Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Basic Res Cardiol ; 118(1): 43, 2023 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-37801130

RESUMEN

Altered autonomic balance is a hallmark of numerous cardiovascular diseases, including myocardial infarction (MI). Although device-based vagal stimulation is cardioprotective during chronic disease, a non-invasive approach to selectively stimulate the cardiac parasympathetic system immediately after an infarction does not exist and is desperately needed. Cardiac vagal neurons (CVNs) in the brainstem receive powerful excitation from a population of neurons in the paraventricular nucleus (PVN) of the hypothalamus that co-release oxytocin (OXT) and glutamate to excite CVNs. We tested if chemogenetic activation of PVN-OXT neurons following MI would be cardioprotective. The PVN of neonatal rats was transfected with vectors to selectively express DREADDs within OXT neurons. At 6 weeks of age, an MI was induced and DREADDs were activated with clozapine-N-oxide. Seven days following MI, patch-clamp electrophysiology confirmed the augmented excitatory neurotransmission from PVN-OXT neurons to downstream nuclei critical for parasympathetic activity with treatment (43.7 ± 10 vs 86.9 ± 9 pA; MI vs. treatment), resulting in stark improvements in survival (85% vs. 95%; MI vs. treatment), inflammation, fibrosis assessed by trichrome blue staining, mitochondrial function assessed by Seahorse assays, and reduced incidence of arrhythmias (50% vs. 10% cumulative incidence of ventricular fibrillation; MI vs. treatment). Myocardial transcriptomic analysis provided molecular insight into potential cardioprotective mechanisms, which revealed the preservation of beneficial signaling pathways, including muscarinic receptor activation, in treated animals. These comprehensive results demonstrate that the PVN-OXT network could be a promising therapeutic target to quickly activate beneficial parasympathetic-mediated cellular pathways within the heart during the early stages of infarction.


Asunto(s)
Infarto del Miocardio , Oxitocina , Ratas , Animales , Oxitocina/farmacología , Oxitocina/metabolismo , Ratas Sprague-Dawley , Hipotálamo , Infarto del Miocardio/metabolismo , Neuronas/metabolismo , Arritmias Cardíacas/metabolismo
2.
Hypertension ; 80(4): 882-894, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36794581

RESUMEN

BACKGROUND: Obstructive sleep apnea is a prevalent and poorly treated cardiovascular disease that leads to hypertension and autonomic imbalance. Recent studies that restore cardiac parasympathetic tone using selective activation of hypothalamic oxytocin neurons have shown beneficial cardiovascular outcomes in animal models of cardiovascular disease. This study aimed to determine if chemogenetic activation of hypothalamic oxytocin neurons in animals with existing obstructive sleep apnea-induced hypertension would reverse or blunt the progression of autonomic and cardiovascular dysfunction. METHODS: Two groups of rats were exposed to chronic intermittent hypoxia (CIH), a model of obstructive sleep apnea, for 4 weeks to induce hypertension. During an additional 4 weeks of exposure to CIH, 1 group was treated with selective activation of hypothalamic oxytocin neurons while the other group was untreated. RESULTS: Hypertensive animals exposed to CIH and treated with daily hypothalamic oxytocin neuron activation had lower blood pressure, faster heart rate recovery times after exercise, and improved indices of cardiac function compared with untreated hypertensive animals. Microarray analysis suggested that, compared with treated animals, untreated animals had gene expression profiles associated with cellular stress response activation, hypoxia-inducible factor stabilization, and myocardial extracellular matrix remodeling and fibrosis. CONCLUSIONS: In animals already presenting with CIH-induced hypertension, chronic activation of hypothalamic oxytocin neurons blunted the progression of hypertension and conferred cardioprotection after an additional 4 weeks of CIH exposure. These results have significant clinical translation for the treatment of cardiovascular disease in patients with obstructive sleep apnea.


Asunto(s)
Enfermedades Cardiovasculares , Cardiopatías , Hipertensión , Apnea Obstructiva del Sueño , Ratas , Animales , Oxitocina/farmacología , Ratas Sprague-Dawley , Enfermedades Cardiovasculares/complicaciones , Modelos Animales de Enfermedad , Apnea Obstructiva del Sueño/complicaciones , Hipoxia/metabolismo , Neuronas/metabolismo
3.
JACC Clin Electrophysiol ; 6(14): 1827-1840, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33357580

RESUMEN

OBJECTIVES: This study sought to investigate the shift of leading pacemaker locations in healthy and failing mammalian hearts over the entire range of physiological heart rates (HRs), and to molecularly characterize spatial regions of spontaneous activity. BACKGROUND: A normal heartbeat originates as an action potential in a group of pacemaker cells known as the sinoatrial node (SAN), located near the superior vena cava. HRs and the anatomical site of origin of pacemaker activity in the adult heart are known to dynamically change in response to various physiological inputs, yet the mechanism of this pacemaker shift is not well understood. METHODS: Optical mapping was applied to ex vivo rat and human isolated right atrial tissues, and HRs were modulated with acetylcholine and isoproterenol. RNA sequencing was performed on tissue areas that elicited spontaneous activity, and comparisons were made to neighboring myocardial tissues. RESULTS: Functional and molecular evidence identified and confirmed the presence of 2 competing right atrial pacemakers localized near the superior vena cava and the inferior vena cava-the superior SAN (sSAN) and inferior SAN (iSAN), respectively-which preferentially control the fast and slow HRs. Both of these regions were evident in non-failing rat and human hearts and maintained spontaneous activity in the rat heart when physically separated from one another. Molecular analysis of these 2 pacemaker regions revealed unique but similar transcriptional profiles, suggesting iSAN dominance when the sSAN is silent. CONCLUSIONS: The presence of 2 spatially distinct dominant pacemakers, sSAN and iSAN, in the mammalian heart clarifies previous identification of migrating pacemakers and corresponding changes in P-wave morphology in mammalian species.


Asunto(s)
Nodo Sinoatrial , Vena Cava Superior , Potenciales de Acción , Animales , Atrios Cardíacos , Frecuencia Cardíaca , Humanos , Ratas
4.
Artículo en Inglés | MEDLINE | ID: mdl-33006920

RESUMEN

BACKGROUND: Left ventricular (LV) electrical maladaptation to increased heart rate in failing myocardium contributes to morbidity and mortality. Recently, cardiac cholinergic neuron activation reduced loss of contractile function resulting from chronic trans-aortic constriction (TAC) in rats. We hypothesized that chronic activation of cardiac cholinergic neurons would also reduce TAC-induced derangement of cardiac electrical activity. METHODS: We investigated electrophysiological rate adaptation in TAC rat hearts with and without daily chemogenetic activation of hypothalamic oxytocin neurons for downstream cardiac cholinergic neuron stimulation. Sprague Dawley rat hearts were excised, perfused, and optically mapped under dynamic pacing after 16 weeks of TAC with or without 12 weeks of daily chemogenetic treatment. Action potential duration (APD60) and conduction velocity (CV) maps were analyzed for regional rate adaptation to dynamic pacing. RESULTS: At lower pacing rates, untreated TAC induced elevated LV epicardial APD60. Fitted APD60 steady state (APDss) was reduced in treated TAC hearts. At higher pacing rates, treatment heterogeneously reduced APD60 compared to untreated TAC hearts. Variance of conduction loss was reduced in treated hearts compared to untreated hearts during fast pacing. However, CV was markedly reduced in both treated and untreated TAC hearts throughout dynamic pacing. At 150msec pacing cycle length, APD60 v. diastolic interval (DI) dispersion was reduced in treated hearts compared to untreated hearts. CONCLUSIONS: Chronic activation of cardiac cholinergic neurons improved electrophysiological adaptation to increases in pacing rate during development of TAC-induced heart failure. This provides insight into the electrophysiological benefits of cholinergic stimulation as a treatment for heart failure patients.

5.
Am J Physiol Heart Circ Physiol ; 319(6): H1153-H1161, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33035444

RESUMEN

Neural control of the heart is regulated by sympathetic and parasympathetic divisions of the autonomic nervous system, both opposing each other to maintain cardiac homeostasis via regulating heart rate, conduction velocity, force of contraction, and coronary blood flow. Sympathetic hyperactivity and diminished parasympathetic activity are the characteristic features of many cardiovascular disease states including hypertension, myocardial ischemia, and arrhythmias that result in heart failure. Restoring parasympathetic activity to the heart has recently been identified as the promising approach to treat such conditions. However, approaches that used vagal nerve stimulation have been shown to be unsuccessful in heart failure. This review focuses on novel chemogenetic approaches used to identify the cardioprotective nature of activating neural points along the vagal pathway (both central and peripheral) while being selectively therapeutic in heart failure and obstructive sleep apnea.


Asunto(s)
Colina O-Acetiltransferasa/metabolismo , Activadores de Enzimas/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Corazón/inervación , Agonistas Muscarínicos/uso terapéutico , Receptores Muscarínicos/efectos de los fármacos , Apnea Obstructiva del Sueño/tratamiento farmacológico , Nervio Vago/efectos de los fármacos , Animales , Colina O-Acetiltransferasa/genética , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Hemodinámica/efectos de los fármacos , Humanos , Ligandos , Oxitocina/metabolismo , Receptores Muscarínicos/genética , Receptores Muscarínicos/metabolismo , Apnea Obstructiva del Sueño/genética , Apnea Obstructiva del Sueño/metabolismo , Apnea Obstructiva del Sueño/fisiopatología , Nervio Vago/metabolismo , Nervio Vago/fisiopatología
6.
Am J Respir Cell Mol Biol ; 63(4): 502-509, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32603263

RESUMEN

Respiratory depression is the main cause of morbidity and mortality associated with opioids. Obesity increases opioid-related mortality, which is mostly related to comorbid obstructive sleep apnea. Naloxone, a µ-opioid receptor blocker, is an effective antidote, but it reverses analgesia. Like humans with obesity, mice with diet-induced obesity hypoventilate during sleep and develop obstructive sleep apnea, which can be treated with intranasal leptin. We hypothesized that intranasal leptin reverses opioid-induced sleep-disordered breathing in obese mice without decreasing analgesia. To test this hypothesis, mice with diet-induced obesity were treated with morphine at 10 mg/kg subcutaneously and with leptin or placebo intranasally. Sleep and breathing were recorded by barometric plethysmography, and pain sensitivity was measured by the tail-flick test. Excitatory postsynaptic currents were recorded in vitro from hypoglossal motor neurons after the application of the µ-opioid receptor agonist [D-Ala2, N-MePhe4, Gly-ol]-enkephalin and leptin. Morphine dramatically increased the frequency of apneas and greatly increased the severity of hypoventilation and obstructive sleep apnea. Leptin decreased the frequency of apneas, improved obstructive sleep apnea, and completely reversed hypoventilation, whereas morphine analgesia was enhanced. Our in vitro studies demonstrated that [D-Ala2, N-MePhe4, Gly-ol]-enkephalin reduced the frequency of excitatory postsynaptic currents in hypoglossal motoneurons and that application of leptin restored excitatory synaptic neurotransmission. Our findings suggest that intranasal leptin may prevent opioid respiratory depression during sleep in patients with obesity receiving opioids without reducing analgesia.


Asunto(s)
Analgésicos Opioides/efectos adversos , Leptina/administración & dosificación , Respiración/efectos de los fármacos , Síndromes de la Apnea del Sueño/inducido químicamente , Síndromes de la Apnea del Sueño/prevención & control , Sueño/efectos de los fármacos , Administración Intranasal/métodos , Analgesia/métodos , Animales , Modelos Animales de Enfermedad , Encefalinas/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Morfina/farmacología , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Receptores Opioides mu/metabolismo , Síndromes de la Apnea del Sueño/metabolismo , Transmisión Sináptica/efectos de los fármacos
7.
JACC Basic Transl Sci ; 5(5): 484-497, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32478209

RESUMEN

This work shows long-term restoration of the hypothalamic oxytocin (OXT) network preserves OXT release, reduces mortality, cardiac inflammation, fibrosis, and improves autonomic tone and cardiac function in a model of heart failure. Intranasal administration of OXT in patients mimics the short-term changes seen in animals by increasing parasympathetic-and decreasing sympathetic-cardiac activity. This work provides the essential translational foundation to determine if approaches that mimic paraventricular nucleus (PVN) OXT neuron activation, such as safe, noninvasive, and well-tolerated intranasal administration of OXT, can be beneficial in patients with heart failure.

8.
Am J Physiol Heart Circ Physiol ; 319(1): H3-H12, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32412778

RESUMEN

Heart failure (HF) is characterized by autonomic imbalance with sympathetic hyperactivity and loss of parasympathetic tone. Intracardiac ganglia (ICG) neurons represent the final common pathway for vagal innervation of the heart and strongly regulate cardiac functions. This study tests whether ICG cholinergic neuron activation mitigates the progression of cardiac dysfunction and reduces mortality that occurs in HF. HF was induced by transaortic constriction (TAC) in male transgenic Long-Evans rats expressing Cre recombinase within choline acetyltransferase (ChAT) neurons. ChAT neurons were selectively activated by expression and activation of excitatory designer receptors exclusively activated by designer receptors (DREADDs) by clozapine-N-oxide (TAC + treatment and sham-treated groups). Control animals expressed DREADDs but received saline (sham and TAC groups). A separate set of animals were telemetry instrumented to record blood pressure (BP) and heart rate (HR). Acute activation of ICG neurons resulted in robust reductions in BP (∼20 mmHg) and HR (∼100 beats/min). All groups of animals were subjected to weekly echocardiography and treadmill stress tests from 3 to 6 wk post-TAC/sham surgery. Activation of ICG cholinergic neurons reduced the left ventricular systolic dysfunction (reductions in ejection fraction, fractional shortening, stroke volume, and cardiac output) and cardiac autonomic dysfunction [reduced HR recovery (HRR) post peak effort] observed in TAC animals. Additionally, activation of ICG ChAT neurons reduced mortality by 30% compared with untreated TAC animals. These data suggest that ICG cholinergic neuron activation reduces cardiac dysfunction and improves survival in HF, indicating that ICG neuron activation could be a novel target for treating HF.NEW & NOTEWORTHY Intracardiac ganglia form the final common pathway for the parasympathetic innervation of the heart. This study has used a novel chemogenetic approach within transgenic ChAT-Cre rats [expressing only Cre-recombinase in choline acetyl transferase (ChAT) neurons] to selectively increase intracardiac cholinergic parasympathetic activity to the heart in a pressure overload-induced heart failure model. The findings from this study confirm that selective activation of intracardiac cholinergic neurons lessens cardiac dysfunction and mortality seen in heart failure, identifying a novel downstream cardiac-selective target for increasing cardioprotective parasympathetic activity in heart failure.


Asunto(s)
Neuronas Colinérgicas/fisiología , Insuficiencia Cardíaca/fisiopatología , Corazón/inervación , Función Ventricular , Animales , Sistema Nervioso Autónomo/fisiopatología , Presión Sanguínea , Colina O-Acetiltransferasa/genética , Colina O-Acetiltransferasa/metabolismo , Neuronas Colinérgicas/efectos de los fármacos , Neuronas Colinérgicas/metabolismo , Clozapina/análogos & derivados , Clozapina/farmacología , Corazón/fisiopatología , Insuficiencia Cardíaca/etiología , Frecuencia Cardíaca , Masculino , Ratas , Ratas Long-Evans , Obstrucción del Flujo Ventricular Externo/complicaciones
9.
Neuropharmacology ; 165: 107932, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31911104

RESUMEN

The intravenous anaesthetic ketamine, has been demonstrated to inhibit nicotinic acetylcholine receptor (nAChR)-mediated currents in dissociated rat intracardiac ganglion (ICG) neurons (Weber et al., 2005). This effect would be predicted to depress synaptic transmission in the ICG and would account for the inhibitory action of ketamine on vagal transmission to the heart (Inoue and König, 1988). This investigation was designed to examine the activity of ketamine on (i) postsynaptic responses to vagal nerve stimulation, (ii) the membrane potential, and (iii) membrane current responses evoked by exogenous application of ACh and nicotine in ICG neurons in situ. Intracellular recordings were made using sharp intracellular microelectrodes in a whole mount ICG preparation. Preganglionic nerve stimulation and recordings in current- and voltage-clamp modes were used to assess the action of ketamine on ganglionic transmission and nAChR-mediated responses. Ketamine attenuated the postsynaptic responses evoked by nerve stimulation. This reduction was significant at clinically relevant concentrations at high frequencies. The excitatory membrane potential and current responses to focal application of ACh and nicotine were inhibited in a concentration-dependent manner by ketamine. In contrast, ketamine had no effect on either the directly-evoked action potential or excitatory responses evoked by focal application of γ-aminobutyric acid (GABA). Taken together, ketamine inhibits synaptic transmission and nicotine- and ACh-evoked currents in adult rat ICG. Ketamine inhibition of synaptic transmission and nAChR-mediated responses in the ICG contributes significantly to its attenuation of the bradycardia observed in response to vagal stimulation in the mammalian heart.


Asunto(s)
Anestésicos Intravenosos/administración & dosificación , Ganglios Parasimpáticos/efectos de los fármacos , Corazón/fisiología , Ketamina/administración & dosificación , Neuronas/efectos de los fármacos , Receptores Nicotínicos/fisiología , Transmisión Sináptica/efectos de los fármacos , Nervio Vago/efectos de los fármacos , Animales , Femenino , Ganglios Parasimpáticos/fisiología , Corazón/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Neuronas/fisiología , Agonistas Nicotínicos/administración & dosificación , Ratas Wistar , Receptores Nicotínicos/administración & dosificación , Nervio Vago/fisiología , Estimulación del Nervio Vago
10.
Cardiovasc Res ; 113(11): 1318-1328, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28472396

RESUMEN

AIMS: A distinctive hallmark of heart failure (HF) is autonomic imbalance, consisting of increased sympathetic activity, and decreased parasympathetic tone. Recent work suggests that activation of hypothalamic oxytocin (OXT) neurons could improve autonomic balance during HF. We hypothesized that a novel method of chronic selective activation of hypothalamic OXT neurons will improve cardiac function and reduce inflammation and fibrosis in a rat model of HF. METHODS AND RESULTS: Two groups of male Sprague-Dawley rats underwent trans-ascending aortic constriction (TAC) to induce left ventricular (LV) hypertrophy that progresses to HF. In one TAC group, OXT neurons in the paraventricular nucleus of the hypothalamus were chronically activated by selective expression and activation of excitatory DREADDs receptors with daily injections of clozapine N-oxide (CNO) (TAC + OXT). Two additional age-matched groups received either saline injections (Control) or CNO injections for excitatory DREADDs activation (OXT NORM). Heart rate (HR), LV developed pressure (LVDP), and coronary flow rate were measured in isolated heart experiments. Isoproterenol (0.01 nM-1.0 µM) was administered to evaluate ß-adrenergic sensitivity. We found that increases in cellular hypertrophy and myocardial collagen density in TAC were blunted in TAC + OXT animals. Inflammatory cytokine IL-1ß expression was more than twice higher in TAC than all other hearts. LVDP, rate pressure product (RPP), contractility, and relaxation were depressed in TAC compared with all other groups. The response of TAC and TAC + OXT hearts to isoproterenol was blunted, with no significant increase in RPP, contractility, or relaxation. However, HR in TAC + OXT animals increased to match Control at higher doses of isoproterenol. CONCLUSIONS: Activation of hypothalamic OXT neurons to elevate parasympathetic tone reduced cellular hypertrophy, levels of IL-1ß, and fibrosis during TAC-induced HF in rats. Cardiac contractility parameters were significantly higher in TAC + OXT compared with TAC animals. HR sensitivity, but not contractile sensitivity, to ß-adrenergic stimulation was improved in TAC + OXT hearts.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , Miocardio/metabolismo , Neuronas/metabolismo , Oxitocina/metabolismo , Animales , Corazón/fisiopatología , Insuficiencia Cardíaca/etiología , Hipertrofia Ventricular Izquierda/complicaciones , Hipertrofia Ventricular Izquierda/fisiopatología , Isoproterenol/farmacología , Masculino , Contracción Miocárdica/efectos de los fármacos , Neuronas/efectos de los fármacos , Ratas Sprague-Dawley
11.
Am J Physiol Heart Circ Physiol ; 310(11): H1549-57, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27016581

RESUMEN

Hypertension is a common outcome associated with obstructive sleep apnea (OSA), a prevalent yet poorly treated cardiovascular disease. Recent studies showed oxytocin (OXT), released from hypothalamic paraventricular nucleus (PVN) neurons, activates cardiac vagal neurons in the dorsal motor nucleus of the vagus (DMNX) and may blunt cardiovascular responses to stress. This study tests whether the release of OXT from PVN fibers in the DMNX is diminished with chronic intermittent hypoxia-hypercapnia (CIH/H) exposure, an animal model of OSA, and whether activation of PVN OXT neurons restores OXT release in the DMNX and prevents the hypertension resulting from CIH/H. To assess OXT release from PVN fibers, Chinese hamster ovarian (CHO) cells were engineered to be highly sensitive to OXT by stable expression of the human recombinant OXT receptor and the calcium indicator R-GECO1. PVN fibers in the DMNX were selectively photoactivated in vitro by expression of channelrhodopsin. The release of OXT onto CHO cells in the DMNX was blunted in rats exposed to 21 days of CIH/H. Chronic activation of PVN OXT neurons in vivo, using designer receptors exclusively activated by designer drugs, restored the release of OXT onto CHO cells in the DMNX. Chronic PVN OXT neuron activation in vivo also prevented the hypertension that occurred in conscious unrestrained telemetry-equipped sham rats exposed to 3 wk of CIH/H. These results demonstrate that chronic activation of OXT neurons restores the release of OXT from PVN fibers in the DMNX and prevents the hypertension that occurs with 3 wk of CIH/H exposure.


Asunto(s)
Presión Sanguínea , Hipercapnia/complicaciones , Hipertensión/prevención & control , Hipoxia/complicaciones , Neuronas/metabolismo , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Receptores de Oxitocina/metabolismo , Animales , Técnicas Biosensibles , Células CHO , Channelrhodopsins , Enfermedad Crónica , Cricetulus , Modelos Animales de Enfermedad , Hipertensión/etiología , Hipertensión/metabolismo , Hipertensión/fisiopatología , Masculino , Optogenética , Oxitocina/genética , Núcleo Hipotalámico Paraventricular/fisiopatología , Ratas Sprague-Dawley , Receptores de Oxitocina/genética , Transducción de Señal , Telemetría , Factores de Tiempo , Transfección
12.
Curr Hypertens Rep ; 18(3): 22, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26849575

RESUMEN

This brief review focuses on four new topics, with novel and clinically significant consequences, concerning the powerful influence of parasympathetic activity on cardiac function. In this short summary, we will highlight very recent and important work, published in the last 3-4 years, that (1) challenges the paradigm that parasympathetic activity to the heart is involved in the control of heart rate but plays little role in other cardiac functions, (2) characterizes important long-range synaptic pathways to parasympathetic cardiac vagal neurons that are involved in "higher" brain functions (such as arousal and emotional challenges), (3) asks whether implantable chronic vagal nerve stimulation is a promising clinical tool for treating cardiovascular diseases, and (4) describes newly identified neuropeptides and other modulators that can influence the generation and maintenance of parasympathetic activity to the heart.


Asunto(s)
Corazón/fisiología , Animales , Frecuencia Cardíaca , Humanos , Hipertensión/fisiopatología , Neuropéptidos/metabolismo , Nervio Vago
13.
Am J Physiol Heart Circ Physiol ; 309(8): H1281-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26371169

RESUMEN

Hypertension, cardiac hypertrophy, and heart failure (HF) are widespread and debilitating cardiovascular diseases that affect nearly 23 million people worldwide. A distinctive hallmark of these cardiovascular diseases is autonomic imbalance, with increased sympathetic activity and decreased parasympathetic vagal tone. Recent device-based approaches, such as implantable vagal stimulators that stimulate a multitude of visceral sensory and motor fibers in the vagus nerve, are being evaluated as new therapeutic approaches for these and other diseases. However, little is known about how parasympathetic activity to the heart is altered with these diseases, and this lack of knowledge is an obstacle in the goal of devising selective interventions that can target and selectively restore parasympathetic activity to the heart. To identify the changes that occur within the brain stem to diminish the parasympathetic cardiac activity, left ventricular hypertrophy was elicited in rats by aortic pressure overload using a transaortic constriction approach. Cardiac vagal neurons (CVNs) in the brain stem that generate parasympathetic activity to the heart were identified with a retrograde tracer and studied using patch-clamp electrophysiological recordings in vitro. Animals with left cardiac hypertrophy had diminished excitation of CVNs, which was mediated both by an augmented frequency of spontaneous inhibitory GABAergic neurotransmission (with no alteration of inhibitory glycinergic activity) as well as a diminished amplitude and frequency of excitatory neurotransmission to CVNs. Opportunities to alter these network pathways and neurotransmitter receptors provide future targets of intervention in the goal to restore parasympathetic activity and autonomic balance to the heart in cardiac hypertrophy and other cardiovascular diseases.


Asunto(s)
Tronco Encefálico/fisiopatología , Insuficiencia Cardíaca/etiología , Corazón/inervación , Hipertrofia Ventricular Izquierda/complicaciones , Transmisión Sináptica , Nervio Vago/fisiopatología , Animales , Tronco Encefálico/metabolismo , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores , Neuronas GABAérgicas/metabolismo , Ácido Glutámico/metabolismo , Glicina/metabolismo , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/fisiopatología , Hipertrofia Ventricular Izquierda/diagnóstico , Hipertrofia Ventricular Izquierda/fisiopatología , Técnicas In Vitro , Masculino , Inhibición Neural , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología , Técnicas de Trazados de Vías Neuroanatómicas , Técnicas de Placa-Clamp , Ratas Sprague-Dawley , Nervio Vago/metabolismo , Ácido gamma-Aminobutírico/metabolismo
14.
Prog Brain Res ; 212: 39-58, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25194192

RESUMEN

Parasympathetic cardiac vagal neurons (CVNs) in the brainstem dominate the control of heart rate. Previous work has determined that these neurons are inherently silent, and their activity is largely determined by synaptic inputs to CVNs that include four major types of synapses that release glutamate, GABA, glycine, or serotonin. Whereas prior reviews have focused on glutamatergic, GABAergic and glycinergic pathways, and the receptors in CVNs activated by these neurotransmitters, this review focuses on the alterations in CVN activity with hypoxia-, sleep-, and sleep-related cardiovascular diseases including obstructive sleep apnea.


Asunto(s)
Tronco Encefálico/fisiología , Frecuencia Cardíaca/fisiología , Corazón/fisiología , Sistema Nervioso Parasimpático/fisiología , Apnea Obstructiva del Sueño/fisiopatología , Sueño/fisiología , Animales , Corazón/inervación , Humanos , Hipoxia/fisiopatología , Neuronas/fisiología , Nervio Vago/fisiología
15.
Hypertension ; 64(3): 597-603, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24958501

RESUMEN

Obstructive sleep apnea is associated with chronic intermittent hypoxia/hypercapnia (CIHH) episodes during sleep that heighten sympathetic and diminish parasympathetic activity to the heart. Although one population of neurons in the paraventricular nucleus of the hypothalamus strongly influences sympathetic tone and has increased activity after CIHH, little is known about the role of this pathway to parasympathetic neurons and how this network is altered in CIHH. We hypothesized that CIHH inhibits the excitatory pathway from the paraventricular nucleus of the hypothalamus to parasympathetic cardiac vagal neurons in the brain stem. To test this hypothesis, channelrhodopsin was selectively expressed, using viral vectors, in neurons in the paraventricular nucleus of the hypothalamus and channelrhodopsin-expressing fibers were photoactivated to evoke postsynaptic currents in cardiac vagal neurons in brain stem slices. Excitatory postsynaptic currents were diminished in animals exposed to CIHH. The paired-pulse and prolonged facilitation of the postsynaptic current amplitudes and frequencies evoked by paired and bursts of photoactivation of channelrhodopsin fibers, respectively, occurred in unexposed rats but were blunted in CIHH animals. In response to an acute challenge of hypoxia/hypercapnia, the amplitude of postsynaptic events was unchanged during, but increased after hypoxia/hypercapnia in unexposed animals. In contrast, postsynaptic currents were inhibited during hypoxia/hypercapnia in rats exposed to CIHH. In conclusion, the excitatory pathway to cardiac vagal neurons is diminished in response to both acute and chronic exposures to hypoxia/hypercapnia. This could elicit a reduced cardioprotective parasympathetic activity and an enhanced risk of adverse cardiovascular events in episodes of apnea and chronic obstructive sleep apnea.


Asunto(s)
Tronco Encefálico/fisiopatología , Corazón/inervación , Hipercapnia/fisiopatología , Hipoxia/fisiopatología , Sistema Nervioso Parasimpático/fisiopatología , Núcleo Hipotalámico Paraventricular/fisiopatología , Animales , Enfermedades Cardiovasculares/epidemiología , Modelos Animales de Enfermedad , Femenino , Masculino , Proteínas del Tejido Nervioso/fisiología , Ratas , Ratas Sprague-Dawley , Factores de Riesgo , Apnea Obstructiva del Sueño/fisiopatología , Transmisión Sináptica/fisiología
16.
J Physiol ; 592(13): 2799-811, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24835174

RESUMEN

Patients with obstructive sleep apnoea experience chronic intermittent hypoxia-hypercapnia (CIHH) during sleep that elicit sympathetic overactivity and diminished parasympathetic activity to the heart, leading to hypertension and depressed baroreflex sensitivity. The parasympathetic control of heart rate arises from pre-motor cardiac vagal neurons (CVNs) located in nucleus ambiguus (NA) and dorsal motor nucleus of the vagus (DMNX). The mechanisms underlying diminished vagal control of heart rate were investigated by studying the changes in blood pressure, heart rate, and neurotransmission to CVNs evoked by acute hypoxia-hypercapnia (H-H) and CIHH. In vivo telemetry recordings of blood pressure and heart rate were obtained in adult rats during 4 weeks of CIHH exposure. Retrogradely labelled CVNs were identified in an in vitro brainstem slice preparation obtained from adult rats exposed either to air or CIHH for 4 weeks. Postsynaptic inhibitory or excitatory currents were recorded using whole cell voltage clamp techniques. Rats exposed to CIHH had increases in blood pressure, leading to hypertension, and blunted heart rate responses to acute H-H. CIHH induced an increase in GABAergic and glycinergic neurotransmission to CVNs in NA and DMNX, respectively; and a reduction in glutamatergic neurotransmission to CVNs in both nuclei. CIHH blunted the bradycardia evoked by acute H-H and abolished the acute H-H evoked inhibition of GABAergic transmission while enhancing glycinergic neurotransmission to CVNs in NA. These changes with CIHH inhibit CVNs and vagal outflow to the heart, both in acute and chronic exposures to H-H, resulting in diminished levels of cardioprotective parasympathetic activity to the heart as seen in OSA patients.


Asunto(s)
Neuronas GABAérgicas/fisiología , Frecuencia Cardíaca , Hipercapnia/fisiopatología , Hipoxia/fisiopatología , Apnea Obstructiva del Sueño/fisiopatología , Transmisión Sináptica , Nervio Vago/fisiopatología , Animales , Presión Sanguínea , Corazón/inervación , Ratas , Ratas Sprague-Dawley , Nervio Vago/citología
17.
Am J Physiol Regul Integr Comp Physiol ; 299(1): R42-54, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20445155

RESUMEN

We have investigated the effects of the reactive oxygen species (ROS) donors hydrogen peroxide (H(2)O(2)) and tert-butyl hydroperoxide (t-BHP) on the intrinsic electrophysiological characteristics: ganglionic transmission and resting [Ca(2+)](i) in neonate and adult rat intracardiac ganglion (ICG) neurons. Intracellular recordings were made using sharp microelectrodes filled with either 0.5 M KCl or Oregon Green 488 BAPTA-1, allowing recording of electrical properties and measurement of [Ca(2+)](i). H(2)O(2) and t-BHP both hyperpolarized the resting membrane potential and reduced membrane resistance. In adult ICG neurons, the hyperpolarizing action of H(2)O(2) was reversed fully by Ba(2+) and partially by tetraethylammonium, muscarine, and linopirdine. H(2)O(2) and t-BHP reduced the action potential afterhyperpolarization (AHP) amplitude but had no impact on either overshoot or AHP duration. ROS donors evoked an increase in discharge adaptation to long depolarizing current pulses. H(2)O(2) blocked ganglionic transmission in most ICG neurons but did not alter nicotine-evoked depolarizations. By contrast, t-BHP had no significant action on ganglionic transmission. H(2)O(2) and t-BHP increased resting intracellular Ca(2+) levels to 1.6 ( +/- 0.6, n = 11, P < 0.01) and 1.6 ( +/- 0.3, n = 8, P < 0.001), respectively, of control value (1.0, approximately 60 nM). The ROS scavenger catalase prevented the actions of H(2)O(2), and this protection extended beyond the period of application. Superoxide dismutase partially shielded against the action of H(2)O(2), but this was limited to the period of application. These data demonstrate that ROS decreases the excitability and ganglionic transmission of ICG neurons, attenuating parasympathetic control of the heart.


Asunto(s)
Calcio/fisiología , Fenómenos Electrofisiológicos , Neuronas/fisiología , Especies Reactivas de Oxígeno/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Compuestos de Anilina , Animales , Animales Recién Nacidos , Femenino , Fluoresceínas , Ganglios , Corazón , Peróxido de Hidrógeno/farmacología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Microelectrodos , Neuronas/efectos de los fármacos , Ratas , Ratas Wistar , terc-Butilhidroperóxido/farmacología
18.
Exp Physiol ; 94(2): 201-12, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18978036

RESUMEN

We have investigated the action of two elements of acute ischaemia, high potassium and aglycaemia, on the electrophysiological properties and ganglionic transmission of adult rat intracardiac ganglion (ICG) neurones. We used a whole-mount ganglion preparation of the right atrial ganglion plexus and sharp microelectrode recording techniques. Increasing extracellular K(+) from its normal value of 4.7 mm to 10 mm decreased membrane potential and action potential after-hyperpolarization amplitude but otherwise had no effect on postganglionic membrane properties. It did, however, reduce the ability of synaptically evoked action potentials to follow high-frequency (100 Hz) repetitive stimulation. A further increase in K(+) changed both the passive and the active membrane properties of the postganglionic neurone: time constant, membrane resistance and action potential overshoot were all decreased in high K(+) (20 mm). The ICG neurones display a predominantly phasic discharge in response to prolonged depolarizing current pulses. High K(+) had no impact on this behaviour but reduced the time-dependent rectification response to hyperpolarizing currents. At 20 mm, K(+) practically blocked ganglionic transmission in most neurones at all frequencies tested. Aglycaemia, nominally glucose-free physiological saline solution (PSS), increased the time constant and membrane resistance of ICG neurones but otherwise had no action on their passive or active properties or ganglionic transmission. However, the combination of aglycaemia and 20 mm K(+) displayed an improvement in passive properties and ganglionic transmission when compared with 20 mm K(+) PSS. These data indicate that the presynaptic terminal is the primary target of high extracellular potassium and that aglycaemia may have protective actions against this challenge.


Asunto(s)
Glucemia/fisiología , Fenómenos Electrofisiológicos/fisiología , Ganglios/fisiología , Corazón/inervación , Potasio/farmacología , Transmisión Sináptica/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Isquemia Miocárdica/fisiopatología , Técnicas de Placa-Clamp , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/fisiología , Ratas , Ratas Wistar , Transmisión Sináptica/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...