Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Complementary Medicines
Publication year range
1.
J Med Toxicol ; 16(4): 465-469, 2020 10.
Article in English | MEDLINE | ID: mdl-32656624

ABSTRACT

INTRODUCTION: Overdoses of beta-adrenergic antagonists and calcium channel antagonists represent an uncommonly encountered but highly morbid clinical presentation. Potential therapies include fluids, calcium salts, vasopressors, intravenous lipid emulsion, methylene blue, and high-dose insulin. Although high-dose insulin is commonly used, the kinetics of insulin under these conditions are unknown. CASE REPORT: We present a case of a 51-year-old male who sustained a life-threatening overdose after ingesting approximately 40 tablets of a mixture of amlodipine 5 mg and metoprolol tartrate 25 mg. Due to severe bradycardia and hypotension, he was started on high-dose insulin (HDI) therapy; this was augmented with epinephrine. Despite the degree of his initial shock state, he ultimately recovered, and HDI was discontinued. Insulin was infused for a total of approximately 37 hours, most of which was dosed at 10 U/kg/hour; following discontinuation, serial serum insulin levels were drawn and remained at supraphysiologic levels for at least 24 hours and well above reference range for multiple days thereafter. CONCLUSION: The kinetics of insulin following discontinuation of high-dose insulin therapy are largely unknown, but supraphysiologic insulin levels persist for some time following therapy; this may allow for simple discontinuation rather than titration of insulin at the end of therapy. Dextrose replacement is frequently needed; although the duration is often difficult to predict, prolonged infusions may not be necessary.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/poisoning , Amlodipine/poisoning , Bradycardia/drug therapy , Calcium Channel Blockers/poisoning , Hyperinsulinism/chemically induced , Hypoglycemic Agents/administration & dosage , Hypotension/drug therapy , Insulin/administration & dosage , Metoprolol/poisoning , Bradycardia/chemically induced , Bradycardia/diagnosis , Bradycardia/physiopathology , Drug Administration Schedule , Drug Overdose , Humans , Hyperinsulinism/blood , Hyperinsulinism/diagnosis , Hypoglycemic Agents/blood , Hypoglycemic Agents/pharmacokinetics , Hypotension/chemically induced , Hypotension/diagnosis , Hypotension/physiopathology , Infusions, Intravenous , Insulin/blood , Insulin/pharmacokinetics , Male , Middle Aged , Suicide, Attempted
2.
Neurosci Lett ; 735: 135196, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32585256

ABSTRACT

Docosahexaenoic acid (DHA) an omega-3 polyunsaturated fatty acid, is an agonist of FFA1 receptor. DHA administration reduces the heart rate via unclear mechanisms. We examined the effect of DHA on neurons of nucleus ambiguus that provide the parasympathetic control of heart rate. DHA produced a dose-dependent increase in cytosolic Ca2+ concentration in cardiac-projecting nucleus ambiguus neurons; the effect was prevented by GW1100, a FFA1 receptor antagonist. DHA depolarized cultured nucleus ambiguus neurons via FFA1 activation. Bilateral microinjection of DHA into nucleus ambiguus produced bradycardia in conscious rats. Our results indicate that DHA decreases heart rate by activation of FFA1 receptor on cardiac-projecting nucleus ambiguus neurons.


Subject(s)
Bradycardia/chemically induced , Fatty Acids, Omega-3/administration & dosage , Heart Rate/drug effects , Heart Rate/physiology , Medulla Oblongata/drug effects , Medulla Oblongata/physiology , Animals , Animals, Newborn , Bradycardia/physiopathology , Cells, Cultured , Male , Microinjections , Rats , Rats, Sprague-Dawley
3.
J Cardiovasc Med (Hagerstown) ; 21(6): 420-427, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32332379

ABSTRACT

BACKGROUND: Temporary transvenous cardiac pacing (TTCP) is a standard procedure in current practice, despite limited coverage in consensus guidelines. However, many authors reported several complications associated with TTCP, especially development of infections of cardiac implantable electronic devices (CIED). The aim of this survey was to provide a country-wide picture of current practice regarding TTCP. METHODS: Data were collected using an online survey that was administered to members of the Italian Association of Arrhythmology and Cardiac Pacing. RESULTS: We collected data from 102 physicians, working in 81 Italian hospitals from 17/21 regions. Our data evidenced that different strategies are adopted in case of acute bradycardia with a tendency to limit TTCP mainly to advanced atrioventricular block. However, some centers reported a greater use in elective procedures. TTCP is usually performed by electrophysiologists or interventional cardiologists and, differently from previous reports, mainly by a femoral approach and with nonfloating catheters. We found high inhomogeneity regarding prevention of infections and thromboembolic complications and in post-TTCP management, associated with different TTCP volumes and a strategy for management of acute bradyarrhythmias. CONCLUSION: This survey evidenced a high inhomogeneity in the approaches adopted by Italian cardiologists for TTCP. Further studies are needed to explore if these divergences are associated with different long-term outcomes, especially incidence of CIED-related infections.


Subject(s)
Bradycardia/therapy , Cardiac Electrophysiology/trends , Cardiac Pacing, Artificial/trends , Cardiologists/trends , Electrophysiologic Techniques, Cardiac/trends , Pacemaker, Artificial/trends , Practice Patterns, Physicians'/trends , Bradycardia/diagnosis , Bradycardia/physiopathology , Health Care Surveys , Healthcare Disparities/trends , Humans , Italy
4.
Prog Biophys Mol Biol ; 157: 76-83, 2020 11.
Article in English | MEDLINE | ID: mdl-31726065

ABSTRACT

It has long been known that the electrophysiological effects of many cardioactive drugs strongly depend on the rate dependent frequency. This was recognized first for class I antiarrhythmic agents: their Vmax suppressive effect was attenuated at long cycle lengths. Later many Ca2+ channel blockers were also found to follow such kinetics. The explanation was provided by the modulated and the guarded receptor theories. Regarding the duration of cardiac action potentials (APD) an opposite frequency-dependence was observed, i.e. the drug-induced changes in APD were proportional with the cycle length of stimulation, therefore it was referred as "reverse rate-dependency". The beat-to-beat, or short term variability of APD (SV) has been recognized as an important proarrhythmic mechanism, its magnitude can be used as an arrhythmia predictor. SV is modulated by several cardioactive agents, however, these drugs modify also APD itself. In order to clear the drug-specific effects on SV from the concomitant unspecific APD-change related ones, the term of "relative variability" was introduced. Relative variability is increased by ion channel blockers that decrease the negative feedback control of APD (i.e. blockers of ICa, IKr and IKs) and also by elevation of cytosolic Ca2+. Cardiac arrhythmias are also often categorized according to the characteristic heart rate (tachy- and bradyarrhythmias). Tachycardia is proarrhythmic primarily due to the concomitant Ca2+ overload causing delayed afterdepolarizations. Early afterdepolarizations (EADs) are complications of the bradycardic heart. What is common in the reverse rate-dependent nature of drug action on APD, increased SV and EAD incidence associated with bradycardia.


Subject(s)
Action Potentials/physiology , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Heart Rate/drug effects , Heart Ventricles/drug effects , Potassium Channel Blockers/pharmacology , Animals , Bradycardia/physiopathology , Dogs , Drug Evaluation, Preclinical , Electrophysiology , Guinea Pigs , Humans , Ions , Kinetics , Male , Mice , Myocytes, Cardiac/physiology , Pharmaceutical Preparations , Rabbits , Tachycardia/physiopathology
5.
J Cardiovasc Electrophysiol ; 30(10): 2164-2169, 2019 10.
Article in English | MEDLINE | ID: mdl-31456266

ABSTRACT

BACKGROUND: His bundle pacing (HBP) is a physiological pacing modality, but HBP implantation remains a challenge. OBJECTIVE: This study explored the feasibility of using visualization of the tricuspid valve annulus (TVA) to locate the site for HBP. METHODS: During the lead placement in eight patients with symptomatic bradycardia, the TVA and tricuspid septal leaflet was revealed by contrast injection in the right ventricle under the fluoroscopic right anterior oblique view, and the target site for HBP was identified near the intersection of the tricuspid septal leaflet and the interventricular septum. On the basis of the imaging marker, the pacing lead was placed for HBP at either the atrial (aHBP) or ventricular side (vHBP). RESULTS: During the implantation, the pacing lead placement was attempted for aHBP in two patients, vHBP in five patients, and first for aHBP then vHBP in one patient. The aHBP was selective and had a capture threshold of 1.6 ± 0.5 V@ 1.0ms and R-wave amplitude of 1.2 ± 0.4 mV. Ventricular-side His bundle capture was selective in four patients and nonselective in two patients. The vHBP capture threshold was 0.8 ± 0.4 V@ 1.0ms (P < .05 vs aHBP) and R-wave amplitude was 4.1 ± 1.5 mV (P < .05 vs aHBP). At the final pacing programming of 3.0 V@ 1.0ms, vHBP was nonselective in all six patients and aHBP remained selective in two patients. Pacing parameters remained stable at 3 months. CONCLUSION: The location of the TVA and tricuspid septal leaflet revealed by right ventriculography can be used as a landmark to identify the HBP site.


Subject(s)
Anatomic Landmarks , Bradycardia/surgery , Bundle of His/physiopathology , Cardiac Pacing, Artificial , Pacemaker, Artificial , Tricuspid Valve/diagnostic imaging , Action Potentials , Aged , Bradycardia/diagnosis , Bradycardia/physiopathology , Contrast Media/administration & dosage , Electrocardiography , Electrophysiologic Techniques, Cardiac , Feasibility Studies , Female , Fluoroscopy , Heart Rate , Humans , Male , Middle Aged , Treatment Outcome
6.
Circ Arrhythm Electrophysiol ; 12(6): e007415, 2019 06.
Article in English | MEDLINE | ID: mdl-31113233

ABSTRACT

Background His-bundle pacing (HBP) is a physiological form of pacing. Although high capture thresholds are common, few predictors of low HBP threshold have been determined. We aimed to identify electrophysiological predictors. Methods Fifty-one patients (53% with atrioventricular block) underwent HBP for bradycardia with an intrinsic QRS duration of <120 ms. Attempts to anchor the HBP lead were guided by unipolar His-bundle electrograms (HB EGMs) recorded with an electrophysiology recording system. Patients were followed-up for >6 months. Results In total, 153 attempts at anchoring the HBP lead were made, of which, 45 achieved acceptable HBP thresholds (≤2.5 V at 1 ms). The amplitude of negative deflection in HB EGM and the selective HBP form at fixation were independently associated with achieving an acceptable threshold. A negative amplitude of ≥0.060 mV in HB EGM was determined as the optimal value for identifying the acceptable threshold. This deep negative HB EGM was recorded with an HBP threshold of 1.4±1.3 V (in 34 attempts), significantly lower than that of positive HB EGM without deep negative deflection (2.8±1.3 V, in 31 trials; or >5 V, in 38 trials). The permanent HBP lead remained with deep negative (≥0.060 mV) or positive HB EGMs in 28 and 14 patients, respectively, and with positive or negative HB injury current in 19 and 23 patients, respectively. During follow-up, increased HBP threshold of >1 V was significantly more prevalent in the positive HB EGM group. The HBP thresholds of deep negative HB EGM and HB injury current, but not of the selective HBP group, were significantly lower than the other subgroups during follow-up. Conclusions Deep negative HB EGM at fixation was associated with an excellent short-term HBP threshold, similar to HB injury current. Analysis of unipolar HB EGM postfixation may enable prediction of permanent HBP threshold.


Subject(s)
Action Potentials , Atrioventricular Block/diagnosis , Atrioventricular Block/therapy , Bradycardia/diagnosis , Bundle of His/physiopathology , Cardiac Pacing, Artificial , Electrophysiologic Techniques, Cardiac , Heart Rate , Aged , Aged, 80 and over , Atrioventricular Block/physiopathology , Bradycardia/physiopathology , Bradycardia/therapy , Cardiac Pacing, Artificial/adverse effects , Female , Humans , Male , Predictive Value of Tests , Prospective Studies , Registries , Risk Factors , Time Factors , Treatment Outcome
7.
Circulation ; 139(18): 2157-2169, 2019 04 30.
Article in English | MEDLINE | ID: mdl-30764634

ABSTRACT

BACKGROUND: Bradyarrhythmia is a common clinical manifestation. Although the majority of cases are acquired, genetic analysis of families with bradyarrhythmia has identified a growing number of causative gene mutations. Because the only ultimate treatment for symptomatic bradyarrhythmia has been invasive surgical implantation of a pacemaker, the discovery of novel therapeutic molecular targets is necessary to improve prognosis and quality of life. METHODS: We investigated a family containing 7 individuals with autosomal dominant bradyarrhythmias of sinus node dysfunction, atrial fibrillation with slow ventricular response, and atrioventricular block. To identify the causative mutation, we conducted the family-based whole exome sequencing and genome-wide linkage analysis. We characterized the mutation-related mechanisms based on the pathophysiology in vitro. After generating a transgenic animal model to confirm the human phenotypes of bradyarrhythmia, we also evaluated the efficacy of a newly identified molecular-targeted compound to upregulate heart rate in bradyarrhythmias by using the animal model. RESULTS: We identified one heterozygous mutation, KCNJ3 c.247A>C, p.N83H, as a novel cause of hereditary bradyarrhythmias in this family. KCNJ3 encodes the inwardly rectifying potassium channel Kir3.1, which combines with Kir3.4 (encoded by KCNJ5) to form the acetylcholine-activated potassium channel ( IKACh channel) with specific expression in the atrium. An additional study using a genome cohort of 2185 patients with sporadic atrial fibrillation revealed another 5 rare mutations in KCNJ3 and KCNJ5, suggesting the relevance of both genes to these arrhythmias. Cellular electrophysiological studies revealed that the KCNJ3 p.N83H mutation caused a gain of IKACh channel function by increasing the basal current, even in the absence of m2 muscarinic receptor stimulation. We generated transgenic zebrafish expressing mutant human KCNJ3 in the atrium specifically. It is interesting to note that the selective IKACh channel blocker NIP-151 repressed the increased current and improved bradyarrhythmia phenotypes in the mutant zebrafish. CONCLUSIONS: The IKACh channel is associated with the pathophysiology of bradyarrhythmia and atrial fibrillation, and the mutant IKACh channel ( KCNJ3 p.N83H) can be effectively inhibited by NIP-151, a selective IKACh channel blocker. Thus, the IKACh channel might be considered to be a suitable pharmacological target for patients who have bradyarrhythmia with a gain-of-function mutation in the IKACh channel.


Subject(s)
Atrial Fibrillation , Atrioventricular Block , Bradycardia , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Genetic Diseases, Inborn , Mutation, Missense , Amino Acid Substitution , Animals , Animals, Genetically Modified , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Atrial Fibrillation/physiopathology , Atrioventricular Block/genetics , Atrioventricular Block/metabolism , Atrioventricular Block/pathology , Atrioventricular Block/physiopathology , Benzopyrans/pharmacology , Bradycardia/genetics , Bradycardia/metabolism , Bradycardia/pathology , Bradycardia/physiopathology , Electrophysiologic Techniques, Cardiac , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Genetic Diseases, Inborn/physiopathology , Humans , Male , Xenopus laevis , Zebrafish
10.
Heart Rhythm ; 16(6): 853-860, 2019 06.
Article in English | MEDLINE | ID: mdl-30550835

ABSTRACT

BACKGROUND: The relationships between hemodynamic consequences of premature ventricular contractions (PVCs) and development of premature ventricular contraction-induced cardiomyopathy (PVC-CM) have not been investigated. OBJECTIVE: The purpose of this study was to correlate concealed mechanical bradycardia and/or postextrasystolic potentiation (PEP) to PVC-CM. METHODS: Invasive arterial pressure measurements from 17 patients with PVC-CM and 16 controls with frequent PVCs were retrospectively analyzed. PVCs were considered efficient (ejecting PVCs) when generating a measurable systolic arterial pressure. PEP was defined by a systolic arterial pressure of the post-PVC beat ≥5 mm Hg higher than the preceding sinus beat. Every PVC was analyzed for 10 minutes before ablation, and the electromechanical index (EMi = number of ejecting PVCs/total PVC) and postextrasystolic potentiation index (PEPi = number of PVCs with PEP/total PVC) were calculated. RESULTS: EMi was 29% ± 31% in PVC-CM and 78% ± 20% in controls (P <.0001). PEPi was 41% ± 28% in PVC-CM and 14% ± 10% in controls (P = .001). There was no control in groups of low EMi or high PEPi. EMi and PEPi were not significantly correlated to left ventricular dimensions or function in PVC-CM patients. PVC coupling interval was related to both ejecting PVCs and PEP. CONCLUSION: Patients with PVC-CM more often display nonejecting PVCs and PEP compared to controls.


Subject(s)
Arterial Pressure , Bradycardia , Cardiomyopathies , Heart Ventricles , Ventricular Premature Complexes , Anti-Arrhythmia Agents/therapeutic use , Blood Pressure , Blood Pressure Determination/methods , Bradycardia/diagnosis , Bradycardia/physiopathology , Cardiac Electrophysiology , Cardiomyopathies/diagnosis , Cardiomyopathies/etiology , Cardiomyopathies/physiopathology , Correlation of Data , Echocardiography/methods , Electrophysiologic Techniques, Cardiac/methods , Female , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Male , Middle Aged , Organ Size , Ventricular Premature Complexes/complications , Ventricular Premature Complexes/diagnosis , Ventricular Premature Complexes/drug therapy , Ventricular Premature Complexes/physiopathology
11.
Int Heart J ; 59(3): 601-606, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29628472

ABSTRACT

A biological pacemaker is expected to solve the persisting problems of an artificial cardiac pacemaker including short battery life, lead breaks, infection, and electromagnetic interference. We previously reported HCN4 overexpression enhances pacemaking ability of mouse embryonic stem cell-derived cardiomyocytes (mESC-CMs) in vitro. However, the effect of these cells on bradycardia in vivo has remained unclear. Therefore, we transplanted HCN4-overexpressing mESC-CMs into bradycardia model animals and investigated whether they could function as a biological pacemaker. The rabbit Hcn4 gene was transfected into mouse embryonic stem cells and induced HCN4-overexpressing mESC-CMs. Non-cardiomyocytes were removed under serum/glucose-free and lactate-supplemented conditions. Cardiac balls containing 5 × 103 mESC-CMs were made by using the hanging drop method. One hundred cardiac balls were injected into the left ventricular free wall of complete atrioventricular block (CAVB) model rats. Heart beats were evaluated using an implantable telemetry system 7 to 30 days after cell transplantation. The result showed that ectopic ventricular beats that were faster than the intrinsic escape rhythm were often observed in CAVB model rats transplanted with HCN4-overexpressing mESC-CMs. On the other hand, the rats transplanted with non-overexpressing mESC-CMs showed sporadic single premature ventricular contraction but not sustained ectopic ventricular rhythms. These results indicated that HCN4-overexpressing mESC-CMs produce rapid ectopic ventricular rhythms as a biological pacemaker.


Subject(s)
Biological Clocks/physiology , Bradycardia/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Animals , Bradycardia/physiopathology , Cell Line , Disease Models, Animal , Fluorescent Antibody Technique , Heart Rate/physiology , Mice , Mouse Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Rabbits , Rats , Telemetry , Transfection
12.
Sci Rep ; 8(1): 1900, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29382866

ABSTRACT

Stimulation of vagal afferent endings with intravenous phenylbiguanide (PBG) causes both bradycardia and vasodepression, simulating neurally mediated syncope. Activation of µ-opioid receptors in the nucleus tractus solitarius (NTS) increases blood pressure. Electroacupuncture (EA) stimulation of somatosensory nerves underneath acupoints P5-6, ST36-37, LI6-7 or G37-39 selectively but differentially modulates sympathoexcitatory responses. We therefore hypothesized that EA-stimulation at P5-6 or ST36-37, but not LI6-7 or G37-39 acupoints, inhibits the bradycardia and vasodepression through a µ-opioid receptor mechanism in the NTS. We observed that stimulation at acupoints P5-6 and ST36-37 overlying the deep somatosensory nerves and LI6-7 and G37-39 overlying cutaneous nerves differentially evoked NTS neural activity in anesthetized and ventilated animals. Thirty-min of EA-stimulation at P5-6 or ST36-37 reduced the depressor and bradycardia responses to PBG while EA at LI6-7 or G37-39 did not. Congruent with the hemodynamic responses, EA at P5-6 and ST36-37, but not at LI6-7 and G37-39, reduced vagally evoked activity of cardiovascular NTS cells. Finally, opioid receptor blockade in the NTS with naloxone or a specific µ-receptor antagonist reversed P5-6 EA-inhibition of the depressor, bradycardia and vagally evoked NTS activity. These data suggest that point specific EA stimulation inhibits PBG-induced vasodepression and bradycardia responses through a µ-opioid mechanism in the NTS.


Subject(s)
Analgesics, Opioid/pharmacology , Bradycardia/drug therapy , Bradycardia/physiopathology , Solitary Nucleus/drug effects , Solitary Nucleus/physiopathology , Vasodilator Agents/pharmacology , Acupuncture Points , Animals , Blood Pressure/drug effects , Bradycardia/metabolism , Cardiovascular System/drug effects , Cardiovascular System/metabolism , Cardiovascular System/physiopathology , Cats , Electroacupuncture/methods , Female , Heart Rate/drug effects , Hemodynamics/drug effects , Male , Naloxone/pharmacology , Receptors, Opioid/metabolism , Reflex/drug effects , Solitary Nucleus/metabolism , Vagus Nerve/drug effects , Vagus Nerve/metabolism , Vagus Nerve/physiopathology
13.
Undersea Hyperb Med ; 44(6): 521-533, 2017.
Article in English | MEDLINE | ID: mdl-29281189

ABSTRACT

OBJECTIVE: Hyperoxia is known to influence cardiovascular and endothelial function, but it is unknown if there are differences between younger and older persons. The aim of this study was to monitor changes in myocardial diastolic function and flow-mediated dilatation (FMD) in younger and elderly volunteers, before and after exposure to relevant hyperbaric hyperoxia. METHODS: 51 male patients were separated into two groups for this study. Volunteers in Group 1 (n=28, mean age 26 ±6, "juniors") and Group 2 (n=23, mean age 53 ±9, "seniors") received standard HBO2 protocol (240kPa oxygen). Directly before and after hyperoxic exposure in a hyperbaric chamber we took blood samples (BNP, hs-troponin-t), assessed the FMD and echocardiographic parameters with focus on diastolic function. RESULTS: After hyperoxia we observed a high significant decrease in heart rate and systolic/diastolic FMD. Diastolic function varied in both groups: E/A ratio showed a statistically significant increase in Group 1 and remained unchanged in Group 2. E/e' ratio showed a slight but significant increase in Group 1, whereas e'/a' ratio increased in both groups. Deceleration time increased significantly in all volunteers. Isovolumetric relaxation time remained unchanged and ejection fraction showed a decrease only in Group 2. There were no changes in levels of BNP and hs-troponin-t in either group. CONCLUSION: Hyperoxia seems to influence endothelial function differently in juniors and seniors: FMD decreases more in seniors, possibly attributable to pre-existing reduced vascular compliance. Hyperoxia-induced bradycardia induced a more pronounced improvement in diastolic function in juniors. The ability of Group 1 to cope with hyperoxia-induced effects did not work in the same manner as with Group 2.


Subject(s)
Endothelium, Vascular/physiopathology , Hyperoxia/physiopathology , Adult , Aging/physiology , Arteries/physiopathology , Bradycardia/etiology , Bradycardia/physiopathology , Diastole/physiology , Echocardiography , Heart/physiopathology , Humans , Hyperbaric Oxygenation/adverse effects , Hyperoxia/complications , Male , Middle Aged , Vascular Resistance/physiology , Vasoconstriction/physiology , Young Adult
14.
Sci Rep ; 7(1): 9040, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28831114

ABSTRACT

This research was conducted to verify the structural and functional characteristics of mast cells in the electroacupuncture (EA) effects on bradycardia. First, we examined the mast cell density at PC 6, adjacent acupoint LU 7, and a non-acupoint. We tested the effects of EA at PC 6 on heart rate (HR) and blood pressure (BP) in rabbits with pituitrin-induced bradycardia. We also injected sodium cromolyn (Cro), a mast cell membrane stabilizer, at PC 6 30 min before EA to investigate if it affected the EA effects. The results showed that in both PC 6 and LU 7, the mast cell densities were higher than in the non-acupoint (P < 0.05). EA could induce mast cell degranulation at PC 6, which could be suppressed by sodium cromolyn (P < 0.05). EA improved HR, though the change was relatively small in the initial stage with a significant change at 35 min after modelling (P < 0.05). BP significantly improved at 10 min after the onset of pituitrin-induced bradycardia (P < 0.05). The EA effects on both HR and BP were suppressed by sodium cromolyn (P < 0.05). Therefore, we concluded that mast cells in the acupoint are important for the EA effects against pituitrin-induced bradycardia in rabbits.


Subject(s)
Acupuncture Points , Bradycardia/etiology , Bradycardia/physiopathology , Cell Degranulation/immunology , Mast Cells/immunology , Pituitary Hormones, Posterior/adverse effects , Animals , Blood Pressure , Bradycardia/diagnosis , Bradycardia/therapy , Cell Count , Disease Models, Animal , Electroacupuncture , Electrocardiography , Heart Rate , Male , Mast Cells/metabolism , Mast Cells/pathology , Rabbits
15.
Heart Rhythm ; 14(5): 694-699, 2017 05.
Article in English | MEDLINE | ID: mdl-28089877

ABSTRACT

BACKGROUND: Syncope is a common problem in the elderly, and a permanent pacemaker is a therapeutic option when a bradycardic etiology is revealed. However, the benefit of pacing when no association of symptoms to bradycardia has been shown is not clear, especially in the elderly. OBJECTIVE: The aim of this study was to evaluate the effect of pacing on syncope-free mortality in patients aged 80 years or older with unexplained syncope and "positive" invasive electrophysiologic testing (EPT). METHODS: This was an observational study. A positive EPT for the purposes of this study was defined by at least 1 of the following: a corrected sinus node recovery time of >525 ms, a basic HV interval of >55 ms, detection of infra-Hisian block, or appearance of second-degree atrioventricular block on atrial decremental pacing at a paced cycle length of >400 ms. RESULTS: Among the 2435 screened patients, 228 eligible patients were identified, 145 of whom were implanted with a pacemaker. Kaplan-Meier analysis determined that time to event (syncope or death) was 50.1 months (95% confidence interval 45.4-54.8 months) with a pacemaker vs 37.8 months (95% confidence interval 31.3-44.4 months) without a pacemaker (log-rank test, P = .001). The 4-year time-dependent estimate of the rate of syncope was 12% vs 44% (P < .001) and that of any-cause death was 41% vs 56% (P = .023), respectively. The multivariable odds ratio was 0.25 (95% confidence interval 0.15-0.40) after adjustment for potential confounders. CONCLUSION: In patients with unexplained syncope and signs of sinus node dysfunction or impaired atrioventricular conduction on invasive EPT, pacemaker implantation was independently associated with longer syncope-free survival. Significant differences were also shown in the individual components of the primary outcome measure (syncope and death from any cause).


Subject(s)
Bradycardia/therapy , Cardiac Pacing, Artificial , Heart Block/diagnosis , Heart Block/therapy , Pacemaker, Artificial , Syncope/prevention & control , Aged, 80 and over , Bradycardia/complications , Bradycardia/physiopathology , Electrophysiologic Techniques, Cardiac , Heart Block/physiopathology , Humans , Syncope/etiology , Syncope/mortality , Syncope/physiopathology
16.
Chin Med J (Engl) ; 130(2): 179-186, 2017 01 20.
Article in English | MEDLINE | ID: mdl-28091410

ABSTRACT

BACKGROUND: The molecular mechanisms of Shenxianshengmai (SXSM), a traditional Chinese medicine, on bradycardia have been incompletely understood. The study tried to investigate the gene expression profile and proteomics of bradycardia rabbits' hearts after SXSM treatment. METHODS: Twenty-four adult rabbits were randomly assigned in four groups: sham, model, model plus SXSM treatment, and sham plus SXSM treatment groups. Heart rate was recorded in all rabbits. Then, total RNA of atria and proteins of ventricle were isolated and quantified, respectively. Gene expression profiling was conducted by gene expression chip, and quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR) was performed to confirm the results of gene expression chip. We used isobaric tags for elative and absolute quantitation and Western blotting to identify altered proteins after SXSM treatment. RESULTS: There was a constant decrease in the mean heart rate (32%, from 238 ± 6 beats/min to 149 ± 12 beats/min) after six weeks in model compared with that in sham group. This effect was partially reversed by 4-week SXSM treatment. Complementary DNA microarray demonstrated that the increased acetylcholinesterase and reduced nicotinic receptor were take responsibility for the increased heart rate. In addition, proteins involved in calcium handling and signaling were affected by SXSM treatment. Real-time RT-PCR verified the results from gene chip. Results from proteomics demonstrated that SXSM enhanced oxidative phosphorylation and tricarboxylic acid (TCA) cycle in ventricular myocardium to improve ATP generation. CONCLUSIONS: Long-term SXSM stimulates sympathetic transmission by increasing the expression of acetylcholinesterase and reduces the expression of nicotinic receptor to increase heart rate. SXSM also restored the calcium handling genes and altered genes involved in signaling. In addition, SXSM improves the ATP supply of ventricular myocardium by increasing proteins involved in TCA cycle and oxidation-respiratory chain.


Subject(s)
Bradycardia/drug therapy , Drugs, Chinese Herbal/therapeutic use , Animals , Bradycardia/metabolism , Bradycardia/physiopathology , Heart Rate/drug effects , Proteomics , Rabbits , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction
17.
Epilepsy Behav ; 64(Pt A): 212-215, 2016 11.
Article in English | MEDLINE | ID: mdl-27750160

ABSTRACT

BACKGROUND: Seizures can lead to cardiac arrhythmias by a number of mechanisms including activation/inhibition of cortical autonomic centers, increase in vagal tone through activation of brainstem reflex centers, and respiratory failure. Ictal asystole (IA) is a potential mechanism underlying sudden unexpected death in epilepsy (SUDEP). We analyzed the clinical features of 5 patients who developed IA requiring pacemaker implantation. METHODS: Patients with ictal arrhythmias were identified from the video-telemetry and ambulatory EEG database at Greater Manchester Neurosciences Centre, as well as an independent epilepsy residential care facility. Only those who had IA requiring pacemaker implantation were included in the analysis. A total of 5 patients were identified. RESULTS: Of the 5 patients with IA, 4 were female. All 5 patients had focal epilepsy, and four had temporal lobe epilepsy. Ictal asystole occurred with focal seizures with impairment of awareness. Seizure onset was left-sided in 2 patients, right-sided in one, left-sided onset with switch of lateralization in one, and nonlateralized in one patient. Three patients had hippocampal sclerosis, one of whom had undergone epilepsy surgery, one had traumatic encephalomalacia of the temporal lobe, and one patient had no lesions detected on MRI. Interictal epileptiform activity was more pronounced during sleep in all patients. Asystole occurred in association with sleep-related seizures in 4 of 5 patients. CONCLUSIONS: Ictal asystole (IA) occurred in association with sleep-related seizures in 4 out of 5 cases, predominantly in patients with temporal lobe epilepsy. These findings may be of relevance to SUDEP.


Subject(s)
Arrhythmias, Cardiac/therapy , Bradycardia/therapy , Epilepsies, Partial/complications , Heart Arrest/therapy , Pacemaker, Artificial , Seizures/complications , Adult , Arrhythmias, Cardiac/complications , Arrhythmias, Cardiac/physiopathology , Bradycardia/complications , Bradycardia/physiopathology , Electrocardiography , Electroencephalography , Epilepsies, Partial/physiopathology , Female , Heart Arrest/complications , Heart Arrest/physiopathology , Humans , Male , Middle Aged , Plant Extracts , Seizures/physiopathology , Temporal Lobe/physiopathology , Treatment Outcome
18.
Naunyn Schmiedebergs Arch Pharmacol ; 389(10): 1073-80, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27405774

ABSTRACT

In several case reports, proarrhythmic effects of antipsychotic drugs have been reported. The aim of the present study was to investigate if application of risperidone or quetiapine has the potential to provoke polymorphic ventricular tachycardia in a sensitive model of proarrhythmia. In 24 isolated rabbit hearts, risperidone (5 and 10 µM, n = 12) or quetiapine (5 and 10 µM, n = 12) was infused after obtaining baseline data. Eight endocardial and epicardial monophasic action potentials and a simultaneously recorded 12-lead ECG showed a significant QT prolongation after application of risperidone as compared with baseline (5 µM: +29 ms, 10 µM: +35 ms, p < 0.01) accompanied by an increase of action potential duration. Administration of risperidone also significantly increased spatial dispersion of repolarization (5 µM: +16 ms, 4 µM: +19 ms; p < 0.05) as well as temporal dispersion of repolarization. Lowering of potassium concentration in bradycardic AV-blocked hearts provoked early afterdepolarizations (EADs) in 8 of 12 hearts and polymorphic ventricular tachycardia resembling torsade de pointes in 6 of 12 hearts (10 µM, 49 episodes). The results were compared with hearts treated with quetiapine (5 and 10 µM). Quetiapine led to an increase in QT interval (5 µM: +10 ms; 10 µM: +28 ms; p < 0.05) and a similar increase of APD90. However, treatment with quetiapine did not result in significant alterations of spatial and temporal dispersion of repolarization. No ventricular arrhythmias were observed in this group. In the present study, quetiapine demonstrated a safe electrophysiologic profile despite significant QT prolongation. In contrast, risperidone led to a more marked prolongation of myocardial repolarization combined with a more marked increase of dispersion of repolarization.


Subject(s)
Antipsychotic Agents/toxicity , Heart Rate/drug effects , Heart Ventricles/drug effects , Quetiapine Fumarate/toxicity , Risperidone/toxicity , Tachycardia, Ventricular/chemically induced , Action Potentials/drug effects , Animals , Atrioventricular Block/physiopathology , Bradycardia/physiopathology , Cardiac Pacing, Artificial , Electrocardiography , Electrophysiologic Techniques, Cardiac , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Isolated Heart Preparation , Potassium/metabolism , Rabbits , Risk Assessment , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Time Factors , Torsades de Pointes/chemically induced , Torsades de Pointes/metabolism , Torsades de Pointes/physiopathology
19.
J Physiol ; 594(20): 5869-5879, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27374078

ABSTRACT

Pacemaker activity of the sino-atrial node generates the heart rate. Disease of the sinus node and impairment of atrioventricular conduction induce an excessively low ventricular rate (bradycardia), which cannot meet the needs of the organism. Bradycardia accounts for about half of the total workload of clinical cardiologists. The 'sick sinus' syndrome (SSS) is characterized by sinus bradycardia and periods of intermittent atrial fibrillation. Several genetic or acquired risk factors or pathologies can lead to SSS. Implantation of an electronic pacemaker constitutes the only available therapy for SSS. The incidence of SSS is forecast to double over the next 50 years, with ageing of the general population thus urging the development of complementary or alternative therapeutic strategies. In recent years an increasing number of mutations affecting ion channels involved in sino-atrial automaticity have been reported to underlie inheritable SSS. L-type Cav 1.3 channels play a major role in the generation and regulation of sino-atrial pacemaker activity and atrioventricular conduction. Mutation in the CACNA1D gene encoding Cav 1.3 channels induces loss-of-function in channel activity and underlies the sino-atrial node dysfunction and deafness syndrome (SANDD). Mice lacking Cav 1.3 channels (Cav 1.3-/- ) fairly recapitulate SSS and constitute a precious model to test new therapeutic approaches to handle this disease. Work in our laboratory shows that targeting G protein-gated K+ (IKACh ) channels effectively rescues SSS of Cav 1.3-/- mice. This new concept of 'compensatory' ion channel targeting shines new light on the principles underlying the pacemaker mechanism and may open the way to new therapies for SSS.


Subject(s)
Calcium Channels, L-Type/metabolism , Channelopathies/metabolism , Heart Ventricles/metabolism , Animals , Bradycardia/genetics , Bradycardia/metabolism , Bradycardia/physiopathology , Calcium Channels, L-Type/genetics , Channelopathies/genetics , Channelopathies/physiopathology , Heart Rate/genetics , Heart Rate/physiology , Heart Ventricles/physiopathology , Humans , Mutation/genetics , Sick Sinus Syndrome/genetics , Sick Sinus Syndrome/metabolism , Sick Sinus Syndrome/physiopathology , Sinoatrial Node/metabolism , Sinoatrial Node/physiopathology
20.
Neuroscience ; 322: 464-78, 2016 May 13.
Article in English | MEDLINE | ID: mdl-26947128

ABSTRACT

This study tested the hypothesis that the hypothalamus participates in the decompensatory phase of hemorrhage by measuring Fos immunoreactivity and by inhibiting neuronal activity in selected hypothalamic nuclei with lidocaine or cobalt chloride. Previously, we reported that inactivation of the arcuate nucleus inhibited, but did not fully prevent, the fall in arterial pressure evoked by hypotensive hemorrhage. Here, we report that hemorrhage (2.2 ml/100g body weight over 20 min) induced Fos expression in a high percentage of cells in the paraventricular, supraoptic and arcuate nuclei of the hypothalamus as shown previously. Lower densities of Fos immunoreactive cells were also found in the medial preoptic area (mPOA), anterior hypothalamus, lateral hypothalamus (LH), dorsomedial hypothalamus, ventromedial hypothalamus (VMH) and posterior hypothalamus. Bilateral injection of lidocaine (2%; 0.1 µl or 0.3 µl) or cobalt chloride (5mM; 0.3 µl) into the tuberal portion of the LH immediately before hemorrhage was initiated reduced the magnitude of hemorrhagic hypotension and bradycardia significantly. Lidocaine injection into the VMH also attenuated the fall in arterial pressure and heart rate evoked by hemorrhage although inactivation of the mPOA or rostral LH was ineffective. These findings indicate that hemorrhage activates neurons throughout much of the hypothalamus and that a relatively broad area of the hypothalamus, extending from the arcuate nucleus laterally through the caudal VMH and tuberal LH, plays an important role in the decompensatory phase of hemorrhage.


Subject(s)
Hemorrhage/physiopathology , Hypothalamus/metabolism , Hypovolemia/physiopathology , Proto-Oncogene Proteins c-fos/metabolism , Animals , Bradycardia/pathology , Bradycardia/physiopathology , Cobalt/pharmacology , Disease Models, Animal , Hemorrhage/pathology , Hemostatics/pharmacology , Hypothalamus/pathology , Hypovolemia/pathology , Lidocaine/pharmacology , Male , Neurons/metabolism , Neurons/pathology , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL