Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 472
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(13)2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-39000591

RESUMEN

Experimental evidence suggests that chronic intermittent hypoxia (CIH), a major hallmark of obstructive sleep apnea (OSA), boosts carotid body (CB) responsiveness, thereby causing increased sympathetic activity, arterial and pulmonary hypertension, and cardiovascular disease. An enhanced circulatory chemoreflex, oxidative stress, and NO signaling appear to play important roles in these responses to CIH in rodents. Since the guinea pig has a hypofunctional CB (i.e., it is a natural CB knockout), in this study we used it as a model to investigate the CB dependence of the effects of CIH on pulmonary vascular responses, including those mediated by NO, by comparing them with those previously described in the rat. We have analyzed pulmonary artery pressure (PAP), the hypoxic pulmonary vasoconstriction (HPV) response, endothelial function both in vivo and in vitro, and vascular remodeling (intima-media thickness, collagen fiber content, and vessel lumen area). We demonstrate that 30 days of the exposure of guinea pigs to CIH (FiO2, 5% for 40 s, 30 cycles/h) induces pulmonary artery remodeling but does not alter endothelial function or the contractile response to phenylephrine (PE) in these arteries. In contrast, CIH exposure increased the systemic arterial pressure and enhanced the contractile response to PE while decreasing endothelium-dependent vasorelaxation to carbachol in the aorta without causing its remodeling. We conclude that since all of these effects are independent of CB sensitization, there must be other oxygen sensors, beyond the CB, with the capacity to alter the autonomic control of the heart and vascular function and structure in CIH.


Asunto(s)
Modelos Animales de Enfermedad , Hipoxia , Arteria Pulmonar , Apnea Obstructiva del Sueño , Vasoconstricción , Animales , Cobayas , Apnea Obstructiva del Sueño/fisiopatología , Apnea Obstructiva del Sueño/metabolismo , Hipoxia/fisiopatología , Hipoxia/metabolismo , Arteria Pulmonar/fisiopatología , Arteria Pulmonar/metabolismo , Masculino , Fenilefrina/farmacología , Remodelación Vascular , Cuerpo Carotídeo/fisiopatología , Cuerpo Carotídeo/metabolismo , Endotelio Vascular/fisiopatología , Endotelio Vascular/metabolismo , Vasodilatación
2.
J Neurophysiol ; 125(6): 2054-2067, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33909496

RESUMEN

Chronic intermittent hypoxia (CIH) is a hallmark manifestation of obstructive sleep apnea (OSA), a widespread breathing disorder. CIH-treated rodents exhibit activation of the sympathetic nervous system and hypertension. Heightened carotid body (CB) activity has been implicated in CIH-induced hypertension. CB expresses high abundance of olfactory receptor (Olfr) 78, a G-protein coupled receptor. Olfr 78 null mice exhibit impaired CB sensory nerve response to acute hypoxia. Present study examined whether Olfr78 participates in CB-dependent activation of the sympathetic nervous system and hypertension in CIH-treated mice and in hemeoxygenase (HO)-2 null mice experiencing CIH as a consequence of naturally occurring OSA. CIH-treated wild-type (WT) mice showed hypertension, biomarkers of sympathetic nerve activation, and enhanced CB sensory nerve response to hypoxia and sensory long-term facilitation (sLTF), and these responses were absent in CIH-treated Olfr78 null mice. HO-2 null mice showed higher apnea index (AI) (58 ± 1.2 apneas/h) than WT mice (AI = 8 ± 0.8 apneas/h) and exhibited elevated blood pressure (BP), elevated plasma norepinephrine (NE) levels, and heightened CB sensory nerve response to hypoxia and sLTF. The magnitude of hypertension correlated with AI in HO-2 null mice. In contrast, HO-2/Olfr78 double null mice showed absence of elevated BP and plasma NE levels and augmented CB response to hypoxia and sLTF. These results demonstrate that Olfr78 participates in sympathetic nerve activation and hypertension and heightened CB activity in two murine models of CIH.NEW & NOTEWORTHY Carotid body (CB) sensory nerve activation is essential for sympathetic nerve excitation and hypertension in rodents treated with chronic intermittent hypoxia (CIH) simulating blood O2 profiles during obstructive sleep apnea (OSA). Here, we report that CIH-treated mice and hemeoxygenase (HO)-2-deficient mice, which show OSA phenotype, exhibit sympathetic excitation, hypertension, and CB activation. These effects are absent in Olfr78 null and Olfr78/HO-2 double null mice.


Asunto(s)
Cuerpo Carotídeo , Hipertensión , Hipoxia , Receptores Odorantes/metabolismo , Apnea Obstructiva del Sueño , Sistema Nervioso Simpático , Animales , Cuerpo Carotídeo/metabolismo , Cuerpo Carotídeo/fisiopatología , Enfermedad Crónica , Modelos Animales de Enfermedad , Hemo Oxigenasa (Desciclizante)/genética , Hipertensión/metabolismo , Hipertensión/fisiopatología , Hipoxia/etiología , Hipoxia/metabolismo , Hipoxia/fisiopatología , Masculino , Ratones , Ratones Noqueados , Norepinefrina/sangre , Receptores Odorantes/genética , Apnea Obstructiva del Sueño/complicaciones , Apnea Obstructiva del Sueño/metabolismo , Apnea Obstructiva del Sueño/fisiopatología , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología
3.
Am J Physiol Heart Circ Physiol ; 320(2): H511-H519, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33275519

RESUMEN

In sleep apnea, airway obstruction causes intermittent hypoxia (IH). In animal studies, IH-dependent hypertension is associated with loss of vasodilator hydrogen sulfide (H2S), and increased H2S activation of sympathetic nervous system (SNS) activity in the carotid body. We previously reported that inhibiting cystathionine γ-lyase (CSE) to prevent H2S synthesis augments vascular resistance in control rats. The goal of this study was to evaluate the contribution of IH-induced changes in CSE signaling to increased blood pressure and vascular resistance. We hypothesized that chronic IH exposure eliminates CSE regulation of blood pressure (BP) and vascular resistance. In rats instrumented with venous catheters, arterial telemeters, and flow probes on the main mesenteric artery, the CSE inhibitor dl-propargylglycine (PAG, 50 mg/kg/day i.v. for 5 days) increased BP in Sham rats but decreased BP in IH rats [in mmHg, Sham (n = 11): 114 ± 4 to 131 ± 6; IH (n = 8): 131 ± 8 to 115 ± 7 mmHg, P < 0.05]. PAG treatment increased mesenteric vascular resistance in Sham rats but decreased it in IH rats (day 5/day 1: Sham: 1.50 ± 0.07; IH: 0.85 ± 0.19, P < 0.05). Administration of the ganglionic blocker hexamethonium (to evaluate SNS activity) decreased mesenteric resistance in PAG-treated Sham rats more than in saline-treated Sham rats or PAG-treated IH rats. CSE immunoreactivity in IH carotid bodies compared with those from Sham rats. However, CSE staining in small mesenteric arteries was less in arteries from IH than in Sham rats but not different in larger arteries (inner diameter > 200 µm). These results suggest endogenous H2S regulates blood pressure and vascular resistance, but this control is lost after IH exposure with decreased CSE expression in resistance size arteries. IH exposure concurrently increases carotid body CSE expression and relative SNS control of blood pressure, suggesting both vascular and carotid body H2S generation contribute to blood pressure regulation.NEW & NOTEWORTHY These results suggest that CSE's protective role in the vasculature is impaired by simulated sleep apnea, which also upregulates CSE in the carotid body. Thus, this enzyme system can exert both pro- and antihypertensive effects and may contribute to elevated SNS outflow in sleep apnea.


Asunto(s)
Circulación Sanguínea , Presión Sanguínea , Gasotransmisores/metabolismo , Sulfuro de Hidrógeno/metabolismo , Síndromes de la Apnea del Sueño/metabolismo , Alquinos/farmacología , Animales , Antihipertensivos/farmacología , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/metabolismo , Cuerpo Carotídeo/fisiopatología , Cistationina gamma-Liasa/antagonistas & inhibidores , Cistationina gamma-Liasa/genética , Cistationina gamma-Liasa/metabolismo , Inhibidores Enzimáticos/farmacología , Gasotransmisores/sangre , Glicina/análogos & derivados , Glicina/farmacología , Hexametonio/farmacología , Sulfuro de Hidrógeno/sangre , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiopatología , Ratas , Ratas Sprague-Dawley , Síndromes de la Apnea del Sueño/fisiopatología , Resistencia Vascular
4.
Am J Physiol Regul Integr Comp Physiol ; 321(2): R197-R207, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34133244

RESUMEN

Tonic carotid body (CB) activity is reduced during exposure to cold and hyperoxia. We tested the hypotheses that cold water diving lowers CB chemosensitivity and augments CO2 retention more than thermoneutral diving. Thirteen subjects [age: 26 ± 4 yr; body mass index (BMI): 26 ± 2 kg/m2) completed two 4-h head-out water immersion protocols in a hyperbaric chamber (1.6 ATA) in cold (15°C) and thermoneutral (25°C) water. CB chemosensitivity was assessed with brief hypercapnic ventilatory response ([Formula: see text]) and hypoxic ventilatory response ([Formula: see text]) tests before dive, 80 and 160 min into the dive (D80 and D160, respectively), and immediately after and 60 min after dive. Data are reported as an absolute mean (SD) change from predive. End-tidal CO2 pressure increased during both the thermoneutral water dive [D160: +2 (3) mmHg; P = 0.02] and the cold water dive [D160: +1 (2) mmHg; P = 0.03]. Ventilation increased during the cold water dive [D80: 4.13 (4.38) and D160: 7.75 (5.23) L·min-1; both P < 0.01] and was greater than the thermoneutral water dive at both time points (both P < 0.01). [Formula: see text] was unchanged during the dive (P = 0.24) and was not different between conditions (P = 0.23). [Formula: see text] decreased during the thermoneutral water dive [D80: -3.45 (3.61) and D160: -2.76 (4.04) L·min·mmHg-1; P < 0.01 and P = 0.03, respectively] but not the cold water dive. However, [Formula: see text] was not different between conditions (P = 0.17). In conclusion, CB chemosensitivity was not attenuated during the cold stress diving condition and does not appear to contribute to changes in ventilation or CO2 retention.


Asunto(s)
Dióxido de Carbono/sangre , Cuerpo Carotídeo/fisiopatología , Frío , Reflejo de Inmersión , Buceo , Hipercapnia/fisiopatología , Hipoxia/fisiopatología , Pulmón/fisiopatología , Ventilación Pulmonar , Adulto , Cuerpo Carotídeo/metabolismo , Hemodinámica , Humanos , Hipercapnia/sangre , Hipoxia/sangre , Inmersión , Masculino , Oxígeno/sangre , Adulto Joven
5.
Circ Res ; 125(11): 989-1002, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31545149

RESUMEN

RATIONALE: Obesity leads to resistant hypertension and mechanisms are poorly understood, but high plasma levels of leptin have been implicated. Leptin increases blood pressure acting both centrally in the dorsomedial hypothalamus and peripherally. Sites of the peripheral hypertensive effect of leptin have not been identified. We previously reported that leptin enhanced activity of the carotid sinus nerve, which transmits chemosensory input from the carotid bodies (CBs) to the medullary centers, and this effect was abolished by nonselective blockers of Trp (transient receptor potential) channels. We searched our mouse CB transcriptome database and found that the Trpm7 (transient receptor potential melastatin 7) channel was the most abundant Trp channel. OBJECTIVE: To examine if leptin induces hypertension acting on the CB Trpm7. METHODS AND RESULTS: C57BL/6J (n=79), leptin receptor (LepRb) deficient db/db mice (n=22), and LepRb-EGFP (n=4) mice were used. CB Trpm7 and LepRb gene expression was determined and immunohistochemistry was performed; CB glomus cells were isolated and Trpm7-like current was recorded. Blood pressure was recorded continuously in (1) leptin-treated C57BL/6J mice with intact and denervated CB; (2) leptin-treated C57BL/6J mice, which also received a nonselective Trpm7 blocker FTY720 administered systemically or topically to the CB area; (3) leptin-treated C57BL/6J mice transfected with Trpm7 small hairpin RNA to the CB, and (4) Leprb deficient obese db/db mice before and after Leprb expression in CB. Leptin receptor and Trpm7 colocalized in the CB glomus cells. Leptin induced a nonselective cation current in these cells, which was inhibited by Trpm7 blockers. Leptin induced hypertension in C57BL/6J mice, which was abolished by CB denervation, Trpm 7 blockers, and Trpm7 small hairpin RNA applied to CBs. Leprb overexpression in CB of Leprb-deficient db/db mice demethylated the Trpm7 promoter, increased Trpm7 gene expression, and induced hypertension. CONCLUSIONS: We conclude that leptin induces hypertension acting on Trmp7 in CB, which opens horizons for new therapy.


Asunto(s)
Presión Sanguínea , Cuerpo Carotídeo/metabolismo , Hipertensión/inducido químicamente , Leptina , Receptores de Leptina/metabolismo , Canales Catiónicos TRPM/metabolismo , Animales , Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/fisiopatología , Desnervación , Modelos Animales de Enfermedad , Hipertensión/metabolismo , Hipertensión/fisiopatología , Hipertensión/prevención & control , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/complicaciones , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Transducción de Señal , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/genética
6.
J Neurosci ; 39(49): 9725-9737, 2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31641048

RESUMEN

The combination of hypoxia and hypercapnia during sleep produces arousal, which helps restore breathing and normalizes blood gases. Hypercapnia and hypoxia produce arousal in mammals by activating central (pH-sensitive) and peripheral (primarily O2-sensitive) chemoreceptors. The relevant chemoreceptors and the neuronal circuits responsible for arousal are largely unknown. Here we examined the contribution of two lower brainstem nuclei that could be implicated in CO2 and hypoxia-induced arousal: the retrotrapezoid nucleus (RTN), a CO2-responsive nucleus, which mediates the central respiratory chemoreflex; and the C1 neurons, which are hypoxia activated and produce arousal and blood pressure increases when directly stimulated. Additionally, we assessed the contribution of the carotid bodies (CBs), the main peripheral chemoreceptors in mammals, to hypoxia and CO2-induced arousal. In unanesthetized male rats, we tested whether ablation of the RTN, CBs, or C1 neurons affects arousal from sleep and respiratory responses to hypercapnia or hypoxia. The sleep-wake pattern was monitored by EEG and neck EMG recordings and breathing by whole-body plethysmography. The latency to arousal in response to hypoxia or hypercapnia was determined along with changes in ventilation coincident with the arousal. RTN lesions impaired CO2-induced arousal but had no effect on hypoxia-induced arousal. CB ablation impaired arousal to hypoxia and, to a lesser extent, hypercapnia. C1 neuron ablation had no effect on arousal. Thus, the RTN contributes to CO2-induced arousal, whereas the CBs contribute to both hypoxia and CO2-induced arousal. Asphyxia-induced arousal likely requires the combined activation of RTN, CBs and other central chemoreceptors.SIGNIFICANCE STATEMENT Hypercapnia and hypoxia during sleep elicit arousal, which facilitates airway clearing in the case of obstruction and reinstates normal breathing in the case of hypoventilation or apnea. Arousal can also be detrimental to health by interrupting sleep. We sought to clarify how CO2 and hypoxia cause arousal. We show that the retrotrapezoid nucleus, a brainstem nucleus that mediates the effect of brain acidification on breathing, also contributes to arousal elicited by CO2 but not hypoxia. We also show that the carotid bodies contribute predominantly to hypoxia-induced arousal. Lesions of the retrotrapezoid nucleus or carotid bodies attenuate, but do not eliminate, arousal to CO2 or hypoxia; therefore, we conclude that these structures are not the sole trigger of CO2 or hypoxia-induced arousal.


Asunto(s)
Nivel de Alerta , Cuerpo Carotídeo/fisiopatología , Hipercapnia/fisiopatología , Hipoxia/fisiopatología , Bulbo Raquídeo/fisiopatología , Síndromes de la Apnea del Sueño/fisiopatología , Animales , Análisis de los Gases de la Sangre , Presión Sanguínea , Electroencefalografía , Electromiografía , Concentración de Iones de Hidrógeno , Masculino , Pletismografía , Ratas , Ratas Sprague-Dawley , Mecánica Respiratoria
7.
Am J Physiol Regul Integr Comp Physiol ; 318(5): R961-R971, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32267729

RESUMEN

We examined the effect of acute intermittent hypoxia (IH) on sympathetic neural firing patterns and the role of the carotid chemoreceptors. We hypothesized exposure to acute IH would increase muscle sympathetic nerve activity (MSNA) via an increase in action potential (AP) discharge rates and within-burst firing. We further hypothesized any change in discharge patterns would be attenuated during acute chemoreceptor deactivation (hyperoxia). MSNA (microneurography) was assessed in 17 healthy adults (11 male/6 female; 31 ± 1 yr) during normoxic rest before and after 30 min of experimental IH. Prior to and following IH, participants were exposed to 2 min of 100% oxygen (hyperoxia). AP patterns were studied from the filtered raw MSNA signal using wavelet-based methodology. Compared with baseline, multiunit MSNA burst incidence (P < 0.01), AP incidence (P = 0.01), and AP content per burst (P = 0.01) were increased following IH. There was an increase in the probability of a particular AP cluster firing once (P < 0.01) and more than once (P = 0.03) per burst following IH. There was no effect of hyperoxia on multiunit MSNA at baseline or following IH (P > 0.05); however, hyperoxia following IH attenuated the probability of particular AP clusters firing more than once per burst (P < 0.01). Acute IH increases MSNA by increasing AP discharge rates and within-burst firing. A portion of the increase in within-burst firing following IH can be attributed to the carotid chemoreceptors. These data advance the mechanistic understanding of sympathetic activation following acute IH in humans.


Asunto(s)
Cuerpo Carotídeo/fisiopatología , Hipoxia/fisiopatología , Contracción Muscular , Músculo Esquelético/inervación , Oxígeno/sangre , Reclutamiento Neurofisiológico , Sistema Nervioso Simpático/fisiopatología , Potenciales de Acción , Adulto , Biomarcadores/sangre , Cuerpo Carotídeo/metabolismo , Femenino , Humanos , Hipoxia/sangre , Hipoxia/diagnóstico , Masculino , Factores de Tiempo
8.
Curr Hypertens Rep ; 22(4): 27, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32166464

RESUMEN

PURPOSE OF REVIEW: Patients with true resistant hypertension (RH) are characterized by having high sympathetic activity and therefore potentially benefit from treatments such as baroreflex amplification (baroreflex activation therapy (BAT) or endovascular baroreflex amplification therapy (EVBA)) or carotid body (CB) modulation. This review aims at providing an up-to-date overview of the available evidence regarding these two therapies. RECENT FINDINGS: In recent years, increasing evidence has confirmed the potential of baroreflex amplification, either electrically (Barostim neo) or mechanically (MobiusHD), to improve blood pressure control on short- and long-term with only few side effects, in patients with RH. Two studies regarding unilateral CB resection did not show a significant change in blood pressure. Only limited studies regarding CB modulation showed promising results for transvenous CB ablation, but not for unilateral CB resection. Despite promising results from mostly uncontrolled studies, more evidence regarding the safety and efficacy from ongoing large randomized sham-controlled trials is needed before baroreflex amplification and CB modulation can be implemented in routine clinical practice.


Asunto(s)
Barorreflejo/fisiología , Cuerpo Carotídeo , Terapia por Estimulación Eléctrica , Hipertensión/terapia , Implantación de Prótesis , Presión Sanguínea/fisiología , Cuerpo Carotídeo/fisiopatología , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/fisiopatología , Presorreceptores
9.
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
10.
Int J Mol Sci ; 21(17)2020 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-32825527

RESUMEN

The carotid body (CB) is an important organ located at the carotid bifurcation that constantly monitors the blood supplying the brain. During hypoxia, the CB immediately triggers an alarm in the form of nerve impulses sent to the brain. This activates protective reflexes including hyperventilation, tachycardia and vasoconstriction, to ensure blood and oxygen delivery to the brain and vital organs. However, in certain conditions, including obstructive sleep apnea, heart failure and essential/spontaneous hypertension, the CB becomes hyperactive, promoting neurogenic hypertension and arrhythmia. G-protein-coupled receptors (GPCRs) are very highly expressed in the CB and have key roles in mediating baseline CB activity and hypoxic sensitivity. Here, we provide a brief overview of the numerous GPCRs that are expressed in the CB, their mechanism of action and downstream effects. Furthermore, we will address how these GPCRs and signaling pathways may contribute to CB hyperactivity and cardiovascular and respiratory disease. GPCRs are a major target for drug discovery development. This information highlights specific GPCRs that could be targeted by novel or existing drugs to enable more personalized treatment of CB-mediated cardiovascular and respiratory disease.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Cuerpo Carotídeo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Enfermedades Respiratorias/metabolismo , Adenosina/metabolismo , Animales , Enfermedades Cardiovasculares/fisiopatología , Cuerpo Carotídeo/fisiopatología , Dopamina/metabolismo , Epinefrina/metabolismo , Humanos , Hipoxia/metabolismo , Transducción de Señal , Apnea Obstructiva del Sueño/metabolismo , Apnea Obstructiva del Sueño/fisiopatología
11.
Int J Mol Sci ; 21(14)2020 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-32698380

RESUMEN

The carotid body (CB) is responsible for the peripheral chemoreflex by sensing blood gases and pH. The CB also appears to act as a peripheral sensor of metabolites and hormones, regulating the metabolism. CB malfunction induces aberrant chemosensory responses that culminate in the tonic overactivation of the sympathetic nervous system. The sympatho-excitation evoked by CB may contribute to the pathogenesis of metabolic syndrome, inducing systemic hypertension, insulin resistance and sleep-disordered breathing. Several molecular pathways are involved in the modulation of CB activity, and their pharmacological manipulation may lead to overall benefits for cardiometabolic diseases. In this review, we will discuss the role of the CB in the regulation of metabolism and in the pathogenesis of the metabolic dysfunction induced by CB overactivity. We will also explore the potential pharmacological targets in the CB for the treatment of metabolic syndrome.


Asunto(s)
Cuerpo Carotídeo/fisiopatología , Síndrome Metabólico/fisiopatología , Animales , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/metabolismo , Descubrimiento de Drogas , Glucosa/metabolismo , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/metabolismo , Hipertensión/fisiopatología , Hipoxia/tratamiento farmacológico , Hipoxia/metabolismo , Hipoxia/fisiopatología , Resistencia a la Insulina , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/metabolismo , Terapia Molecular Dirigida
12.
J Cell Physiol ; 234(8): 13534-13543, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30609027

RESUMEN

Chronic intermittent hypoxia (CIH) is known to induce hypertension, but the mechanism is not well understood. We hypothesized that sensory plasticity of the carotid body (CB) and oxidative stress in the paraventricular nucleus (PVN) are involved in CIH-induced hypertension. In this study, rats were exposed to CIH for 28 days (intermittent hypoxia of 21% O2 for 60 s and 5% O2 for 30 s, cyclically repeated for 8 hr/day) and then randomly grouped for intracerebroventricular injection of 5-HT2 receptor antagonist ritanserin, Rho-associated protein kinase (ROCK) inhibitor Y-27632, and NADPH oxidase (NOX) inhibitor diphenyleneiodonium (DPI), respectively. We found that CIH increased blood pressure (BP), elevated carotid sinus nerve (CSN) and renal sympathetic nerve (RSN) activities, oxidative stress, and cell apoptosis in PVN. NOX-derived reactive oxygen species (ROS) production and cell apoptosis decreased when CIH-induced activation of 5-HT/5-HT2AR/PKC signaling was inhibited by ritanserin. In addition, RhoA expression was downregulated when oxidative stress was attenuated by DPI, while Y-27632 decreased the expression of endothelin-1, which is overexpressed in the vascular wall during hypertension. Moreover, treatment with ritanserin, DPI or Y-27632 attenuated the sensory plasticity and sympathetic hyperactivity as well as CIH-induced elevation of BP. In conclusion, CIH-induced activation of 5-HT/5-HT2AR/PKC signaling contributes to NOX-derived oxidative stress in PVN, which may cause sensory plasticity of CB, RSN hyperactivity, and elevated BP.


Asunto(s)
Cuerpo Carotídeo/fisiopatología , Hipoxia/fisiopatología , Plasticidad Neuronal/fisiología , Núcleo Hipotalámico Paraventricular/fisiopatología , Amidas/farmacología , Animales , Apoptosis/fisiología , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Cuerpo Carotídeo/efectos de los fármacos , Modelos Animales de Enfermedad , Endotelina-1/metabolismo , Inhibidores Enzimáticos/farmacología , Hipertensión/etiología , Hipertensión/fisiopatología , Hipoxia/complicaciones , Masculino , NADPH Oxidasas/antagonistas & inhibidores , Plasticidad Neuronal/efectos de los fármacos , Compuestos Onio/farmacología , Estrés Oxidativo , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/patología , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT2A/metabolismo , Transducción de Señal , Quinasas Asociadas a rho/antagonistas & inhibidores
13.
Physiology (Bethesda) ; 33(4): 281-297, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29897299

RESUMEN

Advances in our understanding of brain mechanisms for the hypoxic ventilatory response, coordinated changes in blood pressure, and the long-term consequences of chronic intermittent hypoxia as in sleep apnea, such as hypertension and heart failure, are giving impetus to the search for therapies to "erase" dysfunctional memories distributed in the carotid bodies and central nervous system. We review current network models, open questions, sex differences, and implications for translational research.


Asunto(s)
Cuerpo Carotídeo/fisiopatología , Hipoxia/fisiopatología , Presión Sanguínea/fisiología , Humanos , Hipertensión/fisiopatología , Caracteres Sexuales , Síndromes de la Apnea del Sueño/fisiopatología
14.
J Neurophysiol ; 122(4): 1386-1396, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31389742

RESUMEN

We sought to examine the effect of varying chemoreflex stress on sympathetic neural recruitment strategies during end-expiratory apnea. We hypothesized that increases in the firing frequency and probability of low-threshold axons at the asphyxic "break point" would be exaggerated during hypoxia and attenuated during hyperoxia. Multiunit muscle sympathetic nervous system activity (MSNA) (peroneal nerve microneurography) was measured in 10 healthy male subjects (31 ± 2 yr, 25 ± 1 kg/m2). Individuals completed maximal voluntary end-expiratory apnea under normoxic, hypoxic (inspired O2 fraction: 0.17 ± 0.01), and hyperoxic (inspired O2 fraction: 0.92 ± 0.03) conditions. Action potential (AP) patterns were examined from the filtered raw signal with wavelet-based methodology. Multiunit MSNA was increased (P ≤ 0.05) during normoxic apnea, because of an increase in the frequency and incidence of AP spikes (243 ± 75 to 519 ± 134 APs/min, P = 0.048; 412 ± 133 to 733 ± 185 APs/100 heartbeats, P = 0.02). Multiunit MSNA increased from baseline (P < 0.01) during hypoxic apnea, which was due to an increase in the frequency and incidence of APs (192 ± 59 to 952 ± 266 APs/min, P < 0.01; 326 ± 89 to 1,212 ± 327 APs/100 heartbeats, P < 0.01). Hypoxic apnea also resulted in an increase in the probability of a particular AP cluster firing more than once per burst (P < 0.01). Hyperoxia attenuated any increase in MSNA with apnea, such that no changes in multiunit MSNA or frequency or incidence of AP spikes were observed (P > 0.05). We conclude that increases in frequency and incidence of APs during apnea are potentiated during hypoxia and suppressed when individuals are hyperoxic, highlighting the important impact of chemoreflex stress in AP discharge patterns. The results may have implications for neural control of the circulation in recreational activities and/or clinical conditions prone to apnea.NEW & NOTEWORTHY Our results demonstrate that, compared with normoxic end-expiratory apnea, hypoxic apnea increases the frequency and incidence of action potential spikes as well as the probability of multiple firing. We further show that this response is suppressed when individuals are hyperoxic. These data highlight the potentially important role of chemoreflex stress in neural firing and recruitment and may have implications for neural control of the circulation in recreational and/or clinical conditions prone to apnea.


Asunto(s)
Apnea/fisiopatología , Dióxido de Carbono/metabolismo , Oxígeno/metabolismo , Reclutamiento Neurofisiológico , Reflejo , Sistema Nervioso Simpático/fisiopatología , Potenciales de Acción , Adulto , Cuerpo Carotídeo/fisiopatología , Humanos , Masculino , Nervio Peroneo/fisiopatología , Estrés Fisiológico
15.
Curr Hypertens Rep ; 21(11): 89, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31599367

RESUMEN

PURPOSE OF REVIEW: Obstructive sleep apnea (OSA), featured by chronic intermittent hypoxia (CIH), is an independent risk for systemic hypertension (HTN) and is associated with pulmonary hypertension (PH). The precise mechanisms underlying pulmonary vascular remodeling and PH in OSA are not fully understood. However, it has been suggested that lung tissue hypoxia, oxidative stress, and pro-inflammatory mediators following CIH exposure may contribute to PH. RECENT FINDINGS: New evidences obtained in preclinical OSA models support that an enhanced carotid body (CB) chemosensory reactiveness to oxygen elicits sympathetic and renin-angiotensin system (RAS) overflow, which contributes to HTN. Moreover, the ablation of the CBs abolished the sympathetic hyperactivity and HTN in rodents exposed to CIH. Accordingly, it is plausible that the enhanced CB chemosensory reactivity may contribute to the pulmonary vascular remodeling and PH through the overactivation of the sympathetic-RAS axis. This hypothesis is supported by the facts that (i) CB stimulation increases pulmonary arterial pressure, (ii) denervation of sympathetic fibers in pulmonary arteries reduces pulmonary remodeling and pulmonary arterial hypertension (PAH) in humans, and (iii) administration of angiotensin-converting enzyme (ACE) or blockers of Ang II type 1 receptor (ATR1) ameliorates pulmonary remodeling and PH in animal models. In this review, we will discuss the supporting evidence for a plausible contribution of the CB-induced sympathetic-RAS axis overflow on pulmonary vascular remodeling and PH induced by CIH, the main characteristic of OSA.


Asunto(s)
Cuerpo Carotídeo/fisiopatología , Hipertensión Pulmonar/fisiopatología , Hipoxia/fisiopatología , Sistema Renina-Angiotensina/fisiología , Apnea Obstructiva del Sueño/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Animales , Humanos , Hipertensión/etiología , Hipertensión/fisiopatología , Estrés Oxidativo , Apnea Obstructiva del Sueño/complicaciones
16.
Br J Neurosurg ; 33(5): 500-503, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31130023

RESUMEN

Purpose: The carotid body functions as a chemoreceptor and receives richer blood supply, by weight, than any other organ in the body. We review the literature regarding the anatomy, histology, and function of the carotid body and the incidence, functionality, and clinical relevance of carotid body tumors and paragangliomas. These lesions are often nonfunctional but can be associated with catecholamine secretion. Most patients are asymptomatic or present initially with a cervical mass. As the tumors grow, they can impinge on nearby cranial nerves. Although there is some debate, the dominant clinical strategy is to surgically resect these tumors as early as possible. If they are resected early, the risk of postoperative neurovascular injury is minimized. Methods: Literature search was performed using the PubMed database with focus on articles including descriptions of the carotid body and associated tumors. Results: We reviewed recent literature that related to the anatomy of the carotid body while also including carotid pargangliomas and associated diagnosis with treatment interventions. Conclusion: As the carotid body serves as a vital modulator of cardiovascular and respiratory functions, illustrates the importance of identifying potential carotid paragangliomas due its ability to impede function of the carotid body. By understanding carotid paraganglioma's distinct etiologies while also understanding proper diagnosis of tumors allows for early detection and appropriate treatment options.


Asunto(s)
Tumor del Cuerpo Carotídeo/cirugía , Cuerpo Carotídeo/cirugía , Paraganglioma/cirugía , Cuerpo Carotídeo/anatomía & histología , Cuerpo Carotídeo/fisiopatología , Tumor del Cuerpo Carotídeo/patología , Tumor del Cuerpo Carotídeo/fisiopatología , Humanos , Paraganglioma/patología , Paraganglioma/fisiopatología
17.
Exp Physiol ; 103(4): 473-482, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29359403

RESUMEN

NEW FINDINGS: What is the central question of this study? Chronic intermittent hypoxia (CIH) and one-kidney, one-clip experimental models lead to sympathetic overactivity and hypertension. The present study explored the impact of previous exposure to CIH on one-kidney, one-clip renal hypertension; we hypothesized that CIH potentiates its development. What is the main finding and its importance? The development of one-kidney, one-clip renal hypertension was attenuated by previous exposure to CIH, and this protective effect was eliminated by carotid body denervation. These findings indicate that inputs from peripheral chemoreceptors in CIH-preconditioned rats play a role in preventing the increase in sympathetic activity and arterial pressure induced by one-kidney, one-clip renal hypertension. ABSTRACT: Chronic intermittent hypoxia (CIH) and one-kidney, one-clip (1K, 1C) experimental models lead to sympathetic overactivity and hypertension. We hypothesized that previous exposure to CIH potentiates the development of 1K, 1C renal hypertension. Male rats were divided into the following four groups: Control-1K, 1C, maintained under normoxia followed by 1K, 1C surgery (n = 19); Control-Sham, maintained under normoxia, followed by sham surgery (n = 19); CIH-1K, 1C, exposed to CIH (10 days) and 1K, 1C surgery (n = 19); and CIH-Sham, exposed to CIH and sham surgery (n = 18). Animals were catheterized 8 days after 1K, 1C or Sham surgeries and cardiovascular and respiratory parameters recorded on the following day. Baseline mean arterial pressure was higher in Control-1K, 1C than in Control-Sham rats (P < 0.05) and was higher in CIH-1K, 1C than in CIH-Sham rats (P < 0.05). However, the increase in mean arterial pressure in CIH-1K, 1C animals was significantly blunted in comparison to Con-1K, 1C rats (P < 0.05), indicating that previous exposure to CIH attenuates the development of renal hypertension. Systemic administration of hexamethonium, a ganglionic blocker, promoted a larger hypotensive response in Con-1K, 1C compared with CIH-1K, 1C rats (P < 0.05), suggesting that sympathetic activity was attenuated in rats previously exposed to the CIH protocol. In addition, removal of the carotid bodies before 1K, 1C renal hypertension eliminated the protective effect of CIH preconditioning on the development of the 1K, 1C hypertension. We conclude that previous exposure to CIH attenuates the development of renal hypertension via a carotid body-dependent mechanism.


Asunto(s)
Hipertensión Renal/fisiopatología , Hipoxia/fisiopatología , Riñón/fisiopatología , Animales , Presión Arterial/efectos de los fármacos , Presión Arterial/fisiología , Sistema Cardiovascular/efectos de los fármacos , Sistema Cardiovascular/fisiopatología , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/fisiopatología , Bloqueadores Ganglionares/farmacología , Hexametonio/farmacología , Hipertensión Renal/inducido químicamente , Riñón/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiopatología
18.
Curr Hypertens Rep ; 20(6): 53, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29789952

RESUMEN

PURPOSE OF REVIEW: An abnormal heightened carotid body (CB) chemoreflex, which produces autonomic dysfunction and sympathetic overactivation, is the common hallmark of obstructive sleep apnea (OSA), resistant hypertension, systolic heart failure (HF), and cardiometabolic diseases. Accordingly, it has been proposed that the elimination of the CB chemosensory input to the brainstem may reduce the autonomic and cardiorespiratory alterations in sympathetic-associated diseases in humans. RECENT FINDINGS: A growing body of evidence obtained in preclinical animal models support that an enhanced CB discharge produces sympathetic hyperactivity, baroreflex sensitivity and heart rate variability impairment, breathing instability, hypertension, and insulin resistance. The elimination CB chemosensory input reduces the sympathetic hyperactivity, the elevated arterial blood pressure in OSA and hypertensive models, abolishes breathing instability and improves animal survival in HF models, and restores insulin tolerance in metabolic models. These results highlight the role played by the enhanced CB drive in the progression of sympathetic-related diseases and support the proposal that the surgical ablation of the CB is useful to restore the autonomic balance and normal cardiorespiratory function in humans. Accordingly, the CB ablation has been used in pilot human studies as a therapeutic treatment for resistant hypertension and HF-induced sympathetic hyperactivity. In this review, I will discuss the supporting evidence for a crucial contribution of the CB in the central autonomic dysfunction and the pros and cons of the CB ablation as a therapy to revert autonomic overactivation. The CB ablation could be a useful method to reverse the enhanced chemoreflex in HF and severe hypertension, but caution is required before extensive use of bilateral CB ablation, which abolished ventilatory responses to hypoxia and may impair baroreceptor function.


Asunto(s)
Técnicas de Ablación/métodos , Enfermedades del Sistema Nervioso Autónomo/cirugía , Cuerpo Carotídeo/cirugía , Animales , Sistema Nervioso Autónomo/fisiopatología , Enfermedades del Sistema Nervioso Autónomo/fisiopatología , Cuerpo Carotídeo/fisiopatología , Humanos , Hipertensión/fisiopatología , Hipertensión/terapia , Hipoxia/fisiopatología , Modelos Animales , Proyectos Piloto , Reflejo/fisiología
19.
Adv Exp Med Biol ; 1071: 103-108, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30357740

RESUMEN

The carotid body (CB) is organized in clusters of lobules containing type I cells and type II cells, in a ratio of approximately 4:1. The CB undergoes structural and functional changes during perinatal development, in response to a variety of environmental stimuli and in pathological conditions. Knowing that the CB acts as a metabolic sensor involved in the control of peripheral insulin sensitivity and that its overactivation contributes to the genesis of metabolic disturbances, herein we tested if diet-induced insulin resistance is associated with morphological alterations in the proportion of type I and type II cells in the CB. Diet induced insulin resistant model (HFHSu) was obtained by submitting Wistar rats to 14 weeks of 60% lipid-rich diet and 35% of sucrose in drinking water. The HFHSu group was compared with an aged-matched control group. Glucose tolerance and insulin sensitivity were measured in conscious animals before diet administration and 14 weeks after the diet protocol. The expression of tyrosine hydroxylase (TH) and nestin were assessed by immunohistochemistry to identify type I and type II cells, respectively. TH expression was also quantified by Western blot. As expected, 14 weeks of HFHSu diet induced a decrease in insulin sensitivity as well as in glucose tolerance. HFHsu diet increased the number of TH-positive type I cells by 192% and decreased nestin-postive type 2 cells by 74%. This increase in type II cells observed by immunohistochemistry correlates with an increase by 107% in TH expression quantified by Western blot. These results suggest that changes in CB morphology are associated with metabolic disturbances invoked by administration of a hypercaloric diet.


Asunto(s)
Cuerpo Carotídeo/fisiopatología , Dieta , Resistencia a la Insulina , Animales , Glucemia , Insulina , Ratas , Ratas Wistar
20.
Adv Exp Med Biol ; 1071: 137-142, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30357744

RESUMEN

The carotid bodies (CB) respond to changes in blood gases with neurotransmitter release, thereby increasing carotid sinus nerve firing frequency and ultimately correcting the pattern of breathing. It has previously been demonstrated that acute application of the adipokine leptin augments the hypoxic sensory response of the intact in-vitro CB (Pye RL, Roy A, Wilson RJ, Wyatt CN. FASEB J 30(1 Supplement):983.1, 2016) and isolated CB type I cell (Pye RL, Dunn EJ, Ricker EM, Jurcsisn JG, Barr BL, Wyatt CN. Arterial chemoreceptors in physiology and pathophysiology. Advances in experimental medicine and biology. Springer, Cham, 2015). This study's aim was to examine, in-vivo, if elevated leptin modulated CB function and breathing.Rats were fed high fat or control chow for 16-weeks. High fat fed (HFF) animals gained significantly more weight compared to control fed (CF) animals and had significantly higher serum leptin levels compared to CF. Utilizing whole-body plethysmography, HFF animals demonstrated significantly depressed breathing compared to CF at rest and during hypoxia. However, amplitudes in the change in breathing from rest to hypoxia were not significantly different between groups. CB type I cells were isolated and intracellular calcium levels recorded. Averaged and peak cellular hypoxic responses were not significantly different.Despite a small but significant rise in leptin, differences in breathing caused by high fat feeding are unlikely caused by an effect of leptin on CB type I cells. However, the possibility remains that leptin may have in-vivo postsynaptic effects on the carotid sinus nerve; this remains to be investigated.


Asunto(s)
Cuerpo Carotídeo/fisiopatología , Células Quimiorreceptoras/citología , Dieta Alta en Grasa , Hipoxia/fisiopatología , Respiración , Animales , Análisis de los Gases de la Sangre , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA