Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Cardiovasc Drugs Ther ; 33(6): 649-660, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31916131

RESUMEN

PURPOSE: Several studies have indicated a potential role for SCN10A/NaV1.8 in modulating cardiac electrophysiology and arrhythmia susceptibility. However, by which mechanism SCN10A/NaV1.8 impacts on cardiac electrical function is still a matter of debate. To address this, we here investigated the functional relevance of NaV1.8 in atrial and ventricular cardiomyocytes (CMs), focusing on the contribution of NaV1.8 to the peak and late sodium current (INa) under normal conditions in different species. METHODS: The effects of the NaV1.8 blocker A-803467 were investigated through patch-clamp analysis in freshly isolated rabbit left ventricular CMs, human left atrial CMs and human-induced pluripotent stem cell-derived CMs (hiPSC-CMs). RESULTS: A-803467 treatment caused a slight shortening of the action potential duration (APD) in rabbit CMs and hiPSC-CMs, while it had no effect on APD in human atrial cells. Resting membrane potential, action potential (AP) amplitude, and AP upstroke velocity were unaffected by A-803467 application. Similarly, INa density was unchanged after exposure to A-803467 and NaV1.8-based late INa was undetectable in all cell types analysed. Finally, low to absent expression levels of SCN10A were observed in human atrial tissue, rabbit ventricular tissue and hiPSC-CMs. CONCLUSION: We here demonstrate the absence of functional NaV1.8 channels in non-diseased atrial and ventricular CMs. Hence, the association of SCN10A variants with cardiac electrophysiology observed in, e.g. genome wide association studies, is likely the result of indirect effects on SCN5A expression and/or NaV1.8 activity in cell types other than CMs.


Asunto(s)
Apéndice Atrial/metabolismo , Ventrículos Cardíacos/metabolismo , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.8/deficiencia , Potenciales de Acción , Animales , Apéndice Atrial/citología , Apéndice Atrial/efectos de los fármacos , Línea Celular , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cinética , Masculino , Miocitos Cardíacos/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.8/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.8/genética , Conejos , Especificidad de la Especie , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología
11.
Heart Lung Circ ; 28(6): 923-931, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29861319

RESUMEN

BACKGROUND: Left atrial appendage (LAA) is gaining increasing attention in patients with atrial fibrillation (AF) in the context of cardioembolic stroke. Galectin-3 (Gal-3) is a mediator of profibrotic pathways and is associated with an increased incidence of heart failure. However, the role of Gal-3 in LAA remodelling and thrombus formation in AF has not been evaluated. METHODS: This prospective study included 153 consecutive patients with paroxysmal (n=58), persistent (n=55) or permanent (n=40) nonvalvular AF. The serum level of Gal-3 was measured by enzyme-linked immunosorbent assay. The morphology and function of LAA were determined by transoesophageal echocardiography. RESULTS: Left atrial appendage thrombus was observed in 22 patients (2 in paroxysmal AF, 11 in persistent AF and 9 in permanent AF). Significant differences among patients with different types of AF were found in terms of LAA morphology (orifice diameter and depth) and function (flow velocity and tissue Doppler contracting velocity) as well as serum levels of Gal-3. Furthermore, patients with persistent or permanent AF had higher levels of Gal-3. High Gal-3 level was closely related to LAA flow velocity and occurrence of LAA thrombus. Multivariate logistic regression analysis revealed that Gal-3 was an independent determinant of LAA thrombus in patients with AF. Receiver operating characteristic (ROC) curves related to LAA thrombus formation established a cut-off point for Gal-3 >18.95ng/ml. CONCLUSIONS: Cardiac rhythm disturbances caused by AF may lead to morphologic and functional remodelling of LAA. The serum level of Gal-3 was significantly correlated with LAA remodelling in patients with AF. High levels of Gal-3 were also a predicator for LAA thrombus formation.


Asunto(s)
Apéndice Atrial , Fibrilación Atrial , Remodelación Atrial , Ecocardiografía Transesofágica , Galectina 3/sangre , Trombosis , Anciano , Apéndice Atrial/diagnóstico por imagen , Apéndice Atrial/metabolismo , Apéndice Atrial/fisiopatología , Fibrilación Atrial/sangre , Fibrilación Atrial/complicaciones , Fibrilación Atrial/diagnóstico por imagen , Fibrilación Atrial/fisiopatología , Proteínas Sanguíneas , Femenino , Galectinas , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Trombosis/sangre , Trombosis/diagnóstico por imagen , Trombosis/etiología , Trombosis/fisiopatología
12.
J Mol Cell Cardiol ; 121: 25-32, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29885959

RESUMEN

Atrial fibrillation (AF) following on-pump coronary artery bypass grafting (CABG) is a common condition associated with increased morbidity and mortality. We investigated the possibility that miRs may play a contributory role in postoperative AF and associated apoptosis. A total of 42 patients (31 males and 11 females, mean age 65.0 ±â€¯1.3 years) with sinus rhythm and without a history of AF were prospectively enrolled. We examined the levels of the muscle-specific miRs 1 and 133A and markers of apoptosis including TUNEL staining, caspase-3 activation, Bcl2 and Bax mRNAs in right atrial appendage (RAA) biopsies and blood plasma taken before aortic cross-clamping and after reperfusion. After reperfusion, indices of apoptosis increased the RAA. There was no change in tissue or plasma miR -1 and -133A levels compared to pre CABG. However, in patients who postoperatively developed AF (n = 14, 7 males and 7 females), compared to patients that remained in SR (n = 28, 24 males and 4 females) post CABG, tissue miR-1 increased whereas miR-133A decreased and negatively correlated with RAA apoptosis. Mechanistically, overexpression of miR-133A inhibited hypoxia-induced rat neonatal cardiomyocyte apoptosis and phosphorylated pro-survival Akt, responses abolished by a miR-133A antisense inhibitor oligonucleotide or by pre-treatment with an Akt inhibitor. In postoperative AF, differential regulation of pro- and anti-apoptotic miRs-1 and -133A respectively in the RAA, may contribute to postoperative apoptosis. These results provide new insights into molecular mechanisms of postoperative AF with potential therapeutic implications.


Asunto(s)
Apéndice Atrial/patología , Fibrilación Atrial/genética , MicroARNs/genética , Anciano , Apoptosis/genética , Apéndice Atrial/metabolismo , Fibrilación Atrial/sangre , Fibrilación Atrial/etiología , Fibrilación Atrial/patología , Biopsia , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Diferenciación Celular/genética , Puente de Arteria Coronaria/efectos adversos , Femenino , Regulación de la Expresión Génica/genética , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Humanos , Masculino , MicroARNs/sangre
13.
Am J Physiol Heart Circ Physiol ; 314(5): H895-H916, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351467

RESUMEN

Variability refers to differences in physiological function between individuals, which may translate into different disease susceptibility and treatment efficacy. Experiments in human cardiomyocytes face wide variability and restricted tissue access; under these conditions, computational models are a useful complementary tool. We conducted a computational and experimental investigation in cardiomyocytes isolated from samples of the right atrial appendage of patients undergoing cardiac surgery to evaluate the impact of variability in action potentials (APs) and subcellular ionic densities on Ca2+ transient dynamics. Results showed that 1) variability in APs and ionic densities is large, even within an apparently homogenous patient cohort, and translates into ±100% variation in ionic conductances; 2) experimentally calibrated populations of models with wide variations in ionic densities yield APs overlapping with those obtained experimentally, even if AP characteristics of the original generic model differed significantly from experimental APs; 3) model calibration with AP recordings restricts the variability in ionic densities affecting upstroke and resting potential, but redundancy in repolarization currents admits substantial variability in ionic densities; and 4) model populations constrained with experimental APs and ionic densities exhibit three Ca2+ transient phenotypes, differing in intracellular Ca2+ handling and Na+/Ca2+ membrane extrusion. These findings advance our understanding of the impact of variability in human atrial electrophysiology. NEW & NOTEWORTHY Variability in human atrial electrophysiology is investigated by integrating for the first time cellular-level and ion channel recordings in computational electrophysiological models. Ion channel calibration restricts current densities but not cellular phenotypic variability. Reduced Na+/Ca2+ exchanger is identified as a primary mechanism underlying diastolic Ca2+ fluctuations in human atrial myocytes.


Asunto(s)
Apéndice Atrial/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio , Simulación por Computador , Modelos Cardiovasculares , Miocitos Cardíacos/metabolismo , Potenciales de Acción , Anciano , Variación Biológica Poblacional , Femenino , Humanos , Cinética , Masculino , Persona de Mediana Edad , Fenotipo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Intercambiador de Sodio-Calcio/metabolismo
14.
J Cardiovasc Pharmacol ; 69(6): 369-373, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28328743

RESUMEN

Atropine (ATr) is well known as a cholinergic antagonist, however, at low concentrations ATr could paradoxically accentuate the parasympathetic actions of acetylcholine (ACh). In 22 pentobarbital anesthetized dogs, via a left and right thoracotomy, a leak-proof barrier was attached to isolate the atrial appendages (AAs) from the rest of the atria. In group 1 (Ach+ATr+Ach), ACh, 100 mM, was placed on the AA followed by the application of ATr, 2 mg/mL. The average atrial fibrillation (AF) duration was 17 ± 7 minutes. After ATr was applied to the AA and ACh again tested, the AF duration was markedly attenuated (2 ± 2 minutes, P < 0.05). In group 2 (ATr+Ach), ATr was initially applied to the AA followed by the application of ACh, 100 mM. There was no significant difference in AF duration (16 ± 4 minutes vs. 18 ± 2 minutes, P = NS). The inhibitory effect of ATr on induced HR reduction (electrical stimulation of the anterior right ganglionated plexi and vagal nerves) was similar between groups 1 and 2. These observations suggest that when ATr is initially administered it attaches to the allosteric site of the muscarinic ACh receptor (M2) leaving the orthosteric site free to be occupied by ACh. The M3 receptor that controls HR slowing does not show the same allosteric properties.


Asunto(s)
Acetilcolina/farmacología , Antiarrítmicos/farmacología , Apéndice Atrial/efectos de los fármacos , Fibrilación Atrial/tratamiento farmacológico , Atropina/farmacología , Agonistas Colinérgicos/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Antagonistas Muscarínicos/farmacología , Acetilcolina/metabolismo , Animales , Antiarrítmicos/metabolismo , Apéndice Atrial/metabolismo , Apéndice Atrial/fisiopatología , Fibrilación Atrial/etiología , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Atropina/metabolismo , Sitios de Unión , Estimulación Cardíaca Artificial , Agonistas Colinérgicos/metabolismo , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Antagonistas Muscarínicos/metabolismo , Unión Proteica , Receptor Muscarínico M2/efectos de los fármacos , Receptor Muscarínico M2/metabolismo , Factores de Tiempo
15.
Europace ; 18(10): 1521-1527, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26850746

RESUMEN

AIMS: We previously reported omega-3 polyunsaturated fatty acids (n-3PUFAs) supplementation does not reduce atrial fibrillation (AF) following coronary artery bypass graft (CABG) surgery. The aim of the present study is to evaluate the impact of n-3 PUFAs on electrocardiogram (ECG) atrial arrhythmic markers and compare with expression of gap-junction proteins, Connexins. METHODS AND RESULTS: Subset of clinical trial subjects with right atrial sampling during CABG surgery included. Twelve-lead ECG performed at recruitment and at surgery [after supplementation with n-3 PUFA (∼1.8 g/day) or matched placebo] for ∼14 days. Electrocardiograms analysed for maximum P-wave duration (P-max) and difference between P-max and minimum P-wave duration, P-wave dispersion (PWD). Right atrial specimens analysed for expression of Connexins 40 and 43 using real-time quantitative polymerase chain reaction (qPCR) and western blot. Serum levels of n-3 PUFA at baseline, at surgery, and atrial tissue levels at surgery collated from file. Postoperative AF was quantified by analysing data from stored continuous electrograms. A total of 61 patients (n-3 PUFA 34, Placebo 27) had ECG analysis and AF burden, of which 52 patients (26 in each group) had qPCR and 16 (8 in each group) had western blot analyses for Connexins 40 and 43. No difference between the two groups in ECG parameters or expression of Connexin 40 or 43. P-wave dispersion in the preoperative ECG independently predicted occurrence of AF following CABG surgery. CONCLUSIONS: Omega-3 polyunsaturated fatty acids supplementation does not alter pro-arrhythmic P-wave parameters in ECG or connexin expression in human atrium with no effect on the incidence of AF following CABG surgery.


Asunto(s)
Fibrilación Atrial/epidemiología , Conexina 43/metabolismo , Conexinas/metabolismo , Puente de Arteria Coronaria/efectos adversos , Ácidos Grasos Omega-3/administración & dosificación , Anciano , Apéndice Atrial/metabolismo , Método Doble Ciego , Electrocardiografía , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Análisis Multivariante , Complicaciones Posoperatorias/epidemiología , Cuidados Preoperatorios , ARN Mensajero/análisis , Análisis de Regresión , Reino Unido , Proteína alfa-5 de Unión Comunicante
16.
Nucleic Acids Res ; 42(6): 3515-28, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24445802

RESUMEN

Differences in methylation across tissues are critical to cell differentiation and are key to understanding the role of epigenetics in complex diseases. In this investigation, we found that locus-specific methylation differences between tissues are highly consistent across individuals. We developed a novel statistical model to predict locus-specific methylation in target tissue based on methylation in surrogate tissue. The method was evaluated in publicly available data and in two studies using the latest IlluminaBeadChips: a childhood asthma study with methylation measured in both peripheral blood leukocytes (PBL) and lymphoblastoid cell lines; and a study of postoperative atrial fibrillation with methylation in PBL, atrium and artery. We found that our method can greatly improve accuracy of cross-tissue prediction at CpG sites that are variable in the target tissue [R(2) increases from 0.38 (original R(2) between tissues) to 0.89 for PBL-to-artery prediction; from 0.39 to 0.95 for PBL-to-atrium; and from 0.81 to 0.98 for lymphoblastoid cell line-to-PBL based on cross-validation, and confirmed using cross-study prediction]. An extended model with multiple CpGs further improved performance. Our results suggest that large-scale epidemiology studies using easy-to-access surrogate tissues (e.g. blood) could be recalibrated to improve understanding of epigenetics in hard-to-access tissues (e.g. atrium) and might enable non-invasive disease screening using epigenetic profiles.


Asunto(s)
Metilación de ADN , Arterias/metabolismo , Apéndice Atrial/metabolismo , Línea Celular Transformada , Niño , Islas de CpG , Femenino , Humanos , Leucocitos/metabolismo , Masculino , Modelos Estadísticos
17.
Genet Mol Res ; 15(1)2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26909933

RESUMEN

Previous studies have used microarray technology to explore gene expression differences between the atrium and the ventricle. However, selection criteria for the differentially expressed genes (DEGs) based only on either the fold change or the P value in these studies. Here, we aim to further identify the DEGs by setting a P value threshold of <0.05 and a fold change of >2, which may yield more specific gene expression differences between the atrium and the ventricle. Gene expression profiling of the atrial appendages and the ventricular free walls in 13 normal male Sprague Dawley rats were obtained from the Gene Expression Omnibus data base (accession No.: GSE5266). DEGs between the atrial and the ventricular samples were screened using the microarray significance analysis. The underlying functions of DEGs were predicted by gene ontology and pathway enrichment analyses. In addition, we also constructed protein interactions networks, and analyzed the function modules of the interacting proteins by MCODE. A total of 757DEGs between the atria and the ventricles were found. The genes highly expressed in the ventricular myocytes were associated with muscle contraction (e.g., Myl1, Myl2, Myl3, and Myh7) and energy production (e.g., Acadm and Acsl6), while the genes preferentially expressed in the atrial myocytes were involved in the integration of neurohumoral signals (e.g., Cldn1). These conclusions were confirmed by pathway enrichment and function module analyses. Our present study provides an overview of the transcript level differences between the atrium and the ventricle, which may be useful for determination of potential biomarkers.


Asunto(s)
Apéndice Atrial/metabolismo , Ventrículos Cardíacos/metabolismo , Miocardio/metabolismo , Mapas de Interacción de Proteínas , Transcriptoma , Animales , Metabolismo Energético/genética , Perfilación de la Expresión Génica , Corazón/fisiología , Masculino , Contracción Miocárdica/genética , Especificidad de Órganos , Ratas , Ratas Sprague-Dawley
18.
Tsitologiia ; 58(5): 340-8, 2016.
Artículo en Inglés, Ruso | MEDLINE | ID: mdl-30188625

RESUMEN

Resident stem cells of the heart are denoted as heterogeneous population of immature cells, which reside in the myocardium and characterized by their ability to self-renewal and are multipotent differentiation capacity into cardiomyocyte-like and vascular like cells. CSCs were originally isolated directly by long enzymatic digestion of heart tissue and selection using stem cell markers. However, long exposure to enzymatic digestion and small myocardial sample size can affect the possibility of obtaining a significant amount of viable cells. To avoid these problems, we developed a method consisting of growing of the CPC in explant culture and subsequent immunomagnetic selection.


Asunto(s)
Apéndice Atrial , Separación Celular , Miocardio , Células Madre , Antígenos de Diferenciación/metabolismo , Apéndice Atrial/citología , Apéndice Atrial/metabolismo , Humanos , Miocardio/citología , Miocardio/metabolismo , Células Madre/citología , Células Madre/metabolismo
19.
Am J Physiol Heart Circ Physiol ; 308(12): H1564-74, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25862829

RESUMEN

Tetrandrine (TTD) is a calcium channel blocker with documented antifibrotic actions. In this study, for the first time, we identified that TTD can directly prevent in vitro human cardiac myofibroblast activation and limit in vivo myocardial fibrosis. In vitro, cardiac myofibroblasts from human atrial biopsies (N = 10) were seeded in three-dimensional collagen matrices. Cell-collagen constructs were exposed to transforming growth factor-ß1 (10 ng/ml), with or without TTD (1 and 5 µM) for 48 h. Collagen gel contraction, myofibroblast activation (α-smooth muscle actin expression), expression of profibrotic mRNAs, and rate of collagen protein synthesis were compared. TTD decreased collagen gel contraction (79.7 ± 1.3 vs 60.1 ± 8.9%, P < 0.01), α-smooth muscle actin expression (flow cytometry), collagen synthesis ([(3)H]proline incorporation), and collagen mRNA expression. Cell viability was similar between groups (annexin positive cells: 1.7 vs. 1.4%). TTD inhibited collagen gel contraction in the presence of T-type and L-type calcium channel blockers, and the intracellular calcium chelator BAPTA-AM (15 µM), suggesting that the observed effects are not mediated by calcium homeostasis. In vivo, Dahl salt-sensitive hypertensive rats were treated with variable doses of TTD (by intraperitoneal injection over 4 wk) and compared with untreated controls (N = 12). Systemic blood pressure was monitored by tail cuff. Myocardial fibrosis and left ventricular compliance were assessed by histology and passive pressure-volume analysis. Myocardial fibrosis was attenuated compared with untreated controls (%collagen area: 9.4 ± 7.3 vs 2.1 ± 1.0%, P < 0.01). Left ventricular compliance was preserved. In conclusion, TTD reverses human cardiac myofibroblast activation and myocardial fibrosis, independent of calcium channel blockade.


Asunto(s)
Apéndice Atrial/efectos de los fármacos , Bencilisoquinolinas/farmacología , Miofibroblastos/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Actinas/genética , Actinas/metabolismo , Animales , Apéndice Atrial/metabolismo , Apéndice Atrial/patología , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Quelantes del Calcio/farmacología , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Cardiomiopatías/fisiopatología , Cardiomiopatías/prevención & control , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Relación Dosis-Respuesta a Droga , Fibrosis , Geles , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/metabolismo , Hipertensión/patología , Hipertensión/fisiopatología , Masculino , Miofibroblastos/metabolismo , Miofibroblastos/patología , Ratas Endogámicas Dahl , Factores de Tiempo , Función Ventricular Izquierda/efectos de los fármacos
20.
Free Radic Biol Med ; 222: 531-538, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38977138

RESUMEN

BACKGROUND: Myocardial infarction (MI) is a significant cause of death in diabetic patients. Growing evidence suggests that mitochondrial dysfunction contributes to heart failure in diabetes. However, the molecular mechanisms of mitochondrial dysfunction mediating heart failure in diabetes are still poorly understood. METHODS: We examined MRPL12 levels in right atrial appendage tissues from diabetic patients undergoing coronary artery bypass graft (CABG) surgery. Using AC-16 cells overexpressing MRPL12 under normal and hyperglycemic conditions we performed mitochondrial functional assays OXPHOS, bioenergetics, mitochondrial membrane potential, ATP production and cell death. RESULTS: We observed elevated MRPL12 levels in heart tissue samples from diabetic patients with ischemic heart disease compared to non-diabetic patients. Overexpression of MRPL12 under hyperglycemic conditions did not affect oxidative phosphorylation (OXPHOS) levels, cellular ATP levels, or cardiomyocyte cell death. However, notable impairment in mitochondrial membrane potential (MMP) was observed under hyperglycemic conditions, along with alterations in both basal respiration oxygen consumption rate (OCR) and maximal respiratory capacity OCR. CONCLUSIONS: Overall, our results suggest that MRPL12 may have a compensatory role in the diabetic myocardium with ischemic heart disease, suggesting that MRPL12 may implicate in the pathophysiology of MI in diabetes.


Asunto(s)
Proteínas de Ciclo Celular , Potencial de la Membrana Mitocondrial , Isquemia Miocárdica , Proteínas Nucleares , Fosforilación Oxidativa , Proteínas Ribosómicas , Anciano , Animales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adenosina Trifosfato/metabolismo , Apéndice Atrial/metabolismo , Apéndice Atrial/patología , Puente de Arteria Coronaria , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/complicaciones , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Mitocondrias Cardíacas/genética , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Isquemia Miocárdica/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Ribosómicas/metabolismo , Proteínas Ribosómicas/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA