxidative phosphorylation. Collectively, our findings show diurnal gene regulation in neuronal and non-neuronal PVN cells and that BMAL1 contributes to diurnal OXT secretion, which is important for systemic diurnal rhythms.


Assuntos
Fatores de Transcrição ARNTL , Relógios Circadianos , Ritmo Circadiano , Camundongos Knockout , Neurônios , Núcleo Hipotalâmico Paraventricular , Animais , Fatores de Transcrição ARNTL/metabolismo , Fatores de Transcrição ARNTL/genética , Núcleo Hipotalâmico Paraventricular/metabolismo , Relógios Circadianos/genética , Camundongos , Neurônios/metabolismo , Ritmo Circadiano/fisiologia , Ocitocina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Regulação da Expressão Gênica , Astrócitos/metabolismo , Oligodendroglia/metabolismo
6.
Neurosci Lett ; 791: 136920, 2022 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-36272558

RESUMO

Follicular development and ovulation are profoundly suppressed during lactation. This suppression is suggested to be due to the suckling-induced inhibition of the kisspeptin gene (the master regulator of reproduction) in the arcuate nucleus (ARC) and subsequent inhibition of pulsatile gonadotropin-releasing hormone (GnRH)/gonadotropin release. The present study examined whether hypothalamic κ-opioid receptor (KOR) or µ-opioid receptor (MOR) signaling mediates the suppression of luteinizing hormone (LH) release induced by suckling stimulus during late lactation in rats. Central administration of a selective KOR antagonist blocked the suppression of LH release on Day 16 of lactation; however, central administration of a selective MOR antagonist failed to block the suppression. The suckling stimulus significantly increased the number of fos (a marker for neural activation)-positive Pdyn (dynorphin gene)-expressing cells in the paraventricular nucleus (PVN) and supraoptic nucleus (SON) but not in the ARC. Taken together, these results suggest that central KOR signaling, but not MOR signaling, at least partly, mediates the suppression of LH release induced by suckling stimulus during late lactation, and PVN and SON Dyn neurons may be involved in the suppression in rats.


Assuntos
Dinorfinas , Receptores Opioides kappa , Feminino , Ratos , Animais , Dinorfinas/metabolismo , Receptores Opioides kappa/metabolismo , Hormônio Luteinizante , Núcleo Arqueado do Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Lactação/fisiologia , Receptores Opioides
7.
Cardiovasc Toxicol ; 21(4): 286-300, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33165770

RESUMO

Hypertension, as one of the major risk factors for cardiovascular disease, significantly affects human health. Prostaglandin E2 (PGE2) and the E3-class prostanoid (EP3) receptor have previously been demonstrated to modulate blood pressure and hemodynamics in various animal models of hypertension. The PGE2-evoked pressor and biochemical responses can be blocked with the EP3 receptor antagonist, L-798106 (N-[(5-bromo-2methoxyphenyl)sulfonyl]-3-[2-(2-naphthalenylmethyl) phenyl]-2-propenamide). In the hypothalamic paraventricular nucleus (PVN), sympathetic excitation can be introduced by PGE2, which can activate EP3 receptors located in the PVN. In such a case, the central knockdown of EP3 receptor can be considered as a potential therapeutic modality for hypertension management. The present study examined the efficacy of the PVN infusion of L-798106, by performing experiments on spontaneously hypertensive rats (SHRs) and normotensive Wistar-Kyoto rats (WKYs). The rats were administered with chronic bilateral PVN infusion of L-798106 (10 µg/day) or the vehicle for 28 days. The results indicated that the SHRs had a higher mean arterial pressure (MAP), an increased Fra-like (Fra-LI) activity in the PVN, as well as a higher expression of gp91phox, mitogen-activated protein kinase (MAPK), and proinflammatory cytokines in the PVN compared with the WKYs. Additionally, the expression of Cu/Zn-SOD in the PVN of the SHRs was reduced compared with the WKYs. The bilateral PVN infusion of L-798106 significantly reduced MAP, as well as plasma norepinephrine (NE) levels in the SHRs. It also inhibited Fra-LI activity and reduced the expression of gp91phox, proinflammatory cytokines, and MAPK, whereas it increased the expression of Cu/Zn-SOD in the PVN of SHRs. In addition, L-798106 restored the balance of the neurotransmitters in the PVN. On the whole, the findings of the present study demonstrate that the PVN blockade of EP3 receptor can ameliorate hypertension and cardiac hypertrophy partially by attenuating ROS and proinflammatory cytokines, and modulating neurotransmitters in the PVN.


Assuntos
Anti-Hipertensivos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Hipertensão/prevenção & controle , Mediadores da Inflamação/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Antagonistas de Prostaglandina/farmacologia , Receptores de Prostaglandina E Subtipo EP3/antagonistas & inibidores , Sulfonamidas/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Cardiomegalia/prevenção & controle , Modelos Animais de Doenças , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Masculino , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Espécies Reativas de Oxigênio/metabolismo , Receptores de Prostaglandina E Subtipo EP3/metabolismo , Transdução de Sinais
8.
Front Pharmacol ; 12: 806012, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35095514

RESUMO

Background: Hydrogen sulfide (H2S) is a new type of gas neurotransmitter discovered in recent years. It plays an important role in various physiological activities. The hypothalamus paraventricular nucleus (PVN) is an important nucleus that regulates gastric function. This study aimed to clarify the role of H2S in the paraventricular nucleus of the hypothalamus on the gastric function of rats. Methods: An immunofluorescence histochemistry double-labelling technique was used to determine whether cystathionine-beta-synthase (CBS) and c-Fos neurons are involved in PVN stress. Through microinjection of different concentrations of NaHS, physiological saline (PS), D-2-Amino-5-phosphonovaleric acid (D-AP5), and pyrrolidine dithiocarbamate (PDTC), we observed gastric motility and gastric acid secretion. Results: c-Fos and CBS co-expressed the most positive neurons after 1 h of restraint and immersion, followed by 3 h, and the least was at 0 h. After injection of different concentrations of NaHS into the PVN, gastric motility and gastric acid secretion in rats were significantly inhibited and promoted, respectively (p < 0.01); however, injection of normal saline, D-AP5, and PDTC did not cause any significant change (p > 0.05). The suppressive effect of NaHS on gastrointestinal motility and the promotional effect of NaHS on gastric acid secretion could be prevented by D-AP5, a specific N-methyl-D-aspartic acid (NMDA) receptor antagonist, and PDTC, an NF-κB inhibitor. Conclusion: There are neurons co-expressing CBS and c-Fos in the PVN, and the injection of NaHS into the PVN can inhibit gastric motility and promote gastric acid secretion in rats. This effect may be mediated by NMDA receptors and the NF-κB signalling pathway.

9.
Cardiovasc Toxicol ; 21(6): 472-489, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33582931

RESUMO

Oxidative stress in the hypothalamic paraventricular nucleus (PVN) contributes greatly to the development of hypertension. The recombinant nuclear respiratory factor 1 (Nrf1) regulates the transcription of several genes related to mitochondrial respiratory chain function or antioxidant expression, and thus may be involved in the pathogenesis of hypertension. Here we show that in the two-kidney, one-clip (2K1C) hypertensive rats the transcription level of Nrf1 was elevated comparing to the normotensive controls. Knocking down of Nrf1 in the PVN of 2K1C rats can significantly reduce their blood pressure and level of plasma norepinephrine (NE). Analysis revealed significant reduction of superoxide production level in both whole cell and mitochondria, along with up-regulation of superoxide dismutase 1 (Cu/Zn-SOD), NAD(P)H: quinone oxidoreductase 1 (NQO1), thioredoxin-dependent peroxiredoxin 3 (Prdx3), cytochrome c (Cyt-c) and glutathione synthesis rate-limiting enzyme (glutamyl-cysteine ligase catalytic subunit (Gclc) and modifier subunit (Gclm)), and down-regulation of cytochrome c oxidase subunit VI c (Cox6c) transcription after Nrf1 knock-down. In addition, the reduced ATP production and elevated mitochondrial membrane potential in the PVN of 2K1C rats were reinstated with Nrf1 knock-down, together with restored expression of peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α), mitochondrial transcription factor A (Tfam), coiled-coil myosin-like BCL2-interacting protein (Beclin1), and Mitofusin 1 (Mfn1), which are related to the mitochondrial biogenesis, fusion, and autophagy. Together, the results indicate that the PVN Nrf1 is associated with the development of 2K1C-induced hypertension, and Nrf1 knock-down in the PVN can alleviate hypertension through intervention of mitochondrial function and restorement of the production-removal balance of superoxide.


Assuntos
Pressão Sanguínea , Hipertensão Renovascular/metabolismo , Mitocôndrias/metabolismo , Fator 1 Nuclear Respiratório/metabolismo , Estresse Oxidativo , Núcleo Hipotalâmico Paraventricular/metabolismo , Superóxidos/metabolismo , Animais , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Hipertensão Renovascular/genética , Hipertensão Renovascular/fisiopatologia , Hipertensão Renovascular/prevenção & controle , Masculino , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Fator 1 Nuclear Respiratório/genética , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Interferência de RNA , Ratos Sprague-Dawley
10.
Endocrinology ; 161(1)2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31754709

RESUMO

To limit excessive glucocorticoid secretion following hypothalamic-pituitary-adrenal (HPA) axis stimulation, circulating glucocorticoids inhibit corticotropin-releasing hormone (CRH) expression in paraventricular nucleus (PVN) neurons. As HPA function differs between sexes and depends on circulating estradiol (E2) levels in females, we investigated sex/estrous stage-dependent glucocorticoid regulation of PVN Crh. Using NanoString nCounter technology, we first demonstrated that adrenalectomized (ADX'd) diestrous female (low E2), but not male or proestrous female (high E2), mice exhibited a robust decrease in PVN CRH mRNA following 2-day treatment with the glucocorticoid receptor (GR) agonist RU28362. Immunohistochemical analysis of PVN CRH neurons in Crh-IRES-Cre;Ai14 mice, where TdTomato fluorescence permanently tags CRH-expressing neurons, showed similarly abundant co-expression of GR-immunoreactivity in males, diestrous females, and proestrous females. However, we identified sex/estrous stage-related glucocorticoid regulation or expression of GR transcriptional coregulators. Out of 17 coregulator genes examined using nCounter multiplex analysis, mRNAs that were decreased by RU28362 in ADX'd mice in a sex/estrous stage-dependent fashion included: GR (males = diestrous females > proestrous females), signal transducer and activator of transcription 3 (STAT3) (males < diestrous = proestrous), and HDAC1 (males < diestrous > proestrous). Steroid receptor coactivator 3 (SRC-3), nuclear corepressor 1 (NCoR1), heterogeneous nuclear ribonucleoprotein U (hnrnpu), CREB binding protein (CBP) and CREB-regulated transcription coactivator 2 (CRTC2) mRNAs were lower in ADX'd diestrous and proestrous females versus males. Additionally, most PVN CRH neurons co-expressed methylated CpG binding protein 2 (MeCP2)-immunoreactivity in diestrous female and male Crh-IRES-Cre;Ai14 mice. Our findings collectively suggest that GR's sex-dependent regulation of PVN Crh may depend upon differences in the GR transcriptional machinery and an underlying influence of E2 levels in females.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Estradiol/sangue , Glucocorticoides/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Adrenalectomia , Androstanóis/farmacologia , Animais , Hormônio Liberador da Corticotropina/genética , Ciclo Estral , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/agonistas , Hipotálamo/citologia , Masculino , Camundongos , Camundongos Endogâmicos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , RNA Mensageiro , Fatores Sexuais , Vagina/citologia
11.
PeerJ ; 8: e8528, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32175184

RESUMO

BACKGROUND: High dietary salt intake is strongly correlated with cardiovascular (CV) diseases and it is regarded as a major risk factor associated with the pathogenesis of hypertension. The CV control centres in the brainstem (the nucleus tractus solitarii (NTS) and the rostral ventrolateral medulla (RVLM)) and hypothalamic forebrain (the subfornical organ, SFO; the supraoptic nucleus, SON and the paraventricular nucleus, PVN) have critical roles in regulating CV autonomic motor outflows, and thus maintaining blood pressure (BP). Growing evidence has implicated autonomic regulatory networks in salt-sensitive HPN (SSH), but the genetic basis remains to be delineated. We hypothesized that the development and/ or maintenance of SSH is reliant on the change in the expression of genes in brain regions controlling the CV system. METHODOLOGY: We used RNA-Sequencing (RNA-Seq) to describe the differential expression of genes in SFO, SON, PVN, NTS and RVLM of rats being chronically fed with high-salt (HS) diet. Subsequently, a selection of putatively regulated genes was validated with quantitative reverse transcription polymerase chain reaction (qRT-PCR) in both Spontaneously Hypertensive rats (SHRs) and Wistar Kyoto (WKY) rats. RESULTS: The findings enabled us to identify number of differentially expressed genes in SFO, SON, PVN, NTS and RVLM; that are either up-regulated in both strains of rats (SON- Caprin2, Sctr), down-regulated in both strains of rats (PVN- Orc, Gkap1), up-regulated only in SHRs (SFO- Apopt1, Lin52, AVP, OXT; SON- AVP, OXT; PVN- Caprin2, Sclt; RVLM- A4galt, Slc29a4, Cmc1) or down-regulated only in SHRs (SON- Ndufaf2, Kcnv1; PVN- Pi4k2a; NTS- Snrpd2l, Ankrd29, St6galnac6, Rnf157, Iglon5, Csrnp3, Rprd1a; RVLM- Ttr, Faim). CONCLUSIONS: These findings demonstrated the adverse effects of HS diet on BP, which may be mediated via modulating the signaling systems in CV centers in the hypothalamic forebrain and brainstem.

12.
Brain Struct Funct ; 225(5): 1459-1482, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32394093

RESUMO

Human obesity epidemic is increasing worldwide with major adverse consequences on health. Among other possible causes, the hypothesis of an infectious contribution is worth it to be considered. Here, we report on an animal model of virus-induced obesity which might help to better understand underlying processes in human obesity. Eighty Wistar rats, between 30 and 60 days of age, were intracerebrally inoculated with Borna disease virus (BDV-1), a neurotropic negative-strand RNA virus infecting an unusually broad host spectrum including humans. Half of the rats developed fatal encephalitis, while the other half, after 3-4 months, continuously gained weight. At tripled weights, rats were sacrificed by trans-cardial fixative perfusion. Neuropathology revealed prevailing inflammatory infiltrates in the median eminence (ME), progressive degeneration of neurons of the paraventricular nucleus, the entorhinal cortex and the amygdala, and a strikingly high-grade involution of the hippocampus with hydrocephalus. Immune histology revealed that major BDV-1 antigens were preferentially present at glutamatergic receptor sites, while GABAergic areas remained free from BDV-1. Virus-induced suppression of the glutamatergic system caused GABAergic predominance. In the hypothalamus, this shifted the energy balance to the anabolic appetite-stimulating side governed by GABA, allowing for excessive fat accumulation in obese rats. Furthermore, inflammatory infiltrates in the ME and ventro-medial arcuate nucleus hindered free access of appetite-suppressing hormones leptin and insulin. The hormone transport system in hypothalamic areas outside the ME became blocked by excessively produced leptin, leading to leptin resistance. The resulting hyperleptinemic milieu combined with suppressed glutamatergic mechanisms was a characteristic feature of the found metabolic pathology. In conclusion, the study provided clear evidence that BDV-1 induced obesity in the rat model is the result of interdependent structural and functional metabolic changes. They can be explained by an immunologically induced hypothalamic microcirculation-defect, combined with a disturbance of neurotransmitter regulatory systems. The proposed mechanism may also have implications for human health. BDV-1 infection has been frequently found in depressive patients. Independently, comorbidity between depression and obesity has been reported, either. Future studies should address the exciting question of whether BDV-1 infection could be a link, whatsoever, between these two conditions.


Assuntos
Doença de Borna/complicações , Vírus da Doença de Borna/fisiologia , Encefalite Viral/patologia , Hipotálamo/patologia , Hipotálamo/virologia , Neuropeptídeos/metabolismo , Obesidade/virologia , Animais , Doença de Borna/metabolismo , Doença de Borna/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/virologia , Hipotálamo/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Neurônios/virologia , Obesidade/metabolismo , Obesidade/patologia , Ratos Wistar
13.
Cardiovasc Toxicol ; 19(5): 451-464, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31037602

RESUMO

Carbon monoxide (CO) presents anti-inflammatory and antioxidant activities as a new gaseous neuromessenger produced by heme oxygenase-1 (HO-1) in the body. High salt-induced hypertension is relevant to the levels of pro-inflammatory cytokines (PICs) and oxidative stress in the hypothalamic paraventricular nucleus (PVN). We explored whether CO in PVN can attenuate high salt-induced hypertension by regulating PICs or oxidative stress. Male Dahl Salt-Sensitive rats were fed high-salt (8% NaCl) or normal-salt (0.3% NaCl) diet for 4 weeks. CORM-2, ZnPP IX, or vehicle was microinjected into bilateral PVN for 6 weeks. High-salt diet increased the levels of MAP, plasma norepinephrine (NE), reactive oxygen species (ROS), and the expressions of COX2, IL-1ß, IL-6, NOX2, and NOX4 significantly in PVN (p < 0.05), but decreased the expressions of HO-1 and Cu/Zn-SOD in PVN (p < 0.05). Salt increased sympathetic activity as measured by circulating norepinephrine, and increased the ratio of basal RSNA to max RSNA, in part by decreasing max RSNA. PVN microinjection of CORM-2 decreased the levels of MAP, NE, RSNA, ROS and the expressions of COX2, IL-1ß, IL-6, NOX2, NOX4 significantly in PVN of hypertensive rat (p < 0.05), but increased the expressions of HO-1 and Cu/Zn-SOD significantly (p < 0.05), which were all opposite to the effects of ZnPP IX microinjected in PVN (p < 0.05). We concluded that exogenous or endogenous CO attenuates high salt-induced hypertension by regulating PICs and oxidative stress in PVN.


Assuntos
Anti-Inflamatórios/farmacologia , Anti-Hipertensivos/farmacologia , Antioxidantes/farmacologia , Pressão Arterial/efeitos dos fármacos , Monóxido de Carbono/farmacologia , Citocinas/metabolismo , Hipertensão/prevenção & controle , Mediadores da Inflamação/metabolismo , Compostos Organometálicos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Animais , Anti-Inflamatórios/metabolismo , Anti-Hipertensivos/metabolismo , Antioxidantes/metabolismo , Monóxido de Carbono/metabolismo , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Heme Oxigenase (Desciclizante)/metabolismo , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Masculino , Compostos Organometálicos/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Ratos Endogâmicos Dahl , Cloreto de Sódio na Dieta
14.
Basic Clin Neurosci ; 9(4): 269-274, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30519385

RESUMO

INTRODUCTION: Carbon Dioxide (CO2) and diethyl ether are used as light anesthetics. However, experimental data about their side effects are scarce. In addition, in all our previous works on regulatory mechanisms of hypothalamus during food intake, including the effect of Paraventricular Nucleus (PVN) D1 and D2 dopamine receptors and glucosensitive neurons, the drug injections were performed under brief diethyl ether anesthesia. In the current study, we tested the hypothesis which postulates that CO2 and diethyl ether as light anesthetic agents affect the stimulatory effect of PVN dopamine receptors and glucosensitive neurons in feeding behavior. METHODS: Male Wistar rats were implanted with guide cannula directed to their PVN. Glucose (0.8 µg), SKF38393 (D1 agonist, 0.5 µg), quinpirole (D2 agonist, 0.3 µg) and saline (0.3 µL) were microinjected into the PVN and food intake was measured over 1 hour. RESULTS: Our results showed that CO2 but not diethyl ether decreased food intake compared to intact animals. The PVN injections of glucose, SKF38393, and quinpirole increased food intake under brief diethyl ether anesthesia. In contrast, the PVN microinjected glucose-induced and dopamine receptor agonists-induced food intake were inhibited under light CO2 anesthesia. CONCLUSION: Our results suggest that brief exposure to CO2 and diethyl ether as light anesthetic agents may affect PVN glucosensing neurons-induced and dopamine receptors-induced food intake in fasted rats.

15.
Neurosci Lett ; 638: 5-11, 2017 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-27923665

RESUMO

We recently found that extracellular administration of nicotine indirectly excited hypothalamic paraventricular nucleus (PVN) corticotropin-releasing hormone (CRH) mRNA-expressing neurons. In this study, we studied the effect of nicotine on PVN oxytocin (OT) mRNA-expressing neuron in vitro in rats, by whole-cell patch-clamp recording technique, immunohistochemistry methods and single-cell reverse-transcription multiplex polymerase chain reaction (SC-RT-mPCR) methods Our results showed that 79.3% (73/92) of the 92 PVN putative magnocellular neurons co-expressed GAPDH mRNA and OT mRNA. Under current-clamp recording conditions, local micro application of nicotine (1-300µM) induced a decrease in spontaneous firing rate accompanied with a hyperpolarization of membrane potential in 76.7% (56/73) of PVN OT mRNA-expressing magnocellular neurons. The nicotine induced inhibition in spontaneous activity of PVN OT mRNA-expressing magnocellular neurons was dose-dependent. The half-inhibitory concentration (IC50) is 2.9µM. The nicotine induced hyperpolarization of PVN OT mRNA-expressing magnocellular neurons was sensitive to GABAA receptor antagonist, SR95531 (10µM) and tetrodotoxin (TTX, 1µM). In addition, local micro application of nicotine induced a significant increase in frequency of spontaneous inhibitory postsynaptic potentials (sIPSPs), but without changes in the sEPSPs amplitude of the OT-mRNA expressing neurons. Biocytin staining confirmed that the nicotine-sensitive OT-mRNA expressing neurons were the PVN magnocellular neurons. These results demonstrated that nicotine enhances the GABAergic inhibition, resulting in a decrease in spontaneous firing rate of the PVN OT-mRNA expressing neurons. These findings suggested that nicotine modulated PVN OT secretion via enhancement of both presynaptic action potential drive and quantal GABA release.


Assuntos
Neurônios/efeitos dos fármacos , Nicotina/farmacologia , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ácido gama-Aminobutírico/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Antagonistas de Receptores de GABA-A/farmacologia , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Masculino , Neurônios/metabolismo , Ocitocina/genética , Núcleo Hipotalâmico Paraventricular/metabolismo , Piridazinas/farmacologia , Ratos Wistar , Tetrodotoxina/farmacologia
16.
Psychoneuroendocrinology ; 71: 73-85, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27243477

RESUMO

Neuropeptide FF (NPFF) is a morphine-modulating peptide that regulates the analgesic effect of opioids, and also controls food consumption and cardiovascular function through its interaction with two cognate receptors, NPFFR1 and NPFFR2. In the present study, we explore a novel modulatory role for NPFF-NPFFR2 in stress-related depressive behaviors. In a mouse model of chronic mild stress (CMS)-induced depression, the expression of NPFF significantly increased in the hypothalamus, hippocampus, medial prefrontal cortex (mPFC) and amygdala. In addition, transgenic (Tg) mice over-expressing NPFFR2 displayed clear depression and anxiety-like behaviors with hyperactivity in the hypothalamic-pituitary-adrenal (HPA) axis, reduced expression of glucocorticoid receptor (GR) and neurogenesis in the hippocampus. Furthermore, acute treatment of NPFFR2 agonists in wild-type (WT) mice enhanced the activity of the HPA axis, and chronic administration resulted in depressive and anxiety-like behaviors. Chronic stimulation of NPFFR2 also decreased the expression of hippocampal GR and led to persistent activation of the HPA axis. Strikingly, bilateral intra-paraventricular nucleus (PVN) injection of NPFFR2 shRNA predominately inhibits the depressive-like behavior in CMS-exposed mice. Antidepressants, fluoxetine and ketamine, effectively relieved the depressive behaviors of NPFFR2-Tg mice. We speculate that persistent NPFFR2 activation, in particular in the hypothalamus, up-regulates the HPA axis and results in long-lasting increases in circulating corticosterone (CORT), consequently damaging hippocampal function. This novel role of NPFFR2 in regulating the HPA axis and hippocampal function provides a new avenue for combating depression and anxiety-like disorder.


Assuntos
Depressão/fisiopatologia , Receptores de Neuropeptídeos/metabolismo , Animais , Antidepressivos/farmacologia , Ansiedade , Transtornos de Ansiedade/metabolismo , Doença Crônica/psicologia , Corticosterona/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Transtorno Depressivo/fisiopatologia , Expressão Gênica , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiopatologia , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Animais , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/metabolismo , Sistema Hipófise-Suprarrenal/fisiopatologia , Córtex Pré-Frontal/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Neuropeptídeos/fisiologia , Estresse Fisiológico/efeitos dos fármacos , Estresse Psicológico/metabolismo
17.
Biomed Mater Eng ; 26 Suppl 1: S487-95, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26406040

RESUMO

Previous literatures have indicated that hypothalamic paraventricular nucleus (PVN) neurons are important for regulating the level of sympathetic and vagal nervous activity. Sympathovagal balance is closely related to heart rate variability (HRV). However, it still requires further elucidation regarding the effect of PVN on HRV by regulating sympathovagal balance. To detect the influence of the PVN on HRV, we evaluated the changes in time domain (including standard deviation of the R-R intervals (SDNN), and the root mean square of successive differences (RMSSD)) and frequency domain (including low frequency (LFnu), high frequency (HFnu) and the ratio of LF/HF) in HRV upon appropriate electronic stimulation, and lesions on the PVN of the rat in vivo. Cardiac vagal modulation was evaluated by HFnu; sympathetic modulation was evaluated by LFnu. Sympathovagal balance was evaluated by LF/HF and SDNN. Upon electronic stimulating (less than 0.6 mA) to the PVN of rats, we found that LFnu and HFnu changed correspondingly but recovered after the stimulation. When the PVN of the rats was injured, the RR intervals were enhanced with the rats' unilaterally or bilaterally injured PVN, especially the bilateral lesion. Meanwhile, LFnu, LF/HF and SDNN decreased gradually, accompanied with an increase of HFnu levels. So these PVN changes may indicate alterations of the sympathovagal balance.


Assuntos
Adaptação Fisiológica/fisiologia , Frequência Cardíaca/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Sistema Nervoso Simpático/fisiologia , Nervo Vago/fisiologia , Animais , Simulação por Computador , Masculino , Modelos Biológicos , Modelos Estatísticos , Ratos , Ratos Endogâmicos F344
18.
Front Neuroanat ; 9: 130, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26500509

RESUMO

Conventional neuroanatomical, immunohistochemical techniques, and electrophysiological recording, as well as in vitro labeling methods may fail to detect long range extra-neurohypophyseal-projecting axons from vasopressin (AVP)-containing magnocellular neurons (magnocells) in the hypothalamic paraventricular nucleus (PVN). Here, we used in vivo extracellular recording, juxtacellular labeling, post-hoc anatomo-immunohistochemical analysis and camera lucida reconstruction to address this question. We demonstrate that all well-labeled AVP immunopositive neurons inside the PVN possess main axons joining the tract of Greving and multi-axon-like processes, as well as axonal collaterals branching very near to the somata, which project to extra-neurohypophyseal regions. The detected regions in this study include the medial and lateral preoptical area, suprachiasmatic nucleus (SCN), lateral habenula (LHb), medial and central amygdala and the conducting systems, such as stria medullaris, the fornix and the internal capsule. Expression of vesicular glutamate transporter 2 was observed in axon-collaterals. These results, in congruency with several previous reports in the literature, provided unequivocal evidence that AVP magnocells have an uncommon feature of possessing multiple axon-like processes emanating from somata or proximal dendrites. Furthermore, the long-range non-neurohypophyseal projections are more common than an "occasional" phenomenon as previously thought.

19.
Brain Res ; 1560: 46-59, 2014 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-24642272

RESUMO

Based on the overlapping physiological roles of cocaine- and amphetamine-regulated transcript (CART) peptides and serotonin, the present study examined the anatomical connection between the hypothalamic paraventricular nucleus (PVN) and the dorsal raphe (DR). The first series of experiments were performed to investigate descending projections from the CART-immunoreactive (CART-ir) PVN to serotonergic DR cells. CART-ir varicosities made contact with serotonergic DR neurons. An anterograde tracing study revealed that varicosities originating from the PVN formed close appositions to serotonergic neuronal profiles along the entire rostro-caudal extent of the DR. A retrograde study demonstrated that CART neurons projecting to the DR were mainly localized in the caudal parvicellular PVN, comprising approximately 3.0%±0.4% (n=8) of total CART cells. A second series of experiments was performed to investigate ascending projections from the DR to CART-ir PVN cells. Serotonin transporter-ir boutons made contact with CART-ir PVN neurons. Anterograde tracing revealed that varicosities originating from the DR formed close appositions to CART-ir PVN cells. Retrograde examination demonstrated that serotonergic neurons projecting to the parvicellular PVN were located along the entire rostro-caudal extent of the DR. The present observation provided an anatomical basis for accumulating evidence in the literature that suggests a functional interaction between the CART and serotonin systems during the regulation of energy balance, emotional behavior, and arousal.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Núcleo Hipotalâmico Paraventricular/anatomia & histologia , Núcleos da Rafe/anatomia & histologia , Neurônios Serotoninérgicos/citologia , Animais , Axônios/metabolismo , Feminino , Imunofluorescência , Masculino , Microscopia Confocal , Vias Neurais/anatomia & histologia , Vias Neurais/metabolismo , Técnicas de Rastreamento Neuroanatômico , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Fotomicrografia , Terminações Pré-Sinápticas/metabolismo , Núcleos da Rafe/metabolismo , Ratos Sprague-Dawley , Neurônios Serotoninérgicos/metabolismo
20.
Laser Ther ; 23(1): 31-40, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24771969

RESUMO

BACKGROUND: In our activity in the Ambulance Service of Bucharest Municipality during March 2002 - March 2003 we studied a casuistry of patients who had fallen in a coma of varying degrees. To aid better understanding of coma, the concept of "pre-coma stage" or diencephalic "0 stage" was introduced. This concept complements the Arseni classification already used in medical practice, because some doctors alternatively use the term of "inaugural coma" for the same condition that we call diencephalic "0 stage". In the median hypothalamus and on the retino-hypothalamic path (SCN - AN) optical waves are transmitted, probably in the near infrared spectral range (800-1000 nm). These waves would constitute a means of transmitting information about the infradian biorhythm of coordination (frequencies below 1 cycle/28 hours), essential for the modulation and pre-processing of the consciousness and wakefulness, a fact which has already been demonstrated in animals. METHODS: The current work is based on observations made on a group of 51 patients with the precoma and coma conditions, and on a thorough study of the specialized (especially Romanian) literature. Also, we used validated scientific proof of torture in conditions of lack of light. RESULTS: We found a perfect interpenetration between the ARAS and the following two complementary subsystems: 1. The hypothalamic suprachiasmatic nucleus, that has the role of coordinating the periodicity of some biological functions depending on the circadian rhythm; and 2. The main photoperiodic organ, the pineal gland (epiphysis), which together with the ARAS has a role in the photoperiodicity modulation of some biological functions during the state of vigilance. CONCLUSION: From the above mentioned one may conclude that the consciousness condition is the unitary result of action of all human brain systems and especially sub-systems, which are controlled and led by a psycho-neurological process of integration at the cortex level. "Everyone knows what the consciousness is, until one tries to define it" (William James: The Stream of Consciousness", 1892).

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa