Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Adv Ther ; 41(1): 130-151, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37851297

RESUMEN

INTRODUCTION: Pulmonary arterial hypertension (PAH) is a rare, progressive disease associated with significant morbidity and mortality. The phase 3 STELLAR trial tested sotatercept plus background therapy (BGT) versus placebo plus BGT. BGT was comprised of mono-, double-, or triple-PAH targeted therapy. Building on STELLAR findings, we employed a population health model to assess the potential long-term clinical impact of sotatercept. METHODS: Based on the well-established ESC/ERS 4-strata risk assessment approach, we developed a six-state Markov-type model (low risk, intermediate-low risk, intermediate-high risk, high risk, lung/heart-lung transplant, and death) to compare the clinical outcomes of sotatercept plus BGT versus BGT alone over a lifetime horizon. State-transition probabilities were obtained from STELLAR. Risk stratum-adjusted mortality and lung/heart-lung transplant probabilities were based on COMPERA PAH registry data, and the post-transplant mortality probability was obtained from existing literature. Model outcomes were discounted at 3% annually. Sensitivity analyses were conducted to examine model robustness. RESULTS: In the base case, sotatercept plus BGT was associated with longer life expectancy from model baseline (16.5 vs 5.1 years) versus BGT alone, leading to 11.5 years gained per patient. Compared with BGT alone, sotatercept plus BGT was further associated with a gain in infused prostacyclin-free life years per patient, along with 683 PAH hospitalizations and 4 lung/heart-lung transplant avoided per 1000 patients. CONCLUSIONS: According to this model, adding sotatercept to BGT increased life expectancy by roughly threefold among patients with PAH while reducing utilization of infused prostacyclin, PAH hospitalizations, and lung/heart-lung transplants. Real-world data are needed to confirm these findings. TRIAL REGISTRATION: ClinicalTrials.gov identifier, NCT04576988 (STELLAR).


Asunto(s)
Hipertensión Arterial Pulmonar , Humanos , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Proteínas Recombinantes de Fusión/efectos adversos , Medición de Riesgo , Morbilidad
2.
Gene Ther ; 30(7-8): 543-551, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-35102273

RESUMEN

Ischemic cardiomyopathy is a leading cause of death and an unmet clinical need. Adeno-associated virus (AAV) gene-based therapies hold great promise for treating and preventing heart failure. Previously we showed that muscle A-kinase Anchoring Protein ß (mAKAPß, AKAP6ß), a scaffold protein that organizes perinuclear signalosomes in the cardiomyocyte, is a critical regulator of pathological cardiac hypertrophy. Here, we show that inhibition of mAKAPß expression in stressed adult cardiomyocytes in vitro was cardioprotective, while conditional cardiomyocyte-specific mAKAP gene deletion in mice prevented pathological cardiac remodeling due to myocardial infarction. We developed a new self-complementary serotype 9 AAV gene therapy vector expressing a short hairpin RNA for mAKAPß under the control of a cardiomyocyte-specific promoter (AAV9sc.shmAKAP). This vector efficiently downregulated mAKAPß expression in the mouse heart in vivo. Expression of the shRNA also inhibited mAKAPß expression in human induced cardiomyocytes in vitro. Following myocardial infarction, systemic administration of AAV9sc.shmAKAP prevented the development of pathological cardiac remodeling and heart failure, providing long-term restoration of left ventricular ejection fraction. Our findings provide proof-of-concept for mAKAPß as a therapeutic target for ischemic cardiomyopathy and support the development of a translational pipeline for AAV9sc.shmAKAP for the treatment of heart failure.


Asunto(s)
Cardiomiopatías , Insuficiencia Cardíaca , Infarto del Miocardio , Ratones , Humanos , Animales , Volumen Sistólico , Remodelación Ventricular/genética , Función Ventricular Izquierda , Miocitos Cardíacos/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/terapia , ARN Interferente Pequeño/genética , Infarto del Miocardio/genética , Infarto del Miocardio/terapia , Infarto del Miocardio/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/terapia
3.
Sci Rep ; 12(1): 7326, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35513431

RESUMEN

Fibroblast growth factor (FGF) 21, a hormone that increases insulin sensitivity, has shown promise as a therapeutic agent to improve metabolic dysregulation. Here we report that FGF21 directly targets cardiac myocytes by binding ß-klotho and FGF receptor (FGFR) 4. In combination with high glucose, FGF21 induces cardiac myocyte growth in width mediated by extracellular signal-regulated kinase 1/2 (ERK1/2) signaling. While short-term FGF21 elevation can be cardio-protective, we find that in type 2 diabetes (T2D) in mice, where serum FGF21 levels are elevated, FGFR4 activation induces concentric cardiac hypertrophy. As T2D patients are at risk for heart failure with preserved ejection fraction (HFpEF), we propose that induction of concentric hypertrophy by elevated FGF21-FGFR4 signaling may constitute a novel mechanism promoting T2D-associated HFpEF such that FGFR4 blockade might serve as a cardio-protective therapy in T2D. In addition, potential adverse cardiac effects of FGF21 mimetics currently in clinical trials should be investigated.


Asunto(s)
Diabetes Mellitus Tipo 2 , Insuficiencia Cardíaca , Animales , Cardiomegalia/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Factores de Crecimiento de Fibroblastos/metabolismo , Insuficiencia Cardíaca/metabolismo , Humanos , Ratones , Miocitos Cardíacos/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Volumen Sistólico
4.
Circulation ; 142(22): 2138-2154, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32933333

RESUMEN

BACKGROUND: Concentric and eccentric cardiac hypertrophy are associated with pressure and volume overload, respectively, in cardiovascular disease both conferring an increased risk of heart failure. These contrasting forms of hypertrophy are characterized by asymmetrical growth of the cardiac myocyte in mainly width or length, respectively. The molecular mechanisms determining myocyte preferential growth in width versus length remain poorly understood. Identification of the mechanisms governing asymmetrical myocyte growth could provide new therapeutic targets for the prevention or treatment of heart failure. METHODS: Primary adult rat ventricular myocytes, adeno-associated virus (AAV)-mediated gene delivery in mice, and human tissue samples were used to define a regulatory pathway controlling pathological myocyte hypertrophy. Chromatin immunoprecipitation assays with sequencing and precision nuclear run-on sequencing were used to define a transcriptional mechanism. RESULTS: We report that asymmetrical cardiac myocyte hypertrophy is modulated by SRF (serum response factor) phosphorylation, constituting an epigenomic switch balancing the growth in width versus length of adult ventricular myocytes in vitro and in vivo. SRF Ser103 phosphorylation is bidirectionally regulated by RSK3 (p90 ribosomal S6 kinase type 3) and PP2A (protein phosphatase 2A) at signalosomes organized by the scaffold protein mAKAPß (muscle A-kinase anchoring protein ß), such that increased SRF phosphorylation activates AP-1 (activator protein-1)-dependent enhancers that direct myocyte growth in width. AAV are used to express in vivo mAKAPß-derived RSK3 and PP2A anchoring disruptor peptides that block the association of the enzymes with the mAKAPß scaffold. Inhibition of RSK3 signaling prevents concentric cardiac remodeling induced by pressure overload, while inhibition of PP2A signaling prevents eccentric cardiac remodeling induced by myocardial infarction, in each case improving cardiac function. SRF Ser103 phosphorylation is significantly decreased in dilated human hearts, supporting the notion that modulation of the mAKAPß-SRF signalosome could be a new therapeutic approach for human heart failure. CONCLUSIONS: We have identified a new molecular switch, namely mAKAPß signalosome-regulated SRF phosphorylation, that controls a transcriptional program responsible for modulating changes in cardiac myocyte morphology that occur secondary to pathological stressors. Complementary AAV-based gene therapies constitute rationally-designed strategies for a new translational modality for heart failure.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Aumento de la Célula , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/metabolismo , Factor de Respuesta Sérica/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Adenoviridae/genética , Animales , Animales Recién Nacidos , Células Cultivadas , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/patología , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley
5.
Circulation ; 142(10): 948-962, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32611257

RESUMEN

BACKGROUND: The Ca2+/calmodulin-dependent phosphatase calcineurin is a key regulator of cardiac myocyte hypertrophy in disease. An unexplained paradox is how the ß isoform of the calcineurin catalytic A-subunit (CaNAß) is required for induction of pathological myocyte hypertrophy, despite calcineurin Aα expression in the same cells. It is unclear how the pleiotropic second messenger Ca2+ drives excitation-contraction coupling while not stimulating hypertrophy by calcineurin in the normal heart. Elucidation of the mechanisms conferring this selectivity in calcineurin signaling should reveal new strategies for targeting the phosphatase in disease. METHODS: Primary adult rat ventricular myocytes were studied for morphology and intracellular signaling. New Förster resonance energy transfer reporters were used to assay Ca2+ and calcineurin activity in living cells. Conditional gene deletion and adeno-associated virus-mediated gene delivery in the mouse were used to study calcineurin signaling after transverse aortic constriction in vivo. RESULTS: CIP4 (Cdc42-interacting protein 4)/TRIP10 (thyroid hormone receptor interactor 10) was identified as a new polyproline domain-dependent scaffold for CaNAß2 by yeast 2-hybrid screen. Cardiac myocyte-specific CIP4 gene deletion in mice attenuated pressure overload-induced pathological cardiac remodeling and heart failure. Blockade of CaNAß polyproline-dependent anchoring using a competing peptide inhibited concentric hypertrophy in cultured myocytes; disruption of anchoring in vivo using an adeno-associated virus gene therapy vector inhibited cardiac hypertrophy and improved systolic function after pressure overload. Live cell Förster resonance energy transfer biosensor imaging of cultured myocytes revealed that Ca2+ levels and calcineurin activity associated with the CIP4 compartment were increased by neurohormonal stimulation, but minimally by pacing. Conversely, Ca2+ levels and calcineurin activity detected by nonlocalized Förster resonance energy transfer sensors were induced by pacing and minimally by neurohormonal stimulation, providing functional evidence for differential intracellular compartmentation of Ca2+ and calcineurin signal transduction. CONCLUSIONS: These results support a structural model for Ca2+ and CaNAß compartmentation in cells based on an isoform-specific mechanism for calcineurin protein-protein interaction and localization. This mechanism provides an explanation for the specific role of CaNAß in hypertrophy and its selective activation under conditions of pathologic stress. Disruption of CaNAß polyproline-dependent anchoring constitutes a rational strategy for therapeutic targeting of CaNAß-specific signaling responsible for pathological cardiac remodeling in cardiovascular disease deserving of further preclinical investigation.


Asunto(s)
Calcineurina/metabolismo , Cardiomegalia/enzimología , Ventrículos Cardíacos/enzimología , Miocitos Cardíacos/enzimología , Animales , Calcineurina/genética , Cardiomegalia/genética , Cardiomegalia/patología , Ventrículos Cardíacos/patología , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Miocitos Cardíacos/patología
6.
Sci Rep ; 10(1): 12368, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32704023

RESUMEN

Preterm infants with bronchopulmonary dysplasia (BPD) and pulmonary hypertension (PH) have accelerated lung aging and poor long-term outcomes. Klotho is an antiaging protein that modulates oxidative stress, angiogenesis and fibrosis. Here we test the hypothesis that decreased cord Klotho levels in preterm infants predict increased BPD-PH risk and early Klotho supplementation prevents BPD-like phenotype and PH in rodents exposed to neonatal hyperoxia. In experiment 1, Klotho levels were measured in cord blood of preterm infants who were enrolled in a longitudinal cohort study. In experiment 2, using an experimental BPD-PH model, rat pups exposed to room air or hyperoxia (85% O2) were randomly assigned to receive every other day injections of recombinant Klotho or placebo. The effect of Klotho on lung structure, PH and cardiac function was assessed. As compared to controls, preterm infants with BPD or BPD-PH had decreased cord Klotho levels. Early Klotho supplementation in neonatal hyperoxia-exposed rodents preserved lung alveolar and vascular structure, attenuated PH, reduced pulmonary vascular remodeling and improved cardiac function. Together, these findings have important implications as they suggest that perinatal Klotho deficiency contributes to BPD-PH risk and strategies that preserve Klotho levels, may improve long-term cardiopulmonary outcomes in preterm infants.


Asunto(s)
Displasia Broncopulmonar/sangre , Sangre Fetal/metabolismo , Glucuronidasa/sangre , Hipertensión Pulmonar/sangre , Recien Nacido Prematuro/sangre , Biomarcadores/sangre , Displasia Broncopulmonar/fisiopatología , Femenino , Humanos , Hipertensión Pulmonar/fisiopatología , Recién Nacido , Proteínas Klotho , Estudios Longitudinales , Masculino
7.
Physiol Rep ; 8(1): e14334, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31925922

RESUMEN

Supplemental oxygen (O2 ) therapy in preterm infants impairs lung development, but the impact of O2 on long-term systemic vascular structure and function has not been well-explored. The present study tested the hypothesis that neonatal O2 therapy induces long-term structural and functional alterations in the systemic vasculature, resulting in vascular stiffness observed in children and young adults born preterm. Newborn Sprague-Dawley rats were exposed to normoxia (21% O2 ) or hyperoxia (85% O2 ) for 1 and 3 weeks. A subgroup exposed to 3 weeks hyperoxia was recovered in normoxia for an additional 3 weeks. Aortic stiffness was assessed by pulse wave velocity (PWV) using Doppler ultrasound and pressure myography. Aorta remodeling was assessed by collagen deposition and expression. Left ventricular (LV) function was assessed by echocardiography. We found that neonatal hyperoxia exposure increased vascular stiffness at 3 weeks, which persisted after normoxic recovery at 6 weeks of age. These findings were accompanied by increased PWV, aortic remodeling, and altered LV function as evidenced by decreased ejection fraction, cardiac output, and stroke volume. Importantly, these functional changes were associated with increased collagen deposition in the aorta. Together, these findings demonstrate that neonatal hyperoxia induces early and sustained biomechanical alterations in the systemic vasculature and impairs LV function. Early identification of preterm infants who are at risk of developing systemic vascular dysfunction will be crucial in developing targeted prevention strategies that may improve the long-term cardiovascular outcomes in this vulnerable population.


Asunto(s)
Aorta/fisiopatología , Hiperoxia/fisiopatología , Terapia por Inhalación de Oxígeno/efectos adversos , Remodelación Vascular/fisiología , Rigidez Vascular/fisiología , Disfunción Ventricular Izquierda/fisiopatología , Animales , Animales Recién Nacidos , Presión Arterial , Fenómenos Biomecánicos , Peso Corporal , Gasto Cardíaco , Ecocardiografía , Femenino , Hiperoxia/complicaciones , Masculino , Mortalidad , Miografía , Análisis de la Onda del Pulso , Ratas , Ratas Sprague-Dawley , Volumen Sistólico , Ultrasonografía Doppler , Disfunción Ventricular Izquierda/etiología
8.
JACC Basic Transl Sci ; 3(2): 163-175, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30062203

RESUMEN

We identified a plasma signature of 11 C14 to C26 ceramides and 1 C16 dihydroceramide predictive of major adverse cardiovascular events in patients with acute myocardial infarction (AMI). Among patients undergoing coronary artery bypass surgery, those with recent AMI, compared with those without recent AMI, showed a significant increase in 5 of the signature's 12 ceramides in plasma but not simultaneously-biopsied aortic tissue. In contrast, a rat AMI model, compared with sham control, showed a significant increase in myocardial concentrations of all 12 ceramides and up-regulation of 3 ceramide-producing enzymes, suggesting ischemic myocardium as a possible source of this ceramide signature.

9.
J Mol Cell Cardiol ; 112: 27-39, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28865712

RESUMEN

RATIONALE: Myocardial infarction (MI) triggers a dynamic microRNA response with the potential of yielding therapeutic targets. OBJECTIVE: We aimed to identify novel aberrantly expressed cardiac microRNAs post-MI with potential roles in adverse remodeling in a rat model, and to provide post-ischemic therapeutic inhibition of a candidate pathological microRNA in vivo. METHODS AND RESULTS: Following microRNA array profiling in rat hearts 2 and 14days post-MI, we identified a time-dependent up-regulation of miR-31 compared to sham-operated rats. A progressive increase of miR-31 (up to 91.4±11.3 fold) was detected in the infarcted myocardium by quantitative real-time PCR. Following target prediction analysis, reporter gene assays confirmed that miR-31 targets the 3´UTR of cardiac troponin-T (Tnnt2), E2F transcription factor 6 (E2f6), mineralocorticoid receptor (Nr3c2) and metalloproteinase inhibitor 4 (Timp4) mRNAs. In vitro, hypoxia and oxidative stress up-regulated miR-31 and suppressed target genes in cardiac cell cultures, whereas LNA-based oligonucleotide inhibition of miR-31 (miR-31i) reversed its repressive effect on target mRNAs. Therapeutic post-ischemic administration of miR-31i in rats silenced cardiac miR-31 and enhanced expression of target genes, while preserving cardiac structure and function at 2 and 4weeks post-MI. Left ventricular ejection fraction (EF) improved by 10% (from day 2 to 30 post-MI) in miR-31i-treated rats, whereas controls receiving scrambled LNA inhibitor or placebo incurred a 17% deterioration in EF. miR-31i decreased end-diastolic pressure and infarct size; attenuated interstitial fibrosis in the remote myocardium and enhanced cardiac output. CONCLUSION: miR-31 induction after MI is deleterious to cardiac function while its therapeutic inhibition in vivo ameliorates cardiac dysfunction and prevents the development of post-ischemic adverse remodeling.


Asunto(s)
MicroARNs/metabolismo , Isquemia Miocárdica/genética , Remodelación Ventricular/genética , Animales , Secuencia de Bases , Hipoxia de la Célula/genética , Línea Celular , Perfilación de la Expresión Génica , Silenciador del Gen/efectos de los fármacos , Masculino , Isquemia Miocárdica/patología , Miocardio/metabolismo , Oligonucleótidos/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Ratas , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Remodelación Ventricular/efectos de los fármacos
10.
J Mol Cell Cardiol ; 93: 98-105, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26940993

RESUMEN

Noonan syndrome (NS) is a congenital disorder resulting from mutations of the Ras-Raf signaling pathway. Hypertrophic cardiomyopathy associated with RAF1 "RASopathy" mutations is a major risk factor for heart failure and death in NS and has been attributed to activation of MEK1/2-ERK1/2 mitogen-activated protein kinases. We recently discovered that type 3 p90 ribosomal S6 kinase (RSK3) is an ERK effector that is required, like ERK1/2, for concentric myocyte hypertrophy in response to pathological stress such as pressure overload. In order to test whether RSK3 also contributes to NS-associated hypertrophic cardiomyopathy, RSK3 knock-out mice were crossed with mice bearing the Raf1(L613V) human NS mutation. We confirmed that Raf1(L613V) knock-in confers a NS-like phenotype, including cardiac hypertrophy. Active RSK3 was increased in Raf1(L613V) mice. Constitutive RSK3 gene deletion prevented the Raf1(L613V)-dependent concentric growth in width of the cardiac myocyte and attenuated cardiac hypertrophy in female mice. These results are consistent with RSK3 being an important mediator of ERK1/2-dependent growth in RASopathy. In conjunction with previously published data showing that RSK3 is important for pathological remodeling of the heart, these data suggest that targeting of this downstream MAP-kinase pathway effector should be considered in the treatment of RASopathy-associated hypertrophic cardiomyopathy.


Asunto(s)
Cardiomiopatía Hipertrófica/etiología , Síndrome de Noonan/complicaciones , Síndrome de Noonan/genética , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Animales , Cardiomiopatía Hipertrófica/diagnóstico , Cardiomiopatía Hipertrófica/metabolismo , Modelos Animales de Enfermedad , Ecocardiografía , Activación Enzimática , Femenino , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Noqueados , Síndrome de Noonan/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Remodelación Ventricular
11.
J Mol Cell Cardiol ; 87: 27-37, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26254181

RESUMEN

AIMS: Myocardial infarction followed by adverse left ventricular (LV) remodeling is the most frequent proximate cause of heart failure. Hydrogen sulfide (H2S) is an important endogenous modulator of diverse physiological and pathophysiological processes. Its role in post-ischemic ventricular remodeling and the associated neurohormonal responses has not been defined. Here, we aimed at evaluating whether the slow-releasing water-soluble H2S donor GYY4137 (GYY) exerts cardioprotective effects and modulates the neurohormonal response to cardiac ischemic injury. METHODS AND RESULTS: Treatment for 2 or 7 days with GYY (100 mg/Kg/48 h, IP) after acute myocardial infarction (MI) in rats preserved LV dimensions and function in vivo, compared to untreated infarcted (MI), placebo- and dl-propargylglycine- (PAG, an inhibitor of endogenous H2S synthesis) treated animals (n=9/group/time-point). LV dimensions and function in GYY-treated animals were comparable to healthy sham-operated rats. GYY-treated hearts had significantly less LV fibrosis than MI, placebo and PAG hearts. A higher density of blood vessels was found in the LV scar area of GYY-treated animals compared to all other infarcted groups. Despite preserved LV structure and function, treatment with GYY increased the levels of the natriuretic peptides ANP and BNP in association with enhanced cyclic GMP levels, paralleled by higher cGMP-dependent protein kinase type I (cGKI) protein levels. CONCLUSIONS: Our data suggest that the slow-releasing H2S donor, GYY4137, preserves cardiac function, attenuates adverse remodeling and may exert post-ischemic cardioprotective (pro-angiogenic, anti-apoptotic, anti-hypertrophic and anti-fibrotic) effects in part through enhanced early post-ischemic endogenous natriuretic peptide activation.


Asunto(s)
Factor Natriurético Atrial/metabolismo , Sulfuro de Hidrógeno/metabolismo , Isquemia/tratamiento farmacológico , Infarto del Miocardio/tratamiento farmacológico , Péptido Natriurético Encefálico/metabolismo , Animales , Cardiotónicos/administración & dosificación , Humanos , Isquemia/fisiopatología , Morfolinas/administración & dosificación , Infarto del Miocardio/fisiopatología , Compuestos Organotiofosforados/administración & dosificación , Ratas , Disfunción Ventricular Izquierda/tratamiento farmacológico , Disfunción Ventricular Izquierda/fisiopatología , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/fisiología
12.
IUBMB Life ; 67(5): 331-7, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25988524

RESUMEN

The family of p90 ribosomal S6 kinases (RSKs) are pleiotropic effectors for extracellular signal-regulated kinase signaling pathways. Recently, RSK3 was shown to be important for pathological remodeling of the heart. Although cardiac myocyte hypertrophy can be compensatory for increased wall stress, in chronic heart diseases, this nonmitotic cell growth is usually associated with interstitial fibrosis, increased cell death, and decreased cardiac function. Although RSK3 is less abundant in the cardiac myocyte than other RSK family members, RSK3 appears to serve a unique role in cardiac myocyte stress responses. A potential mechanism conferring the unique function of RSK3 in the heart is anchoring by the scaffold protein muscle A-kinase anchoring protein ß (mAKAPß). Recent findings suggest that RSK3 should be considered as a therapeutic target for the prevention of heart failure, a clinical syndrome of major public health significance.


Asunto(s)
Miocitos Cardíacos/patología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Animales , Humanos , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Estrés Fisiológico
13.
Biomaterials ; 53: 12-24, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25890702

RESUMEN

The limited efficacy of cardiac cell-based therapy is thought to be due to poor cell retention within the myocardium. Hence, there is an urgent need for biomaterials that aid in long-term cell retention. This study describes the development of injectable microcapsules for the delivery of mesenchymal stem cells (MSCs) into the infarcted cardiac wall. These microcapsules comprise of low concentrations of agarose supplemented with extracellular matrix (ECM) proteins collagen and fibrin. Dextran sulfate, a negatively charged polycarbohydrate, was added to mimic glycosaminoglycans in the ECM. Cell viability assays showed that a combination of all components is necessary to support long-term survival and proliferation of MSCs within microcapsules. Following intramyocardial transplantation, microcapsules degraded slowly in vivo and did not induce a fibrotic foreign body response. Pre-labeling of encapsulated MSCs with iron oxide nanoparticles allowed continued cell-tracking by MRI over several weeks following transplantation into infarcted myocardium. In contrast, MSCs injected as cell suspension were only detectable for two days post transplantation by MRI. Histological analysis confirmed integration of transplanted cells at the infarct site. Therefore, microcapsules proved to be suitable for stem cell delivery into the infarcted myocardium and can overcome current limitations of poor cell retention in cardiac cell-based therapy.


Asunto(s)
Cápsulas , Proliferación Celular , Supervivencia Celular , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/patología , Animales , Masculino , Ratas , Ratas Wistar
14.
J Mol Cell Cardiol ; 82: 13-21, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25736855

RESUMEN

Natriuretic peptide receptor 3 (NPR3) is the clearance receptor for the cardiac natriuretic peptides (NPs). By modulating the level of NPs, NPR3 plays an important role in cardiovascular homeostasis. Although the physiological functions of NPR3 have been explored, little is known about its regulation in health or disease. MicroRNAs play an essential role in the post-transcriptional expression of many genes. Our aim was to investigate potential microRNA-based regulation of NPR3 in multiple models. Hypoxic challenge elevated levels of NPPB and ADM mRNA, as well as NT-proBNP and MR-proADM in human left ventricle derived cardiac cells (HCMa), and in the corresponding conditioned medium, as revealed by qRT-PCR and ELISA. NPR3 was decreased while NPR1 was increased by hypoxia at mRNA and protein levels in HCMa. Down-regulation of NPR3 mRNA was also observed in infarct and peri-infarct cardiac tissue from rats undergoing myocardial infarction. From microRNA microarray analyses and microRNA target predictive databases, miR-100 was selected as a candidate regulator of NPR3 expression. Further analyses confirmed up-regulation of miR-100 in hypoxic cells and associated conditioned media. Antagomir-based silencing of miR-100 enhanced NPR3 expression in HCMa. Furthermore, miR-100 levels were markedly up-regulated in rat hearts and in peripheral blood after myocardial infarction and in the blood from heart failure patients. Results from this study point to a role for miR-100 in the regulation of NPR3 expression, and suggest a possible therapeutic target for modulation of NP bioactivity in heart disease.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , Receptores del Factor Natriurético Atrial/genética , Regiones no Traducidas 3' , Adrenomedulina/genética , Adrenomedulina/metabolismo , Anciano , Animales , Secuencia de Bases , Sitios de Unión , Estudios de Casos y Controles , Medios de Cultivo Condicionados/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Masculino , MicroARNs/química , Persona de Mediana Edad , Infarto del Miocardio/sangre , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Péptido Natriurético Encefálico/metabolismo , Fragmentos de Péptidos/metabolismo , Precursores de Proteínas/metabolismo , Interferencia de ARN , ARN Mensajero/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores del Factor Natriurético Atrial/química , Receptores del Factor Natriurético Atrial/metabolismo , Factores de Tiempo
15.
Biomaterials ; 45: 27-35, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25662492

RESUMEN

AIMS: Cell-based myocardial restoration has not penetrated broad clinical practice yet due to poor cell retention and survival rates. In this study, we attempt a translational, large-scale restorative but minimally invasive approach in the pig, aiming at both structurally stabilizing the left ventricular (LV) wall and enhancing function following ischemic injury. METHODS AND RESULTS: A myocardial infarction (MI) was created by permanent ligation of left circumflex coronary artery through a small lateral thoracotomy. Thirty-six Yorkshire pigs were randomized to receive transthoracic intramyocardial injection into both infarct and border zone areas with different compounds: 1) Hyaluronic acid-based hydrogel; 2) autologous platelet-rich plasma (PRP); 3) ascorbic acid-enriched hydrogel (50 mg/L), combined with IV ibuprofen (25 mg/kg) and allopurinol (25 mg/kg) (cocktail group); 4) PRP and cocktail (full-compound); or 5) saline (control). The latter two groups received daily oral ibuprofen (25 mg/kg) for 7 days and allopurinol (25 mg/kg) for 30 days, postoperatively. Hemodynamic and echocardiographic studies were carried out at baseline, immediately after infarction and at end-point. Eight weeks after MI, the full-compound group had better LV fractional area change, ejection fraction and smaller LV dimensions than the control group. Also, dp/dtmax was significantly higher in the full-compound group when the heart rate increased from 100 bpm to 160bpm in stress tests. Blood vessel density was higher in the full-compound group, compared to the other treatment groups. CONCLUSIONS: A combination of PRP, anti-oxidant and anti-inflammatory factors with intramyocardial injection of hydrogel has the potential to structurally and functionally improve the injured heart muscle while attenuating adverse cardiac remodeling after acute myocardial infarction.


Asunto(s)
Ventrículos Cardíacos/patología , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacología , Infarto del Miocardio/terapia , Plasma Rico en Plaquetas/metabolismo , Investigación Biomédica Traslacional , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Cicatriz/patología , Colágeno/metabolismo , Diástole/efectos de los fármacos , Femenino , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Inyecciones , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/fisiopatología , Miocardio/patología , Neovascularización Fisiológica/efectos de los fármacos , Sus scrofa , Trasplante Autólogo , Ultrasonografía
16.
Tissue Eng Part A ; 21(5-6): 1106-15, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25370818

RESUMEN

Clinical applications of tissue engineering are constrained by the ability of the implanted construct to invoke vascularization in adequate extent and velocity. To overcome the current limitations presented by local delivery of single angiogenic factors, we explored the incorporation of prolyl hydroxylase inhibitors (PHIs) into scaffolds as an alternative vascularization strategy. PHIs are small molecule drugs that can stabilize the alpha subunit of hypoxia-inducible factor-1 (HIF-1), a key transcription factor that regulates a variety of angiogenic mechanisms. In this study, we conjugated the PHI pyridine-2,4-dicarboxylic acid (PDCA) through amide bonds to a gelatin sponge (Gelfoam(®)). Fibroblasts cultured on PDCA-Gelfoam were able to infiltrate and proliferate in these scaffolds while secreting significantly more vascular endothelial growth factor than cells grown on Gelfoam without PDCA. Reporter cells expressing green fluorescent protein-tagged HIF-1α exhibited dose-dependent stabilization of this angiogenic transcription factor when growing within PDCA-Gelfoam constructs. Subsequently, we implanted PDCA-Gelfoam scaffolds into the perirenal fat tissue of Sprague Dawley rats for 8 days. Immunostaining of explants revealed that the PDCA-Gelfoam scaffolds were amply infiltrated by cells and promoted vascular ingrowth in a dose-dependent manner. Thus, the incorporation of PHIs into scaffolds appears to be a feasible strategy for improving vascularization in regenerative medicine applications.


Asunto(s)
Neovascularización Fisiológica/efectos de los fármacos , Inhibidores de Prolil-Hidroxilasa/farmacología , Andamios del Tejido/química , Animales , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Crioultramicrotomía , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Esponja de Gelatina Absorbible/farmacología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Porosidad , Proteolisis/efectos de los fármacos , Piridinas/farmacología , Ratas Sprague-Dawley , Espectrofotometría Ultravioleta , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Stem Cells Dev ; 22(23): 3087-99, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23869939

RESUMEN

A crucial question in post-ischemic cell therapy refers to the ideal method of cell delivery to the heart. We hypothesized that epicardial implantation of subamnion-cord-lining mesenchymal stem cells (CL-MSC) angiogenic spheroids embedded within fibrin grafts (SASG) facilitates donor cell survival and enhances cardiac function in failing rat hearts. Furthermore, we compared the efficacy of this approach applied through two delivery methods. Spheroids made of 1.5×10(4) human CL-MSC coated with 2×10(3) human umbilical vein endothelial cells were self-assembled in hanging drops. SASG were constructed by embedding 150 spheroids in fibrin matrix. Except for untreated rats (MI, n=8), grafts were implanted 2 weeks after myocardial infarction upon confirmation of ensued heart failure through thoracotomy: SASG (n=8) and fibrin graft (FG, n=8); or video-assisted thoracoscopic surgery (VATS): SASG-VATS (n=8) and FG-VATS (n=7). In vivo CL-MSC survival was comparable between both SASG-treated groups throughout the study. SASG and SASG-VATS animals had decreased left ventricular end-diastolic pressure relative to untreated animals, and increased fractional shortening compared to MI and FG controls, 4 weeks after treatment. A 14.1% and 6.2% enhancement in ejection fraction from week 2 to 6 after injury was observed in SASG/SASG-VATS, paralleled by improvement in cardiac output. Treated hearts had smaller scar size, and more blood vessels than MI, while donor CL-MSC contributed to arteriogenesis within the graft and infarct areas. Taken together, our data suggest that SASG treatment has the potential to restore failing hearts by preserving cardiac function and inducing myocardial revascularization, while attenuating cardiac fibrosis. Furthermore, we introduce a method for minimally invasive in situ graft assembly.


Asunto(s)
Amnios/citología , Trasplante de Células Madre Mesenquimatosas , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/terapia , Revascularización Miocárdica , Neovascularización Fisiológica , Cordón Umbilical/citología , Animales , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/cirugía , Insuficiencia Cardíaca/terapia , Pruebas de Función Cardíaca , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/cirugía , Infarto del Miocardio/terapia , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/cirugía , Miocardio/patología , Fenotipo , Multimerización de Proteína , Ratas , Ratas Desnudas , Esferoides Celulares/citología , Cirugía Torácica Asistida por Video , Toracotomía , Factor A de Crecimiento Endotelial Vascular/metabolismo , Remodelación Ventricular
18.
Tissue Eng Part A ; 19(11-12): 1303-15, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23448654

RESUMEN

Myocardial restoration using tissue-engineered grafts to regenerate the ischemic myocardium offers improved donor cell retention, yet a limited cell survival resulting from poor vascularization needs to be addressed. A cell type derived from the subamnion, namely, cord-lining mesenchymal stem cells (CL-MSC), has recently been identified. Here we present a restorative strategy that combines a fibrin graft containing human CL-MSC and omental flap providing, thereby, cell-, structural-, and angiogenic support to the injured myocardium. The graft consisted of a mixture of 2×10(6) CL-MSC-GFP-Fluc and fibrin. Myocardial infarction (MI) was induced in nude rats and following confirmation of ensued heart failure with echocardiography 2 weeks after injury, therapeutic intervention was performed as follows: untreated (MI, n=7), CL-MSC graft (CL-MSCG, n=8), CL-MSCG and omental flap (CL-MSCG+OM, n=11), and omental flap (OM, n=8). In vivo bioluminescence imaging at 1, 3, 7, and 14 days post-treatment indicated comparable early donor cell viability between the CL-MSCG and CL-MSCG+OM. Treatment with CL-MSCG+OM improved the myocardial function as assessed by the measurement of end-diastolic left ventricular (LV) pressure (3.53±0.34 vs. 5.21±0.54 mmHg, p<0.05), contractility (+dP/dt, 3383.8±250.78 mmHg vs. 2464.9±191.8 mmHg, p<0.05), and the relaxation rate (-dP/dt, -2707.2±250.7 mmHg vs. 1948.7±207.8 mmHg, p<0.05), compared to MI control 6 weeks after ischemic injury. Furthermore, evidence of a 20.32% increase in the ejection fraction was observed in CL-MSCG+OM rats from week 2 to 6 after injury. Both CL-MSCG and CL-MSCG+OM led to an enhanced cardiac output (p<0.05), and attenuated the infarct size (35.7%±4.2% and 34.7%±4.8%), as compared to MI (60.7%±3.1%; p<0.01 and p<0.001, respectively). All treated groups had a higher arteriole density than controls. Yet, a higher amount of functional blood vessels, and a 20-fold increase in arteriole numbers were found in CL-MSCG+OM. Altogether, CL-MSCGs supplemented with vascular supply have the potential to repair the failing, chronically ischemic heart by improving myocardial revascularization, attenuating remodeling, and ameliorating cardiac dysfunction.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Revascularización Miocárdica , Epiplón/cirugía , Colgajos Quirúrgicos , Cordón Umbilical/citología , Animales , Supervivencia Celular , Enfermedad Crónica , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/terapia , Pruebas de Función Cardíaca , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Hemodinámica , Humanos , Células Madre Mesenquimatosas/metabolismo , Microscopía Confocal , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/terapia , Fenotipo , Ratas , Ultrasonografía , Función Ventricular Izquierda
19.
J Tissue Eng Regen Med ; 7(3): 203-12, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22034461

RESUMEN

Angiogenesis plays a key role in post-ischaemic myocardial repair. We hypothesized that epicardial implantation of an ascorbic acid (AA)-enriched myocardial artificial graft (MAG), which has been prevascularized in the recipients' own body, promotes restoration of the ischaemic heart. Gelatin patches were seeded with GFP-luciferase-expressing rat cardiomyoblasts and enriched with 5 µm AA. Grafts were prevascularized in vivo for 3 days, using a renal pouch model in rats. The MAG patch was then implanted into the same rat's ischaemic heart following myocardial infarction (MI). MAG-treated animals (MAG group, n = 6) were compared to untreated infarcted animals as injury controls (MI group, n = 6) and sham-operated rats as healthy controls (healthy group, n = 7). In vivo bioluminescence imaging indicated a decrease in donor cell survival by 83% during the first week post-implantation. Echocardiographic and haemodynamic assessment 4 weeks after MI revealed that MAG treatment attenuated left ventricular (LV) remodelling (LV end-systolic volume, 0.31 ± 0.13 vs 0.81 ± 0.01 ml, p < 0.05; LV end-diastolic volume 0.79 ± 0.33 vs 1.83 ± 0.26 ml, p < 0.076) and preserved LV wall thickness (0.21 ± 0.03 vs 0.09 ± 0.005 cm, p < 0.05) compared to the MI group. Cardiac output was higher in MAG than MI (51.59 ± 6.5 vs 25.06 ± 4.24 ml/min, p < 0.01) and comparable to healthy rats (47.08 ± 1.9 ml/min). Histology showed decreased fibrosis, and a seven-fold increase in blood vessel density in the scar area of MAG compared to MI group (15.3 ± 1.1 vs 2.1 ± 0.3 blood vessels/hpf, p < 0.0001). Implantation of AA-enriched prevascularized grafts enhanced vascularity in ischaemic rat hearts, attenuated LV remodelling and preserved LV function.


Asunto(s)
Ácido Ascórbico/farmacología , Trasplante de Corazón , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/terapia , Miocardio/patología , Neovascularización Fisiológica/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos , Animales , Antígenos/metabolismo , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Electrocardiografía , Fibrosis , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Hemodinámica/efectos de los fármacos , Masculino , Isquemia Miocárdica/diagnóstico por imagen , Ratas , Ratas Wistar , Ultrasonografía
20.
Cardiol Res Pract ; 2012: 240497, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22400122

RESUMEN

Myocardial infarction is the leading cause of death in developed countries. Cardiac cell therapy has been introduced to clinical trials for more than ten years but its results are still controversial. Tissue engineering has addressed some limitations of cell therapy and appears to be a promising solution for cardiac regeneration. In this review, we would like to summarize the current understanding about the therapeutic effect of cell therapy and tissue engineering under purview of functional and structural aspects, highlighting actual roles of each therapy towards clinical application.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...