Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 322
Filtrar
1.
Reprod Toxicol ; 100: 35-41, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33383163

RESUMO

We have reported that smoking during pregnancy is associated with deficit in neonatal central chemoreception. However, the underlying mechanism is not well clarified. In this study, we developed a rat model of maternal cigarette smoke (CS) exposure. Pregnant rats were exposed to CS during gestational day 1-20. Offspring were studied on postnatal day 2. Reactive oxygen species (ROS) content and expressions of antioxidant proteins in retrotrapezoid nucleus/parafacial respiratory group (RTN/pFRG) were examined by fluorogenic dye MitoSOX™ Red and Western blotting, respectively. The response of hypoglossal rootlets discharge to acidification was also detected with micro-injection of H2O2 into RTN/pFRG of offspring brainstem slices in vitro. Results showed that maternal CS exposure led to an increase in ROS production, and brought about decreases in mitochondrial superoxide dismutase and Kelch-like ECH-associated protein-1, and an increase in NF-E2-related factor 2 in offspring RTN/pFRG. Catalase and glutathione reductase expressions were not significantly changed. Moreover, oxidative stress induced by micro-injection of H2O2 into RTN/pFRG in vitro inhibited the discharge response of hypoglossal rootlets to acidification. These findings suggest that maternal CS exposure results in oxidative stress in RTN/pFRG of rat offspring, which might play a role in the impairment of central chemoreception.


Assuntos
Bulbo/metabolismo , Nicotiana , Estresse Oxidativo/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Fumaça/efeitos adversos , Animais , Animais Recém-Nascidos , Células Quimiorreceptoras/efeitos dos fármacos , Feminino , Bulbo/efeitos dos fármacos , Bulbo/ultraestrutura , Mitocôndrias/química , Nicotina/efeitos adversos , Gravidez , Complicações na Gravidez/etiologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/análise , Fumar/efeitos adversos
2.
Pflugers Arch ; 473(1): 107-120, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33074398

RESUMO

Oxaliplatin is a platinum-based alkylating chemotherapeutic agent used for cancer treatment. Neurotoxicity is one of its major adverse effects that often demands dose limitation. However, the effects of chronic oxaliplatin on the toxicity of the autonomic nervous system regulating cardiorespiratory function and adaptive reflexes are unknown. Male Sprague Dawley rats were treated with intraperitoneal oxaliplatin (3 mg kg-1 per dose) 3 times a week for 14 days. The effects of chronic oxaliplatin treatment on baseline mean arterial pressure (MAP); heart rate (HR); splanchnic sympathetic nerve activity (sSNA); phrenic nerve activity (PNA) and its amplitude (PNamp) and frequency (PNf); and sympathetic reflexes were investigated in anaesthetised, vagotomised and artificially ventilated rats. The same parameters were evaluated after acute oxaliplatin injection, and in the chronic treatment group following a single dose of oxaliplatin. The amount of platinum in the brain was determined with atomic absorption spectrophotometry. Chronic oxaliplatin treatment significantly increased MAP, sSNA and PNf and decreased HR and PNamp, while acute oxaliplatin had no effects. Platinum was accumulated in the brain after chronic oxaliplatin treatment. In the chronic oxaliplatin treatment group, further administration of a single dose of oxaliplatin increased MAP and sSNA. The baroreceptor sensitivity and somatosympathetic reflex were attenuated at rest while the sympathoexcitatory response to hypercapnia was increased in the chronic treatment group. This is the first study to reveal oxaliplatin-induced alterations in the central regulation of cardiovascular and respiratory functions as well as reflexes that may lead to hypertension and breathing disorders which may be mediated via accumulated platinum in the brain.


Assuntos
Fenômenos Fisiológicos Cardiovasculares/efeitos dos fármacos , Oxaliplatina/efeitos adversos , Oxaliplatina/farmacocinética , Platina/metabolismo , Fenômenos Fisiológicos Respiratórios/efeitos dos fármacos , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Barorreflexo/efeitos dos fármacos , Sangue/efeitos dos fármacos , Células Quimiorreceptoras/efeitos dos fármacos , Esquema de Medicação , Frequência Cardíaca , Masculino , Oxaliplatina/administração & dosagem , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Nervos Esplâncnicos/efeitos dos fármacos , Sistema Nervoso Simpático/efeitos dos fármacos , Distribuição Tecidual
3.
PLoS One ; 15(10): e0237643, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33064729

RESUMO

We previously reported that maternal cigarette smoke (CS) exposure resulted in impairment of central chemoreception and induced mitochondrial dysfunction in offspring parafacial respiratory group (pFRG), the kernel for mammalian central chemoreception. We also found that hydrogen sulfide (H2S) could attenuate maternal CS exposure-induced impairment of central chemoreception in the rat offspring in vivo. Mitochondrial ATP sensitive potassium (mitoKATP) channel has been reported to play a significant role in mitochondrial functions and protect against apoptosis in neurons. Thus, we hypothesize here that mitoKATP channel plays a role in the protective effects of H2S on neonatal central chemoreception in maternal CS-exposed rats. Our findings revealed that pretreatment with NaHS (donor of H2S, 22.4mM) reversed the central chemosensitivity decreased by maternal CS exposure, and also inhibited cell apoptosis in offspring pFRG, however, 5-HD (blocker of mitoKATP channels, 19mM) attenuated the protective effects of NaHS. In addition, NaHS declined pro-apoptotic proteins related to mitochondrial pathway apoptosis in CS rat offspring pFRG, such as Bax, Cytochrome C, caspase9 and caspase3. NaHS or 5-HD alone had no significant effect on above indexes. These results suggest that mitoKATP channels play an important role in the protective effect of H2S against impairment of central chemoreception via anti-apoptosis in pFRG of rat offspring exposed to maternal CS.


Assuntos
Células Quimiorreceptoras/efeitos dos fármacos , Fumar Cigarros/efeitos adversos , Sulfeto de Hidrogênio/metabolismo , Exposição Materna/efeitos adversos , Canais de Potássio/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Células Quimiorreceptoras/patologia , Células Quimiorreceptoras/fisiologia , Feminino , Bulbo/efeitos dos fármacos , Bulbo/patologia , Bulbo/fisiopatologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Substâncias Protetoras/metabolismo , Substâncias Protetoras/farmacologia , Ratos , Ratos Sprague-Dawley , Mecânica Respiratória/efeitos dos fármacos , Mecânica Respiratória/fisiologia , Sulfetos/metabolismo , Sulfetos/farmacologia
4.
Respir Physiol Neurobiol ; 274: 103383, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31923590

RESUMO

We previously found that maternal cigarette smoke (CS) exposure resulted in impairment of central chemoreception and oxidative stress and mitochondrial dysfunction of parafacial respiratory group (pFRG, a critical site for mammalian central chemoreception) in neonatal rats. The present work was carried out to identify if maternal CS exposure could disturb the glutamate (GLU)-ergic and γ-aminobutyric acid (GABA)-ergic balance in pFRG of neonatal rats. We found that maternal CS exposure induced a decrease in GLU content and consequently in GLU/GABA ratio in pFRG of neonatal rats. Maternal CS exposure also decreased glutamine content and glutaminase and glutamine synthetase activity in offspring pFRG. In addition, expression of vesicular glutamate transporter 2 was depressed, and those of glutamate transporter 1 and GABA transporter 3 were elevated by maternal CS exposure. These results indicate that maternal CS exposure leads to a disturbance of GLU/GABA balance in pFRG of the neonatal rats, which might contribute to the suppression of central chemoreception in maternal CS-exposed offspring.


Assuntos
Relógios Biológicos/efeitos dos fármacos , Células Quimiorreceptoras , Fumar Cigarros/efeitos adversos , Ácido Glutâmico/metabolismo , Bulbo , Efeitos Tardios da Exposição Pré-Natal , Centro Respiratório , Ácido gama-Aminobutírico/metabolismo , Animais , Animais Recém-Nascidos , Células Quimiorreceptoras/efeitos dos fármacos , Células Quimiorreceptoras/metabolismo , Feminino , Bulbo/efeitos dos fármacos , Bulbo/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos , Ratos Sprague-Dawley , Centro Respiratório/efeitos dos fármacos , Centro Respiratório/metabolismo
5.
Am J Physiol Heart Circ Physiol ; 318(1): H78-H89, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31675256

RESUMO

The role of the acid-sensing ion channel 1a (ASIC1a) in evoking the exercise pressor reflex is unknown, despite the fact that ASIC1a is opened by decreases in pH in the physiological range. This fact prompted us to test the hypothesis that ASIC1a plays an important role in evoking the exercise pressor reflex in decerebrated rats with freely perfused hindlimb muscles. To test this hypothesis, we measured the effect of injecting two ASIC1a blockers into the arterial supply of the triceps surae muscles on the reflex pressor responses to four maneuvers, namely 1) static contraction of the triceps surae muscles (i.e., the exercise pressor reflex), 2) calcaneal tendon stretch, 3) intra-arterial injection of lactic acid, and 4) intra-arterial injection of diprotonated phosphate. We found that the 2 ASIC1a blockers, psalmotoxin-1 (200 ng/kg) and mambalgin-1 (6.5 µg/kg), decreased the pressor responses to static contraction as well as the peak pressor responses to injection of lactic acid and diprotonated phosphate. In contrast, neither ASIC1a blocker had any effect on the pressor responses to tendon stretch. Importantly, we found that ASIC1a blockade significantly decreased the pressor response to static contraction after a latency of at least 8 s. Our results support the hypothesis that ASIC1a plays a key role in evoking the metabolic component of the exercise pressor reflex.NEW & NOTEWORTHY The role played by acid-sensing ion channel 1a (ASIC1a) in evoking the exercise pressor reflex remains unknown. In decerebrated rats with freely perfused femoral arteries, blocking ASIC1a with psalmotoxin-1 or mambalgin-1 significantly attenuated the pressor response to static contraction, lactic acid, and diprotonated phosphate injection but had no effect on the pressor response to stretch. We conclude that ASIC1a plays a key role in evoking the exercise pressor reflex by responding to contraction-induced metabolites, such as protons.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Sistema Nervoso Autônomo/fisiologia , Células Quimiorreceptoras/metabolismo , Contração Muscular , Fusos Musculares/metabolismo , Músculo Esquelético/inervação , Músculo Esquelético/metabolismo , Reflexo , Canais Iônicos Sensíveis a Ácido/efeitos dos fármacos , Animais , Células Quimiorreceptoras/efeitos dos fármacos , Estado de Descerebração , Venenos Elapídicos/farmacologia , Membro Posterior , Concentração de Íons de Hidrogênio , Masculino , Moduladores de Transporte de Membrana/farmacologia , Fusos Musculares/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Peptídeos/farmacologia , Ratos Sprague-Dawley , Venenos de Aranha/farmacologia
6.
Exp Physiol ; 104(9): 1335-1342, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31161612

RESUMO

NEW FINDINGS: What is the central question of this study? The traditional surgical approach for sino-aortic denervation in rats leads to simultaneous carotid baroreceptor and chemoreceptor deactivation, which does not permit their individual study in different situations. What is the main finding and its importance? We have described a new surgical approach capable of selective denervation of the arterial (aortic and carotid) baroreceptors, keeping the carotid bodies (chemoreceptors) intact. It is understood that this technique might be a useful tool for investigating the relative role of the baro- and chemoreceptors in several physiological and pathophysiological conditions. ABSTRACT: Studies have demonstrated that the traditional surgical approach for sino-aortic denervation in rats leads to simultaneous carotid baroreceptor and chemoreceptor deactivation. The present study reports a new surgical approach to denervate the aortic and the carotid baroreceptors selectively, keeping the carotid bodies (peripheral chemoreceptors) intact. Wistar rats were subjected to specific aortic and carotid baroreceptor denervation (BAROS-X) or sham surgery (SHAM). Baroreflex activation was achieved by i.v. administration of phenylephrine, whereas peripheral chemoreflex activation was produced by i.v. administration of potassium cyanide. The SHAM and BAROS-X rats displayed significant hypertensive responses to phenylephrine administration. However, the reflex bradycardia following the hypertensive response caused by phenylephrine was remarkable in SHAM, but not significant in the BAROS-X animals, confirming the efficacy of the surgical procedure to abolish the baroreflex. In addition, the baroreflex activation elicited by phenylephrine increased carotid sinus nerve activity only in SHAM, but not in the BAROS-X animals, providing support to the notion that the baroreceptor afferents were absent. Instead, the classical peripheral chemoreflex hypertensive and bradycardic responses to potassium cyanide were similar in both groups, suggesting that the carotid body chemoreceptors were preserved after BAROS-X. In summary, we describe a new surgical approach in which only the baroreceptors are eliminated, while the carotid chemoreceptors are preserved. Therefore, it is understood that this procedure is potentially a useful tool for examining the relative roles of the arterial baroreceptors versus the chemoreceptors in several pathophysiological conditions, for instance, arterial hypertension and heart failure.


Assuntos
Aorta/cirurgia , Artérias/cirurgia , Corpo Carotídeo/cirurgia , Animais , Aorta/efeitos dos fármacos , Aorta/fisiologia , Artérias/efeitos dos fármacos , Barorreflexo/efeitos dos fármacos , Barorreflexo/fisiologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Corpo Carotídeo/efeitos dos fármacos , Corpo Carotídeo/fisiologia , Células Quimiorreceptoras/efeitos dos fármacos , Células Quimiorreceptoras/fisiologia , Denervação/métodos , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Hipertensão/fisiopatologia , Masculino , Fenilefrina/farmacologia , Pressorreceptores/efeitos dos fármacos , Pressorreceptores/fisiologia , Ratos , Ratos Wistar
7.
Science ; 364(6436)2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30872534

RESUMO

Chemogenetics enables noninvasive chemical control over cell populations in behaving animals. However, existing small-molecule agonists show insufficient potency or selectivity. There is also a need for chemogenetic systems compatible with both research and human therapeutic applications. We developed a new ion channel-based platform for cell activation and silencing that is controlled by low doses of the smoking cessation drug varenicline. We then synthesized subnanomolar-potency agonists, called uPSEMs, with high selectivity for the chemogenetic receptors. uPSEMs and their receptors were characterized in brains of mice and a rhesus monkey by in vivo electrophysiology, calcium imaging, positron emission tomography, behavioral efficacy testing, and receptor counterscreening. This platform of receptors and selective ultrapotent agonists enables potential research and clinical applications of chemogenetics.


Assuntos
Células Quimiorreceptoras/efeitos dos fármacos , Antagonistas Nicotínicos/farmacologia , Agentes de Cessação do Hábito de Fumar/farmacologia , Vareniclina/análogos & derivados , Vareniclina/farmacologia , Receptor Nicotínico de Acetilcolina alfa7/agonistas , Animais , Células Quimiorreceptoras/fisiologia , Engenharia Genética , Haplorrinos , Humanos , Ligantes , Camundongos , Mutação , Domínios Proteicos , Receptores de Glicina/agonistas , Receptores de Glicina/genética , Receptores 5-HT3 de Serotonina/genética , Tropizetrona/farmacologia , Receptor Nicotínico de Acetilcolina alfa7/genética
8.
Exp Physiol ; 103(12): 1679-1691, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30242927

RESUMO

NEW FINDINGS: What is the central question of this study? ATP is known to modulate the chemosensitivity of some brain areas. However, whether the ATP contributes specifically to the mechanism of chemoreception in the lateral hypothalamus/perifornical area (LH/PFA) remains to be determined. What is the main finding and its importance? ATP, acting on the LH/PFA, enhances the hypercapnic ventilatory response in rats during wakefulness, in the dark period. Our results highlight the importance of ATP as a modulator of central chemoreception and provide new insight regarding the mechanisms involved in LH/PFA chemosensitivity and the sleep-wake differences in the CO2 /H+ -dependent drive to breathe. ABSTRACT: The lateral hypothalamus/perifornical area (LH/PFA) is a central chemoreceptor site, which acts in an arousal state-dependent manner. It has been shown that purinergic signalling through ATP influences the CO2 /H+ responsiveness of other chemosensitive regions, but it is unknown whether ATP is also involved in the mechanisms that underlie LH/PFA chemoreception. Here, we studied the effects of microdialysis of a P2X-receptor agonist [α,ß-methylene ATP (α,ß-meATP), 10 mm] and a non-selective P2-receptor antagonist [pyridoxal-phosphate-6-azophenyl-2',4'-disulfonate (PPADS), 1 mm] into the LH/PFA of conscious rats on ventilation in room air and in 7% CO2 . In the dark (active) phase, but not in the light, microdialysis of α,ß-meATP caused an augmented hypercapnic ventilatory response during wakefulness, but not during non-REM sleep (P < 0.001). PPADS caused no change in CO2 ventilatory responses in either the dark period or the light period. Our data suggest that ATP in LH/PFA contributes to the hypercapnic ventilatory response in conscious rats during wakefulness in the dark phase of the diurnal cycle.


Assuntos
Trifosfato de Adenosina/metabolismo , Dióxido de Carbono/metabolismo , Células Quimiorreceptoras/metabolismo , Região Hipotalâmica Lateral/metabolismo , Ventilação Pulmonar/fisiologia , Trifosfato de Adenosina/análogos & derivados , Animais , Células Quimiorreceptoras/efeitos dos fármacos , Hipercapnia/metabolismo , Região Hipotalâmica Lateral/efeitos dos fármacos , Masculino , Ventilação Pulmonar/efeitos dos fármacos , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacologia , Ratos , Ratos Wistar , Respiração/efeitos dos fármacos , Sono/efeitos dos fármacos , Sono/fisiologia , Vigília/efeitos dos fármacos , Vigília/fisiologia
9.
Neuropharmacology ; 138: 47-56, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29857188

RESUMO

The brain regulates breathing in response to changes in tissue CO2/H+ via a process called central chemoreception. Neurons and astrocytes in the retrotrapezoid nucleus (RTN) function as respiratory chemoreceptors. The role of astrocytes in this process appears to involve CO2/H+-dependent release of ATP to enhance activity of chemosensitive RTN neurons. Considering that in most brain regions extracellular ATP is rapidly broken down to adenosine by ectonucleotidase activity and since adenosine is a potent neuromodulator, we wondered whether adenosine signaling contributes to RTN chemoreceptor function. To explore this possibility, we pharmacologically manipulated activity of adenosine receptors in the RTN under control conditions and during inhalation of 7-10% CO2 (hypercapnia). In urethane-anesthetized or unrestrained conscious rats, bilateral injections of adenosine into the RTN blunted the hypercapnia ventilatory response. The inhibitory effect of adenosine on breathing was blunted by prior RTN injection of a broad spectrum adenosine receptor blocker (8-PT) or a selective A1-receptor blocker (DPCPX). Although RTN injections of 8PT, DPCPX or the ectonucleotidase inhibitor ARL67156 did not affected baseline breathing in either anesthetized or awake rats. We did find that RTN application of DPCPX or ARL67156 potentiated the respiratory frequency response to CO2, suggesting a portion of ATP released in the RTN during high CO2/H+ is converted to adenosine and serves to limit chemoreceptor function. These results identify adenosine as a novel purinergic regulator of RTN chemoreceptor function during hypercapnia.


Assuntos
Adenosina/metabolismo , Hipercapnia/metabolismo , Bulbo/metabolismo , Receptores Purinérgicos P1/metabolismo , Reflexo/fisiologia , Respiração , Adenosina/administração & dosagem , Animais , Células Quimiorreceptoras/efeitos dos fármacos , Células Quimiorreceptoras/metabolismo , Hipercapnia/tratamento farmacológico , Masculino , Bulbo/efeitos dos fármacos , Antagonistas de Receptores Purinérgicos P1/farmacologia , Ratos Wistar , Reflexo/efeitos dos fármacos , Respiração/efeitos dos fármacos , Vigília
10.
Physiol Genomics ; 50(4): 255-262, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29521602

RESUMO

Mammalian carotid bodies (CB) are chemosensory organs that mediate compensatory cardiorespiratory reflexes in response to low blood PO2 (hypoxemia) and elevated CO2/H+ (acid hypercapnia). The chemoreceptors are glomus or type I cells that occur in clusters enveloped by neighboring glial-like type II cells. During chemoexcitation type I cells depolarize, leading to Ca2+-dependent release of several neurotransmitters, some excitatory and others inhibitory, that help shape the afferent carotid sinus nerve (CSN) discharge. Among the predominantly excitatory neurotransmitters are the purines ATP and adenosine, whereas dopamine (DA) is inhibitory in most species. There is a consensus that ATP and adenosine, acting via postsynaptic ionotropic P2X2/3 receptors and pre- and/or postsynaptic A2 receptors respectively, are major contributors to the increased CSN discharge during chemoexcitation. However, it has been proposed that the CB sensory output is also tuned by paracrine signaling pathways, involving glial-like type II cells. Indeed, type II cells express functional receptors for several excitatory neurochemicals released by type I cells including ATP, 5-HT, ACh, angiotensin II, and endothelin-1. Stimulation of the corresponding G protein-coupled receptors increases intracellular Ca2+, leading to the further release of ATP through pannexin-1 channels. Recent evidence suggests that other CB neurochemicals, e.g., histamine and DA, may actually inhibit Ca2+ signaling in subpopulations of type II cells. Here, we review evidence supporting neurotransmitter-mediated crosstalk between type I and type II cells of the rat CB. We also consider the potential contribution of paracrine signaling and purinergic catabolic pathways to the integrated sensory output of the CB during chemotransduction.


Assuntos
Corpo Carotídeo/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/metabolismo , Corpo Carotídeo/efeitos dos fármacos , Células Quimiorreceptoras/efeitos dos fármacos , Células Quimiorreceptoras/metabolismo , Endotelina-1/farmacologia , Humanos , Receptores Muscarínicos/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Serotonina/farmacologia
11.
eNeuro ; 5(6)2018.
Artigo em Inglês | MEDLINE | ID: mdl-30627640

RESUMO

A subset of neurons in the retrotrapezoid nucleus (RTN) function as respiratory chemoreceptors by regulating depth and frequency of breathing in response to changes in tissue CO2/H+. The activity of chemosensitive RTN neurons is also subject to modulation by CO2/H+-dependent purinergic signaling. However, mechanisms contributing to purinergic regulation of RTN chemoreceptors are not entirely clear. Recent evidence suggests adenosine inhibits RTN chemoreception in vivo by activation of A1 receptors. The goal of this study was to characterize effects of adenosine on chemosensitive RTN neurons and identify intrinsic and synaptic mechanisms underlying this response. Cell-attached recordings from RTN chemoreceptors in slices from rat or wild-type mouse pups (mixed sex) show that exposure to adenosine (1 µM) inhibits chemoreceptor activity by an A1 receptor-dependent mechanism. However, exposure to a selective A1 receptor antagonist (8-cyclopentyl-1,3-dipropylxanthine, DPCPX; 30 nM) alone did not potentiate CO2/H+-stimulated activity, suggesting activation of A1 receptors does not limit chemoreceptor activity under these reduced conditions. Whole-cell voltage-clamp from chemosensitive RTN neurons shows that exposure to adenosine activated an inward rectifying K+ conductance, and at the network level, adenosine preferentially decreased frequency of EPSCs but not IPSCs. These results show that adenosine activation of A1 receptors inhibits chemosensitive RTN neurons by direct activation of a G-protein-regulated inward-rectifier K+ (GIRK)-like conductance, and presynaptically, by suppression of excitatory synaptic input to chemoreceptors.


Assuntos
Adenosina/metabolismo , Células Quimiorreceptoras/fisiologia , Receptores Purinérgicos P1/metabolismo , Centro Respiratório/citologia , Transdução de Sinais/fisiologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Adenosina/farmacologia , Animais , Animais Recém-Nascidos , Bário/farmacologia , Dióxido de Carbono/farmacologia , Células Quimiorreceptoras/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasticidade Neuronal/efeitos dos fármacos , Neurotransmissores/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Purinérgicos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P1/genética , Transdução de Sinais/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia
12.
Respir Physiol Neurobiol ; 246: 98-106, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28851593

RESUMO

Cystathionine-γ-lyase (CSE) is a multifunctional enzyme, and hydrogen sulfide (H2S) is one of its products. CSE and H2S have recently been proposed to be critical signaling molecules in hypoxia-induced excitation of carotid body (CB) glomus cells and the chemosensory response. Because the role of H2S in arterial chemoreception is still debated, we further examined the role of CSE by studying the effects of hypoxia on TASK K+ channel activity, cell depolarization, [Ca2+]i and ventilation using CSE+/+ and CSE-/- mice. As predicted, hypoxia reduced TASK activity and depolarized glomus cells isolated from CSE+/+ mice. These effects of hypoxia were not significantly altered in glomus cells from CSE-/- mice. Basal [Ca2+]i and hypoxia-induced elevation of [Ca2+] were also not significantly different in glomus cells from CSE+/+ and CSE-/- mice. In whole-body plethysmography, hypoxia (10%O2) increased minute ventilation in both CSE+/+ and CSE-/- mice equally well, and no significant differences were found in either males or females when adjusted by body weight. Together, these results show that deletion of the CSE gene has no effects on hypoxia-induced changes in TASK, cell depolarization, [Ca2+]i and ventilation, and therefore do not support the idea that CSE/H2S signaling is important for CB chemoreceptor activity in mice.


Assuntos
Cálcio/metabolismo , Corpo Carotídeo/patologia , Cistationina gama-Liase/metabolismo , Hipóxia , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Ventilação , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Animais , Animais Geneticamente Modificados , Bicarbonatos/farmacologia , Células Quimiorreceptoras/efeitos dos fármacos , Células Quimiorreceptoras/fisiologia , Cistationina gama-Liase/genética , Modelos Animais de Doenças , Feminino , Sulfeto de Hidrogênio/metabolismo , Hipóxia/metabolismo , Hipóxia/patologia , Hipóxia/terapia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/farmacologia , Cloreto de Potássio/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Volume de Ventilação Pulmonar/efeitos dos fármacos , Volume de Ventilação Pulmonar/fisiologia
13.
J Physiol ; 595(13): 4261-4277, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28332205

RESUMO

KEY POINTS: 5-HT is a neuromodulator released from carotid body (CB) chemoreceptor (type I) cells and facilitates the sensory discharge following chronic intermittent hypoxia (CIH). In the present study, we show that, in addition to type I cells, adjacent glial-like type II cells express functional, ketanserin-sensitive 5-HT2 receptors, and their stimulation increases cytoplasmic Ca2+ derived from intracellular stores. In type II cells, 5-HT activated a ketanserin-sensitive inward current (I5-HT ) that was similar to that (IUTP ) activated by the P2Y2R agonist, UTP. As previously shown for IUTP , I5-HT was inhibited by BAPTA-AM and carbenoxolone (5 µm), a putative blocker of ATP-permeable pannexin (Panx)-1 channels; IUTP was reversibly inhibited by the specific Panx-1 mimetic peptide channel blocker, 10 Panx peptide. Paracrine stimulation of type II cells by 5-HT, leading to ATP release via Panx-1 channels, may contribute to CB excitability, especially in pathophysiological conditions associated with CIH (e.g. obstructive sleep apnoea). ABSTRACT: Carotid body (CB) chemoreceptor (type I) cells can synthesize and release 5-HT and increased autocrine-paracrine 5-HT2 receptor signalling contributes to sensory long-term facilitation during chronic intermittent hypoxia (CIH). However, recent studies suggest that adjacent glial-like type II cells can respond to CB paracrine signals by elevating intracellular calcium (Δ[Ca2+ ]i ) and activating carbenoxolone-sensitive, ATP-permeable, pannexin (Panx)-1-like channels. In the present study, using dissociated rat CB cultures, we found that 5-HT induced Δ[Ca2+ ]i responses in a subpopulation of type I cells, as well as in most (∼67%) type II cells identified by their sensitivity to the P2Y2 receptor agonist, UTP. The 5-HT-induced Ca2+ response in type II cells was dose-dependent (EC50 ∼183 nm) and largely inhibited by the 5-HT2A receptor blocker, ketanserin (1 µm), and also arose mainly from intracellular stores. 5-HT also activated an inward current (I5-HT ) in type II cells (EC50 ∼200 nm) that was reversibly inhibited by ketanserin (1-10 nm), the Ca2+ chelator BAPTA-AM (5 µm), and low concentrations of carbenoxolone (5 µm), a putative Panx-1 channel blocker. I5-HT reversed direction at approximately -11 mV and was indistinguishable from the UTP-activated current (IUTP ). Consistent with a role for Panx-1 channels, IUTP was reversibly inhibited by the specific Panx-1 mimetic peptide blocker 10 Panx (100 µm), although not by its scrambled control peptide (sc Panx). Because ATP is an excitatory CB neurotransmitter, it is possible that the contribution of enhanced 5-HT signalling to the increased sensory discharge during CIH may occur, in part, by a boosting of ATP release from type II cells via Panx-1 channels.


Assuntos
Potenciais de Ação , Sinalização do Cálcio , Corpo Carotídeo/metabolismo , Células Quimiorreceptoras/metabolismo , Conexinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Agonistas do Receptor de Serotonina/farmacologia , Serotonina/farmacologia , Animais , Carbenoxolona/farmacologia , Corpo Carotídeo/citologia , Células Cultivadas , Células Quimiorreceptoras/efeitos dos fármacos , Conexinas/antagonistas & inibidores , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Ratos , Ratos Wistar
14.
Respir Physiol Neurobiol ; 237: 13-21, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28013061

RESUMO

A recent study showed that hypoxia activates a Ca2+-sensitive, Na+-permeable non-selective cation channel (NSC) in carotid body glomus cells. We studied the effects of mitochondrial inhibitors that increase Ca2+ influx via Ca2+ channel (Cav), and receptor agonists that release Ca2+ from endoplasmic reticulum (ER) on NSC. Mitochondrial inhibitors (NaCN, FCCP, H2S, NO) elevated [Ca2+]i and activated NSC. Angiotensin II and acetylcholine that elevate [Ca2+]i via the Gq-IP3 pathway activated NSC. However, endothelin-1 (Gq) and 5-HT (Gq) showed little or no effect on [Ca2+]i and did not activate NSC. Adenosine (Gs) caused a weak rise in [Ca2+]i but did not activate NSC. Dopamine (Gs) and γ-aminobytyric acid (Gi) were ineffective in raising [Ca2+]i and failed to activate NSC. Store-operated Ca2+ entry (SOCE) produced by depletion of Ca2+ stores with cyclopiazonic acid activated NSC. Our results show that Ca2+ entry via Cav, ER Ca2+ release and SOCE can activate NSC. Thus, NSC contributes to both voltage- and receptor-mediated excitation of glomus cells.


Assuntos
Cálcio/metabolismo , Corpo Carotídeo/citologia , Células Quimiorreceptoras/fisiologia , Canais Iônicos/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Angiotensina II/farmacologia , Anilidas/farmacologia , Animais , Animais Recém-Nascidos , Cafeína/farmacologia , Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Carbonil Cianeto p-Trifluormetoxifenil Hidrazona/farmacologia , Células Quimiorreceptoras/efeitos dos fármacos , Relação Dose-Resposta a Droga , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Inibidores Enzimáticos/farmacologia , Hipóxia/fisiopatologia , Ionóforos de Próton/farmacologia , Ratos , Ratos Sprague-Dawley , S-Nitroso-N-Acetilpenicilamina/farmacologia , Cianeto de Sódio/farmacologia , Tiadiazóis/farmacologia
15.
Exp Physiol ; 102(1): 34-47, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27763697

RESUMO

NEW FINDINGS: What is the central question of this study? In this study, we sought to investigate whether cardiovascular responses to peripheral chemoreflex activation of rats recovered from protein restriction are related to activation of AT1 receptors. What is the main finding and its importance? This study highlights the fact that angiotensinergic mechanisms activated by AT1 receptors do not support increased responses to peripheral chemoreflex activation by KCN in rats recovered from protein restriction. Also, we found that protein restriction led to increased resting ventilation in adult rats, even after recovery. The effects of a low-protein diet followed by recovery on cardiorespiratory responses to peripheral chemoreflex activation were tested before and after systemic angiotensin II type 1 (AT1 ) receptor antagonism. Male Fischer rats were divided into control and recovered (R-PR) groups after weaning. The R-PR rats were fed a low-protein (8%) diet for 35 days and recovered with a normal protein (20%) diet for 70 days. Control rats received a normal protein diet for 105 days (CG105 ). After cannulation surgery, mean arterial pressure, heart rate, respiratory frequency, tidal volume and minute ventilation were acquired using a digital recording system in freely moving rats. The role of angintensin II was evaluated by systemic antagonism of AT1 receptors with losartan (20 mg kg-1 i.v.). The peripheral chemoreflex was elicited by increasing doses of KCN (20-160 µg kg min-1 , i.v.). At baseline, R-PR rats presented increased heart rate and minute ventilation (372 ± 34 beats min-1 and 1.274 ± 377 ml kg-1  min-1 ) compared with CG105 animals (332 ± 22 beats min-1 and 856 ± 112 ml kg-1  min-1 ). Mean arterial pressure was not different between the groups. Pressor and bradycardic responses evoked by KCN (60 µg kg-1 ) were increased in R-PR (+45 ± 13 mmHg and -77 ± 47 beats min-1 ) compared with CG105 rats (+25 ± 17 mmHg and -27 ± 28 beats min-1 ), but no difference was found in the tachypnoeic response. These differences were preserved after losartan. The data suggest that angiotensin II acting on AT1 receptors may not be associated with the increased heart rate, increased minute ventilation and acute cardiovascular responses to peripheral chemoreflex activation in rats that underwent postweaning protein restriction followed by recovery.


Assuntos
Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatologia , Células Quimiorreceptoras/metabolismo , Células Quimiorreceptoras/fisiologia , Receptor Tipo 1 de Angiotensina/metabolismo , Reflexo/fisiologia , Angiotensina II/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Pressão Arterial/efeitos dos fármacos , Pressão Arterial/fisiologia , Bradicardia/metabolismo , Sistema Cardiovascular/efeitos dos fármacos , Células Quimiorreceptoras/efeitos dos fármacos , Dieta com Restrição de Proteínas/métodos , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Losartan/farmacologia , Masculino , Ratos , Ratos Endogâmicos F344 , Reflexo/efeitos dos fármacos , Taquicardia/metabolismo , Volume de Ventilação Pulmonar/efeitos dos fármacos , Volume de Ventilação Pulmonar/fisiologia
16.
Respir Physiol Neurobiol ; 234: 47-59, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27595979

RESUMO

Chronic exposure to intermittent hypoxia (CIH) elicits plasticity of the carotid sinus and phrenic nerves via reactive oxygen species (ROS). To determine whether CIH-induced alterations in ventilation, metabolism, and heart rate are also dependent on ROS, we measured responses to acute hypoxia in conscious rats after 14 and 21 d of either CIH or normoxia (NORM), with or without concomitant administration of allopurinol (xanthine oxidase inhibitor), combined allopurinol plus losartan (angiotensin II type 1 receptor antagonist), or apocynin (NADPH oxidase inhibitor). Carotid body nitrotyrosine production was measured by immunohistochemistry. CIH produced an increase in the ventilatory response to acute hypoxia that was virtually eliminated by all three pharmacologic interventions. CIH caused a robust increase in carotid body nitrotyrosine production that was greatly attenuated by allopurinol plus losartan and by apocynin but unaffected by allopurinol. CIH caused a decrease in metabolic rate and a reduction in hypoxic bradycardia. Both of these effects were prevented by allopurinol, allopurinol plus losartan, and apocynin.


Assuntos
Seio Carotídeo/metabolismo , Células Quimiorreceptoras/metabolismo , Hipóxia/patologia , Estresse Oxidativo/fisiologia , Respiração , Acetofenonas/farmacologia , Alopurinol/farmacologia , Análise de Variância , Animais , Antiarrítmicos/farmacologia , Antioxidantes/farmacologia , Peso Corporal/efeitos dos fármacos , Dióxido de Carbono/metabolismo , Seio Carotídeo/efeitos dos fármacos , Catecolaminas/sangue , Células Quimiorreceptoras/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Hipóxia/fisiopatologia , Losartan/farmacologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Consumo de Oxigênio/fisiologia , Pletismografia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Análise de Regressão , Respiração/efeitos dos fármacos , Volume de Ventilação Pulmonar/fisiologia , Fatores de Tempo , Tirosina/análogos & derivados , Tirosina/metabolismo
17.
Int J Mol Sci ; 17(6)2016 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-27258252

RESUMO

Scientific efforts to elucidate the mechanisms of chemical communication between organisms in marine environments are increasing. This study applied novel molecular technology to outline the effects of two xenobiotic drugs, deltamethrin (DM) and azamethiphos (AZA), on the neurotransmission system of the copepod ectoparasite Caligus rogercresseyi. Transcriptome sequencing and bioinformatics analyses were conducted to evaluate treatment effects on the glutamatergic synaptic pathway of the parasite, which is closely related to chemoreception and neurotransmission. After drug treatment with DM or AZA, stochastic mRNA expression patterns of glutamatergic synapse pathway components were observed. Both DM and AZA promoted a down-regulation of the glutamate-ammonia ligase, and DM activated a metabotropic glutamate receptor that is a suggested inhibitor of neurotransmission. Furthermore, the delousing drugs drove complex rearrangements in the distribution of mapped reads for specific metabotropic glutamate receptor domains. This study introduces a novel methodological approach that produces high-quality results from transcriptomic data. Using this approach, DM and AZA were found to alter the expression of numerous mRNAs tightly linked to the glutamatergic signaling pathway. These data suggest possible new targets for xenobiotic drugs that play key roles in the delousing effects of antiparasitics in sea lice.


Assuntos
Copépodes/genética , Perfilação da Expressão Gênica/métodos , Glutamato-Amônia Ligase/genética , Praguicidas/farmacologia , Receptores de Glutamato Metabotrópico/genética , Análise de Sequência de RNA/métodos , Animais , Células Quimiorreceptoras/efeitos dos fármacos , Copépodes/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Nitrilas/farmacologia , Organotiofosfatos/farmacologia , Piretrinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
18.
J Physiol ; 594(2): 407-19, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26572090

RESUMO

KEY POINTS: ACh is an important modulator of breathing, including at the level of the retrotrapezoid nucleus (RTN), where evidence suggests that ACh is essential for the maintenance of breathing. Despite this potentially important physiological role, little is known about the mechanisms responsible for the cholinergic control of RTN function. In the present study, we show at the cellular level that ACh increases RTN chemoreceptor activity by a CO2/H(+) independent mechanism involving M1/M3 receptor-mediated inositol 1,4,5-trisphosphate/Ca(+2) signalling and downstream inhibition of KCNQ channels. These results dispel the theory that ACh is required for RTN chemoreception by showing that ACh, similar to serotonin and other modulators, controls the activity of RTN chemoreceptors without interfering with the mechanisms by which these cells sense H(+). By identifying the mechanisms by which wake-on neurotransmitters such as ACh modulate RTN chemoreception, the results of the present study provide a framework for understanding the molecular basis of the sleep-wake state-dependent control of breathing. ABSTRACT: ACh has long been considered important for the CO2/H(+)-dependent drive to breathe produced by chemosensitive neurons in the retrotrapezoid nucleus (RTN). However, despite this potentially important physiological role, almost nothing is known about the mechanisms responsible for the cholinergic control of RTN function. In the present study, we used slice-patch electrophysiology and pharmacological tools to characterize the effects of ACh on baseline activity and CO2/H(+)-sensitivity of RTN chemoreceptors, as well as to dissect the signalling pathway by which ACh activates these neurons. We found that ACh activates RTN chemoreceptors in a dose-dependent manner (EC50 = 1.2 µm). The firing response of RTN chemoreceptors to ACh was mimicked by a muscarinic receptor agonist (oxotremorine; 1 µm), and blunted by M1- (pirezenpine; 2 µm) and M3- (diphenyl-acetoxy-N-methyl-piperidine; 100 nm) receptor blockers, but not by a nicotinic-receptor blocker (mecamylamine; 10 µm). Furthermore, pirenzepine, diphenyl-acetoxy-N-methyl-piperidine and mecamylamine had no measurable effect on the CO2/H(+)-sensitivity of RTN chemoreceptors. The effects of ACh on RTN chemoreceptor activity were also blunted by inhibition of inositol 1,4,5-trisphosphate receptors with 2-aminoethoxydiphenyl borate (100 µm), depletion of intracellular Ca(2+) stores with thapsigargin (10 µm), inhibition of casein kinase 2 (4,5,6,7-tetrabromobenzotriazole; 10 µm) and blockade of KCNQ channels (XE991; 10 µm). These results show that ACh activates RTN chemoreceptors by a CO2/H(+) independent mechanism involving M1/M3 receptor-mediated inositol 1,4,5-trisphosphate/Ca(+2) signalling and downstream inhibition of KCNQ channels. Identifying the components of the signalling pathway coupling muscarinic receptor activation to changes in chemoreceptor activity may provide new potential therapeutic targets for the treatment of respiratory control disorders.


Assuntos
Acetilcolina/metabolismo , Células Quimiorreceptoras/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Canais de Potássio KCNQ/metabolismo , Bulbo/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Potenciais de Ação , Animais , Cálcio/metabolismo , Sinalização do Cálcio , Dióxido de Carbono/metabolismo , Células Quimiorreceptoras/efeitos dos fármacos , Células Quimiorreceptoras/fisiologia , Bulbo/citologia , Bulbo/fisiologia , Agonistas Muscarínicos/farmacologia , Antagonistas Nicotínicos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Ratos
19.
PLoS One ; 10(9): e0137362, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26333001

RESUMO

It has been postulated that prenatal cigarette smoke exposure (CSE) increases the risk for sudden infant death syndrome. The victims of infant death syndrome suffer from respiratory abnormalities, such as central apnea, diminished chemoreflex and alteration in respiratory pattern during sleep. However, no experimental evidence on CSE model exists to confirm whether prenatal CSE gives rise to reduction of neonatal central chemoreception in in vitro preparations in absence of peripheral sensory feedback. The aim of the present study was to test the hypothesis that maternal CSE during pregnancy depresses central chemoreception of the neonatal rats. The pregnant rats were divided into two groups, control (n = 8) and CSE (n = 8). Experiments were performed on neonatal (0-3days) rat pups. Fictive respiratory activity was monitored by recording the rhythmic discharge from the hypoglossal rootlets of the medullary slices obtained from the neonatal rats. The burst frequency (BF) and integrated amplitude (IA) of the discharge were analyzed. Their responses to acidified artificial cerebrospinal fluid (aCSF) were tested to indicate the change of the central chemosensitivity. Under condition of perfusing with standard aCSF (pH 7.4), no significant difference was detected between the two groups in either BF or IA (P>0.05). Under condition of perfusing with acidified aCSF (pH 7.0), BF was increased and IA was decreased in both groups (P<0.01). However, their change rates in the CSE group were obviously smaller than that in the control group, 66.98 ± 10.11% vs. 143.75 ± 15.41% for BF and -22.38 ± 2.51% vs. -44.90 ± 3.92% for IA (P<0.01). In conclusion, these observations, in a prenatal CSE model, provide important evidence that maternal smoking during pregnancy exerts adverse effects on central chemoreception of neonates.


Assuntos
Tronco Encefálico/efeitos dos fármacos , Células Quimiorreceptoras/efeitos dos fármacos , Exposição Materna , Nicotiana , Fumaça , Animais , Animais Recém-Nascidos , Tronco Encefálico/fisiopatologia , Feminino , Gravidez , Ratos , Ratos Sprague-Dawley
20.
Respir Physiol Neurobiol ; 215: 30-8, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25956223

RESUMO

Acute hypoxia depolarizes carotid body chemoreceptor (glomus) cells and elevates intracellular Ca(2+) concentration ([Ca(2+)]i). Recent studies suggest that hydrogen sulfide (H2S) may serve as an oxygen sensor/signal in the carotid body during acute hypoxia. To further test such a role for H2S, we studied the effects of H2S on the activity of TASK channel and [Ca(2+)]i, which are considered important for mediating the glomus cell response to hypoxia. Like hypoxia, NaHS (a H2S donor) inhibited TASK activity and elevated [Ca(2+)]i. To inhibit the production of H2S, glomus cells were incubated (3h) with inhibitors of cystathionine-ß-synthase and cystathionine-γ-lyase (DL-propargylglycine, aminooxyacetic acid, ß-cyano-L-alanine; 0.3 mM). SF7 fluorescence was used to assess the level of H2S production. The inhibitors blocked L-cysteine- and hypoxia-induced elevation of SF7 fluorescence intensity. In cells treated with the inhibitors, hypoxia produced an inhibition of TASK activity and a rise in [Ca(2+)]i, similar in magnitude to those observed in control cells. L-cysteine produced no effect on TASK activity or [Ca(2+)]i and did not affect hypoxia-induced inhibition of TASK and elevation of [Ca(2+)]i. These findings suggest that under normal conditions, H2S is not a major signal in hypoxia-induced modulation of TASK channels and [Ca(2+)]i in isolated glomus cells.


Assuntos
Cálcio/metabolismo , Corpo Carotídeo/citologia , Células Quimiorreceptoras/efeitos dos fármacos , Gasotransmissores/farmacologia , Sulfeto de Hidrogênio/farmacologia , Hipóxia/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Alcinos/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Células Quimiorreceptoras/citologia , Relação Dose-Resposta a Droga , Glicina/análogos & derivados , Glicina/farmacologia , Líquido Intracelular/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Proteínas do Tecido Nervoso , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Compostos de Sódio , Tacrolimo/análogos & derivados , Tacrolimo/farmacologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA