Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Circ Arrhythm Electrophysiol ; 16(11): e012199, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37933567

RESUMEN

BACKGROUND: ß-AR (ß-adrenergic receptor) stimulation regulates atrial electrophysiology and Ca2+ homeostasis via cAMP-dependent mechanisms; however, enhanced ß-AR signaling can promote atrial fibrillation (AF). CNP (C-type natriuretic peptide) can also regulate atrial electrophysiology through the activation of NPR-B (natriuretic peptide receptor B) and cGMP-dependent signaling. Nevertheless, the role of NPR-B in regulating atrial electrophysiology, Ca2+ homeostasis, and atrial arrhythmogenesis is incompletely understood. METHODS: Studies were performed using atrial samples from human patients with AF or sinus rhythm and in wild-type and NPR-B-deficient (NPR-B+/-) mice. Studies were conducted in anesthetized mice by intracardiac electrophysiology, in isolated mouse atrial preparations using high-resolution optical mapping, in isolated mouse and human atrial myocytes using patch-clamping and Ca2+ imaging, and in mouse and human atrial tissues using molecular biology. RESULTS: Atrial NPR-B protein levels were reduced in patients with AF, and NPR-B+/- mice were more susceptible to AF. Atrial cGMP levels and PDE2 (phosphodiesterase 2) activity were reduced in NPR-B+/- mice leading to larger increases in atrial cAMP in the presence of the ß-AR agonist isoproterenol. NPR-B+/- mice displayed larger increases in action potential duration and L-type Ca2+ current in the presence of isoproterenol. This resulted in the occurrence of spontaneous sarcoplasmic reticulum Ca2+ release events and delayed afterdepolarizations in NPR-B+/- atrial myocytes. Phosphorylation of the RyR2 (ryanodine receptor) and phospholamban was increased in NPR-B+/- atria in the presence of isoproterenol compared with the wildtypes. C-type natriuretic peptide inhibited isoproterenol-stimulated L-type Ca2+ current through PDE2 in mouse and human atrial myocytes. CONCLUSIONS: NPR-B protects against AF by preventing enhanced atrial responses to ß-adrenergic receptor agonists.


Asunto(s)
Fibrilación Atrial , Humanos , Ratones , Animales , Fibrilación Atrial/prevención & control , Fibrilación Atrial/metabolismo , Isoproterenol/farmacología , Péptido Natriurético Tipo-C/farmacología , Atrios Cardíacos , Miocitos Cardíacos/metabolismo
2.
JACC Basic Transl Sci ; 8(8): 922-936, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37719430

RESUMEN

Atrial fibrillation (AF) is highly prevalent in type 2 diabetes where it increases morbidity and mortality. Glucagon-like peptide (GLP)-1 receptor agonists are used in the treatment of type 2 diabetes (T2DM), but their effects on AF in T2DM are poorly understood. The present study demonstrates type 2 diabetic db/db mice are highly susceptible to AF in association with atrial electrical and structural remodeling. GLP-1, as well as the long-acting GLP-1 analogue liraglutide, reduced AF and prevented atrial remodeling in db/db mice. These data suggest that GLP-1 and related analogues could protect against AF in patients with T2DM.

3.
Cardiovasc Res ; 119(17): 2697-2711, 2023 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-37643895

RESUMEN

AIMS: The sympathetic nervous system increases HR by activating ß-adrenergic receptors (ß-ARs) and increasing cAMP in sinoatrial node (SAN) myocytes while phosphodiesterases (PDEs) degrade cAMP. Chronotropic incompetence, the inability to regulate heart rate (HR) in response to sympathetic nervous system activation, is common in hypertensive heart disease; however, the basis for this is poorly understood. The objective of this study was to determine the mechanisms leading to chronotropic incompetence in mice with angiotensin II (AngII)-induced hypertensive heart disease. METHODS AND RESULTS: C57BL/6 mice were infused with saline or AngII (2.5 mg/kg/day for 3 weeks) to induce hypertensive heart disease. HR and SAN function in response to the ß-AR agonist isoproterenol (ISO) were studied in vivo using telemetry and electrocardiography, in isolated atrial preparations using optical mapping, in isolated SAN myocytes using patch-clamping, and using molecular biology. AngII-infused mice had smaller increases in HR in response to physical activity and during acute ISO injection. Optical mapping of the SAN in AngII-infused mice demonstrated impaired increases in conduction velocity and altered conduction patterns in response to ISO. Spontaneous AP firing responses to ISO in isolated SAN myocytes from AngII-infused mice were impaired due to smaller increases in diastolic depolarization (DD) slope, hyperpolarization-activated current (If), and L-type Ca2+ current (ICa,L). These changes were due to increased localization of PDE4D surrounding ß1- and ß2-ARs in the SAN, increased SAN PDE4 activity, and reduced cAMP generation in response to ISO. Knockdown of PDE4D using a virus-delivered shRNA or inhibition of PDE4 with rolipram normalized SAN sensitivity to ß-AR stimulation in AngII-infused mice. CONCLUSIONS: AngII-induced hypertensive heart disease results in impaired HR responses to ß-AR stimulation due to up-regulation of PDE4D and reduced effects of cAMP on spontaneous AP firing in SAN myocytes.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Hipertensión , Receptores Adrenérgicos beta , Nodo Sinoatrial , Animales , Ratones , Arritmias Cardíacas , Isoproterenol/farmacología , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal , Hipertensión/inducido químicamente , Hipertensión/complicaciones , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo
4.
Can J Physiol Pharmacol ; 101(1): 27-40, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36342379

RESUMEN

Smoothelin-like 1 (SMTNL1) modulates the contractile performance of smooth muscle and thus has a key role in vascular homeostasis. Elevated vascular tone, recognized as a contributor to the development of progressive cardiac dysfunction, was previously found with SMTNL1 deletion. In this study, we assessed cardiac morphology and function of male and female, wild-type (Smtnl1+/+) and global SMTNL1 knockout (Smtnl1-/-) mice at 10 weeks of age. Gross dissection revealed distinct cardiac morphology only in males; Smtnl1-/- hearts were significantly smaller than Smtnl1+/+, but the left ventricle (LV) proportion of heart mass was greater. Male Smtnl1-/- mice also displayed increased ejection fraction and fractional shortening, as well as elevated aortic and pulmonary flow velocities. The impact of cardiac stress with pressure overload by transverse aortic constriction (TAC) was examined in male mice. With TAC banding, systolic function was preserved, but the LV filling pressure was selectively elevated due to relaxation impairment. Smtnl1-/- mice displayed higher early/passive filling velocity of LV/early mitral annulus velocity ratio (E/E' ratio) and myocardial performance index along with a prolonged isovolumetric relaxation time. Taken together, the findings support a novel, sex-dimorphic role for SMTNL1 in modulating cardiac structure and function of mice.


Asunto(s)
Proteínas Musculares , Músculo Liso , Factores Sexuales , Función Ventricular Izquierda , Animales , Femenino , Masculino , Ratones , Ratones Noqueados , Contracción Muscular , Volumen Sistólico , Proteínas Musculares/genética , Fosfoproteínas/genética
5.
J Gerontol A Biol Sci Med Sci ; 77(5): 902-908, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-34865023

RESUMEN

Heart rate (HR) is controlled by the sinoatrial node (SAN). SAN dysfunction is highly prevalent in aging; however, not all individuals age at the same rate. Rather, health status during aging is affected by frailty. Natriuretic peptides regulate SAN function in part by activating natriuretic peptide receptor C (NPR-C). The impacts of NPR-C on HR and SAN function in aging and as a function of frailty are unknown. Frailty was measured in aging wild-type and NPR-C knockout (NPR-C-/-) mice using a mouse clinical frailty index (FI). HR and SAN structure and function were investigated using intracardiac electrophysiology in anesthetized mice, high-resolution optical mapping in intact atrial preparations, histology, and molecular biology. NPR-C-/- mice rapidly became frail leading to shortened life span. HR was reduced and SAN recovery time was increased in older versus younger mice, and these changes were exacerbated in NPR-C-/- mice; however, there was substantial variability among age groups and genotypes. HR and SAN recovery time were correlated with FI score and fell along a continuum regardless of age or genotype. Optical mapping demonstrates impairments in SAN function that were also correlated with FI score. SAN fibrosis was increased in aged and NPR-C-/- mice and was graded by FI score. Loss of NPR-C results in accelerated aging and rapid decline in health status in association with impairments in HR and SAN function. Frailty assessment was effective and better able to distinguish aging-dependent changes in SAN function in the setting of shortened life span due to loss of NPR-C.


Asunto(s)
Fragilidad , Nodo Sinoatrial , Anciano , Envejecimiento/fisiología , Animales , Anciano Frágil , Humanos , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/patología , Nodo Sinoatrial/fisiología
6.
Cardiovasc Res ; 118(8): 1917-1931, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34273155

RESUMEN

AIMS: Heart rate (HR) is a critical indicator of cardiac performance that is determined by sinoatrial node (SAN) function and regulation. Natriuretic peptides, including C-type NP (CNP), have been shown to modulate ion channel function in the SAN when applied exogenously. CNP is the only NP that acts as a ligand for natriuretic peptide receptor-B (NPR-B). Despite these properties, the ability of CNP and NPR-B to regulate HR and intrinsic SAN automaticity in vivo, and the mechanisms by which it does so, are incompletely understood. Thus, the objective of this study was to determine the role of NPR-B signalling in regulating HR and SAN function. METHODS AND RESULTS: We have used NPR-B deficient mice (NPR-B+/-) to study HR regulation and SAN function using telemetry in conscious mice, intracardiac electrophysiology in anaesthetized mice, high-resolution optical mapping in isolated SAN preparations, patch-clamping in isolated SAN myocytes, and molecular biology in isolated SAN tissue. These studies demonstrate that NPR-B+/- mice exhibit slow HR, increased corrected SAN recovery time, and slowed SAN conduction. Spontaneous AP firing frequency in isolated SAN myocytes was impaired in NPR-B+/- mice due to reductions in the hyperpolarization activated current (If) and L-type Ca2+ current (ICa,L). If and ICa,L were reduced due to lower cGMP levels and increased hydrolysis of cAMP by phosphodiesterase 3 (PDE3) in the SAN. Inhibiting PDE3 or restoring cGMP signalling via application of 8-Br-cGMP abolished the reductions in cAMP, AP firing, If, and ICa,L, and normalized SAN conduction, in the SAN in NPR-B+/- mice. NPR-B+/- mice did not exhibit changes in SAN fibrosis and showed no evidence of cardiac hypertrophy or changes in ventricular function. CONCLUSIONS: NPR-B plays an essential physiological role in maintaining normal HR and SAN function by modulating ion channel function in SAN myocytes via a cGMP/PDE3/cAMP signalling mechanism.


Asunto(s)
Péptido Natriurético Tipo-C , Receptores del Factor Natriurético Atrial , Nodo Sinoatrial , Animales , GMP Cíclico , Guanilato Ciclasa , Frecuencia Cardíaca , Ratones , Péptido Natriurético Tipo-C/farmacología , Péptidos Natriuréticos , Receptores del Factor Natriurético Atrial/genética
7.
Curr Eye Res ; 47(3): 426-435, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34674590

RESUMEN

PURPOSE: Retinoblastoma is the most frequent intraocular cancer in children. It is also one of the most common causes for enucleation and carries a significant morbidity rate in affected individuals. Hence, studies on its pathophysiological and growth regulatory mechanisms are urgently needed to identify more effective novel therapeutics. METHODS: Using the Y79 retinoblastoma cell line, we investigated the electrophysiological and functional activities of the T-type voltage-gated calcium channel Cav3.1, that is constitutively expressed in these cells. We also analyzed the Akt and MAPK signaling pathways downstream of the epidermal growth factor receptor (EGFR) to understand the mechanism responsible for the inhibition of Cav3.1. RESULTS: We demonstrate that the EGFR inhibitor Afatinib significantly reduced cell viability and Cav3.1 mRNA expression and electrophysiological activity. At low concentrations (1 µM), Afatinib reduced the amplitude of Cav3.1 current density, whereas at a high concentration (10 µM), it completely abolished the voltage-gated calcium current. Our results show that inhibition of the MAPK pathway by a specific inhibitor VX-11e affected the Cav3.1 current in a dose-dependent manner. VX-11e (50 nM-1 µM) treatment reduced Cav3.1 current densities in Y79 cells, with complete abolishment of Cav3.1 current at higher concentrations (5 µM). We also demonstrate that the specific inhibition of the Akt kinase (using MK-2206) had no effect on the Cav3.1 currents. CONCLUSION: Our study provides a functional relationship between the MAPK pathway and EGFR signaling and indicates that the MAPK signaling pathway mediates the control of Cav3.1 by EGFR in retinoblastoma.


Asunto(s)
Canales de Calcio Tipo T , Receptores ErbB , Sistema de Señalización de MAP Quinasas , Neoplasias de la Retina , Retinoblastoma , Afatinib , Canales de Calcio Tipo T/genética , Canales de Calcio Tipo T/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias de la Retina/tratamiento farmacológico , Retinoblastoma/tratamiento farmacológico
8.
J Vis Exp ; (175)2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34542537

RESUMEN

This protocol has shown that the pericardium and its contents play an essential anti-fibrotic role in the ischemic rodent model (coronary ligation to induce myocardial injury). The majority of pre-clinical myocardial infarction models require the disruption of pericardial integrity with loss of the homeostatic cellular milieu. However, recently a methodology has been developed by us to induce myocardial infarction, which minimizes pericardial damage and retains the heart's resident immune cell population. An improved cardiac functional recovery in mice with an intact pericardial space following coronary ligation has been observed. This method provides an opportunity to study inflammatory responses in the pericardial space following myocardial infarction. Further development of the labeling techniques can be combined with this model to understand the fate and function of pericardial immune cells in regulating the inflammatory mechanisms that drive remodeling in the heart, including fibrosis.


Asunto(s)
Infarto del Miocardio , Roedores , Animales , Fibrosis , Ratones , Infarto del Miocardio/patología , Pericardio
9.
Heart Rhythm ; 18(1): 118-129, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32911049

RESUMEN

BACKGROUND: Atrial fibrillation (AF) is highly prevalent in diabetes mellitus (DM), yet the basis for this finding is poorly understood. Type 2 DM may be associated with unique patterns of atrial electrical and structural remodeling; however, this has not been investigated in detail. OBJECTIVE: The purpose of this study was to investigate AF susceptibility and atrial electrical and structural remodeling in type 2 diabetic db/db mice. METHODS: AF susceptibility and atrial function were assessed in male and female db/db mice and age-matched wildtype littermates. Electrophysiological studies were conducted in vivo using intracardiac electrophysiology and programmed stimulation. Atrial electrophysiology was also investigated in isolated atrial preparations using high-resolution optical mapping and in isolated atrial myocytes using patch-clamping. Molecular biology studies were performed using quantitative polymerase chain reaction and western blotting. Atrial fibrosis was assessed using histology. RESULTS: db/db mice were highly susceptible to AF in association with reduced atrial conduction velocity, action potential duration prolongation, and increased heterogeneity in repolarization in left and right atria. In db/db mice, atrial K+ currents, including the transient outward current (Ito) and the ultrarapid delayed rectifier current (IKur), were reduced. The reduction in Ito occurred in association with reductions in Kcnd2 mRNA expression and KV4.2 protein levels. The reduction in IKur was not related to gene or protein expression changes. Interstitial atrial fibrosis was increased in db/db mice. CONCLUSION: Our study demonstrates that increased susceptibility to AF in db/db mice occurs in association with impaired electrical conduction as well as electrical and structural remodeling of the atria.


Asunto(s)
Fibrilación Atrial/fisiopatología , Remodelación Atrial/fisiología , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2/complicaciones , Atrios Cardíacos/fisiopatología , Miocitos Cardíacos/metabolismo , Canales de Potasio/metabolismo , Potenciales de Acción/fisiología , Animales , Fibrilación Atrial/etiología , Fibrilación Atrial/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Femenino , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Masculino , Ratones , Ratones Endogámicos , Miocitos Cardíacos/patología , Imagen Óptica
10.
Sci Rep ; 10(1): 9459, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32528051

RESUMEN

Coronary heart disease is a leading cause of death. Tissue remodeling and fibrosis results in cardiac pump dysfunction and ischemic heart failure. Cardiac fibroblasts may rebuild damaged tissues when prompted by suitable environmental cues. Here, we use acellular biologic extracellular matrix scaffolds (bioscaffolds) to stimulate pathways of muscle repair and restore tissue function. We show that acellular bioscaffolds with bioinductive properties can redirect cardiac fibroblasts to rebuild microvascular networks and avoid tissue fibrosis. Specifically, when human cardiac fibroblasts are combined with bioactive scaffolds, gene expression is upregulated and paracrine mediators are released that promote vasculogenesis and prevent scarring. We assess these properties in rodents with myocardial infarction and observe bioscaffolds to redirect fibroblasts, reduce tissue fibrosis and prevent maladaptive structural remodeling. Our preclinical data confirms that acellular bioscaffold therapy provides an appropriate microenvironment to stimulate pathways of functional repair. We translate our observations to patients with coronary heart disease by conducting a first-in-human observational cohort study. We show that bioscaffold therapy is associated with improved perfusion of infarcted myocardium, reduced myocardial scar burden, and reverse structural remodeling. We establish that clinical use of acellular bioscaffolds is feasible and offers a new frontier to enhance surgical revascularization of ischemic heart muscle.


Asunto(s)
Fibroblastos/patología , Lesiones Cardíacas/patología , Infarto del Miocardio/patología , Miocardio/patología , Animales , Línea Celular , Cicatriz/patología , Estudios de Cohortes , Matriz Extracelular/patología , Fibrosis/patología , Corazón/fisiopatología , Humanos , Masculino , Ratas , Roedores , Andamios del Tejido , Remodelación Ventricular/fisiología
11.
Proc Natl Acad Sci U S A ; 117(14): 7990-8000, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32198206

RESUMEN

Atrial fibrillation (AF) is prevalent in diabetes mellitus (DM); however, the basis for this is unknown. This study investigated AF susceptibility and atrial electrophysiology in type 1 diabetic Akita mice using in vivo intracardiac electrophysiology, high-resolution optical mapping in atrial preparations, and patch clamping in isolated atrial myocytes. qPCR and western blotting were used to assess ion channel expression. Akita mice were highly susceptible to AF in association with increased P-wave duration and slowed atrial conduction velocity. In a second model of type 1 DM, mice treated with streptozotocin (STZ) showed a similar increase in susceptibility to AF. Chronic insulin treatment reduced susceptibility and duration of AF and shortened P-wave duration in Akita mice. Atrial action potential (AP) morphology was altered in Akita mice due to a reduction in upstroke velocity and increases in AP duration. In Akita mice, atrial Na+ current (INa) and repolarizing K+ current (IK) carried by voltage gated K+ (Kv1.5) channels were reduced. The reduction in INa occurred in association with reduced expression of SCN5a and voltage gated Na+ (NaV1.5) channels as well as a shift in INa activation kinetics. Insulin potently and selectively increased INa in Akita mice without affecting IK Chronic insulin treatment increased INa in association with increased expression of NaV1.5. Acute insulin also increased INa, although to a smaller extent, due to enhanced insulin signaling via phosphatidylinositol 3,4,5-triphosphate (PIP3). Our study reveals a critical, selective role for insulin in regulating atrial INa, which impacts susceptibility to AF in type 1 DM.


Asunto(s)
Fibrilación Atrial/metabolismo , Remodelación Atrial/fisiología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 1/complicaciones , Insulina/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/etiología , Fibrilación Atrial/fisiopatología , Remodelación Atrial/inmunología , Células Cultivadas , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animales de Enfermedad , Ecocardiografía , Electrocardiografía , Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Atrios Cardíacos/fisiopatología , Humanos , Insulina/administración & dosificación , Insulina/genética , Canal de Potasio Kv1.5/metabolismo , Masculino , Ratones , Ratones Transgénicos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Técnicas de Placa-Clamp , Potasio/metabolismo , Cultivo Primario de Células , Sodio/metabolismo , Estreptozocina/toxicidad
12.
Can J Cardiol ; 36(4): 543-553, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31837891

RESUMEN

BACKGROUND: Empagliflozin, an SGLT2 inhibitor, has shown remarkable reductions in cardiovascular mortality and heart failure admissions (EMPA-REG OUTCOME). However, the mechanism underlying the heart failure protective effects of empagliflozin remains largely unknown. Cardiac fibroblasts play an integral role in the progression of structural cardiac remodelling and heart failure, in part, by regulating extracellular matrix (ECM) homeostasis. The objective of this study was to determine if empagliflozin has a direct effect on human cardiac myofibroblast-mediated ECM remodelling. METHODS: Cardiac fibroblasts were isolated via explant culture from human atrial tissue obtained at open heart surgery. Collagen gel contraction assay was used to assess myofibroblast activity. Cell morphology and cell-mediated ECM remodelling was examined with the use of confocal microscopy. Gene expression of profibrotic markers was assessed with the use of reverse-transcription quantitative polymerase chain reaction. RESULTS: Empagliflozin significantly attenuated transforming growth factor ß1-induced fibroblast activation via collagen gel contraction after 72-hour exposure, with escalating concentrations (0.5 µmol/L, 1 µmol/L, and 5 µmol/L) resulting in greater attenuation. Morphologic assessment showed that myofibroblasts exposed to empagliflozin were smaller in size with shorter and fewer number of extensions, indicative of a more quiescent phenotype. Moreover, empagliflozin significantly attenuated cell-mediated ECM remodelling as measured by collagen fibre alignment index. Gene expression profiling revealed significant suppression of critical profibrotic markers by empagliflozin, including COL1A1, ACTA2, CTGF, FN1, and MMP-2. CONCLUSIONS: We provide novel data showing a direct effect of empagliflozin on human cardiac myofibroblast phenotype and function by attenuation of myofibroblast activity and cell-mediated collagen remodelling. These data provide critical insights into the profound effects of empagliflozin as noted in the EMPA-REG OUTCOME study.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/fisiología , Glucósidos/farmacología , Miocardio/citología , Miofibroblastos/efectos de los fármacos , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Células Cultivadas , Humanos
13.
J Thorac Cardiovasc Surg ; 157(1): 109-119.e2, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30528439

RESUMEN

OBJECTIVES: Fluoroquinolone (FQ) antibiotics are associated with adverse aortic clinical events. We assessed human aortic myofibroblast-mediated extracellular matrix (ECM) dysregulation as a possible cellular mechanism underlying FQ-associated aortopathy. METHODS: Human aortic myofibroblasts were isolated from patients with aortopathy undergoing elective ascending aortic resection (N = 9). The capacity for extracellular matrix degradation in cells exposed to FQ was assessed by multiplex analysis of secreted matrix metalloproteinases relative to tissue inhibitors of matrix metalloproteinases (TIMPs). Direct evaluation of extracellular matrix degradation was investigated in human aortic cells using a 3-dimensional gelatin-fluorescein isothiocyanate fluorescence microgel assay. Aortic cellular collagen-1 expression following FQ exposure was determined by immunoblotting and immunofluorescent staining. Cell apoptosis, necrosis, and metabolic viability was determined by annexin-V, propidium iodide staining, and water-soluble tetrazolium salt (WST1) assay. RESULTS: FQ exposure significantly decreased aortic cell TIMP-1 (P = .004) and TIMP-2 (P = .0004) protein expression compared with vehicle control. The ratio of matrix metalloproteinase-9/TIMP-2 was increased suggesting an increased capacity for extracellular matrix degradation (P = .01). In collagen gels, we show a trend toward increased aortic myofibroblast-mediated collagen fiber degradation with FQ exposure (P = .09). Similarly, FQ exposure attenuated collagen-1 expression as assessed by immunoblotting (P = .002) and immunofluorescence (P = .02). Cell apoptosis, necrosis, and metabolic viability was not significantly influenced by FQ exposure. CONCLUSIONS: For the first time, we document a putative mechanism underlying FQ-associated aortopathy whereby decreased TIMP expression with impaired compensatory collagen-1 expression results in human aortic myofibroblast-mediated extracellular matrix dysregulation. These novel data may provide a cellular and molecular mechanism to explain the established clinical association between FQ exposure and acute aortic events.


Asunto(s)
Aorta/citología , Enfermedades de la Aorta/inducido químicamente , Matriz Extracelular/efectos de los fármacos , Fluoroquinolonas/efectos adversos , Miofibroblastos/efectos de los fármacos , Aorta/efectos de los fármacos , Aorta/fisiopatología , Enfermedades de la Aorta/fisiopatología , Apoptosis/efectos de los fármacos , Células Cultivadas , Matriz Extracelular/fisiología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Masculino , Persona de Mediana Edad , Miofibroblastos/fisiología , Inhibidores Tisulares de Metaloproteinasas/antagonistas & inhibidores
14.
J Mol Cell Cardiol ; 124: 12-25, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30273558

RESUMEN

Atrial fibrillation (AF) is prevalent in hypertension and elevated angiotensin II (Ang II); however, the mechanisms by which Ang II leads to AF are poorly understood. Here, we investigated the basis for this in mice treated with Ang II or saline for 3 weeks. Ang II treatment increased susceptibility to AF compared to saline controls in association with increases in P wave duration and atrial effective refractory period, as well as reductions in right and left atrial conduction velocity. Patch-clamp studies demonstrate that action potential (AP) duration was prolonged in right atrial myocytes from Ang II treated mice in association with a reduction in repolarizing K+ currents. In contrast, APs in left atrial myocytes from Ang II treated mice showed reductions in upstroke velocity and overshoot, as well as greater prolongations in AP duration. Ang II reduced Na+ current (INa) in the left, but not the right atrium. This reduction in INa was reversible following inhibition of protein kinase C (PKC) and PKCα expression was increased selectively in the left atrium in Ang II treated mice. The transient outward K+ current (Ito) showed larger reductions in the left atrium in association with a shift in the voltage dependence of activation. Finally, Ang II caused fibrosis throughout the atria in association with changes in collagen expression and regulators of the extracellular matrix. This study demonstrates that hypertension and elevated Ang II cause distinct patterns of electrical and structural remodeling in the right and left atria that collectively create a substrate for AF.


Asunto(s)
Potenciales de Acción , Angiotensina II/metabolismo , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/etiología , Remodelación Atrial , Angiotensina II/farmacología , Animales , Biomarcadores , Presión Sanguínea , Ecocardiografía , Electrocardiografía , Inmunohistoquímica , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo
15.
PLoS One ; 12(3): e0174019, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28339476

RESUMEN

BACKGROUND: Arrhythmogenic cardiomyopathy is an inherited heart muscle disorder leading to ventricular arrhythmias and heart failure, mainly as a result of mutations in cardiac desmosomal genes. Desmosomes are cell-cell junctions mediating adhesion of cardiomyocytes; however, the molecular and cellular mechanisms underlying the disease remain widely unknown. Desmocollin-2 is a desmosomal cadherin serving as an anchor molecule required to reconstitute homeostatic intercellular adhesion with desmoglein-2. Cardiac specific lack of desmoglein-2 leads to severe cardiomyopathy, whereas overexpression does not. In contrast, the corresponding data for desmocollin-2 are incomplete, in particular from the view of protein overexpression. Therefore, we developed a mouse model overexpressing desmocollin-2 to determine its potential contribution to cardiomyopathy and intercellular adhesion pathology. METHODS AND RESULTS: We generated transgenic mice overexpressing DSC2 in cardiac myocytes. Transgenic mice developed a severe cardiac dysfunction over 5 to 13 weeks as indicated by 2D-echocardiography measurements. Corresponding histology and immunohistochemistry demonstrated fibrosis, necrosis and calcification which were mainly localized in patches near the epi- and endocardium of both ventricles. Expressions of endogenous desmosomal proteins were markedly reduced in fibrotic areas but appear to be unchanged in non-fibrotic areas. Furthermore, gene expression data indicate an early up-regulation of inflammatory and fibrotic remodeling pathways between 2 to 3.5 weeks of age. CONCLUSION: Cardiac specific overexpression of desmocollin-2 induces necrosis, acute inflammation and patchy cardiac fibrotic remodeling leading to fulminant biventricular cardiomyopathy.


Asunto(s)
Cardiomiopatías/genética , Glicoproteínas de Membrana/genética , Miocarditis/genética , Miocardio/patología , Miocitos Cardíacos/patología , Animales , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Desmocolinas , Desmosomas/metabolismo , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , Miocarditis/metabolismo , Miocarditis/patología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Regulación hacia Arriba
16.
JACC Basic Transl Sci ; 2(4): 450-464, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30062163

RESUMEN

Structural cardiac remodeling after ischemic injury can induce a transition to heart failure from progressive loss of cardiac function. Cellular regenerative therapies are promising but face significant translational hurdles. Tissue extracellular matrix (ECM) holds the necessary environmental cues to stimulate cell-based endogenous myocardial repair pathways and promote adaptive remodeling toward functional recovery. Heart epicardium has emerged as an important anatomic niche for endogenous repair pathways including vasculogenesis and cardiogenesis. We show that acellular ECM scaffolds surgically implanted on the epicardium following myocardial infarction (MI) can attenuate structural cardiac remodeling and improve functional recovery. We assessed the efficacy of this strategy on post-MI functional recovery by comparing intact bioactive scaffolds with biologically inactivated ECM scaffolds. We confirm that bioactive properties within the acellular ECM biomaterial are essential for the observed functional benefits. We show that interaction of human cardiac fibroblasts with bioactive ECM can induce a robust cell-mediated vasculogenic paracrine response capable of functional blood vessel assembly. Fibroblast growth factor-2 is uncovered as a critical regulator of this novel bioinductive effect. Acellular bioactive ECM scaffolds surgically implanted on the epicardium post-MI can reprogram resident fibroblasts and stimulate adaptive pro-reparative pathways enhancing functional recovery. We introduce a novel surgical strategy for tissue repair that can be performed as an adjunct to conventional surgical revascularization with minimal translational challenges.

17.
Artículo en Inglés | MEDLINE | ID: mdl-27154360

RESUMEN

BACKGROUND: We have recently shown that Calanus oil, which is extracted from the marine copepod Calanus finmarchicus, reduces fat deposition, suppresses adipose tissue inflammation and improves insulin sensitivity in high fat-fed rodents. This study expands upon our previous observations by examining whether dietary supplementation with Calanus oil could antagonize angiotensin II (Ang II)-induced hypertension and ventricular remodeling in mice given a high fat diet (HFD). METHODS: C57BL/6J mice were initially subjected to 8 weeks of HFD with or without 2% (w/w) Calanus oil. Thereafter, animals within each group were randomized for the administration of either Ang II (1µg/kg/min) or saline for another two weeks, while still on the same dietary regimen. RESULTS: Ang II caused a marked decline in body and organ weights in mice receiving non-supplemented HFD, a response which was clearly attenuated in mice receiving Calanus oil supplementation. Furthermore, Ang II-induced elevation in blood pressure was also attenuated in the Calanus oil-supplemented group. As expected, infusion of Ang II produced hypertrophy and up-regulation of marker genes (mRNA level) of both hypertrophy and fibrosis in cardiac muscle, but this response was unaffected by dietary Calanus oil. Fibrosis and inflammation were up-regulated also in the aorta following Ang II infusion. However, the inflammatory response was blocked by Calanus oil supplementation. A final, and unexpected, finding was that dietary intake of Calanus oil caused a robust increase in the level of O-GlcNAcylation in cardiac tissue. CONCLUSION: These results suggest that dietary intake of oil from the marine copepod Calanus finmarchicus could be a beneficial addition to conventional hypertension treatment. The compound attenuates inflammation and the severe metabolic stress caused by Ang II infusion. Although the present study suggests that the anti-hypertensive effect of the oil (or its n-3 PUFAs constituents) is related to its anti-inflammatory action in the vessel wall, other mechanisms such as interaction with intracellular calcium mechanisms or a direct antagonistic effect on Ang II receptors should be examined.


Asunto(s)
Angiotensina II/efectos adversos , Antiinflamatorios/administración & dosificación , Grasas Insaturadas en la Dieta/administración & dosificación , Hipertensión/dietoterapia , Animales , Antiinflamatorios/farmacología , Aorta/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Copépodos/química , Dieta Alta en Grasa , Grasas Insaturadas en la Dieta/farmacología , Hipertensión/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Tamaño de los Órganos/efectos de los fármacos , Distribución Aleatoria , Resultado del Tratamiento
18.
J Am Coll Cardiol ; 66(8): 892-900, 2015 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-26293758

RESUMEN

BACKGROUND: Suspected genetic causes for extracellular matrix (ECM) dysregulation in the ascending aorta in patients with bicuspid aortic valves (BAV) have influenced strategies and thresholds for surgical resection of BAV aortopathy. Using 4-dimensional (4D) flow cardiac magnetic resonance imaging (CMR), we have documented increased regional wall shear stress (WSS) in the ascending aorta of BAV patients. OBJECTIVES: This study assessed the relationship between WSS and regional aortic tissue remodeling in BAV patients to determine the influence of regional WSS on the expression of ECM dysregulation. METHODS: BAV patients (n = 20) undergoing ascending aortic resection underwent pre-operative 4D flow CMR to regionally map WSS. Paired aortic wall samples (i.e., within-patient samples obtained from regions of elevated and normal WSS) were collected and compared for medial elastin degeneration by histology and ECM regulation by protein expression. RESULTS: Regions of increased WSS showed greater medial elastin degradation compared to adjacent areas with normal WSS: decreased total elastin (p = 0.01) with thinner fibers (p = 0.00007) that were farther apart (p = 0.001). Multiplex protein analyses of ECM regulatory molecules revealed an increase in transforming growth factor ß-1 (p = 0.04), matrix metalloproteinase (MMP)-1 (p = 0.03), MMP-2 (p = 0.06), MMP-3 (p = 0.02), and tissue inhibitor of metalloproteinase-1 (p = 0.04) in elevated WSS regions, indicating ECM dysregulation in regions of high WSS. CONCLUSIONS: Regions of increased WSS correspond with ECM dysregulation and elastic fiber degeneration in the ascending aorta of BAV patients, implicating valve-related hemodynamics as a contributing factor in the development of aortopathy. Further study to validate the use of 4D flow CMR as a noninvasive biomarker of disease progression and its ability to individualize resection strategies is warranted.


Asunto(s)
Aorta/fisiopatología , Válvula Aórtica/anomalías , Enfermedades de las Válvulas Cardíacas/fisiopatología , Adulto , Aorta/metabolismo , Válvula Aórtica/metabolismo , Válvula Aórtica/fisiopatología , Enfermedad de la Válvula Aórtica Bicúspide , Estudios de Casos y Controles , Elastina/metabolismo , Femenino , Enfermedades de las Válvulas Cardíacas/metabolismo , Hemodinámica , Humanos , Imagen por Resonancia Magnética , Masculino , Metaloproteinasas de la Matriz/metabolismo , Persona de Mediana Edad , Estrés Mecánico , Inhibidores Tisulares de Metaloproteinasas/metabolismo
19.
Appl Physiol Nutr Metab ; 40(7): 749-52, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25962839

RESUMEN

Cecal microbiota from type 2 diabetic (db/db) and control (db/(+)) mice was obtained following 6 weeks of sedentary or exercise activity. qPCR analysis revealed a main effect of exercise, with greater abundance of select Firmicutes species and lower Bacteroides/Prevotella spp. in both normal and diabetic exercised mice compared with sedentary counterparts. Conversely, Bifidobacterium spp. was greater in exercised normal but not diabetic mice (exercise × diabetes interaction). How exercise influences gut microbiota requires further investigation.


Asunto(s)
Diabetes Mellitus Experimental/microbiología , Microbioma Gastrointestinal , Condicionamiento Físico Animal/estadística & datos numéricos , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL
20.
J Transl Med ; 13: 147, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25948488

RESUMEN

BACKGROUND: Tissue fibrosis and chamber remodeling is a hallmark of the failing heart and the final common pathway for heart failure of diverse etiologies. Sustained elevation of pro-fibrotic cytokine transforming growth factor-beta1 (TGFß1) induces cardiac myofibroblast-mediated fibrosis and progressive structural tissue remodeling. OBJECTIVES: We examined the effects of low molecular weight fibroblast growth factor (LMW-FGF-2) on human cardiac myofibroblast-mediated extracellular matrix (ECM) dysregulation and remodeling. METHODS: Human cardiac biopsies were obtained during open-heart surgery and myofibroblasts were isolated, passaged, and seeded within type I collagen matrices. To induce myofibroblast activation and ECM remodeling, myofibroblast-seeded collagen gels were exposed to TGFß1. The extent of ECM contraction, myofibroblast activation, ECM dysregulation, and cell apoptosis was determined in the presence of LMW-FGF-2 and compared to its absence. Using a novel floating nylon-grid supported thin collagen gel culture platform system, myofibroblast activation and local ECM remodeling around isolated single cells was imaged using confocal microscopy and quantified by image analysis. RESULTS: TGFß1 induced significant myofibroblast activation and ECM dysregulation as evidenced by collagen gel contraction, structural ECM remodeling, collagen synthesis, ECM degradation, and altered TIMP expression. LMW-FGF-2 significantly attenuated TGFß1 induced myofibroblast-mediated ECM remodeling. These observations were similar using either ventricular or atrial-derived cardiac myofibroblasts. In addition, for the first time using individual cells, LMW-FGF-2 was observed to attenuate cardiac myofibroblast activation and prevent local cell-mediated ECM perturbations. CONCLUSIONS: LMW-FGF-2 attenuates human cardiac myofibroblast-mediated ECM remodeling and may prevent progressive maladaptive chamber remodeling and tissue fibrosis for patients with diverse structural heart diseases.


Asunto(s)
Matriz Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Miofibroblastos/metabolismo , Apoptosis , Biopsia , Diferenciación Celular , Colágeno/metabolismo , Femenino , Fibrosis , Corazón/fisiología , Insuficiencia Cardíaca/fisiopatología , Humanos , Inmunohistoquímica , Masculino , Microscopía Confocal , Miocardio/metabolismo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...