Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 114(6): 1413-1418, 2017 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-28115703

RESUMEN

Sleep apnea, which is the periodic cessation of breathing during sleep, is a major health problem affecting over 10 million people in the United States and is associated with several sequelae, including hypertension and stroke. Clinical studies suggest that abnormal carotid body (CB) activity may be a driver of sleep apnea. Because gaseous molecules are important determinants of CB activity, aberrations in their signaling could lead to sleep apnea. Here, we report that mice deficient in heme oxygenase-2 (HO-2), which generates the gaseous molecule carbon monoxide (CO), exhibit sleep apnea characterized by high apnea and hypopnea indices during rapid eye movement (REM) sleep. Similar high apnea and hypopnea indices were also noted in prehypertensive spontaneously hypertensive (SH) rats, which are known to exhibit CB hyperactivity. We identified the gaseous molecule hydrogen sulfide (H2S) as the major effector molecule driving apneas. Genetic ablation of the H2S-synthesizing enzyme cystathionine-γ-lyase (CSE) normalized breathing in HO-2-/- mice. Pharmacologic inhibition of CSE with l-propargyl glycine prevented apneas in both HO-2-/- mice and SH rats. These observations demonstrate that dysregulated CO and H2S signaling in the CB leads to apneas and suggest that CSE inhibition may be a useful therapeutic intervention for preventing CB-driven sleep apnea.


Asunto(s)
Monóxido de Carbono/metabolismo , Gasotransmisores/metabolismo , Sulfuro de Hidrógeno/metabolismo , Síndromes de la Apnea del Sueño/metabolismo , Animales , Cuerpo Carotídeo/metabolismo , Cuerpo Carotídeo/fisiopatología , Cistationina gamma-Liasa/genética , Cistationina gamma-Liasa/metabolismo , Femenino , Hemo Oxigenasa (Desciclizante)/genética , Hemo Oxigenasa (Desciclizante)/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Respiración/genética , Síndromes de la Apnea del Sueño/genética , Síndromes de la Apnea del Sueño/fisiopatología
2.
J Physiol ; 595(1): 63-77, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27506145

RESUMEN

KEY POINTS: The effects of short-term (ST; 10 days) and long-term (LT; 30 days) intermittent hypoxia (IH) on blood pressure (BP), breathing and carotid body (CB) chemosensory reflex were examined in adult rats. ST- and LT-IH treated rats exhibited hypertension, irregular breathing with apnoea and augmented the CB chemosensory reflex, with all these responses becoming normalized during recovery from ST- but not from LT-IH. The persistent cardiorespiratory responses to LT-IH were associated with elevated reactive oxygen species (ROS) levels in the CB and adrenal medulla, which were a result of DNA methylation-dependent suppression of genes encoding anti-oxidant enzymes (AOEs). Treating rats with decitabine either during LT-IH or during recovery from LT-IH prevented DNA methylation of AOE genes, normalized the expression of AOE genes and ROS levels, reversed the heightened CB chemosensory reflex and hypertension, and also stabilized breathing. ABSTRACT: Rodents exposed to chronic intermittent hypoxia (IH), simulating blood O2 saturation profiles during obstructive sleep apnoea (OSA), have been shown to exhibit a heightened carotid body (CB) chemosensory reflex and hypertension. CB chemosensory reflex activation also results in unstable breathing with apnoeas. However, the effect of chronic IH on breathing is not known. In the present study, we examined the effects of chronic IH on breathing along with blood pressure (BP) and assessed whether the autonomic responses are normalized after recovery from chronic IH. Studies were performed on adult, male, Sprague-Dawley rats exposed to either short-term (ST; 10 days) or long-term (LT, 30 days) IH. Rats exposed to either ST- or LT-IH exhibited hypertension, irregular breathing with apnoeas, an augmented CB chemosensory reflex as indicated by elevated CB neural activity and plasma catecholamine levels, and elevated reactive oxygen species (ROS) levels in the CB and adrenal medulla (AM). All these effects were normalized after recovery from ST-IH but not from LT-IH. Analysis of the molecular mechanisms underlying the persistent effects of LT-IH revealed increased DNA methylation of genes encoding anti-oxidant enzymes (AOEs). Treatment with decitabine, a DNA methylation inhibitor, either during LT-IH or during recovery from LT-IH, prevented DNA methylation, normalized the expression of AOE genes, ROS levels, CB chemosensory reflex and BP, and also stabilized breathing. These results suggest that persistent cardiorespiratory abnormalities caused by LT-IH are mediated by epigenetic re-programming of the redox state in the CB chemosensory reflex pathway.


Asunto(s)
Hipertensión/fisiopatología , Hipoxia/fisiopatología , Trastornos Respiratorios/fisiopatología , Aconitato Hidratasa/metabolismo , Médula Suprarrenal/metabolismo , Animales , Presión Sanguínea , Cuerpo Carotídeo/metabolismo , Cuerpo Carotídeo/fisiología , Catalasa/genética , Metilación de ADN , Epigénesis Genética , Expresión Génica , Glutatión Peroxidasa/genética , Hipertensión/sangre , Hipertensión/genética , Hipertensión/metabolismo , Hipoxia/sangre , Hipoxia/genética , Hipoxia/metabolismo , Masculino , Malondialdehído/metabolismo , Norepinefrina/sangre , Oxidación-Reducción , Peroxirredoxinas/genética , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Trastornos Respiratorios/sangre , Trastornos Respiratorios/genética , Trastornos Respiratorios/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/genética
3.
Proc Natl Acad Sci U S A ; 111(3): 1174-9, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24395806

RESUMEN

Oxygen (O2) sensing by the carotid body and its chemosensory reflex is critical for homeostatic regulation of breathing and blood pressure. Humans and animals exhibit substantial interindividual variation in this chemosensory reflex response, with profound effects on cardiorespiratory functions. However, the underlying mechanisms are not known. Here, we report that inherent variations in carotid body O2 sensing by carbon monoxide (CO)-sensitive hydrogen sulfide (H2S) signaling contribute to reflex variation in three genetically distinct rat strains. Compared with Sprague-Dawley (SD) rats, Brown-Norway (BN) rats exhibit impaired carotid body O2 sensing and develop pulmonary edema as a consequence of poor ventilatory adaptation to hypobaric hypoxia. Spontaneous Hypertensive (SH) rat carotid bodies display inherent hypersensitivity to hypoxia and develop hypertension. BN rat carotid bodies have naturally higher CO and lower H2S levels than SD rat, whereas SH carotid bodies have reduced CO and greater H2S generation. Higher CO levels in BN rats were associated with higher substrate affinity of the enzyme heme oxygenase 2, whereas SH rats present lower substrate affinity and, thus, reduced CO generation. Reducing CO levels in BN rat carotid bodies increased H2S generation, restoring O2 sensing and preventing hypoxia-induced pulmonary edema. Increasing CO levels in SH carotid bodies reduced H2S generation, preventing hypersensitivity to hypoxia and controlling hypertension in SH rats.


Asunto(s)
Monóxido de Carbono/química , Cuerpo Carotídeo/fisiología , Sulfuro de Hidrógeno/química , Hipertensión/metabolismo , Oxígeno/química , Edema Pulmonar/metabolismo , Animales , Peso Corporal , Catecolaminas/metabolismo , Cistationina gamma-Liasa/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Hipoxia , Inmunohistoquímica , Masculino , Consumo de Oxígeno , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Respiración , Transducción de Señal , Especificidad de la Especie , Nervios Esplácnicos/patología
4.
J Neurophysiol ; 115(1): 345-54, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26561606

RESUMEN

Chronic intermittent hypoxia (CIH) is a hallmark manifestation of sleep apnea. A heightened carotid body activity and the resulting chemosensory reflex mediate increased sympathetic nerve activity by CIH. However, the mechanisms underlying heightened carotid body activity by CIH are not known. An elevation of intracellular calcium ion concentration ([Ca(2+)]i) in glomus cells, the primary oxygen-sensing cells, is an essential step for carotid body activation by hypoxia. In the present study, we examined the effects of CIH on the glomus cell [Ca(2+)]i response to hypoxia and assessed the underlying mechanisms. Glomus cells were harvested from adult rats or wild-type mice treated with 10 days of either room air (control) or CIH (alternating cycles of 15 s of hypoxia and 5 min of room air; 9 episodes/h; 8 h/day). CIH-treated glomus cells exhibited an enhanced [Ca(2+)]i response to hypoxia, and this effect was absent in the presence of 2-(4-cyclopropylphenyl)-N-((1R)-1-[5-[(2,2,2-trifluoroethyl)oxo]-pyridin-2-yl]ethyl)acetamide (TTA-A2), a specific inhibitor of T-type Ca(2+) channels, and in voltage-gated calcium channel, type 3.2 (CaV3.2), null glomus cells. CaV3.2 knockout mice exhibited an absence of CIH-induced hypersensitivity of the carotid body. CIH increased reactive oxygen species (ROS) levels in glomus cells. A ROS scavenger prevented the exaggerated TTA-A2-sensitive [Ca(2+)]i response to hypoxia. CIH had no effect on CaV3.2 mRNA levels. CIH augmented Ca(2+) currents and increased CaV3.2 protein in plasma membrane fractions of human embryonic kidney-293 cells stably expressing CaV3.2, and either a ROS scavenger or brefeldin-A, an inhibitor of protein trafficking, prevented these effects. These findings suggest that CIH leads to an augmented Ca(2+) influx via ROS-dependent facilitation of CaV3.2 protein trafficking to the plasma membrane.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Calcio/metabolismo , Cuerpo Carotídeo/metabolismo , Hipoxia/metabolismo , Animales , Bencenoacetamidas/administración & dosificación , Canales de Calcio Tipo T/fisiología , Cuerpo Carotídeo/efectos de los fármacos , Hipoxia de la Célula , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas , Piridinas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(19): E1788-96, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23610397

RESUMEN

Breathing and blood pressure are under constant homeostatic regulation to maintain optimal oxygen delivery to the tissues. Chemosensory reflexes initiated by the carotid body and catecholamine secretion from the adrenal medulla are the principal mechanisms for maintaining respiratory and cardiovascular homeostasis; however, the underlying molecular mechanisms are not known. Here, we report that balanced activity of hypoxia-inducible factor-1 (HIF-1) and HIF-2 is critical for oxygen sensing by the carotid body and adrenal medulla, and for their control of cardio-respiratory function. In Hif2α(+/-) mice, partial HIF-2α deficiency increased levels of HIF-1α and NADPH oxidase 2, leading to an oxidized intracellular redox state, exaggerated hypoxic sensitivity, and cardio-respiratory abnormalities, which were reversed by treatment with a HIF-1α inhibitor or a superoxide anion scavenger. Conversely, in Hif1α(+/-) mice, partial HIF-1α deficiency increased levels of HIF-2α and superoxide dismutase 2, leading to a reduced intracellular redox state, blunted oxygen sensing, and impaired carotid body and ventilatory responses to chronic hypoxia, which were corrected by treatment with a HIF-2α inhibitor. None of the abnormalities observed in Hif1α(+/-) mice or Hif2α(+/-) mice were observed in Hif1α(+/-);Hif2α(+/-) mice. These observations demonstrate that redox balance, which is determined by mutual antagonism between HIF-α isoforms, establishes the set point for hypoxic sensing by the carotid body and adrenal medulla, and is required for maintenance of cardio-respiratory homeostasis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cuerpo Carotídeo/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Oxígeno/metabolismo , Médula Suprarrenal/fisiología , Animales , Presión Sanguínea , Sistema Cardiovascular , Cuerpo Carotídeo/metabolismo , Catecolaminas/metabolismo , Heterocigoto , Homeostasis , Hipoxia , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Células PC12 , Ratas , Superóxido Dismutasa/metabolismo
6.
Am J Physiol Cell Physiol ; 308(2): C146-54, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25377087

RESUMEN

Arterial blood O2 levels are detected by specialized sensory organs called carotid bodies. Voltage-gated Ca(2+) channels (VGCCs) are important for carotid body O2 sensing. Given that T-type VGCCs contribute to nociceptive sensation, we hypothesized that they participate in carotid body O2 sensing. The rat carotid body expresses high levels of mRNA encoding the α1H-subunit, and α1H protein is localized to glomus cells, the primary O2-sensing cells in the chemoreceptor tissue, suggesting that CaV3.2 is the major T-type VGCC isoform expressed in the carotid body. Mibefradil and TTA-A2, selective blockers of the T-type VGCC, markedly attenuated elevation of hypoxia-evoked intracellular Ca(2+) concentration, secretion of catecholamines from glomus cells, and sensory excitation of the rat carotid body. Similar results were obtained in the carotid body and glomus cells from CaV3.2 knockout (Cacna1h(-/-)) mice. Since cystathionine-γ-lyase (CSE)-derived H2S is a critical mediator of the carotid body response to hypoxia, the role of T-type VGCCs in H2S-mediated O2 sensing was examined. Like hypoxia, NaHS, a H2S donor, increased intracellular Ca(2+) concentration and augmented carotid body sensory nerve activity in wild-type mice, and these effects were markedly attenuated in Cacna1h(-/-) mice. In wild-type mice, TTA-A2 markedly attenuated glomus cell and carotid body sensory nerve responses to hypoxia, and these effects were absent in CSE knockout mice. These results demonstrate that CaV3.2 T-type VGCCs contribute to the H2S-mediated carotid body response to hypoxia.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Canales de Calcio/metabolismo , Cuerpo Carotídeo/metabolismo , Sulfuro de Hidrógeno/farmacología , Hipoxia/metabolismo , Animales , Calcio/metabolismo , Cuerpo Carotídeo/efectos de los fármacos , Catecolaminas/metabolismo , Células Cultivadas , Células Quimiorreceptoras/efectos de los fármacos , Células Quimiorreceptoras/metabolismo , Cistationina gamma-Liasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxígeno/metabolismo , Ratas , Ratas Sprague-Dawley , Sulfuros
7.
Biochem J ; 458(2): 203-11, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24328859

RESUMEN

Increased catalytic activity of CBS (cystathionine ß-synthase) was recently shown to mediate vasodilation of the cerebral microcirculation, which is initiated within minutes of the onset of acute hypoxia. To test whether chronic hypoxia was a stimulus for increased CBS expression, U87-MG human glioblastoma and PC12 rat phaeochromocytoma cells were exposed to 1% or 20% O2 for 24-72 h. CBS mRNA and protein expression were increased in hypoxic cells. Hypoxic induction of CBS expression was abrogated in cells transfected with vector encoding shRNA targeting HIF (hypoxia-inducible factor) 1α or 2α. Exposure of rats to hypobaric hypoxia (0.35 atm; 1 atm=101.325 kPa) for 3 days induced increased CBS mRNA, protein and catalytic activity in the cerebral cortex and cerebellum, which was blocked by administration of the HIF inhibitor digoxin. HIF-binding sites, located 0.8 and 1.2 kb 5' to the transcription start site of the human CBS and rat Cbs genes respectively, were identified by ChIP assays. A 49-bp human sequence, which encompassed an inverted repeat of the core HIF-binding site, functioned as a hypoxia-response element in luciferase reporter transcription assays. Thus HIFs mediate tissue-specific CBS expression, which may augment cerebral vasodilation as an adaptive response to chronic hypoxia.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Cistationina betasintasa/biosíntesis , Regulación Enzimológica de la Expresión Génica , Hipoxia Encefálica/enzimología , Animales , Encéfalo/irrigación sanguínea , Células Cultivadas , Cistationina betasintasa/genética , Células HEK293 , Humanos , Hipoxia Encefálica/genética , Hipoxia Encefálica/patología , Factor 1 Inducible por Hipoxia/fisiología , Masculino , Células PC12 , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Distribución Tisular/genética , Vasodilatación/genética
8.
Adv Exp Med Biol ; 860: 195-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26303481

RESUMEN

Intermittent hypoxia (IH) increases reactive oxygen species generation resulting in oxidative stress in the adrenal medulla (AM), a major end-organ of the sympathetic nervous system which facilitates catecholamine secretion by hypoxia. Here, we show that carotid body chemoreflex contributes to IH-induced oxidative stress in the AM. Carotid bodies were ablated by cryocoagulation of glomus cells, the putative O(2) sensing cells. Carotid body ablated (CBA) and control rats were exposed to IH and the redox state of the AM was assessed biochemically. We found that IH raised reactive oxygen species levels along with an increase in NADPH oxidase (Nox), a pro-oxidant enzyme and a decrease in superoxide dismutase-2 (SOD2), an anti-oxidant enzyme. Further, IH increased hypoxia-inducible factor (HIF)-1α, whereas decreased HIF-2α, the transcriptional regulator of Nox and SOD-2, respectively. These IH-induced changes in the AM were absent in CBA rats. Moreover, IH increased splanchnic nerve activity and facilitated hypoxia-evoked catecholamine efflux from the AM and CBA prevented these effects. These findings suggest that IH-induced oxidative stress and catecholamine efflux in the AM occurs via carotid body chemoreflex involving HIF α isoform mediated imbalance in pro-, and anti-oxidant enzymes.


Asunto(s)
Médula Suprarrenal/metabolismo , Cuerpo Carotídeo/fisiología , Hipoxia/metabolismo , Estrés Oxidativo , Reflejo/fisiología , Animales , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Masculino , Ratas , Ratas Sprague-Dawley
9.
Proc Natl Acad Sci U S A ; 109(7): 2515-20, 2012 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-22232674

RESUMEN

Recurrent apnea with intermittent hypoxia is a major clinical problem in preterm infants. Recent studies, although limited, showed that adults who were born preterm exhibit increased incidence of sleep-disordered breathing and hypertension, suggesting that apnea of prematurity predisposes to autonomic dysfunction in adulthood. Here, we demonstrate that adult rats that were exposed to intermittent hypoxia as neonates exhibit exaggerated responses to hypoxia by the carotid body and adrenal chromaffin cells, which regulate cardio-respiratory function, resulting in irregular breathing with apneas and hypertension. The enhanced hypoxic sensitivity was associated with elevated oxidative stress, decreased expression of genes encoding antioxidant enzymes, and increased expression of pro-oxidant enzymes. Decreased expression of the Sod2 gene, which encodes the antioxidant enzyme superoxide dismutase 2, was associated with DNA hypermethylation of a single CpG dinucleotide close to the transcription start site. Treating neonatal rats with decitabine, an inhibitor of DNA methylation, during intermittent hypoxia exposure prevented oxidative stress, enhanced hypoxic sensitivity, and autonomic dysfunction. These findings implicate a hitherto uncharacterized role for DNA methylation in mediating neonatal programming of hypoxic sensitivity and the ensuing autonomic dysfunction in adulthood.


Asunto(s)
Epigénesis Genética , Corazón/fisiología , Homeostasis , Hipoxia/fisiopatología , Pulmón/fisiología , Animales , Animales Recién Nacidos , Islas de CpG , Metilación de ADN , Ratas
10.
Am J Physiol Cell Physiol ; 306(8): C745-52, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24477234

RESUMEN

The objective of the present study was to determine the impact of simulated apnea with intermittent hypoxia (IH) on endothelial barrier function and assess the underlying mechanism(s). Experiments were performed on human lung microvascular endothelial cells exposed to IH-consisting alternating cycles of 1.5% O2 for 30s followed by 20% O2 for 5 min. IH decreased transendothelial electrical resistance (TEER) suggesting attenuated endothelial barrier function. The effect of IH on TEER was stimulus dependent and reversible after reoxygenation. IH-exposed cells exhibited stress fiber formation and redistribution of cortactin, vascular endothelial-cadherins, and zona occludens-1 junction proteins along with increased intercellular gaps at cell-cell boundaries. Extracellular signal-regulated kinase (ERK) and c-jun NH2-terminal kinase (JNK) were phosphorylated in IH-exposed cells. Inhibiting either ERK or JNK prevented the IH-induced decrease in TEER and the reorganization of the cytoskeleton and junction proteins. IH increased reactive oxygen species (ROS) levels, and manganese (III) tetrakis (1-methyl-4-pyridyl) porphyrin pentachloride, a membrane-permeable antioxidant, prevented ERK and JNK phosphorylation as well as IH-induced changes in endothelial barrier function. These results demonstrate that IH via ROS-dependent activation of MAP kinases leads to reorganization of cytoskeleton and junction proteins resulting in endothelial barrier dysfunction.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Oxígeno/farmacología , Especies Reactivas de Oxígeno/metabolismo , Células Cultivadas , Citoesqueleto/fisiología , Impedancia Eléctrica , Activación Enzimática , Regulación Enzimológica de la Expresión Génica , Humanos , Hipoxia/metabolismo , Uniones Intercelulares/metabolismo , Pulmón/irrigación sanguínea , Estrés Oxidativo , Oxígeno/metabolismo , Fosforilación , Factores de Tiempo
11.
J Physiol ; 592(17): 3841-58, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24973414

RESUMEN

Previous studies reported that chronic intermittent hypoxia (CIH) results in an imbalanced expression of hypoxia-inducible factor-α (HIF-α) isoforms and oxidative stress in rodents, which may be due either to the direct effect of CIH or indirectly via hitherto uncharacterized mechanism(s). As neural activity is a potent regulator of gene transcription, we hypothesized that carotid body (CB) neural activity contributes to CIH-induced HIF-α isoform expression and oxidative stress in the chemoreflex pathway. Experiments were performed on adult rats exposed to CIH for 10 days. Rats exposed to CIH exhibited: increased HIF-1α and decreased HIF-2α expression; increased NADPH oxidase 2 and decreased superoxide dismutase 2 expression; and oxidative stress in the nucleus tractus solitarius and rostral ventrolateral medulla as well as in the adrenal medulla (AM), a major end organ of the sympathetic nervous system. Selective ablation of the CB abolished these effects. In the AM, sympathetic activation by the CB chemoreflex mediates CIH-induced HIF-α isoform imbalance via muscarinic acetylcholine receptor-mediated Ca(2+) influx, and the resultant activation of mammalian target of rapamycin pathway and calpain proteases. Rats exposed to CIH presented with hypertension, elevated sympathetic activity and increased circulating catecholamines. Selective ablation of either the CB (afferent pathway) or sympathetic innervation to the AM (efferent pathway) abolished these effects. These observations uncover CB neural activity-dependent regulation of HIF-α isoforms and the redox state by CIH in the central and peripheral nervous systems associated with the chemoreflex.


Asunto(s)
Cuerpo Carotídeo/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Estrés Oxidativo , Médula Suprarrenal/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Calcio/metabolismo , Calpaína/metabolismo , Hipoxia/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Reflejo , Núcleo Solitario/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
12.
Proc Natl Acad Sci U S A ; 108(7): 3065-70, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21288809

RESUMEN

Cardiorespiratory functions in mammals are exquisitely sensitive to changes in arterial O(2) levels. Hypoxia-inducible factors (e.g., HIF-1 and HIF-2) mediate transcriptional responses to reduced oxygen availability. We demonstrate that haploinsufficiency for the O(2)-regulated HIF-2α subunit results in augmented carotid body sensitivity to hypoxia, irregular breathing, apneas, hypertension, and elevated plasma norepinephrine levels in adult Hif-2α(+/-) mice. These dysregulated autonomic responses were associated with increased oxidative stress and decreased mitochondrial electron transport chain complex I activity in adrenal medullae as a result of decreased expression of major cytosolic and mitochondrial antioxidant enzymes. Systemic administration of a membrane-permeable antioxidant prevented oxidative stress, normalized hypoxic sensitivity of the carotid body, and restored autonomic functions in Hif-2α(+/-) mice. Thus, HIF-2α-dependent redox regulation is required for maintenance of carotid body function and cardiorespiratory homeostasis.


Asunto(s)
Médula Suprarrenal/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cuerpo Carotídeo/fisiología , Hipertensión/fisiopatología , Hipoxia/fisiopatología , Estrés Oxidativo/fisiología , Mecánica Respiratoria/fisiología , Análisis de Varianza , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Presión Sanguínea , Western Blotting , Complejo I de Transporte de Electrón/metabolismo , Perfilación de la Expresión Génica , Inmunohistoquímica , Ratones , Ratones Noqueados , Norepinefrina/metabolismo , Oxidación-Reducción , Consumo de Oxígeno , Pletismografía Total , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Exp Physiol ; 98(11): 1620-30, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23913764

RESUMEN

Chronic intermittent hypoxia (CIH) leads to remodelling of the carotid body function, manifested by an augmented sensory response to hypoxia and induction of sensory long-term facilitation (LTF). It was proposed that endothelin-1 (ET-1) contributes to CIH-induced hypoxic hypersensitivity of the carotid body. The objectives of the present study were as follows: (i) to delineate the mechanisms by which CIH upregulates ET-1 expression in the carotid body; and (ii) to assess whether ET-1 also contributes to sensory LTF. Experiments were performed on adult, male rats exposed to alternating cycles of 5% O2 (15 s) and room air (5 min), nine episodes per hour and 8 h per day for 10 days. Chronic intermittent hypoxia increased ET-1 levels in glomus cells without significantly altering prepro-endothelin-1 mRNA levels. The activity of endothelin-converting enzyme increased with concomitant elevation of ET-1 levels in CIH-exposed carotid bodies, and MnTMPyP, a membrane-permeable antioxidant, prevented these effects. Hypoxia facilitated ET-1 release from CIH-treated carotid bodies, which is a prerequisite for activation of ET receptors; however, hypoxia had no effect on ET-1 release from control carotid bodies. In CIH-exposed carotid bodies, mRNAs encoding ETA receptor were upregulated, and an ETA receptor-specific antagonist abolished CIH-induced hypersensitivity of the hypoxic response, whereas it had no effect on the sensory LTF. These results suggest that ECE-dependent increased production of ET-1 coupled with hypoxia-evoked ET-1 release and the ensuing ETA receptor activation mediate the CIH-induced carotid body hypersensitivity to hypoxia, but the ETA signalling pathway is not associated with sensory LTF elicited by CIH.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Cuerpo Carotídeo/fisiopatología , Hipoxia/fisiopatología , Metaloendopeptidasas/metabolismo , Estrés Oxidativo/fisiología , Animales , Antioxidantes/farmacología , Endotelina-1/metabolismo , Enzimas Convertidoras de Endotelina , Activación Enzimática , Masculino , Metaloporfirinas/farmacología , Plasticidad Neuronal/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Endotelina A/genética
14.
Proc Natl Acad Sci U S A ; 107(23): 10719-24, 2010 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-20556885

RESUMEN

Gaseousmessengers, nitric oxide and carbon monoxide, have been implicated in O2 sensing by the carotid body, a sensory organ that monitors arterial blood O2 levels and stimulates breathing in response to hypoxia. We now show that hydrogen sulfide (H2S) is a physiologic gasotransmitter of the carotid body, enhancing its sensory response to hypoxia. Glomus cells, the site of O2 sensing in the carotid body, express cystathionine gamma-lyase (CSE), an H2S-generating enzyme, with hypoxia increasing H2S generation in a stimulus-dependent manner. Mice with genetic deletion of CSE display severely impaired carotid body response and ventilatory stimulation to hypoxia, as well as a loss of hypoxia-evoked H2S generation. Pharmacologic inhibition of CSE elicits a similar phenotype in mice and rats. Hypoxia-evoked H2S generation in the carotid body seems to require interaction of CSE with hemeoxygenase-2, which generates carbon monoxide. CSE is also expressed in neonatal adrenal medullary chromaffin cells of rats and mice whose hypoxia-evoked catecholamine secretion is greatly attenuated by CSE inhibitors and in CSE knockout mice.


Asunto(s)
Cuerpo Carotídeo/fisiología , Sulfuro de Hidrógeno/metabolismo , Oxígeno/fisiología , Animales , Cistationina gamma-Liasa/deficiencia , Cistationina gamma-Liasa/metabolismo , Hipoxia/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratas
15.
Sci Adv ; 9(27): eadf3026, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37406126

RESUMEN

Oxygen (O2) sensing by the carotid body is critical for maintaining cardiorespiratory homeostasis during hypoxia. Hydrogen sulfide (H2S) signaling is implicated in carotid body activation by low O2. Here, we show that persulfidation of olfactory receptor 78 (Olfr78) by H2S is an integral component of carotid body activation by hypoxia. Hypoxia and H2S increased persulfidation in carotid body glomus cells and persulfidated cysteine240 in Olfr78 protein in heterologous system. Olfr78 mutants manifest impaired carotid body sensory nerve, glomus cell, and breathing responses to H2S and hypoxia. Glomus cells are positive for GOlf, adenylate cyclase 3 (Adcy3) and cyclic nucleotide-gated channel alpha 2 (Cnga2), key molecules of odorant receptor signaling. Adcy3 or Cnga2 mutants exhibited impaired carotid body and glomus cell responses to H2S and breathing responses to hypoxia. These results suggest that H2S through redox modification of Olfr78 participates in carotid body activation by hypoxia to regulate breathing.


Asunto(s)
Cuerpo Carotídeo , Sulfuro de Hidrógeno , Receptores Odorantes , Humanos , Receptores Odorantes/metabolismo , Hipoxia/metabolismo , Sulfuro de Hidrógeno/metabolismo , Cuerpo Carotídeo/metabolismo , Oxígeno/metabolismo
16.
Am J Physiol Cell Physiol ; 303(9): C916-23, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22744006

RESUMEN

H(2)S generated by the enzyme cystathionine-γ-lyase (CSE) has been implicated in O(2) sensing by the carotid body. The objectives of the present study were to determine whether glomus cells, the primary site of hypoxic sensing in the carotid body, generate H(2)S in an O(2)-sensitive manner and whether endogenous H(2)S is required for O(2) sensing by glomus cells. Experiments were performed on glomus cells harvested from anesthetized adult rats as well as age and sex-matched CSE(+/+) and CSE(-/-) mice. Physiological levels of hypoxia (Po(2) ∼30 mmHg) increased H(2)S levels in glomus cells, and dl-propargylglycine (PAG), a CSE inhibitor, prevented this response in a dose-dependent manner. Catecholamine (CA) secretion from glomus cells was monitored by carbon-fiber amperometry. Hypoxia increased CA secretion from rat and mouse glomus cells, and this response was markedly attenuated by PAG and in cells from CSE(-/-) mice. CA secretion evoked by 40 mM KCl, however, was unaffected by PAG or CSE deletion. Exogenous application of a H(2)S donor (50 µM NaHS) increased cytosolic Ca(2+) concentration ([Ca(2+)](i)) in glomus cells, with a time course and magnitude that are similar to that produced by hypoxia. [Ca(2+)](i) responses to NaHS and hypoxia were markedly attenuated in the presence of Ca(2+)-free medium or cadmium chloride, a pan voltage-gated Ca(2+) channel blocker, or nifedipine, an L-type Ca(2+) channel inhibitor, suggesting that both hypoxia and H(2)S share common Ca(2+)-activating mechanisms. These results demonstrate that H(2)S generated by CSE is a physiologic mediator of the glomus cell's response to hypoxia.


Asunto(s)
Cuerpo Carotídeo/metabolismo , Cistationina gamma-Liasa/metabolismo , Sulfuro de Hidrógeno/metabolismo , Hipoxia/metabolismo , Alquinos/farmacología , Animales , Cloruro de Cadmio/farmacología , Calcio/análisis , Bloqueadores de los Canales de Calcio/farmacología , Cuerpo Carotídeo/efectos de los fármacos , Catecolaminas/metabolismo , Cistationina gamma-Liasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Glicina/análogos & derivados , Glicina/farmacología , Masculino , Ratones , Nifedipino/farmacología , Cloruro de Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Sulfuros/farmacología
17.
Adv Exp Med Biol ; 758: 381-5, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23080186

RESUMEN

Although continuous hypoxia (CH) and intermittent hypoxia (IH) cause reduction in oxygen availability, organisms adapt to the effects of chronic CH whereas IH adversely impacts autonomic functions. Catecholamines are expressed both in the central and peripheral nervous systems and they play important roles in the regulation of cardio-respiratory functions during hypoxia. Tyrosine hydroxylase (TH) is the rate-limiting enzyme for catecholamine synthesis. Several studies have examined the effects of hypoxia on catecholamines by focusing on the regulation of TH. In this article, we present a brief overview of the impact of chronic CH and IH on TH expression, activity and the associated cellular mechanism(s).


Asunto(s)
Hipoxia/enzimología , Tirosina 3-Monooxigenasa/metabolismo , Animales , Humanos , Fosfoproteínas Fosfatasas/fisiología , Proteínas Quinasas/fisiología , Especies Reactivas de Oxígeno/metabolismo
18.
Proc Natl Acad Sci U S A ; 106(4): 1199-204, 2009 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-19147445

RESUMEN

Intermittent hypoxia (IH) occurs in many pathological conditions including recurrent apneas. Hypoxia-inducible factors (HIFs) 1 and 2 mediate transcriptional responses to low O(2). A previous study showed that HIF-1 mediates some of the IH-evoked physiological responses. Because HIF-2alpha is an orthologue of HIF-1alpha, we examined the effects of IH on HIF-2alpha, the O(2)-regulated subunit expression, in pheochromocytoma 12 cell cultures. In contrast to the up-regulation of HIF-1alpha, HIF-2alpha was down-regulated by IH. Similar down-regulation of HIF-2alpha was also seen in carotid bodies and adrenal medullae from IH-exposed rats. Inhibitors of calpain proteases (ALLM, ALLN) prevented IH-evoked degradation of HIF-2alpha whereas inhibitors of prolyl hydroxylases or proteosome were ineffective. IH activated calpain proteases and down-regulated the endogenous calpain inhibitor calpastatin. IH-evoked HIF-2alpha degradation led to inhibition of SOD2 transcription, resulting in oxidative stress. Over-expression of transcriptionally active HIF-2alpha prevented IH-evoked oxidative stress and restored SOD2 activity. Systemic treatment of IH-exposed rats with ALLM rescued HIF-2alpha degradation and restored SOD2 activity, thereby preventing oxidative stress and hypertension. These observations demonstrate that, unlike continuous hypoxia, IH leads to down-regulation of HIF-2alpha via a calpain-dependent signaling pathway and results in oxidative stress as well as autonomic morbidities.


Asunto(s)
Apnea/enzimología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Calpaína/metabolismo , Estrés Oxidativo , Procesamiento Proteico-Postraduccional , Animales , Apnea/mortalidad , Sistema Nervioso Autónomo/efectos de los fármacos , Sistema Nervioso Autónomo/patología , Señalización del Calcio/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Calpaína/antagonistas & inhibidores , Hipoxia de la Célula/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacología , Estrés Oxidativo/efectos de los fármacos , Células PC12 , Unión Proteica/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/antagonistas & inhibidores
19.
J Neurosci ; 30(32): 10763-72, 2010 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-20705601

RESUMEN

Nearly 90% of premature infants experience the stress of intermittent hypoxia (IH) as a consequence of recurrent apneas (periodic cessation of breathing). In neonates, catecholamine secretion from the adrenal medulla is critical for maintaining homeostasis under hypoxic stress. We recently reported that IH treatment enhanced hypoxia-evoked catecholamine secretion and [Ca2+]i responses in neonatal rat adrenal chromaffin cells and involves reactive oxygen species (ROS). The purpose of the present study was to identify the source(s) of ROS generation and examine the mechanisms underlying the enhanced catecholamine secretion by IH. Neonatal rats of either sex (postal day 0-5) were exposed to either IH or normoxia. IH treatment increased NADPH oxidase (NOX) activity, upregulated NOX2 and NOX4 transcription in adrenal medullae, and a NOX inhibitor prevented the effects of IH on hypoxia-evoked chromaffin cell secretion. IH upregulated Cav3.1 and Cav3.2 T-type Ca2+ channel mRNAs via NOX/ROS signaling and augmented T-type Ca2+ current in IH-treated chromaffin cells. Mibefradil, a blocker of T-type Ca2+ channels attenuated the effects of hypoxia on [Ca2+]i and catecholamine secretion in IH-treated cells. In Ca2+-free medium, IH-treated cells exhibited higher basal [Ca2+]i levels and more pronounced [Ca2+]i responses to hypoxia compared with controls, and blockade of ryanodine receptors (RyRs) prevented these effects. RyR2 and RyR3 mRNAs were upregulated, RyR2 was S-glutathionylated in IH-treated adrenal medullae, and NOX/ROS inhibitors prevented these effects. These results demonstrate that neonatal IH treatment leads to NOX/ROS-dependent recruitment of T-type Ca2+ channels and RyRs, resulting in augmented [Ca2+]i mobilization and catecholamine secretion.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Catecolaminas/metabolismo , Células Cromafines/fisiología , Exocitosis/fisiología , Hipoxia/patología , NADPH Oxidasas/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Animales Recién Nacidos , Cloruro de Cadmio/farmacología , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/genética , Células Cromafines/efectos de los fármacos , Células Cromafines/metabolismo , Electroquímica/métodos , Inhibidores Enzimáticos/farmacología , Exocitosis/efectos de los fármacos , Femenino , Hipoxia/fisiopatología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Técnicas de Placa-Clamp , ARN Mensajero/metabolismo , Ratas , Canal Liberador de Calcio Receptor de Rianodina/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
20.
Am J Physiol Cell Physiol ; 300(4): C743-51, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21270298

RESUMEN

Central and peripheral neurons as well as neuroendocrine cells express a variety of neurotransmitters/modulators that play critical roles in regulation of physiological systems. The synthesis of several neurotransmitters/modulators is regulated by O(2)-requiring rate-limiting enzymes. Consequently, hypoxia resulting from perturbations in O(2) homeostasis can affect neuronal functions by altering neurotransmitter synthesis. Two broad categories of hypoxia are frequently encountered: continuous hypoxia (CH) and intermittent hypoxia (IH). CH is often seen during high altitude sojourns, whereas IH is experienced in sleep-disordered breathing with recurrent apneas (i.e., brief, repetitive cessations of breathing). This article presents what is currently known on the effects of both forms of hypoxia on neurotransmitter levels and neurotransmitter synthesizing enzymes in the central and peripheral nervous systems.


Asunto(s)
Hipoxia/metabolismo , Neurotransmisores/biosíntesis , Animales , Transmisión Sináptica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA