Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38617374

RESUMEN

Background: Right ventricular dysfunction (RVD) portends increased death risk for heart failure (HF) and pulmonary arterial hypertension (PAH) patients, regardless of left ventricular function or etiology. In both, RVD arises from the chronic RV pressure overload, and represents advanced cardiopulmonary disease. RV remodeling responses and survival rates of HF and PAH patients, however, differ by sex. Men develop more severe RVD and die at younger ages than do women. Mechanistic details of this sexual dimorphism in RV remodeling are incompletely understood. We sought to elucidate the cardiac pathophysiology underlying the sex-specific RV remodeling phenotypes, RV failure (RVF) versus compensated RVD. Methods: We subjected male (M-) and female (F-) adult mice to moderate pulmonary artery banding (PAB) for 9wks. Mice underwent serial echocardiography, cardiac MRI, RV pressure-volume loop recordings, histologic and molecular analyses. Results: M-PAB developed severe RVD with RVF, increased RV collagen deposition and degradation, extracellular matrix (ECM) instability, and activation and recruitment of macrophages. Despite the same severity and chronicity of RV pressure overload, F-PAB had more stable ECM, lacked chronic inflammation, and developed mild RVD without RVF. Conclusions: ECM destabilization and chronic activation of recruited macrophages are associated with maladaptive RV remodeling and RVF in male PAB mice. Adaptive RV remodeling of female PAB mice lacked these histopathologic changes. Our findings suggest that these two pathophysiologic processes likely contribute to the sexual dimorphism of RV pressure overload remodeling. Further mechanistic studies are needed to assess their pathogenic roles and potential as targets for RVD therapy and RVF prevention. CLINICAL PERSPECTIVE: What is new?: In a mouse model of pure PH, males but not females showed an association between ECM instability, chronic inflammation with activation of recruited macrophages, and severe RV dysfunction and failure.What are the clinical implications?: In male HF and PH patients, enhancing ECM stability and countering the recruitment and activation of macrophages may help preserve RV function such that RVF can be prevented or delayed. Further preclinical mechanistic studies are needed to assess the therapeutic potential of such approaches. RESEARCH PERSPECTIVE: What new question does this study raise? What question should be addressed next?: What mechanisms regulate RV ECM stability and macrophage recruitment and activation in response to chronic RV pressure overload? Are these regulatory mechanisms dependent upon or independent of sex hormone signaling?

2.
Circ Res ; 132(10): 1290-1301, 2023 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-37167361

RESUMEN

From the onset of the pandemic, evidence of cardiac involvement in acute COVID-19 abounded. Cardiac presentations ranged from arrhythmias to ischemia, myopericarditis/myocarditis, ventricular dysfunction to acute heart failure, and even cardiogenic shock. Elevated serum cardiac troponin levels were prevalent among hospitalized patients with COVID-19; the higher the magnitude of troponin elevation, the greater the COVID-19 illness severity and in-hospital death risk. Whether these consequences were due to direct SARS-CoV-2 infection of cardiac cells or secondary to inflammatory responses steered early cardiac autopsy studies. SARS-CoV-2 was reportedly detected in endothelial cells, cardiac myocytes, and within the extracellular space. However, findings were inconsistent and different methodologies had their limitations. Initial autopsy reports suggested that SARS-CoV-2 myocarditis was common, setting off studies to find and phenotype inflammatory infiltrates in the heart. Nonetheless, subsequent studies rarely detected myocarditis. Microthrombi, cardiomyocyte necrosis, and inflammatory infiltrates without cardiomyocyte damage were much more common. In vitro and ex vivo experimental platforms have assessed the cellular tropism of SARS-CoV-2 and elucidated mechanisms of viral entry into and replication within cardiac cells. Data point to pericytes as the primary target of SARS-CoV-2 in the heart. Infection of pericytes can account for the observed pericyte and endothelial cell death, innate immune response, and immunothrombosis commonly observed in COVID-19 hearts. These processes are bidirectional and synergistic, rendering a definitive order of events elusive. Single-cell/nucleus analyses of COVID-19 myocardial tissue and isolated cardiac cells have provided granular data about the cellular composition and cell type-specific transcriptomic signatures of COVID-19 and microthrombi-positive COVID-19 hearts. Still, much remains unknown and more in vivo studies are needed. This review seeks to provide an overview of the current understanding of COVID-19 cardiac pathophysiology. Cell type-specific mechanisms and the studies that provided such insights will be highlighted. Given the unprecedented pace of COVID-19 research, more mechanistic details are sure to emerge since the writing of this review. Importantly, our current knowledge offers significant clues about the cardiac pathophysiology of long COVID-19, the increased postrecovery risk of cardiac events, and thus, the future landscape of cardiovascular disease.


Asunto(s)
COVID-19 , Cardiopatías , Miocarditis , Humanos , COVID-19/complicaciones , SARS-CoV-2 , Células Endoteliales , Mortalidad Hospitalaria , Síndrome Post Agudo de COVID-19 , Corazón , Troponina , Miocitos Cardíacos
3.
Curr Cardiol Rep ; 25(3): 171-184, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36897483

RESUMEN

PURPOSE OF REVIEW: Cardiac consequences occur in both acute COVID-19 and post-acute sequelae of COVID-19 (PASC). Here, we highlight the current understanding about COVID-19 cardiac effects, based upon clinical, imaging, autopsy, and molecular studies. RECENT FINDINGS: COVID-19 cardiac effects are heterogeneous. Multiple, concurrent cardiac histopathologic findings have been detected on autopsies of COVID-19 non-survivors. Microthrombi and cardiomyocyte necrosis are commonly detected. Macrophages often infiltrate the heart at high density but without fulfilling histologic criteria for myocarditis. The high prevalences of microthrombi and inflammatory infiltrates in fatal COVID-19 raise the concern that recovered COVID-19 patients may have similar but subclinical cardiac pathology. Molecular studies suggest that SARS-CoV-2 infection of cardiac pericytes, dysregulated immunothrombosis, and pro-inflammatory and anti-fibrinolytic responses underlie COVID-19 cardiac pathology. The extent and nature by which mild COVID-19 affects the heart is unknown. Imaging and epidemiologic studies of recovered COVID-19 patients suggest that even mild illness confers increased risks of cardiac inflammation, cardiovascular disorders, and cardiovascular death. The mechanistic details of COVID-19 cardiac pathophysiology remain under active investigation. The ongoing evolution of SARS-CoV-2 variants and vast numbers of recovered COVID-19 patients portend a burgeoning global cardiovascular disease burden. Our ability to prevent and treat cardiovascular disease in the future will likely depend on comprehensive understanding of COVID-19 cardiac pathophysiologic phenotypes.


Asunto(s)
COVID-19 , Cardiopatías , Miocarditis , Trombosis , Humanos , COVID-19/complicaciones , COVID-19/epidemiología , SARS-CoV-2/genética , Corazón/diagnóstico por imagen , Miocarditis/etiología , Cardiopatías/complicaciones , Trombosis/complicaciones
4.
Nat Cardiovasc Res ; 1(11): 1022-1038, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36424916

RESUMEN

Fight-or-flight responses involve ß-adrenergic-induced increases in heart rate and contractile force. In the present study, we uncover the primary mechanism underlying the heart's innate contractile reserve. We show that four protein kinase A (PKA)-phosphorylated residues in Rad, a calcium channel inhibitor, are crucial for controlling basal calcium current and essential for ß-adrenergic augmentation of calcium influx in cardiomyocytes. Even with intact PKA signaling to other proteins modulating calcium handling, preventing adrenergic activation of calcium channels in Rad-phosphosite-mutant mice (4SA-Rad) has profound physiological effects: reduced heart rate with increased pauses, reduced basal contractility, near-complete attenuation of ß-adrenergic contractile response and diminished exercise capacity. Conversely, expression of mutant calcium-channel ß-subunits that cannot bind 4SA-Rad is sufficient to enhance basal calcium influx and contractility to adrenergically augmented levels of wild-type mice, rescuing the failing heart phenotype of 4SA-Rad mice. Hence, disruption of interactions between Rad and calcium channels constitutes the foundation toward next-generation therapeutics specifically enhancing cardiac contractility.

5.
ASAIO J ; 68(2): e29-e33, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33883506

RESUMEN

Decompensated right ventricular failure (RVF) in patients with pulmonary hypertension (PH) is fatal, with limited treatment options. Novel mechanical circulatory support systems have therapeutic potential for RVF, but the development of these devices requires a large animal disease model that replicates the pathophysiology observed in humans. We previously reported an effective disease model of PH in sheep through ligation of the left pulmonary artery (PA) and progressive occlusion of the main PA. Herein, we report a case of acute decompensation with this model of chronic RVF. Gradual PA banding raised the RV pressure (maximum RV systolic/mean pressure = 95 mmHg/56 mmHg). Clinical findings and laboratory serum parameters suggested appropriate physiologic compensation for 7 weeks. However, mixed venous saturation declined precipitously on week 7, and creatinine increased markedly on week 9. By the 10th week, the animal developed dependent, subcutaneous edema. Subsequently, the animal expired during the induction of general anesthesia. Post-mortem evaluation revealed several liters of pleural effusion and ascites, RV dilatation, eccentric RV hypertrophy, and myocardial fibrosis. The presented case supports this model's relevance to the human pathophysiology of RVF secondary to PH and its value in the development of novel devices, therapeutics, and interventions.


Asunto(s)
Insuficiencia Cardíaca , Hipertensión Pulmonar , Disfunción Ventricular Derecha , Animales , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/etiología , Humanos , Hipertensión Pulmonar/etiología , Hipertrofia Ventricular Derecha/etiología , Arteria Pulmonar , Ovinos , Disfunción Ventricular Derecha/etiología
6.
JCI Insight ; 7(2)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-34905515

RESUMEN

Acute cardiac injury is prevalent in critical COVID-19 and associated with increased mortality. Its etiology remains debated, as initially presumed causes - myocarditis and cardiac necrosis - have proved uncommon. To elucidate the pathophysiology of COVID-19-associated cardiac injury, we conducted a prospective study of the first 69 consecutive COVID-19 decedents at CUIMC in New York City. Of 6 acute cardiac histopathologic features, presence of microthrombi was the most commonly detected among our cohort. We tested associations of cardiac microthrombi with biomarkers of inflammation, cardiac injury, and fibrinolysis and with in-hospital antiplatelet therapy, therapeutic anticoagulation, and corticosteroid treatment, while adjusting for multiple clinical factors, including COVID-19 therapies. Higher peak erythrocyte sedimentation rate and C-reactive protein were independently associated with increased odds of microthrombi, supporting an immunothrombotic etiology. Using single-nuclei RNA-sequencing analysis on 3 patients with and 4 patients without cardiac microthrombi, we discovered an enrichment of prothrombotic/antifibrinolytic, extracellular matrix remodeling, and immune-potentiating signaling among cardiac fibroblasts in microthrombi-positive, relative to microthrombi-negative, COVID-19 hearts. Non-COVID-19, nonfailing hearts were used as reference controls. Our study identifies a specific transcriptomic signature in cardiac fibroblasts as a salient feature of microthrombi-positive COVID-19 hearts. Our findings warrant further mechanistic study as cardiac fibroblasts may represent a potential therapeutic target for COVID-19-associated cardiac microthrombi.


Asunto(s)
COVID-19 , Lesiones Cardíacas , RNA-Seq , SARS-CoV-2/metabolismo , Trombosis , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/genética , COVID-19/metabolismo , COVID-19/patología , Femenino , Lesiones Cardíacas/genética , Lesiones Cardíacas/metabolismo , Lesiones Cardíacas/patología , Humanos , Masculino , Persona de Mediana Edad , Miocardio/metabolismo , Miocardio/patología , Estudios Prospectivos , Trombosis/genética , Trombosis/metabolismo , Trombosis/patología
8.
Circ Heart Fail ; 14(6)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34422205

RESUMEN

Right ventricular dysfunction is a hallmark of advanced pulmonary vascular, lung parenchymal, and left heart disease, yet the underlying mechanisms that govern (mal)adaptation remain incompletely characterized. Owing to the knowledge gaps in our understanding of the right ventricle (RV) in health and disease, the National Heart, Lung, and Blood Institute (NHLBI) commissioned a working group to identify current challenges in the field. These included a need to define and standardize normal RV structure and function in populations; access to RV tissue for research purposes and the development of complex experimental platforms that recapitulate the in vivo environment; and the advancement of imaging and invasive methodologies to study the RV within basic, translational, and clinical research programs. Specific recommendations were provided, including a call to incorporate precision medicine and innovations in prognosis, diagnosis, and novel RV therapeutics for patients with pulmonary vascular disease.


Asunto(s)
Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/terapia , Hipertensión Pulmonar/terapia , Circulación Pulmonar/fisiología , Función Ventricular Derecha/inmunología , Insuficiencia Cardíaca/fisiopatología , Humanos , Hipertensión Pulmonar/diagnóstico , Hipertensión Pulmonar/fisiopatología , National Heart, Lung, and Blood Institute (U.S.) , Estados Unidos , Disfunción Ventricular Derecha/fisiopatología
9.
bioRxiv ; 2021 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-34341789

RESUMEN

Cardiac injury is associated with critical COVID-19, yet its etiology remains debated. To elucidate the pathogenic mechanisms of COVID-19-associated cardiac injury, we conducted a single-center prospective cohort study of 69 COVID-19 decedents. Of six cardiac histopathologic features, microthrombi was the most commonly detected (n=48, 70%). We tested associations of cardiac microthrombi with biomarkers of inflammation, cardiac injury, and fibrinolysis and with in-hospital antiplatelet therapy, therapeutic anticoagulation, and corticosteroid treatment, while adjusting for multiple clinical factors, including COVID-19 therapies. Higher peak ESR and CRP during hospitalization were independently associated with higher odds of microthrombi. Using single nuclei RNA-sequence analysis, we discovered an enrichment of pro-thrombotic/anti-fibrinolytic, extracellular matrix remodeling, and immune-potentiating signaling amongst cardiac fibroblasts in microthrombi-positive COVID-19 hearts relative to microthrombi-negative COVID-19. Non-COVID-19 non-failing hearts were used as reference controls. Our cumulative findings identify the specific transcriptomic changes in cardiac fibroblasts as salient features of COVID-19-associated cardiac microthrombi.

10.
J Vis Exp ; (173)2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34338672

RESUMEN

Decompensated right ventricular failure (RVF) in pulmonary hypertension (PH) is fatal, with limited medical treatment options. Developing and testing novel therapeutics for PH requires a clinically relevant large animal model of increased pulmonary vascular resistance and RVF. This manuscript discusses the latest development of the previously published ovine PH-RVF model that utilizes left pulmonary artery (PA) ligation and main PA occlusion. This model of PH-RVF is a versatile platform to control not only the disease severity but also the RV's phenotypic response. Adult sheep (60-80 kg) underwent left PA (LPA) ligation, placement of main PA cuff, and insertion of RV pressure monitor. PA cuff and RV pressure monitor were connected to subcutaneous ports. Subjects underwent progressive PA banding twice per week for 9 weeks with sequential measures of RV pressure, PA cuff pressures, and mixed venous blood gas (SvO2). At the initiation and endpoint of this model, ventricular function and dimensions were assessed using echocardiography. In a representative group of 12 animal subjects, RV mean and systolic pressure increased from 28 ± 5 and 57 ± 7 mmHg at week 1, respectively, to 44 ± 7 and 93 ± 18 mmHg (mean ± standard deviation) by week 9. Echocardiography demonstrated characteristic findings of PH-RVF, notably RV dilation, increased wall thickness, and septal bowing. The longitudinal trend of SvO2 and PA cuff pressure demonstrates that the rate of PA banding can be titrated to elicit varying RV phenotypes. A faster PA banding strategy led to a precipitous decline in SvO2 < 65%, indicating RV decompensation, whereas a slower, more paced strategy led to the maintenance of physiologic SvO2 at 70%-80%. One animal that experienced the accelerated strategy developed several liters of pleural effusion and ascites by week 9. This chronic PH-RVF model provides a valuable tool for studying molecular mechanisms, developing diagnostic biomarkers, and enabling therapeutic innovation to manage RV adaptation and maladaptation from PH.


Asunto(s)
Insuficiencia Cardíaca , Hipertensión Pulmonar , Disfunción Ventricular Derecha , Animales , Modelos Animales de Enfermedad , Hipertensión Pulmonar/etiología , Arteria Pulmonar/cirugía , Ovinos , Disfunción Ventricular Derecha/etiología , Función Ventricular Derecha
11.
Nature ; 595(7865): 114-119, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33915568

RESUMEN

Respiratory failure is the leading cause of death in patients with severe SARS-CoV-2 infection1,2, but the host response at the lung tissue level is poorly understood. Here we performed single-nucleus RNA sequencing of about 116,000 nuclei from the lungs of nineteen individuals who died of COVID-19 and underwent rapid autopsy and seven control individuals. Integrated analyses identified substantial alterations in cellular composition, transcriptional cell states, and cell-to-cell interactions, thereby providing insight into the biology of lethal COVID-19. The lungs from individuals with COVID-19 were highly inflamed, with dense infiltration of aberrantly activated monocyte-derived macrophages and alveolar macrophages, but had impaired T cell responses. Monocyte/macrophage-derived interleukin-1ß and epithelial cell-derived interleukin-6 were unique features of SARS-CoV-2 infection compared to other viral and bacterial causes of pneumonia. Alveolar type 2 cells adopted an inflammation-associated transient progenitor cell state and failed to undergo full transition into alveolar type 1 cells, resulting in impaired lung regeneration. Furthermore, we identified expansion of recently described CTHRC1+ pathological fibroblasts3 contributing to rapidly ensuing pulmonary fibrosis in COVID-19. Inference of protein activity and ligand-receptor interactions identified putative drug targets to disrupt deleterious circuits. This atlas enables the dissection of lethal COVID-19, may inform our understanding of long-term complications of COVID-19 survivors, and provides an important resource for therapeutic development.


Asunto(s)
COVID-19/patología , COVID-19/virología , Pulmón/patología , SARS-CoV-2/patogenicidad , Análisis de la Célula Individual , Anciano , Anciano de 80 o más Años , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/virología , Atlas como Asunto , Autopsia , COVID-19/inmunología , Estudios de Casos y Controles , Femenino , Fibroblastos/patología , Fibrosis/patología , Fibrosis/virología , Humanos , Inflamación/patología , Inflamación/virología , Macrófagos/patología , Macrófagos/virología , Macrófagos Alveolares/patología , Macrófagos Alveolares/virología , Masculino , Persona de Mediana Edad , Células Plasmáticas/inmunología , Linfocitos T/inmunología
12.
Circ Res ; 128(8): 1214-1236, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33856918

RESUMEN

A pandemic of historic impact, coronavirus disease 2019 (COVID-19) has potential consequences on the cardiovascular health of millions of people who survive infection worldwide. Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), the etiologic agent of COVID-19, can infect the heart, vascular tissues, and circulating cells through ACE2 (angiotensin-converting enzyme 2), the host cell receptor for the viral spike protein. Acute cardiac injury is a common extrapulmonary manifestation of COVID-19 with potential chronic consequences. This update provides a review of the clinical manifestations of cardiovascular involvement, potential direct SARS-CoV-2 and indirect immune response mechanisms impacting the cardiovascular system, and implications for the management of patients after recovery from acute COVID-19 infection.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/virología , Enfermedades Cardiovasculares/virología , Miocitos Cardíacos/virología , SARS-CoV-2/fisiología , Internalización del Virus , Biomarcadores/metabolismo , COVID-19/complicaciones , COVID-19/epidemiología , COVID-19/terapia , Cardiomiopatías/virología , Expresión Génica , Humanos , Sistema Inmunológico/fisiología , Miocardio/enzimología , Miocitos Cardíacos/enzimología , Neuropilina-1/metabolismo , Activación Plaquetaria , ARN Mensajero/metabolismo , Sistema Renina-Angiotensina/fisiología , Volver al Deporte , Factores de Riesgo , SARS-CoV-2/ultraestructura , Glicoproteína de la Espiga del Coronavirus/metabolismo , Troponina/metabolismo , Remodelación Ventricular , Acoplamiento Viral , Internalización del Virus/efectos de los fármacos
13.
Clin Transplant ; 34(10): e14028, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32623785

RESUMEN

Light-chain (AL) cardiac amyloidosis (CA) has a worse prognosis than transthyretin (ATTR) CA. In this single-center study, we compared post-heart transplant (OHT, orthotopic heart transplantation) survival for AL and ATTR amyloidosis, hypothesizing that these differences would persist post-OHT. Thirty-nine patients with CA (AL, n = 18; ATTR, n = 21) and 1023 non-amyloidosis subjects undergoing OHT were included. Cox proportional hazards modeling was used to evaluate the impact of amyloid subtype and era (early era: from 2001 to 2007; late era: from 2008 to 2018) on survival post-OHT. Survival for non-amyloid patients was greater than ATTR (P = .034) and AL (P < .001) patients in the early era. One, 3-, and 5-year survival rates were higher for ATTR patients than AL patients in the early era (100% vs 75%, 67% vs 50%, and 67% vs 33%, respectively, for ATTR and AL patients). Survival in the non-amyloid cohort was 87% at 1 year, 81% at 3 years, and 76% at 5 years post-OHT. In the late era, AL and ATTR patients had unadjusted 1-year, 3-year, and 5-year survival rates of 100%, which was comparable to non-amyloid patients (90% vs 84% vs 81%). Overall, these findings demonstrate that in the current era, differences in post-OHT survival for AL compared to ATTR are diminishing; OHT outcomes for selected patients with CA do not differ from non-amyloidosis patients.


Asunto(s)
Neuropatías Amiloides Familiares , Amiloidosis , Cardiomiopatías , Trasplante de Corazón , Neuropatías Amiloides Familiares/cirugía , Cardiomiopatías/etiología , Humanos , Prealbúmina , Pronóstico , Tasa de Supervivencia
14.
Circ Heart Fail ; 13(1): e006426, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31916447

RESUMEN

BACKGROUND: The failing right ventricle (RV) does not respond like the left ventricle (LV) to guideline-directed medical therapy of heart failure, perhaps due to interventricular differences in their molecular pathophysiology. METHODS: Using the canine tachypacing-induced biventricular heart failure (HF) model, we tested the hypothesis that interventricular differences in microRNAs (miRs) expression distinguish failing RV from failing LV. RESULTS: Severe RV dysfunction was indicated by elevated end-diastolic pressure (11.3±2.5 versus 5.7±2.0 mm Hg; P<0.0001) and diminished fractional area change (24.9±7.1 versus 48.0±3.6%; P<0.0001) relative to prepacing baselines. Microarray analysis of ventricular tissue revealed that miR-21 and miR-221, 2 activators of profibrotic and proliferative processes, increased the most, at 4- and 2-fold, respectively, in RV-HF versus RV-Control. Neither miR-21 or miR-221 was statistically significantly different in LV-HF versus LV-Control. These changes were accompanied by more extensive fibrosis in RV-HF than LV-HF. To test whether miR-21 and miR-221 upregulation is specific to RV cellular response to mechanical and hormonal stimuli associated with HF, we subjected fibroblasts and cardiomyocytes isolated from normal canine RV and LV to cyclic overstretch and aldosterone. These 2 stressors markedly upregulated miR-21 and miR-221 in RV fibroblasts but not in LV fibroblasts nor cardiomyocytes of either ventricle. Furthermore, miR-21/221 knockdown significantly attenuated RV but not LV fibroblast proliferation. CONCLUSIONS: We identified a novel, biological difference between RV and LV fibroblasts that might underlie distinctions in pathological remodeling of the RV in biventricular HF.


Asunto(s)
Fibroblastos/metabolismo , Insuficiencia Cardíaca/metabolismo , Ventrículos Cardíacos/metabolismo , MicroARNs/metabolismo , Disfunción Ventricular Derecha/metabolismo , Animales , Perros , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/fisiopatología , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Miocitos Cardíacos/metabolismo , Regulación hacia Arriba , Disfunción Ventricular Derecha/fisiopatología , Función Ventricular Izquierda/fisiología
15.
JCI Insight ; 52019 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-31021818

RESUMEN

Right ventricular dysfunction is highly prevalent across cardiopulmonary diseases and independently predicts death in both heart failure (HF) and pulmonary hypertension (PH). Progression towards right ventricular failure (RVF) can occur in spite of optimal medical treatment of HF or PH, highlighting current insufficient understanding of RVF molecular pathophysiology. To identify molecular mechanisms that may distinctly underlie RVF, we investigated the cardiac ventricular transcriptome of advanced HF patients, with and without RVF. Using an integrated systems genomic and functional biology approach, we identified an RVF-specific gene module, for which WIPI1 served as a hub and HSPB6 and MAP4 as drivers, and confirmed the ventricular specificity of Wipi1, Hspb6, and Map4 transcriptional changes in adult murine models of pressure overload induced RV- versus LV- failure. We uncovered a shift towards non-canonical autophagy in the failing RV that correlated with RV-specific Wipi1 upregulation. In vitro siRNA silencing of Wipi1 in neonatal rat ventricular myocytes limited non-canonical autophagy and blunted aldosterone-induced mitochondrial superoxide levels. Our findings suggest that Wipi1 regulates mitochondrial oxidative signaling and non-canonical autophagy in cardiac myocytes. Together with our human transcriptomic analysis and corroborating studies in an RVF mouse model, these data render Wipi1 a potential target for RV-directed HF therapy.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Redes Reguladoras de Genes , Insuficiencia Cardíaca/genética , Ventrículos Cardíacos/patología , Proteínas de la Membrana/metabolismo , Disfunción Ventricular Derecha/genética , Adulto , Animales , Animales Recién Nacidos , Autofagia/genética , Proteínas Relacionadas con la Autofagia/genética , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Proteínas del Choque Térmico HSP20/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/cirugía , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/cirugía , Humanos , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , Mitocondrias/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/patología , Estrés Oxidativo/genética , Cultivo Primario de Células , RNA-Seq , Transducción de Señal/genética , Regulación hacia Arriba , Disfunción Ventricular Derecha/patología
16.
J Am Heart Assoc ; 7(9)2018 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-29686029

RESUMEN

BACKGROUND: Outcomes in peripartum cardiomyopathy (PPCM) vary. We sought to determine whether severity of left or right ventricular dysfunction (RVD) at PPCM diagnosis differentially associates with adverse outcomes. METHODS AND RESULTS: We conducted a single-center retrospective cohort study of 53 patients with PPCM. The primary outcome was a composite of left ventricular assist device implantation, cardiac transplantation, or death. We used Kaplan-Meier curves to examine event-free survival and Cox proportional hazards models to examine associations of left ventricular (LV) ejection fraction <30%, LV end-diastolic diameter ≥60 mm, and moderate-to-severe RVD at PPCM diagnosis with the primary outcome. Median (interquartile range) follow-up time was 3.6 (1.4-7.3) years. Seventeen patients (32%) experienced the primary outcome, of whom 11 had moderate-to-severe RVD at time of PPCM diagnosis. Overall event-free survival differed by initial RVD severity and LV ejection fraction <30%, but not by LV end-diastolic diameter ≥60 mm. In univariable analyses, LV ejection fraction <30% and moderate-to-severe RVD were associated with the outcome (hazard ratios [95% confidence intervals] of 4.85 [1.11-21.3] and 4.26 [1.47-11.6], respectively). In a multivariable model with LV ejection fraction <30%, LV end-diastolic diameter ≥60 mm, and moderate-to-severe RVD, only moderate-to-severe RVD was independently associated with the outcome (hazard ratio [95% confidence interval], 3.21 [1.13-9.10]). Although most outcomes occurred within the first year, nearly a third occurred years after PPCM diagnosis. CONCLUSIONS: Initial moderate-to-severe RVD is associated with a more advanced cardiomyopathy phenotype and increased risk of adverse outcomes in PPCM, within and beyond the first year of diagnosis. By identifying a worse PPCM phenotype, initial moderate-to-severe RVD may prompt earlier consideration of advanced heart replacement therapies.


Asunto(s)
Cardiomiopatías/fisiopatología , Disfunción Ventricular Derecha/fisiopatología , Función Ventricular Derecha , Adulto , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/mortalidad , Cardiomiopatías/terapia , Femenino , Trasplante de Corazón , Corazón Auxiliar , Humanos , Periodo Periparto , Fenotipo , Embarazo , Supervivencia sin Progresión , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Índice de Severidad de la Enfermedad , Factores de Tiempo , Disfunción Ventricular Derecha/diagnóstico por imagen , Disfunción Ventricular Derecha/mortalidad , Disfunción Ventricular Derecha/terapia , Función Ventricular Izquierda , Adulto Joven
17.
Nature ; 545(7652): 93-97, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28445457

RESUMEN

Mitochondrial calcium (mCa2+) has a central role in both metabolic regulation and cell death signalling, however its role in homeostatic function and disease is controversial. Slc8b1 encodes the mitochondrial Na+/Ca2+ exchanger (NCLX), which is proposed to be the primary mechanism for mCa2+ extrusion in excitable cells. Here we show that tamoxifen-induced deletion of Slc8b1 in adult mouse hearts causes sudden death, with less than 13% of affected mice surviving after 14 days. Lethality correlated with severe myocardial dysfunction and fulminant heart failure. Mechanistically, cardiac pathology was attributed to mCa2+ overload driving increased generation of superoxide and necrotic cell death, which was rescued by genetic inhibition of mitochondrial permeability transition pore activation. Corroborating these findings, overexpression of NCLX in the mouse heart by conditional transgenesis had the beneficial effect of augmenting mCa2+ clearance, preventing permeability transition and protecting against ischaemia-induced cardiomyocyte necrosis and heart failure. These results demonstrate the essential nature of mCa2+ efflux in cellular function and suggest that augmenting mCa2+ efflux may be a viable therapeutic strategy in disease.


Asunto(s)
Calcio/metabolismo , Homeostasis , Mitocondrias/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Animales , Supervivencia Celular , Muerte Súbita , Femenino , Eliminación de Gen , Células HeLa , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Masculino , Ratones , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Necrosis , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Intercambiador de Sodio-Calcio/genética , Superóxidos/metabolismo , Tamoxifeno/farmacología , Remodelación Ventricular
18.
N Engl J Med ; 374(3): 233-41, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26735901

RESUMEN

Background Peripartum cardiomyopathy shares some clinical features with idiopathic dilated cardiomyopathy, a disorder caused by mutations in more than 40 genes, including TTN, which encodes the sarcomere protein titin. Methods In 172 women with peripartum cardiomyopathy, we sequenced 43 genes with variants that have been associated with dilated cardiomyopathy. We compared the prevalence of different variant types (nonsense, frameshift, and splicing) in these women with the prevalence of such variants in persons with dilated cardiomyopathy and with population controls. Results We identified 26 distinct, rare truncating variants in eight genes among women with peripartum cardiomyopathy. The prevalence of truncating variants (26 in 172 [15%]) was significantly higher than that in a reference population of 60,706 persons (4.7%, P=1.3×10(-7)) but was similar to that in a cohort of patients with dilated cardiomyopathy (55 of 332 patients [17%], P=0.81). Two thirds of identified truncating variants were in TTN, as seen in 10% of the patients and in 1.4% of the reference population (P=2.7×10(-10)); almost all TTN variants were located in the titin A-band. Seven of the TTN truncating variants were previously reported in patients with idiopathic dilated cardiomyopathy. In a clinically well-characterized cohort of 83 women with peripartum cardiomyopathy, the presence of TTN truncating variants was significantly correlated with a lower ejection fraction at 1-year follow-up (P=0.005). Conclusions The distribution of truncating variants in a large series of women with peripartum cardiomyopathy was remarkably similar to that found in patients with idiopathic dilated cardiomyopathy. TTN truncating variants were the most prevalent genetic predisposition in each disorder.


Asunto(s)
Cardiomiopatías/genética , Cardiomiopatía Dilatada/genética , Conectina/genética , Predisposición Genética a la Enfermedad , Mutación , Periodo Periparto , Complicaciones Cardiovasculares del Embarazo/genética , Adulto , Estudios de Casos y Controles , Conectina/química , Femenino , Humanos , Embarazo , Isoformas de Proteínas , Análisis de Secuencia de ADN , Volumen Sistólico
19.
J Am Coll Cardiol ; 65(2): 133-47, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25593054

RESUMEN

BACKGROUND: Functional decline in stem cell-mediated regeneration contributes to aging associated with cellular senescence in c-kit+ cardiac progenitor cells (CPCs). Clinical implementation of CPC-based therapy in elderly patients would benefit tremendously from understanding molecular characteristics of senescence to antagonize aging. Nucleostemin (NS) is a nucleolar protein regulating stem cell proliferation and pluripotency. OBJECTIVES: This study sought to demonstrate that NS preserves characteristics associated with "stemness" in CPCs and antagonizes myocardial senescence and aging. METHODS: CPCs isolated from human fetal (fetal human cardiac progenitor cell [FhCPC]) and adult failing (adult human cardiac progenitor cell [AhCPC]) hearts, as well as young (young cardiac progenitor cell [YCPC]) and old mice (old cardiac progenitor cell [OCPC]), were studied for senescence characteristics and NS expression. Heterozygous knockout mice with 1 functional allele of NS (NS+/-) were used to demonstrate that NS preserves myocardial structure and function and slows characteristics of aging. RESULTS: NS expression is decreased in AhCPCs relative to FhCPCs, correlating with lowered proliferation potential and shortened telomere length. AhCPC characteristics resemble those of OCPCs, which have a phenotype induced by NS silencing, resulting in cell flattening, senescence, multinucleated cells, decreased S-phase progression, diminished expression of stemness markers, and up-regulation of p53 and p16. CPC senescence resulting from NS loss is partially p53 dependent and is rescued by concurrent silencing of p53. Mechanistically, NS induction correlates with Pim-1 kinase-mediated stabilization of c-Myc. Engineering OCPCs and AhCPCs to overexpress NS decreases senescent and multinucleated cells, restores morphology, and antagonizes senescence, thereby preserving phenotypic properties of "stemness." Early cardiac aging with a decline in cardiac function, an increase in senescence markers p53 and p16, telomere attrition, and accompanied CPC exhaustion is evident in NS+/- mice. CONCLUSIONS: Youthful properties and antagonism of senescence in CPCs and the myocardium are consistent with a role for NS downstream from Pim-1 signaling that enhances cardiac regeneration.


Asunto(s)
Proteínas Portadoras/biosíntesis , Senescencia Celular/fisiología , Miocardio/citología , Proteínas Nucleares/biosíntesis , Rejuvenecimiento/fisiología , Células Madre/citología , Animales , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al GTP , Humanos , Masculino , Ratones , Ratones Noqueados , Miocardio/metabolismo , Proteínas de Unión al ARN , Células Madre/metabolismo
20.
Basic Res Cardiol ; 110(1): 456, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25480109

RESUMEN

The ß1-adrenergic antagonist metoprolol improves cardiac function in animals and patients with chronic heart failure, isolated mitral regurgitation (MR), and ischemic heart disease, though the molecular mechanisms remain incompletely understood. Metoprolol has been reported to upregulate cardiac expression of ß3-adrenergic receptors (ß3AR) in animal models. Myocardial ß3AR signaling via neuronal nitric oxide synthase (nNOS) activation has recently emerged as a cardioprotective pathway. We tested whether chronic ß1-adrenergic blockade with metoprolol enhances myocardial ß3AR coupling with nitric oxide-stimulated cyclic guanosine monophosphate (ß3AR/NO-cGMP) signaling in the MR-induced, volume-overloaded heart. We compared the expression, distribution, and inducible activation of ß3AR/NO-cGMP signaling proteins within myocardial membrane microdomains in dogs (canines) with surgically induced MR, those also treated with metoprolol succinate (MR+ßB), and unoperated controls. ß3AR mRNA transcripts, normalized to housekeeping gene RPLP1, increased 4.4 × 10(3)- and 3.2 × 10(2)-fold in MR and MR+ßB hearts, respectively, compared to Control. Cardiac ß3AR expression was increased 1.4- and nearly twofold in MR and MR+ßB, respectively, compared to Control. ß3AR was detected within caveolae-enriched lipid rafts (Cav3(+)LR) and heavy density, non-lipid raft membrane (NLR) across all groups. However, in vitro selective ß3AR stimulation with BRL37344 (BRL) triggered cGMP production within only NLR of MR+ßB. BRL induced Ser (1412) phosphorylation of nNOS within NLR of MR+ßB, but not Control or MR, consistent with detection of NLR-specific ß3AR/NO-cGMP coupling. Treatment with metoprolol prevented MR-associated oxidation of NO biosensor soluble guanylyl cyclase (sGC) within NLR. Metoprolol therapy also prevented MR-induced relocalization of sGCß1 subunit away from caveolae, suggesting preserved NO-sGC-cGMP signaling, albeit without coupling to ß3AR, within MR+ßB caveolae. Chronic ß1-blockade is associated with myocardial ß3AR/NO-cGMP coupling in a microdomain-specific fashion. Our canine study suggests that microdomain-targeted enhancement of myocardial ß3AR/NO-cGMP signaling may explain, in part, ß1-adrenergic antagonist-mediated preservation of cardiac function in the volume-overloaded heart.


Asunto(s)
Antagonistas de Receptores Adrenérgicos beta 1/farmacología , GMP Cíclico/fisiología , Insuficiencia de la Válvula Mitral/tratamiento farmacológico , Óxido Nítrico/fisiología , Receptores Adrenérgicos beta 3/fisiología , Transducción de Señal/fisiología , Antagonistas de Receptores Adrenérgicos beta 1/uso terapéutico , Animales , Enfermedad Crónica , Perros , Etanolaminas/farmacología , Guanilato Ciclasa/metabolismo , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/fisiología , Metoprolol/farmacología , Insuficiencia de la Válvula Mitral/fisiopatología , Óxido Nítrico Sintasa de Tipo I/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Guanilil Ciclasa Soluble , Función Ventricular Izquierda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...