Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 351
Filter
Add more filters

Publication year range
1.
Physiol Meas ; 44(1)2023 01 12.
Article in English | MEDLINE | ID: mdl-36343372

ABSTRACT

Objective.To conduct a systematic review of the possible effects of passive heating protocols on cardiovascular autonomic control in healthy individuals.Approach.The studies were obtained from MEDLINE (PubMed), LILACS (BVS), EUROPE PMC (PMC), and SCOPUS databases, simultaneously. Studies were considered eligible if they employed passive heating protocols and investigated cardiovascular autonomic control by spontaneous methods, such as heart rate variability (HRV), systolic blood pressure variability (SBPV), and baroreflex sensitivity (BRS), in healthy adults. The revised Cochrane risk-of-bias tool (RoB-2) was used to assess the risk of bias in each study.Main results.Twenty-seven studies were included in the qualitative synthesis. Whole-body heating protocols caused a reduction in cardiac vagal modulation in 14 studies, and two studies reported both increased sympathetic modulation and vagal withdrawal. Contrariwise, local-heating protocols and sauna bathing seem to increase cardiac vagal modulation. A reduction of BRS was reported in most of the studies that used whole-body heating protocols. However, heating effects on BRS remain controversial due to methodological differences among baroreflex analysis and heating protocols.Significance.Whole-body heat stress may increase sympathetic and reduce vagal modulation to the heart in healthy adults. On the other hand, local-heating therapy and sauna bathing seem to increase cardiac vagal modulation, opposing sympathetic modulation. Nonetheless, further studies should investigate acute and chronic effects of thermal therapy on cardiovascular autonomic control.


Subject(s)
Autonomic Nervous System , Cardiovascular System , Hyperthermia, Induced , Adult , Humans , Autonomic Nervous System/physiology , Autonomic Nervous System/physiopathology , Baroreflex/physiology , Blood Pressure/physiology , Cardiovascular System/innervation , Cardiovascular System/physiopathology , Heart/innervation , Heart/physiology , Heart Rate/physiology , Hot Temperature/adverse effects , Hyperthermia, Induced/adverse effects , Hyperthermia, Induced/methods
2.
Clin Exp Hypertens ; 44(5): 427-435, 2022 Jul 04.
Article in English | MEDLINE | ID: mdl-35438014

ABSTRACT

OBJECTIVES: This crossover study design aimed to assess hemodynamic, cardiac autonomic, and vascular responses to high-intensity interval (HIIE) vs moderate-intensity continuous exercise (MICE) in older individuals with hypertension. METHODS: Twenty (67 ± 7 y) older individuals with hypertension were randomly assigned to perform HIIE, MICE, or control (CON) sessions in the heated swimming pool (30-32°C). Blood pressure (BP), arterial stiffness, endothelial reactivity, and heart rate variability (HRV) were measured pre, post, and 45 min (recovery) after each intervention followed by 24-h ambulatory BP and HRV. RESULTS: One single aerobic exercise session was not effective to provoke post-exercise hypotension and vascular improvements. HIIE was superior to MICE and CON to increasing parasympathetic modulation at post and recovery. Exercise sessions showed to disturb the autonomic system at nighttime compared to CON. CONCLUSIONS: These results may have important implications in water-based therapy and the elderly with hypertension.


Subject(s)
Aquatic Therapy , High-Intensity Interval Training , Hypertension , Aged , Aquatic Therapy/methods , Autonomic Nervous System/physiopathology , Blood Pressure/physiology , Blood Vessels/physiopathology , Cross-Over Studies , Exercise/physiology , Heart/innervation , Heart/physiopathology , Heart Rate/physiology , Heating , Hemodynamics/physiology , High-Intensity Interval Training/methods , Humans , Hypertension/physiopathology , Hypertension/therapy , Middle Aged
3.
Acupunct Med ; 39(6): 681-690, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34056953

ABSTRACT

BACKGROUND: Sympathetic and parasympathetic nerve remodeling play an important role in cardiac function after myocardial ischemia (MI) injury. Increasing evidence indicates that electroacupuncture (EA) can regulate cardiac function by modulating the autonomic nervous system (ANS), but little is known about its effectiveness on neural remodeling post-MI. OBJECTIVES: To investigate the role of EA in ANS remodeling post-MI. METHODS: Adult male C57/BL6 mice were equally divided into the Control (Ctrl), MI and EA groups after generating the MI model by ligating the left anterior descending (LAD) coronary artery. Echocardiography and 2,3,5-triphenyltetrazolium (TTC) staining were employed to evaluate cardiac function and infarct size after EA treatment for five consecutive days. Serum norepinephrine (NE) levels were measured by ELISA to quantify sympathetic activation. Then, ANS remodeling was detected by immunohistochemistry (IHC), RT-qPCR, and Western blotting. RESULTS: Our preliminary findings showed that EA increased ejection fraction and fractional shortening and reduced infarct area after MI injury. Serum NE levels in the EA group were significantly decreased compared with those in the MI group. IHC staining results demonstrated that the density of growth associated protein (GAP)43 and tyrosine hydroxylase (TH) positive nerve fibers in the EA group were decreased with increased choline acetyltransferase (CHAT) and vesicular acetylcholine transporter (VACHT). Meanwhile, the results verified that mRNA and protein expression of GAP43 and TH were significantly inhibited by EA treatment in the MI mice, accompanied by elevated CHAT and VACHT. CONCLUSIONS: EA treatment could improve cardiac function and reduce infarct size by modulating sympathetic and parasympathetic nerve remodeling post-MI, thus helping the cardiac ANS reach a new balance to try to protect the heart from further possible injury.


Subject(s)
Autonomic Nervous System/physiopathology , Electroacupuncture , Myocardial Ischemia/therapy , Animals , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Heart/innervation , Heart/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Myocardial Ischemia/blood , Myocardial Ischemia/physiopathology , Norepinephrine/blood
4.
Cardiovasc Res ; 117(7): 1732-1745, 2021 06 16.
Article in English | MEDLINE | ID: mdl-33989382

ABSTRACT

The cardiac autonomic nervous system (ANS) plays an integral role in normal cardiac physiology as well as in disease states that cause cardiac arrhythmias. The cardiac ANS, comprised of a complex neural hierarchy in a nested series of interacting feedback loops, regulates atrial electrophysiology and is itself susceptible to remodelling by atrial rhythm. In light of the challenges of treating atrial fibrillation (AF) with conventional pharmacologic and myoablative techniques, increasingly interest has begun to focus on targeting the cardiac neuraxis for AF. Strong evidence from animal models and clinical patients demonstrates that parasympathetic and sympathetic activity within this neuraxis may trigger AF, and the ANS may either induce atrial remodelling or undergo remodelling itself to serve as a substrate for AF. Multiple nexus points within the cardiac neuraxis are therapeutic targets, and neuroablative and neuromodulatory therapies for AF include ganglionated plexus ablation, epicardial botulinum toxin injection, vagal nerve (tragus) stimulation, renal denervation, stellate ganglion block/resection, baroreceptor activation therapy, and spinal cord stimulation. Pre-clinical and clinical studies on these modalities have had promising results and are reviewed here.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/therapy , Autonomic Denervation , Autonomic Nervous System/physiopathology , Electric Stimulation Therapy , Heart/innervation , Neurotransmitter Agents/therapeutic use , Action Potentials , Animals , Anti-Arrhythmia Agents/adverse effects , Atrial Fibrillation/diagnosis , Atrial Fibrillation/physiopathology , Atrial Remodeling , Autonomic Denervation/adverse effects , Electric Stimulation Therapy/adverse effects , Heart Rate , Humans , Neurotransmitter Agents/adverse effects , Spinal Cord Stimulation , Treatment Outcome , Vagus Nerve Stimulation
5.
Circ Arrhythm Electrophysiol ; 14(4): e009668, 2021 04.
Article in English | MEDLINE | ID: mdl-33858178

ABSTRACT

Symptomatic heart failure (HF) patients despite optimal medical therapy and advances such as invasive hemodynamic monitoring remain challenging to manage. While cardiac resynchronization therapy remains a highly effective therapy for a subset of HF patients with wide QRS, a majority of symptomatic HF patients are poor candidates for such. Recently, cardiac contractility modulation, neuromodulation based on carotid baroreceptor stimulation, and phrenic nerve stimulation have been approved by the US Food and Drug Administration and are emerging as therapeutic options for symptomatic HF patients. This state-of-the-art review examines the role of these evolving electrical therapies in advanced HF.


Subject(s)
Autonomic Nervous System/physiopathology , Electric Stimulation Therapy , Heart Failure/therapy , Heart/innervation , Myocardial Contraction , Stroke Volume , Ventricular Function, Left , Animals , Cardiac Pacing, Artificial , Electric Stimulation Therapy/adverse effects , Electric Stimulation Therapy/instrumentation , Heart Failure/diagnosis , Heart Failure/epidemiology , Heart Failure/physiopathology , Humans , Pacemaker, Artificial , Prevalence , Recovery of Function , Spinal Cord Stimulation , Treatment Outcome , Vagus Nerve Stimulation
6.
J Am Heart Assoc ; 10(2): e018371, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33441022

ABSTRACT

Background Cardiac sympathetic denervation (CSD) has been used as a bailout strategy for refractory ventricular tachycardia (VT). Risk of VT recurrence in patients with scar-related monomorphic VT referred for CSD and the extent to which CSD can modify this risk is unknown. We aimed to quantify arrhythmia recurrence risk and impact of CSD in this population. Methods and Results Adjusted competing risk time to event models were developed to adjust for risk of VT recurrence and sustained VT/implantable cardioverter-defibrillator shocks after VT ablation based on patient comorbidities at the time of VT ablation. Adjusted VT and implantable cardioverter-defibrillator shock recurrence rates were estimated for the subgroup who subsequently required CSD after ablation. The expected adjusted recurrence rates were then compared with the observed rates after CSD. Data from 381 patients with scar-mediated monomorphic VT who underwent VT ablation were analyzed, excluding patients with polymorphic VT. Sixty eight patients underwent CSD for recurrent VT. CSD reduced the expected adjusted VT recurrence rate by 36% (expected rate of 5.61 versus observed rate of 3.58 per 100 person-months, P=0.01) and the sustained VT/implantable cardioverter-defibrillator shock rates by 34% (expected rate of 4.34 versus observed 2.85 per 100 person-months, P=0.03). The median number of sustained VT/implantable cardioverter-defibrillator shocks in the year before versus the year after CSD was reduced by 90% (10 versus 1, P<0.0001). Conclusions Patients referred for CSD for refractory scar-mediated monomorphic VT are at a higher risk of VT recurrence after ablation as compared with those not requiring CSD, mostly because of their cardiac comorbidities. CSD significantly reduced both the expected risk of recurrences and VT burden.


Subject(s)
Catheter Ablation , Cicatrix , Defibrillators, Implantable , Sympathectomy , Tachycardia, Ventricular , Anti-Arrhythmia Agents/therapeutic use , Catheter Ablation/adverse effects , Catheter Ablation/methods , Cicatrix/etiology , Cicatrix/physiopathology , Comorbidity , Electrophysiologic Techniques, Cardiac/methods , Female , Heart/innervation , Humans , Male , Middle Aged , Retrospective Studies , Risk Adjustment/methods , Secondary Prevention/methods , Sympathectomy/adverse effects , Sympathectomy/methods , Tachycardia, Ventricular/physiopathology , Tachycardia, Ventricular/prevention & control , Tachycardia, Ventricular/surgery , United States/epidemiology
7.
J Neurophysiol ; 124(4): 1241-1256, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32755328

ABSTRACT

The American lobster, Homarus americanus, cardiac neuromuscular system is controlled by the cardiac ganglion (CG), a central pattern generator consisting of four premotor and five motor neurons. Here, we show that the premotor and motor neurons can establish independent bursting patterns when decoupled by a physical ligature. We also show that mRNA encoding myosuppressin, a cardioactive neuropeptide, is produced within the CG. We thus asked whether myosuppressin modulates the decoupled premotor and motor neurons, and if so, how this modulation might underlie the role(s) that these neurons play in myosuppressin's effects on ganglionic output. Although myosuppressin exerted dose-dependent effects on burst frequency and duration in both premotor and motor neurons in the intact CG, its effects on the ligatured ganglion were more complex, with different effects and thresholds on the two types of neurons. These data suggest that the motor neurons are more important in determining the changes in frequency of the CG elicited by low concentrations of myosuppressin, whereas the premotor neurons have a greater impact on changes elicited in burst duration. A single putative myosuppressin receptor (MSR-I) was previously described from the Homarus nervous system. We identified four additional putative MSRs (MSR-II-V) and investigated their individual distributions in the CG premotor and motor neurons using RT-PCR. Transcripts for only three receptors (MSR-II-IV) were amplified from the CG. Potential differential distributions of the receptors were observed between the premotor and motor neurons; these differences may contribute to the distinct physiological responses of the two neuron types to myosuppressin.NEW & NOTEWORTHY Premotor and motor neurons of the Homarus americanus cardiac ganglion (CG) are normally electrically and chemically coupled, and generate rhythmic bursting that drives cardiac contractions; we show that they can establish independent bursting patterns when physically decoupled by a ligature. The neuropeptide myosuppressin modulates different aspects of the bursting pattern in these neuron types to determine the overall modulation of the intact CG. Differential distribution of myosuppressin receptors may underlie the observed responses to myosuppressin.


Subject(s)
Ganglia, Invertebrate/metabolism , Motor Neurons/metabolism , Neuropeptides/metabolism , Synaptic Potentials , Animals , Ganglia, Invertebrate/cytology , Ganglia, Invertebrate/physiology , Heart/innervation , Motor Neurons/physiology , Nephropidae , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism
8.
JACC Cardiovasc Interv ; 13(1): 1-19, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31918927

ABSTRACT

Despite optimal combination of guideline-directed anti-ischemic therapies and myocardial revascularization, a substantial proportion of patients with stable coronary artery disease continues to experience disabling symptoms and is often referred as "no-option." The appraisal of the pathways linking ischemia to symptom perception indicates a complex model of heart-brain interactions in the generation of the subjective anginal experience and inspired novel approaches that may be clinically effective in alleviating the angina burden of this population. Conversely, the prevailing ischemia-centered view of angina, with the focus on traditional myocardial revascularization as the sole option to address ischemia on top of medical therapy, hinders the experimental characterization and broad-scale clinical implementation of strongly needed therapeutic options. The interventionist, often the first physician to establish the diagnosis of refractory angina pectoris (RAP) following coronary angiography, should be aware of the numerous emerging technologies with the potential to improve quality of life in the growing population of RAP patients. This review describes the current landscape and the future perspectives on nonpharmacological treatment technologies for patients with RAP, with a view on the underlying physiopathological rationale and current clinical evidence.


Subject(s)
Angina Pectoris/therapy , Coronary Artery Disease/therapy , Counterpulsation , Electric Stimulation Therapy , Extracorporeal Shockwave Therapy , Genetic Therapy , Heart/innervation , Laser Therapy , Stem Cell Transplantation , Angina Pectoris/genetics , Angina Pectoris/metabolism , Angina Pectoris/physiopathology , Animals , Brain/metabolism , Brain/physiopathology , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/physiopathology , Coronary Circulation , Counterpulsation/adverse effects , Electric Stimulation Therapy/adverse effects , Energy Metabolism , Extracorporeal Shockwave Therapy/adverse effects , Genetic Therapy/adverse effects , Humans , Laser Therapy/adverse effects , Myocardium/metabolism , Stem Cell Transplantation/adverse effects , Treatment Outcome
9.
Nutrients ; 11(12)2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31757095

ABSTRACT

Resting heart rate (rHR) and heart rate variability (HRV) are non-invasive measurements that predict the risk of sudden cardiac death (SCD). Marine n-3 polyunsaturated fatty acid (PUFA) supplementation may decrease rHR, increase HRV, and reduce the risk of SCD. To date, no studies have investigated the effect of marine n-3 PUFA on HRV in renal transplant recipients. In a randomized controlled trial, 132 renal transplant recipients were randomized to receive either three 1 g capsules of marine n-3 PUFA, each containing 460 mg/g EPA and 380 mg/g DHA, or control (olive oil) for 44 weeks. HRV was calculated in the time and frequency domains during a conventional cardiovascular reflex test (response to standing, deep breathing, and Valsalva maneuver) and during 2 min of resting in the supine position. There was no significant effect of marine n-3 PUFA supplementation on time-domain HRV compared with controls. rHR decreased 3.1 bpm (± 13.1) for patients receiving marine n-3 PUFA compared to 0.8 (± 11.0) in controls (p = 0.28). In the frequency domain HRV analyses, there was a significant change in response to standing in both high and low frequency measures, 2.9 (p = 0.04, 95% CI (1.1;8)) and 2.7 (p = 0.04, 95% CI (1.1;6.5)), respectively. In conclusion, 44 weeks of supplemental marine n-3 PUFAs in renal transplant recipients significantly improved the cardiac autonomic function, assessed by measuring HRV during conventional cardiovascular reflex tests.


Subject(s)
Autonomic Nervous System/drug effects , Death, Sudden, Cardiac/prevention & control , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Heart Rate/drug effects , Heart/innervation , Kidney Transplantation , Transplant Recipients , Adult , Aged , Autonomic Nervous System/physiopathology , Death, Sudden, Cardiac/etiology , Dietary Supplements/adverse effects , Docosahexaenoic Acids/adverse effects , Double-Blind Method , Drug Combinations , Eicosapentaenoic Acid/adverse effects , Female , Humans , Kidney Transplantation/adverse effects , Kidney Transplantation/mortality , Male , Middle Aged , Norway , Time Factors , Treatment Outcome
10.
Rev Cardiovasc Med ; 20(3): 129-137, 2019 Sep 30.
Article in English | MEDLINE | ID: mdl-31601087

ABSTRACT

Atrial fibrillation is the most common symptomatic arrhythmia that is associated with stroke. Contemporary management of the disease is focused on anticoagulation to prevent stroke, coupled with catheter ablation to limit symptoms and prevent deleterious cardiac remodeling. Emerging data highlights the importance of lifestyle modification by managing sleep apnea, increasing physical activity, and weight loss. There is significant data that supports a link between the autonomic nervous system, arrhythmia development, and atrial fibrillation therapy. It is likely that lifestyle modification through these techniques that are aimed to reduce stress may also mediate atrial fibrillation development through this mechanism. This review examines how mind and body practices such as meditation, yoga, and acupuncture may influence the autonomic nervous system and mitigate atrial fibrillation progression and regression. Available evidence from molecular and anatomical levels through to clinical observations and translational clinical trials were scrutinized and a case established for these interventions as potential powerful mediators of anti-arrhythmic benefit.


Subject(s)
Atrial Fibrillation/therapy , Autonomic Nervous System/physiopathology , Heart Rate , Heart/innervation , Mind-Body Therapies , Animals , Atrial Fibrillation/diagnosis , Atrial Fibrillation/physiopathology , Humans , Treatment Outcome
11.
J Cardiovasc Electrophysiol ; 30(12): 2920-2928, 2019 12.
Article in English | MEDLINE | ID: mdl-31625219

ABSTRACT

BACKGROUND: Inappropriate sinus tachycardia (IST) remains a clinical challenge because patients often are highly symptomatic and not responsive to medical therapy. OBJECTIVE: To study the safety and efficacy of stellate ganglion (SG) block and cardiac sympathetic denervation (CSD) in patients with IST. METHODS: Twelve consecutive patients who had drug-refractory IST (10 women) were studied. According to a prospectively initiated protocol, five patients underwent an electrophysiologic study before and after SG block (electrophysiology study group). The subsequent seven patients had ambulatory Holter monitoring before and after SG block (ambulatory group). All patients underwent SG block on the right side first, and then on the left side. Selected patients who had heart rate reduction ≥15 beats per minute (bpm) were recommended to consider CSD. RESULTS: The mean (SD) baseline heart rate (HR) was 106 (21) bpm. The HR significantly decreased to 93 (20) bpm (P = .02) at 10 minutes after right SG block and remained significantly slower at 97(19) bpm at 60 minutes. Left SG block reduced HR from 99 (21) to 87(16) bpm (P = .02) at 60 minutes. SG block had no significant effect on blood pressure or HR response to isoproterenol or exercise (all P > .05). Five patients underwent right (n = 4) or bilateral (n = 1) CSD. The clinical outcomes were heterogeneous: one patient had complete and two had partial symptomatic relief, and two did not have improvement. CONCLUSION: SG blockade modestly reduces resting HR but has no significant effect on HR during exercise. Permanent CSD may have a modest role in alleviating symptoms in selected patients with IST.


Subject(s)
Anesthetics, Combined/administration & dosage , Anesthetics, Local/administration & dosage , Autonomic Nerve Block , Bupivacaine/administration & dosage , Heart Rate/drug effects , Heart/innervation , Lidocaine/administration & dosage , Stellate Ganglion/drug effects , Sympathectomy , Tachycardia, Sinus/therapy , Adult , Anesthetics, Combined/adverse effects , Anesthetics, Local/adverse effects , Autonomic Nerve Block/adverse effects , Bupivacaine/adverse effects , Electrocardiography, Ambulatory , Electrophysiologic Techniques, Cardiac , Female , Humans , Lidocaine/adverse effects , Male , Middle Aged , Pilot Projects , Prospective Studies , Stellate Ganglion/physiopathology , Sympathectomy/adverse effects , Tachycardia, Sinus/diagnosis , Tachycardia, Sinus/physiopathology , Time Factors , Treatment Outcome , Young Adult
13.
JACC Clin Electrophysiol ; 5(8): 881-896, 2019 08.
Article in English | MEDLINE | ID: mdl-31439288

ABSTRACT

Autonomic dysregulation in cardiovascular disease plays a major role in the pathogenesis of arrhythmias. Cardiac neural control relies on complex feedback loops consisting of efferent and afferent limbs, which carry sympathetic and parasympathetic signals from the brain to the heart and sensory signals from the heart to the brain. Cardiac disease leads to neural remodeling and sympathovagal imbalances with arrhythmogenic effects. Preclinical studies of modulation at central and peripheral levels of the cardiac autonomic nervous system have yielded promising results, leading to early stage clinical studies of these techniques in atrial fibrillation and refractory ventricular arrhythmias, particularly in patients with inherited primary arrhythmia syndromes and structural heart disease. However, significant knowledge gaps in basic cardiac neurophysiology limit the success of these neuromodulatory therapies. This review discusses the recent advances in neuromodulation for cardiac arrhythmia management, with a clinical scenario-based approach aimed at bringing neurocardiology closer to the realm of the clinical electrophysiologist.


Subject(s)
Atrial Fibrillation , Electric Stimulation Therapy , Heart Conduction System/physiology , Tachycardia, Ventricular , Atrial Fibrillation/physiopathology , Atrial Fibrillation/therapy , Autonomic Nervous System/physiology , Heart/innervation , Heart/physiology , Humans , Tachycardia, Ventricular/physiopathology , Tachycardia, Ventricular/therapy
14.
Nat Rev Cardiol ; 16(12): 707-726, 2019 12.
Article in English | MEDLINE | ID: mdl-31197232

ABSTRACT

Research into cardiac autonomic control has received great interest in the past 20 years, and we are now at a critical juncture with regard to the clinical translation of the experimental findings. A rush to develop clinical interventions and implant a range of devices aimed at cardiac neuromodulation therapy has occurred. This interest has been driven by research, superimposed on commercial opportunities and perhaps the more relaxed regulatory framework governing implantable devices and interventions compared with that for pharmacotherapy. However, many of the results of the clinical trials into these therapies have been disappointing or conflicting. This lack of positive results is partly attributable to a scramble to find simple solutions for complex problems that we do not yet fully understand. Are there reasons to be optimistic? In this Review, we highlight areas in the field of cardiac autonomic control that we feel show the most promise for clinical translation and areas in which our current range of blunt tools need to be refined to bring about long-term success in treating arrhythmias.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/therapy , Autonomic Nervous System/physiopathology , Anti-Arrhythmia Agents/therapeutic use , Catheter Ablation , Death, Sudden, Cardiac/etiology , Heart/innervation , Humans , Transcutaneous Electric Nerve Stimulation
15.
Am J Physiol Heart Circ Physiol ; 317(2): H226-H233, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31149841

ABSTRACT

Cardiac sympathetic overdrive provides inotropic support to the failing heart. However, as myocardial insult evolves, this compensatory response impairs contractile function and constitutes an independent mortality predictor and a primary target in the treatment of heart failure (HF). In this prospective, randomized, double-blind, controlled crossover trial, we proposed cervicothoracic transcutaneous electrical nerve stimulation (CTENS) as a nonpharmacological therapy on cardiac sympathetic activity in patients with HF. Seventeen patients with HF were randomly assigned to an in-home CTENS (30 min twice daily, 80-Hz frequency, and 150-µs pulse duration) or a control intervention (Sham) for 14 consecutive days. Following a 60-day washout phase, patients were crossed over to the opposite intervention. The heart-to-mediastinum ratio (HMR) and washout rate (WR) (indexes of sympathetic innervation density and activity from planar 123iodo-metaiodobenzylguanidine myocardial scintigraphy images, respectively), as well as blood pressure (BP) and heart rate (HR), were quantified before and after each intervention. HMR, BP, and HR did not change throughout the study. Nonetheless, CTENS reduced WR (CTENS -4 ± 10 vs. Sham +5 ± 15%, P = 0.03) when compared with Sham. When allocated in two independent groups, preserved (PCSI, HMR > 1.6, n = 10) and impaired cardiac sympathetic innervation (ICSI, HRM ≤1.6, n = 7), PCSI patients showed an important attenuation of WR (-11 ± 9 vs. Sham +8 ± 19%, P = 0.007) after CTENS. Nonetheless, neither Sham nor CTENS evoked changes in WR of the ICSI patients (P > 0.05). These findings indicate that CTENS attenuates the cardiac sympathetic overdrive in patients with HF and a preserved innervation constitutes an essential factor for this beneficial neuromodulatory impact. Clinical Trial Registration: URL: https://www.clinicaltrials.gov . Identifier: NCT03354689. NEW & NOTEWORTHY We found that short-term cervicothoracic transcutaneous electrical nerve stimulation (CTENS) attenuates cardiac sympathetic overdrive in patients with heart failure and a preserved autonomic innervation may constitute an essential factor to maximize this beneficial neuromodulatory effect. CTENS then emerges as an alternative noninvasive and nonpharmacological strategy to attenuate exaggerated cardiac sympathetic drive in patients with heart failure.


Subject(s)
3-Iodobenzylguanidine/administration & dosage , Heart Failure/therapy , Heart/innervation , Iodine Radioisotopes/administration & dosage , Myocardial Contraction , Radiopharmaceuticals/administration & dosage , Sympathetic Nervous System/physiopathology , Transcutaneous Electric Nerve Stimulation , Aged , Blood Pressure , Brazil , Cross-Over Studies , Double-Blind Method , Female , Heart Failure/diagnostic imaging , Heart Failure/physiopathology , Heart Rate , Humans , Male , Middle Aged , Prospective Studies , Sympathetic Nervous System/diagnostic imaging , Time Factors , Transcutaneous Electric Nerve Stimulation/adverse effects , Treatment Outcome
16.
J Cardiovasc Electrophysiol ; 30(7): 1138-1147, 2019 07.
Article in English | MEDLINE | ID: mdl-31104349

ABSTRACT

BACKGROUND: Sympathetic neural activation plays a key role in the incidence and maintenance of acute myocardial infarction (AMI) induced ventricular arrhythmia (VA). Furthermore, previous studies showed that AMI might induce microglia and sympathetic activation and that microglial activation might contribute to sympathetic activation. Recently, studies showed that light emitting diode (LED) therapy might attenuate microglial activation. Therefore, we hypothesized that LED therapy might reduce AMI-induced VA by attenuating microglia and sympathetic activation. METHODS: Thirty anesthetized rats were randomly divided into three groups: the Control group (n = 6), AMI group (n = 12), and AMI + LED group (n = 12). Electrocardiogram (ECG) and left stellate ganglion (LSG) neural activity were continuously recorded. The incidence of VAs was recorded during the first hour after AMI. Furthermore, we sampled the brain and myocardium tissue of the different groups to examine the microglial activation and expression of nerve growth factor (NGF), interleukin-18 (IL-18), and IL-1ß, respectively. RESULTS: Compared to the AMI group, LED therapy significantly reduced the incidence of AMI-induced VAs (ventricular premature beats [VPB] number: 85.08 ± 13.91 vs 27.5 ± 9.168, P < .01; nonsustained ventricular tachycardia (nSVT) duration: 34.39 ± 8.562 vs 9.005 ± 3.442, P < .05; nSVT number: 18.92 ± 4.52 vs 7.583 ± 3.019, P < .05; incidence rate of SVT/VF: 58.33% vs. 8.33%, P < .05) and reduced the LSG neural activity (P < .01) in the AMI + LED group. Furthermore, LED significantly attenuated microglial activation and reduced IL-18, IL-1ß, and NGF expression in the peri-infarct myocardium. CONCLUSION: LED therapy may protect against AMI-induced VAs by suppressing sympathetic neural activity and the inflammatory response.


Subject(s)
Heart/innervation , Lasers, Semiconductor , Low-Level Light Therapy/instrumentation , Myocardial Infarction/radiotherapy , Neuroimmunomodulation , Paraventricular Hypothalamic Nucleus/physiopathology , Stellate Ganglion/physiopathology , Tachycardia, Supraventricular/prevention & control , Tachycardia, Ventricular/prevention & control , Ventricular Premature Complexes/prevention & control , Action Potentials , Animals , Disease Models, Animal , Heart Rate , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Male , Microglia/metabolism , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/metabolism , Nerve Growth Factor/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Rats, Sprague-Dawley , Tachycardia, Supraventricular/etiology , Tachycardia, Supraventricular/metabolism , Tachycardia, Supraventricular/physiopathology , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Time Factors , Ventricular Premature Complexes/etiology , Ventricular Premature Complexes/metabolism , Ventricular Premature Complexes/physiopathology
17.
Medicine (Baltimore) ; 98(8): e14657, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30813209

ABSTRACT

Recent studies reported that high doses of short-acting loop diuretics are associated with poor outcomes in patients with heart failure (HF). Short-acting loop diuretics have been shown to activate the renin-angiotensin system (RAS) and have no favorable effects on cardiac sympathetic nervous system (SNS) activity. The goal of this study is to investigate the relationship between daily doses of furosemide and the outcomes of patients with left ventricular dysfunction (LVD) from the viewpoint of cardiac SNS abnormalities using iodine-123-labeled metaiodobenzylguanidine (l-MIBG) myocardial scintigraphy.We enrolled 137 hospitalized patients (62.5 ±â€Š14.2 years old, 103 men) with LVEF < 45% who underwent l-MIBG myocardial scintigraphy. A delayed heart-to-mediastinum ratio (delayed HMR) was assessed using l-MIBG scintigraphy. Cardiac events were defined as cardiac death or re-hospitalization due to the deterioration of HF. Cox proportional hazard analysis was used to identify predictors of cardiac events.Cardiac events occurred in 57 patients in a follow-up period of 33.1 ±â€Š30 months. In a multivariate Cox proportional hazard analysis, delayed HMR and furosemide doses were identified as independent predictors of cardiac events (P = .0042, P = .033, respectively). Inverse probability of treatment weighting Cox modeling showed that the use of furosemide (≥40 mg /day) was associated with cardiac events with a hazard ratio of 1.96 (P = .003). In the Kaplan-Mayer analysis, the cardiac event-free survival rate was significantly lower in patients treated with high doses of furosemide (≥60 mg/day vs 40-60 mg/day vs <40 mg/day, the Log-rank test P < .0001). In a receiver-operating characteristic (ROC) analysis, the cut-off value for cardiac events was 40 mg/day of furosemide. The cardiac event-free rate was significantly lower in patients with delayed HMR <1.8 (median value) and receiving furosemide ≥40 mg/day than in other patients (the Log-rank test P < .0001). Significant differences in cardiac event rates according to furosemide doses among patients with delayed HMR <1.8 were observed among patients without ß-blocker therapy (P = .001), but not among those with ß-blocker therapy (P = .127).The present results indicate that a relationship exists between higher doses of furosemide and poor outcomes. The prognosis of HF patients with severe cardiac SNS abnormalities receiving high-dose short-acting loop diuretics is poor.


Subject(s)
Furosemide , Heart Failure , Heart , Sympathetic Nervous System/drug effects , Ventricular Dysfunction, Left , 3-Iodobenzylguanidine/pharmacology , Aged , Biological Availability , Dose-Response Relationship, Drug , Female , Furosemide/administration & dosage , Furosemide/adverse effects , Furosemide/pharmacokinetics , Heart/diagnostic imaging , Heart/innervation , Heart Failure/diagnosis , Heart Failure/drug therapy , Heart Failure/physiopathology , Humans , Japan , Male , Middle Aged , Myocardial Perfusion Imaging/methods , Outcome and Process Assessment, Health Care , Radiopharmaceuticals/pharmacology , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Sodium Potassium Chloride Symporter Inhibitors/adverse effects , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Stroke Volume/drug effects , Ventricular Dysfunction, Left/diagnosis , Ventricular Dysfunction, Left/drug therapy
18.
Can J Physiol Pharmacol ; 97(2): 90-98, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30462556

ABSTRACT

Although depression and cardiovascular diseases are related, the role of antidepressants such as fluoxetine (increasing serotonin levels) within cardiac regulation remains unclear. We aimed to determine whether fluoxetine modifies the pharmacological profile of serotonergic influence on vagal cardiac outflow. Rats were treated with fluoxetine (10 mg/kg per day; p.o.) for 14 days or equivalent volumes of drinking water (control group); then, they were pithed and prepared for vagal stimulation. Bradycardic responses were obtained by electrical stimulation of the vagal fibers (3, 6, and 9 Hz) or i.v. acetylcholine (ACh; 1, 5, and 10 µg/kg). The i.v. administration of 5-hydroxytryptamine (5-HT; 10 and 50 µg/kg) inhibited the vagally induced bradycardia. 5-CT (5-HT1/7 agonist) and L-694,247 (5-HT1D agonist) mimicked the serotonin inhibitory effect while α-methyl-5-HT (5-HT2 agonist) was devoid of any action. SB269970 (5-HT7 antagonist) did not abolish 5-CT inhibitory action on the electrically induced bradycardia. Pretreatment with LY310762 (5-HT1D antagonist) blocked the effects induced by L-694,247 and 5-CT. 5-HT and 5-CT failed to modify the bradycardia induced by exogenous ACh. Our outcomes suggest that fluoxetine treatment modifies 5-HT modulation on heart parasympathetic neurotransmission in rats, evoking inhibition of the bradycardia via prejunctional 5-HT1D in pithed rats.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Bradycardia/drug therapy , Fluoxetine/pharmacology , Receptor, Serotonin, 5-HT1D/metabolism , Vagus Nerve/drug effects , Administration, Oral , Animals , Antidepressive Agents, Second-Generation/therapeutic use , Bradycardia/etiology , Depression/complications , Depression/drug therapy , Disease Models, Animal , Drug Evaluation, Preclinical , Fluoxetine/therapeutic use , Heart/innervation , Heart Rate/drug effects , Humans , Oxadiazoles/pharmacology , Phenols/pharmacology , Rats , Rats, Wistar , Serotonin/pharmacology , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Serotonin Receptor Agonists/metabolism , Sulfonamides/pharmacology , Tryptamines/pharmacology , Vagus Nerve/metabolism
19.
Nutrients ; 10(9)2018 Sep 17.
Article in English | MEDLINE | ID: mdl-30227610

ABSTRACT

Marine n-3 polyunsaturated fatty acids (PUFA) may improve autonomic dysfunction, as indicated by an increase in heart rate variability (HRV) and reduce the risk of sudden cardiac death. Hence, the aim of this study was to investigate the effects of marine n-3 PUFA on 24-h HRV in patients on chronic dialysis, who have a high risk of sudden cardiac death. Between June 2014 and March 2016, 112 patients on chronic dialysis from Denmark were allocated to a daily supplement of 2 g marine n-3 PUFA or control for three months in a randomized, double-blinded, controlled trial. A 48-h Holter monitoring was performed and mean 24-h HRV indices for the two days were available in 85 patients. The mean age was 62.3 years (SD: 14.3) and median dialysis vintage was 1.7 years (IQR: 0.5, 6.4). Within-group and between-group changes in outcome were evaluated by a paired and two sample t-test, respectively. Marine n-3 PUFA did not change the primary endpoint SDNN (SD of all RR-intervals) reflecting overall HRV, but other HRV indices increased and the mean RR-interval increased significantly, corresponding to a decrease in heart rate by 2.5 beats per minute (p = 0.04). In conclusion, marine n-3 PUFA did not change SDNN, but the mean heart rate was significantly reduced and changes in other HRV-indices were also observed, indicating an increase in vagal modulation that might be protective against malignant ventricular arrhythmias.


Subject(s)
Autonomic Nervous System/physiopathology , Death, Sudden, Cardiac/prevention & control , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Heart Rate , Heart/innervation , Renal Dialysis , Renal Insufficiency, Chronic/therapy , Aged , Denmark , Double-Blind Method , Female , Humans , Male , Middle Aged , Renal Dialysis/adverse effects , Renal Dialysis/mortality , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/mortality , Renal Insufficiency, Chronic/physiopathology , Risk Factors , Time Factors , Treatment Outcome
20.
Trials ; 19(1): 459, 2018 Aug 25.
Article in English | MEDLINE | ID: mdl-30144819

ABSTRACT

BACKGROUND: Recuperative techniques have been used to anticipate and potentiate recovery. The massage is one of the most widely used in sports. Among the ways to demonstrate the recovery of the organism is the resumption of autonomic modulation of heart rate, which can be analyzed in situations that cause disturbances in the behavior of the cardiovascular system with the objective of verifying the responsiveness of the autonomic nervous system (ANS). Recovery can be assessed through heart rate variability (HRV) which analyzes the oscillations in consecutive heartbeats, thus allowing an important non-invasive alternative for the study of modulation of the ANS. The objective of the study will be to measure the effects of massage as a recuperative technique on the autonomic modulation of heart rate and cardiorespiratory parameters at different moments of application. METHODS: This is a randomized, cross-over clinical trial. Forty men aged 18 to 30 years, healthy and physically active according to the International Physical Activity Questionnaire will participate in the study. Participants will be randomized into groups, which will perform the five interventions of the study at randomized moments, one intervention per session: Intervention 1: control; Intervention 2: participants will receive the massage protocol; Intervention 3: performance of the stress protocol; Intervention 4: participants will perform the stress protocol and immediately after receive the massage; Intervention 5: participants will perform the stress protocol and 1 h after conclusion of the protocol will receive the massage. The sessions will occur with an interval of 1 week between them and, due to the technique used, blinding participants and therapists is not possible. The primary outcome measure is HRV that will be measured 2 h after the conclusion of each intervention, and secondary outcome measures, which include heart rate, respiratory rate, blood pressure, oxygen saturation, and individual touch perception, will be measured at specific moments in the course of each intervention. DISCUSSION: The implementation and use of this standardized protocol should provide important and reliable information regarding the use of massage in post-exercise recovery, with the identification of its effects on the ANS and the best timing and form of massage application. The data obtained in the present study will provide subsidies for the best management of application of the technique in sports clinical practice, considering periods of training and, mainly, of competitions. TRIAL REGISTRATION: ClinicalTrials.gov, ID: NCT03094676 . Pre-results. 12 March 2018.


Subject(s)
Autonomic Nervous System/physiology , Exercise/physiology , Heart Rate , Heart/innervation , Lung/innervation , Massage , Respiratory Rate , Stress, Physiological , Adolescent , Adult , Brazil , Cross-Over Studies , Humans , Male , Randomized Controlled Trials as Topic , Recovery of Function , Time Factors , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL