Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Circ Heart Fail ; 13(9): e007099, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32900234

RESUMEN

BACKGROUND: Risk stratifying patients with cardiogenic shock (CS) is a major unmet need. The recently proposed Society for Cardiovascular Angiography and Interventions (SCAI) stages as an approach to identify patients at risk for in-hospital mortality remains under investigation. We studied the utility of the SCAI stages and further explored the impact of hemodynamic congestion on clinical outcomes. METHODS: The CS Working Group registry includes patients with CS from 8 medical centers enrolled between 2016 and 2019. Patients were classified by the maximum SCAI stage (B-E) reached during their hospital stay according to drug and device utilization. In-hospital mortality was evaluated for association with SCAI stages and hemodynamic congestion. RESULTS: Of the 1414 patients with CS, the majority were due to decompensated heart failure (50%) or myocardial infarction (MI; 35%). In-hospital mortality was 31% for the total cohort, but higher among patients with MI (41% versus 26%, MI versus heart failure, P<0.0001). Risk for in-hospital mortality was associated with increasing SCAI stage (odds ratio [95% CI], 3.25 [2.63-4.02]) in both MI and heart failure cohorts. Hemodynamic data was available in 1116 (79%) patients. Elevated biventricular filling pressures were common among patients with CS, and right atrial pressure was associated with increased mortality and higher SCAI Stage. CONCLUSIONS: Our findings support an association between the proposed SCAI staging system and in-hospital mortality among patient with heart failure and MI. We further identify that venous congestion is common and identifies patients with CS at high risk for in-hospital mortality. These findings provide may inform future management protocols and clinical studies.


Asunto(s)
Hemodinámica , Mortalidad Hospitalaria , Choque Cardiogénico/clasificación , Choque Cardiogénico/mortalidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sistema de Registros , Medición de Riesgo , Factores de Riesgo , Choque Cardiogénico/fisiopatología , Estados Unidos
2.
FASEB J ; 34(1): 930-944, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914588

RESUMEN

The pathophysiology of pulmonary hypertension (PH) and heart failure (HF) includes fibrogenic remodeling associated with the loss of pulmonary arterial (PA) and cardiac compliance. We and others have previously identified transglutaminase 2 (TG2) as a participant in adverse fibrogenic remodeling. However, little is known about the biologic mechanisms that regulate TG2 function. We examined physiological mouse models of experimental PH, HF, and type 1 diabetes that are associated with altered glucose metabolism/glycolysis and report here that TG2 expression and activity are elevated in pulmonary and cardiac tissues under all these conditions. We additionally used PA adventitial fibroblasts to test the hypothesis that TG2 is an intermediary between enhanced tissue glycolysis and fibrogenesis. Our in vitro results show that glycolytic enzymes and TG2 are upregulated in fibroblasts exposed to high glucose, which stimulates cellular glycolysis as measured by Seahorse analysis. We examined the relationship of TG2 to a terminal glycolytic enzyme, pyruvate kinase M2 (PKM2), and found that PKM2 regulates glucose-induced TG2 expression and activity as well as fibrogenesis. Our studies further show that TG2 inhibition blocks glucose-induced fibrogenesis and cell proliferation. Our findings support a novel role for glycolysis-mediated TG2 induction and tissue fibrosis associated with experimental PH, HF, and hyperglycemia.


Asunto(s)
Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Regulación Enzimológica de la Expresión Génica , Glucólisis , Hipertensión Pulmonar/metabolismo , Transglutaminasas/genética , Transglutaminasas/metabolismo , Animales , Proteínas Portadoras/metabolismo , Proliferación Celular , Fibroblastos/metabolismo , Glucosa/metabolismo , Humanos , Hiperglucemia/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína Glutamina Gamma Glutamiltransferasa 2 , Arteria Pulmonar/metabolismo , Piruvato Quinasa/metabolismo , Transducción de Señal , Hormonas Tiroideas/metabolismo , Regulación hacia Arriba , Proteínas de Unión a Hormona Tiroide
3.
Artif Organs ; 43(2): 125-131, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30216467

RESUMEN

Hemolysis is a potential limitation of percutaneously delivered left-sided mechanical circulatory support pumps, including trans valvular micro-axial flow pumps (TVP). Hemolytic biomarkers among durable left ventricular assist devices include lactate dehydrogenase (LDH) >2.5 times the upper limit of normal (ULN) and plasma-free hemoglobin (pf-Hb) >20 mg/dL. We examined the predictive value of these markers among patients with cardiogenic shock (CS) receiving a TVP. We retrospectively studied records of 116 consecutive patients receiving an Impella TVP at our institution between 2012 and 2017 for CS. Twenty-three met inclusion/exclusion criteria, and had sufficient pf-Hb data for analysis. Area under receiver-operator characteristic (ROC) curve for diagnosing hemolysis were calculated. Mean age was 62 ± 14 years and ejection fraction was 15 ± 5%. Mean duration of support was 5.4 ± 3.5 days. Pre-device LDH levels were >2.5x ULN in 71% (n = 5/7) of 5.0 and 29% of CP patients, while pre-device pf-Hb levels were >20 mg/dL in 14% (n = 1/7) of 5.0 and 25% (n = 4/16) of CP patients. Given elevated baseline LDH and pf-Hb levels, we defined hemolysis as a pf-Hb level >40 mg/dL within 72 h post-implant plus clinical evidence of device-related hemolysis. We identified that 30% (n = 7/23) had device-related hemolysis. Using ROC curve-derived cut-points, an increase in delta pf-Hb by >27mg/dL, not delta LDH, within 24 h after TVP implant (delta pf-Hb: C-statistic = 0.79, sensitivity: 57%, specificity: 93%, p <0.05) was highly predictive of hemolysis. In conclusion, we identified a change in pf-Hb, not LDH, levels is highly sensitive and specific for hemolysis in patients treated with a TVP for CS.


Asunto(s)
Corazón Auxiliar/efectos adversos , Hemoglobinas/análisis , Hemólisis/fisiología , Choque Cardiogénico/terapia , Anciano , Femenino , Pruebas Hematológicas , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Choque Cardiogénico/sangre
4.
J Am Coll Cardiol ; 72(5): 501-514, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-30049311

RESUMEN

BACKGROUND: Heart failure after an acute myocardial infarction (AMI) is a major cause of morbidity and mortality worldwide. We recently reported that activation of a transvalvular axial-flow pump in the left ventricle and delaying myocardial reperfusion, known as primary unloading, limits infarct size after AMI. The mechanisms underlying the cardioprotective benefit of primary unloading and whether the acute decrease in infarct size results in a durable reduction in LV scar and improves cardiac function remain unknown. OBJECTIVES: This study tested the importance of LV unloading before reperfusion, explored cardioprotective mechanisms, and determined the late-term impact of primary unloading on myocardial function. METHODS: Adult male swine were subjected to primary reperfusion or primary unloading after 90 min of percutaneous left anterior descending artery occlusion. RESULTS: Compared with primary reperfusion, 30 min of LV unloading was necessary and sufficient before reperfusion to limit infarct size 28 days after AMI. Compared with primary reperfusion, primary unloading increased expression of genes associated with cellular respiration and mitochondrial integrity within the infarct zone. Primary unloading for 30 min further reduced activity levels of proteases known to degrade the cardioprotective cytokine, stromal-derived factor (SDF)-1α, thereby increasing SDF-1α signaling via reperfusion injury salvage kinases, which limits apoptosis within the infarct zone. Inhibiting SDF-1α activity attenuated the cardioprotective effect of primary unloading. Twenty-eight days after AMI, primary unloading reduced LV scar size, improved cardiac function, and limited expression of biomarkers associated with heart failure and maladaptive remodeling. CONCLUSIONS: The authors report for the first time that first mechanically reducing LV work before coronary reperfusion with a transvalvular pump is necessary and sufficient to reduce infarct size and to activate a cardioprotective program that includes enhanced SDF-1α activity. Primary unloading further improved LV scar size and cardiac function 28 days after AMI.


Asunto(s)
Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Reperfusión Miocárdica/métodos , Recuperación de la Función/fisiología , Función Ventricular Izquierda/fisiología , Animales , Masculino , Reperfusión Miocárdica/tendencias , Porcinos
5.
Circulation ; 138(5): 513-526, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-29487140

RESUMEN

BACKGROUND: Heart failure is a growing cause of morbidity and mortality worldwide. Transforming growth factor beta (TGF-ß1) promotes cardiac fibrosis, but also activates counterregulatory pathways that serve to regulate TGF-ß1 activity in heart failure. Bone morphogenetic protein 9 (BMP9) is a member of the TGFß family of cytokines and signals via the downstream effector protein Smad1. Endoglin is a TGFß coreceptor that promotes TGF-ß1 signaling via Smad3 and binds BMP9 with high affinity. We hypothesized that BMP9 limits cardiac fibrosis by activating Smad1 and attenuating Smad3, and, furthermore, that neutralizing endoglin activity promotes BMP9 activity. METHODS: We examined BMP9 expression and signaling in human cardiac fibroblasts and human subjects with heart failure. We used the transverse aortic constriction-induced model of heart failure to evaluate the functional effect of BMP9 signaling on cardiac remodeling. RESULTS: BMP9 expression is increased in the circulation and left ventricle (LV) of human subjects with heart failure and is expressed by cardiac fibroblasts. Next, we observed that BMP9 attenuates type I collagen synthesis in human cardiac fibroblasts using recombinant human BMP9 and a small interfering RNA approach. In BMP9-/- mice subjected to transverse aortic constriction, loss of BMP9 activity promotes cardiac fibrosis, impairs LV function, and increases LV levels of phosphorylated Smad3 (pSmad3), not pSmad1. In contrast, treatment of wild-type mice subjected to transverse aortic constriction with recombinant BMP9 limits progression of cardiac fibrosis, improves LV function, enhances myocardial capillary density, and increases LV levels of pSmad1, not pSmad3 in comparison with vehicle-treated controls. Because endoglin binds BMP9 with high affinity, we explored the effect of reduced endoglin activity on BMP9 activity. Neutralizing endoglin activity in human cardiac fibroblasts or in wild-type mice subjected to transverse aortic constriction-induced heart failure limits collagen production, increases BMP9 protein levels, and increases levels of pSmad1, not pSmad3. CONCLUSIONS: Our results identify a novel functional role for BMP9 as an endogenous inhibitor of cardiac fibrosis attributable to LV pressure overload and further show that treatment with either recombinant BMP9 or disruption of endoglin activity promotes BMP9 activity and limits cardiac fibrosis in heart failure, thereby providing potentially novel therapeutic approaches for patients with heart failure.


Asunto(s)
Factor 2 de Diferenciación de Crecimiento/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocardio/metabolismo , Función Ventricular Izquierda , Remodelación Ventricular , Animales , Modelos Animales de Enfermedad , Endoglina/deficiencia , Endoglina/genética , Endoglina/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Factor 2 de Diferenciación de Crecimiento/deficiencia , Factor 2 de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/genética , Haploinsuficiencia , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Fosforilación , Recuperación de la Función , Transducción de Señal , Proteína Smad1/metabolismo , Proteína smad3/metabolismo
6.
Cardiovasc Pathol ; 31: 26-33, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28820968

RESUMEN

INTRODUCTION: Activin receptor-like kinase 1 (ALK1) mediates signaling via the transforming growth factor beta-1 (TGFß1), a pro-fibrogenic cytokine. No studies have defined a role for ALK1 in heart failure. HYPOTHESIS: We tested the hypothesis that reduced ALK1 expression promotes maladaptive cardiac remodeling in heart failure. METHODS AND RESULTS: In patients with advanced heart failure referred for left ventricular (LV) assist device implantation, LV Alk1 mRNA and protein levels were lower than control LV obtained from patients without heart failure. To investigate the role of ALK1 in heart failure, Alk1 haploinsufficient (Alk1+/-) and wild-type (WT) mice were studied 2 weeks after severe transverse aortic constriction (TAC). LV and lung weights were higher in Alk1+/- mice after TAC. Cardiomyocyte area and LV mRNA levels of brain natriuretic peptide and ß-myosin heavy chain were increased similarly in Alk1+/- and WT mice after TAC. Alk-1 mice exhibited reduced Smad 1 phosphorylation and signaling compared to WT mice after TAC. Compared to WT, LV fibrosis and Type 1 collagen mRNA and protein levels were higher in Alk1+/- mice. LV fractional shortening was lower in Alk1+/- mice after TAC. CONCLUSIONS: Reduced expression of ALK1 promotes cardiac fibrosis and impaired LV function in a murine model of heart failure. Further studies examining the role of ALK1 and ALK1 inhibitors on cardiac remodeling are required.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocardio/patología , Remodelación Ventricular/fisiología , Receptores de Activinas Tipo I/metabolismo , Adulto , Animales , Femenino , Fibrosis/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Miocardio/metabolismo
7.
Circulation ; 136(3): 314-326, 2017 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-28716832

RESUMEN

Right ventricular (RV) failure remains a major cause of global morbidity and mortality for patients with advanced heart failure, pulmonary hypertension, or acute myocardial infarction and after major cardiac surgery. Over the past 2 decades, percutaneously delivered acute mechanical circulatory support pumps specifically designed to support RV failure have been introduced into clinical practice. RV acute mechanical circulatory support now represents an important step in the management of RV failure and provides an opportunity to rapidly stabilize patients with cardiogenic shock involving the RV. As experience with RV devices grows, their role as mechanical therapies for RV failure will depend less on the technical ability to place the device and more on improved algorithms for identifying RV failure, patient monitoring, and weaning protocols for both isolated RV failure and biventricular failure. In this review, we discuss the pathophysiology of acute RV failure and both the mechanism of action and clinical data exploring the utility of existing RV acute mechanical circulatory support devices.


Asunto(s)
Insuficiencia Cardíaca/cirugía , Corazón Auxiliar , Disfunción Ventricular Derecha/cirugía , Enfermedad Aguda , Insuficiencia Cardíaca/diagnóstico , Corazón Auxiliar/tendencias , Humanos , Intervención Coronaria Percutánea/métodos , Intervención Coronaria Percutánea/tendencias , Disfunción Ventricular Derecha/diagnóstico
8.
Heart Vessels ; 32(5): 628-636, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28213819

RESUMEN

Activin like kinase-1 (AlK-1) mediates signaling via the transforming growth factor beta (TGFß) family of ligands. AlK-1 activity promotes endothelial proliferation and migration. Reduced AlK-1 activity is associated with arteriovenous malformations. No studies have examined the effect of global AlK-1 deletion on indices of cardiac remodeling. We hypothesized that reduced levels of AlK-1 promote maladaptive cardiac remodeling. To test this hypothesis, we employed AlK-1 conditional knockout mice (cKO) harboring the ROSA26-CreER knock-in allele, whereby a single dose of intraperitoneal tamoxifen triggered ubiquitous Cre recombinase-mediated excision of floxed AlK-1 alleles. Tamoxifen treated wild-type (WT-TAM; n = 5) and vehicle treated AlK-1-cKO mice (cKO-CON; n = 5) served as controls for tamoxifen treated AlK-1-cKO mice (cKO-TAM; n = 15). AlK-1 cKO-TAM mice demonstrated reduced 14-day survival compared to cKO-CON controls (13 vs 100%, respectively, p < 0.01). Seven days after treatment, cKO-TAM mice exhibited reduced left ventricular (LV) fractional shortening, progressive LV dilation, and gastrointestinal bleeding. After 14 days total body mass was reduced, but LV and lung mass increased in cKO-TAM not cKO-CON mice. Peak LV systolic pressure, contractility, and arterial elastance were reduced, but LV end-diastolic pressure and stroke volume were increased in cKO-TAM, not cKO-CON mice. LV AlK-1 mRNA levels were reduced in cKO-TAM, not cKO-CON mice. LV levels of other TGFß-family ligands and receptors (AlK5, TBRII, BMPRII, Endoglin, BMP7, BMP9, and TGFß1) were unchanged between groups. Cardiomyocyte area and LV levels of BNP were increased in cKO-TAM mice, but LV levels of ß-MHC and SERCA were unchanged. No increase in markers of cardiac fibrosis, Type I collagen, CTGF, or PAI-1, were observed between groups. No differences were observed for any variable studied between cKO-CON and WT-TAM mice. Global deletion of AlK-1 is associated with the development of high output heart failure without maladaptive remodeling. Future studies exploring the functional role of AlK-1 in cardiac remodeling independent of systemic AVMs are required.


Asunto(s)
Receptores de Activinas Tipo I/genética , Regulación de la Expresión Génica , Insuficiencia Cardíaca/genética , ARN/genética , Función Ventricular Izquierda/fisiología , Remodelación Ventricular/fisiología , Receptores de Activinas Tipo I/biosíntesis , Receptores de Activinas Tipo II , Alelos , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal
9.
Cardiovasc Pathol ; 25(6): 478-482, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27614169

RESUMEN

INTRODUCTION: Transient receptor potential (TRP) channels are broadly expressed cation channels that mediate diverse physiological stimuli and include canonical (TRPC), melastatin (TRPM), and vanilloid (TRPV) subtypes. Recent studies have implicated a role for TRPC6 channels as an important component of signaling via the cytokine, transforming growth factor beta 1 (TGFß1) in right (RV) or left ventricular (LV) failure. Endoglin (Eng) is a transmembrane glycoprotein that promotes TRPC6 expression and TGFß1 activity. No studies have defined biventricular expression of all TRP channel family members in heart failure. HYPOTHESIS: We hypothesized that heart failure is associated with distinct patterns of TRP channel expression in the LV and RV. METHODS: Paired viable LV and RV free wall tissue was obtained from human subjects with end-stage heart failure (n=12) referred for cardiac transplantation or biventricular assist device implantation. Paired LV and RV samples from human subjects without heart failure served as controls (n=3). To explore a functional role for Eng as a regulator of TRP expression in response to RV or LV pressure overload, wild-type (Eng+/+) and Eng haploinsufficient (Eng+/-) mice were exposed to thoracic aortic (TAC) or pulmonary arterial (PAC) constriction for 8weeks. Biventricular tissue was analyzed by real-time polymerase chain reaction. RESULTS: Compared to nonfailing human LV and RV samples, mRNA levels of TRPC1, 3, 4, 6, and TRPV-2 were increased and TRPM2, 3, and 8 were decreased in failing LV and RV samples. TRPC1 and 6 levels were higher in failing RV compared to failing LV samples. After TAC, murine LV levels of TPRC1 and 6 were increased in both Eng+/+ and Eng+/- mice compared to sham controls. LV levels of TRPC4, TRPM3 and 7, TRPV2 and 4 were increased in Eng+/+, not in Eng+/- mice after TAC. After PAC, all TRP channel family members were increased in the RV, but not LV, of Eng+/+ compared to sham controls. In contrast to Eng+/+, PAC did not increase RV or LV levels of TRP channels in Eng+/- mice. CONCLUSIONS: This is the first study to demonstrate that TRP channels exhibit distinct profiles of expression in the LV and RV of patients with heart failure and in murine models of univentricular pressure overload. We further introduce that the TGFß1 coreceptor Eng selectively regulates expression of multiple TRP channels in the setting of LV or RV pressure overload.


Asunto(s)
Endoglina/metabolismo , Insuficiencia Cardíaca/metabolismo , Canales de Potencial de Receptor Transitorio/biosíntesis , Adulto , Anciano , Animales , Femenino , Regulación de la Expresión Génica , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma , Adulto Joven
10.
Artículo en Inglés | MEDLINE | ID: mdl-26758053

RESUMEN

OPINION STATEMENT: The use of percutaneous, non-durable mechanical circulatory support (MCS) for cardiogenic shock (CS) is growing; however, large, randomized clinical trials confirming benefit in this population do not exist. Guidelines and recommendations regarding optimal timing for MCS implementation, patient selection, device selection, and post-implantation management are beginning to emerge. A better understanding of (1) the distinct hemodynamic effects of each device option, (2) the need for early implementation of the appropriate device option for a particular clinical scenario, (3) the definition of non-salvageable CS to help clinicians know when to say "no" to non-durable MCS, and (4) best practices to monitor, wean, and optimize metabolic parameters while using non-durable MCS are required to continue improving clinical outcomes for patients with CS.

11.
J Card Fail ; 22(2): 110-6, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26564619

RESUMEN

BACKGROUND: Right ventricular failure (RVF) is a major cause of morbidity and mortality after CF-LVAD implantation. We explored the association of pulmonary artery compliance (PAC), pulmonary artery elastance (PAE), and pulmonary artery pulsatility index (PAPi) in addition to established parameters as preoperative determinants of postoperative RVF after CF-LVAD surgery. METHODS AND RESULTS: We retrospectively reviewed 132 consecutive CF-LVAD implantations at Tufts Medical Center from 2008 to 2013. Clinical, hemodynamic, and echocardiographic data were studied. RVF was defined as the unplanned need for a right ventricular assist device or inotrope dependence for ≥14 days. Univariate analysis was performed. RVF occurred in 32 of 132 patients (24%). PAC and PAE were not changed, whereas the PAPi was lower among patients with versus without postoperative RVF (1.32 ± 0.46 vs 2.77 ± 1.16; P < .001). RA pressure, RA to pulmonary capillary wedge pressure ratio (RA:PCWP), and RV stroke work index (RVSWI) were also associated with RVF. Using receiver operating characteristic curve-derived cut-points, PAPi < 1.85 provided 94% sensitivity and 81% specificity (C-statistic = 0.942) for identifying RVF and exceeded the predictive value of RA:PCWP, RVSWI, or RA pressure alone. CONCLUSIONS: PAPi is a simple hemodynamic variable that may help to identify patients at high risk of developing RVF after LVAD implantation.


Asunto(s)
Insuficiencia Cardíaca/etiología , Ventrículos Cardíacos/cirugía , Corazón Auxiliar/efectos adversos , Arteria Pulmonar/fisiopatología , Presión Esfenoidal Pulmonar , Disfunción Ventricular Derecha/etiología , Adulto , Anciano , Ecocardiografía , Femenino , Insuficiencia Cardíaca/prevención & control , Humanos , Masculino , Persona de Mediana Edad , Curva ROC , Estudios Retrospectivos , Disfunción Ventricular Derecha/prevención & control
12.
ASAIO J ; 62(1): 33-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26479464

RESUMEN

Left ventricular assist device (LVAD) thrombosis is associated with high morbidity and mortality because of device malfunction, embolic events, and hemolysis. There remains uncertainty as to whether immediate device exchange versus an initial trial of anticoagulant and antiplatelet therapy is most appropriate in hemodynamically stable patients. We conducted a retrospective analysis of all LVAD implantations at a single center between January 2009 and June 2013 with follow-up through December 2013. Suspected LVAD thrombosis occurred in 20% of patients (N = 25) over a median follow-up of 275 days. Medical therapy led to resolution of hemolysis, and discharge to home, in 15 of 25 (60%) cases; however, this strategy was associated with intracranial hemorrhage in 4 patients and readmission with recurrent thrombosis in 10 patients. The 30 day, 6 month, and 1 year freedom from suspected LVAD thrombosis was 96.5, 85.9, and 80.3% in HeartMate II devices and 100, 92.9, and 87.1% in HeartWare ventricular assist devices, respectively (p = 0.11). Although medical treatment with intravenous heparin, antiplatelet agents, antithrombotic agents, or thrombolytic therapy can lead to initial resolution of hemolysis, the risks of recurrence after transition to warfarin and adverse events are high.


Asunto(s)
Insuficiencia Cardíaca/cirugía , Corazón Auxiliar/efectos adversos , Trombosis/etiología , Anticoagulantes/uso terapéutico , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Inhibidores de Agregación Plaquetaria/uso terapéutico , Estudios Retrospectivos , Trombosis/tratamiento farmacológico
13.
Biomarkers ; 21(1): 91-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26667393

RESUMEN

BACKGROUND: Heart failure (HF) involves myocardial fibrosis and dysregulated angiogenesis. OBJECTIVE: We explored whether biomarkers of fibrosis and angiogenesis correlate with HF severity. METHODS: Biomarkers of fibrosis [procollagen types I and III (PIP and P3NP), carboxyterminal-telopeptide of type I collagen (ICTP), matrix metalloproteases (MMP2 and MMP9), tissue inhibitor of MMP1 (TIMP1)]; and angiogenesis [placental growth factor (PGF), vascular endothelial growth factor (VEGF), soluble Fms-like tyrosine kinase-1 (sFlt1)] were measured in 52 HF patients and 19 controls. RESULTS: P3NP, ICTP, MMP2, TIMP1, PGF, and sFlt1 levels were elevated in HF, while PIP/ICTP, PGF/sFlt1, and VEGF/sFlt1 ratios were reduced. PIP/ICTP, MMP-9/TIMP1, and VEGF/sFlt1 ratios were lowest among patients with severe HF. CONCLUSIONS: Severe HF is associated with collagen breakdown and reduced angiogenesis. A multimarker approach may guide therapeutic targeting of fibrosis and angiogenesis in HF.


Asunto(s)
Insuficiencia Cardíaca/sangre , Miocardio/patología , Anciano , Proteínas Angiogénicas/sangre , Biomarcadores/sangre , Estudios de Casos y Controles , Femenino , Fibrosis , Humanos , Masculino , Persona de Mediana Edad , Neovascularización Fisiológica , Estudios Prospectivos
14.
JACC Heart Fail ; 3(11): 873-82, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26541785

RESUMEN

OBJECTIVES: This study tested the hypothesis that first reducing myocardial work by unloading the left ventricle (LV) with a novel intracorporeal axial flow catheter while delaying coronary reperfusion activates a myocardial protection program and reduces infarct size. BACKGROUND: Ischemic heart disease is a major cause of morbidity and mortality worldwide. Primary myocardial reperfusion remains the gold standard for the treatment of an acute myocardial infarction (AMI); however, ischemia-reperfusion injury contributes to residual myocardial damage and subsequent heart failure. Stromal cell-derived factor (SDF)-1α is a chemokine that activates cardioprotective signaling via Akt, extracellular regulated kinase, and glycogen synthase kinase-3ß. METHODS: AMI was induced by occlusion of the left anterior descending artery (LAD) via angioplasty for 90 min in 50-kg male Yorkshire swine (n = 5/group). In the primary reperfusion (1° Reperfusion) group, the LAD was reperfused for 120 min. In the primary unloading (1° Unloading) group, after 90 min of ischemia the axial flow pump was activated and the LAD left occluded for an additional 60 min, followed by 120 min of reperfusion. Myocardial infarct size and kinase activity were quantified. RESULTS: Compared with 1° Reperfusion, 1° Unloading reduced LV wall stress and increased myocardial levels of SDF-1α, CXCR4, and phosphorylated Akt, extracellular regulated kinase, and glycogen synthase kinase-3ß in the infarct zone. 1° Unloading increased antiapoptotic signaling and reduced myocardial infarct size by 43% compared with 1° Reperfusion (73 ± 13% vs. 42 ± 8%; p = 0.005). Myocardial levels of SDF-1 correlated inversely with infarct size (R = 0.89; p < 0.01). CONCLUSIONS: Compared with the contemporary strategy of primary reperfusion, mechanically conditioning the myocardium using a novel axial flow catheter while delaying coronary reperfusion decreases LV wall stress and activates a myocardial protection program that up-regulates SDF-1α/CXCR4 expression, increases cardioprotective signaling, reduces apoptosis, and limits myocardial damage in AMI.


Asunto(s)
Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Daño por Reperfusión Miocárdica/prevención & control , Reperfusión Miocárdica , Animales , Biomarcadores/metabolismo , Quimiocina CXCL12/metabolismo , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Hemodinámica , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/mortalidad , Reperfusión Miocárdica/instrumentación , Reperfusión Miocárdica/métodos , Reperfusión Miocárdica/mortalidad , Fosfotransferasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores CXCR4/metabolismo , Estrés Mecánico , Sus scrofa
15.
J Am Heart Assoc ; 3(4)2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-25015075

RESUMEN

BACKGROUND: Right ventricular (RV) failure is a major cause of mortality worldwide and is often a consequence of RV pressure overload (RVPO). Endoglin is a coreceptor for the profibrogenic cytokine, transforming growth factor beta 1 (TGF-ß1). TGF-ß1 signaling by the canonical transient receptor protein channel 6 (TRPC-6) was recently reported to stimulate calcineurin-mediated myofibroblast transformation, a critical component of cardiac fibrosis. We hypothesized that reduced activity of the TGF-ß1 coreceptor, endoglin, limits RV calcineurin expression and improves survival in RVPO. METHODS AND RESULTS: We first demonstrate that endoglin is required for TGF-ß1-mediated calcineurin/TRPC-6 expression and up-regulation of alpha-smooth muscle antigen (α-SMA), a marker of myofibroblast transformation, in human RV fibroblasts. Using endoglin haploinsufficient mice (Eng(+/-)) we show that reduced endoglin activity preserves RV function, limits RV fibrosis, and attenuates activation of the calcineurin/TRPC-6/α-SMA pathway in a model of angio-obliterative pulmonary hypertension. Next, using Eng(+/-) mice or a neutralizing antibody (Ab) against endoglin (N-Eng) in wild-type mice, we show that reduced endoglin activity improves survival and attenuates RV fibrosis in models of RVPO induced by pulmonary artery constriction. To explore the utility of targeting endoglin, we observed a reversal of RV fibrosis and calcineurin levels in wild-type mice treated with a N-Eng Ab, compared to an immunoglobulin G control. CONCLUSION: These data establish endoglin as a regulator of TGF-ß1 signaling by calcineurin and TRPC-6 in the RV and identify it as a potential therapeutic target to limit RV fibrosis and improve survival in RVPO, a common cause of death in cardiac and pulmonary disease.


Asunto(s)
Calcineurina/genética , Hipertensión Pulmonar/genética , Péptidos y Proteínas de Señalización Intracelular/genética , ARN Mensajero/metabolismo , Canales Catiónicos TRPC/genética , Disfunción Ventricular Derecha/genética , Actinas/genética , Actinas/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Calcineurina/metabolismo , Modelos Animales de Enfermedad , Endoglina , Fibroblastos/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Humanos , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Noqueados , Miofibroblastos/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Tasa de Supervivencia , Canales Catiónicos TRPC/metabolismo , Canal Catiónico TRPC6 , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Disfunción Ventricular Derecha/metabolismo , Disfunción Ventricular Derecha/fisiopatología
16.
Vasc Health Risk Manag ; 9: 195-206, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23662065

RESUMEN

Endoglin (CD105) is a type III auxiliary receptor for the transforming growth factor beta (TGFß) superfamily. Several lines of evidence suggest that endoglin plays a critical role in maintaining cardiovascular homeostasis. Seemingly disparate disease conditions, including hereditary hemorrhagic telangiectasia, pre-eclampsia, and cardiac fibrosis, have now been associated with endoglin. Given the central role of the TGFß superfamily in multiple disease conditions, this review provides a detailed update on endoglin as an evolving therapeutic target in the management of cardiovascular disease.


Asunto(s)
Antígenos CD/metabolismo , Enfermedades Cardiovasculares/metabolismo , Sistema Cardiovascular/metabolismo , Receptores de Superficie Celular/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/terapia , Sistema Cardiovascular/patología , Sistema Cardiovascular/fisiopatología , Endoglina , Femenino , Homeostasis , Humanos , Ligandos , Masculino , Preeclampsia/metabolismo , Embarazo , Transducción de Señal , Telangiectasia Hemorrágica Hereditaria/metabolismo , Remodelación Ventricular
17.
PLoS One ; 7(1): e30063, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22253880

RESUMEN

Increased utrophin expression is known to reduce pathology in dystrophin-deficient skeletal muscles. Transgenic over-expression of PGC-1α has been shown to increase levels of utrophin mRNA and improve the histology of mdx muscles. Other reports have shown that PGC-1α signaling can lead to increased oxidative capacity and a fast to slow fiber type shift. Given that it has been shown that slow fibers produce and maintain more utrophin than fast skeletal muscle fibers, we hypothesized that over-expression of PGC-1α in post-natal mdx mice would increase utrophin levels via a fiber type shift, resulting in more slow, oxidative fibers that are also more resistant to contraction-induced damage. To test this hypothesis, neonatal mdx mice were injected with recombinant adeno-associated virus (AAV) driving expression of PGC-1α. PGC-1α over-expression resulted in increased utrophin and type I myosin heavy chain expression as well as elevated mitochondrial protein expression. Muscles were shown to be more resistant to contraction-induced damage and more fatigue resistant. Sirt-1 was increased while p38 activation and NRF-1 were reduced in PGC-1α over-expressing muscle when compared to control. We also evaluated if the use a pharmacological PGC-1α pathway activator, resveratrol, could drive the same physiological changes. Resveratrol administration (100 mg/kg/day) resulted in improved fatigue resistance, but did not achieve significant increases in utrophin expression. These data suggest that the PGC-1α pathway is a potential target for therapeutic intervention in dystrophic skeletal muscle.


Asunto(s)
Fibras Musculares de Contracción Rápida/patología , Fibras Musculares de Contracción Lenta/patología , Distrofia Muscular Animal/fisiopatología , Recuperación de la Función/fisiología , Transactivadores/metabolismo , Animales , Fenómenos Biomecánicos , Peso Corporal/efectos de los fármacos , Dependovirus/efectos de los fármacos , Dependovirus/metabolismo , Suplementos Dietéticos , Técnicas de Transferencia de Gen , Ratones , Ratones Endogámicos mdx , Contracción Muscular , Fatiga Muscular , Fibras Musculares de Contracción Rápida/efectos de los fármacos , Fibras Musculares de Contracción Lenta/efectos de los fármacos , Distrofia Muscular Animal/complicaciones , Miosinas/metabolismo , Tamaño de los Órganos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Recuperación de la Función/efectos de los fármacos , Resveratrol , Estilbenos/administración & dosificación , Estilbenos/farmacología , Factores de Transcripción
18.
Mol Ther ; 20(3): 580-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22146342

RESUMEN

Although restoration of dystrophin expression via exon skipping in both cardiac and skeletal muscle has been successfully demonstrated in the mdx mouse, restoration of cardiac dystrophin expression in large animal models of Duchenne muscular dystrophy (DMD) has proven to be a challenge. In large animals, investigators have focused on using intravenous injection of antisense oligonucleotides (AO) to mediate exon skipping. In this study, we sought to optimize restoration of cardiac dystrophin expression in the golden retriever muscular dystrophy (GRMD) model using percutaneous transendocardial delivery of recombinant AAV6 (rAAV6) to deliver a modified U7 small nuclear RNA (snRNA) carrying antisense sequence to target the exon splicing enhancers of exons 6 and 8 and correct the disrupted reading frame. We demonstrate restoration of cardiac dystrophin expression at 13 months confirmed by reverse transcription-PCR (RT-PCR) and immunoblot as well as membrane localization by immunohistochemistry. This was accompanied by improved cardiac function as assessed by cardiac magnetic resonance imaging (MRI). Percutaneous transendocardial delivery of rAAV6 expressing a modified U7 exon skipping construct is a safe, effective method for restoration of dystrophin expression and improvement of cardiac function in the GRMD canine and may be easily translatable to human DMD patients.


Asunto(s)
Empalme Alternativo , Dependovirus/genética , Distrofina/genética , Vectores Genéticos/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Animales , Línea Celular , Modelos Animales de Enfermedad , Perros , Distrofina/metabolismo , Ecocardiografía , Exones , Fibrosis , Expresión Génica , Orden Génico , Técnicas de Transferencia de Gen , Vectores Genéticos/farmacocinética , Genoma Viral , Humanos , Imagen por Resonancia Magnética , Distrofia Muscular de Duchenne/diagnóstico , Miocardio/patología , ARN Mensajero/metabolismo
19.
PLoS One ; 6(6): e20856, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21731628

RESUMEN

Duchenne muscular dystrophy (DMD) is a degenerative disorder affecting skeletal and cardiac muscle for which there is no effective therapy. Angiotension receptor blockade (ARB) has excellent therapeutic potential in DMD based on recent data demonstrating attenuation of skeletal muscle disease progression during 6-9 months of therapy in the mdx mouse model of DMD. Since cardiac-related death is major cause of mortality in DMD, it is important to evaluate the effect of any novel treatment on the heart. Therefore, we evaluated the long-term impact of ARB on both the skeletal muscle and cardiac phenotype of the mdx mouse. Mdx mice received either losartan (0.6 g/L) (n = 8) or standard drinking water (n = 9) for two years, after which echocardiography was performed to assess cardiac function. Skeletal muscle weight, morphology, and function were assessed. Fibrosis was evaluated in the diaphragm and heart by Trichrome stain and by determination of tissue hydroxyproline content. By the study endpoint, 88% of treated mice were alive compared to only 44% of untreated (p = 0.05). No difference in skeletal muscle morphology, function, or fibrosis was noted in losartan-treated animals. Cardiac function was significantly preserved with losartan treatment, with a trend towards reduction in cardiac fibrosis. We saw no impact on the skeletal muscle disease progression, suggesting that other pathways that trigger fibrosis dominate over angiotensin II in skeletal muscle long term, unlike the situation in the heart. Our study suggests that ARB may be an important prophylactic treatment for DMD-associated cardiomyopathy, but will not impact skeletal muscle disease.


Asunto(s)
Corazón/fisiopatología , Losartán/administración & dosificación , Losartán/uso terapéutico , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/mortalidad , Distrofia Muscular Animal/fisiopatología , Animales , Fibrosis , Corazón/efectos de los fármacos , Sistema de Conducción Cardíaco/diagnóstico por imagen , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/metabolismo , Sistema de Conducción Cardíaco/fisiopatología , Pruebas de Función Cardíaca , Losartán/farmacología , Masculino , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Distrofia Muscular Animal/tratamiento farmacológico , Miocardio/metabolismo , Miocardio/patología , Análisis de Supervivencia , Ultrasonografía
20.
Hum Gene Ther ; 22(12): 1499-509, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21787232

RESUMEN

Duchenne muscular dystrophy (DMD) is a lethal, X-linked recessive disease affecting 1 in 3,500 newborn boys for which there is no effective treatment or cure. One novel strategy that has therapeutic potential for DMD is inhibition of myostatin, a negative regulator of skeletal muscle mass that may also promote fibrosis. Therefore, our goal in this study was to evaluate systemic myostatin inhibition in the golden retriever model of DMD (GRMD). GRMD canines underwent liver-directed gene transfer of a self-complementary adeno-associated virus type 8 vector designed to express a secreted dominant-negative myostatin peptide (n = 4) and were compared with age-matched, untreated GRMD controls (n = 3). Dogs were followed with serial magnetic resonance imaging (MRI) for 13 months to assess cross-sectional area and volume of skeletal muscle, then euthanized so that tissue could be harvested for morphological and histological analysis. We found that systemic myostatin inhibition resulted in increased muscle mass in GRMD dogs as assessed by MRI and confirmed at tissue harvest. We also found that hypertrophy of type IIA fibers was largely responsible for the increased muscle mass and that reductions in serum creatine kinase and muscle fibrosis were associated with long-term myostatin inhibition in GRMD. This is the first report describing the effects of long-term, systemic myostatin inhibition in a large-animal model of DMD, and we believe that the simple and effective nature of our liver-directed gene-transfer strategy makes it an ideal candidate for evaluation as a novel therapeutic approach for DMD patients.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/uso terapéutico , Hígado/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/terapia , Miostatina/antagonistas & inhibidores , Animales , Western Blotting , Creatina Quinasa/genética , Creatina Quinasa/metabolismo , Perros , Fibrosis/metabolismo , Fibrosis/patología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hígado/patología , Imagen por Resonancia Magnética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/patología , Miostatina/genética , Miostatina/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...