Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Clin Sleep Med ; 19(8): 1447-1456, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37082823

RESUMO

STUDY OBJECTIVES: The coexistence of obstructive sleep apnea (OSA) and chronic obstructive pulmonary disease (COPD) in a single individual, also known as overlap syndrome (OVS), is associated with higher cardiovascular risk and mortality than either OSA or COPD alone. However, the underlying mechanisms remain unclear. We hypothesized that patients with OVS have elevated systemic inflammatory biomarkers relative to patients with either disease alone, which could explain greater cardiovascular risk observed in OVS. METHODS: We included 255 participants in the study, 55 with COPD alone, 100 with OSA alone, 50 with OVS, and 50 healthy controls. All participants underwent a home sleep study, spirometry, and a blood draw for high-sensitivity C-reactive protein and total blood count analysis. In a randomly selected subset of 186 participants, inflammatory protein profiling was performed using Bio-Rad Bio-Plex Pro Human Cytokine 27-Plex Assays. Biomarker level differences across groups were identified using a mixed linear model. RESULTS: Levels of interleukin 6 (IL-6), high-sensitivity C-reactive protein (hs-CRP), and granulocyte colony stimulating factor (G-CSF) were higher in participants with OVS and COPD compared with healthy controls and participants with OSA. Furthermore, participants with OVS had higher circulating levels of leukocytes and neutrophils than those with COPD, OSA, and controls. CONCLUSIONS: COPD and OVS are associated with higher systemic inflammation relative to OSA and healthy controls. This work proposes the potential utilization of interleukin 6, granulocyte colony stimulating factor, and high-sensitivity C-reactive protein as screening biomarkers for COPD in patients with OSA. Inflammatory pathways may not fully explain the higher cardiovascular risk observed in OVS, indicating the need for further investigation. CITATION: Sanchez-Azofra A, Gu W, Masso-Silva JA, et al. Inflammation biomarkers in OSA, chronic obstructive pulmonary disease, and chronic obstructive pulmonary disease/OSA overlap syndrome. J Clin Sleep Med. 2023;19(8):1447-1456.


Assuntos
Doenças Autoimunes , Doença Pulmonar Obstrutiva Crônica , Síndromes da Apneia do Sono , Apneia Obstrutiva do Sono , Humanos , Proteína C-Reativa , Interleucina-6 , Apneia Obstrutiva do Sono/diagnóstico , Síndromes da Apneia do Sono/complicações , Doença Pulmonar Obstrutiva Crônica/complicações , Inflamação/complicações , Biomarcadores , Doenças Autoimunes/complicações , Fator Estimulador de Colônias de Granulócitos
2.
Dev Cell ; 57(7): 839-853.e6, 2022 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-35303432

RESUMO

Although increased neuropeptides are often detected in lungs that exhibit respiratory distress, whether they contribute to the condition is unknown. Here, we show in a mouse model of neuroendocrine cell hyperplasia of infancy, a pediatric disease with increased pulmonary neuroendocrine cells (PNECs), excess PNEC-derived neuropeptides are responsible for pulmonary manifestations including hypoxemia. In mouse postnatal lung, prolonged signaling from elevated neuropeptides such as calcitonin gene-related peptide (CGRP) activate receptors enriched on endothelial cells, leading to reduced cellular junction gene expression, increased endothelium permeability, excess lung fluid, and hypoxemia. Excess fluid and hypoxemia were effectively attenuated by either prevention of PNEC formation, inactivation of CGRP gene, endothelium-specific inactivation of CGRP receptor gene, or treatment with CGRP receptor antagonist. Neuropeptides were increased in human lung diseases with excess fluid such as acute respiratory distress syndrome. Our findings suggest that restricting neuropeptide function may limit fluid and improve gas exchange in these conditions.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina , Neuropeptídeos , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Células Endoteliais/metabolismo , Humanos , Hipóxia/metabolismo , Pulmão/metabolismo , Camundongos , Neuropeptídeos/metabolismo
3.
J Physiol ; 598(10): 2021-2034, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32026480

RESUMO

KEY POINTS: We hypothesized that hypoxia inducible factor 1α (HIF-1α) in CNS respiratory centres is necessary for ventilatory acclimatization to hypoxia (VAH); VAH is a time-dependent increase in baseline ventilation and the hypoxic ventilatory response (HVR) occurring over days to weeks of chronic sustained hypoxia (CH). Constitutive deletion of HIF-1α in CNS neurons in transgenic mice tended to blunt the increase in HVR that occurs in wild-type mice with CH. Conditional deletion of HIF-1α in glutamatergic neurons of the nucleus tractus solitarius during CH significantly decreased ventilation in acute hypoxia but not normoxia in CH mice. These effects are not explained by changes in metabolic rate, nor CO2 , and there were no changes in the HVR in normoxic mice. HIF-1α mediated changes in gene expression in CNS respiratory centres are necessary in addition to plasticity of arterial chemoreceptors for normal VAH. ABSTRACT: Chronic hypoxia (CH) produces a time-dependent increase of resting ventilation and the hypoxic ventilatory response (HVR) that is called ventilatory acclimatization to hypoxia (VAH). VAH involves plasticity in arterial chemoreceptors and the CNS [e.g. nucleus tractus solitarius (NTS)], although the signals for this plasticity are not known. We hypothesized that hypoxia inducible factor 1α (HIF-1α), an O2 -sensitive transcription factor, is necessary in the NTS for normal VAH. We tested this in two mouse models using loxP-Cre gene deletion. First, HIF-1α was constitutively deleted in CNS neurons (CNS-HIF-1α-/- ) by breeding HIF-1α floxed mice with mice expressing Cre-recombinase driven by the calcium/calmodulin-dependent protein kinase IIα promoter. Second, HIF-1α was deleted in NTS neurons in adult mice (NTS-HIF-1α-/- ) by microinjecting adeno-associated virus that expressed Cre-recombinase in HIF-1α floxed mice. In normoxic control mice, HIF-1α deletion in the CNS or NTS did not affect ventilation, nor the acute HVR (10-15 min hypoxic exposure). In mice acclimatized to CH for 1 week, ventilation in hypoxia was blunted in CNS-HIF-1α-/- and significantly decreased in NTS-HIF-1α-/- compared to control mice (P < 0.0001). These changes were not explained by differences in metabolic rate or CO2 . Immunofluorescence showed that HIF-1α deletion in NTS-HIF-1α-/- was restricted to glutamatergic neurons. The results indicate that HIF-1α is a necessary signal for VAH and the previously described plasticity in glutamatergic neurotransmission in the NTS with CH. HIF-1α deletion had no effect on the increase in normoxic ventilation with acclimatization to CH, indicating this is a distinct mechanism from the increased HVR with VAH.


Assuntos
Hipóxia , Núcleo Solitário , Aclimatação , Animais , Camundongos , Neurônios , Centro Respiratório
4.
Front Physiol ; 9: 860, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30072908

RESUMO

Different patterns of hypoxia evoke different forms of plasticity in the neural control of ventilation. For example, acute intermittent hypoxia produces long term facilitation (LTF) of ventilation, while chronic sustained hypoxia (CH) causes ventilatory acclimatization to hypoxia (VAH). In both LTF and VAH, ventilation in normoxia is greater than normal after the hypoxic stimulus is removed and the acute hypoxic ventilatory response can increase. However, the mechanisms of LTF and VAH are thought to be different based on previous results showing serotonin 5HT2 receptors, which are G protein coupled receptors (GPCR) that activate GQ signaling, contribute to LTF but not VAH. Newer results show that a different GPCR, namely adenosine A2A receptors and the GS signaling pathway, cause LTF with more severe intermittent hypoxia, i.e., PaO2 = 25-30 Torr for GS versus 35-45 Torr for LTF with the GQ signaling pathway. We hypothesized adenosine A2A receptors and GS signaling are involved in establishing VAH with longer term moderate CH and tested this in adult male rats by measuring ventilatory responses to O2 and CO2 with barometric pressure plethysmography after administering MSX-3 or ketanserin (A2A and 5HT2 antagonists, respectively, both 1 mg/Kg i.p.) during CH for 7 days. Blocking GS or GQ signals throughout CH exposure, significantly decreased VAH. After VAH was established, GQ blockade did not affect ventilation while GS blockade increased VAH. Similar to LTF, data support roles for both GQ and GS pathways in the development of VAH but after VAH has been established, the GS pathway inhibits VAH.

5.
Biol. Res ; 51: 57, 2018. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1011401

RESUMO

BACKGROUND: chronic hypoxia increases basal ventilation and pulmonary vascular resistance, with variable changes in arterial blood pressure and heart rate, but it's impact on heart rate variability and autonomic regulation have been less well examined. We studied changes in arterial blood pressure, heart rate and heart rate variability (HRV) in rabbits subjected to chronic normobaric hypoxia (CNH; PB ~ 719 mmHg; FIO2 ~ 9.2%) for 14 days and assess the effect of autonomic control by acute bilateral vagal denervation. RESULTS: exposure to CNH stalled animal weight gain and increased the hematocrit, without affecting heart rate or arterial blood pressure. Nevertheless, Poincaré plots of the electrocardiographic R-R intervals showed a reduced distribution parallel to the line of identity, which interpreted as reduced long-term HRV. In the frequency domain, CNH reduced the very-low- (< 0.2 Hz) and high-frequency components (> 0.8 Hz) of the R-R spectrograms and produced a prominent component in the low-frequency component (0.2-0.5 Hz) of the power spectrum. In control and CNH exposed rabbits, bilateral vagotomy had no apparent effect on the short- and long-term HRV in the Poincaré plots. However, bilateral vagotomy differentially affected higher-frequency components (> 0.8 Hz); reducing it in control animals without modifying it in CNH-exposed rabbits. CONCLUSIONS: These results suggest that CNH exposure shifts the autonomic balance of heart rate towards a sympathetic predominance without modifying resting heart rate or arterial blood pressure.


Assuntos
Animais , Masculino , Coelhos , Vagotomia , Pressão Sanguínea/fisiologia , Frequência Cardíaca/fisiologia , Hipóxia/fisiopatologia , Glicemia/fisiologia , Peso Corporal/fisiologia , Doença Crônica , Modelos Animais de Doenças , Hematócrito
6.
J Physiol ; 595(17): 5797-5813, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28688178

RESUMO

KEY POINTS: Changes in gene expression that occur within hours of exposure to hypoxia in in vivo skeletal muscles remain unexplored. Two hours of hypoxia caused significant down-regulation of extracellular matrix genes followed by a shift at 6 h to altered expression of genes associated with the nuclear lumen while respiratory and blood gases were stabilized. Enrichment analysis of mRNAs classified by stability rates suggests an attenuation of post-transcriptional regulation within hours of hypoxic exposure, where PI3K-Akt signalling was suggested to have a nodal role by pathway analysis. Experimental measurements and bioinformatic analyses suggested that the dephosphorylation of Akt after 2 h of hypoxic exposure might deactivate RNA-binding protein BRF1, hence resulting in the selective degradation of mRNAs. ABSTRACT: The effects of acute hypoxia have been widely studied, but there are few studies of transcriptional responses to hours of hypoxia in vivo, especially in hypoxia-tolerant tissues like skeletal muscles. We used RNA-seq to analyse gene expression in plantaris muscles while monitoring respiration, arterial blood gases, and blood glucose in mice exposed to 8% O2 for 2 or 6 h. Rapid decreases in blood gases and a slower reduction in blood glucose suggest stress, which was accompanied by widespread changes in gene expression. Early down-regulation of genes associated with the extracellular matrix was followed by a shift to genes associated with the nuclear lumen. Most of the early down-regulated genes had mRNA half-lives longer than 2 h, suggesting a role for post-transcriptional regulation. These transcriptional changes were enriched in signalling pathways in which the PI3K-Akt signalling pathway was identified as a hub. Our analyses indicated that gene targets of PI3K-Akt but not HIF were enriched in early transcriptional responses to hypoxia. Among the PI3K-Akt targets, 75% could be explained by a deactivation of adenylate-uridylate-rich element (ARE)-binding protein BRF1, a target of PI3K-Akt. Consistent decreases in the phosphorylation of Akt and BRF1 were experimentally confirmed following 2 h of hypoxia. These results suggest that the PI3K-Akt signalling pathway might play a role in responses induced by acute hypoxia in skeletal muscles, partially through the dephosphorylation of ARE-binding protein BRF1.


Assuntos
Hipóxia/genética , Músculo Esquelético/metabolismo , Fosfatidilinositol 3-Quinases/genética , Animais , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hipóxia/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais
7.
Exp Physiol ; 100(2): 149-55, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25523440

RESUMO

NEW FINDINGS: What is the topic of this review? This article describes the contribution of oxidative stress and pro-inflammatory cytokines to the enhanced carotid body chemosensory responsiveness to the hypoxia and systemic hypertension induced by chronic intermittent hypoxia. What advances does it highlight? Chronic intermittent hypoxia enhances the carotid body chemosensory discharge during normoxia and hypoxia, leading to sympathetic overactivity and hypertension. New evidence suggests that chronic intermittent hypoxia increases pro-inflammatory cytokines. Here, we discuss the role of inflammation in the alterations of the carotid chemoreceptor function as well as the cardiorespiratory alterations following chronic intermittent hypoxia. Chronic intermittent hypoxia (CIH), the main characteristic of obstructive sleep apnoea, enhances carotid body (CB) chemosensory discharges during normoxia and hypoxia and elicits hypertension. These alterations are attributed to oxidative stress, because antioxidants prevent the enhanced CB chemosensory discharges and the hypertension. In this report, we discuss new evidence supporting the suggestion that oxidative stress-induced upregulation of pro-inflammatory cytokines (i.e. tumour necrosis factor-α and interleukin-1ß) in the CB is involved in the chemosensory potentiation and the hypertension following CIH. Anti-inflammatory treatment with ibuprofen prevents the increased tumour necrosis factor-α and interleukin-1ß levels in the CB and the hypertension, but does not reduce the enhanced chemosensory hypoxic response and the local oxidative stress in the CB. In contrast, antioxidant treatment with ascorbic acid prevents the increase in cytokine concentrations and CB oxidative stress, the chemosensory potentiation and the hypertension. Thus, the enhanced CB chemosensory responses to hypoxia depend critically on the oxidative stress, but not on the increased tumour necrosis factor-α and interleukin-1ß in the CB. We discuss a possible role for pro-inflammatory cytokines in development of the hypertension produced by CIH, acting on cardiorespiratory centres located in the CNS.


Assuntos
Corpo Carotídeo/fisiopatologia , Hipóxia/fisiopatologia , Inflamação/fisiopatologia , Estresse Oxidativo/fisiologia , Animais , Corpo Carotídeo/metabolismo , Citocinas/metabolismo , Humanos , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Hipóxia/metabolismo , Inflamação/metabolismo
8.
Behav Brain Res ; 253: 60-7, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23860120

RESUMO

The present work was aimed to evaluate the contribution of interoception to the autonomic and behavioral responses to hypoxia. To address this issue, we studied whether the inactivation of the primary interoceptive posterior insular cortex (pIC) may disrupt the autonomic and behavioral effects of hypoxia in conscious rats. Rats were implanted with telemetric transmitters and microinjection cannulae placed bilaterally in the pIC. After one week, rats were injected with bupivacaine (26.5µM 1µL/side) and saline (1µL/side) into the pIC, and exposed to hypoxia (∼6% O2) for 150s, and autonomic and behavioral responses were recorded. Hypoxia produces hypertension, tachycardia followed by bradycardia, and hypothermia. When O2 dropped to ∼8%, rats showed escape behavior. Baseline cardiovascular variables and the pattern of hypoxia-induced autonomic and behavioral responses were not disrupted by pIC inactivation. However, pIC inactivation produced a modest but significant temperature decrease, higher bradycardic and hypertensive responses to hypoxia, and a minimal delay in escape onset. In addition, we measured the hypoxia-induced Fos activation in the nucleus tractus solitarius (NTS), the periaqueductal gray matter (PAG) and the pIC, which are key components of the interoceptive pathway. Hypoxia increased the number of Fos-positive neurons in the NTS and PAG, but not in the pIC. Present results suggest that pIC is not involved in the hypoxia-induced behavioral response, which seems to be processed in the NTS and PAG, but has a role in the efferent control of autonomic changes coping with hypoxia.


Assuntos
Sistema Nervoso Autônomo/fisiologia , Comportamento Animal/fisiologia , Córtex Cerebral/fisiologia , Hipóxia Encefálica/fisiopatologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Temperatura Corporal/efeitos dos fármacos , Bupivacaína/farmacologia , Interpretação Estatística de Dados , Frequência Cardíaca/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Atividade Motora/fisiologia , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Bloqueadores dos Canais de Sódio/farmacologia , Núcleo Solitário/fisiologia , Telemetria
9.
Adv Exp Med Biol ; 758: 199-205, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23080163

RESUMO

Exposure to chronic intermittent hypoxia (CIH) produces hypertension. A critical process involved in the CIH-induced hypertension is the potentiation of the carotid body (CB) chemosensory responses to acute hypoxia. The CIH-induced changes in the CB chemosensory process have been related to an enhanced reactive oxygen species (ROS) production. However, it is still a matter of debate where ROS could directly modify the CB chemosensory discharge. Recently, we found that CIH-induced increase expression of TNF-a and IL-1b within the CB. Thus, we studied the contribution of these pro-inflammatory cytokines on the enhanced CB chemosensory response to acute hypoxia in rats exposed to CIH. To study the role of TNF-a and IL-1b, male Sprague-Dawley rats were submitted to CIH (5% O(2), 12 times/hr for 8 hr/day) and received chronic ibuprofen treatment (40 mg/kg). Following 21 days of CIH, rats were anaesthetized and the CB chemosensory discharge was recorded in response to several levels FiO2 (5-100%). Exposure to CIH significantly increases the immunorreactive levels of TNF-a and IL-1b in the CB, along with an increase accumulation of the p65 NF-kb subunit. Treating rats with ibuprofen significantly prevents the CIH-induced increases in TNF-a and IL-1b in the CB chemoreceptor cells but failed to decrease the enhanced CB chemosensory reactivity to hypoxia. Our results suggest that the mechanisms underlying the potentiation of the CB chemosensory response to acute hypoxia are not linked to the increased expression of TNF-a and IL-1b within the CBs of CIH-exposed rats.


Assuntos
Corpo Carotídeo/fisiologia , Hipóxia/fisiopatologia , Inflamação/fisiopatologia , Animais , Ibuprofeno/farmacologia , Interleucina-1beta/análise , Ratos , Fator de Transcrição RelA/análise , Fator de Necrose Tumoral alfa/análise
11.
Eur Respir J ; 39(6): 1492-500, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22183481

RESUMO

Chronic intermittent hypoxia (CIH), a main feature of obstructive sleep apnoea (OSA), increases hypoxic ventilatory responses and elicits hypertension, partially attributed to an enhance carotid body (CB) responsiveness to hypoxia. As inflammation has been involved in CIH-induced hypertension and chemosensory potentiation, we tested whether ibuprofen may block CB chemosensory and cardiorespiratory alterations induced by CIH in a rat model of OSA. We studied the effects of ibuprofen (40 mg · kg(-1) · day(-1)) on immunohistochemical interleukin (IL)-1ß and tumour necrosis factor (TNF)-α levels in the CB, the number of c-fos-positive neurons in the nucleus tractus solitarii (NTS), CB chemosensory and ventilatory responses to hypoxia, and arterial blood pressure in male rats either exposed for 21 days to 5% O(2) (12 episodes · h(-1), 8 h · day(-1)) or kept under sham condition. CIH increased CB TNF-α and IL-1ß and c-fos-positive neurons in the NTS, enhanced carotid chemosensory and ventilatory hypoxic responses, and produced hypertension. Ibuprofen prevented CB cytokine overexpression and CIH-induced increases in c-fos-positive neurons in the NTS, the enhanced hypoxic ventilatory responses and hypertension, but failed to impede the CB chemosensory potentiation. Results suggest that pro-inflammatory cytokines may contribute to the CIH-induced cardiorespiratory alterations, acting at several levels of the hypoxic chemoreflex and cardiovascular control pathways.


Assuntos
Corpo Carotídeo/fisiopatologia , Hipóxia/fisiopatologia , Inflamação/fisiopatologia , Apneia Obstrutiva do Sono/fisiopatologia , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Sistema Cardiovascular/efeitos dos fármacos , Sistema Cardiovascular/fisiopatologia , Corpo Carotídeo/efeitos dos fármacos , Doença Crônica , Modelos Animais de Doenças , Hipertensão/tratamento farmacológico , Hipertensão/fisiopatologia , Hipóxia/tratamento farmacológico , Ibuprofeno/uso terapêutico , Inflamação/tratamento farmacológico , Interleucina-1beta/análise , Masculino , Proteínas Proto-Oncogênicas c-fos/análise , Ratos , Ratos Sprague-Dawley , Respiração/efeitos dos fármacos , Apneia Obstrutiva do Sono/tratamento farmacológico , Núcleo Solitário/química , Fator de Necrose Tumoral alfa/análise
12.
Am J Physiol Lung Cell Mol Physiol ; 301(5): L702-11, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21821731

RESUMO

Chronic intermittent hypoxia (CIH), a characteristic of sleep obstructive apnea, enhances carotid body (CB) chemosensory responses to hypoxia, but its consequences on CB vascular area and VEGF expression are unknown. Accordingly, we studied the effect of CIH on CB volume, glomus cell numbers, blood vessel diameter and number, and VEGF immunoreactivity (VEGF-ir) in male Sprague-Dawley rats exposed to 5% O(2), 12 times/h for 8 h or sham condition for 21 days. We found that CIH did not modify the CB volume or the number of glomus cells but increased VEGF-ir and enlarged the vascular area by increasing the size of the blood vessels, whereas the number of the vessels was unchanged. Because oxidative stress plays an essential role in the CIH-induced carotid chemosensory potentiation, we tested whether antioxidant treatment with ascorbic acid may impede the vascular enlargement and the VEGF upregulation. Ascorbic acid, which prevents the CB chemosensory potentiation, failed to impede the vascular enlargement and the increased VEGF-ir. Thus present results suggest that the CB vascular enlargement induced by CIH is a direct effect of intermittent hypoxia and not secondary to the oxidative stress. Accordingly, the subsequent capillary changes may be secondary to the mechanisms involved in the neural chemosensory plasticity induced by intermittent hypoxia.


Assuntos
Ácido Ascórbico/farmacologia , Corpo Carotídeo , Hipóxia/complicações , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Antioxidantes/farmacologia , Corpo Carotídeo/metabolismo , Corpo Carotídeo/patologia , Corpo Carotídeo/fisiopatologia , Contagem de Células , Células Quimiorreceptoras/citologia , Células Quimiorreceptoras/efeitos dos fármacos , Células Quimiorreceptoras/metabolismo , Hipóxia/tratamento farmacológico , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Imuno-Histoquímica , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Apneia Obstrutiva do Sono/complicações , Apneia Obstrutiva do Sono/tratamento farmacológico , Apneia Obstrutiva do Sono/metabolismo , Apneia Obstrutiva do Sono/fisiopatologia , Regulação para Cima
13.
Respir Physiol Neurobiol ; 164(3): 401-10, 2008 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-18824142

RESUMO

Histamine has been proposed to be an excitatory transmitter between the carotid body (CB) chemoreceptor (glomus) cells and petrosal ganglion (PG) neurons. The histamine biosynthetic pathway, its storage and release, as well as the presence of histamine H1, H2 and H3 receptors have been found in the CB. However, there is only indirect evidence showing the presence of histamine in glomus cells, or weather its application produces chemosensory excitation. Thus, we studied the histamine immunocytochemical localization in the cat CB, and the effects of histamine, and H1, H2 and H3 receptor blockers on carotid sinus nerve (CSN) discharge, using CB and PG preparations in vitro. We found histamine immunoreactivity in dense-cored vesicles of glomus cells. Histamine induced dose-dependent increases in CSN discharge in the CB, but not in the PG. The H1-antagonist pyrilamine reduced the CB responses induced by histamine, the H2-antagonists cimetidine and ranitidine had no effect, while the H3-antagonist thioperamide enhanced histamine-induced responses. Present data suggests that histamine plays an excitatory modulatory role in the generation of cat CB chemosensory activity.


Assuntos
Corpo Carotídeo/citologia , Corpo Carotídeo/metabolismo , Células Quimiorreceptoras/metabolismo , Histamina/metabolismo , Acetilcolina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Gatos , Células Quimiorreceptoras/efeitos dos fármacos , Células Quimiorreceptoras/ultraestrutura , Relação Dose-Resposta a Droga , Histamina/farmacologia , Agonistas dos Receptores Histamínicos/farmacologia , Antagonistas dos Receptores Histamínicos H1/farmacologia , Antagonistas dos Receptores Histamínicos H3/farmacologia , Hipóxia/fisiopatologia , Técnicas In Vitro , Masculino , Microscopia Imunoeletrônica/métodos , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Piperidinas/farmacologia , Pirilamina/farmacologia , Tirosina 3-Mono-Oxigenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA