Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 831
Filtrar
1.
Neuroreport ; 32(16): 1307-1310, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34605451

RESUMEN

OBJECTIVE: The combination of opioids and ethanol can synergistically depress breathing and the acute ventilatory response to hypoxia. Multiple studies have shown that the underlying mechanisms for this may involve calcium channel inhibition in central neurons. But we have previously identified opioid receptors in the carotid bodies and shown that their activation inhibits calcium influx into the chemosensitive cells. Given that the carotid bodies contribute to the drive to breathe and underpin the acute hypoxic ventilatory response, we hypothesized that ethanol and opioids may act synergistically in these peripheral sensory organs to further inhibit calcium influx and therefore inhibit ventilation. METHODS: Carotid bodies were removed from 56 Sprague-Dawley rats (1021 days old) and then enzymatically dissociated to allow calcium imaging of isolated chemosensitive type I cells. Cells were stimulated with high K+ in the presence and absence of the µ-opioid agonist [D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) (10 µM), a maximal sublethal concentration of ethanol (3 g L-1, 65.1 mM) or a combination of both. RESULTS: DAMGO alone significantly inhibited Ca2+ influx but this effect was not potentiated by the high concentration of ethanol. CONCLUSION: These results indicate for the first time that while opioids may suppress breathing via an action at the level of the carotid bodies, ethanol is unlikely to potentiate inhibition via this pathway. Thus, the synergistic effects of ethanol and opioids on ventilatory parameters are likely mediated by central rather than peripheral actions.


Asunto(s)
Analgésicos Opioides/farmacología , Cuerpo Carotídeo/efectos de los fármacos , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Etanol/farmacología , Animales , Calcio/metabolismo , Cuerpo Carotídeo/metabolismo , Sinergismo Farmacológico , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
2.
J Neuroinflammation ; 18(1): 191, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34465362

RESUMEN

The carotid bodies are multimodal sensors that regulate various autonomic reflexes. Recent evidence demonstrates their role in immune reflex regulation. Our previous studies using the allergen (ovalbumin) sensitised and exposed Brown Norway rat model of asthma suggest that carotid bodies mediate asthmatic bronchoconstriction through a lysophosphatidic acid (LPA) receptor (LPAr)-protein kinase C epsilon (PKCε)-transient receptor potential vanilloid one channel (TRPV1) pathway. Whilst naïve carotid bodies respond to LPA, whether their response to LPA is enhanced in asthma is unknown. Here, we show that asthmatic sensitisation of Brown Norway rats involving repeated aerosolised allergen challenges over 6 days, results in an augmentation of the carotid bodies' acute sensitivity to LPA. Increased expression of LPAr in the carotid bodies and petrosal ganglia likely contributed to this sensitivity. Importantly, allergen sensitisation of the carotid bodies to LPA did not alter their hypoxic response, nor did hypoxia augment LPA sensitivity acutely. Our data demonstrate the ability of allergens to sensitise the carotid bodies, highlighting the likely role of the carotid bodies and blood-borne inflammatory mediators in asthma.


Asunto(s)
Asma/metabolismo , Cuerpo Carotídeo/efectos de los fármacos , Lisofosfolípidos/farmacología , Alérgenos , Animales , Cuerpo Carotídeo/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Endogámicas BN , Ratas Sprague-Dawley
4.
J Clin Pharm Ther ; 46(5): 1189-1193, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33460173

RESUMEN

WHAT IS KNOWN AND OBJECTIVE: Treating an opioid overdose using an opioid receptor antagonist (such as naloxone) makes mechanistic sense and can be effective. Unfortunately, the majority of current drug overdose deaths involve polysubstance use (i.e., an opioid plus a non-opioid). COMMENT: Respiratory depression induced by opioids results from excessive opioid molecules binding to opioid receptors. This effect can be reversed by an opioid receptor antagonist. However, the respiratory depression induced by non-opioid drugs is not due to action at opioid receptors; thus, an opioid receptor antagonist is ineffective in many of these cases. For respiratory depression induced by non-opioids, receptor antagonists are either not available (e.g., for propofol overdose) or there may be attendant risks with their use (e.g., seizures with flumazenil). This gives rise to a need for more effective ways to treat polysubstance overdose. WHAT IS NEW AND CONCLUSION: A new approach to treating opioid-induced respiratory depression due to drug overdose focuses on agents that stimulate respiratory drive rather than competing for opioid receptors. Such an approach is "agnostic" to the cause of the respiratory depression, so might be a potential way to treat polysubstance overdose.


Asunto(s)
Analgésicos Opioides/toxicidad , Sobredosis de Droga/tratamiento farmacológico , Naloxona/uso terapéutico , Antagonistas de Narcóticos/farmacología , Antagonistas de Narcóticos/uso terapéutico , Insuficiencia Respiratoria/tratamiento farmacológico , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/metabolismo , Sobredosis de Droga/fisiopatología , Humanos , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Atención Perioperativa
5.
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
6.
Respir Physiol Neurobiol ; 285: 103593, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33276092

RESUMEN

The carotid body's glomus cells are the primary sensors of hypoxia in mammals. Previous studies suggested that the glomus cells' hypoxia sensitivity is mediated by lactate in mice. This molecule increases the intracellular [Ca2+] and induces exocytosis in glomus cells, activating the carotid sinus nerve (the axons of chemoreceptive petrosal neurons). On the other hand, how lactate affects the activity of carotid body of rats is still unknown. We hypothesized that lactate activates the carotid body of rats. In Wistar rats, we measured the changes in the electrical properties of isolated glomus cells and petrosal chemoreceptive neurons in in situ preparations in response to different concentrations of lactate. Superfusion of both physiological and supraphysiological concentrations of lactate did not affect the membrane conductance and potential of glomus cells. Moreover, lactate injected into the carotid body did not activate the anatomically and physiologically identified chemoreceptive petrosal neurons. We conclude that the carotid body of Wistar rats is not sensitive to lactate.


Asunto(s)
Cuerpo Carotídeo/metabolismo , Células Quimiorreceptoras/fisiología , Ácido Láctico/metabolismo , Potenciales de la Membrana/fisiología , Animales , Cuerpo Carotídeo/efectos de los fármacos , Células Quimiorreceptoras/efectos de los fármacos , Ácido Láctico/farmacología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas , Ratas Wistar
7.
Pflugers Arch ; 473(1): 37-51, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33210151

RESUMEN

Carotid body (CB) hyperactivity promotes hypertension in response to chronic intermittent hypoxia (CIH). The plasma concentration of adrenaline is reported to be elevated in CIH and our previous work suggests that adrenaline directly activates the CB. However, a role for chronic adrenergic stimulation in mediating CB hyperactivity is currently unknown. This study evaluated whether beta-blocker treatment with propranolol (Prop) prevented the development of CB hyperactivity, vascular sympathetic nerve growth and hypertension caused by CIH. Adult male Wistar rats were assigned into 1 of 4 groups: Control (N), N + Prop, CIH and CIH + Prop. The CIH paradigm consisted of 8 cycles h-1, 8 h day-1, for 3 weeks. Propranolol was administered via drinking water to achieve a dose of 40 mg kg-1 day-1. Immunohistochemistry revealed the presence of both ß1 and ß2-adrenoceptor subtypes on the CB type I cell. CIH caused a 2-3-fold elevation in basal CB single-fibre chemoafferent activity and this was prevented by chronic propranolol treatment. Chemoafferent responses to hypoxia and mitochondrial inhibitors were attenuated by propranolol, an effect that was greater in CIH animals. Propranolol decreased respiratory frequency in normoxia and hypoxia in N and CIH. Propranolol also abolished the CIH mediated increase in vascular sympathetic nerve density. Arterial blood pressure was reduced in propranolol groups during hypoxia. Propranolol exaggerated the fall in blood pressure in most (6/7) CIH animals during hypoxia, suggestive of reduced sympathetic tone. These findings therefore identify new roles for ß-adrenergic stimulation in evoking CB hyperactivity, sympathetic vascular hyperinnervation and altered blood pressure control in response to CIH.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Cuerpo Carotídeo/efectos de los fármacos , Hipoxia , Propranolol/farmacología , Antagonistas Adrenérgicos beta , Animales , Dióxido de Carbono , Esquema de Medicación , Masculino , Ratas , Ratas Wistar , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Sistema Nervioso Simpático/efectos de los fármacos
8.
Am J Physiol Regul Integr Comp Physiol ; 320(3): R203-R212, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33206558

RESUMEN

Activation of the carotid body (CB) using intracarotid potassium cyanide (KCN) injection increases coronary blood flow (CoBF). This increase in CoBF is considered to be mediated by co-activation of both the sympathetic and parasympathetic nerves to the heart. However, whether cardiac sympathetic nerve activity (cardiac SNA) actually increases during CB activation has not been determined previously. We hypothesized that activation of the CB would increase directly recorded cardiac SNA, which would cause coronary vasodilatation. Experiments were conducted in conscious sheep implanted with electrodes to record cardiac SNA and diaphragmatic electromyography (dEMG), flow probes to record CoBF and cardiac output, and a catheter to record arterial pressure. Intracarotid KCN injection was used to activate the CB. To eliminate the contribution of metabolic demand on coronary flow, the heart was paced at a constant rate during CB chemoreflex stimulation. Intracarotid KCN injection resulted in a significant increase in directly recorded cardiac SNA frequency (from 24 ± 2 to 40 ± 4 bursts/min; P < 0.05) as well as a dose-dependent increase in mean arterial pressure (79 ± 15 to 88 ± 14 mmHg; P < 0.01) and CoBF (75 ± 37 vs. 86 ± 42 mL/min; P < 0.05). The increase in CoBF and coronary vascular conductance to intracarotid KCN injection was abolished after propranolol infusion, suggesting that the increased cardiac SNA mediates coronary vasodilatation. The pressor response to activation of the CB was abolished by pretreatment with intravenous atropine, but there was no change in the coronary flow response. Our results indicate that CB activation increases directly recorded cardiac SNA, which mediates vasodilatation of the coronary vasculature.


Asunto(s)
Cuerpo Carotídeo/efectos de los fármacos , Circulación Coronaria/efectos de los fármacos , Corazón/inervación , Cianuro de Potasio/farmacología , Sistema Nervioso Simpático/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Animales , Presión Arterial/efectos de los fármacos , Cuerpo Carotídeo/metabolismo , Estado de Conciencia , Femenino , Oveja Doméstica , Sistema Nervioso Simpático/fisiología , Factores de Tiempo
9.
Molecules ; 25(24)2020 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-33348537

RESUMEN

General anesthesia is obtained by administration of potent hypnotics, analgesics and muscle relaxants. Apart from their intended effects (loss of consciousness, pain relief and muscle relaxation), these agents profoundly affect the control of breathing, in part by an effect within the peripheral chemoreflex loop that originates at the carotid bodies. This review assesses the role of cholinergic chemotransmission in the peripheral chemoreflex loop and the mechanisms through which muscle relaxants and hypnotics interfere with peripheral chemosensitivity. Additionally, consequences for clinical practice are discussed.


Asunto(s)
Anestésicos Generales/farmacología , Cuerpo Carotídeo/efectos de los fármacos , Colinérgicos/farmacología , Neuronas Colinérgicas/efectos de los fármacos , Retraso en el Despertar Posanestésico/prevención & control , Hipnóticos y Sedantes/farmacología , Relajantes Musculares Centrales/farmacología , Acetilcolina/metabolismo , Anestesia General/efectos adversos , Anestesia General/métodos , Humanos , Propofol/farmacología , Receptores Nicotínicos/efectos de los fármacos , Respiración/efectos de los fármacos , Respiración Artificial
10.
Anesthesiology ; 133(5): 1046-1059, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32826405

RESUMEN

BACKGROUND: The degree to which different volatile anesthetics depress carotid body hypoxic response relates to their ability to activate TASK potassium channels. Most commonly, volatile anesthetic pairs act additively at their molecular targets. We examined whether this applied to carotid body TASK channels. METHODS: We studied halothane and isoflurane effects on hypoxia-evoked rise in intracellular calcium (Ca2+i, using the indicator Indo-1) in isolated neonatal rat glomus cells, and TASK single-channel activity (patch clamping) in native glomus cells and HEK293 cell line cells transiently expressing TASK-1. RESULTS: Halothane (5%) depressed glomus cell Ca2+i hypoxic response (mean ± SD, 94 ± 4% depression; P < 0.001 vs. control). Isoflurane (5%) had a less pronounced effect (53 ± 10% depression; P < 0.001 vs. halothane). A mix of 3% isoflurane/1.5% halothane depressed cell Ca2+i response (51 ± 17% depression) to a lesser degree than 1.5% halothane alone (79 ± 15%; P = 0.001), but similar to 3% isoflurane alone (44 ± 22%; P = 0.224), indicating subadditivity. Halothane and isoflurane increased glomus cell TASK-1/TASK-3 activity, but mixes had a lesser effect than that seen with halothane alone: 4% halothane/4% isoflurane yielded channel open probabilities 127 ± 55% above control, versus 226 ± 12% for 4% halothane alone (P = 0.009). Finally, in HEK293 cell line cells, progressively adding isoflurane (1.5 to 5%) to halothane (2.5%) reduced TASK-1 channel activity from 120 ± 38% above control, to 88 ± 48% (P = 0.034). CONCLUSIONS: In all three experimental models, the effects of isoflurane and halothane combinations were quantitatively consistent with the modeling of weak and strong agonists competing at a common receptor on the TASK channel.


Asunto(s)
Anestésicos por Inhalación/metabolismo , Cuerpo Carotídeo/metabolismo , Halotano/metabolismo , Isoflurano/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Cuerpo Carotídeo/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Combinación de Medicamentos , Interacciones Farmacológicas/fisiología , Células HEK293 , Halotano/farmacología , Humanos , Isoflurano/farmacología
11.
Int J Mol Sci ; 21(15)2020 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-32751703

RESUMEN

Dopamine (DA) is a well-studied neurochemical in the mammalian carotid body (CB), a chemosensory organ involved in O2 and CO2/H+ homeostasis. DA released from receptor (type I) cells during chemostimulation is predominantly inhibitory, acting via pre- and post-synaptic dopamine D2 receptors (D2R) on type I cells and afferent (petrosal) terminals respectively. By contrast, co-released ATP is excitatory at postsynaptic P2X2/3R, though paracrine P2Y2R activation of neighboring glial-like type II cells may boost further ATP release. Here, we tested the hypothesis that DA may also inhibit type II cell function. When applied alone, DA (10 µM) had negligible effects on basal [Ca2+]i in isolated rat type II cells. However, DA strongly inhibited [Ca2+]i elevations (Δ[Ca2+]i) evoked by the P2Y2R agonist UTP (100 µM), an effect opposed by the D2/3R antagonist, sulpiride (1-10 µM). As expected, acute hypercapnia (10% CO2; pH 7.4), or high K+ (30 mM) caused Δ[Ca2+]i in type I cells. However, these stimuli sometimes triggered a secondary, delayed Δ[Ca2+]i in nearby type II cells, attributable to crosstalk involving ATP-P2Y2R interactions. Interestingly sulpiride, or DA store-depletion using reserpine, potentiated both the frequency and magnitude of the secondary Δ[Ca2+]i in type II cells. In functional CB-petrosal neuron cocultures, sulpiride potentiated hypercapnia-induced Δ[Ca2+]i in type I cells, type II cells, and petrosal neurons. Moreover, stimulation of type II cells with UTP could directly evoke Δ[Ca2+]i in nearby petrosal neurons. Thus, dopaminergic inhibition of purinergic signalling in type II cells may help control the integrated sensory output of the CB during hypercapnia.


Asunto(s)
Cuerpo Carotídeo/metabolismo , Dopamina/metabolismo , Receptores de Dopamina D2/genética , Receptores Purinérgicos P2Y2/genética , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Dióxido de Carbono/metabolismo , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/crecimiento & desarrollo , Homeostasis/genética , Hidrógeno/metabolismo , Oxígeno/metabolismo , Agonistas del Receptor Purinérgico P2Y/farmacología , Ratas , Transducción de Señal/efectos de los fármacos , Sulpirida/farmacología , Uridina Trifosfato/farmacología
12.
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
13.
Int J Mol Sci ; 21(14)2020 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-32664461

RESUMEN

The sleep apnea-hypopnea syndrome (SAHS) involves periods of intermittent hypoxia, experimentally reproduced by exposing animal models to oscillatory PO2 patterns. In both situations, chronic intermittent hypoxia (CIH) exposure produces carotid body (CB) hyperactivation generating an increased input to the brainstem which originates sympathetic hyperactivity, followed by hypertension that is abolished by CB denervation. CB has dopamine (DA) receptors in chemoreceptor cells acting as DA-2 autoreceptors. The aim was to check if blocking DA-2 receptors could decrease the CB hypersensitivity produced by CIH, minimizing CIH-related effects. Domperidone (DOM), a selective peripheral DA-2 receptor antagonist that does not cross the blood-brain barrier, was used to examine its effect on CIH (30 days) exposed rats. Arterial pressure, CB secretory activity and whole-body plethysmography were measured. DOM, acute or chronically administered during the last 15 days of CIH, reversed the hypertension produced by CIH, an analogous effect to that obtained with CB denervation. DOM marginally decreased blood pressure in control animals and did not affect hypoxic ventilatory response in control or CIH animals. No adverse effects were observed. DOM, used as gastrokinetic and antiemetic drug, could be a therapeutic opportunity for hypertension in SAHS patients' resistant to standard treatments.


Asunto(s)
Antagonistas de Dopamina/farmacología , Hipertensión/tratamiento farmacológico , Hipoxia/tratamiento farmacológico , Receptores Dopaminérgicos/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/metabolismo , Células Quimiorreceptoras/efectos de los fármacos , Células Quimiorreceptoras/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Hipertensión/metabolismo , Hipoxia/metabolismo , Masculino , Ratas , Ratas Wistar , Apnea Obstructiva del Sueño/tratamiento farmacológico , Apnea Obstructiva del Sueño/metabolismo
14.
Am J Physiol Heart Circ Physiol ; 318(5): H1325-H1336, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32330089

RESUMEN

Unilateral carotid body denervation has been proposed as treatment for sympathetic-related human diseases such as systolic heart failure, hypertension, obstructive sleep apnea, and cardiometabolic diseases. The long-term therapeutic effects of carotid body removal will be maintained if the remnant "buffer nerves," that is, the contralateral carotid nerve and the aortic nerves that innervate second-order neurons at the solitary tract nuclei (NTS), do not modify their contributions to the cardiovascular chemoreflexes. Here, we studied the cardiovascular chemoreflexes 1 mo after unilateral carotid body denervation either by excision of the petrosal ganglion (petrosal ganglionectomy, which eliminates central carotid afferents) or exeresis of a segment of one carotid nerve (carotid neurectomy, which preserves central afferents). Cardiovascular chemoreflexes were induced by intravenous (iv) injections of sodium cyanide in pentobarbitone-anesthetized adult cats. After 1 mo of unilateral petrosal ganglionectomy, without significant changes in basal arterial pressure, the contribution of the contralateral carotid nerve to the chemoreflex increases in arterial pressure was enhanced without changes in the contribution provided by the aortic nerves. By contrast, after 1 mo of unilateral carotid neurectomy, the contribution of remnant buffer nerves to cardiovascular chemoreflexes remained unmodified. These results indicate that a carotid nerve interruption involving denervation of second-order chemosensory neurons at the NTS will trigger cardiovascular chemoreflex plasticity on the contralateral carotid pathway. Then, unilateral carotid body denervation as therapeutic tool should consider the maintenance of the integrity of carotid central chemoafferents to prevent plasticity on remnant buffer nerves.NEW & NOTEWORTHY Unilateral carotid body denervation has been proposed as treatment for sympathetic hyperactivity-related human disorders. Its therapeutic effectiveness for maintaining a persistent decrease in the sympathetic outflow activity will depend on the absence of compensatory chemoreflex plasticity in the remnant carotid and aortic afferents. Here, we suggest that the integrity of central afferents after carotid body denervation is essential to prevent the emergence of plastic functional changes on the contralateral "intact" carotid nerve.


Asunto(s)
Presión Arterial , Cuerpo Carotídeo/fisiología , Reflejo , Animales , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/cirugía , Gatos , Desnervación , Ganglio Geniculado/fisiología , Cianuro de Sodio/farmacología
15.
Respir Physiol Neurobiol ; 277: 103438, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32259688

RESUMEN

Hyperreflexia of the peripheral chemoreceptors is a potential contributor of apnoeas of prematurity (AoP). Recently, it was shown that elevated P2X3 receptor expression was associated with elevated carotid body afferent sensitivity. Therefore, we tested whether P2X3 receptor antagonism would reduce AoP known to occur in newborn rats. Unrestrained whole-body plethysmography was used to record breathing and from this the frequency of apnoeas at baseline and following administration of either a P2X3 receptor antagonist - AF-454 (5 mg/kg or 10 mg/kg s.c.) or vehicle was derived. In a separate group, we tested the effects of AF-454 (10 mg/kg) on the hypoxic ventilatory response (10 % FiO2). Ten but not 5 mg/kg AF-454 reduced the frequency of AoP and improved breathing regularity significantly compared to vehicle. Neither AF-454 (both 5 and 10 mg/kg) nor vehicle affected baseline respiration. However, P2X3 receptor antagonism (10 mg/kg) powerfully blunted hypoxic ventilatory response to 10 % FiO2. These data suggest that P2X3 receptors contribute to AoP and the hypoxic ventilatory response in newborn rats but play no role in the drive to breathe at rest.


Asunto(s)
Apnea/prevención & control , Antagonistas del Receptor Purinérgico P2X/uso terapéutico , Receptores Purinérgicos P2X3/fisiología , Animales , Animales Recién Nacidos , Apnea/fisiopatología , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/fisiopatología , Hipoxia/tratamiento farmacológico , Hipoxia/fisiopatología , Masculino , Pletismografía Total/métodos , Antagonistas del Receptor Purinérgico P2X/farmacología , Ratas , Ratas Wistar
16.
Am J Physiol Cell Physiol ; 318(2): C430-C438, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31913694

RESUMEN

We studied the mechanisms by which carotid body glomus (type 1) cells produce spontaneous Ca2+ oscillations in normoxia and hypoxia. In cells perfused with normoxic solution at 37°C, we observed relatively uniform, low-frequency Ca2+ oscillations in >60% of cells, with each cell showing its own intrinsic frequency and amplitude. The mean frequency and amplitude of Ca2+ oscillations were 0.6 ± 0.1 Hz and 180 ± 42 nM, respectively. The duration of each Ca2+ oscillation ranged from 14 to 26 s (mean of ∼20 s). Inhibition of inositol (1,4,5)-trisphosphate receptor and store-operated Ca2+ entry (SOCE) using 2-APB abolished Ca2+ oscillations. Inhibition of endoplasmic reticulum Ca2+-ATPase (SERCA) using thapsigargin abolished Ca2+ oscillations. ML-9, an inhibitor of STIM1 translocation, also strongly reduced Ca2+ oscillations. Inhibitors of L- and T-type Ca2+ channels (Cav; verapamil>nifedipine>TTA-P2) markedly reduced the frequency of Ca2+ oscillations. Thus, Ca2+ oscillations observed in normoxia were caused by cyclical Ca2+ fluxes at the ER, which was supported by Ca2+ influx via Ca2+ channels. Hypoxia (2-5% O2) increased the frequency and amplitude of Ca2+ oscillations, and Cav inhibitors (verapamil>nifedipine>>TTA-P2) reduced these effects of hypoxia. Our study shows that Ca2+ oscillations represent the basic Ca2+ signaling mechanism in normoxia and hypoxia in CB glomus cells.


Asunto(s)
Calcio/metabolismo , Cuerpo Carotídeo/metabolismo , Hipoxia/metabolismo , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Cuerpo Carotídeo/efectos de los fármacos , Línea Celular , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Femenino , Masculino , Nifedipino/farmacología , Ratas , Ratas Sprague-Dawley , Molécula de Interacción Estromal 1/metabolismo , Tapsigargina/farmacología
17.
Sci Rep ; 9(1): 18136, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31792232

RESUMEN

Recent research supports that over-activation of the carotid body plays a key role in metabolic diseases like type 2 diabetes. Supressing carotid body signalling through carotid sinus nerve (CSN) modulation may offer a therapeutic approach for treating such diseases. Here we anatomically and histologically characterised the CSN in the farm pig as a recommended path to translational medicine. We developed an acute in vivo porcine model to assess the application of kilohertz frequency alternating current (KHFAC) to the CSN of evoked chemo-afferent CSN responses. Our results demonstrate the feasibility of this approach in an acute setting, as KHFAC modulation was able to successfully, yet variably, block evoked chemo-afferent responses. The observed variability in blocking response is believed to reflect the complex and diverse anatomy of the porcine CSN, which closely resembles human anatomy, as well as the need for optimisation of electrodes and parameters for a human-sized nerve. Overall, these results demonstrate the feasibility of neuromodulation of the CSN in an anesthetised large animal model, and represent the first steps in driving KHFAC modulation towards clinical translation. Chronic recovery disease models will be required to assess safety and efficacy of this potential therapeutic modality for application in diabetes treatment.


Asunto(s)
Seno Carotídeo/inervación , Animales , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/fisiología , Seno Carotídeo/anatomía & histología , Seno Carotídeo/efectos de los fármacos , Electrodos Implantados , Femenino , Humanos , Conducción Nerviosa , Respiración , Cianuro de Sodio/farmacología , Porcinos
18.
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
19.
Exp Physiol ; 104(9): 1335-1342, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31161612

RESUMEN

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.


Asunto(s)
Aorta/cirugía , Arterias/cirugía , Cuerpo Carotídeo/cirugía , Animales , Aorta/efectos de los fármacos , Aorta/fisiología , Arterias/efectos de los fármacos , Barorreflejo/efectos de los fármacos , Barorreflejo/fisiología , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/fisiología , Células Quimiorreceptoras/efectos de los fármacos , Células Quimiorreceptoras/fisiología , Desnervación/métodos , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Hipertensión/fisiopatología , Masculino , Fenilefrina/farmacología , Presorreceptores/efectos de los fármacos , Presorreceptores/fisiología , Ratas , Ratas Wistar
20.
Br J Anaesth ; 122(6): e168-e179, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30915997

RESUMEN

BACKGROUND: Opioids are potent painkillers but come with serious adverse effects ranging from addiction to potentially lethal respiratory depression. A variety of drugs with separate mechanisms of action are available to prevent or reverse opioid-induced respiratory depression (OIRD). METHODS: The authors reviewed human studies on reversal of OIRD using models that describe and predict the time course of pharmacokinetics (PK) and pharmacodynamics (PD) of opioids and reversal agents and link PK to PD. RESULTS: The PKPD models differ in their basic structure to capture the specific pharmacological mechanisms by which reversal agents interact with opioid effects on breathing. The effect of naloxone, a competitive opioid receptor antagonist, is described by the combined effect-compartment receptor-binding model to quantify rate limitation at the level of drug distribution and receptor kinetics. The effects of reversal agents that act through non-opioidergic pathways, such as ketamine and the experimental drug GAL021, are described by physiological models, in which stimulants act at CO2 chemosensitivity, CO2-independent ventilation, or both. The PKPD analyses show that although all reversal strategies may be effective under certain circumstances, there are conditions at which reversal is less efficacious and sometimes even impossible. CONCLUSIONS: Model-based drug development is needed to design an 'ideal' reversal agent-that is, one that is not influenced by opioid receptor kinetics, does not interfere with opioid analgesia, has a rapid onset of action with long-lasting effects, and is devoid of adverse effects.


Asunto(s)
Analgésicos Opioides/efectos adversos , Antagonistas de Narcóticos/farmacología , Insuficiencia Respiratoria/inducido químicamente , Analgésicos Opioides/antagonistas & inhibidores , Cuerpo Carotídeo/efectos de los fármacos , Doxapram/farmacología , Diseño de Fármacos , Humanos , Modelos Biológicos , Naloxona/farmacología , Insuficiencia Respiratoria/prevención & control , Triazinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...