Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.548
Filtrar
1.
PLoS One ; 19(9): e0309940, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39255304

RESUMEN

Given the limited capacity of intensive care units in many countries, it is crucial to identify reliable prognostic markers to optimize poisoning patients management and improve outcomes. This study aimed to assess the predictive value of three variables, namely the initial QTc interval (iQTc) measured within two hours of admission, the delayed QTc interval (dQTc) measured between 6 and 12 hours of entry, and the QTc interval trend over time (ΔQTc), for mortality in patients with undifferentiated poisoning. A retrospective case series was conducted on 70 patients with undifferentiated poisoning admitted to the intensive care unit (ICU) of Afzalipour Hospital between March 21, 2021, and March 20, 2023. The results of the multivariate analysis revealed that dQTc, base deficit, and creatinine were independently associated with mortality (P value < 0.001). The dQTc had the highest predictive ability, with an area under the curve (AUC) of 0.84, followed by ΔQTc with an AUC of 0.76, and iQTc with an AUC of 0.67. Additionally, the results of the Generalized Estimating Equation model with repeated measurements revealed a higher odds ratio for dQTc (OR, 6.33; 95% CI, 2.54-15.79) compared to iQTc (OR, 4.92; 95% CI, 1.71-14.17). The study concluded that monitoring the dQTc interval could provide valuable prognostic information in acute poisoning cases.


Asunto(s)
Síndrome de QT Prolongado , Intoxicación , Humanos , Masculino , Femenino , Pronóstico , Persona de Mediana Edad , Estudios Retrospectivos , Adulto , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/diagnóstico , Electrocardiografía , Unidades de Cuidados Intensivos , Anciano , Enfermedad Aguda
2.
BMC Anesthesiol ; 24(1): 307, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39232657

RESUMEN

BACKGROUND: Fentanyl is an opioid analgesic frequently used in the emergency department (ED) and is usually administered without knowing the QTC values of the patients or being monitored. However, the effect of fentanyl on QTC, prolongation or shortening, has not been elucidated. This study aimed to determine the effect of fentanyl on QTC. METHODS: This is a prospective observational study in the ED of a tertiary hospital on patients who received intravenous fentanyl for procedures other than intubation. ECG was performed before and at 1, 5, 15, 30, and 60 min after the initiation of fentanyl administration, and QTC value was calculated. Primary outcomes were QTC prolongation, defined as an increase in the QTC to ≥ 500 ms or any increase in QTC by ≥ 60 ms. RESULTS: The study included 109 patients. Of these, 60 patients were male, and the median age was 40. Compared with the baseline QTC value, statistically significant prolongation was detected at the 5th, 15th, 30th, and 60th minutes, with the maximum prolongation at 30 min, and the median was 13.08 ms. Most patients with QTC prolongation were female and over 40 years of age. Clinically, none of these patients developed malignant arrhythmias during the 60-minute monitored observation period. CONCLUSION: Fentanyl prolonged the QTC value statistically significantly. Although no patient developed malignant arrhythmia clinically, our results suggest that this QTC-prolonging effect should be considered when using fentanyl in patients at risk of torsades.


Asunto(s)
Analgésicos Opioides , Electrocardiografía , Servicio de Urgencia en Hospital , Fentanilo , Humanos , Fentanilo/administración & dosificación , Fentanilo/efectos adversos , Masculino , Femenino , Estudios Prospectivos , Adulto , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/administración & dosificación , Electrocardiografía/efectos de los fármacos , Persona de Mediana Edad , Síndrome de QT Prolongado/inducido químicamente , Anciano , Adulto Joven , Administración Intravenosa
3.
Nat Commun ; 15(1): 7507, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39209900

RESUMEN

Rare genetic diseases are typically studied in referral populations, resulting in underdiagnosis and biased assessment of penetrance and phenotype. To address this, we develop a generalizable method of genotype inference based on distant relatedness and deploy this to identify undiagnosed Type 5 Long QT Syndrome (LQT5) rare variant carriers in a non-referral population. We identify 9 LQT5 families referred to a single specialty clinic, each carrying p.Asp76Asn, the most common LQT5 variant. We uncover recent common ancestry and a single shared haplotype among probands. Application to a non-referral population of 69,819 BioVU biobank subjects identifies 22 additional subjects sharing this haplotype, which we confirm to carry p.Asp76Asn. Referral and non-referral carriers have prolonged QT interval corrected for heart rate (QTc) compared to controls, and, among carriers, the QTc polygenic score is independently associated with QTc prolongation. Thus, our innovative analysis of shared chromosomal segments identifies undiagnosed cases of genetic disease and refines the understanding of LQT5 penetrance and phenotype.


Asunto(s)
Bancos de Muestras Biológicas , Haplotipos , Síndrome de QT Prolongado , Humanos , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/diagnóstico , Femenino , Masculino , Adulto , Penetrancia , Persona de Mediana Edad , Fenotipo , Linaje , Predisposición Genética a la Enfermedad , Genotipo , Electrocardiografía
6.
PLoS One ; 19(8): e0308999, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39159176

RESUMEN

Heart failure (HF) with reduced ejection fraction (HFrEF) is a risk factor for drug-induced QT interval prolongation. It is unknown if HF with preserved ejection fraction (HFpEF) is also associated with an increased risk. Dofetilide and sotalol are potent QT interval-prolonging agents that are frequently used in patients with HFpEF, in whom atrial fibrillation is a common comorbidity. We tested the hypothesis that the risk of QT interval prolongation associated with dofetilide and sotalol is increased in patients with HFpEF. We conducted a retrospective cohort study conducted using electronic health records from the Indiana Network for Patient Care (January 31, 2010 -May 3, 2021). After removing patients with overlapping diagnoses of HFpEF and HFrEF, no diagnosis code, and absence of QT interval records, we identified patients taking dofetilide or sotalol among three groups: HFrEF (n = 138), HFpEF (n = 109), and no HF (n = 729). QT prolongation was defined as heart rate-corrected QT (QTc) > 500 ms during dofetilide/sotalol therapy. Unadjusted odds ratios (OR) for QT prolongation were determined by univariate analysis. Adjusted ORs were determined by generalized estimating equations (GEE) with logit link to account for an individual cluster with different times of hospitalization and covariates. QTc prolongation associated with dofetilide or sotalol occurred in 53.2%, 71.7% and 30.0% of patients with HFpEF, HFrEF, and patients with no HF, respectively. After adjusting for age, sex, race, serum potassium and magnesium concentrations, kidney function, concomitant drug therapy, and comorbid conditions, the adjusted odds of QTc prolongation were significantly higher in patients with HFpEF [OR = 1.98 (95% CI 1.17-3.33)], and in those with HFrEF [OR = 5.23, (3.15-8.67)], compared to those with no evidence of HF. The odds of QT prolongation among inpatients receiving dofetilide or sotalol were increased in patients with HFpEF and HFrEF compared to those who did not have HF.


Asunto(s)
Insuficiencia Cardíaca , Síndrome de QT Prolongado , Fenetilaminas , Sotalol , Volumen Sistólico , Sulfonamidas , Humanos , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/tratamiento farmacológico , Femenino , Masculino , Anciano , Fenetilaminas/efectos adversos , Sotalol/efectos adversos , Volumen Sistólico/efectos de los fármacos , Estudios Retrospectivos , Sulfonamidas/efectos adversos , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/fisiopatología , Síndrome de QT Prolongado/epidemiología , Persona de Mediana Edad , Anciano de 80 o más Años , Electrocardiografía , Antiarrítmicos/efectos adversos , Factores de Riesgo
7.
Medicine (Baltimore) ; 103(32): e39039, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39121326

RESUMEN

Amidst the COVID-19 pandemic, hydroxychloroquine (HCQ) was widely administered despite limited data on its safety and efficacy. This study assesses the acute and chronic impacts of HCQ on electrocardiography (ECG) parameters alongside the effects of azithromycin and levofloxacin coadministration in patients with COVID-19. A comprehensive analysis was conducted on 109 COVID-19 patients receiving HCQ, with or without Azithromycin and/or Levofloxacin, and 51 long-term HCQ-treated Sjogren's syndrome (SS) patients. ECG parameters, including QTc interval, were meticulously evaluated against a control group of 109 COVID-19 patients without HCQ treatment. HCQ monotherapy, in combination with Levofloxacin, significantly prolonged the QTc interval in COVID-19 patients compared to controls. Notably, the combination of HCQ and Azithromycin demonstrated a mitigated impact on QTc prolongation. Long-term HCQ use in SS patients did not significantly affect QTc intervals, illustrating a distinct safety profile from short-term use in COVID-19 treatment. HCQ's impact on QTc prolongation is influenced by therapeutic context, coadministered drugs, and patient demographics. The findings underscore the necessity of cautious HCQ use, particularly in acute settings like COVID-19, where monitoring and consideration of drug interactions and patient-specific factors are critical.


Asunto(s)
Azitromicina , Tratamiento Farmacológico de COVID-19 , Electrocardiografía , Hidroxicloroquina , Síndrome de QT Prolongado , Humanos , Hidroxicloroquina/efectos adversos , Hidroxicloroquina/uso terapéutico , Hidroxicloroquina/administración & dosificación , Electrocardiografía/efectos de los fármacos , Femenino , Masculino , Persona de Mediana Edad , Azitromicina/uso terapéutico , Azitromicina/efectos adversos , Azitromicina/administración & dosificación , Síndrome de QT Prolongado/inducido químicamente , Anciano , Síndrome de Sjögren/tratamiento farmacológico , Quimioterapia Combinada , Levofloxacino/uso terapéutico , Levofloxacino/administración & dosificación , Levofloxacino/efectos adversos , Adulto , SARS-CoV-2 , COVID-19
8.
Circ Res ; 135(7): 722-738, 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39166328

RESUMEN

BACKGROUND: The KCNQ1+KCNE1 (IKs) potassium channel plays a crucial role in cardiac adaptation to stress, in which ß-adrenergic stimulation phosphorylates the IKs channel through the cyclic adenosine monophosphate (cAMP)/PKA (protein kinase A) pathway. Phosphorylation increases the channel current and accelerates repolarization to adapt to an increased heart rate. Variants in KCNQ1 can cause long-QT syndrome type 1 (LQT1), and those with defective cAMP effects predispose patients to the highest risk of cardiac arrest and sudden death. However, the molecular connection between IKs channel phosphorylation and channel function, as well as why high-risk LQT1 mutations lose cAMP sensitivity, remain unclear. METHODS: Regular patch clamp and voltage clamp fluorometry techniques were utilized to record pore opening and voltage sensor movement of wild-type and mutant KCNQ1/IKs channels. The clinical phenotypic penetrance of each LQT1 mutation was analyzed as a metric for assessing their clinical risk. The patient-specific-induced pluripotent stem-cell model was used to test mechanistic findings in physiological conditions. RESULTS: By systematically elucidating mechanisms of a series of LQT1 variants that lack cAMP sensitivity, we identified molecular determinants of IKs channel regulation by phosphorylation. These key residues are distributed across the N-terminus of KCNQ1 extending to the central pore region of IKs. We refer to this pattern as the IKs channel PKA phosphorylation axis. Next, by examining LQT1 variants from clinical databases containing 10 579 LQT1 carriers, we found that the distribution of the most high-penetrance LQT1 variants extends across the IKs channel PKA phosphorylation axis, demonstrating its clinical relevance. Furthermore, we found that a small molecule, ML277, which binds at the center of the phosphorylation axis, rescues the defective cAMP effects of multiple high-risk LQT1 variants. This finding was then tested in high-risk patient-specific induced pluripotent stem cell-derived cardiomyocytes, where ML277 remarkably alleviates the beating abnormalities. CONCLUSIONS: Our findings not only elucidate the molecular mechanism of PKA-dependent IKs channel phosphorylation but also provide an effective antiarrhythmic strategy for patients with high-risk LQT1 variants.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico , Células Madre Pluripotentes Inducidas , Canal de Potasio KCNQ1 , Humanos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosforilación , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Síndrome de Romano-Ward/genética , Síndrome de Romano-Ward/metabolismo , AMP Cíclico/metabolismo , Miocitos Cardíacos/metabolismo , Mutación , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/metabolismo , Células HEK293 , Canales de Potasio con Entrada de Voltaje
10.
J Clin Anesth ; 98: 111574, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39121785

RESUMEN

STUDY OBJECTIVE: Although a prolonged heart rate-corrected QT interval (QTcI) is associated with an increased risk of mortality in the general population, its prognostic value in surgical patients remains unclear. We aimed to examine whether preoperative QTcI prolongation predicts short-term postoperative outcomes in elderly patients undergoing noncardiac surgery. DESIGN: The study was a retrospective analysis using the TriNetX network database. SETTING: Operating room. INTERVENTION: Assessment and categorization of preoperative QTcI. PATIENTS: Data of patients aged ≥65 years who underwent non-cardiac surgery between 2010 and 2023 were analyzed. MEASUREMENTS: Patients were categorized into four groups based on preoperative QTcI: long (500-600 ms), borderline (460-500 ms), high-normal (420-460 ms) and control (370-420 ms) groups. The groups were compared using a propensity score-matched analysis. The primary outcome was the all-cause 90-day mortality risk. The secondary outcomes included 90-day risks of postoperative new-onset atrial fibrillation (Af), ventricular arrhythmias (VAs), emergency visits, hospital readmissions, and pneumonia. RESULTS: In total, data on 519,929 patients were collected in this study. Pairwise comparisons showed that all QTcI prolongation groups demonstrated a heightened incidence of postoperative mortality, arrhythmias, and other complications compared to the control group. Patients with a long QTcI had a 3-fold higher risk of mortality (hazard ratio [HR] = 3.124, p < 0.001), Af (HR = 3.059, p < 0.001), and VAs (HR = 3.617, p < 0.001) than controls. The risks of emergency visits (HR = 1.287, p < 0.001), hospital readmissions (HR = 1.591, p < 0.001), and pneumonia (HR = 1.672, p < 0.001) were also higher in the long QTcI group than in the control group. A dose-dependent response was evident between QTcI and mortality as well as arrhythmia risk. CONCLUSION: Preoperative QTcI screening effectively risk-stratifies elderly surgical patients, with a QTcI≥500 ms being strongly predictive of short-term postoperative mortality and other complications. Incorporating QTcI assessment into the preoperative evaluation may guide perioperative monitoring and management.


Asunto(s)
Electrocardiografía , Síndrome de QT Prolongado , Complicaciones Posoperatorias , Humanos , Estudios Retrospectivos , Femenino , Anciano , Masculino , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/mortalidad , Síndrome de QT Prolongado/epidemiología , Anciano de 80 o más Años , Readmisión del Paciente/estadística & datos numéricos , Frecuencia Cardíaca , Periodo Preoperatorio , Arritmias Cardíacas/etiología , Arritmias Cardíacas/epidemiología , Arritmias Cardíacas/diagnóstico , Factores de Riesgo , Medición de Riesgo/métodos , Medición de Riesgo/estadística & datos numéricos , Fibrilación Atrial/diagnóstico , Incidencia , Pronóstico
11.
Biochim Biophys Acta Biomembr ; 1866(7): 184377, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39103068

RESUMEN

KCNQ1, also known as Kv7.1, is a voltage gated potassium channel that associates with the KCNE protein family. Mutations in this protein has been found to cause a variety of diseases including Long QT syndrome, a type of cardiac arrhythmia where the QT interval observed on an electrocardiogram is longer than normal. This condition is often aggravated during strenuous exercise and can cause fainting spells or sudden death. KCNE1 is an ancillary protein that interacts with KCNQ1 in the membrane at varying molar ratios. This interaction allows for the flow of potassium ions to be modulated to facilitate repolarization of the heart. The interaction between these two proteins has been studied previously with cysteine crosslinking and electrophysiology. In this study, electron paramagnetic resonance (EPR) spectroscopy line shape analysis in tandem with site directed spin labeling (SDSL) was used to observe changes in side chain dynamics as KCNE1 interacts with KCNQ1. KCNE1 was labeled at different sites that were found to interact with KCNQ1 based on previous literature, along with sites outside of that range as a control. Once labeled KCNE1 was incorporated into vesicles, KCNQ1 (helices S1-S6) was titrated into the vesicles. The line shape differences observed upon addition of KCNQ1 are indicative of an interaction between the two proteins. This method provides a first look at the interactions between KCNE1 and KCNQ1 from a dynamics perspective using the full transmembrane portion of KCNQ1.


Asunto(s)
Canal de Potasio KCNQ1 , Canales de Potasio con Entrada de Voltaje , Canales de Potasio con Entrada de Voltaje/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/química , Canal de Potasio KCNQ1/metabolismo , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/química , Espectroscopía de Resonancia por Spin del Electrón/métodos , Unión Proteica , Humanos , Animales , Síndrome de QT Prolongado/metabolismo , Síndrome de QT Prolongado/genética
12.
J Biol Chem ; 300(8): 107526, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38960041

RESUMEN

The human ether-a-go-go-related gene (hERG) encodes the Kv11.1 (or hERG) channel that conducts the rapidly activating delayed rectifier potassium current (IKr). Naturally occurring mutations in hERG impair the channel function and cause long QT syndrome type 2. Many missense hERG mutations lead to a lack of channel expression on the cell surface, representing a major mechanism for the loss-of-function of mutant channels. While it is generally thought that a trafficking defect underlies the lack of channel expression on the cell surface, in the present study, we demonstrate that the trafficking defective mutant hERG G601S can reach the plasma membrane but is unstable and quickly degrades, which is akin to WT hERG channels under low K+ conditions. We previously showed that serine (S) residue at 624 in the innermost position of the selectivity filter of hERG is involved in hERG membrane stability such that substitution of serine 624 with threonine (S624T) enhances hERG stability and renders hERG insensitive to low K+ culture. Here, we report that the intragenic addition of S624T substitution to trafficking defective hERG mutants G601S, N470D, and P596R led to a complete rescue of the function of these otherwise loss-of-function mutant channels to a level similar to the WT channel, representing the most effective rescue means for the function of mutant hERG channels. These findings not only provide novel insights into hERG mutation-mediated channel dysfunction but also point to the critical role of S624 in hERG stability on the plasma membrane.


Asunto(s)
Membrana Celular , Canal de Potasio ERG1 , Síndrome de QT Prolongado , Humanos , Síndrome de QT Prolongado/metabolismo , Síndrome de QT Prolongado/genética , Membrana Celular/metabolismo , Canal de Potasio ERG1/metabolismo , Canal de Potasio ERG1/genética , Células HEK293 , Mutación Missense , Estabilidad Proteica , Canales de Potasio Éter-A-Go-Go/metabolismo , Canales de Potasio Éter-A-Go-Go/genética , Transporte de Proteínas , Sustitución de Aminoácidos , Animales
13.
Antimicrob Agents Chemother ; 68(8): e0046424, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38953364

RESUMEN

Islatravir is a deoxynucleoside analog being developed for the treatment of HIV-1 infection. Clinical studies are being conducted to evaluate islatravir, administered in combination with other antiretroviral therapies, at doses of 0.25 mg once daily and 2 mg once weekly. In multiple previous clinical studies, islatravir was generally well tolerated, with no clear trend in cardiac adverse events. A trial was conducted to evaluate the effect of islatravir on cardiac repolarization. A randomized, double-blind, active- and placebo-controlled phase 1 trial was conducted, in which a single dose of islatravir 0.75 mg, islatravir 240 mg (supratherapeutic dose), moxifloxacin 400 mg (active control), or placebo was administered. Continuous 12-lead electrocardiogram monitoring was performed before dosing through 24 hours after dosing. QT interval measurements were collected, and safety and pharmacokinetics were evaluated. Sixty-three participants were enrolled, and 59 completed the study. Fridericia's QT correction for heart rate was inadequate; therefore, a population-specific correction was applied (QTcP). The placebo-corrected change from baseline in QTcP (ΔΔQTcP) interval at the observed geometric mean maximum plasma concentration associated with islatravir 0.75 mg and islatravir 240 mg was <10 ms at all time points. Assay sensitivity was confirmed because the use of moxifloxacin 400 mg led to a ΔΔQTcP >10 ms. The pharmacokinetic profile of islatravir was consistent with that of previous studies, and islatravir was generally well tolerated. Results from the current trial suggest that single doses of islatravir as high as 240 mg do not lead to QTc interval prolongation.


Asunto(s)
Electrocardiografía , Fluoroquinolonas , Moxifloxacino , Humanos , Adulto , Masculino , Electrocardiografía/efectos de los fármacos , Método Doble Ciego , Femenino , Persona de Mediana Edad , Fluoroquinolonas/efectos adversos , Fluoroquinolonas/farmacocinética , Moxifloxacino/efectos adversos , Moxifloxacino/farmacocinética , Frecuencia Cardíaca/efectos de los fármacos , Síndrome de QT Prolongado/inducido químicamente , Adulto Joven , Fármacos Anti-VIH/farmacocinética , Fármacos Anti-VIH/efectos adversos , Fármacos Anti-VIH/uso terapéutico , Compuestos Aza/efectos adversos , Compuestos Aza/farmacocinética , Desoxiadenosinas
14.
Circulation ; 150(7): 516-530, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39051104

RESUMEN

BACKGROUND: Whether vigorous exercise increases risk of ventricular arrhythmias for individuals diagnosed and treated for congenital long QT syndrome (LQTS) remains unknown. METHODS: The National Institutes of Health-funded LIVE-LQTS study (Lifestyle and Exercise in the Long QT Syndrome) prospectively enrolled individuals 8 to 60 years of age with phenotypic and/or genotypic LQTS from 37 sites in 5 countries from May 2015 to February 2019. Participants (or parents) answered physical activity and clinical events surveys every 6 months for 3 years with follow-up completed in February 2022. Vigorous exercise was defined as ≥6 metabolic equivalents for >60 hours per year. A blinded Clinical Events Committee adjudicated the composite end point of sudden death, sudden cardiac arrest, ventricular arrhythmia treated by an implantable cardioverter defibrillator, and likely arrhythmic syncope. A National Death Index search ascertained vital status for those with incomplete follow-up. A noninferiority hypothesis (boundary of 1.5) between vigorous exercisers and others was tested with multivariable Cox regression analysis. RESULTS: Among the 1413 participants (13% <18 years of age, 35% 18-25 years of age, 67% female, 25% with implantable cardioverter defibrillators, 90% genotype positive, 49% with LQT1, 91% were treated with beta-blockers, left cardiac sympathetic denervation, and/or implantable cardioverter defibrillator), 52% participated in vigorous exercise (55% of these competitively). Thirty-seven individuals experienced the composite end point (including one sudden cardiac arrest and one sudden death in the nonvigorous group, one sudden cardiac arrest in the vigorous group) with overall event rates at 3 years of 2.6% in the vigorous and 2.7% in the nonvigorous exercise groups. The unadjusted hazard ratio for experience of events for the vigorous group compared with the nonvigorous group was 0.97 (90% CI, 0.57-1.67), with an adjusted hazard ratio of 1.17 (90% CI, 0.67-2.04). The upper 95% one-sided confidence level extended beyond the 1.5 boundary. Neither vigorous or nonvigorous exercise was found to be superior in any group or subgroup. CONCLUSIONS: Among individuals diagnosed with phenotypic and/or genotypic LQTS who were risk assessed and treated in experienced centers, LQTS-associated cardiac event rates were low and similar between those exercising vigorously and those not exercising vigorously. Consistent with the low event rate, CIs are wide, and noninferiority was not demonstrated. These data further inform shared decision-making discussions between patient and physician about exercise and competitive sports participation. REGISTRATION: URL: https://www.clinicaltrials.gov; Unique identifier: NCT02549664.


Asunto(s)
Ejercicio Físico , Síndrome de QT Prolongado , Humanos , Síndrome de QT Prolongado/terapia , Síndrome de QT Prolongado/congénito , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/fisiopatología , Síndrome de QT Prolongado/mortalidad , Femenino , Masculino , Adolescente , Niño , Estudios Prospectivos , Adulto , Persona de Mediana Edad , Adulto Joven , Muerte Súbita Cardíaca/prevención & control , Muerte Súbita Cardíaca/epidemiología , Factores de Riesgo
15.
Circ Arrhythm Electrophysiol ; 17(8): e012036, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39069900

RESUMEN

BACKGROUND: CaM (calmodulin)-mediated long-QT syndrome is a genetic arrhythmia disorder (calmodulinopathies) characterized by a high prevalence of life-threatening ventricular arrhythmias occurring early in life. Three distinct genes (CALM1, CALM2, and CALM3) encode for the identical CaM protein. Conventional pharmacotherapies fail to adequately protect against potentially lethal cardiac events in patients with calmodulinopathy. METHODS: Five custom-designed CALM1-, CALM2-, and CALM3-targeting short hairpin RNAs (shRNAs) were tested for knockdown (KD) efficiency using TSA201 cells and reverse transcription-quantitative polymerase chain reaction. A dual-component suppression and replacement (SupRep) CALM gene therapy (CALM-SupRep) was created by cloning into a single construct CALM1-, CALM2-, and CALM3-specific shRNAs that produce KD (suppression) of each respective gene and a shRNA-immune CALM1 cDNA (replacement). CALM1-F142L, CALM2-D130G, and CALM3-D130G induced pluripotent stem cell-derived CMs were generated from patients with CaM-mediated long-QT syndrome. A voltage-sensing dye was used to measure action potential duration at 90% repolarization (APD90). RESULTS: Following shRNA KD efficiency testing, a candidate shRNA was identified for CALM1 (86% KD), CALM2 (71% KD), and CALM3 (94% KD). The APD90 was significantly prolonged in CALM2-D130G (647±9 ms) compared with CALM2-WT (359±12 ms; P<0.0001). Transfection with CALM-SupRep shortened the average APD90 of CALM2-D130G to 457±19 ms (66% attenuation; P<0.0001). Additionally, transfection with CALM-SupRep shortened the APD90 of CALM1-F142L (665±9 to 410±15 ms; P<0.0001) and CALM3-D130G (978±81 to 446±6 ms; P<0.001). CONCLUSIONS: We provide the first proof-of-principle suppression-replacement gene therapy for CaM-mediated long-QT syndrome. The CALM-SupRep gene therapy shortened the pathologically prolonged APD90 in CALM1-, CALM2-, and CALM3-variant CaM-mediated long-QT syndrome induced pluripotent stem cell-derived CM lines. The single CALM-SupRep construct may be able to treat all calmodulinopathies, regardless of which of the 3 CaM-encoding genes are affected.


Asunto(s)
Calmodulina , Terapia Genética , Síndrome de QT Prolongado , Humanos , Calmodulina/genética , Calmodulina/metabolismo , Terapia Genética/métodos , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/terapia , Síndrome de QT Prolongado/fisiopatología , Síndrome de QT Prolongado/metabolismo , Síndrome de QT Prolongado/diagnóstico , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Potenciales de Acción , Predisposición Genética a la Enfermedad , Mutación , Interferencia de ARN , Frecuencia Cardíaca/genética
17.
Stem Cell Res ; 80: 103513, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39079396

RESUMEN

Timothy syndrome, an extremely rare disease, is closely associated with a mutation in CACNA1C gene, which encodes the cardiac L-type voltage-gated calcium channel (Cav1.2). In this study, we generated a human induced pluripotent stem cell (iPSC) line from a Timothy syndrome infant carrying heterozygous CACNA1C mutation (transcript variant NM_000719.7c.1216G>A: p.G406R). The generated iPSC line showed typical stem cell morphology, positively expressed pluripotency and proliferation markers, normal karyotype, and trilineage differentiation potential. Therefore, this patient-specific iPSC can be of great significance in investigating the mechanisms underlying Timothy syndrome, and hence establishing effective intervention strategies.


Asunto(s)
Trastorno Autístico , Canales de Calcio Tipo L , Heterocigoto , Células Madre Pluripotentes Inducidas , Síndrome de QT Prolongado , Sindactilia , Humanos , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Sindactilia/genética , Sindactilia/patología , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/patología , Síndrome de QT Prolongado/metabolismo , Trastorno Autístico/genética , Trastorno Autístico/patología , Mutación , Línea Celular , Diferenciación Celular , Lactante
18.
Am J Cardiovasc Drugs ; 24(5): 685-691, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38976171

RESUMEN

BACKGROUND: Heart failure (HF) is a pervasive global health concern, with acute decompensated heart failure (ADHF) contributing significantly to morbidity and mortality. Medications used in patients with HF may exacerbate HF or prolong the QT interval, posing additional risks. OBJECTIVE: The objective is to assess the prevalence and utilization patterns of medications known to cause or exacerbate HF and prolong the QT interval among patients with ADHF. Understanding these patterns is crucial for optimizing patient care and minimizing potential risks. METHODS: A retrospective chart review was conducted at Huntsville Hospital, Huntsville, USA, covering 602 patients with ADHF over a 40-month period. Inclusion criteria involved age ≥ 18 years, a history of HF, and ADHF admission. The 2016 American Heart Association Scientific Statement was used to identify drugs that may cause or exacerbate HF and those that could prolong the QT interval RESULTS: Among the 602 patients, 57.3% received medications causing or exacerbating HF, notably albuterol (34.9%) and diabetes medications (20.4%), primarily metformin, followed by urologic agents (14.3%), mostly tamsulosin, and nonsteroidal anti-inflammatory drugs (NSAIDs) (6.1%). Moreover, 82.9% were on medications prolonging the QT interval, with loop diuretics, amiodarone, ondansetron, and famotidine most prevalent. Furthermore, 42.1% of the patients received more than two concomitant medications that prolong the QT interval, which can further exacerbate the risk of torsades de pointes. CONCLUSION: This study underscores the high prevalence of HF-causing or HF-exacerbating medications and QT-prolonging drugs in patients with ADHF. Healthcare professionals must be cognizant of these patterns, advocating for safer prescribing practices to optimize patient outcomes and reduce the burden of HF-related hospitalizations.


Asunto(s)
Insuficiencia Cardíaca , Hospitalización , Humanos , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/epidemiología , Estudios Retrospectivos , Masculino , Femenino , Anciano , Hospitalización/estadística & datos numéricos , Persona de Mediana Edad , Anciano de 80 o más Años , Enfermedad Aguda , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/epidemiología
19.
Stem Cell Res ; 79: 103496, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39018827

RESUMEN

The KCNQ1 gene encodes a voltage-gated potassium channel required for cardiac action potentials. Mutations in this gene have been associated with hereditary long QT syndrome 1, Jervell and Lange-Nielsen syndromes, and familial atrial fibrillation. The NM_000218.3(KCNQ1): c.604 + 2T > C mutation has been categorized as the causative variant leading to LQT1. In this study, we generated a KCNQ1 (c.644 + 2T > C) mutation human embryonic stem cell line WAe009-A-1L based on CRISPR base editing system. WAe009-A-1L cell has the potential to differentiate cardiomyocytes and would be used as an in vitro disease model for mechanism exploration and drug screening.


Asunto(s)
Edición Génica , Células Madre Embrionarias Humanas , Canal de Potasio KCNQ1 , Mutación , Humanos , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/citología , Edición Génica/métodos , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/metabolismo , Línea Celular , Sistemas CRISPR-Cas , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/citología , Diferenciación Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética
20.
Cardiovasc Toxicol ; 24(10): 1053-1066, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38954228

RESUMEN

The studies regarding prevalence, outcomes, and predictors of prolonged corrected QT (QTc) among COVID-19 patients not on QTc-prolonging medication are not available in the literature. In this retrospective cohort study, the QTc of 295 hospital-admitted COVID-19 patients was analyzed and its association with in-hospital mortality was determined. The QTc was prolonged in 14.6% (43/295) of the study population. Prolonged QTc was seen in patients with older age (P = 0.018), coronary artery disease (P = 0.001), congestive heart failure (P = 0.042), elevated N-terminal-pro-B-type natriuretic peptide (NT-ProBNP) (P < 0.0001), and on remdesivir (P = 0.046). No episode of torsades de pointes arrhythmia or any arrhythmic death was observed among patients with prolonged QTc. The mortality was significantly high in patients with prolonged QTc (P = 0.003). The multivariate logistic regression analysis showed coronary artery disease (odds ratio (OR): 4.153, 95% CI 1.37-14.86; P = 0.013), and NT-ProBNP (ng/L) (OR: 1.000, 95% CI 1.000-1.000; P = 0.007) as predictors of prolonged QTc. The prolonged QTc was associated with the worst in-hospital survival (p by log-rank 0.001). A significant independent association was observed between prolonged QTc and in-hospital mortality in multivariate cox-regression analysis (adjusted hazard ratio: 3.861; (95% CI 1.719-6.523), P < 0.0001). QTc was found to be a marker of underlying comorbidities among COVID-19 patients. Prolonged QTc in hospitalized COVID-19 patients was independently associated with in-hospital mortality.


Asunto(s)
COVID-19 , Mortalidad Hospitalaria , Síndrome de QT Prolongado , Humanos , Masculino , Femenino , COVID-19/mortalidad , COVID-19/diagnóstico , COVID-19/epidemiología , COVID-19/complicaciones , Estudios Retrospectivos , Anciano , Persona de Mediana Edad , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/epidemiología , Síndrome de QT Prolongado/mortalidad , Síndrome de QT Prolongado/fisiopatología , Prevalencia , Factores de Riesgo , Hidroxicloroquina/uso terapéutico , Hidroxicloroquina/efectos adversos , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19 , Electrocardiografía , Hospitalización , Medición de Riesgo , Anciano de 80 o más Años , Frecuencia Cardíaca
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA