Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Am J Physiol Cell Physiol ; 310(5): C329-36, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26659724

RESUMO

Human ether-a-go-go-related gene (hERG) channels conduct delayed rectifier K(+) current. However, little information is available on physiological situations affecting hERG channel protein and function. In the present study we examined the effects of intermittent hypoxia (IH), which is a hallmark manifestation of sleep apnea, on hERG channel protein and function. Experiments were performed on SH-SY5Y neuroblastoma cells, which express hERG protein. Cells were exposed to IH consisting of alternating cycles of 30 s of hypoxia (1.5% O2) and 5 min of 20% O2. IH decreased hERG protein expression in a stimulus-dependent manner. A similar reduction in hERG protein was also seen in adrenal medullary chromaffin cells from IH-exposed neonatal rats. The decreased hERG protein was associated with attenuated hERG K(+) current. IH-evoked hERG protein degradation was not due to reduced transcription or increased proteosome/lysomal degradation. Rather it was mediated by calcium-activated calpain proteases. Both COOH- and NH2-terminal sequences of the hERG protein were the targets of calpain-dependent degradation. IH increased reactive oxygen species (ROS) levels, intracellular Ca(2+) concentration ([Ca(2+)]i), calpain enzyme activity, and hERG protein degradation, and all these effects were prevented by manganese-(111)-tetrakis-(1-methyl-4-pyridyl)-porphyrin pentachloride, a membrane-permeable ROS scavenger. These results demonstrate that activation of calpains by ROS-dependent elevation of [Ca(2+)]i mediates hERG protein degradation by IH.


Assuntos
Calpaína/metabolismo , Canais de Potássio Éter-A-Go-Go/genética , Processamento de Proteína Pós-Traducional/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Medula Suprarrenal/metabolismo , Animais , Animais Recém-Nascidos , Hipóxia Celular , Células Cultivadas , Canal de Potássio ERG1 , Ativação Enzimática , Feminino , Humanos , Masculino , Proteólise , Ratos Sprague-Dawley
2.
J Neurosci ; 29(15): 4903-10, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19369559

RESUMO

Respiratory motoneuron response to hypoxia is reflex in nature and carotid body sensory receptor constitutes the afferent limb of this reflex. Recent studies showed that repetitive exposures to hypoxia evokes long term facilitation of sensory nerve discharge (sLTF) of the carotid body in rodents exposed to chronic intermittent hypoxia (CIH). Although studies with anti-oxidants suggested the involvement of reactive oxygen species (ROS)-mediated signaling in eliciting sLTF, the source of and the mechanisms associated with ROS generation have not yet been investigated. We tested the hypothesis that ROS generated by NADPH oxidase (NOX) mediate CIH-evoked sLTF. Experiments were performed on ex vivo carotid bodies from rats and mice exposed either to 10 d of CIH or normoxia. Acute repetitive hypoxia evoked a approximately 12-fold increase in NOX activity in CIH but not in control carotid bodies, and this effect was associated with upregulation of NOX2 mRNA and protein, which was primarily localized to glomus cells of the carotid body. sLTF was prevented by NOX inhibitors and was absent in mice deficient in NOX2. NOX activation by CIH required 5-HT release and activation of 5-HT(2) receptors coupled to PKC signaling. Studies with ROS scavengers revealed that H(2)O(2) generated from O(2).(-) contributes to sLTF. Priming with H(2)O(2) elicited sLTF of carotid bodies from normoxic control rats and mice, similar to that seen in CIH-treated animals. These observations reveal a novel role for NOX-induced ROS signaling in mediating sensory plasticity of the carotid body.


Assuntos
Corpo Carotídeo/enzimologia , Hipóxia Encefálica/enzimologia , NADPH Oxidases/fisiologia , Plasticidade Neuronal/fisiologia , Animais , Corpo Carotídeo/metabolismo , Doença Crônica , Hipóxia Encefálica/metabolismo , Hipóxia Encefálica/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vias Neurais/enzimologia , Vias Neurais/metabolismo , Proteína Quinase C/fisiologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Receptores 5-HT2 de Serotonina/fisiologia , Serotonina/fisiologia
3.
Adv Exp Med Biol ; 648: 307-17, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19536494

RESUMO

Physiological responses to hypoxia either continuous (CH) or intermittent (IH) depend on the O(2)-sensing ability of the peripheral arterial chemoreceptors, especially the carotid bodies, and the ensuing reflexes play important roles in maintaining homeostasis. The purpose of this article is to summarize the effects of CH and IH on carotid body function and the underlying mechanisms. CH increases baseline carotid body activity and sensitizes the response to acute hypoxia. These effects are associated with hyperplasia of glomus cells and neovascularization. Enhanced hypoxic sensitivity is due to alterations in ion current densities as well as changes in neurotransmitter dynamics and recruitment of additional neuromodulators (endothelin-1, ET-1) in glomus cells. Morphological alterations are in part due to up-regulation of growth factors (e.g. VEGF). Hypoxia-inducible factor-1 (HIF-1), a transcriptional activator might underlie the remodeling of carotid body structure and function by CH. Chronic IH, on the other hand, is associated with recurrent apneas in adults and premature infants. Two major effects of chronic IH on the adult carotid body are sensitization of the hypoxic sensory response and long-lasting increase in baseline activity i.e., sensory long-term facilitation (LTF) which involve reactive oxygen species (ROS) and HIF-1. In neonates, chronic IH leads to sensitization of the hypoxic response but does not induce sensory LTF. Chronic IH-induced sensitization of the carotid body response to hypoxia increases the likelihood of unstable breathing perpetuating in more number of apneas, whereas sensory LTF may contribute to increased sympathetic tone and systemic hypertension associated with recurrent apneas.


Assuntos
Adaptação Fisiológica , Corpo Carotídeo/fisiologia , Hipóxia/fisiopatologia , Aclimatação , Animais , Humanos , Hipóxia/metabolismo , Fatores de Tempo
4.
Prog Biophys Mol Biol ; 91(3): 249-86, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16137743

RESUMO

This review is divided into three parts: (a) The primary site of oxygen sensing is the carotid body which instantaneously respond to hypoxia without involving new protein synthesis, and is historically known as the first oxygen sensor and is therefore placed in the first section (Lahiri, Roy, Baby and Hoshi). The carotid body senses oxygen in acute hypoxia, and produces appropriate responses such as increases in breathing, replenishing oxygen from air. How this oxygen is sensed at a relatively high level (arterial PO2 approximately 50 Torr) which would not be perceptible by other cells in the body, is a mystery. This response is seen in afferent nerves which are connected synaptically to type I or glomus cells of the carotid body. The major effect of oxygen sensing is the increase in cytosolic calcium, ultimately by influx from extracellular calcium whose concentration is 2 x 10(4) times greater. There are several contesting hypotheses for this response: one, the mitochondrial hypothesis which states that the electron transport from the substrate to oxygen through the respiratory chain is retarded as the oxygen pressure falls, and the mitochondrial membrane is depolarized leading to the calcium release from the complex of mitochondria-endoplasmic reticulum. This is followed by influx of calcium. Also, the inhibitors of the respiratory chain result in mitochondrial depolarization and calcium release. The other hypothesis (membrane model) states that K(+) channels are suppressed by hypoxia which depolarizes the membrane leading to calcium influx and cytosolic calcium increase. Evidence supports both the hypotheses. Hypoxia also inhibits prolyl hydroxylases which are present in all the cells. This inhibition results in membrane K(+) current suppression which is followed by cell depolarization. The theme of this section covers first what and where the oxygen sensors are; second, what are the effectors; third, what couples oxygen sensors and the effectors. (b) All oxygen consuming cells have a built-in mechanism, the transcription factor HIF-1, the discovery of which has led to the delineation of oxygen-regulated gene expression. This response to chronic hypoxia needs new protein synthesis, and the proteins of these genes mediate the adaptive physiological responses. HIF-1alpha, which is a part of HIF-1, has come to be known as master regulator for oxygen homeostasis, and is precisely regulated by the cellular oxygen concentration. Thus, the HIF-1 encompasses the chronic responses (gene expression in all cells of the body). The molecular biology of oxygen sensing is reviewed in this section (Semenza). (c) Once oxygen is sensed and Ca(2+) is released, the neurotransmittesr will be elaborated from the glomus cells of the carotid body. Currently it is believed that hypoxia facilitates release of one or more excitatory transmitters from glomus cells, which by depolarizing the nearby afferent terminals, leads to increases in the sensory discharge. The transmitters expressed in the carotid body can be classified into two major categories: conventional and unconventional. The conventional neurotransmitters include those stored in synaptic vesicles and mediate their action via activation of specific membrane bound receptors often coupled to G-proteins. Unconventional neurotransmitters are those that are not stored in synaptic vesicles, but spontaneously generated by enzymatic reactions and exert their biological responses either by interacting with cytosolic enzymes or by direct modifications of proteins. The gas molecules such as NO and CO belong to this latter category of neurotransmitters and have unique functions. Co-localization and co-release of neurotransmitters have also been described. Often interactions between excitatory and inhibitory messenger molecules also occur. Carotid body contains all kinds of transmitters, and an interplay between them must occur. But very little has come to be known as yet. Glimpses of these interactions are evident in the discussion in the last section (Prabhakar).


Assuntos
Oxigênio/metabolismo , Animais , Monóxido de Carbono/metabolismo , Corpo Carotídeo/metabolismo , Membrana Celular/metabolismo , Humanos , Hipóxia/metabolismo , Fator 1 Induzível por Hipóxia/metabolismo , Mitocôndrias/metabolismo , Modelos Biológicos , Neurotransmissores/metabolismo , Óxido Nítrico/metabolismo , Canais de Potássio/metabolismo
5.
J Appl Physiol (1985) ; 101(6): 1795-802, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16888052

RESUMO

The objective of the present study was to examine the impact of early stages of lung injury on ventilatory control by hypoxia and hypercapnia. Lung injury was induced with intratracheal instillation of bleomycin (BM; 1 unit) in adult, male Sprague-Dawley rats. Control animals underwent sham surgery with saline instillation. Five days after the injections, lung injury was present in BM-treated animals as evidenced by increased neutrophils and protein levels in bronchoalveolar lavage fluid, as well as by changes in lung histology and computed tomography images. There was no evidence of pulmonary fibrosis, as indicated by lung collagen content. Basal core body temperature, arterial Po(2), and arterial Pco(2) were comparable between both groups of animals. Ventilatory responses to hypoxia (12% O(2)) and hypercapnia (7% CO(2)) were measured by whole body plethysmography in unanesthetized animals. Baseline respiratory rate and the hypoxic ventilatory response were significantly higher in BM-injected compared with control animals (P = 0.003), whereas hypercapnic ventilatory response was not statistically different. In anesthetized, spontaneously breathing animals, response to brief hyperoxia (Dejours' test, an index of peripheral chemoreceptor sensitivity) and neural hypoxic ventilatory response were augmented in BM-exposed relative to control animals, as measured by diaphragmatic electromyelograms. The enhanced hypoxic sensitivity persisted following bilateral vagotomy, but was abolished by bilateral carotid sinus nerve transection. These data demonstrate that afferent sensory input from the carotid body contributes to a selective enhancement of hypoxic ventilatory drive in early lung injury in the absence of pulmonary fibrosis and arterial hypoxemia.


Assuntos
Corpo Carotídeo/fisiopatologia , Células Quimiorreceptoras/fisiopatologia , Hipóxia/fisiopatologia , Troca Gasosa Pulmonar , Reflexo , Síndrome do Desconforto Respiratório/fisiopatologia , Doença Aguda , Animais , Hipóxia/complicações , Masculino , Ratos , Ratos Sprague-Dawley , Síndrome do Desconforto Respiratório/complicações
6.
Respir Physiol Neurobiol ; 145(2-3): 135-42, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15705529

RESUMO

Previous studies have shown that glomus cells of the carotid body express 5-hydroxytryptamine (5-HT). The aim of this study was to elucidate the role of 5-HT on the hypoxic sensory response (HSR) of the carotid body. Sensory activity was recorded from multi-fiber (n=16) and single-fiber (n=8) preparations of ex vivo carotid bodies harvested from anesthetized, adult rats. 5-HT (3 microM) had no significant effect on the magnitude or on the onset of the HSR. However, 5-HT consistently prolonged the time necessary for the sensory activity to return to baseline following the termination of the hypoxic challenge. Ketanserin (40 microM), a 5-HT2 receptor antagonist completely prevented 5-HT-induced prolongation of the HSR, whereas had no effect on the control HSR (onset, magnitude, and time for decay without 5-HT). Carotid bodies expressed 5-HT, but hypoxia did not facilitate 5-HT release. These observations suggest that 5-HT is not critical for the HSR of the rat carotid body, but it modulates the dynamics of the HSR via its action on 5-HT2 receptors.


Assuntos
Corpo Carotídeo/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Hipóxia/fisiopatologia , Receptores 5-HT2 de Serotonina/fisiologia , Serotonina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/efeitos da radiação , Animais , Corpo Carotídeo/metabolismo , Corpo Carotídeo/fisiopatologia , Cromatografia Líquida de Alta Pressão , Dopamina/metabolismo , Eletroquímica , Técnicas In Vitro , Ketanserina/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley , Serotonina/metabolismo , Antagonistas do Receptor 5-HT2 de Serotonina , Fatores de Tempo
7.
Brain Res Mol Brain Res ; 59(1): 74-83, 1998 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-9729288

RESUMO

Previous studies have demonstrated that hypoxia stimulates expression of the c-fos gene in intact animals and isolated cells. The purpose of the present study was to assess the functional significance of c-fos activation during hypoxia. Using antisense c-fos strategy, we tested the hypothesis that c-fos is essential for activation of activator protein-1 transcription factor complex (AP-1) and subsequent stimulation of down stream genes such as tyrosine hydroxylase (TH) gene during hypoxia. Experiments were performed on rat pheochromocytoma 12 (PC12) cells. AP-1 activity was determined by a reporter gene assay using a luciferase expression vector driven by two copies of an AP-1 cis-element (AP-1-Luc). Cells transfected with AP-1-Luc construct were exposed to normoxia (21% O2) or to varying intensities and/or durations of hypoxia. AP-1 activity increased in response to hypoxia. The magnitude of the response depended on the intensity and duration of the hypoxic stimulus. Increases in AP-1 activity could not be elicited in neuroblastoma cells, indicating that hypoxia-induced increase in AP-1 activity is a cell selective phenomenon. Antisense c-fos abolished hypoxia-induced AP-1 activation in PC12 cells. Hypoxia increased tyrosine hydroxylase-chloramphenicol acetyl transferase activity (TH-CAT), and antisense c-fos and mutations at AP-1 binding sites in TH promoter abolished this effect. These results provide direct evidence that c-fos is essential for functional activation of AP-1 and subsequent activation of delayed response genes such as TH in PC12 cells.


Assuntos
Elementos Facilitadores Genéticos , Regulação Enzimológica da Expressão Gênica , Genes fos/fisiologia , Fator de Transcrição AP-1/fisiologia , Tirosina 3-Mono-Oxigenase/biossíntese , Animais , Elementos Antissenso (Genética)/genética , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Ativação Enzimática/genética , Ativação Enzimática/fisiologia , Genes Reporter , Células PC12 , Ratos , Fator de Transcrição AP-1/genética , Células Tumorais Cultivadas , Tirosina 3-Mono-Oxigenase/genética
8.
Prog Brain Res ; 81: 215-20, 1989.
Artigo em Inglês | MEDLINE | ID: mdl-2616783

RESUMO

Interventions confined to the region adjacent to the VMS can produce both respiratory and circulatory effects. Although it has been suggested that both breathing and vasomotor changes arise from the same neural elements near the VMS, our own investigations indicate that the neurons involved are closely linked but not identical. This belief is supported by recent studies which show that AII and angiotensin antagonists microinjected into the rostral portion of the VMS can significantly modify blood pressure and respiration but can produce effects of different sign. These observations, coupled with previous studies of the VMS, indicate the possibility that regions near the VMS may contribute to integration of circulatory and respiratory responses.


Assuntos
Pressão Sanguínea/fisiologia , Bulbo/fisiologia , Respiração/fisiologia , Animais , Gatos , Ratos
9.
Brain Res Mol Brain Res ; 43(1-2): 341-6, 1996 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-9037552

RESUMO

In the present study we examined the effects of hypobaric hypoxia on neuronal (n) and endothelial (e) nitric oxide synthase (NOS) gene expression in the central and peripheral nervous system. Adult rats were exposed either to normoxia (room air) on to hypobaric hypoxia (0.4 atm) for 4, 12 or 24 h and cerebellum and nodose ganglion representing the central and peripheral neurons, respectively, were removed. Messenger RNAs encoding n- and eNOS as well as beta-actin were analyzed by reverse transcriptase polymerase chain reaction (RT-PCR) technique. Hypoxia increased nNOS mRNA expression with maximal changes occurring after 12 h wherein mRNA levels were increased by 10.4 +/- 1.3 and 2 +/- 0.4 fold in nodose ganglion and cerebellum, respectively. Hypoxia, on the other hand, had no significant effect on eNOS and beta-actin mRNA levels. Analysis of nNOS protein and enzyme activity showed near doubling of these variables in both tissues after 24 h of hypoxia, indicating that nNOS protein levels are increased and that the protein is functionally active. These observations demonstrate that 12-24 h of hypobaric hypoxia selectively activates nNOS gene expression, which is reflected in an increase in nNOS protein in central and peripheral neurons. It is suggested that up-regulation of nNOS leads to increased generation of nitric oxide, which in turn may contribute to the readjustments of cardio-respiratory systems during the early stages of chronic hypoxia.


Assuntos
Sistema Nervoso Central/metabolismo , Hipóxia/metabolismo , Neurônios/metabolismo , Óxido Nítrico Sintase/metabolismo , Sistema Nervoso Periférico/metabolismo , Animais , Feminino , Expressão Gênica/genética , Masculino , Ratos , Ratos Sprague-Dawley
10.
J Appl Physiol (1985) ; 88(6): 2287-95, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10846047

RESUMO

Carotid bodies are sensory organs that detect changes in arterial blood oxygen, and the ensuing reflexes are critical for maintaining homeostasis during hypoxemia. During the past decade, tremendous progress has been made toward understanding the cellular mechanisms underlying oxygen sensing at the carotid body. The purpose of this minireview is to highlight some recent concepts on sensory transduction and transmission at the carotid body. A bulk of evidence suggests that glomus (type I) cells are the initial site of transduction and that they release transmitters in response to hypoxia, which causes depolarization of nearby afferent nerve endings, leading to an increase in sensory discharge. There are two main hypotheses to explain the transduction process that triggers transmitter release. One hypothesis assumes that a biochemical event associated with a heme protein triggers the transduction cascade. The other hypothesis suggests that a K(+) channel protein is the oxygen sensor and that inhibition of this channel by hypoxia leading to depolarization is a seminal event in transduction. Although there is body of evidence supporting and questioning each of these, this review will try to point out that the truth lies somewhere in an interrelation between the two. Several transmitters have been identified in glomus cells, and they are released in response to hypoxia. However, their precise roles in sensory transmission remain uncertain. It is hoped that future studies involving transgenic animals with targeted disruption of genes encoding transmitters and their receptors may resolve some of the key issues surrounding the sensory transmission at the carotid body. Further studies are necessary to identify whether a single sensor or multiple oxygen sensors are needed for the transduction process.


Assuntos
Corpo Carotídeo/fisiologia , Células Quimiorreceptoras/fisiologia , Oxigênio/sangue , Animais , Corpo Carotídeo/citologia , Corpo Carotídeo/fisiopatologia , Células Quimiorreceptoras/fisiopatologia , Hipóxia/sangue
11.
J Appl Physiol (1985) ; 90(5): 1986-94, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11299293

RESUMO

To the majority of the population, recurrent episodes of hypoxia are more likely encountered in life than sustained hypoxia. Until recently, much of the information on the long-term effects of intermittent hypoxia has come from studies on human subjects experiencing chronic recurrent apneas. Recent development of animal models of intermittent hypoxia and techniques for exposing cell cultures to alternating cycles of hypoxia have led to new information on the effects of episodic hypoxia on oxygen-sensing mechanisms in the carotid body chemoreceptors and regulation of gene expression. The purpose of this review is to highlight some recent studies on the effects of intermittent hypoxia on oxygen sensing at the carotid bodies and regulation of gene expression. In a rodent model, chronic intermittent hypoxia selectively enhances hypoxic sensitivity of the carotid body chemoreceptors. More interestingly, chronic intermittent hypoxia also induces a novel form of plasticity in the carotid body, leading to long-term facilitation in the sensory discharge. Studies on cell cultures reveal that intermittent hypoxia is more potent in activating activator protein-1 and hypoxia-inducible factor-1 transcription factors than sustained hypoxia. Moreover, some evidence suggests that intermittent hypoxia utilizes intracellular signaling pathways distinct from sustained hypoxia. Reactive oxygen species generated during the reoxygenation phase of intermittent hypoxia might play a key role in the effects of intermittent hypoxia on carotid body function and gene expression. Global gene profile analysis in cell cultures suggests that certain genes are selectively affected by intermittent hypoxia, some upregulated and some downregulated. It is suggested that, in intact animals, coordinated gene regulation of gene expression might be critical for eliciting phenotypic changes in the cardiorespiratory systems in response to intermittent hypoxia. It is hoped that future studies will unravel new mechanisms that are unique to intermittent hypoxia that may lead to a better understanding of the changes in the cardiorespiratory systems and new therapies for diseases associated with chronic recurrent episodes of hypoxia.


Assuntos
Apneia/fisiopatologia , Hipóxia/fisiopatologia , Oxigênio/fisiologia , Altitude , Animais , Apneia/sangue , Apneia/genética , Pressão Sanguínea , Sistema Cardiovascular/fisiopatologia , Corpo Carotídeo/fisiopatologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Hipóxia/sangue , Hipóxia/genética , Espécies Reativas de Oxigênio/fisiologia , Sistema Respiratório/fisiopatologia
12.
J Appl Physiol (1985) ; 63(6): 2418-25, 1987 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-2449416

RESUMO

Experiments were performed on 17 anesthetized, paralyzed, and artificially ventilated cats to evaluate the importance of substance P-like peptide (SP) on the carotid body responses to CO2. Single or paucifiber carotid chemoreceptor activity was recorded from the peripheral end of the cut carotid sinus nerve. In eight of the cats the influence of SP on hyperoxic hypercapnic responses was studied. While the animals breathed 100% O2, intracarotid infusion of SP (1 microgram.kg-1.min-1, 3 min) increased chemoreceptor activity by +4.8 +/- 0.3 impulses/s. After SP infusion, inhalation of CO2 in O2 caused a rapid increase in activity that reached a peak and then adapted to a lower level, whereas similar levels of CO2 before SP caused only a gradual increase in carotid body discharge rate without any overshoot in response. Furthermore SP significantly increased the magnitude and slope of the CO2 response. In the other nine cats the effect of intracarotid infusion of an SP antagonist, [D-Pro2,D-Trp7,9] SP (10-15 micrograms.kg-1.min-1), on carotid body responses to 1) hyperoxic hypercapnia (7% CO2-93% O2), 2) isocapnic hypoxia (11% O2-89% N2), and 3) hypoxic hypercapnia (11% O2-7% CO2-82% N2) was examined. SP antagonist had no effect on carotid body response to hyperoxic hypercapnia but significantly attenuated the chemoreceptor excitation caused by isocapnic hypoxia and hypoxic hypercapnia. These results suggest that 1) SP may play an important role in carotid body responses to hypoxia but not to CO2, and 2) the mechanisms of stimulation of the carotid body by hypercapnia and by hypoxia differ.


Assuntos
Corpo Carotídeo/fisiologia , Hipercapnia/fisiopatologia , Substância P/fisiologia , Animais , Gasometria , Gatos , Feminino , Hipóxia/fisiopatologia , Masculino , Oxigênio/fisiologia , Substância P/antagonistas & inibidores
13.
J Appl Physiol (1985) ; 58(6): 1755-60, 1985 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-4008397

RESUMO

The effect of stimulation of afferent mesenteric nerves on tidal volume (VT), phrenic nerve, and external intercostal muscle activities was studied in anesthetized spontaneously breathing cats. Both mechanical distension of the small intestine and electrical stimulation of the mesenteric nerves resulted in an initial inspiratory inhibition of VT followed by a gradual recovery above the prestimulus controls. Changes in VT were accompanied by a depression of phrenic nerve activity and an excitation of external intercostal muscle activity. During the recovery phase of VT, the amplitude of phrenic nerve activity returned only partially, whereas the activity of the external intercostal muscle was greater than the prestimulus controls. In a second group of experiments, brief tetanic stimulation at the beginning of inspiration led to a complete and maintained inhibition of phrenic nerve activity but with a simultaneous excitation of external intercostal muscle activity and without any change in VT; whereas expiratory stimulation caused a decrease in expiratory abdominal muscle activity, without changing the peak amplitude of phrenic nerve activity. The respiratory changes observed with distension of the small intestine were abolished after denervation of the mesenteric plexus. It is concluded that activation of the visceral afferents of the mesenteric region reflexly changes diaphragmatic breathing to intercostal breathing. It is assumed that such a type of breathing pattern may occur in pregnancy and in pathophysiological situations involving splanchnic viscera.


Assuntos
Músculos Intercostais/fisiologia , Mesentério/inervação , Nervo Frênico/fisiologia , Respiração , Animais , Gatos , Diafragma/inervação , Diafragma/fisiologia , Estimulação Elétrica , Intestino Delgado/fisiologia , Neurônios Motores/fisiologia , Neurônios Aferentes/fisiologia , Estimulação Física , Pressão , Volume de Ventilação Pulmonar , Fatores de Tempo
14.
J Appl Physiol (1985) ; 61(4): 1499-509, 1986 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-3096939

RESUMO

We examined the effects of chemical and reflex drives on the postinspiratory inspiratory activity (PIIA) of phrenic motoneurons using a single-fiber technique. Action potentials from "single" fibers were recorded from the C5 phrenic root together with contralateral mass phrenic activity (also from C5) in anesthetized, paralyzed, and artificially ventilated cats with intact vagus and carotid sinus nerves. Nerve fibers were classified as "early" or "late" based on their onset of discharge in relation to mass phrenic activity during hyperoxic ventilation. Only the early fibers displayed PIIA but not the late fibers, even when their activity began earlier in inspiration with increased chemical drives. Isocapnic hypoxia increased, whereas hyperoxic hypercapnia shortened the duration of PIIA. Pulmonary stretch and "irritant" receptors inhibited PIIA. Hypercapnia and stimulation of peripheral chemoreceptors by lobeline excited both early and late units to the same extent, but hypoxic ventilation had a less marked excitatory effect on late fiber activity. Irritant receptor activation increased the activity of early more than late fibers. Hyperoxic hyperventilation eliminated late phrenic fiber activity, whereas early fibers became tonically active. Bilateral vagotomy abolished this sustained discharge in eight of nine early units, suggesting the importance of vagal afferents in producing tonic firing during hyperventilation. These results suggest that early and late phrenic fibers have different responses to chemical stimuli and to vagally mediated reflexes; late units do not discharge in postinspiratory period, whereas early fibers do; the PIIA is not affected in the same way by various chemical and vagal inputs; and early units that exhibit PIIA display tonic activity with hyperoxic hypocapnia.


Assuntos
Neurônios Motores/fisiologia , Nervo Frênico/fisiologia , Reflexo , Respiração , Nervo Vago/fisiologia , Potenciais de Ação , Animais , Biguanidas/farmacologia , Capsaicina/farmacologia , Dióxido de Carbono/sangue , Gatos , Hipóxia/fisiopatologia , Lobelina/farmacologia , Neurônios Motores/efeitos dos fármacos , Pressão Parcial , Nervo Frênico/efeitos dos fármacos , Nervo Frênico/fisiopatologia
15.
J Appl Physiol (1985) ; 67(5): 1814-9, 1989 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-2574718

RESUMO

We studied the central effects of N-methyl-D-aspartate (NMDA) on respiration in 18 artificially ventilated cats anesthetized with alpha-chloralose. Unilateral topical application of NMDA (1 x 10(-8) mol) to the intermediate region of the ventrolateral medulla exaggerates the phrenic response to CO2 at end-tidal PCO2 levels of less than 50.0 Torr. At higher end-tidal PCO2 levels, however, such differences disappear. Unilateral NMDA application increases the activity of the right and left phrenic nerves equally. Furthermore, the magnitude of the phrenic response after unilateral application of NMDA was not different from that after bilateral application. NMDA also had a vasopressor action when applied to the ventrolateral medullary surface. In contrast to respiratory responses, bilateral application of NMDA caused a significant increase in blood pressure compared with unilateral application of NMDA. Application of the NMDA antagonist 2-amino-5-phosphonovaleric acid abolished both the blood pressure and respiratory effects of NMDA. These results suggest that CO2 and NMDA may act on a common respiratory premotoneuron to produce stimulation of breathing. Because blood pressure responses, unlike respiratory responses, were greater after bilateral application than after unilateral application of NMDA, it is suggested that the neural substrates for the two effects of NMDA seem to be different.


Assuntos
Ácido Aspártico/análogos & derivados , Bulbo/efeitos dos fármacos , Respiração/efeitos dos fármacos , 2-Amino-5-fosfonovalerato/farmacologia , Animais , Ácido Aspártico/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Dióxido de Carbono/metabolismo , Seio Carotídeo/inervação , Gatos , Feminino , Masculino , Bulbo/fisiologia , N-Metilaspartato , Nervo Frênico/efeitos dos fármacos , Nervo Frênico/fisiologia , Respiração/fisiologia , Volume de Ventilação Pulmonar , Vagotomia
16.
J Appl Physiol (1985) ; 66(2): 598-605, 1989 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-2708191

RESUMO

We examined the contribution of the neural elements near the ventral medullary surface (VMS) to the respiratory response caused by 2,4-dinitrophenol (DNP). Two series of experiments were performed on 12 vagotomized and sinoaortic denervated cats. The first series examined the effect of focal cooling of the VMS on the respiratory response to DNP in four spontaneously breathing, anesthetized cats. When the VMS temperature was 37 degrees C, systemic administration of DNP increased minute ventilation under nearly isocapnic conditions, and focal cooling of the intermediate area of VMS to 20 degrees C attenuated the ventilatory augmentation caused by DNP. To eliminate the influence of anesthetics, a second group of experiments was performed on eight decerebrate, artificially ventilated cats while phrenic nerve activity was monitored as an index of respiration. AgNO3 (10%) was topically applied to the VMS until the respiratory response to inhaled CO2 was abolished. Apnea occurred in seven of eight cats after AgNO3, whereas in the remaining one animal, tidal phrenic activity decreased substantially. Systemic administration of DNP produced no respiratory excitation in any of the animals. On the other hand, rhythmic respiratory activity could be provoked by electrical stimulation of the mesencephalic locomotor area and carotid sinus nerve and by excitation of somatic afferents. Histological examination of the brain stem showed that the AgNO3 had penetrated no more than 350 microns from the ventral medullary surface. These results indicate superficial structures of the VMS are of potential importance in mediating the respiratory responses to hypermetabolism.


Assuntos
Dinitrofenóis/farmacologia , Bulbo/efeitos dos fármacos , Respiração/efeitos dos fármacos , 2,4-Dinitrofenol , Animais , Tronco Encefálico/patologia , Gatos , Células Quimiorreceptoras/efeitos dos fármacos , Células Quimiorreceptoras/fisiologia , Temperatura Baixa , Estado de Descerebração/fisiopatologia , Estimulação Elétrica , Feminino , Masculino , Bulbo/fisiologia , Nervo Frênico/fisiologia , Nitrato de Prata/farmacologia
17.
J Appl Physiol (1985) ; 67(2): 541-6, 1989 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-2793655

RESUMO

The role of adenosine in the ventilatory depression induced by hypoxia was studied in 82 spontaneously breathing urethan-anesthetized 4-day-old rabbit pups. Respiration was monitored with a pneumotachograph. The animals were exposed to hypoxia (6% O2 in N2) for 30 min or until the occurrence of terminal apnea. In all animals hypoxia produced an initial increase in ventilation followed by a decrease. In the control group 52% of the animals became apneic after 7 min of hypoxic exposure. By contrast, pretreatment with dipyridamole (10 or 20 mg/kg), an adenosine uptake blocker, significantly shortened the time needed to reach apnea. Thus at 7 min of hypoxia 93% of the animals that received dipyridamole became apneic. On the other hand, administration of adenosine antagonists 8-p-sulfophenyltheophylline (5 or 8 mg/kg) and aminophylline (10 or 25 mg/kg) significantly prolonged the time required to produce apnea. Only 20% of the animals that received these antagonists became apneic at 7 min of hypoxia. These results suggest that adenosine is potentially involved in the ventilatory depression produced by hypoxia in neonatal rabbit pups.


Assuntos
Adenosina/fisiologia , Consumo de Oxigênio , Respiração/efeitos dos fármacos , Aminofilina/farmacologia , Animais , Pressão Sanguínea , Dipiridamol/farmacologia , Coelhos , Sistema Respiratório/metabolismo , Fatores de Tempo
18.
J Appl Physiol (1985) ; 88(1): 195-202, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10642381

RESUMO

Previously, we showed that carotid bodies express neutral endopeptidase (NEP)-like enzyme activity and that phosphoramidon, a potent inhibitor of NEP, potentiates the chemosensory response of the carotid body to hypoxia in vivo. NEP has been shown to hydrolyze methionine enkephalin (Met-Enk) and substance P (SP) in neuronal tissues. The purpose of the present study is to determine whether NEP hydrolyzes Met-Enk and SP in the carotid body and if so whether these peptides contribute to phosphoramidon-induced potentiation of the sensory response to hypoxia. Experiments were performed on carotid bodies excised from anesthetized adult cats (n = 72 carotid bodies). The hydrolysis of Met-Enk and SP was analyzed by HPLC. The results showed that both SP and Met-Enk were hydrolyzed by the carotid body, but the rate of Met-Enk hydrolysis was approximately fourfold higher than that of SP. Phosphoramidon (400 microM) markedly inhibited SP hydrolysis ( approximately 90%) but had only a marginal effect on Met-Enk hydrolysis ( approximately 15% inhibition). Hypoxia (PO(2), 68 +/- 6 Torr) as well as exogenous administration of SP (10 and 20 nmol) increased the sensory discharge of the carotid body in vitro. Sensory responses to hypoxia and SP (10 nmol) were potentiated by approximately 80 and approximately 275%, respectively (P < 0.01), in the presence of phosphoramidon. SP-receptor antagonists Spantide (peptidyl) and CP-96345 (nonpeptidyl) either abolished or markedly attenuated the phosphoramidon-induced potentiation of the sensory response of the carotid body to hypoxia as well as to SP. These results demonstrate that SP is a preferred substrate for NEP in the carotid body and that SP is involved in the potentiation of the hypoxic response of the carotid body by phosphoramidon.


Assuntos
Corpo Carotídeo/enzimologia , Corpo Carotídeo/fisiologia , Neprilisina/metabolismo , Oxigênio/fisiologia , Substância P/metabolismo , Animais , Compostos de Bifenilo/farmacologia , Corpo Carotídeo/efeitos dos fármacos , Corpo Carotídeo/metabolismo , Gatos , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Encefalina Metionina/metabolismo , Feminino , Glicopeptídeos/antagonistas & inibidores , Glicopeptídeos/farmacologia , Hidrólise/efeitos dos fármacos , Hipóxia/fisiopatologia , Cinética , Masculino , Neprilisina/antagonistas & inibidores , Antagonistas dos Receptores de Neurocinina-1 , Receptores da Neurocinina-1/metabolismo , Substância P/análogos & derivados , Substância P/antagonistas & inibidores , Substância P/farmacologia
19.
J Appl Physiol (1985) ; 88(4): 1496-508, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10749847

RESUMO

In the present study, the role of nitric oxide (NO) generated by endothelial nitric oxide synthase (NOS-3) in the control of respiration during hypoxia and hypercapnia was assessed using mutant mice deficient in NOS-3. Experiments were performed on awake and anesthetized mutant and wild-type (WT) control mice. Respiratory responses to 100, 21, and 12% O(2) and 3 and 5% CO(2)-balance O(2) were analyzed. In awake animals, respiration was monitored by body plethysmography along with O(2) consumption (VO(2)) and CO(2) production (VCO(2)). In anesthetized, spontaneously breathing mice, integrated efferent phrenic nerve activity was monitored as an index of neural respiration along with arterial blood pressure and blood gases. Under both experimental conditions, WT mice responded with greater increases in respiration during 12% O(2) than mutant mice. Respiratory responses to hyperoxic hypercapnia were comparable between both groups of mice. Arterial blood gases, changes in blood pressure, VO(2), and VCO(2) during hypoxia were comparable between both groups of mice. Respiratory responses to cyanide and brief hyperoxia were attenuated in mutant compared with WT mice, indicating reduced peripheral chemoreceptor sensitivity. cGMP levels in the brain stem during 12% O(2), taken as an index of NO production, were greater in mutant compared with WT mice. These observations demonstrate that NOS-3 mutant mice exhibit selective blunting of the respiratory responses to hypoxia but not to hypercapnia, which in part is due to reduced peripheral chemosensitivity. These results support the idea that NO generated by NOS-3 is an important physiological modulator of respiration during hypoxia.


Assuntos
Hipóxia/fisiopatologia , Óxido Nítrico Sintase/metabolismo , Fenômenos Fisiológicos Respiratórios , Animais , Corpo Carotídeo/patologia , Corpo Carotídeo/fisiologia , Corpo Carotídeo/fisiopatologia , Vias Eferentes/fisiologia , Vias Eferentes/fisiopatologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase/deficiência , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Consumo de Oxigênio , Nervo Frênico/fisiologia , Nervo Frênico/fisiopatologia , Valores de Referência
20.
J Appl Physiol (1985) ; 90(6): 2508-13, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11356820

RESUMO

Intermittent hypoxia (IH), associated with obstructive sleep apnea, initiates adaptive physiological responses in a variety of organs. Little is known about its influence on diaphragm. IH was simulated by exposing rats to alternating 15-s cycles of 5% O2 and 21% O2 for 5 min, 9 sets/h, 8 h/day, for 10 days. Controls did not experience IH. Diaphragms were excised 20-36 h after IH. Diaphragm bundles were studied in vitro or analyzed for myosin heavy chain isoform composition. No differences in maximum tetanic stress were observed between groups. However, peak twitch stress (P < 0.005), twitch half-relaxation time (P < 0.02), and tetanic stress at 20 or 30 Hz (P < 0.05) were elevated in IH. No differences in expression of myosin heavy chain isoforms or susceptibility to fatigue were seen. Contractile function after 30 min of anoxia (95% N2-5% CO2) was markedly preserved at all stimulation frequencies during IH and at low frequencies after 15 min of reoxygenation. Anoxia-induced increases in passive muscle force were eliminated in the IH animals (P < 0.01). These results demonstrate that IH induces adaptive responses in the diaphragm that preserve its function in anoxia.


Assuntos
Diafragma/fisiopatologia , Hipóxia/fisiopatologia , Adaptação Fisiológica , Animais , Estimulação Elétrica , Cinética , Masculino , Contração Muscular/fisiologia , Fadiga Muscular/fisiologia , Cadeias Pesadas de Miosina/metabolismo , Ratos , Ratos Sprague-Dawley , Síndromes da Apneia do Sono/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA