Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
J Cell Mol Med ; 28(12): e18483, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-39051629

RESUMEN

The development of high-throughput technologies has enhanced our understanding of small non-coding RNAs (sncRNAs) and their crucial roles in various diseases, including atrial fibrillation (AF). This study aimed to systematically delineate sncRNA profiles in AF patients. PANDORA-sequencing was used to examine the sncRNA profiles of atrial appendage tissues from AF and non-AF patients. Differentially expressed sncRNAs were identified using the R package DEGseq 2 with a fold change >2 and p < 0.05. The target genes of the differentially expressed sncRNAs were predicted using MiRanda and RNAhybrid. Gene Ontology (GO) categories and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed. In AF patients, the most abundant sncRNAs were ribosomal RNA-derived small RNAs (rsRNAs), followed by transfer RNA-derived small RNAs (tsRNAs), and microRNAs (miRNAs). Compared with non-AF patients, 656 rsRNAs, 45 miRNAs, 191 tsRNAs and 51 small nucleolar RNAs (snoRNAs) were differentially expressed in AF patients, whereas no significantly differentially expressed piwi-interacting RNAs were identified. Two out of three tsRNAs were confirmed to be upregulated in AF patients by quantitative reverse transcriptase polymerase chain reaction, and higher plasma levels of tsRNA 5006c-LysCTT were associated with a 2.55-fold increased risk of all-cause death in AF patients (hazard ratio: 2.55; 95% confidence interval, 1.56-4.17; p < 0.001). Combined with our previous transcriptome sequencing results, 32 miRNA, 31 snoRNA, 110 nucleus-encoded tsRNA, and 33 mitochondria-encoded tsRNA target genes were dysregulated in AF patients. GO and KEGG analyses revealed enrichment of differentially expressed sncRNA target genes in AF-related pathways, including the 'calcium signaling pathway' and 'adrenergic signaling in cardiomyocytes.' The dysregulated sncRNA profiles in AF patients suggest their potential regulatory roles in AF pathogenesis. Further research is needed to investigate the specific mechanisms of sncRNAs in the development of AF and to explore potential biomarkers for AF treatment and prognosis.


Asunto(s)
Apéndice Atrial , Fibrilación Atrial , Perfilación de la Expresión Génica , ARN Pequeño no Traducido , Humanos , Fibrilación Atrial/genética , ARN Pequeño no Traducido/genética , Apéndice Atrial/metabolismo , Masculino , Femenino , MicroARNs/genética , Ontología de Genes , Anciano , Persona de Mediana Edad , ARN Nucleolar Pequeño/genética , ARN Nucleolar Pequeño/metabolismo , Regulación de la Expresión Génica , Transcriptoma/genética , Biología Computacional/métodos , Pronóstico
2.
Clin Transl Med ; 13(6): e1297, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37278111

RESUMEN

BACKGROUND: Atrial fibrillation (AF) is associated with an increased risk of thrombosis of the left atrial appendage (LAA). However, the molecular mechanisms underlying this site-specificity remain poorly understood. Here, we present a comparative single-cell transcriptional profile of paired atrial appendages from patients with AF and illustrate the chamber-specific properties of the main cell types. METHODS: Single-cell RNA sequencing analysis of matched atrial appendage samples from three patients with persistent AF was evaluated by 10× genomics. The AF mice model was created using Tbx5 knockout mice. Validation experiments were performed by glutathione S-transferase pull-down assays, coimmunoprecipitation (Co-IP), cleavage assays and shear stress experiments in vitro. RESULTS: In LAA, phenotype switching from endothelial cells to fibroblasts and inflammation associated with proinflammatory macrophage infiltration were observed. Importantly, the coagulation cascade is highly enriched in LAA endocardial endothelial cells (EECs), accompanying the up-regulation of a disintegrin and metalloproteinase with thrombospondin motifs 1 (ADAMTS1) and the down-regulation of the tissue factor pathway inhibitor (TFPI) and TFPI2. Similar alterations were verified in an AF mouse model (Tbx5+/- ) and EECs treated with simulated AF shear stress in vitro. Furthermore, we revealed that the cleavage of both TFPI and TFPI2 based on their interaction with ADAMTS1 would lead to loss of anticoagulant activities of EECs. CONCLUSIONS: This study highlights the decrease in the anticoagulant status of EECs in LAA as a potential mechanism underlying the propensity for thrombosis, which may aid the development of anticoagulation therapeutic approaches targeting functionally distinct cell subsets or molecules during AF.


Asunto(s)
Apéndice Atrial , Fibrilación Atrial , Trombosis , Animales , Ratones , Fibrilación Atrial/genética , Fibrilación Atrial/complicaciones , Apéndice Atrial/metabolismo , Células Endoteliales/metabolismo , Trombosis/genética , Anticoagulantes/metabolismo , Análisis de Secuencia de ARN
3.
Cardiovasc Diabetol ; 20(1): 149, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34301258

RESUMEN

BACKGROUND: Diabetic patients have an increased predisposition to thromboembolic events, in most cases originating from thrombi in the left atrial appendage (LAA). Remodeling of the LAA, which predisposes to thrombi formation, has been previously described in diabetic patients with atrial fibrillation, but whether remodeling of the LAA occurs in diabetics also in the absence of atrial fibrillation is unknown. To investigate the contribution of diabetes, as opposed to atrial fibrillation, to remodeling of the LAA, we went from humans to the animal model. METHODS: We studied by echocardiography the structure and function of the heart over multiple time points during the evolution of diabetes in the Cohen diabetic sensitive rat (CDs/y) provided diabetogenic diet over a period of 4 months; CDs/y provided regular diet and the Cohen diabetic resistant (CDr/y), which do not develop diabetes, served as controls. All animals were in sinus rhythm throughout the study period. RESULTS: Compared to controls, CDs/y developed during the evolution of diabetes a greater heart mass, larger left atrial diameter, wider LAA orifice, increased LAA depth, greater end-diastolic and end-systolic diameter, and lower E/A ratio-all indicative of remodeling of the LAA and left atrium (LA), as well as the development of left ventricular diastolic dysfunction. To investigate the pathophysiology involved, we studied the histology of the hearts at the end of the study. We found in diabetic CDs/y, but not in any of the other groups, abundance of glycogen granules in the atrial appendages , atria  and ventricles, which may be of significance as glycogen granules have previously been associated with cell and organ dysfunction in the diabetic heart. CONCLUSIONS: We conclude that our rodent model of diabetes, which was in sinus rhythm, reproduced structural and functional alterations previously observed in hearts of human diabetics with atrial fibrillation. Remodeling of the LAA and of the LA in our model was unrelated to atrial fibrillation and associated with accumulation of glycogen granules. We suggest that myocardial accumulation of glycogen granules is related to the development of diabetes and may play a pathophysiological role in remodeling of the LAA and LA, which predisposes to atrial fibrillation, thromboembolic events and left ventricular diastolic dysfunction in the diabetic heart.


Asunto(s)
Apéndice Atrial/fisiopatología , Función del Atrio Izquierdo , Remodelación Atrial , Diabetes Mellitus Tipo 2/complicaciones , Cardiomiopatías Diabéticas/etiología , Animales , Apéndice Atrial/diagnóstico por imagen , Apéndice Atrial/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Cardiomiopatías Diabéticas/diagnóstico por imagen , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ecocardiografía Doppler en Color , Glucógeno/metabolismo , Frecuencia Cardíaca , Masculino , Ratas Endogámicas , Factores de Tiempo , Función Ventricular Izquierda
4.
J Mol Cell Cardiol ; 155: 10-20, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33631188

RESUMEN

AIM: To obtain a quantitative expression profile of the main genes involved in the cAMP-signaling cascade in human control atria and in different cardiac pathologies. METHODS AND RESULTS: Expression of 48 target genes playing a relevant role in the cAMP-signaling cascade was assessed by RT-qPCR. 113 samples were obtained from right atrial appendages (RAA) of patients in sinus rhythm (SR) with or without atrium dilation, paroxysmal atrial fibrillation (AF), persistent AF or heart failure (HF); and left atrial appendages (LAA) from patients in SR or with AF. Our results show that right and left atrial appendages in donor hearts or from SR patients have similar expression values except for AC7 and PDE2A. Despite the enormous chamber-dependent variability in the gene-expression changes between pathologies, several distinguishable patterns could be identified. PDE8A, PI3Kγ and EPAC2 were upregulated in AF. Different phosphodiesterase (PDE) families showed specific pathology-dependent changes. CONCLUSION: By comparing mRNA-expression patterns of the cAMP-signaling cascade related genes in right and left atrial appendages of human hearts and across different pathologies, we show that 1) gene expression is not significantly affected by cardioplegic solution content, 2) it is appropriate to use SR atrial samples as controls, and 3) many genes in the cAMP-signaling cascade are affected in AF and HF but only few of them appear to be chamber (right or left) specific. TOPIC: Genetic changes in human diseased atria. TRANSLATIONAL PERSPECTIVE: The cyclic AMP signaling pathway is important for atrial function. However, expression patterns of the genes involved in the atria of healthy and diseased hearts are still unclear. We give here a general overview of how different pathologies affect the expression of key genes in the cAMP signaling pathway in human right and left atria appendages. Our study may help identifying new genes of interest as potential therapeutic targets or clinical biomarkers for these pathologies and could serve as a guide in future gene therapy studies.


Asunto(s)
AMP Cíclico/metabolismo , Variación Genética , Atrios Cardíacos/metabolismo , Sistemas de Mensajero Secundario/genética , Anciano , Alelos , Apéndice Atrial/metabolismo , Fibrilación Atrial/complicaciones , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Biomarcadores , Susceptibilidad a Enfermedades , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/etiología , Humanos , Masculino , Persona de Mediana Edad , Proteoma , Proteómica/métodos
5.
Pflugers Arch ; 473(3): 461-475, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33454842

RESUMEN

Atrial fibrillation (AF) is strongly associated with risk of stroke and heart failure. AF promotes atrial remodeling that increases risk of stroke due to left atrial thrombogenesis, and increases energy demand to support high rate electrical activity and muscle contraction. While many transcriptomic studies have assessed AF-related changes in mRNA abundance, fewer studies have assessed proteomic changes. We performed a proteomic analysis on left atrial appendage (LAA) tissues from 12 patients with a history of AF undergoing elective surgery; atrial rhythm was documented at time of surgery. Proteomic analysis was performed using liquid chromatography with mass spectrometry (LC/MS-MS). Data-dependent analysis identified 3090 unique proteins, with 408 differentially expressed between sinus rhythm and AF. Ingenuity Pathway Analysis of differentially expressed proteins identified mitochondrial dysfunction, oxidative phosphorylation, and sirtuin signaling among the most affected pathways. Increased abundance of electron transport chain (ETC) proteins in AF was accompanied by decreased expression of ETC complex assembly factors, tricarboxylic acid cycle proteins, and other key metabolic modulators. Discordant changes were also evident in the contractile unit with both up and downregulation of key components. Similar pathways were affected in a comparison of patients with a history of persistent vs. paroxysmal AF, presenting for surgery in sinus rhythm. Together, these data suggest that while the LAA attempts to meet the energetic demands of AF, an uncoordinated response may reduce ATP availability, contribute to tissue contractile and electrophysiologic heterogeneity, and promote a progression of AF from paroxysmal episodes to development of a substrate amenable to persistent arrhythmia.


Asunto(s)
Apéndice Atrial/metabolismo , Fibrilación Atrial/metabolismo , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteómica
6.
Cardiovasc Res ; 117(7): 1790-1801, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32520995

RESUMEN

AIMS: Atrial fibrillation (AF) is a commonly occurring arrhythmia after cardiac surgery (postoperative AF, poAF) and is associated with poorer outcomes. Considering that reduced atrial contractile function is a predictor of poAF and that Ca2+ plays an important role in both excitation-contraction coupling and atrial arrhythmogenesis, this study aims to test whether alterations of intracellular Ca2+ handling contribute to impaired atrial contractility and to the arrhythmogenic substrate predisposing patients to poAF. METHODS AND RESULTS: Right atrial appendages were obtained from patients in sinus rhythm undergoing open-heart surgery. Cardiomyocytes were investigated by simultaneous measurement of [Ca2+]i and action potentials (APs, patch-clamp). Patients were followed-up for 6 days to identify those with and without poAF. Speckle-tracking analysis of preoperative echocardiography revealed reduced left atrial contraction strain in poAF patients. At the time of surgery, cellular Ca2+ transients (CaTs) and the sarcoplasmic reticulum (SR) Ca2+ content were smaller in the poAF group. CaT decay was slower in poAF, but the decay of caffeine-induced Ca2+ transients was unaltered, suggesting preserved sodium-calcium exchanger function. In agreement, western blots revealed reduced SERCA2a expression in poAF patients but unaltered phospholamban expression/phosphorylation. Computational modelling indicated that reduced SERCA activity promotes occurrence of CaT and AP alternans. Indeed, alternans of CaT and AP occurred more often and at lower stimulation frequencies in atrial myocytes from poAF patients. Resting membrane potential and AP duration were comparable between both groups at various pacing frequencies (0.25-8 Hz). CONCLUSIONS: Biochemical, functional, and modelling data implicate reduced SERCA-mediated Ca2+ reuptake into the SR as a major contributor to impaired preoperative atrial contractile function and to the pre-existing arrhythmogenic substrate in patients developing poAF.


Asunto(s)
Potenciales de Acción , Apéndice Atrial/metabolismo , Fibrilación Atrial/etiología , Señalización del Calcio , Calcio/metabolismo , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Frecuencia Cardíaca , Miocitos Cardíacos/metabolismo , Anciano , Apéndice Atrial/fisiopatología , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Proteínas de Unión al Calcio/metabolismo , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fosforilación , Retículo Sarcoplasmático/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Factores de Tiempo
7.
Cardiovasc Ther ; 2020: 5925342, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33294032

RESUMEN

Several studies have indicated that long noncoding RNAs (lncRNAs)-HOX transcript antisense RNA (HOTAIR) is involved in some cardiovascular diseases by regulating gene expression as a competitive endogenous RNA (ceRNA). GJA1 encoding Cx43 is one potential target gene of microRNA-613 (miR-613). Meanwhile, there is a potential target regulatory relationship between HOTAIR and miR-613. The present study is aimed at investigating whether HOTAIR functions as a ceRNA to regulate the Cx43 expression in atrial fibrillation (AF) by sponging miR-613. The expressions of HOTAIR, miR-613, and Cx43 were detected in the right atrial appendages of 45 patients with heart valve disease, including 23 patients with chronic AF. The HOTAIR overexpressed and underexpressed HL-1 cell model were constructed to confirm the effect of HOTAIR on Cx43. Then, the Cx43 expression was detected to testify the interplay between HOTAIR and miR-613 after cotransfecting HOTAIR and miR-613. Furthermore, luciferase assays were performed to verify that HOTAIR could regulate Cx43 remolding as a ceRNA by sponging miR-613. The expression of HOTAIR and Cx43 was significantly downregulated in chronic AF group. HOTAIR regulated positively the Cx43 expression in HL-1 cells. The upregulated effect of HOTAIR on the Cx43 expression could be remarkably attenuated by miR-613. Moreover, the inhibitory effect of miR-613 on the Cx43 expression could be obviously mitigated by HOTAIR. At last, luciferase assays confirmed HOTAIR functioned as a ceRNA in the Cx43 expression by sponging miR-613. Our study suggests that HOTAIR, functioning as a ceRNA by sponging miR-613, is an important contributor to Cx43 remolding in AF.


Asunto(s)
Apéndice Atrial/metabolismo , Fibrilación Atrial/metabolismo , Remodelación Atrial , Conexina 43/metabolismo , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/metabolismo , Adulto , Anciano , Animales , Apéndice Atrial/fisiopatología , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Línea Celular , Conexina 43/genética , Femenino , Regulación de la Expresión Génica , Frecuencia Cardíaca , Humanos , Masculino , Ratones , MicroARNs/genética , Persona de Mediana Edad , ARN Largo no Codificante/genética , Transducción de Señal
8.
JCI Insight ; 5(16)2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32814717

RESUMEN

BACKGROUNDGenomic and experimental studies suggest a role for PITX2 in atrial fibrillation (AF). To assess if this association is relevant for recurrent AF in patients, we tested whether left atrial PITX2 affects recurrent AF after AF ablation.METHODSmRNA concentrations of PITX2 and its cardiac isoform, PITX2c, were quantified in left atrial appendages (LAAs) from patients undergoing thoracoscopic AF ablation, either in whole LAA tissue (n = 83) or in LAA cardiomyocytes (n = 52), and combined with clinical parameters to predict AF recurrence. Literature suggests that BMP10 is a PITX2-repressed, atrial-specific, secreted protein. BMP10 plasma concentrations were combined with 11 cardiovascular biomarkers and clinical parameters to predict recurrent AF after catheter ablation in 359 patients.RESULTSReduced concentrations of cardiomyocyte PITX2, but not whole LAA tissue PITX2, were associated with AF recurrence after thoracoscopic AF ablation (16% decreased recurrence per 2-(ΔΔCt) increase in PITX2). RNA sequencing, quantitative PCR, and Western blotting confirmed that BMP10 is one of the most PITX2-repressed atrial genes. Left atrial size (HR per mm increase [95% CI], 1.055 [1.028, 1.082]); nonparoxysmal AF (HR 1.672 [1.206, 2.318]), and elevated BMP10 (HR 1.339 [CI 1.159, 1.546] per quartile increase) were predictive of recurrent AF. BMP10 outperformed 11 other cardiovascular biomarkers in predicting recurrent AF.CONCLUSIONSReduced left atrial cardiomyocyte PITX2 and elevated plasma concentrations of the PITX2-repressed, secreted atrial protein BMP10 identify patients at risk of recurrent AF after ablation.TRIAL REGISTRATIONClinicalTrials.gov NCT01091389, NL50069.018.14, Dutch National Registry of Clinical Research Projects EK494-16.FUNDINGBritish Heart Foundation, European Union (H2020), Leducq Foundation.


Asunto(s)
Apéndice Atrial/citología , Fibrilación Atrial/etiología , Fibrilación Atrial/cirugía , Proteínas Morfogenéticas Óseas/sangre , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Anciano , Apéndice Atrial/metabolismo , Biomarcadores/sangre , Proteínas Morfogenéticas Óseas/metabolismo , Ablación por Catéter , Femenino , Proteínas de Homeodominio/genética , Humanos , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Toracoscopía , Factores de Transcripción/genética , Proteína del Homeodomínio PITX2
9.
Circ Arrhythm Electrophysiol ; 13(8): e008382, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32654517

RESUMEN

BACKGROUND: Postoperative atrial fibrillation (POAF) occurs in 30% to 50% of patients undergoing cardiac surgery and is associated with increased morbidity and mortality. Prospective identification of structural/molecular changes in atrial myocardium that correlate with myocardial injury and precede and predict risk of POAF may identify new molecular pathways and targets for prevention of this common morbid complication. METHODS: Right atrial appendage samples were prospectively collected during cardiac surgery from 239 patients enrolled in the OPERA trial (Omega-3 Fatty Acids for Prevention of Post-Operative Atrial Fibrillation), fixed in 10% buffered formalin, and embedded in paraffin for histology. We assessed general tissue morphology, cardiomyocyte diameters, myocytolysis (perinuclear myofibril loss), accumulation of perinuclear glycogen, interstitial fibrosis, and myocardial gap junction distribution. We also assayed NT-proBNP (N-terminal pro-B-type natriuretic peptide), hs-cTnT, CRP (C-reactive protein), and circulating oxidative stress biomarkers (F2-isoprostanes, F3-isoprostanes, isofurans) in plasma collected before, during, and 48 hours after surgery. POAF was defined as occurrence of postcardiac surgery atrial fibrillation or flutter of at least 30 seconds duration confirmed by rhythm strip or 12-lead ECG. The follow-up period for all arrhythmias was from surgery until hospital discharge or postoperative day 10. RESULTS: Thirty-five percent of patients experienced POAF. Compared with the non-POAF group, they were slightly older and more likely to have chronic obstructive pulmonary disease or heart failure. They also had a higher European System for Cardiac Operative Risk Evaluation and more often underwent valve surgery. No differences in left atrial size were observed between patients with POAF and patients without POAF. The extent of atrial interstitial fibrosis, cardiomyocyte myocytolysis, cardiomyocyte diameter, glycogen score or Cx43 distribution at the time of surgery was not significantly associated with incidence of POAF. None of these histopathologic abnormalities were correlated with levels of NT-proBNP, hs-cTnT, CRP, or oxidative stress biomarkers. CONCLUSIONS: In sinus rhythm patients undergoing cardiac surgery, histopathologic changes in the right atrial appendage do not predict POAF. They also do not correlate with biomarkers of cardiac function, inflammation, and oxidative stress. Graphic Abstract: A graphic abstract is available for this article.


Asunto(s)
Apéndice Atrial/fisiopatología , Fibrilación Atrial/etiología , Aleteo Atrial/etiología , Función del Atrio Derecho , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Frecuencia Cardíaca , Potenciales de Acción , Anciano , Apéndice Atrial/metabolismo , Apéndice Atrial/patología , Fibrilación Atrial/sangre , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/fisiopatología , Aleteo Atrial/sangre , Aleteo Atrial/diagnóstico , Aleteo Atrial/fisiopatología , Remodelación Atrial , Biomarcadores/sangre , Proteína C-Reactiva/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Péptido Natriurético Encefálico/sangre , Estrés Oxidativo , Fragmentos de Péptidos/sangre , Estudios Prospectivos , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento , Troponina T/sangre
10.
Rev Invest Clin ; 72(2): 103-109, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32284626

RESUMEN

BACKGROUND: The left atrial appendage (LAAp) resection is an effective treatment approach to reduce the risk of thromboembolism in patients with atrial fibrillation. OBJECTIVE: To study was to study the impact of removing atrial appendages in the production of natriuretic peptides (NPs) in conditions of volume overload and to develop an experimental model of LAAp resection. MATERIALS AND METHODS: In a swine model of ischemic heart failure (HF), serum NP levels were measured before (Basal-1A) and after (Basal-1B) a fluid overload. Animals were grouped as follows: (0) preserved appendages, (1) resected LAAp, and (2) both atrial appendages resected. Levels of NP were measured before (2A) and after a fluid overload (2B). RESULTS: Furin levels were higher in Group 0-2A than in Group 2-2A, and a significant increase was found in Group 0-2B compared to Groups 1-2B and 2-2B. Corin levels increased in Basal-1B versus Basal-1A. Atrial NP (ANP) decreased in Basal-1B compared to Basal-1A. After HF induction, ANP increased in Groups 2-2A and 2-2B. CONCLUSIONS: Resection of atrial appendages drastically modifies the natriuretic mechanisms of cardiac homeostasis, especially after a fluid overload challenge. Herein, we describe the face and predictive validation of an animal model of atrial appendage resection useful to investigations in translational medicine.


Asunto(s)
Apéndice Atrial/metabolismo , Apéndice Atrial/cirugía , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/cirugía , Homeostasis , Péptidos Natriuréticos/biosíntesis , Péptidos Natriuréticos/fisiología , Centros Médicos Académicos , Animales , Masculino , Porcinos
11.
Rev. invest. clín ; 72(2): 103-109, Mar.-Apr. 2020. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1251841

RESUMEN

ABSTRACT Background: The left atrial appendage (LAAp) resection is an effective treatment approach to reduce the risk of thromboembolism in patients with atrial fibrillation. Objective: To study was to study the impact of removing atrial appendages in the production of natriuretic peptides (NPs) in conditions of volume overload and to develop an experimental model of LAAp resection. Materials and Methods: In a swine model of ischemic heart failure (HF), serum NP levels were measured before (Basal-1A) and after (Basal-1B) a fluid overload. Animals were grouped as follows: (0) preserved appendages, (1) resected LAAp, and (2) both atrial appendages resected. Levels of NP were measured before (2A) and after a fluid overload (2B). Results: Furin levels were higher in Group 0-2A than in Group 2-2A, and a significant increase was found in Group 0-2B compared to Groups 1-2B and 2-2B. Corin levels increased in Basal-1B versus Basal-1A. Atrial NP (ANP) decreased in Basal-1B compared to Basal-1A. After HF induction, ANP increased in Groups 2-2A and 2-2B. Conclusions: Resection of atrial appendages drastically modifies the natriuretic mechanisms of cardiac homeostasis, especially after a fluid overload challenge. Herein, we describe the face and predictive validation of an animal model of atrial appendage resection useful to investigations in translational medicine.


Asunto(s)
Animales , Masculino , Apéndice Atrial/cirugía , Apéndice Atrial/metabolismo , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/cirugía , Insuficiencia Cardíaca/metabolismo , Homeostasis , Porcinos , Péptidos Natriuréticos/biosíntesis , Péptidos Natriuréticos/fisiología , Centros Médicos Académicos
12.
J Mol Cell Cardiol ; 140: 10-21, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32006532

RESUMEN

Hypertension is an independent risk factor for atrial fibrillation (AF), although its specific mechanisms remain unclear. Previous research has been focused on cyclic stretch, ignoring the role of high hydrostatic pressure. The present study aimed to explore the effect of high hydrostatic pressure stimulation on electrical remodeling in atrial myocytes and its potential signaling pathways. Experiments were performed on left atrial appendages from patients with chronic AF or sinus rhythm, spontaneously hypertensive rats (SHRs) treated with or without valsartan (10 mg/kg/day) and HL-1 cells were exposed to high hydrostatic pressure using a self-developed device. Whole-cell patch-clamp recordings and western blots demonstrated that the amplitudes of ICa,L, Ito, and IKur were reduced in AF patients with corresponding changes in protein expression. Angiotensin protein levels increased and Ang1-7 decreased, while focal adhesion kinase (FAK) and Src kinase were enhanced in atrial tissue from AF patients and SHRs. After rapid atrial pacing, AF inducibility in SHR was significantly higher, accompanied by a decrease in ICa,L, upregulation of Ito and IKur, and a shortened action potential duration. Angiotensin upregulation and FAK/Src activation in SHR were inhibited by angiotensin type 1 receptor inhibitor valsartan, thus, preventing electrical remodeling and reducing AF susceptibility. These results were verified in HL-1 cells treated with high hydrostatic pressure, and demonstrated that electrical remodeling regulated by the FAK-Src pathway could be modulated by valsartan. The present study indicated that high hydrostatic pressure stimulation increases AF susceptibility by activating the renin-angiotensin system and FAK-Src pathway in atrial myocytes.


Asunto(s)
Angiotensina II/metabolismo , Angiotensina I/metabolismo , Remodelación Atrial/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Fragmentos de Péptidos/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Familia-src Quinasas/metabolismo , Animales , Antiarrítmicos/farmacología , Apéndice Atrial/metabolismo , Fibrilación Atrial/patología , Línea Celular Tumoral , Humanos , Presión Hidrostática , Ratones , Miocitos Cardíacos/metabolismo , Ratas , Ratas Endogámicas SHR , Receptor de Angiotensina Tipo 1/metabolismo , Valsartán/farmacología
13.
Circ Res ; 126(5): 603-615, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31902278

RESUMEN

RATIONALE: Sleep-disordered breathing (SDB) is frequently associated with atrial arrhythmias. Increased CaMKII (Ca/calmodulin-dependent protein kinase II) activity has been previously implicated in atrial arrhythmogenesis. OBJECTIVE: We hypothesized that CaMKII-dependent dysregulation of Na current (INa) may contribute to atrial proarrhythmic activity in patients with SDB. METHODS AND RESULTS: We prospectively enrolled 113 patients undergoing elective coronary artery bypass grafting for cross-sectional study and collected right atrial appendage biopsies. The presence of SDB (defined as apnea-hypopnea index ≥15/h) was assessed with a portable SDB monitor the night before surgery. Compared with 56 patients without SDB, patients with SDB (57) showed a significantly increased level of activated CaMKII. Patch clamp was used to measure INa. There was a significantly enhanced late INa, but reduced peak INa due to enhanced steady-state inactivation in atrial myocytes of patients with SDB consistent with significantly increased CaMKII-dependent cardiac Na channel phosphorylation (NaV1.5, at serine 571, Western blotting). These gating changes could be fully reversed by acute CaMKII inhibition (AIP [autocamtide-2 related inhibitory peptide]). As a consequence, we observed significantly more cellular afterdepolarizations and more severe premature atrial contractions in atrial trabeculae of patients with SDB, which could be blocked by either AIP or KN93 (N-[2-[[[(E)-3-(4-chlorophenyl)prop-2-enyl]-methylamino]methyl]phenyl]-N-(2-hydroxyethyl)-4-methoxybenzenesulfonamide). In multivariable linear regression models incorporating age, sex, body mass index, existing atrial fibrillation, existing heart failure, diabetes mellitus, and creatinine levels, apnea-hypopnea index was independently associated with increased CaMKII activity, enhanced late INa and correlated with premature atrial contraction severity. CONCLUSIONS: In atrial myocardium of patients with SDB, increased CaMKII-dependent phosphorylation of NaV1.5 results in dysregulation of INa with proarrhythmic activity that was independent from preexisting comorbidities. Inhibition of CaMKII may be useful for prevention or treatment of arrhythmias in SDB. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT02877745. Visual Overview: An online visual overview is available for this article.


Asunto(s)
Arritmias Cardíacas/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Síndromes de la Apnea del Sueño/metabolismo , Potenciales de Acción , Anciano , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/fisiopatología , Apéndice Atrial/efectos de los fármacos , Apéndice Atrial/metabolismo , Apéndice Atrial/fisiopatología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Células Cultivadas , Femenino , Humanos , Activación del Canal Iónico , Masculino , Persona de Mediana Edad , Péptidos/farmacología , Fosforilación , Síndromes de la Apnea del Sueño/complicaciones , Síndromes de la Apnea del Sueño/fisiopatología
14.
Exp Cell Res ; 389(2): 111821, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-31923425

RESUMEN

Circular RNAs (circRNAs) have emerged as a novel type of non-coding RNA (ncRNA) of interest in gene regulation, especially for its vital function underlying many diseases. Atrial fibrillation is the most common sustained arrythmia. However, the expression spectrum and function of circRNAs in atrial appendage of patients with atrial fibrillation (AF) has seldomly been investigated. Human atrial appendage tissues were acquired during cardiac surgery, which were divided into the AF group and the Sinus rhythm (SR) group. The expression characterization of circRNAs of two groups was revealed by high-throughput sequencing. The dysregulated circRNAs were identified and analyzed by bioinformatics methods, and further validated by realtime PCR. A total 18109 circRNAs in human atrial appendage tissues were targeted. Among them, 147 differentially expressed circRNAs (102 up-regulated and 45 down-regulated) were found between AF group and SR group. Gene ontology (GO) and KEGG pathway analysis indicated that many mRNAs transcribed from the host genes of altered circRNAs were implicated in regulation of sequence-specific DNA binding transcription factor activity, as well as nicotinate and nicotinamide metabolism pathways. Analysis of the association between differently expressed circRNA and miRNA were explored, which revealed an ample interaction. Our study firstly revealed the expression spectrum of circRNAs in both left and right atrial appendage of patients with or without AF. Differentially expressed circRNAs in the atrial appendage were also identified, analyzed and validated. The results of this study may provide novel biomarkers and potential therapeutic targets for AF.


Asunto(s)
Apéndice Atrial/metabolismo , Fibrilación Atrial/genética , Biomarcadores/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes , ARN Circular/genética , Apéndice Atrial/patología , Fibrilación Atrial/patología , Estudios de Casos y Controles , Biología Computacional , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , MicroARNs/genética , ARN Mensajero/genética
15.
J Cardiovasc Pharmacol ; 75(2): 141-147, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31789884

RESUMEN

Atrial apoptosis has been found to be majorly involved in the pathogenesis of human atrial fibrillation (AF). Mesencephalic astrocyte-derived neurotrophic factor exerts an antiapoptotic effect for multiple cell types. However, the correlation between MANF and atrial apoptosis in AF is still undefined. In this study, 59 patients with valvular or congenital heart disease were divided into 2 groups: AF group and sinus rhythm (SR) group. We found that the apoptotic atrial myocytes in the right atrial appendage tissues of the AF group were significantly more than those of the SR group, whereas mRNA and protein levels of MANF in the AF group were significantly down-regulated compared with those in the SR group. The serum MANF in patients with AF was markedly lower than that in patients with SR, which was inversely correlated with atrial apoptosis in patients with AF. In addition, the AF group had the greater inflammation and endoplasmic reticulum stress compared with the SR group. These findings suggest that MANF downregulation may lead to more atrial apoptosis in human chronic AF, indicating MANF as a potential therapeutic agent in AF treatment.


Asunto(s)
Apéndice Atrial/metabolismo , Fibrilación Atrial/sangre , Factores de Crecimiento Nervioso/sangre , Adulto , Anciano , Apoptosis , Apéndice Atrial/patología , Apéndice Atrial/fisiopatología , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/fisiopatología , Función del Atrio Derecho , Remodelación Atrial , Biomarcadores/sangre , Estudios de Casos y Controles , Enfermedad Crónica , Regulación hacia Abajo , Estrés del Retículo Endoplásmico , Femenino , Frecuencia Cardíaca , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Persona de Mediana Edad
16.
Int J Cardiol ; 301: 103-107, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31787387

RESUMEN

INTRODUCTION: Atrial fibrillation (AF), a risk factor for stroke and systemic thromboembolism, is associated with unfavorable fibrin clot properties and increased thrombus formation in peripheral blood. The left atrial appendage (LAA) is known to be the primary site of thrombus formation. AIM: We investigated the relative differences in plasma fibrin clot features including plasma fibrin clot permeability (Ks) and clot lysis time (CLT) between the right atrium (RA), right ventricle (RV), left atrium (LA), left ventricle (LV), LAA, and peripheral blood. METHODS: Sixteen patients with nonvalvular AF who stopped oral anticoagulant therapy at least 2 days before a LARIAT procedure participated in a single-center prospective study. We measured fibrinogen and plasminogen levels along with Ks, CLT, and endogenous thrombin potential (ETP) during the LARIAT procedure in blood obtained from the right femoral vein, RA, RV, LA, LV and LAA. RESULTS: LAA clot porosity was reduced by 16.2% compared to peripheral blood (p = 0.026), also after adjustment for fibrinogen levels (p = 0.038). Ks was similar for the RA, RV, LA, LV, and LAA (all p > 0.05). We found 14.7% prolonged CLT for clots prepared from blood samples obtained from the LAA compared to those prepared from peripheral blood, but no differences between the RA, RV, LA and LV (all p > 0.05) were found. There were no significant differences in other parameters, including ETP, between heart chambers. CONCLUSIONS: Patients with AF are characterised by a local prothrombotic state as reflected by formation of compact fibrin clots in the LAA compared to peripheral blood, which may contribute to LAA thrombus formation and device-related thrombi.


Asunto(s)
Apéndice Atrial/metabolismo , Fibrilación Atrial/complicaciones , Fibrilación Atrial/metabolismo , Factores de Coagulación Sanguínea/metabolismo , Cardiopatías/etiología , Trombosis/etiología , Anciano , Estudios de Cohortes , Femenino , Fibrina/metabolismo , Tiempo de Lisis del Coágulo de Fibrina , Cardiopatías/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Trombosis/metabolismo
17.
Gene ; 728: 144286, 2020 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-31838248

RESUMEN

Atrial fibrillation (AF) is an abnormal heart rhythm characterized by rapid and irregular beating of the atria. The non-coding RNAs (ncRNAs) have attracted much attention of AF researchers, as they play a critical role in the transcriptional and post-transcriptional regulation, which could greatly benefit the interpretation of the pathogenesis of AF. However, circRNAs, as a special member of the ncRNAs, and their role in the pathogenesis of AF is less understood. In the present study, we detected a total of 14,215 circRNAs in AF patients and healthy controls. Differential expression analysis of these circRNAs revealed 20 upregulated and 3 downregulated circRNAs, which were differentially expressed in both left and right atrial appendages. The association analysis of the AF-related circRNAs and their parental genes revealed that hsa_circ_0003965 had significantly negative correlation with its parental gene TMEM245 (PCC = -0.51), suggesting that the dysregulation of hsa_circ_0003965 was not regulated by the transcription of its parental gene, but could be associated with glucagon signaling pathway. The competing endogenous RNA (ceRNA) network analysis revealed two upregulated genes, IFNG and GDF7, and one downregulated gene, BMP7, all of which were involved in TGF-beta signaling pathway, which further suggested that these circRNAs, namely hsa_circ_0000075 and hsa_circ_0082096, participated in the AF pathogenesis via TGF-beta signaling pathway. Consistently, TGF-beta signaling pathway was a well-recognized player for its association with atrial fibrosis in AF. In summary, we aimed to discover and provide key circRNAs involved in AF for AF-related researchers, which had the potential to greatly improve our understanding of the underlying mechanism behind circRNAs and AF.


Asunto(s)
Apéndice Atrial/metabolismo , Fibrilación Atrial/diagnóstico , Biomarcadores/análisis , Genoma Humano , MicroARNs/genética , ARN Circular/genética , ARN Mensajero/genética , Fibrilación Atrial/genética , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Pronóstico
18.
J Cardiovasc Med (Hagerstown) ; 20(11): 752-761, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31567634

RESUMEN

BACKGROUND: Atrial fibrillation is the most common cardiac arrhythmia, posing a heavy burden on patients' wellbeing and healthcare budgets. Patients undergoing cardiac surgery are at risk of developing postoperative atrial fibrillation (POAF), new-onset atrial fibrillation and subsequent atrial fibrillation-related complications, including stroke. Sufficient clinical identification of patients at risk fails while the pathological substrate changes that precede atrial fibrillation remain unknown. Here, we describe the PREDICT AF study design, which will be the first study to associate tissue pathophysiology and blood biomarkers with clinical profiling and follow-up of cardiothoracic surgery patients for the prediction of future atrial fibrillation. METHODS: PREDICT AF will include 150 patients without atrial fibrillation and a CHA2DS2-VASc score of at least 2 undergoing cardiac surgery. The left atrial appendage will be excised during surgery and blood samples will be collected before surgery and at 6 and 12 months' follow-up. Tissue and blood analysis will be used for the discovery of biomarkers including microRNAs and protein biomarkers. The primary study endpoint is atrial fibrillation, which will be objectified by 24 h Holters and ECGs after 30 days for POAF and after 6, 12 and 24 months for new-onset atrial fibrillation. Secondary endpoints include the dynamic changes of blood biomarkers over time and other atrial arrhythmias. PREDICT AF participants may benefit from extensive postoperative care with clinical phenotyping, rhythm monitoring and primary prevention of stroke. CONCLUSION: We here describe the PREDICT AF trial design, which will enable the discovery of biomarkers that truly predict POAF and new-onset atrial fibrillation by combining tissue and plasma-derived biomarkers with comprehensive clinical follow-up data. TRIAL REGISTRATION: Retrospectively registered NCT03130985 27 April 2017.


Asunto(s)
Apéndice Atrial/metabolismo , Fibrilación Atrial/etiología , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Apéndice Atrial/cirugía , Fibrilación Atrial/sangre , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/fisiopatología , Biomarcadores/sangre , Electrocardiografía , Femenino , Frecuencia Cardíaca , Humanos , Masculino , Persona de Mediana Edad , Países Bajos , Valor Predictivo de las Pruebas , Estudios Prospectivos , Factores Protectores , Proyectos de Investigación , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
19.
Eur Rev Med Pharmacol Sci ; 23(17): 7605-7610, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31539152

RESUMEN

OBJECTIVE: Recently, long noncoding RNAs (lncRNAs) have caught more attention for their role in the progression of many diseases. Among them, lncRNA GAS5 (Growth Inhibition Specificity 5) was studied in this research to identify how it affects the progression of atrial fibrillation (AF). PATIENTS AND METHODS: In 40 patients with AF and 30 patients with sinus rhythm (SR), the GAS5 expression of the right atrial appendage (RAA) tissues was detected by the quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). Moreover, the cell proliferation assay was conducted in AC16 cells transfected with GAS5 inhibitor and mimics, respectively. Furthermore, the qRT-PCR was performed to uncover the mechanism. RESULTS: In the research, the expression of GAS5 in RAA tissues was decreased significantly in AF patients than that in SR ones. Moreover, overexpression of GAS5 inhibited cell growth in AC16 cells, while knockdown of GAS5 promoted cell growth in AC16 cells. In addition, further experiments revealed that ALK5 was a target of GAS5 and its expression in AF tissues negatively correlated to GAS5 expression. CONCLUSIONS: These results indicate that GAS5 could inhibit cell proliferation of AF via suppressing ALK5, which may offer a new vision for interpreting the mechanism of AF development.


Asunto(s)
Fibrilación Atrial/patología , ARN Largo no Codificante/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Arritmia Sinusal/complicaciones , Arritmia Sinusal/genética , Arritmia Sinusal/patología , Apéndice Atrial/metabolismo , Apéndice Atrial/patología , Fibrilación Atrial/complicaciones , Fibrilación Atrial/genética , Línea Celular , Proliferación Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Interferencia de ARN , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/genética , ARN Interferente Pequeño/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética
20.
Eur Rev Med Pharmacol Sci ; 23(10): 4419-4424, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31173317

RESUMEN

OBJECTIVE: To investigate the specific role of miR-23 in atrial fibrillation (AF) progression and explore the possible underlying mechanism. PATIENTS AND METHODS: Right atrial appendage (RAA) tissues were collected from 30 patients with AF and 30 patients with sinus rhythm (SR), respectively. The expression level of miR-23 was detected by quantitative Real time-polymerase chain reaction (qRT-PCR). Moreover, cell counting kit-8 and flow cytometry were performed to detect the proliferation and cell apoptosis of AC16 cells after transfection with miR-23 inhibitor and mimics. Furthermore, luciferase reporter gene assay and RNA immunoprecipitation assay were performed to uncover the possible underlying mechanism. RESULTS: In the present study, the expression level of miR-23 in RAA tissues of AF patients was significantly higher than that of SR patients. After knockdown of miR-23 in AC16 cells, the proliferation was inhibited and cell apoptosis was induced. However, overexpression of miR-23 significantly promoted cell growth and suppressed cell apoptosis. Further experiments revealed that transforming growth factor-b1 (TGF-ß1) was a direct target of miR-23. In addition, TGF-ß1 expression was positively correlated with miR-23 expression in AF tissues. CONCLUSIONS: Our findings indicated that miR-23 could promote the progression of AF via promoting TGF-ß1, which might serve as a new direction for interpreting the mechanism of AF development.


Asunto(s)
Apoptosis/genética , Fibrilación Atrial/genética , Fibrilación Atrial/patología , Fibroblastos/patología , MicroARNs/genética , Factor de Crecimiento Transformador beta1/genética , Apéndice Atrial/metabolismo , Recuento de Células , Línea Celular , Proliferación Celular , Progresión de la Enfermedad , Marcación de Gen , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA