Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neuroendocrinol ; 29(12)2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29063642

RESUMO

Respiratory manifestations of panic disorder (PD) include a greater respiratory instability and enhanced responsiveness to CO2 compared to normal individuals. Although the prevalence of PD is approximately three times greater in women compared to men, the origins of this sexual dimorphism remain poorly understood. Similar to PD patients, adult female rats previously subjected to neonatal maternal separation (NMS) show an increase in their ventilatory response to CO2 . Because this effect of NMS is not observed in males, we hypothesised that testosterone prevents NMS-induced hyper-responsiveness to CO2 . Pups subjected to NMS were placed in an incubator for 3 h d-1 from postnatal days 3-12. Control pups remained undisturbed. At adulthood (8-10 weeks of age), rats were then subjected either to sham surgery or castration. Fourteen days later, breathing was measured at rest (room air) and during acute exposure to hypercapnia (5 and 10% CO2 for 10 minutes each) using plethysmography. To gain insight into the mechanisms involved, c-fos expression was used as an indicator of neuronal activation. Brains were collected following air or CO2 exposure for quantification of c-fos positive cells by immunohistochemistry in selected regions, including the paraventricular nucleus of the hypothalamus, the dorsomedial hypothalamus and the amygdalar complex. Castration produced a 100% increase of hyperventilatory response to 10% CO2 in control rats. Unexpectedly, castration had no effect on the hyperventilatory response of NMS rats. The intensity of the hypercapnic response was inversely correlated with c-fos expression in the medial amygdala. We conclude that testosterone prevents the hyper-responsiveness to CO2 , whereas NMS attenuates sensitivity to hormone withdrawal. We propose that an inhibitory influence from the medial amygdala contributes to this effect.


Assuntos
Hipercapnia/fisiopatologia , Privação Materna , Transtorno de Pânico/fisiopatologia , Respiração , Testosterona/fisiologia , Animais , Animais Recém-Nascidos , Apneia/fisiopatologia , Complexo Nuclear Corticomedial/metabolismo , Masculino , Orquiectomia , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos Sprague-Dawley
2.
Neuroscience ; 192: 398-412, 2011 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-21745545

RESUMO

In Na(+) appetite neurobiology, it is essential to investigate whether endogenous opioids modulate the output of the neural substrates that are involved in both the detection and integration of Na(+) deficiency and the motivational aspect of Na(+) intake. Thus, evaluating the recruitment dynamics of enkephalin (ENK)-containing and/or mu-opioid receptor (µ-OR)-expressing neurons in close correlation with the hydromineral state of the rat might provide useful information regarding the role of the opioid system in regulating the central network that controls water and Na(+) intake. Furosemide was used to deplete both fluid volume and the Na(+) content of the extracellular fluid (ECF) compartment when combined with water repletion and a short-term Na(+)-free diet. Na(+) restoration in the ECF compartment was achieved by providing unrestricted access to both saline (0.3 M NaCl) and water. Combining in situ hybridization (against ENK and µ-OR mRNA) and immunohistochemistry (against Fos) revealed a specific pattern of hypovolemia-induced Fos expression in the enkephalinergic subpopulations of the central amygdala, in the oval nucleus of the bed nucleus of the stria terminalis and in the nucleus accumbens shell. Hypovolemia also induced transient Fos expression in µ-OR-expressing neurons in the same nuclei and in the median preoptic nucleus and subfornical organ. However, this specific hydromineral state did not activate the ENK and/or µ-OR-expressing neurons in the lateral parabrachial nucleus or in the medial nucleus of the solitary tract. These results implicate the ENK/µ-OR system as a putative facilitator of Na(+) intake in discrete regions of the forebrain, possibly by modulating the hedonic and reward value of Na(+) by increasing ENK release in these regions.


Assuntos
Comportamento de Ingestão de Líquido/fisiologia , Encefalinas/metabolismo , Hipovolemia/metabolismo , Prosencéfalo/metabolismo , Receptores Opioides mu/metabolismo , Sódio/metabolismo , Animais , Apetite/fisiologia , Líquido Extracelular/fisiologia , Imuno-Histoquímica , Hibridização In Situ , Masculino , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar
3.
J Comp Neurol ; 508(4): 663-75, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18381633

RESUMO

The interaction between the stress axis and endogenous opioid systems has gained substantial attention, because it is increasingly recognized that stress alters individual sensitivity to opiates. One site at which opiates and stress substrates may interact to have global effects on behavior is within the locus coeruleus (LC). We have previously described interactions of several opioid peptides [e.g., proopiomelanocortin, enkephalin (ENK)] with the stress-related peptide corticotropin-releasing factor (CRF) in the LC. To examine further the interactions among dynorphin (DYN), ENK, and CRF in the LC, sections were processed for detection of DYN and CRF or DYN and ENK in rat brain. DYN- and CRF-containing axon terminals overlapped noradrenergic dendrites in this region. Dual immunoelectron microscopy showed coexistence of DYN and CRF; 35% of axon terminals containing DYN were also immunoreactive for CRF. In contrast, few axon terminals contained both DYN and ENK. A potential DYN/CRF afferent is the central nucleus of the amygdala (CeA). Dual in situ hybridization showed that, in CeA neurons, 31% of DYN mRNA-positive cells colocalized with CRF mRNA, whereas 53% of CRF mRNA-containing cells colocalized with DYN mRNA. Finally, to determine whether limbic DYN afferents target the LC, the CeA was electrolytically lesioned. Light-level densitometry of DYN labeling in the LC showed a significant decrease in immunoreactivity on the side of the lesion. Taken together, these data indicate that DYN- and CRF-labeled axon terminals, most likely arising from amygdalar sources, are positioned dually to affect LC function, whereas DYN and ENK function in parallel.


Assuntos
Tonsila do Cerebelo/química , Tonsila do Cerebelo/fisiologia , Dinorfinas/análise , Locus Cerúleo/química , Peptídeos/química , Estresse Fisiológico , Animais , Dinorfinas/fisiologia , Vias Eferentes/química , Vias Eferentes/fisiologia , Locus Cerúleo/fisiologia , Masculino , Peptídeos/fisiologia , Ratos , Ratos Sprague-Dawley , Estresse Fisiológico/metabolismo
4.
Neuroscience ; 122(1): 247-57, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14596865

RESUMO

The lamina terminalis (LT) contains three main regions, namely the subfornical organ (SFO), the median preoptic nucleus (MnPO) and the vascular organ of the LT (OVLT). Although LT is recognized of paramount importance in the regulation of hydromineral homeostasis, identity of the neurocircuits interconnecting the SFO and OVLT to the MnPO is not known. Furthermore, the phenotype of neuronal populations activated during acute hydromineral challenge is not yet determined. By using the high cellular resolution of the in situ hybridization histochemistry (ISHH), we investigated whether a furosemide-induced fluid and electrolyte depletion might modify both putative GABAergic and glutamatergic systems within the LT. We show that acute furosemide treatment (4 h) significantly reduced the expression of GAD67 mRNA, the active holoenzyme predictive of GABA synthesis, within the SFO. A strong tendency toward a reduction of GAD67 signal was also observed in the OVLT and MnPO. The hydromineral challenge did not alter the expression of GAD65 and type 2 vesicular glutamate transporter (vGlut2) mRNA in all the structures of the LT. Furosemide treatment was associated with a reduction in the population of GAD67-containing neurons in the periphery of the SFO and dorsal part of the MnPO. Contrastingly, GAD65-containing cells were shown to be increased in the OVLT and no change was observed for the vGlut2-containing neurons in the whole LT. By combining ISHH with immunohistochemistry (Fos immunoreactivity), we report that furosemide-induced water and sodium depletion did essentially recruit a glutamatergic network throughout the LT, although GABAergic neurons were specifically activated in the ring of the SFO and in the OVLT. The MnPO, the region of the LT that is considered as being an integrative area for sensory inputs arising from the SFO and OVLT, showed exclusive activation of excitatory neuronal populations. Taken together these results suggest that acute water and Na(+) depletion diminish the efficacy of the GABAergic system and mainly activates excitatory neuronal pathways in the regions of the LT.


Assuntos
Glutamato Descarboxilase/metabolismo , Ácido Glutâmico , Hipotálamo/metabolismo , Isoenzimas/metabolismo , Proteínas de Membrana Transportadoras , Neurônios/metabolismo , Sódio/deficiência , Proteínas de Transporte Vesicular , Água/metabolismo , Ácido gama-Aminobutírico , Animais , Proteínas de Transporte/metabolismo , Diuréticos/farmacologia , Eletrólitos , Furosemida/farmacologia , Imuno-Histoquímica , Hibridização In Situ , Masculino , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Proteína Vesicular 2 de Transporte de Glutamato
5.
Neuroscience ; 116(2): 539-51, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12559109

RESUMO

Exposure of CD-1 mice to a familiar environment lined with clean shavings (control odor) as well as a familiar environment lined with soiled rat shavings (predator odor) induced anxiety in the light/dark box. Mice exposed to the familiar environment or predator odor displayed decreased latency to enter the dark chamber of the light/dark box and spent less time in the light portion of the apparatus relative to home-caged mice. Mice exposed to the familiar environment lined with clean shavings or predator odor displayed elevated cholecystokinin mRNA levels from the ventral tegmental area, medial and basolateral nuclei of the amygdala relative to home-caged mice. Exposure of CD-1 mice to 2, 5 or 10 min of predator odor increased acoustic startle relative to mice merely exposed to the familiar environment lined with clean shavings at protracted intervals. Mice exposed to the familiar environment lined with clean shavings did not exhibit enhanced startle relative to home-caged mice. Exaggerated startle reactivity was in evidence immediately, 24, and 48 h following a 5-min exposure of mice to predator odor. In contrast, a 10-min exposure of mice to predator odor produced an oscillating pattern of enhanced startle evident during the immediate and 48-h post-stressor intervals only. However, when the startle stimulus was withheld 1 h following odor presentation, mice exhibited enhanced startle patterns reminiscent of the 5-min exposure. The 2-min exposure of mice to predator odor produced a delayed onset of enhanced startle observed at the 168-h test interval only. Potential anxiogenic influences of mesocorticolimbic cholecystokinin availability as well as the time course and underlying neuronal substrates of long-term behavioral disturbances as a result of psychogenic stressor manipulations are discussed.


Assuntos
Ansiedade/fisiopatologia , Encéfalo/fisiologia , Colecistocinina/genética , Estresse Psicológico/fisiopatologia , Estimulação Acústica , Tonsila do Cerebelo/fisiologia , Animais , Escuridão , Comportamento Exploratório/fisiologia , Medo/fisiologia , Expressão Gênica , Iluminação , Masculino , Camundongos , Camundongos Endogâmicos , Odorantes , Comportamento Predatório , Córtex Pré-Frontal/fisiologia , RNA Mensageiro/análise , Reflexo de Sobressalto/fisiologia , Área Tegmentar Ventral/fisiologia
6.
Artigo em Inglês | MEDLINE | ID: mdl-11383975

RESUMO

Numerous studies and reviews support an important contribution of endogenous opioid peptide systems in the mediation, modulation, and regulation of stress responses including endocrine (hypothalamopituitary-adrenal, HPA axis), autonomic nervous system (ANS axis), and behavioral responses. Although several discrepancies exist, the most consistent finding among such studies using different species and stressors is that opioids not only diminish stress-induced neuroendocrine and autonomic responses, but also stimulate these effector systems in the non-stressed state. A distinctive feature of the analgesic action of opioids is the blunting of the distressing, affective component of pain without dulling the sensation itself. Therefore, opioid peptides may diminish the impact of stress by attenuating an array of physiologic responses including emotional and affective states. The widespread distribution of enkephalin (ENK) throughout the limbic system (including the extended amygdala, cingulate cortex, entorhinal cortex, septum, hippocampus, and the hypothalamus) is consistent with a direct role in the modulation the stress responses. The predictability of stressful events reduces the impact of a wide range of stressors and ENK appears to play an important role in this process. Therefore, ENK and its receptors could represent a major modulatory system in the adaptation of an organism to stress, balancing the response that the stressor places on the central stress system with the potentially detrimental effects that a sustained stress may produce. Chronic neurogenic stressors will induce changes in specific components of the stress-induced ENKergic system, including ENK, delta- and mu-opioid receptors. This review presents evidences for adaptive cellular mechanisms underlying the response of the central stress system when assaulted by repeated psychogenic stress, and the involvement of ENK in these processes.


Assuntos
Endorfinas/fisiologia , Estresse Psicológico/fisiopatologia , Adaptação Psicológica/fisiologia , Animais , Humanos
7.
Peptides ; 22(5): 761-7, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11337089

RESUMO

Activation of the hypothalamic-pituitary-adrenal (HPA) axis is the main defining feature of the stress response. The primary mediator of this response is corticotropin-releasing hormone (CRH), a 41-residue peptide acknowledged as the principal hypophysiotropic factor driving stress-induced adrenocorticotropic hormone (ACTH) secretion. Although CRH is widely distributed within the central nervous system (CNS), the paraventricular nucleus (PVN) of the hypothalamus is the principal site of the parvocellular neurosecretory neurons responsible for delivering CRH to the hypophyseal portal system, an event that initiates the activity of the pituitary-adrenal axis. Stress-induced transcriptional activation of CRH takes place quite uniquely in this hypothalamic nucleus, despite the robust constitutive hybridization signal for CRH mRNA across the brain. The fact that CRH itself is capable of mimicking these effects and that de novo but transient expression of its type one receptor occurs in the PVN are data that make this hypothalamic region of great interest to study the mechanisms that lead to such specific transcriptional activity. This review will present evidence of such phenomenon by stressors of different categories as well as the possible neuromediators involved.


Assuntos
Hormônio Liberador da Corticotropina/genética , Sistema Hipotálamo-Hipofisário/fisiologia , Sistemas Neurossecretores/fisiologia , Núcleo Hipotalâmico Paraventricular/química , Receptores de Hormônio Liberador da Corticotropina/genética , Transcrição Gênica , Animais , Encéfalo/metabolismo , Hormônio Liberador da Corticotropina/química , Hormônio Liberador da Corticotropina/metabolismo , Ratos , Receptores de Hormônio Liberador da Corticotropina/química , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Estresse Fisiológico/fisiopatologia
8.
Appl Environ Microbiol ; 67(4): 1700-9, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11282624

RESUMO

pCD4, a small, highly stable theta-replicating lactococcal plasmid, was used to develop a food-grade cloning system. Sequence analysis revealed five open reading frames and two putative cis-acting regions. None appears to code for undesirable phenotypes with regard to food applications. Functional analysis of the replication module showed that only the cis-acting ori region and the repB gene coding for the replication initiator protein were needed for the stable replication and maintenance of pCD4 derivatives in Lactococcus lactis. A two-component food-grade cloning system was derived from the pCD4 replicon. The vector pVEC1, which carries the functional pCD4 replicon, is entirely made up of L. lactis DNA and has no selection marker. The companion pCOM1 is a repB-deficient pCD4 derivative that carries an erythromycin resistance gene as a dominant selection marker. The pCOM1 construct can only replicate in L. lactis if trans complemented by the RepB initiator provided by pVEC1. Since only the cotransformants that carry both pVEC1 and pCOM1 can survive on plates containing erythromycin, pCOM1 can be used transiently to select cells that have acquired pVEC1. Due to the intrinsic incompatibility between these plasmids, pCOM1 can be readily cured from the cells grown on an antibiotic-free medium after the selection step. The system was used to introduce a phage resistance mechanism into the laboratory strain MG1363 of L. lactis and two industrial strains. The introduction of the antiphage barrier did not alter the wild-type plasmid profile of the industrial strains. The phenotype was stable after 100 generations and conferred an effective resistance phenotype against phages of the 936 and c2 species.


Assuntos
Clonagem Molecular , Indústria Alimentícia/normas , Lactococcus lactis/genética , Plasmídeos/genética , Replicon/genética , Sequência de Aminoácidos , Bacteriófagos/patogenicidade , Sequência de Bases , Microbiologia de Alimentos/normas , Dosagem de Genes , Lactococcus lactis/virologia , Dados de Sequência Molecular , Análise de Sequência de DNA
9.
Mol Neurobiol ; 23(2-3): 155-71, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11817217

RESUMO

Opiate dependence and withdrawal involve neuroadaptive responses in the central nervous system. A host of studies have previously implicated the A6 noradrenergic neurons of the pontine nucleus locus coeruleus (LC) as an important mediator of somatic signs observed upon withdrawal from opiates. Recent studies, however, are showing that noradrenergic neurons of the LC may not be solely involved in mediating somatic signs of withdrawal. The A2 noradrenergic neurons of the nucleus of the solitary tract (nucleus tractus solitarius [NTS]) in the caudal brainstem may be another possible site. Neurons in the nucleus paragigantocellularis lateralis (PGi), located in the rostral ventral medulla, which are known to send collateral projections to both the LC and the NTS, may co-modulate both noradrenergic nuclei in a parallel fashion, which may represent an anatomical substrate underlying the behavioral expression of opiate withdrawal. The PGi provides glutamatergic and opioid innervation to LC neurons. Hyperactivity of LC during opiate withdrawal arises, in part, from increased glutamate transmission in this pathway. The authors have recently shown that the excitatory transmitter, glutamate, co-exists with the endogenous opioid peptide, enkephalin, in a subset of axon terminals in the LC. Decreases in endogenous opioids in afferents to LC and NTS, following chronic opiate administration, may be equally important in modulating noradrenergic neurons following chronic opiate exposure, by removing a neurochemical system that would inhibit noradrenergic neurons. A persistent decrease in opioid peptide release from afferents during withdrawal would result in glutamate acting on postsynaptic targets, in an unopposed fashion. A parallel effect in opioid projections from PGi to the NTS would potentially support similar actions in this noradrenergic nucleus. The authors' recent data show that opioid-containing neurons in the PGi project to the NTS, and that enkephalin levels are decreased in opioid afferents to the NTS. This review summarizes data that the authors have collected regarding opioid expression changes in brainstem circuits (PGi-LC and PGi-NTS), following chronic morphine treatment, which may represent a model for understanding of adaptations in endogenous opioid circuits during drug dependence and withdrawal.


Assuntos
Analgésicos Opioides/administração & dosagem , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/fisiologia , Morfina/administração & dosagem , Norepinefrina/fisiologia , Animais , Tronco Encefálico/metabolismo , Humanos , Norepinefrina/metabolismo
10.
Artigo em Inglês | MEDLINE | ID: mdl-11131177

RESUMO

1. The main objective of this work was to investigate the extent of cellular colocalization of dopamine D1 and D2 receptors in the rat brain. A double labeling technique, that combined immunocytochemical labeling of the D2 receptor using polyclonal antibodies raised against the third intracellular loop of the short isoform of the human D2 receptor in combination with in situ hybridization detecting D1 mRNA expression, was designed to accomplish this goal. 2. The specificity of the antisera obtained was confirmed by immunoprecipitation assay, Western blot analysis, and immunocytochemistry on D2R transfected cells and murine brain tissue. Western blot using the D2 receptor antibody revealed a specific broad band centered at 67 kDa in transfected cells and a major protein of 88 kDa corresponding to D2R expressed in the caudate-putamen, to a lesser extent in the cortex, and not at all detected in the hypothalamic region. 3. The content of neurons double-labeled for D1/D2 receptors was observed at in differing intensities in the dorsal endopiroform nucleus, the intercalated nucleus of amygdala, the anterior part of the cortical nucleus amygdala, the nucleus of the lateral olfactory tract, the piriform cortex, the parabrachial nucleus, the supraoptic nucleus and the parabigeminal nucleus. All other regions of the brain revealed neurons expressing either D1 or D2 dopamine receptors but not both at that same time. 4. These results clearly demonstrated that specific neurons expressed both receptors D1 and D2, and that this colocalization was restricted to particular regions of the rat brain.


Assuntos
Encéfalo/fisiologia , Receptores de Dopamina D1/análise , Receptores de Dopamina D2/análise , Animais , Anticorpos , Western Blotting , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Neurônios/fisiologia , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Coelhos , Ratos , Receptores de Dopamina D1/fisiologia , Receptores de Dopamina D2/fisiologia
11.
J Neurochem ; 75(5): 2200-11, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11032909

RESUMO

This study tested the hypothesis that the activation pattern of enkephalinergic (ENKergic) neurons within the paraventricular nucleus of the hypothalamus (PVH) in response to psychogenic stress is identical whether in response to repeated exposure to the same stress (homotypic; immobilization) or to a novel stress (heterotypic; air jet puff). Rats were assigned to either acute or chronic immobilization stress paradigms (90 min/day for 1 or 10 days, respectively). The chronic group was then subjected to an additional 90-min session of either heterotypic or homotypic stress. A single 90-min stress session (immobilization or air jet) increased PVH-ENK heteronuclear (hn) RNA expression. In chronically stressed rats, exposure to an additional stress session (whether homotypic or heterotypic) continued to stimulate ENK hnRNA expression. Acute immobilization caused a marked increase in the numbers of Fos-immunoreactive and Fos-ENK double-labeled cells in the dorsal and ventral medial parvicellular, and lateral parvicellular subdivisions of the PVH. Chronic immobilization caused an attenuated Fos response ( approximately 66%) to subsequent immobilization. In contrast, chronic immobilization did not impair ENKergic neuron activation within the PVH following homotypic or heterotypic stress. These results indicate that within the PVH, chronic psychogenic stress markedly attenuates the Fos response, whereas ENKergic neurons resist habituation, principally within the ventral neuroendocrine portion of the nucleus. This suggests an increase in ENK effect during chronic stress exposure. Homotypic (immobilization) and heterotypic (air jet) psychogenic stressors produce similar responses, including Fos, ENK-Fos, and ENK hnRNA, within each subdivision of the PVH, suggesting similar processing for painless neurogenic stimuli.


Assuntos
Encefalinas/biossíntese , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Estresse Psicológico/metabolismo , Doença Aguda , Hormônio Adrenocorticotrópico/sangue , Animais , Doença Crônica , Corticosterona/sangue , Encefalinas/genética , Teste de Esforço , Masculino , Neurônios/citologia , Núcleo Hipotalâmico Paraventricular/citologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Restrição Física , Estresse Psicológico/sangue
12.
J Neurochem ; 74(6): 2568-75, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10820219

RESUMO

We examined the effects of acute and chronic psychogenic stress on the activation pattern of enkephalin-containing perikarya in the rat ventrolateral medulla. Rats allocated to the chronic stress groups were subjected to 90 min of immobilization for 10 days. On the 11th day, the chronically stressed rats were exposed to homotypic (90-min immobilization) or to heterotypic but still psychogenic (90-min immobilization coupled to air jet stress) stress. The acute stress group was subjected once to an acute 90-min immobilization. For each group, the rats were anesthetized either before stress (time 0) or 90, 180, and 270 min after the onset of stress. Brain sections were then processed using immunocytochemistry (Fos protein) followed by radioactive in situ hybridization histochemistry (enkephalin mRNA). Following immobilization, the acute group displayed a marked increase in the number of activated enkephalin-containing perikarya within the paragigantocellularis and lateral reticular nuclei. This level of activation was sustained up to 180 min following the onset of the immobilization stress and had returned to baseline levels by 270 min from the initiation of the stress. However, this stress-induced activation of enkephalin-containing perikarya of the ventrolateral medulla was not seen following either homotypic or heterotypic stress in the chronically stressed group. These results provide evidence that enkephalin-containing perikarya of the ventrolateral medulla may constitute a potential circuit through which they regulate some aspect of the stress responses. Conversely, this enkephalinergic influence from the ventrolateral medulla was shown to be absent following chronic stress exposure. This would suggest a decrease in enkephalin inhibitory input originating from the ventrolateral medulla, thereby allowing a neuroendocrine and/or autonomic response to chronic stress.


Assuntos
Sistema Nervoso Autônomo/fisiologia , Encefalinas/genética , Bulbo/fisiologia , Estresse Psicológico/fisiopatologia , Doença Aguda , Movimentos do Ar , Animais , Sistema Nervoso Autônomo/química , Sistema Nervoso Autônomo/citologia , Doença Crônica , Expressão Gênica/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Hibridização In Situ , Masculino , Bulbo/química , Bulbo/citologia , Neurônios/química , Neurônios/fisiologia , Sistema Hipófise-Suprarrenal/fisiologia , Proteínas Proto-Oncogênicas c-fos/análise , RNA Complementar , RNA Mensageiro/análise , Ratos , Ratos Wistar , Restrição Física
13.
J Neurosci ; 20(23): 8659-66, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11102471

RESUMO

Several biochemical changes have been described in noradrenergic neurons of the locus coeruleus (LC) after chronic morphine treatment. Changes in neurochemical expression in opioid afferent projections to the LC may be equally important in modulating noradrenergic neurons during chronic opiate exposure. To test the hypothesis that opioid peptides in LC afferents are altered after chronic opiate administration, we exposed adult male rats to either morphine or placebo pellets for 5 d. Tissue sections through the LC were processed for peroxidase or gold-silver labeling of methionine(5)-enkephalin (met-ENK) and analyzed using light or electron microscopy, respectively. Light level densitometry and ultrastructural analysis showed that there was a significant decrease in immunolabeling for ENK in LC-afferent terminals of morphine-treated rats. Western immunoblot analysis confirmed that protein levels for both leucine(5)- and methionine(5)-ENK were significantly decreased in tissue samples containing the LC after chronic morphine treatment. To test whether decreases in ENK protein expression were mirrored by decreases in gene expression, Northern blot analysis of preproenkephalin (PPE) mRNA was conducted in tissue samples obtained through the medulla, a brainstem area that contains the major opioid afferents to the LC. PPE mRNA was reduced in samples obtained from morphine-treated rats. Finally, in situ hybridization experiments confirmed significant decreases in PPE mRNA expression in the nucleus paragigantocellularis, a region known to provide a robust opioid input to the LC. These data suggest that there is a decrease in the synthesis of the opioid peptide mRNA and protein in the medullo-coerulear pathway after chronic exposure to morphine. Such alterations in opioid peptide levels during opiate dependence may contribute to the observed hyperactivity of LC neurons during opiate withdrawal.


Assuntos
Locus Cerúleo/metabolismo , Bulbo/metabolismo , Morfina/administração & dosagem , Vias Neurais/metabolismo , Peptídeos Opioides/metabolismo , Animais , Northern Blotting , Western Blotting , Densitometria , Implantes de Medicamento , Encefalina Leucina/metabolismo , Encefalina Metionina/metabolismo , Encefalinas/genética , Encefalinas/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Locus Cerúleo/citologia , Locus Cerúleo/efeitos dos fármacos , Masculino , Bulbo/citologia , Bulbo/efeitos dos fármacos , Microscopia Eletrônica , Vias Neurais/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
14.
Arch Mal Coeur Vaiss ; 92(8): 1111-3, 1999 Aug.
Artigo em Francês | MEDLINE | ID: mdl-10486675

RESUMO

The dynamic regulation of neurotransmitters and their receptors is an important component of the process of coping and stress adaptation. Among the central neurochemical systems, CRF and the renin-angiotensin may represent major modulatory systems involved in the adaptation of an organism to chronic stress, balancing the response demands that the stressor places on the central nervous system with the potentially detrimental effects that a sustained stress response may produce. As such, the study of these two systems with respect to their neurotransmitters and receptors will allow us to achieve a better perspective on the mechanisms responsible for effective short-term coping with stress as well as long-term adaptation and restoration in response to chronic or repeated stress. It will then be possible to verify the level of activation of different components of the central pathways involved in the mediation of stress responses in rat strains which develop hypertension following chronic exposure to stress. The primary objective of the present paper is to review some facts on the contribution of the central renin-angiotensin system on the regulatory mechanisms involved in the mediation of physiological responses to stress and on the involvement of these neurons in the CNS adaptation in rat strains that are developing hypertension when chronically exposed to stress. Neurons that are expressing angiotensin-receptors may be important in the short-term adaptation to stress by potentiating sympathoadrenal and/or hypophyseo-pituitary-adrenal responses. These same neurons may also participate in long-term stress-adaptation by altering gene expression of their angiotensin receptors. Moreover, these processes represent potential points of dysregulation in the case of extreme, repeated or prolonged stress, and thus in the development of stress-related pathological states such as hypertension and heart diseases.


Assuntos
Adaptação Fisiológica , Sistema Nervoso Central/fisiologia , Hipertensão/fisiopatologia , Sistema Renina-Angiotensina/fisiologia , Estresse Fisiológico/fisiopatologia , Animais , Sistema Nervoso Central/citologia , Neurônios/fisiologia
15.
Neuroscience ; 93(3): 877-84, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10473253

RESUMO

The present study examined the effects of acute and chronic neurogenic stressors on the expression of two distinct angiotensin receptors in two stress-related brain nuclei: angiotensin type 1A receptor in the paraventricular nucleus of the hypothalamus and angiotensin type 2 receptor in the nucleus locus coeruleus. Male Wistar rats were divided into four experimental groups. The first two groups were subjected once to an acute 90-min immobilization or air-jet stress session, respectively. The other two groups were subjected to 10 days of daily 90-min immobilization sessions and, on the 11th day, one group was exposed to an additional 90-min immobilization and the other to a single air-jet stress (heterotypic but still neurogenic) session. In each group, rats were perfused before stress (0 min), immediately following stress (90 min) or 150, 180, 270 or 360 min (and 24 h in chronic immobilization) after the beginning of the last stress session. Basal expression of both angiotensin receptor subtype 1A and angiotensin receptor subtype 2 messenger RNA was minimal in non-stressed animals. Acute immobilization as well as air-jet stress induced similar patterns (time-course and maximal values) of angiotensin receptor subtype 1A messenger RNA expression in the paraventricular nucleus. Angiotensin receptor subtype 1A messenger RNA expression increased 90-150 min after the beginning of the stress and returned to basal levels by 360 min. Chronic stress immobilization slightly modified the pattern, but not maximal values of angiotensin receptor subtype 1A messenger RNA expression to further immobilization (homotypic) or air-jet stress (heterotypic). Acute immobilization and air-jet stress sessions induced similar locus coeruleus-specific angiotensin receptor subtype 2 messenger RNA expression. This expression increased 90 min following the onset of the stress session and remained elevated for at least 360 min. Chronic immobilization stress increased angiotensin receptor subtype 2 messenger RNA expression to levels comparable to those observed in acute stress conditions. Novel acute exposure to neurogenic stressors did not further increase these levels in either homotypic (immobilization) or in heterotypic (air-jet stress) conditions. These results suggest that central angiotensin receptors are targets of regulation in stress; therefore, stress may modulate angiotensin function in the paraventricular nucleus and locus coeruleus during chronic exposure to neurogenic stressors.


Assuntos
Adaptação Fisiológica/fisiologia , Angiotensina II/fisiologia , Locus Cerúleo/fisiopatologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Receptores de Angiotensina/fisiologia , Estresse Fisiológico/fisiopatologia , Ar , Animais , Imobilização/efeitos adversos , Masculino , Ratos , Ratos Wistar , Receptor Tipo 1 de Angiotensina , Receptor Tipo 2 de Angiotensina , Estresse Psicológico/fisiopatologia , Regulação para Cima
16.
J Neurobiol ; 36(3): 357-78, 1998 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-9733072

RESUMO

The recent cloning of a second estrogen receptor (ER) provided a new tool to investigate and clarify how estrogens are capable of communicating with the brain and influence gene expression and neural function. The purpose of the present study was to define the neuroanatomical organization of each receptor subtype using a side-by-side approach and to characterize the cellular population (s) expressing the ERbeta transcript in the endocrine hypothalamus using immunohistochemistry combined with in situ hybridization. Axonal transport inhibition was accomplished to cause neuropeptide accumulation into the cytoplasm and thus facilitate the detection of all positive luteinizing hormone-releasing hormone (LHRH), corticotropin-releasing factor (CRF), vasopressin (AVP), oxytocin (OT), gastrin-related peptide (GRP), and enkephalin (ENK) neurons. The genes encoding either ERalpha or -beta were expressed in numerous limbic-associated structures, and fine differences were found in terms of intensity and positive signal. Such phenomenon is best represented by the bed nucleus of the stria terminalis (BnST) and preoptic area/anterior hypothalamus, where the expression pattern of both transcripts differed across subnuclei. The novel ER was also found to be expressed quite exclusively in other hypothalamic nuclei, including the supraoptic (SON) and selective compartments (magnocellular and autonomic divisions) of the paraventricular nucleus (PVN). A high percentage of the ERbeta-expressing neurons located in the ventro- and dorsomedial PVN are of OT type; 40% of the OT-ir cells forming the medial magnocellular and ventromedial parvocellular PVN showed a clear hybridization signal for ERbeta mRNA, whereas a lower percentage (15-20%) of OT neurons were positive in the caudal parvocellular PVN and no double-labeled cells were found in the rostral PVN and other regions of the brain with the exception of the SON. Very few AVP-ir neurons expressing ERbeta transcript were found throughout the rat brain, although the medial PVN displayed some scattered double-labeled cells (<5%). Quite interestingly, the large majority of the ERbeta-positive cells in the caudal PVN were colocalized within CRF-ir perikarya. Indeed, more than 60-80% of the CRF-containing cells located in the caudolateral division of the parvocellular PVN exhibited a positive hybridization signal for ERbeta mRNA, whereas very few (<5%) neuroendocrine CRF-ir parvocellular neurons of the medial PVN expressed the gene encoding ERbeta. A small percentage of ERbeta-expressing cells in the dorsocaudal and ventromedial zones of the parvocellular PVN were also ENK positive. The ventral zone of the medial parvocellular PVN also displayed GRP-ir neurons, but no convincing hybridization signal for ERbeta was detected in this neuronal population. Finally, as previously described for the gene encoding the classic ER, LHRH neurons of both intact and colchicine-pretreated animals did not express the novel estrogen receptor. This study shows a differential pattern of expression of both receptors in the brain of intact rats and that ERbeta is expressed at various levels in distinct neuropeptidergic populations, including OT, CRF, and ENK. The influence of estrogen in mediating genomic and neuronal responses may therefore take place within these specific cellular groups in the brains of cycling as well as intact male mammals.


Assuntos
Química Encefálica/fisiologia , Encéfalo/anatomia & histologia , Neuropeptídeos/química , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/química , Animais , Colchicina/administração & dosagem , Colchicina/farmacologia , DNA/biossíntese , DNA/genética , Imuno-Histoquímica , Hibridização In Situ , Injeções Intraventriculares , Masculino , Plasmídeos/biossíntese , Plasmídeos/genética , Sondas RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
J Neurochem ; 70(4): 1556-66, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9523573

RESUMO

The effects of immobilization on the expression of immediate-early gene c-fos and nerve growth factor-inducible (NGFI)-B mRNAs, corticotropin-releasing factor (CRF) mRNA, CRF heteronuclear RNA (hnRNA), CRF receptor types 1 and 2alpha mRNA, and enkephalin hn/mRNA were investigated in the hypothalamic paraventricular nucleus of Wistar-Kyoto (WKY) rats and borderline hypertensive rats (BHRs). Rats were deeply anesthetized 0, 30, 60, and 180 min after the beginning of the immobilization session (60 min maximum). BHR paraventricular nuclei displayed slight differences in their resting levels of NGFI-B mRNA and CRF hnRNA, both being significantly elevated compared with those of WKY rats. Conversely, basal levels of enkephalin primary transcript were significantly lower in BHRs. Immobilization, however, induced transient variations in the hybridization signals for all evaluated genes within the paraventricular nucleus (except for CRF 2alpha receptor). Immediate-early gene mRNA levels were higher and more prolonged in BHRs than in WKY rats. This heightened neuronal activation in the BHRs was associated with a more rapid increase in CRF mRNA expression (30 min) compared with that in WKY rats (60 min). It is interesting that a transient rise in CRF hnRNA levels was detected in stressed WKY rats, whereas the BHR group displayed a progressive decline in this transcript, being significantly below resting levels 180 min after the immobilization session. The stress-induced expression of CRF type 1 receptor mRNA was similar in both strains. Moreover, no significant differences were observed for enkephalin hn/mRNA in either strain during the immobilization session. Therefore, the hypothalamic paraventricular nucleus appears to be involved in the functional hyperreactivity of the hypothalamic-pituitary-adrenal and autonomic axes to stress observed in BHRs, which may lead ultimately to a stress-induced hypertensive state.


Assuntos
Regulação da Expressão Gênica/fisiologia , Hipertensão/genética , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Estresse Fisiológico/genética , Transcrição Gênica/fisiologia , Animais , Hormônio Liberador da Corticotropina/genética , Encefalinas/genética , Expressão Gênica/fisiologia , Genes Precoces/genética , Hipertensão/patologia , Imobilização , Masculino , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/patologia , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos , Ratos Endogâmicos WKY , Receptores de Hormônio Liberador da Corticotropina/genética , Estresse Fisiológico/etiologia
18.
J Neuroendocrinol ; 10(2): 119-31, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9535058

RESUMO

It has been reported that corticotropin-releasing factor (CRF) may regulate its own biosynthesis in the paraventricular nucleus of the hypothalamus (PVH). Whether this CRF autoregulation is mediated by local circuitry or from extra-PVH CRF neuronal fibers terminating on CRF perikarya within the PVH is unknown. In the present study, we sought to determine the origin(s) of this CRF innervation using retrograde transport of wheat germ-conjugated-gold particles (WGA-apoHRP-Au) combined with immunohistochemistry for CRF. The rats also received colchicine (100 microg, icv) 5-7 days after tracer injection and were perfused 24 h later. Results of retrograde labeling with pressure injections of WGA-apoHRP-Au centered to PVH and subsequent immunohistochemical staining for CRF demonstrated numerous retrogradely labeled CRF neurons in the perifornical hypothalamic nucleus (PeF), the dorsolateral hypothalamic area (DA) (medial and lateral portions) and the dorsomedial nucleus of the hypothalamus (DMH). Smaller groups of CRF-ir neurons that were retrogradely labeled were found in the bed nuclei of the stria terminalis (BnST), the Barrington's nucleus (Bar) and the dorsal raphé (DR). These CRFergic pathways to the PVH may represent an anatomical substrate underlying the function of the stress-integrative PVH neurons in the autonomic, behavioral and neuroendocrine regulation during the stress response, including CRF autoregulation.


Assuntos
Hormônio Liberador da Corticotropina/fisiologia , Vias Neurais/anatomia & histologia , Vias Neurais/fisiologia , Neurônios Aferentes/fisiologia , Núcleo Hipotalâmico Paraventricular/anatomia & histologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Animais , Hormônio Liberador da Corticotropina/metabolismo , Hormônio Liberador da Corticotropina/farmacologia , Imuno-Histoquímica , Hibridização In Situ , Masculino , Neurônios Aferentes/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Ratos Wistar
19.
Am J Physiol ; 272(3 Pt 2): R813-20, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9087643

RESUMO

The effects of acute foot shock on cardiovascular and sympathoadrenal responses were investigated in chronically stressed borderline hypertensive rats (BHR) and Wistar-Kyoto (WKY) rats. Male BHRs were divided into two groups; the maturation group (Mat) was not stressed, whereas the chronic stress group (AJS) received 8 wk (5 days/wk, 30 min/day) of air-jet stress coupled with immobilization. After chronic stress, the rats were cannulated in the femoral artery and jugular vein. Resting mean arterial pressure (MAP), heart rate (HR), and plasma norepinephrine (NE) levels were higher in the AJS group. In contrast, chronic stress failed to increase basal arterial pressure or HR in WKY rats. In response to acute foot shock, the Mat group had higher MAP increases, at which they plateaued, whereas the AJS rats displayed a progressive decline in MAP. This was associated with higher plasma NE and epinephrine levels but a smaller increase in adrenocorticotropic hormone in AJS versus Mat rats. This hyperactivity in the sympathoadrenal system of chronically stressed BHRs may, in part, account for their mild hypertension.


Assuntos
Epinefrina/sangue , Hemodinâmica , Hipertensão/fisiopatologia , Norepinefrina/sangue , Estresse Psicológico/fisiopatologia , Hormônio Adrenocorticotrópico/sangue , Ar , Animais , Pressão Sanguínea , Eletrochoque , Frequência Cardíaca , Hipertensão/psicologia , Masculino , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos , Ratos Endogâmicos WKY , Valores de Referência , Restrição Física , Fatores de Tempo
20.
Endocrinology ; 137(11): 4619-29, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8895325

RESUMO

The present study sought to examine the effects of intracerebroventricular (icv) administration of corticotropin-releasing factor (CRF) on the expression of CRF1 receptor messenger RNA (mRNA) within the hypothalamus as determined by quantitative in situ hybridization histochemistry. Adult male Sprague-Dawley rats were stereotaxically implanted with guide cannulae directed towards the right lateral ventricle. After 8-10 days of recovery, either 10 microliters CRF (5 micrograms) or vehicle solution was injected into the lateral ventricle over a 2-min period. The rats were then deeply anesthetized at 15, 60, and 180 min after icv injection, transcardially perfused, and their brains cut into 30-micron coronal sections. Brain sections were then processed using standard radioactive in situ hybridization histochemistry revealing the expression of the CRF1 receptor mRNA. Low to moderate basal levels of CRF1 receptor transcript were observed in several regions of the forebrain. However, the hybridization signal for the mRNA encoding the CRF1 receptor was barely detectable in the paraventricular nucleus of the hypothalamus (PVN) of vehicle-injected rats. In contrast, 180 min after icv administration of CRF, a significant increase in CRF1 receptor transcript was measured specifically in the PVN, despite having virtually any hybridization signal before 180 min. This increase in the level of receptor transcription by CRF was restricted to the type 1 receptor subtype because the hybridization signal for the CRF2 alpha receptor mRNA was unaffected in the brain regions in which it was located. Moreover, we confirmed previous findings of a CRF-induced neuronal activation of parvocellular neurosecretory cells of the PVN, as assessed by c-fos mRNA expression. This neuronal activation induced by exogenous CRF was also associated with a rapid and strong induction of CRF heteronuclear RNA selectively in the rat PVN, a phenomenon abolished by a pretreatment with a CRF receptor antagonist. These results provide evidence that elevated levels of central CRF may trigger CRF1 receptor transcription selectively in the PVN. This positive feedback of CRF on its own receptor may represent a functional adaptation of the hypothalamic-pituitary-adrenal axis in response to stress.


Assuntos
Encéfalo/metabolismo , Ventrículos Cerebrais/fisiologia , Hormônio Liberador da Corticotropina/farmacologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores de Hormônio Liberador da Corticotropina/biossíntese , Transcrição Gênica/efeitos dos fármacos , Análise de Variância , Animais , Encéfalo/efeitos dos fármacos , Ventrículos Cerebrais/efeitos dos fármacos , Hormônio Liberador da Corticotropina/administração & dosagem , Retroalimentação , Injeções Intraventriculares , Cinética , Masculino , Especificidade de Órgãos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Prosencéfalo/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Sondas RNA , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Valores de Referência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...