Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 250: 520-35, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-23876319

RESUMO

Neurons and neighboring astrocytic glia are mostly studied in nervous tissues from rodents whereas less is known on their properties and interactions in the human brain. Here, confocal/multiphoton fluorescence imaging for several hours revealed that co-cultured fetal human cortical neurons and astrocytes show pronounced spontaneous rises of cytosolic Ca(2+) which last for up to several minutes without concomitant changes in either movements or membrane potential of mitochondria. Similar Ca(2+) rises were evoked mainly in neurons by bath-applied glutamate or γ-aminobutyric acid (GABA) acting via N-methyl-d-aspartate (NMDA)+AMPA/Kainate and GABAA receptors, respectively. Predominantly in astrocytes, Ca(2+) baseline was elevated by adenosine diphosphate (ADP) and adenosine triphosphate (ATP) acting via P2Y1 and P2X7 receptors, likely causing the release of glutamate and glutamine. Mainly astrocytes responded to histamine, whereas the activation of muscarinic acetylcholine (ACh) receptors raised Ca(2+) in both cell types. Evoked neuronal and astrocytic Ca(2+) rises could last for several minutes without affecting mitochondrial movements or membrane potential. In contrast, reversible depolarization of mitochondrial membrane potential accompanied neuronal Ca(2+) rises induced by cyanide-evoked chemical anoxia or the uncoupling of mitochondrial respiration with carbonyl-cyanide-4-(trifluoromethoxy)-phenylhydrazone (FCCP). During such metabolic perturbation, mitochondrial depolarization also occurred in astrocytes, whereas Ca(2+) was largely unaffected. In summary, fetal human cortical neurons and astrocytes show distinct patterns of neuro/glio-transmitter- and metabolically-evoked Ca(2+) rises and possess active mitochondria. One aspect of our discussion deals with the question of whether the functional mitochondria contribute to cellular Ca(2+) homeostasis that seems to be already well-developed in fetal human cortical brain cells.


Assuntos
Astrócitos/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/fisiologia , Córtex Cerebral/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Astrócitos/fisiologia , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Técnicas de Cocultura , Citosol/metabolismo , Interpretação Estatística de Dados , Feminino , Humanos , Potencial da Membrana Mitocondrial/fisiologia , Microscopia Confocal , Microscopia de Fluorescência , Neuroglia/fisiologia , Neurônios/fisiologia , Neurotransmissores/farmacologia , Gravidez , Cultura Primária de Células , Desacopladores/farmacologia
2.
Mol Neurobiol ; 47(1): 37-63, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22983915

RESUMO

Alzheimer's disease (AD) is a complex multifactorial neurodegenerative disorder believed to be initiated by accumulation of amyloid ß (Aß)-related peptides derived from proteolytic processing of amyloid precursor protein (APP). Research over the past two decades provided a mechanistic link between cholesterol and AD pathogenesis. Genetic polymorphisms in genes regulating the pivotal points in cholesterol metabolism have been suggested to enhance the risk of developing AD. Altered neuronal membrane cholesterol level and/or subcellular distribution have been implicated in aberrant formation, aggregation, toxicity, and degradation of Aß-related peptides. However, the results are somewhat contradictory and we still do not have a complete understanding on how cholesterol can influence AD pathogenesis. In this review, we summarize our current understanding on the role of cholesterol in regulating the production/function of Aß-related peptides and also examine the therapeutic potential of regulating cholesterol homeostasis in the treatment of AD pathology.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Colesterol/metabolismo , Sequência de Aminoácidos , Precursor de Proteína beta-Amiloide/química , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Homeostase , Humanos , Dados de Sequência Molecular
3.
Neuropharmacology ; 54(4): 721-33, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18222494

RESUMO

The invariant characteristic features associated with Alzheimer's disease (AD) brain include the presence of extracellular neuritic plaques composed of amyloid beta (Abeta) peptide, intracellular neurofibrillary tangles containing hyper-phosphorylated tau protein and the loss of basal forebrain cholinergic neurons. Studies of the pathological changes that characterize AD and several other lines of evidence indicate that in vivo accumulation of Abeta(1-42) may initiate the process of neurodegeneration observed in AD brains. However, the cause of degeneration of the basal forebrain cholinergic neurons and their association to Abeta peptides or phosphorylated tau protein have not been clearly established. In the present study, using rat primary septal cultures, we have shown that Abeta(1-42), in a time (1-48 h) and concentration (0.01-20 microM)-dependent manner, induce toxicity in cultured neurons. Subsequently, we have demonstrated that Abeta toxicity is mediated via activation of cysteine proteases, i.e., calpain and caspase, and proteolytic breakdown of their downstream substrates tau, microtubule-associated protein-2 and alpha II-spectrin. Additionally, Abeta-treatment was found to induce phosphorylation of tau protein along with decreased levels of phospho-Akt and phospho-Ser(9)glycogen synthase kinase-3beta. Exposure to specific inhibitors of caspase or calpain can partially protect cultured neurons against Abeta-induced toxicity but their effects are not found to be additive. These results, taken together, suggest that Abeta peptide can induce toxicity in rat septal cultured neurons by activating multiple intracellular signaling molecules. Additionally, evidence that inhibitors of caspase and calpains can partially protect the cultured basal forebrain neurons raised the possibility that their inhibitors could be of therapeutic relevance in the treatment of AD pathology.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Calpaína/metabolismo , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Septo do Cérebro/citologia , Animais , Proteínas de Ligação ao Cálcio/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Microscopia Eletrônica de Varredura/métodos , Proteínas Associadas aos Microtúbulos/metabolismo , Emaranhados Neurofibrilares/ultraestrutura , Neurônios/ultraestrutura , Oligopeptídeos/farmacologia , Gravidez , Ratos , Sais de Tetrazólio , Tiazóis , Fatores de Tempo
4.
Mol Neurobiol ; 35(3): 329-45, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17917122

RESUMO

The G protein-coupled receptor (GPCR) family represents the largest and most versatile group of cell surface receptors. Classical GPCR signaling constitutes ligand binding to a seven-transmembrane domain receptor, receptor interaction with a heterotrimeric G protein, and the subsequent activation or inhibition of downstream intracellular effectors to mediate a cellular response. However, recent reports on direct, receptor-independent G protein activation, G protein-independent signaling by GPCRs, and signaling of nonheptahelical receptors via trimeric G proteins have highlighted the intrinsic complexities of G protein signaling mechanisms. The insulin-like growth factor-II/mannose-6 phosphate (IGF-II/M6P) receptor is a single-transmembrane glycoprotein whose principal function is the intracellular transport of lysosomal enzymes. In addition, the receptor also mediates some biological effects in response to IGF-II binding in both neuronal and nonneuronal systems. Multidisciplinary efforts to elucidate the intracellular signaling pathways that underlie these effects have generated data to suggest that the IGF-II/M6P receptor might mediate transmembrane signaling via a G protein-coupled mechanism. The purpose of this review is to outline the characteristics of traditional and nontraditional GPCRs, to relate the IGF-II/M6P receptor's structure with its role in G protein-coupled signaling and to summarize evidence gathered over the years regarding the putative signaling of the IGF-II/M6P receptor mediated by a G protein.


Assuntos
Comunicação Celular/fisiologia , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Receptor IGF Tipo 2/metabolismo , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular , Receptor IGF Tipo 2/química , Receptor IGF Tipo 2/genética
5.
J Neurochem ; 95(1): 263-72, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16181430

RESUMO

Glycogen synthase kinase-3beta (GSK-3beta) is a multifunctional enzyme involved in a variety of biological events including development, glucose metabolism and cell death. Its activity is inhibited by phosphorylation of the Ser9 residue and up-regulated by Tyr216 phosphorylation. Activated GSK-3beta increases phosphorylation of tau protein and induces cell death in a variety of cultured neurons, whereas phosphorylation of phosphatidylinositol-3 (PI-3) kinase-dependent protein kinase B (Akt), which inhibits GSK-3beta activity, is one of the best characterized cell survival signaling pathways. In the present study, the cholinergic immunotoxin 192 IgG-saporin was used to address the potential role of GSK-3beta in the degeneration of basal forebrain cholinergic neurons, which are preferentially vulnerable in Alzheimer's disease (AD) brain. GSK-3beta co-localized with a subset of forebrain cholinergic neurons and loss of these neurons was accompanied by a transient decrease in PI-3 kinase, phospho-Ser473Akt and phospho-Ser9GSK-3beta levels, as well as an increase in phospho-tau levels, in the basal forebrain and hippocampus. Total Akt, GSK-3beta, tau and phospho-Tyr216GSK-3beta levels were not significantly altered in these brain regions in animals treated with 192 IgG-saporin. Systemic administration of the GSK-3beta inhibitor LiCl did not significantly affect cholinergic marker or phospho-Ser9GSK-3beta levels in control rats but did preclude 192-IgG saporin-induced alterations in PI-3 kinase/phospho-Akt, phospho-Ser9GSK-3beta and phospho-tau levels, and also partly protected cholinergic neurons against the immunotoxin. These results provide the first evidence that increased GSK-3beta activity, via decreased Ser9 phosphorylation, can mediate, at least in part, 192-IgG saporin-induced in vivo degeneration of forebrain cholinergic neurons by enhancing tau phosphorylation. The partial protection of these neurons following inhibition of GSK-3beta kinase activity suggests a possible therapeutic role for GSK-3beta inhibitors in attenuating the loss of basal forebrain cholinergic neurons observed in AD.


Assuntos
Anticorpos Monoclonais/farmacologia , Colinérgicos/farmacologia , Fibras Colinérgicas/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Imunotoxinas/farmacologia , Degeneração Neural/induzido quimicamente , Neurônios/efeitos dos fármacos , Prosencéfalo/efeitos dos fármacos , Animais , Anticorpos Monoclonais/administração & dosagem , Colinérgicos/administração & dosagem , Glicogênio Sintase Quinase 3 beta , Imunotoxinas/administração & dosagem , Injeções Intraventriculares , Cloreto de Lítio/farmacologia , Masculino , N-Glicosil Hidrolases , Fármacos Neuroprotetores/farmacologia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Proteínas Inativadoras de Ribossomos Tipo 1 , Saporinas , Proteínas tau/metabolismo
6.
J Neuroendocrinol ; 15(1): 24-32, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12535166

RESUMO

Neuropeptide FF (NPFF), a morphine modulatory peptide, is emerging as an important neuromodulator in the context of central autonomic and neuroendocrine regulation. NPFF immunoreactivity and receptors have been identified in discrete autonomic regions within the brain and spinal cord, including the hypothalamic paraventricular nucleus (PVN). In this study, we examined the effects of intracerebroventricular (i.c.v.) administration of NPFF on activation of chemically identified PVN neurones that project to the brainstem nucleus of the solitary tract (NTS). In conscious rats, i.c.v. NPFF at a dose of 10 micro g, but not 8 micro g, caused an increase in arterial blood pressure. Immunohistochemical analysis revealed a dose-dependent increase in activated (Fos positive) PVN neurones following i.c.v. NPFF administration compared to controls receiving i.c.v. saline. Activated PVN neurones were located predominantly in the parvocellular compartment of the nucleus with relatively few Fos positive cells in the magnocellular subdivision. Chemical identification of activated neurones revealed significant number of activated cells to be oxytocin positive, whereas only few vasopressin, tyrosine hydroxylase (TH) or corticotrophin-releasing factor (CRF) neurones were double-labelled. Injection of the retrograde tracer fluorogold into the NTS resulted in labelling of significant numbers of parvocellular oxytocin, but not vasopressin, TH or CRF, PVN neurones. We conclude that centrally administered NPFF stimulates brainstem-projecting oxytocin PVN neurones. Oxytocin released from terminals within the NTS oxytocin thus modulate the activity of ascending visceral autonomic pathways that synapse initially within the NTS.


Assuntos
Tronco Encefálico/citologia , Antagonistas de Entorpecentes/farmacologia , Oligopeptídeos/farmacologia , Ocitocina/fisiologia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Estilbamidinas , Animais , Pressão Sanguínea/efeitos dos fármacos , Corantes Fluorescentes , Injeções Intraventriculares , Masculino , Vias Neurais , Núcleo Hipotalâmico Paraventricular/fisiologia , Ratos , Ratos Sprague-Dawley
7.
J Pharmacol Exp Ther ; 303(1): 188-95, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12235250

RESUMO

Nociceptin/orphanin FQ (N/OFQ) is an endogenous opioid-like heptadecapeptide that plays an important role in a variety of physiological functions. N/OFQ and its receptor opioid receptor-like orphan receptor-1 are abundant in the diagonal band of Broca (DBB), a basal forebrain nucleus where the loss of cholinergic neurons is linked to memory and spatial learning deficits. In the whole animal, central injections of N/OFQ have been shown to disrupt spatial learning. In this study, we investigated the basis for these behavioral observations by examining the cellular effects of N/OFQ on chemically identified DBB neurons. Whole cell patch-clamp recordings were performed on enzymatically dissociated DBB neurons. Under voltage-clamp conditions, bath application of N/OFQ (10 pM-1 microM) resulted in a dose-dependent depression of whole cell currents. Single cell reverse transcription-polymerase chain reaction analysis identified cholinergic and fewer GABAergic cells to be N/OFQ-responsive. [Nphe(1)]nociceptin-(1-13)-NH(2) and CompB (J-113397) antagonized the N/OFQ response, but both compounds also displayed partial agonist activity. Using a combination of channel blockers we determined that the effects of N/OFQ were mediated via a suite of Ca(2+) (N- and L-type) and Ca(2+)-dependent K(+) (iberiotoxin-sensitive) conductances. In addition, biophysical analysis of voltage subtraction protocols revealed that N/OFQ reduces transient outward and the delayed rectifier K(+) currents. Because N-type and L-type Ca(2+) channels are important in the context of neurotransmitter release, our observations indicate that N/OFQ inhibition of Ca(2+)-dependent conductances in cholinergic neurons would be expected to result in depression of acetylcholine release, which may explain the behavioral actions of N/OFQ in the brain.


Assuntos
Neurônios/fisiologia , Peptídeos Opioides/farmacologia , Prosencéfalo/fisiologia , Receptores Opioides/fisiologia , Acetilcolina/fisiologia , Actinas/genética , Animais , Sequência de Bases , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/fisiologia , Primers do DNA , Condutividade Elétrica , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neurônios/efeitos dos fármacos , Nimodipina/farmacologia , Técnicas de Patch-Clamp , Potássio/fisiologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ácido gama-Aminobutírico/fisiologia , ômega-Conotoxina GVIA/farmacologia , Receptor de Nociceptina , Nociceptina
8.
J Neurophysiol ; 86(3): 1312-20, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11535679

RESUMO

The deposition of amyloid beta-protein (Abeta) in the brain and the loss of cholinergic neurons in the basal forebrain are two pathological hallmarks of Alzheimer's disease (AD). Although the mechanism of Abeta neurotoxicity is unknown, these cholinergic neurons display a selective vulnerability when exposed to this peptide. In this study, application of Abeta(25-35) or Abeta(1-40) to acutely dissociated rat neurons from the basal forebrain nucleus diagonal band of Broca (DBB), caused a decrease in whole cell voltage-activated currents in a majority of cells. This reduction in whole cell currents occurs through a modulation of a suite of potassium conductances including calcium-activated potassium (I(C)), the delayed rectifier (I(K)), and transient outward potassium (I(A)) conductances, but not calcium or sodium currents. Under current-clamp conditions, Abeta evoked an increase in excitability and a loss of accommodation in cholinergic DBB neurons. Using single-cell RT-PCR technique, we determined that Abeta actions were specific to cholinergic, but not GABAergic DBB neurons. Abeta effects on whole cell currents were occluded in the presence of membrane-permeable protein tyrosine kinase inhibitors, genistein and tyrphostin B-44. Our data indicate that the Abeta actions on specific potassium conductances are modulated through a protein tyrosine kinase pathway and that these effects are selective to cholinergic but not GABAergic cells. These observations provide a cellular basis for the selectivity of Abeta neurotoxicity toward cholinergic basal forebrain neurons that are at the epicenter of AD pathology.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Colina O-Acetiltransferase/genética , Feixe Diagonal de Broca/citologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fragmentos de Peptídeos/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Actinas/genética , Animais , Cálcio/farmacologia , Charibdotoxina/farmacologia , Canais de Potássio de Retificação Tardia , Inibidores Enzimáticos/farmacologia , Expressão Gênica/fisiologia , Genisteína/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Fenótipo , Fosforilação , Potássio/metabolismo , Canais de Potássio/fisiologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sódio/metabolismo , Tetraetilamônio/farmacologia , Tirosina/metabolismo , Tirfostinas/farmacologia
9.
J Chem Neuroanat ; 21(2): 171-9, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11312058

RESUMO

Neuropeptide FF (NPFF), a morphine modulatory peptide, has been identified within discrete autonomic regions in the brainstem and hypothalamus. Triple fluorescence labelling was employed to identify collateral branching projections of NPFF neurons located within the nucleus tractus solitarius (NTS) and in the region of the hypothalamus between the dorsomedial and ventromedial hypothalamus. Injections of two retrograde tracers, rhodamine- and fluorescein-labelled latex microspheres into the pontine parabrachial nucleus (PBN) and the ventrolateral medulla resulted in labelling of NPFF neurons in the NTS that contained one (double-labelled) or both (triple-labelled) tracers. Within the NTS, most double- and triple-labelled NPFF neurons were localized at the level of the area postrema or just rostral to it and within the medial and dorsomedial subdivisions of the nucleus. Injections of tracers into the PBN and hypothalamic paraventricular nucleus revealed double- and triple-labelled NPFF neurons, a majority of which were located in a zone between the dorsomedial and ventromedial hypothalamus. These results indicate that NPFF neurons in the brainstem and hypothalamus may simultaneously transmit signals to their target nuclei in the brainstem and forebrain. This coordinated signalling may lead to synchronized responses of NPFF target sites and provide insights into the role of this peptide in cardiovascular and nociceptive responses.


Assuntos
Tronco Encefálico/fisiologia , Hipotálamo/fisiologia , Vias Neurais/fisiologia , Oligopeptídeos/fisiologia , Animais , Imuno-Histoquímica , Masculino , Bulbo/citologia , Bulbo/fisiologia , Microscopia de Fluorescência , Microesferas , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Ratos , Ratos Sprague-Dawley , Núcleo Solitário/citologia , Núcleo Solitário/fisiologia
10.
J Neurophysiol ; 84(2): 744-51, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10938301

RESUMO

The pontine parabrachial nucleus (PBN) receives both opioid and Neuropeptide FF (NPFF) projections from the lower brain stem and/or the spinal cord. Because of this anatomical convergence and previous evidence that NPFF displays both pro- and anti-opioid activities, this study examined the synaptic effects of NPFF in the PBN and the mechanisms underlying these effects using an in vitro brain slice preparation and the nystatin-perforated patch-clamp recording technique. Under voltage-clamp conditions, NPFF reversibly reduced the evoked excitatory postsynaptic currents (EPSCs) in a dose-dependent fashion. This effect was not accompanied by apparent changes in the holding current, the current-voltage relationship or alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-induced inward currents in the PBN cells. When a paired-pulse protocol was used, NPFF increased the ratio of these synaptic currents. Analysis of miniature EPSCs showed that NPFF caused a rightward shift in the frequency-distribution curve, whereas the amplitude-distribution curve remained unchanged. Collectively, these experiments indicate that NPFF reduces the evoked EPSCs through a presynaptic mechanism of action. The synaptic effects induced by NPFF (5 microM) could not be blocked by the specific mu-opioid receptor antagonist, D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH(2) (1 microM), but application of delta-opioid receptor antagonist Tyr-Tic-Phe-Phe (5 microM) almost completely prevented effects of NPFF. Moreover, the delta-opioid receptor agonist, Deltorphin (1 microM), mimicked the effects as NPFF and also occluded NPFF's actions on synaptic currents. These results indicate that NPFF modulates excitatory synaptic transmission in the PBN through an interaction with presynaptic delta-opioid receptors. These observations provide a cellular basis for NPFF enhancement of the antinociceptive effects consequent to central activation of delta-opioid receptors.


Assuntos
Antagonistas de Entorpecentes/farmacologia , Oligopeptídeos/farmacologia , Ponte/fisiologia , Terminações Pré-Sinápticas/fisiologia , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Tetra-Hidroisoquinolinas , Analgésicos Opioides/farmacologia , Animais , Relação Dose-Resposta a Droga , Condutividade Elétrica , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Encefalinas/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Antagonistas GABAérgicos/farmacologia , Técnicas In Vitro , Ligantes , Masculino , Naloxona/farmacologia , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Picrotoxina/farmacologia , Ponte/química , Ponte/citologia , Potássio/metabolismo , Terminações Pré-Sinápticas/química , Ratos , Ratos Sprague-Dawley , Tetrodotoxina/farmacologia
11.
JAMA ; 282(21): 2019-26, 1999 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-10591384

RESUMO

CONTEXT: Recombinant tissue-type plasminogen activator (rt-PA) improves outcomes for patients with acute ischemic stroke, but current approved use is limited to within 3 hours of symptom onset. This restricts the number of patients who can be treated, since most stroke patients present more than 3 hours after symptom onset. OBJECTIVE: To test the efficacy and safety of rt-PA in patients with acute ischemic stroke when administered between 3 and 5 hours after symptom onset. DESIGN: The Alteplase ThromboLysis for Acute Noninterventional Therapy in Ischemic Stroke (ATLANTIS) study is a phase 3, placebo-controlled, double-blind randomized study conducted between December 1993 and July 1998, with up to 90 days of follow-up. SETTING: One hundred forty university and community hospitals in North America. PATIENTS: An intent-to-treat population of 613 acute ischemic stroke patients was enrolled, with 547 of these treated as assigned within 3 to 5 hours of symptom onset. A total of 39 others were treated within 3 hours of symptom onset, 24 were treated more than 5 hours after symptom onset, and 3 never received any study drug. INTERVENTION: Administration of 0.9 mg/kg of rt-PA (n = 272) or placebo (n = 275) intravenously over 1 hour. MAIN OUTCOME MEASURES: Primary efficacy was an excellent neurologic recovery at day 90 (National Institutes of Health Stroke Scale [NIHSS] score of < or =1); secondary end points included excellent recovery on functional outcome measures (Barthel index, modified Rankin scale, and Glasgow Outcome Scale) at days 30 and 90. Serious adverse events were also assessed. RESULTS: In the target population, 32% of the placebo and 34% of rt-PA patients had an excellent recovery at 90 days (P = .65). There were no differences on any of the secondary functional outcome measures. In the first 10 days treatment with rt-PA significantly increased the rate of symptomatic intracerebral hemorrhage (ICH) (1.1% vs 7.0% [P<.001]), a symptomatic ICH (4.7% vs 11.4% [P = .004]), and fatal ICH (0.3% vs 3.0% [P<.001]). Mortality at 90 days was 6.9% with placebo and 11.0% with rt-PA (P = .09). Results in the intent-to-treat population were similar. CONCLUSIONS: This study found no significant rt-PA benefit on the 90-day efficacy end points in patients treated between 3 and 5 hours. The risk of symptomatic ICH increased with rt-PA treatment. These results do not support the use of intravenous rt-PA for stroke treatment beyond 3 hours.


Assuntos
Transtornos Cerebrovasculares/tratamento farmacológico , Fibrinolíticos/uso terapêutico , Terapia Trombolítica , Ativador de Plasminogênio Tecidual/uso terapêutico , Idoso , Isquemia Encefálica/tratamento farmacológico , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Análise de Sobrevida , Fatores de Tempo , Resultado do Tratamento
12.
Neuroscience ; 94(3): 785-95, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10579569

RESUMO

We examined modulation of ionic currents by Zn2+ in acutely dissociated neurons from the rat's horizontal limb of the diagonal band of Broca using the whole-cell patch-clamp technique. Application of 50 microM Zn2+ increased the peak amplitude of the transiently activated potassium current, I(A) (at + 30 mV), from 2.20+/-0.08 to 2.57+/-0.11 nA (n = 27). This response was reversible and could be repeated in 0 Ca2+/1 microM tetrodotoxin (n = 15). Zn2+ shifted the inactivation curve to the right, resulting in a shift in the half-inactivation voltage from 76.4+/-2.2 to -53.4+/-2.0 mV (n = 11), with no effect on the voltage dependence of activation gating (n = 15). There was no significant difference in the time to peak under control conditions (7.43+/-0.35 ms, n = 14) and in the presence of Zn2+ (8.20+/-0.57 ms, n = 14). Similarly, the time constant of decay of I(A) (tau(d)) at + 30 mV showed no difference (control: 38.68+/-3.68 ms, n = 15; Zn2+: 38.48+/-2.85 ms, n = 15). I(A) was blocked by 0.5-1 mM 4-aminopyridine. In contrast to its effects on I(A), Zn2+ reduced the amplitude of the delayed rectifier potassium current (I(K)). The reduction of outward K+ currents was reproducible when cells were perfused with 1 microM tetrodotoxin in a 0 Ca2+ external solution. The amplitude of the steady-state outward currents at +30 mV under these conditions was reduced from 6.40+/-0.23 (control) to 5.76+/-0.18 nA in the presence of Zn2+ (n = 16). The amplitudes of peak sodium currents (INa) were not significantly influenced (n = 10), whereas barium currents (I(Ba)) passing through calcium channels were potently modulated. Zn2+ reversibly reduced I(Ba) at -10 mV by approximately 85% from -2.06+/-0.14 nA under control conditions to -0.30+/-0.10 nA in the presence of Zn2+ (n = 14). Further analyses of Zn2+ effects on specific calcium channels reveals that it suppresses all types of high-voltage-activated Ca2+ currents. Under current-clamp conditions, application of Zn2+ resulted in an increase in excitability and loss of accommodation (n = 13), which appears to be mediated through its effects on Ca2+-dependent conductances.


Assuntos
Feixe Diagonal de Broca/fisiologia , Potenciais Evocados/efeitos dos fármacos , Neurônios/fisiologia , Canais de Potássio/fisiologia , Zinco/farmacologia , 4-Aminopiridina/farmacologia , Animais , Cálcio/metabolismo , Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Cátions Bivalentes/farmacologia , Feixe Diagonal de Broca/efeitos dos fármacos , Técnicas In Vitro , Masculino , Neurônios/efeitos dos fármacos , Nimodipina/farmacologia , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sódio/metabolismo , Tetrodotoxina/farmacologia , ômega-Conotoxinas/farmacologia
13.
J Neuroendocrinol ; 11(9): 715-23, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10447810

RESUMO

Neuropeptide Y (NPY) is found in autonomic neurones and participates in regulation of autonomic functions. To investigate the role of NPY in the stress response in normo- and hypertensive rats, activation of brainstem and arcuate nucleus (ARC) NPY neurones and levels of NPY mRNA in the ARC were measured in response to restraint stress in adult spontaneously hypertensive rats (SHRs) and two strains of normotensive rats. Controls from each strain were not restrained. Sections of the brain were prepared for Fos immunohistochemistry and NPY in-situ hybridization to identify activated NPY neurones in the nucleus of the tractus solitarii (NTS), ventrolateral medulla (VLM), and ARC. NPY mRNA levels were quantified in the ARC. In the NTS and VLM of restrained rats, approximately 33% and 75%, respectively, of NPY neurones were activated. No differences among strains were found. In the ARC, about 36% of neurones activated by restraint contained NPY mRNA with no differences found among strains. In unrestrained rats, NPY mRNA levels were significantly elevated in SHRs compared to the normotensive rats. Restraint led to significant decreases in mRNA levels in all strains and mRNA levels among strains were no longer different from one another. These data show that NPY likely participates as a neurotransmitter in the autonomic pathways utilized during stress and originating in the NTS, VLM, and ARC. On the other hand, the decreased gene expression of NPY in the ARC in response to restraint stress argues against a role for activation of autonomic pathways or the hypothalamo-pituitary-adrenal (HPA) axis by NPY from the ARC of stressed rats. The elevated NPY gene expression in resting SHRs compared to normotensive rats is abrogated after restraint, suggesting that this gene is differentially regulated in SHRs compared to normotensive rats.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Tronco Encefálico/metabolismo , Hipertensão/metabolismo , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Restrição Física , Estresse Fisiológico/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/patologia , Tronco Encefálico/patologia , Hipertensão/patologia , Imuno-Histoquímica , Hibridização In Situ , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Ratos Sprague-Dawley
14.
Neuroscience ; 89(3): 863-71, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10199619

RESUMO

The present study examined the electrophysiological and kinetic properties of a hyperpolarizing-activated current in neurons located in the lateral parabrachial nucleus. We investigated whether differences observed in the shape of action potential afterhyperpolarizations in lateral parabrachial nucleus neurons, and the ability of these neurons to accommodate, correlated with the presence of this current. A voltage-activated inwardly rectifying current that increased in amplitude with hyperpolarization was observed in 83% of the neurons examined. Under voltage-clamp recording conditions, this current activated at about -70 mV, was half-activated at -96.5 mV and was blocked by bath application of 2 mM cesium, but not by 100 microM barium. In the current-clamp mode, activation of this current resulted in a transient increase in neuronal excitability at the termination of the more negative current injections. The presence of this current did not correlate with specific action potential characteristics or the ability of lateral parabrachial nucleus neurons to accommodate, as the kinetics and voltage-dependent characteristics are such that this hyperpolarizing-activated current does not affect neuronal excitability at or near the resting membrane potential. However, it may act as an important depolarizing mechanism that prevents neurons from becoming unresponsive when they are excessively hyperpolarized, These results provide evidence that the majority of neurons located in the lateral parabrachial nucleus exhibit a mixed cationic current, which is consistent with the H-current or Q-current. This current may function as a negative feedback mechanism that is activated under conditions of intense hyperpolarization so as to ensure that lateral parabrachial nucleus neurons are in a more suitable state of readiness to respond appropriately to afferent input.


Assuntos
Tronco Encefálico/fisiologia , Neurônios/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Bário/farmacologia , Relógios Biológicos/efeitos dos fármacos , Relógios Biológicos/fisiologia , Césio/farmacologia , Eletrofisiologia , Retroalimentação , Ativação do Canal Iônico , Transporte de Íons , Masculino , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley
15.
Brain Res Mol Brain Res ; 65(1): 70-9, 1999 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-10036309

RESUMO

To test the hypothesis that chronically elevated sympathetic drive is associated with hyperreactiveness of autonomic centers in the brain to stress, adult spontaneously hypertensive rats (SHRs) and two strains of normotensive rats (Wistar Kyoto [WKY] and Sprague Dawley [SD] rats) were acutely exposed to restraint stress; controls from each strain were not stressed. Brain sections were prepared for Fos immunohistochemistry to identify activated neurons in the paraventricular nucleus of the hypothalamus, Barrington's nucleus of the pons, nucleus of the tractus solitarius, and ventrolateral medulla, or for combined Fos immunohistochemistry and corticotropin-releasing factor (CRF) in situ hybridization in the paraventricular nucleus and Barrington's nucleus. Restraint led to increased activation of neurons in all nuclei. Strain differences were found only in the caudal and rostral paraventricular nucleus where restraint resulted in greater numbers of activated neurons in SHRs compared to either normotensive strain. Levels of CRF mRNA in Barrington's nucleus of unrestrained rats were similar among strains. After restraint, mRNA levels and double labeled neurons were increased in Barrington's nucleus of SHRs. In unstressed rats, CRF mRNA levels were elevated in some regions of the paraventricular nucleus in SHRs. After restraint, mRNA levels increased throughout the paraventricular nucleus of SHRs. Significantly greater numbers of double labeled neurons were found in the dorsolateral medial and ventral medial parvocellular paraventricular nucleus of stressed SHRs compared to WKY or SD rats. These data show that chronic elevation in sympathetic activity, present in SHRs, is associated with hyperreactiveness of the paraventricular and Barrington's nucleus including recruitment of neurons to express CRF, and may have important implications for the response of the hypothalamo-pituitary-adrenal axis during stress.


Assuntos
Hormônio Liberador da Corticotropina/genética , Neurônios/fisiologia , Ratos Endogâmicos SHR/fisiologia , Estresse Fisiológico/fisiopatologia , Animais , Pressão Sanguínea/fisiologia , Expressão Gênica/fisiologia , Hipertensão/fisiopatologia , Neurônios/química , Núcleo Hipotalâmico Paraventricular/química , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos WKY , Ratos Sprague-Dawley , Restrição Física , Núcleo Solitário/química , Núcleo Solitário/citologia , Núcleo Solitário/fisiologia , Sistema Nervoso Simpático/citologia , Sistema Nervoso Simpático/fisiologia
16.
J Neurophysiol ; 81(1): 234-46, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9914284

RESUMO

Whole cell recordings were performed on acutely dissociated neurons from the horizontal limb of the diagonal band of Broca (hDBB) from rats to elucidate the ionic mechanisms of action of neurotensin. Neurotensin caused a decrease in whole cell voltage-activated outward currents and failed to elicit a response when Ca2+ influx was blocked by changing the external solution to the one containing 0 mM Ca2+ and 50 microM Cd2+, suggesting the involvement of Ca2+-dependent conductances. Charybdotoxin, a specific blocker of voltage-sensitive calcium-activated K+ channels (IC), caused a decrease in outward currents comparable with that caused by blocking calcium influx and occluded the neurotensin-induced decrease in outward currents. Similarly, 50 microM tetraethylammonium ions also blocked the neurotensin response. Also neurotensin reduced whole cell barium currents (IBa) and calcium currents (ICa). Amiloride and omega-conotoxin GVIA, but not nimodipine, were able to eliminate the neurotensin-induced decrease in IBa. Thus T- and N- but not L-type calcium channels are subject to modulation by neurotensin, and this may account for its effects on IC. The predicted changes in action potential as a result of the blockade of currents through calcium channels culminating into changes in IC were confirmed in the bridge current-clamp recordings. Specifically, neurotensin application led to depolarization of the resting membrane potential, broadening of spike and a decrease in afterhyperpolarization and accommodation. These alterations in action potential characteristics that resulted in increased firing rate and excitability of the hDBB neurons also were produced by application of charybdotoxin. Neurotensin effects on these properties were occluded by 2 - [(1 - 7 - chloro - 4 - quinolinyl) - 5 - (2, 6 - di - methoxyphenyl) pyrazol-3-yl) carbonylamino] tricyclo (3.3.1.1.)decan-2-carboxylic acid, a nonpeptide high-affinity neurotensin receptor antagonist. Neurotensin blockade of IC, possibly through ICa, is a potential physiological mechanism whereby this peptide may evoke alterations in the cortical arousal, sleep-wake cycle, and theta rhythm.


Assuntos
Lobo Frontal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurotensina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Apamina/farmacologia , Cálcio/metabolismo , Cálcio/fisiologia , Bloqueadores dos Canais de Cálcio/farmacologia , Estimulação Elétrica , Eletrofisiologia , Lobo Frontal/citologia , Técnicas In Vitro , Masculino , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Pirazóis/farmacologia , Quinolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Neurotensina/antagonistas & inibidores , Compostos de Tetraetilamônio/farmacologia
17.
J Comp Neurol ; 402(2): 210-21, 1998 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-9845244

RESUMO

Neuropeptide FF (NPFF), a morphine modulatory peptide, is localized within discrete autonomic regions including the brainstem nucleus tractus solitarius (NTS) and the parabrachial nucleus (PBN). We investigated the activation of NPFF neurons in the NTS of rats induced by cardiovascular challenge and centrally generated opiate withdrawal. For hypotensive stimulation, we used systemic infusions of sodium nitroprusside (NP) or hemorrhage (HEM), and hypertension was achieved by intravenous phenylephrine (PHENYL) or angiotensin II (AII). In rats that received continuous intracerebroventricular injections of morphine, intraperitoneal injections of naloxone precipitated behavioural signs of opioid withdrawal. Activated NTS neurons were identified by using a combined immunohistochemistry for Fos and NPFF, and neurons projecting to the PBN were determined with a retrograde tracer. HEM, administration of vasoactive drugs, and opiate withdrawal produced a very robust activation of NTS neurons. In NP and HEM groups, 25.6+/-3.2% and 7.6+/-1.3% of NPFF neurons were activated, respectively. Lesser numbers of NPFF neurons were activated in the PHENYL (4.6+/-1.6%) and AII (2.4+/-0.8%) groups. However, following opiate withdrawal, virtually no Fos expression was observed in NPFF neurons. NPFF neurons activated during NP infusion constituted the largest number of cells projecting to the PBN. This study shows that NPFF neurons in NTS that project to the PBN respond selectively to NP as opposed to other cardiovascular challenges or opiate withdrawal. These data support an emerging and important role for NPFF in the context of central cardiovascular regulation.


Assuntos
Hemodinâmica/fisiologia , Hipertensão/fisiopatologia , Hipotensão/fisiopatologia , Morfina/efeitos adversos , Naloxona/farmacologia , Neurônios/fisiologia , Oligopeptídeos/análise , Núcleo Solitário/fisiologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Angiotensina II/toxicidade , Animais , Masculino , Dependência de Morfina/tratamento farmacológico , Naloxona/uso terapêutico , Neurônios/química , Nitroprussiato/toxicidade , Fenilefrina/toxicidade , Ratos , Ratos Sprague-Dawley , Choque Hemorrágico/fisiopatologia , Núcleo Solitário/ultraestrutura
18.
J Neuroendocrinol ; 10(6): 453-9, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9688348

RESUMO

Using the immunohistochemical localization of the protein product of the immediate early gene, c-fos, to localize activated neurons in the paraventricular nucleus of the hypothalamus (PVN), we studied the chemical phenotypes of neurons activated by circulating angiotensin II (AII). We determined the proportions of activated PVN neurons that expressed AII type I receptor-like immunoreactivity (AT1-L) or the neurohormones vasopressin (VP) and oxytocin (OXY). In addition, we identified activated PVN neurons that putatively produce nitric oxide (NO) on the basis of histochemical staining for nicotinamide adenine dinucleotide phosphate diaphorase (NADPH-d). Conscious rats received intravenous AII infusions at a rate sufficient to elevate mean arterial pressure by 40-60 mmHg for 90 min; control rats received infusions of vehicle. Brains were prepared for double immunohistochemistry [Fos-like immunoreactivity (FLI)/AT1-L, FLI/VP or FLI/OXY] or FLI/ NADPH-d histochemistry. Systemic AII infusions led to activation of 149+/-14 PVN neurons per section. In contrast, control animals showed activation of 21+/-6 PVN neurons per section. AII infusions elicited the activation of the following numbers of chemically identified PVN neurons per section: AT1-L, 24+/-5; VP, 26+/-5; OXY, 11+/-2; NADPH-d, 22+/-4. Control animals had few activated PVN neurons per section. For each of the chemically identified populations of PVN neurons, the following proportions were activated: AT1-L, 12.5%; VP, 15.2%; OXY, 7.2%; NADPH-d, 17.3%. The results suggest that PVN neurons producing the AT1 receptor, VP, OXY, and NO, participate in the mediation of the central responses to circulating AII.


Assuntos
Angiotensina II/farmacologia , Neurônios/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Animais , Imuno-Histoquímica , Masculino , NADPH Desidrogenase/metabolismo , Neurônios/enzimologia , Neurônios/metabolismo , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Angiotensina/metabolismo , Vasopressinas/metabolismo
19.
Neuroscience ; 81(1): 57-67, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9300401

RESUMO

The actions of vasopressin on acutely dissociated neurons within the rat horizontal limb of the diagonal band of Broca were examined using the whole-cell patch-clamp technique. Vasopressin elicited two distinct responses in 45 of 62 neurons. In one group of cells, 300 nM vasopressin decreased voltage-activated outward currents (26/45 cells) whereas in a second group, vasopressin increased outward currents (19/45 cells). The vasopressin-mediated decrease in outward currents was blocked by 1 microM Manning compound, a V1 receptor antagonist, suggesting that this response was mediated via V1 receptors. In contrast, the vasopressin-induced increase in outward current was blocked by 1 microM d(CH2)5)1,D-Ile2,Ile4,Arg8,Ala9, a V2 receptor antagonist, indicating that V2 receptor activation underlies this second response. When cells were perfused with 0 Ca2+/50 microM Cd2+, application of vasopressin did not cause any change in voltage-activated outward currents, suggesting that vasopressin modulates a calcium-dependent conductance. In the presence of 25 nM charybdotoxin, an Ic channel antagonist, vasopressin application did not influence outward currents, indicating that vasopressin modulates Ic. Currents through voltage-gated calcium channels which are responsible for activation of Ic were unaffected by vasopressin, suggesting a direct effect of vasopressin on Ic channels. These observations indicate a differential modulation of Ic channels by vasopressin via V1 and V2 receptors in the horizontal limb of the diagonal band of Broca. Our data also demonstrate the ionic mechanisms whereby vasopressin may act at V1 for V2 receptors to influence the excitability of the horizontal limb of the diagonal band of Broca neurons.


Assuntos
Cálcio/farmacologia , Lobo Frontal/química , Canais de Potássio/agonistas , Receptores de Vasopressinas/fisiologia , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos , Arginina Vasopressina/agonistas , Arginina Vasopressina/antagonistas & inibidores , Arginina Vasopressina/fisiologia , Cálcio/metabolismo , Charibdotoxina/farmacologia , Lobo Frontal/citologia , Lobo Frontal/fisiologia , Masculino , Neurônios/química , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Técnicas de Patch-Clamp , Canais de Potássio/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Vasopressinas/agonistas , Vasopressinas/farmacologia
20.
J Comp Neurol ; 385(2): 285-96, 1997 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-9268128

RESUMO

To investigate the involvement of neuronal nitric oxide (NO) in the response of the brain to changes in blood pressure, we studied the activation of putative NO-producing neurons in the paraventricular nucleus of the hypothalamus (PVN) in rats whose mean arterial pressures (MAPs) were decreased by 40-50% with hemorrhage (HEM) or infusion of sodium nitroprusside (NP). Activation was assessed on the basis of expression of the immediate early gene, c-fos; putative NO-producing neurons were identified with the histochemical stain for nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d); and the proportions of neurons projecting to the nucleus of the tractus solitarius (NTS) and/or caudal ventrolateral medulla (CVLM) were determined with retrograde tracing techniques. No differences were found for results obtained from HEM and NP animals. Three to four percent of activated PVN neurons projected to the NTS or CVLM. Conversely, approximately 33% and 16% of neurons projecting to the NTS and CVLM, respectively, were activated. About 43% of NADPH-d neurons in the PVN were activated. Of PVN neurons projecting to the NTS or CVLM, 38% and 32%, respectively, were NADPH-d positive. About 11% of NADPH-d PVN neurons projected to the NTS or CVLM. An average of 3 NADPH-d neurons per section were activated and projected to either target. Finally, 7 PVN cells per section sent collateral branches to the NTS and CVLM; 2 or 3 of these cells per section were also activated by decreases in arterial pressure. No NADPH-d cells were found that sent collateral branches to the NTS and CVLM. This study shows that decreases in MAP activate PVN neurons that project, singly and through collaterals, to the NTS and CVLM. A relatively high proportion of the singly projecting neurons is NADPH-d positive. These results support the contention that descending projections from the PVN to the brainstem play an important role in the physiological response to decreases in arterial pressure and suggest that NO may participate in this response.


Assuntos
Tronco Encefálico/anatomia & histologia , Hipotensão/fisiopatologia , Hipotálamo/anatomia & histologia , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/anatomia & histologia , Animais , Tronco Encefálico/fisiologia , Hipotálamo/fisiologia , Masculino , Óxido Nítrico/metabolismo , Nitroprussiato/farmacologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Ratos , Ratos Sprague-Dawley , Vasodilatadores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...