Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 137(9): 1208-1218, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33181835

RESUMEN

Previous reports indicate that IL18 is a novel candidate gene for diastolic dysfunction in sickle cell disease (SCD)-related cardiomyopathy. We hypothesize that interleukin-18 (IL-18) mediates the development of cardiomyopathy and ventricular tachycardia (VT) in SCD. Compared with control mice, a humanized mouse model of SCD exhibited increased cardiac fibrosis, prolonged duration of action potential, higher VT inducibility in vivo, higher cardiac NF-κB phosphorylation, and higher circulating IL-18 levels, as well as reduced voltage-gated potassium channel expression, which translates to reduced transient outward potassium current (Ito) in isolated cardiomyocytes. Administering IL-18 to isolated mouse hearts resulted in VT originating from the right ventricle and further reduced Ito in SCD mouse cardiomyocytes. Sustained IL-18 inhibition via IL-18-binding protein resulted in decreased cardiac fibrosis and NF-κB phosphorylation, improved diastolic function, normalized electrical remodeling, and attenuated IL-18-mediated VT in SCD mice. Patients with SCD and either myocardial fibrosis or increased QTc displayed greater IL18 gene expression in peripheral blood mononuclear cells (PBMCs), and QTc was strongly correlated with plasma IL-18 levels. PBMC-derived IL18 gene expression was increased in patients who did not survive compared with those who did. IL-18 is a mediator of sickle cell cardiomyopathy and VT in mice and a novel therapeutic target in patients at risk for sudden death.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Cardiomiopatías/etiología , Interleucina-18/sangre , Taquicardia Ventricular/etiología , Adulto , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/fisiopatología , Animales , Arritmias Cardíacas/sangre , Arritmias Cardíacas/etiología , Arritmias Cardíacas/fisiopatología , Cardiomiopatías/sangre , Cardiomiopatías/fisiopatología , Humanos , Interleucina-18/análisis , Masculino , Ratones , Taquicardia Ventricular/sangre , Taquicardia Ventricular/fisiopatología , Adulto Joven
2.
Am J Otolaryngol ; 44(2): 103732, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36682146

RESUMEN

PURPOSE: Second medical opinions (SMO) can improve patient outcomes and change medical decision-making. The purpose was to determine the concordance of initial management of thyroid nodules for patients seeking SMO to established management guidelines. MATERIALS AND METHODS: Cases of patients consulting a single provider via telemedicine for SMO on the workup and management of thyroid nodule(s) were reviewed from September 2011 to February 2022. The primary outcome was the overall rate of adherence to 2015 ATA guidelines (correct/incorrect) and complete agreement (yes/no) between SMO and initial treatment team. RESULTS: Most sought a second opinion for treatment options. Only 14 (29.2 %) cases had followed all the guidelines correctly. Living in North America compared to Asia (10/18 vs. 4/25, p = 0.004) and consulting endocrinology (11/21 vs. 3/26, p = 0.004) was associated with correct following of all guidelines. The most common violations of the guidelines were a lack of Bethesda scoring in pathology reports (31.8 %) and inappropriate initial FNA (25.5 %). The SMO was in complete agreement with the initial treatment recommendation in 31 cases (64.6 %), in partial agreement in 12 cases (25 %), and in disagreement in 5 cases (10.4 %). CONCLUSIONS: In our study, adherence to guidelines was low. However, the SMO agreed with the workup and management of most patients, as most of this discordance with guidelines did not affect the overall treatment. The virtual second opinion consult was valuable in addressing patient-specific concerns, explaining additional treatment options, and, in a few cases, recommending against inappropriate surgical intervention.


Asunto(s)
Endocrinología , Neoplasias de la Tiroides , Nódulo Tiroideo , Humanos , Nódulo Tiroideo/diagnóstico , Nódulo Tiroideo/terapia , Nódulo Tiroideo/patología , Neoplasias de la Tiroides/patología , Estudios Retrospectivos , Ultrasonografía
3.
Exp Brain Res ; 240(11): 2965-2979, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36173425

RESUMEN

Previous history of activity and learning modulates synaptic plasticity and can lead to saturation of synaptic connections. According to the synaptic homeostasis hypothesis, neural oscillations during slow-wave sleep play an important role in restoring plasticity within a functional range. However, it is not known whether slow-wave oscillations-without the concomitant requirement of sleep-play a causal role in human synaptic homeostasis. Here, we aimed to answer this question using transcranial alternating current stimulation (tACS) to induce slow-oscillatory activity in awake human participants. tACS was interleaved between two plasticity-inducing interventions: motor learning, and paired associative stimulation (PAS). The hypothesis tested was that slow-oscillatory tACS would prevent homeostatic interference between motor learning and PAS, and facilitate plasticity from these successive interventions. Thirty-six participants received sham and active fronto-motor tACS in two separate sessions, along with electroencephalography (EEG) recordings, while a further 38 participants received tACS through a control montage. Motor evoked potentials (MEPs) were recorded throughout the session to quantify plasticity changes after the different interventions, and the data were analysed with Bayesian statistics. As expected, there was converging evidence that motor training led to excitatory plasticity. Importantly, we found moderate evidence against an effect of active tACS in restoring PAS plasticity, and no evidence of lasting entrainment of slow oscillations in the EEG. This suggests that, under the conditions tested here, slow-oscillatory tACS does not modulate synaptic homeostasis in the motor system of awake humans.


Asunto(s)
Corteza Motora , Estimulación Transcraneal de Corriente Directa , Humanos , Corteza Motora/fisiología , Teorema de Bayes , Potenciales Evocados Motores/fisiología , Plasticidad Neuronal/fisiología
4.
Cleft Palate Craniofac J ; 59(11): 1371-1376, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-34514869

RESUMEN

OBJECTIVE: To study the efficacy and safety profile of ketorolac in cleft palate surgery. DESIGN: Retrospective analysis of patients who underwent primary cleft palate surgery and received either postoperative ketorolac or opioids. SETTING: Tertiary care children's hospital. PATIENTS, PARTICIPANTS: Eighty-nine patients enrolled who were all younger than 36 months of age, not dependent on a gastrostomy tube, with no history of bleeding disorders, and had undergone their primary cleft palate procedure by one specific surgeon between January 2010 and June 2019. INTERVENTIONS: n/a. MAIN OUTCOME MEASURE: Morphine equivalent dose (MED), Face, Legs, Activity, Cry, Consolability (FLACC) score, length of stay (LOS), total oral intake (mL), total oral intake/LOS, and postoperative adverse events between ketorolac and no ketorolac groups. RESULTS: MED, FLACC score, and LOS were significantly lower in the ketorolac group compared to the no ketorolac group. One patient in the ketorolac group had a bleeding event. CONCLUSIONS: Use of ketorolac significantly decreased narcotic usage and pain scores as reported by the FLACC score. Moreover, postoperative bleeding was rare in both ketorolac and no ketorolac groups.


Asunto(s)
Fisura del Paladar , Ketorolaco , Analgésicos Opioides/uso terapéutico , Antiinflamatorios no Esteroideos/efectos adversos , Antiinflamatorios no Esteroideos/uso terapéutico , Niño , Fisura del Paladar/inducido químicamente , Fisura del Paladar/cirugía , Humanos , Ketorolaco/efectos adversos , Ketorolaco/uso terapéutico , Morfina , Manejo del Dolor , Dolor Postoperatorio/tratamiento farmacológico , Estudios Retrospectivos
5.
J Biol Chem ; 295(52): 18148-18159, 2020 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-33093176

RESUMEN

The QT interval is a recording of cardiac electrical activity. Previous genome-wide association studies identified genetic variants that modify the QT interval upstream of LITAF (lipopolysaccharide-induced tumor necrosis factor-α factor), a protein encoding a regulator of endosomal trafficking. However, it was not clear how LITAF might impact cardiac excitation. We investigated the effect of LITAF on the voltage-gated sodium channel Nav1.5, which is critical for cardiac depolarization. We show that overexpressed LITAF resulted in a significant increase in the density of Nav1.5-generated voltage-gated sodium current INa and Nav1.5 surface protein levels in rabbit cardiomyocytes and in HEK cells stably expressing Nav1.5. Proximity ligation assays showed co-localization of endogenous LITAF and Nav1.5 in cardiomyocytes, whereas co-immunoprecipitations confirmed they are in the same complex when overexpressed in HEK cells. In vitro data suggest that LITAF interacts with the ubiquitin ligase NEDD4-2, a regulator of Nav1.5. LITAF overexpression down-regulated NEDD4-2 in cardiomyocytes and HEK cells. In HEK cells, LITAF increased ubiquitination and proteasomal degradation of co-expressed NEDD4-2 and significantly blunted the negative effect of NEDD4-2 on INa We conclude that LITAF controls cardiac excitability by promoting degradation of NEDD4-2, which is essential for removal of surface Nav1.5. LITAF-knockout zebrafish showed increased variation in and a nonsignificant 15% prolongation of action potential duration. Computer simulations using a rabbit-cardiomyocyte model demonstrated that changes in Ca2+ and Na+ homeostasis are responsible for the surprisingly modest action potential duration shortening. These computational data thus corroborate findings from several genome-wide association studies that associated LITAF with QT interval variation.


Asunto(s)
Endosomas/metabolismo , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo , Potenciales de Acción , Animales , Estudio de Asociación del Genoma Completo , Humanos , Miocitos Cardíacos/citología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Ubiquitina-Proteína Ligasas Nedd4/genética , Proteínas Nucleares/genética , Unión Proteica , Transporte de Proteínas , Conejos , Factores de Transcripción/genética , Ubiquitinación , Pez Cebra
6.
J Biol Chem ; 294(1): 351-360, 2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30401747

RESUMEN

The QT interval is an important diagnostic feature on surface electrocardiograms because it reflects the duration of the ventricular action potential. A previous genome-wide association study has reported a significant linkage between a single-nucleotide polymorphism ∼11.7 kb downstream of the gene encoding the RING finger ubiquitin ligase rififylin (RFFL) and variability in the QT interval. This, along with results in animal studies, suggests that RFFL may have effects on cardiac repolarization. Here, we sought to determine the role of RFFL in cardiac electrophysiology. Adult rabbit cardiomyocytes with adenovirus-expressed RFFL exhibited reduced rapid delayed rectifier current (IKr). Neonatal rabbit cardiomyocytes transduced with RFFL-expressing adenovirus exhibited reduced total expression of the potassium channel ether-a-go-go-related gene (rbERG). Using transfections of 293A cells and Western blotting experiments, we observed that RFFL and the core-glycosylated form of the human ether-a-go-go-related gene (hERG) potassium channel interact. Furthermore, RFFL overexpression led to increased polyubiquitination and proteasomal degradation of hERG protein and to an almost complete disappearance of IKr, which depended on the intact RING domain of RFFL. Blocking the ER-associated degradation (ERAD) pathway with a dominant-negative form of the ERAD core component, valosin-containing protein (VCP), in 293A cells partially abolished RFFL-mediated hERG degradation. We further substantiated the link between RFFL and ERAD by showing an interaction between RFFL and VCP in vitro We conclude that RFFL is an important regulator of voltage-gated hERG potassium channel activity and therefore cardiac repolarization and that this ubiquitination-mediated regulation requires parts of the ERAD pathway.


Asunto(s)
Canal de Potasio ERG1/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Miocitos Cardíacos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Canal de Potasio ERG1/genética , Células HEK293 , Humanos , Transporte de Proteínas , Conejos , Ubiquitina-Proteína Ligasas/genética
7.
J Electrocardiol ; 62: 20-25, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32745731

RESUMEN

INTRODUCTION: Low serum magnesium (Mg) is associated with an increased incidence of atrial and ventricular arrhythmias. A richer phenotyping of arrhythmia indices, such as burden or frequency, may provide etiologic insights. OBJECTIVES: To evaluate cross-sectional associations of serum Mg with burden of atrial arrhythmias [atrial fibrillation (AF), premature atrial contractions (PAC), supraventricular tachycardia (SVT)], and ventricular arrhythmias [premature ventricular contractions (PVC), non-sustained ventricular tachycardia (NSVT)] over 2-weeks of ECG monitoring. METHODS: We included 2513 ARIC Study visit 6 (2016-2017) participants who wore the Zio XT Patch-a leadless, ambulatory ECG-monitor-for up to 2-weeks. Serum Mg was modeled categorically and continuously. AF burden was categorized as intermittent or continuous based on the percent of analyzable time spent in AF. Other arrhythmia burdens were defined by the average number of abnormal beats per day. Linear regression was used for continuous outcomes; logistic and multinomial regression were used for categorical outcomes. RESULTS: Participants were mean ± SD age 79 ± 5 years, 58% were women and 25% black. Mean serum Mg was 0.82 ± 0.08 mmol/L and 19% had hypomagnesemia (<0.75 mmol/L). Serum Mg was inversely associated with PVC burden and continuous AF. The AF association was no longer statistically significant with further adjustment for traditional lifestyle risk factors, only the association with PVC burden remained significant. There were no associations between serum Mg and other arrhythmias examined. CONCLUSIONS: In this community-based cohort of older adults, we found little evidence of independent cross-sectional associations between serum Mg and arrhythmia burden.


Asunto(s)
Aterosclerosis , Fibrilación Atrial , Anciano , Anciano de 80 o más Años , Fibrilación Atrial/epidemiología , Estudios Transversales , Electrocardiografía , Electrocardiografía Ambulatoria , Femenino , Humanos , Magnesio , Masculino
8.
J Mol Cell Cardiol ; 117: 62-71, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29474817

RESUMEN

RATIONALE: Heart failure is characterized by electrical remodeling that contributes to arrhythmic risk. The unfolded protein response (UPR) is active in heart failure and can decrease protein levels by increasing mRNA decay, accelerating protein degradation, and inhibiting protein translation. OBJECTIVE: Therefore, we investigated whether the UPR downregulated cardiac ion channels that may contribute to arrhythmogenic electrical remodeling. METHODS: Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were used to study cardiac ion channels. Action potentials (APs) and ion channel currents were measured by patch clamp recording. The mRNA and protein levels of channels and the UPR effectors were determined by quantitative RT-PCR and Western blotting. Tunicamycin (TM, 50 ng/mL and 5 µg/mL), GSK2606414 (GSK, 300 nmol/L), and 4µ8C (5 µmol/L) were utilized to activate the UPR, inhibit protein kinase-like ER kinase (PERK) and inositol-requiring protein-1 (IRE1), respectively. RESULTS: TM-induced activation of the UPR caused significant prolongation of the AP duration (APD) and a reduction of the maximum upstroke velocity (dV/dtmax) of the AP phase 0 in both acute (20-24 h) and chronic treatment (6 days). These changes were explained by reductions in the sodium, L-type calcium, the transient outward and rapidly/slowly activating delayed rectifier potassium currents. Nav1.5, Cav1.2, Kv4.3, and KvLQT1 channels showed concomitant reductions in mRNA and protein levels under activated UPR. Inhibition of PERK or IRE1 shortened the APD and reinstated dV/dtmax. The PERK branch regulated Nav1.5, Kv4.3, hERG, and KvLQT1. The IRE1 branch regulated Nav1.5, hERG, KvLQT1, and Cav1.2. CONCLUSIONS: Activated UPR downregulates all major cardiac ion currents and results in electrical remodeling in hiPSC-CMs. Both PERK and IRE1 branches downregulate Nav1.5, hERG, and KvLQT1. The PERK branch specifically downregulates Kv4.3, while the IRE1 branch downregulates Cav1.2. Therefore, the UPR contributed to electrical remodeling, and targeting the UPR might be anti-arrhythmic.


Asunto(s)
Regulación hacia Abajo , Células Madre Pluripotentes Inducidas/citología , Canales Iónicos/metabolismo , Miocitos Cardíacos/metabolismo , Respuesta de Proteína Desplegada , Potenciales de Acción/efectos de los fármacos , Adenina/análogos & derivados , Adenina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Endorribonucleasas/metabolismo , Humanos , Indoles/farmacología , Activación del Canal Iónico/efectos de los fármacos , Isoproterenol/farmacología , Miocitos Cardíacos/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Tunicamicina/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , eIF-2 Quinasa/metabolismo
9.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L748-L759, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28258105

RESUMEN

Right ventricular (RV) dysfunction is associated with numerous smoking-related illnesses, including chronic obstructive pulmonary disease (COPD), in which it is present even in the absence of pulmonary hypertension. It is unknown whether exposure to cigarette smoke (CS) has direct effects on RV function and cardiac fibroblast (CF) proliferation or collagen synthesis. In this study, we evaluated cardiac function and fibrosis in mice exposed to CS and determined mechanisms of smoke-induced changes in CF signaling and fibrosis. AKR mice were exposed to CS for 6 wk followed by echocardiography and evaluation of cardiac hypertrophy, collagen content, and pulmonary muscularization. Proliferation and collagen content were evaluated in primary isolated rat CFs exposed to CS extract (CSE) or nicotine. Markers of cell proliferation, fibrosis, and proliferative signaling were determined by immunoblot or Sircol collagen assay. Mice exposed to CS had significantly decreased RV function, as determined by tricuspid annular plane systolic excursion. There were no changes in left ventricular parameters. RV collagen content was significantly elevated, but there was no change in RV hypertrophy or pulmonary vascular muscularization. CSE directly increased CF proliferation and collagen content in CF. Nicotine alone reproduced these effects. CSE and nicotine-induced fibroblast proliferation and collagen content were mediated through α7 nicotinic acetylcholine receptors and were dependent on PKC-α, PKC-δ, and reduced p38-MAPK phosphorylation. CS and nicotine have direct effects on CFs to induce proliferation and fibrosis, which may negatively affect right heart function.


Asunto(s)
Fibroblastos/metabolismo , Fibroblastos/patología , Ventrículos Cardíacos/patología , Miocardio/patología , Fumar/efectos adversos , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Hemodinámica/efectos de los fármacos , Hipertrofia Ventricular Derecha/complicaciones , Hipertrofia Ventricular Derecha/diagnóstico por imagen , Hipertrofia Ventricular Derecha/patología , Hipertrofia Ventricular Derecha/fisiopatología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones Endogámicos AKR , Nicotina/farmacología , Fosforilación/efectos de los fármacos , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-delta/metabolismo , Ratas Sprague-Dawley , Remodelación Vascular/efectos de los fármacos , Disfunción Ventricular Derecha/complicaciones , Disfunción Ventricular Derecha/diagnóstico por imagen , Disfunción Ventricular Derecha/patología , Disfunción Ventricular Derecha/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Circ Res ; 116(12): 1937-55, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-26044249

RESUMEN

Ventricular arrhythmia is the leading cause of sudden cardiac death (SCD). Deranged cardiac metabolism and abnormal redox state during cardiac diseases foment arrhythmogenic substrates through direct or indirect modulation of cardiac ion channel/transporter function. This review presents current evidence on the mechanisms linking metabolic derangement and excessive oxidative stress to ion channel/transporter dysfunction that predisposes to ventricular arrhythmias and SCD. Because conventional antiarrhythmic agents aiming at ion channels have proven challenging to use, targeting arrhythmogenic metabolic changes and redox imbalance may provide novel therapeutics to treat or prevent life-threatening arrhythmias and SCD.


Asunto(s)
Muerte Súbita Cardíaca/etiología , Cardiopatías/metabolismo , Miocardio/metabolismo , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Señalización del Calcio , Fármacos Cardiovasculares/farmacología , Fármacos Cardiovasculares/uso terapéutico , Muerte Súbita Cardíaca/prevención & control , Uniones Comunicantes/fisiología , Sistema de Conducción Cardíaco/fisiopatología , Homeostasis , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Canales Iónicos/efectos de los fármacos , Canales Iónicos/fisiología , Potenciales de la Membrana , Enfermedades Metabólicas/complicaciones , Enfermedades Metabólicas/fisiopatología , Mitocondrias Cardíacas/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Potasio/fisiología , Especies Reactivas de Oxígeno , Sodio/fisiología
11.
Pacing Clin Electrophysiol ; 40(7): 770-778, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28543286

RESUMEN

We investigated the rates and reasons for crossover to alternative treatment strategies and its impact on mortality in patients who were enrolled in the Atrial Fibrillation Follow-up Investigation of Rhythm Management (AFFIRM) trial. Over a mean follow-up period of 3.5 years, 842 patients underwent crossover to the alternative treatment arms in AFFIRM. The rate of crossover from rhythm to rate control (594/2,033, 29.2%) was more frequent than the rate of crossover from rate to rhythm control (248/2,027, 12.2%, P < 0.0001). The leading reasons for crossover from rhythm to rate control were failure to achieve or maintain sinus rhythm (272/594, 45.8%) and intolerable adverse effects (122/594, 20.5%). In comparison, the major reasons for crossover from rate to rhythm control were failure to control atrial fibrillation symptoms (159/248, 64.1%) and intolerable adverse effects (9/248, 3.6%). This difference in crossover pattern was statistically significant (P < 0.0001). There was a significantly decreased risk of all-cause mortality (adjusted HR: 0.61, 95% CI: 0.48-0.78, P < 0.0001) and cardiac mortality (adjusted hazard ratio [HR]: 0.61, 95% confidence interval [CI]: 0.43-0.88, P = 0.008) in the subgroup of patients who crossed over from rhythm to rate control as compared to those who continued in rhythm control. There was a nonsignificant trend toward decreased all-cause (adjusted HR: 0.76, 95% CI: 0.53-1.10, P = 0.14) and cardiac mortality (adjusted HR: 0.70, 95% CI: 0.42-1.18, P = 0.18) in patients who crossed over from rate to rhythm control as compared to those who continued rate control.


Asunto(s)
Antagonistas Adrenérgicos beta/uso terapéutico , Antiarrítmicos/uso terapéutico , Anticoagulantes/uso terapéutico , Fibrilación Atrial/tratamiento farmacológico , Bloqueadores de los Canales de Calcio/uso terapéutico , Anciano , Fibrilación Atrial/mortalidad , Fibrilación Atrial/fisiopatología , Estudios Cruzados , Quimioterapia Combinada , Femenino , Frecuencia Cardíaca/fisiología , Humanos , Masculino , Resultado del Tratamiento
12.
Europace ; 18(9): 1383-90, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26498162

RESUMEN

AIMS: Heart failure patients are at increased risk of ventricular arrhythmias and all-cause mortality. However, existing clinical and serum markers only modestly predict these adverse events. We sought to use metabolic profiling to identify novel biomarkers in two independent prospective cohorts of patients with implantable cardioverter-defibrillators (ICDs) for primary prevention of sudden cardiac death (SCD). METHODS AND RESULTS: Baseline serum was quantitatively profiled for 42 known biologically relevant amine-based metabolites among 402 patients from the Prospective Observational Study of Implantable Cardioverter-Defibrillators (PROSE-ICD) Study (derivation group) and 240 patients from the Genetic Risk Assessment of Defibrillator Events (GRADE) Study (validation group) for ventricular arrhythmia-induced ICD shocks and all-cause mortality. Three amines, N-methyl-l-histidine, symmetric dimethylarginine (SDMA), and l-kynurenine, were derived and validated to be associated with all-cause mortality. The hazard ratios of mortality in PROSE-ICD and GRADE were 1.48 (95% confidence interval 1.14-1.92) and 1.67 (1.22-2.27) for N-methyl-l-histidine, 1.49 (1.17-1.91) and 1.77 (1.27-2.45) for SDMA, 1.31 (1.06-1.63) and 1.73 (1.32-2.27) for l-kynurenine, respectively. l-Histidine, SDMA, and l-kynurenine were associated with ventricular arrhythmia-induced ICD shocks in PROSE-ICD, but they did not reach statistical significance in the GRADE cohort. CONCLUSION: Utilizing metabolic profiling in two independent prospective cohorts of patients undergoing ICD implantation for primary prevention of SCD, we identified several novel amine markers that were associated with appropriate shock and mortality. These findings shed insight into the potential biologic pathways leading to adverse events in ICD patients. Further studies are needed to confirm the prognostic value of these findings.


Asunto(s)
Aminas/sangre , Muerte Súbita Cardíaca/prevención & control , Desfibriladores Implantables , Cardioversión Eléctrica/instrumentación , Insuficiencia Cardíaca/terapia , Prevención Primaria/métodos , Anciano , Arginina/análogos & derivados , Arginina/sangre , Biomarcadores/sangre , Muerte Súbita Cardíaca/etiología , Cardioversión Eléctrica/efectos adversos , Cardioversión Eléctrica/mortalidad , Femenino , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/mortalidad , Humanos , Quinurenina/sangre , Masculino , Metabolómica , Metilhistidinas/sangre , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Estudios Prospectivos , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento , Estados Unidos
13.
Pacing Clin Electrophysiol ; 39(12): 1373-1378, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27807875

RESUMEN

BACKGROUND: During atrial fibrillation (AF), a high rate of myocyte activation causes cellular stress and initiates the process of atrial remodeling, which further promotes persistence of AF. Although heat shock proteins (HSPs) have been shown to prevent atrial remodeling and suppress the occurrence of AF in cellular and animal experimental models, increased levels of HSP-60 have been observed in patients with postoperative AF, likely reflecting a response to cellular stress. To better understand the role of HSP-60 in relation to AF, we examined the association of HSP-60 levels in relation to the future development of AF in the Multi-Ethnic Study of Atherosclerosis (MESA). METHODS: MESA is a cohort study that recruited 6,814 participants aged 45-84 years and free of known cardiovascular disease at baseline (2000-2002) from six field centers. We investigated 983 participants, selected at random from the total cohort, who had HSP-60 measured and were free of AF at baseline. We tested the association of HSP-60 levels with the incidence of AF using multivariate Cox models after adjustment for demographics, clinical characteristics, and biomarkers. RESULTS: During an average of 10.6 years of follow-up, 77 participants developed AF. We did not observe a significant association between the log-transformed HSP-60 levels and development of AF on either unadjusted or multivariate analysis (adjusted hazard ratio: 1.02 per unit difference on natural log scale, 95% confidence interval: 0.77-1.34 ln (ng/mL). CONCLUSION: Contrary to the findings from the preclinical studies, which demonstrated an important role of HSP-60 in the pathogenesis of AF, we did not observe a significant association between HSP-60 and occurrence of AF.


Asunto(s)
Aterosclerosis/sangre , Aterosclerosis/etnología , Fibrilación Atrial/etnología , Chaperonina 60/sangre , Proteínas Mitocondriales/sangre , Anciano , Anciano de 80 o más Años , Fibrilación Atrial/sangre , Biomarcadores/sangre , Comorbilidad , Progresión de la Enfermedad , Diagnóstico Precoz , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Pronóstico , Reproducibilidad de los Resultados , Medición de Riesgo/métodos , Sensibilidad y Especificidad , Estados Unidos/etnología
14.
Circ J ; 79(3): 470-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25746522

RESUMEN

Despite the growing number of patients affected, the understanding of diastolic dysfunction and heart failure with preserved ejection fraction (HFpEF) is still poor. Clinical trials, largely based on successful treatments for systolic heart failure, have been disappointing, suggesting that HFpEF has a different pathology to that of systolic dysfunction. In this review, general concepts, epidemiology, diagnosis, and treatment of diastolic dysfunction are summarized, with an emphasis on new experiments suggesting that oxidative stress plays a crucial role in the pathogenesis of at least some forms of the disease. This observation has lead to potential new diagnostics and therapeutics for diastolic dysfunction and heart failure caused by diastolic dysfunction.


Asunto(s)
Diástole , Insuficiencia Cardíaca , Estrés Oxidativo , Volumen Sistólico , Animales , Ensayos Clínicos como Asunto , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Humanos
15.
J Emerg Med ; 48(6): 756-61, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25802165

RESUMEN

BACKGROUND: Uncontrolled hypertension is a primary risk factor for development of cardiovascular complications. OBJECTIVE: Determine the point prevalence of left ventricular hypertrophy (LVH) and diastolic dysfunction in an urban emergency department (ED) population with elevated blood pressures (BP) and examine correlations between subclinical disease and patient cardiovascular risk profiles. METHODS: A convenience sample of patients with EBP (>140/90 on two measurements) had limited bedside echocardiograms (LBE). Subclinical hypertensive heart disease was classified as the presence of: LVH, abnormal ejection fraction (EF), or diastolic dysfunction. RESULTS: Thirty-nine patients with EBP were enrolled. The mean age was 46 years (SD = 10.9), 59% were women, 21% were smokers, and 92% had a history of hypertension. The average body mass index was 30.7 (SD = 8.7). Patients were 67% African American, 23% Latino, 5% Caucasian, 3% Asian, and 3% Native American. Subclinical disease was found in 39%: 31% had LVH, 15% had diastolic dysfunction, and 8% had abnormal EF. On bivariate analysis, elevated BP (p = 0.039) and blood urea nitrogen (p = 0.016) were correlated with subclinical heart disease. After adjusting for other covariates, receiving oral/intravenous antihypertensive medications in the ED (p = 0.005) was associated with subclinical heart disease. CONCLUSIONS: We found a point prevalence of subclinical heart disease of 39% in this urban ED population, using LBE. Real-time identification of subclinical heart disease at early stages in the ED in conjunction with abnormal renal function can help emergency physicians identify those patients in need of more aggressive therapy and urgent follow-up.


Asunto(s)
Antihipertensivos/uso terapéutico , Presión Sanguínea , Cardiopatías/epidemiología , Hipertrofia Ventricular Izquierda/epidemiología , Disfunción Ventricular Izquierda/epidemiología , Adulto , Enfermedades Asintomáticas/epidemiología , Nitrógeno de la Urea Sanguínea , Diástole , Servicio de Urgencia en Hospital , Femenino , Hospitales Urbanos , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/epidemiología , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Persona de Mediana Edad , Proyectos Piloto , Prevalencia , Factores de Riesgo , Volumen Sistólico , Población Urbana , Disfunción Ventricular Izquierda/fisiopatología
16.
Int J Mol Sci ; 17(1)2015 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-26729106

RESUMEN

The unfolded protein response (UPR) has been extensively investigated in neurological diseases and diabetes, while its function in heart disease is less well understood. Activated UPR participates in multiple cardiac conditions and can either protect or impair heart function. Recently, the UPR has been found to play a role in arrhythmogenesis during human heart failure by affecting cardiac ion channels expression, and blocking UPR has an antiarrhythmic effect. This review will discuss the rationale for and challenges to targeting UPR in heart disease for treatment of arrhythmias.


Asunto(s)
Arritmias Cardíacas/metabolismo , Cardiopatías/metabolismo , Respuesta de Proteína Desplegada , Animales , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/genética , Arritmias Cardíacas/patología , Chaperón BiP del Retículo Endoplásmico , Cardiopatías/tratamiento farmacológico , Cardiopatías/genética , Cardiopatías/patología , Humanos , Terapia Molecular Dirigida , Respuesta de Proteína Desplegada/efectos de los fármacos
17.
Circ Res ; 110(6): 841-50, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22343711

RESUMEN

RATIONALE: Previously, we demonstrated that a deoxycorticosterone acetate (DOCA)-salt hypertensive mouse model produces cardiac oxidative stress and diastolic dysfunction with preserved systolic function. Oxidative stress has been shown to increase late inward sodium current (I(Na)), reducing the net cytosolic Ca(2+) efflux. OBJECTIVE: Oxidative stress in the DOCA-salt model may increase late I(Na), resulting in diastolic dysfunction amenable to treatment with ranolazine. METHODS AND RESULTS: Echocardiography detected evidence of diastolic dysfunction in hypertensive mice that improved after treatment with ranolazine (E/E':sham, 31.9 ± 2.8, sham+ranolazine, 30.2 ± 1.9, DOCA-salt, 41.8 ± 2.6, and DOCA-salt+ranolazine, 31.9 ± 2.6; P=0.018). The end-diastolic pressure-volume relationship slope was elevated in DOCA-salt mice, improving to sham levels with treatment (sham, 0.16 ± 0.01 versus sham+ranolazine, 0.18 ± 0.01 versus DOCA-salt, 0.23 ± 0.2 versus DOCA-salt+ranolazine, 0.17 ± 0.0 1 mm Hg/L; P<0.005). DOCA-salt myocytes demonstrated impaired relaxation, τ, improving with ranolazine (DOCA-salt, 0.18 ± 0.02, DOCA-salt+ranolazine, 0.13 ± 0.01, sham, 0.11 ± 0.01, sham+ranolazine, 0.09 ± 0.02 seconds; P=0.0004). Neither late I(Na) nor the Ca(2+) transients were different from sham myocytes. Detergent extracted fiber bundles from DOCA-salt hearts demonstrated increased myofilament response to Ca(2+) with glutathionylation of myosin binding protein C. Treatment with ranolazine ameliorated the Ca(2+) response and cross-bridge kinetics. CONCLUSIONS: Diastolic dysfunction could be reversed by ranolazine, probably resulting from a direct effect on myofilaments, indicating that cardiac oxidative stress may mediate diastolic dysfunction through altering the contractile apparatus.


Asunto(s)
Acetanilidas/farmacología , Calcio/metabolismo , Diástole/efectos de los fármacos , Insuficiencia Cardíaca Diastólica/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Miofibrillas/efectos de los fármacos , Piperazinas/farmacología , Acetanilidas/sangre , Animales , Desoxicorticosterona/toxicidad , Diástole/fisiología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/farmacología , Insuficiencia Cardíaca Diastólica/inducido químicamente , Insuficiencia Cardíaca Diastólica/fisiopatología , Ratones , Mineralocorticoides/toxicidad , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Miocitos Cardíacos/fisiología , Miofibrillas/metabolismo , Estrés Oxidativo/fisiología , Piperazinas/sangre , Ranolazina , Sodio/metabolismo , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/tratamiento farmacológico , Disfunción Ventricular Izquierda/fisiopatología
18.
JCI Insight ; 9(15)2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38889387

RESUMEN

Diabetes mellitus (DM) is an independent risk factor for atrial fibrillation (AF). The mechanisms underlying DM-associated AF are unclear. AF and DM are both related to inflammation. We investigated whether DM-associated inflammation contributed to AF risk. Mice were fed with high-fat diet to induce type II DM and were subjected to IL-1ß antibodies, macrophage depletion by clodronate liposomes, a mitochondrial antioxidant (mitoTEMPO), or a cardiac ryanodine receptor 2 (RyR2) stabilizer (S107). All tests were performed at 36-38 weeks of age. DM mice presented with increased AF inducibility, enhanced mitochondrial reactive oxygen species (mitoROS) generation, and activated innate immunity in the atria, as evidenced by enhanced monocyte chemoattractant protein-1 (MCP-1) expression, macrophage infiltration, and IL-1ß levels. Signs of aberrant RyR2 Ca2+ leak were observed in the atria of DM mice. IL-1ß neutralization, macrophage depletion, and exposure to mitoTEMPO and S107 significantly ameliorated the AF vulnerability in DM mice. Atrial overexpression of MCP-1 increased AF occurrence in normal mice through the same mechanistic signaling cascade as observed in DM mice. In conclusion, macrophage-mediated IL-1ß contributed to DM-associated AF risk through mitoROS modulation of RyR2 Ca2+ leak.


Asunto(s)
Fibrilación Atrial , Diabetes Mellitus Experimental , Interleucina-1beta , Macrófagos , Animales , Fibrilación Atrial/metabolismo , Fibrilación Atrial/etiología , Fibrilación Atrial/inmunología , Ratones , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Macrófagos/inmunología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/inmunología , Masculino , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/inmunología , Quimiocina CCL2/metabolismo , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ratones Endogámicos C57BL , Dieta Alta en Grasa/efectos adversos , Inflamación/metabolismo
19.
Circ Arrhythm Electrophysiol ; : e013054, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39212055

RESUMEN

BACKGROUND: Increased mitochondrial Ca2+ uptake has been implicated in the QT prolongation and lethal arrhythmias associated with nonischemic cardiomyopathy. We attempted to define the role of mitochondria in ischemic arrhythmic risk and to identify upstream regulators. METHODS: Myocardial infarction (MI) was induced in wild-type FVB/NJ mice by ligation of the left anterior descending coronary artery. Western blot, immunoprecipitation, ECG telemetry, and patch-clamp techniques were used. RESULTS: After MI, c-Src (proto-oncogene tyrosine-protein kinase Src) and its active form (p-Src Y416) were increased. The activation of c-Src was associated with increased diastolic Ca2+ sparks, action potential duration prolongation, and arrhythmia in MI mice. c-Src upregulation and arrhythmia could be reversed by treatment of mice with the Src inhibitor PP1 but not with the inactive analogue PP3. Tyrosine phosphorylated mitochondrial Ca2+ uniporter (MCU) was upregulated in the heart tissues of MI mice and patients with ischemic cardiomyopathy. In a heterologous expression system, c-Src could bind MCU and phosphorylate MCU tyrosines. Overexpression of wild-type c-Src significantly increased the mitochondrial Ca2+ transient while overexpression of dominant-negative c-Src significantly decreased the mitochondrial Ca2+ transient. c-Src inhibition by PP1, MCU inhibition by Ru360, or MCU knockdown could reduce the action potential duration, Ca2+ sparks, and arrhythmia after MI. The human heart tissue showed that patients with ischemic cardiomyopathy had significantly increased c-Src active form associated with increased MCU tyrosine phosphorylation and ventricular arrhythmia. CONCLUSIONS: MI leads to increased c-Src active form that results in MCU tyrosine phosphorylation, increased mitochondrial Ca2+ uptake, QT prolongation, and arrhythmia, suggesting c-Src or MCU may represent novel antiarrhythmic targets.

20.
Cells ; 13(12)2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38920678

RESUMEN

Successful heart development depends on the careful orchestration of a network of transcription factors and signaling pathways. In recent years, in vitro cardiac differentiation using human pluripotent stem cells (hPSCs) has been used to uncover the intricate gene-network regulation involved in the proper formation and function of the human heart. Here, we searched for uncharacterized cardiac-development genes by combining a temporal evaluation of human cardiac specification in vitro with an analysis of gene expression in fetal and adult heart tissue. We discovered that CARDEL (CARdiac DEvelopment Long non-coding RNA; LINC00890; SERTM2) expression coincides with the commitment to the cardiac lineage. CARDEL knockout hPSCs differentiated poorly into cardiac cells, and hPSC-derived cardiomyocytes showed faster beating rates after controlled overexpression of CARDEL during differentiation. Altogether, we provide physiological and molecular evidence that CARDEL expression contributes to sculpting the cardiac program during cell-fate commitment.


Asunto(s)
Diferenciación Celular , Corazón , Homeostasis , Miocitos Cardíacos , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Diferenciación Celular/genética , Corazón/embriología , Corazón/fisiología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/citología , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Linaje de la Célula/genética , Organogénesis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA