Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 135(17)2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35946425

RESUMEN

Mitral and tricuspid valves are essential for unidirectional blood flow in the heart. They are derived from similar cell sources, and yet congenital dysplasia affecting both valves is clinically rare, suggesting the presence of differential regulatory mechanisms underlying their development. Here, we specifically inactivated Dicer1 in the endocardium during cardiogenesis and found that Dicer1 deletion caused congenital mitral valve stenosis and regurgitation, whereas it had no impact on other valves. We showed that hyperplastic mitral valves were caused by abnormal condensation and extracellular matrix (ECM) remodeling. Our single-cell RNA sequencing analysis revealed impaired maturation of mesenchymal cells and abnormal expression of ECM genes in mutant mitral valves. Furthermore, expression of a set of miRNAs that target ECM genes was significantly lower in tricuspid valves compared to mitral valves, consistent with the idea that the miRNAs are differentially required for mitral and tricuspid valve development. We thus reveal miRNA-mediated gene regulation as a novel molecular mechanism that differentially regulates mitral and tricuspid valve development, thereby enhancing our understanding of the non-association of inborn mitral and tricuspid dysplasia observed clinically.


Asunto(s)
MicroARNs , Válvula Tricúspide , Matriz Extracelular/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Válvula Mitral , Válvula Tricúspide/anomalías
2.
Nucleic Acids Res ; 50(4): 2270-2286, 2022 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-35137168

RESUMEN

Human genetic studies identified a strong association between loss of function mutations in RBFOX2 and hypoplastic left heart syndrome (HLHS). There are currently no Rbfox2 mouse models that recapitulate HLHS. Therefore, it is still unknown how RBFOX2 as an RNA binding protein contributes to heart development. To address this, we conditionally deleted Rbfox2 in embryonic mouse hearts and found profound defects in cardiac chamber and yolk sac vasculature formation. Importantly, our Rbfox2 conditional knockout mouse model recapitulated several molecular and phenotypic features of HLHS. To determine the molecular drivers of these cardiac defects, we performed RNA-sequencing in Rbfox2 mutant hearts and identified dysregulated alternative splicing (AS) networks that affect cell adhesion to extracellular matrix (ECM) mediated by Rho GTPases. We identified two Rho GTPase cycling genes as targets of RBFOX2. Modulating AS of these two genes using antisense oligos led to cell cycle and cell-ECM adhesion defects. Consistently, Rbfox2 mutant hearts displayed cell cycle defects and inability to undergo endocardial-mesenchymal transition, processes dependent on cell-ECM adhesion and that are seen in HLHS. Overall, our work not only revealed that loss of Rbfox2 leads to heart development defects resembling HLHS, but also identified RBFOX2-regulated AS networks that influence cell-ECM communication vital for heart development.


Asunto(s)
Empalme Alternativo , Corazón/embriología , Factores de Empalme de ARN/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Organogénesis , ARN/metabolismo , Factores de Empalme de ARN/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
3.
J Mol Cell Cardiol ; 183: 1-13, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37579636

RESUMEN

Cardiovascular calcification can occur in vascular and valvular structures and is commonly associated with calcium deposition and tissue mineralization leading to stiffness and dysfunction. Patients with chronic kidney disease and associated hyperphosphatemia have an elevated risk for coronary artery calcification (CAC) and calcific aortic valve disease (CAVD). However, there is mounting evidence to suggest that the susceptibility and pathobiology of calcification in these two cardiovascular structures may be different, yet clinically they are similarly treated. To better understand diversity in molecular and cellular processes that underlie hyperphosphatemia-induced calcification in vascular and valvular structures, we exposed aortic vascular smooth muscle cells (AVSMCs) and aortic valve interstitial cells (AVICs) to high (2.5 mM) phosphate (Ph) conditions in vitro, and examined cell-specific responses. To further identify hyperphosphatemic-specific responses, parallel studies were performed using osteogenic media (OM) as an alternative calcific stimulus. Consistent with clinical observations made by others, we show that AVSMCs are more susceptible to calcification than AVICs. In addition, bulk RNA-sequencing reveals that AVSMCs and AVICs activate robust ossification-programs in response to high phosphate or OM treatments, however, the signaling pathways, cellular processes and osteogenic-associated markers involved are cell- and treatment-specific. For example, compared to VSMCs, VIC-mediated calcification involves biological processes related to osteo-chondro differentiation and down regulation of 'actin cytoskeleton'-related genes, that are not observed in VSMCs. Furthermore, hyperphosphatemic-induced calcification in AVICs and AVSMCs is independent of P13K signaling, which plays a role in OM-treated cells. Together, this study provides a wealth of information suggesting that the pathogenesis of cardiovascular calcifications is significantly more diverse than previously appreciated.


Asunto(s)
Estenosis de la Válvula Aórtica , Calcinosis , Hiperfosfatemia , Calcificación Vascular , Humanos , Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/metabolismo , Calcinosis/metabolismo , Músculo Liso Vascular/patología , Hiperfosfatemia/metabolismo , Hiperfosfatemia/patología , Células Cultivadas , Fosfatos , Calcificación Vascular/metabolismo
4.
Circ Res ; 128(9): 1330-1343, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33914609

RESUMEN

Aortic stenosis (AS) remains one of the most common forms of valve disease, with significant impact on patient survival. The disease is characterized by left ventricular outflow obstruction and encompasses a series of stenotic lesions starting from the left ventricular outflow tract to the descending aorta. Obstructions may be subvalvar, valvar, or supravalvar and can be present at birth (congenital) or acquired later in life. Bicuspid aortic valve, whereby the aortic valve forms with two instead of three cusps, is the most common cause of AS in younger patients due to primary anatomic narrowing of the valve. In addition, the secondary onset of premature calcification, likely induced by altered hemodynamics, further obstructs left ventricular outflow in bicuspid aortic valve patients. In adults, degenerative AS involves progressive calcification of an anatomically normal, tricuspid aortic valve and is attributed to lifelong exposure to multifactoral risk factors and physiological wear-and-tear that negatively impacts valve structure-function relationships. AS continues to be the most frequent valvular disease that requires intervention, and aortic valve replacement is the standard treatment for patients with severe or symptomatic AS. While the positive impacts of surgical interventions are well documented, the financial burden, the potential need for repeated procedures, and operative risks are substantial. In addition, the clinical management of asymptomatic patients remains controversial. Therefore, there is a critical need to develop alternative approaches to prevent the progression of left ventricular outflow obstruction, especially in valvar lesions. This review summarizes our current understandings of AS cause; beginning with developmental origins of congenital valve disease, and leading into the multifactorial nature of AS in the adult population.


Asunto(s)
Estenosis de la Válvula Aórtica/etiología , Factores de Edad , Animales , Válvula Aórtica/anomalías , Válvula Aórtica/anatomía & histología , Válvula Aórtica/fisiopatología , Válvula Aórtica/cirugía , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/fisiopatología , Estenosis de la Válvula Aórtica/cirugía , Calcinosis/etiología , Progresión de la Enfermedad , Humanos , Ilustración Médica , Ratones , Factores de Riesgo , Obstrucción del Flujo Ventricular Externo/etiología , Obstrucción del Flujo Ventricular Externo/prevención & control
6.
Hum Mol Genet ; 29(16): 2723-2735, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32720677

RESUMEN

The Forkhead Box C1 (FOXC1) gene encodes a forkhead/winged helix transcription factor involved in embryonic development. Mutations in this gene cause dysgenesis of the anterior segment of the eye, most commonly Axenfeld-Rieger syndrome (ARS), often with other systemic features. The developmental mechanisms and pathways regulated by FOXC1 remain largely unknown. There are two conserved orthologs of FOXC1 in zebrafish, foxc1a and foxc1b. To further examine the role of FOXC1 in vertebrates, we generated foxc1a and foxc1b single knockout zebrafish lines and bred them to obtain various allelic combinations. Three genotypes demonstrated visible phenotypes: foxc1a-/- single homozygous and foxc1-/- double knockout homozygous embryos presented with similar characteristics comprised of severe global vascular defects and early lethality, as well as microphthalmia, periocular edema and absence of the anterior chamber of the eye; additionally, fish with heterozygous loss of foxc1a combined with homozygosity for foxc1b (foxc1a+/-;foxc1b-/-) demonstrated craniofacial defects, heart anomalies and scoliosis. All other single and combined genotypes appeared normal. Analysis of foxc1 expression detected a significant increase in foxc1a levels in homozygous and heterozygous mutant eyes, suggesting a mechanism for foxc1a upregulation when its function is compromised; interestingly, the expression of another ARS-associated gene, pitx2, was responsive to the estimated level of wild-type Foxc1a, indicating a possible role for this protein in the regulation of pitx2 expression. Altogether, our results support a conserved role for foxc1 in the formation of many organs, consistent with the features observed in human patients, and highlight the importance of correct FOXC1/foxc1 dosage for vertebrate development.


Asunto(s)
Segmento Anterior del Ojo/anomalías , Anomalías del Ojo/genética , Enfermedades Hereditarias del Ojo/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción/genética , Proteínas de Pez Cebra/genética , Alelos , Animales , Segmento Anterior del Ojo/patología , Desarrollo Embrionario/genética , Anomalías del Ojo/patología , Enfermedades Hereditarias del Ojo/patología , Dosificación de Gen/genética , Regulación del Desarrollo de la Expresión Génica/genética , Genotipo , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Heterocigoto , Homocigoto , Humanos , Mutación/genética , Escoliosis/genética , Escoliosis/patología , Pez Cebra/genética
7.
Arterioscler Thromb Vasc Biol ; 41(12): 2923-2942, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34645278

RESUMEN

OBJECTIVE: Aortic valve disease is a common worldwide health burden with limited treatment options. Studies have shown that the valve endothelium is critical for structure-function relationships, and disease is associated with its dysfunction, damage, or injury. Therefore, therapeutic targets to maintain a healthy endothelium or repair damaged endothelial cells could hold promise. In this current study, we utilize a surgical mouse model of heart valve endothelial cell injury to study the short-term response at molecular and cellular levels. The goal is to determine if the native heart valve exhibits a reparative response to injury and identify the mechanisms underlying this process. Approach and Results: Mild aortic valve endothelial injury and abrogated function was evoked by inserting a guidewire down the carotid artery of young (3 months) and aging (16-18 months) wild-type mice. Short-term cellular responses were examined at 6 hours, 48 hours, and 4 weeks following injury, whereas molecular profiles were determined after 48 hours by RNA-sequencing. Within 48 hours following endothelial injury, young wild-type mice restore endothelial barrier function in association with increased cell proliferation, and upregulation of transforming growth factor beta 1 (Tgfß1) and the glycoprotein, collagen triple helix repeat containing 1 (Cthrc1). Interestingly, this beneficial response to injury was not observed in aging mice with known underlying endothelial dysfunction. CONCLUSIONS: Data from this study suggests that the healthy valve has the capacity to respond to mild endothelial injury, which in short term has beneficial effects on restoring endothelial barrier function through acute activation of the Tgfß1-Cthrc1 signaling axis and cell proliferation.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Endotelio Vascular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Envejecimiento/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Femenino , Masculino , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN , Porcinos , Regulación hacia Arriba
8.
Arterioscler Thromb Vasc Biol ; 38(3): 636-644, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29348122

RESUMEN

OBJECTIVE: Hematopoietic-derived cells have been reported in heart valves but remain poorly characterized. Interestingly, recent studies reveal infiltration of leukocytes and increased macrophages in human myxomatous mitral valves. Nevertheless, timing and contribution of macrophages in normal valves and myxomatous valve disease are still unknown. The objective is to characterize leukocytes during postnatal heart valve maturation and identify macrophage subsets in myxomatous valve disease. APPROACH AND RESULTS: Leukocytes are detected in heart valves after birth, and their numbers increase during postnatal valve development. Flow cytometry and immunostaining analysis indicate that almost all valve leukocytes are myeloid cells, consisting of at least 2 differentially localized macrophage subsets and dendritic cells. Beginning a week after birth, increased numbers of CCR2+ (C-C chemokine receptor type 2) macrophages are present, consistent with infiltrating populations of monocytes, and macrophages are localized in regions of biomechanical stress in the valve leaflets. Valve leukocytes maintain expression of CD (cluster of differentiation) 45 and do not contribute to significant numbers of endothelial or interstitial cells. Macrophage lineages were examined in aortic and mitral valves of Axin2 KO (knockout) mice that exhibit myxomatous features. Infiltrating CCR2+ monocytes and expansion of CD206-expressing macrophages are localized in regions where modified heavy chain hyaluronan is observed in myxomatous valve leaflets. Similar colocalization of modified hyaluronan and increased numbers of macrophages were observed in human myxomatous valve disease. CONCLUSIONS: Our study demonstrates the heterogeneity of myeloid cells in heart valves and highlights an alteration of macrophage subpopulations, notably an increased presence of infiltrating CCR2+ monocytes and CD206+ macrophages, in myxomatous valve disease.


Asunto(s)
Linaje de la Célula , Matriz Extracelular/patología , Enfermedades de las Válvulas Cardíacas/patología , Válvulas Cardíacas/patología , Macrófagos/patología , Factores de Edad , Anciano , Animales , Proteína Axina/genética , Proteína Axina/metabolismo , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/patología , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Enfermedades de las Válvulas Cardíacas/genética , Enfermedades de las Válvulas Cardíacas/metabolismo , Válvulas Cardíacas/metabolismo , Humanos , Ácido Hialurónico/metabolismo , Lectinas Tipo C/metabolismo , Leucocitos/metabolismo , Leucocitos/patología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Macrófagos/metabolismo , Masculino , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Mutación , Fenotipo , Receptores CCR2/metabolismo , Receptores de Superficie Celular/metabolismo
9.
Curr Cardiol Rep ; 20(4): 21, 2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29520694

RESUMEN

PURPOSE OF REVIEW: This review aims to highlight the past and more current literature related to the multifaceted pathogenic programs that contribute to calcific aortic valve disease (CAVD) with a focus on the contribution of developmental programs. RECENT FINDINGS: Calcification of the aortic valve is an active process characterized by calcific nodule formation on the aortic surface leading to a less supple and more stiffened cusp, thereby limiting movement and causing clinical stenosis. The mechanisms underlying these pathogenic changes are largely unknown, but emerging studies have suggested that signaling pathways common to valvulogenesis and bone development play significant roles and include Transforming Growth Factor-ß (TGF-ß), bone morphogenetic protein (BMP), Wnt, Notch, and Sox9. This comprehensive review of the literature highlights the complex nature of CAVD but concurrently identifies key regulators that can be targeted in the development of mechanistic-based therapies beyond surgical intervention to improve patient outcome.


Asunto(s)
Estenosis de la Válvula Aórtica/fisiopatología , Válvula Aórtica/patología , Transducción de Señal , Calcificación Vascular/fisiopatología , Válvula Aórtica/fisiopatología , Estenosis de la Válvula Aórtica/etiología , Biología Evolutiva , Matriz Extracelular/metabolismo , Humanos , Calcificación Vascular/etiología
10.
Curr Opin Cardiol ; 32(3): 239-245, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28157139

RESUMEN

PURPOSE OF REVIEW: Aortic valve disease is relatively common and encompasses both congenital and acquired forms. Bicuspid aortic valve (BAV) is the most common type of cardiac malformation and predisposes to the development of calcific aortic valve disease (CAVD). Since the description of the link between NOTCH1, BAV and CAVD approximately a decade ago, there have been significant advances in the genetic and molecular understanding of these diseases. RECENT FINDINGS: Recent work has defined the congenital cardiac phenotypes linked to mutations in NOTCH1, and in addition, novel etiologic genes for BAV have been discovered using new genetic technologies in humans. Furthermore, several mouse models of BAV have been described defining the role of endothelial Notch1 in aortic valve morphogenesis, whereas others have implicated new genes. These murine models along with other cell-based studies have led to molecular insights in the pathogenesis of CAVD. SUMMARY: These findings provide important insights into the molecular and genetic basis of aortic valve malformations, including BAV, specifically highlighting the etiologic role of endothelial cells. In addition, numerous investigations in to the mechanisms of CAVD demonstrate the importance of developmental origins and signaling pathways as well as communication between valve endothelial cells and the underlying interstitial cells in valve disease onset and progression.

11.
Arterioscler Thromb Vasc Biol ; 36(2): 328-38, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26634652

RESUMEN

OBJECTIVE: Aortic valve disease, including calcification, affects >2% of the human population and is caused by complex interactions between multiple risk factors, including genetic mutations, the environment, and biomechanics. At present, there are no effective treatments other than surgery, and this is because of the limited understanding of the mechanisms that underlie the condition. Previous work has shown that valve interstitial cells within the aortic valve cusps differentiate toward an osteoblast-like cell and deposit bone-like matrix that leads to leaflet stiffening and calcific aortic valve stenosis. However, the mechanisms that promote pathological phenotypes in valve interstitial cells are unknown. APPROACH AND RESULTS: Using a combination of in vitro and in vivo tools with mouse, porcine, and human tissue, we show that in valve interstitial cells, reduced Sox9 expression and nuclear localization precedes the onset of calcification. In vitro, Sox9 nuclear export and calcific nodule formation is prevented by valve endothelial cells. However, in vivo, loss of Tgfß1 in the endothelium leads to reduced Sox9 expression and calcific aortic valve disease. CONCLUSIONS: Together, these findings suggest that reduced nuclear localization of Sox9 in valve interstitial cells is an early indicator of calcification, and therefore, pharmacological targeting to prevent nuclear export could serve as a novel therapeutic tool in the prevention of calcification and stenosis.


Asunto(s)
Estenosis de la Válvula Aórtica/metabolismo , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Calcinosis/metabolismo , Células Endoteliales/metabolismo , Comunicación Paracrina , Factor de Transcripción SOX9/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Transporte Activo de Núcleo Celular , Animales , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/prevención & control , Calcinosis/genética , Calcinosis/patología , Calcinosis/prevención & control , Células Cultivadas , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Células Endoteliales/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción SOX9/genética , Porcinos , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Transfección , Factor de Crecimiento Transformador beta1/genética , Quinasas Asociadas a rho/metabolismo
12.
J Mol Cell Cardiol ; 100: 72-82, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27756541

RESUMEN

Risk factors of heart valve disease are well defined and prolonged exposure throughout life leads to degeneration and dysfunction in up to 33% of the population. While aortic valve replacement remains the most common need for cardiovascular surgery particularly in those aged over 65, the underlying mechanisms of progressive deterioration are unknown. In other cardiovascular systems, a decline in endothelial cell integrity and function play a major role in promoting pathological changes, and while similar mechanisms have been speculated in the valves, studies to support this are lacking. The goal of this study was to examine age-related changes in valve endothelial cell (VEC) distribution, morphology, function and transcriptomes during critical stages of valve development (embryonic), growth (postnatal (PN)), maintenance (young adult) and aging (aging adult). Using a combination of in vivo mouse, and in vitro porcine assays we show that VEC function including, nitric oxide bioavailability, metabolism, endothelial-to-mesenchymal potential, membrane self-repair and proliferation decline with age. In addition, density of VEC distribution along the endothelium decreases and this is associated with changes in morphology, decreased cell-cell interactions, and increased permeability. These changes are supported by RNA-seq analysis showing that focal adhesion-, cell cycle-, and oxidative phosphorylation-associated biological processes are negatively impacted by aging. Furthermore, by performing high-throughput analysis we are able to report the differential and common transcriptomes of VECs at each time point that can provide insights into the mechanisms underlying age-related dysfunction. These studies suggest that maturation of heart valves over time is a multifactorial process and this study has identified several key parameters that may contribute to impairment of the valve to maintain critical structure-function relationships; leading to degeneration and disease.


Asunto(s)
Células Endoteliales/metabolismo , Válvulas Cardíacas/metabolismo , Válvulas Cardíacas/patología , Envejecimiento , Animales , Comunicación Celular , Recuento de Células , Proliferación Celular , Células Cultivadas , Senescencia Celular/genética , Análisis por Conglomerados , Células Endoteliales/ultraestructura , Perfilación de la Expresión Génica , Válvulas Cardíacas/ultraestructura , Humanos , Ratones , Ratones Transgénicos , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transcriptoma
13.
J Heart Valve Dis ; 24(5): 531-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26897831

RESUMEN

BACKGROUND AND AIM OF THE STUDY: Minimally invasive aortic valve replacement via ministernotomy (ministernotomy-AVR) or minithoracotomy (minithoracotomy-AVR) is gaining popularity. To date, a direct comparison of ministernotomy-AVR versus minithoracotomy-AVR is lacking. The study aim was to compare these two procedures from a cost-benefit perspective. METHODS: Eight reports from the United States were selected from amongst 33,494 literature citations based on sample size and data completeness. Perioperative variables were collected for each surgical approach. Fixed and variable costs were estimated as cost per case in excess of full sternotomy AVR procedures. RESULTS: Ministernotomy-AVR patients were of a significantly lower mean age (59.8 years versus 67.9 years), ejection fraction (50.4-51.6% versus 56.1-57.8%), shorter cardiopulmonary bypass time (97.2 min versus 125.6 min) and cross-clamp time (69.9 min versus 87.9 min), a lower rate of blood transfusion (25.9% versus 64.4%), and a shorter length of hospital stay (5.7 versus 6.2 days). There were no significant inter-group differences in 30-day mortality, conversion to sternotomy, neurologic events, arrhythmia, wound infection, or postoperative bleeding. Assuming a volume of 50 cases per year, the added operative cost per case for a minithoracotomy-AVR was US$ 4,254 compared to US$ 290 for a ministernotomy-AVR. The added costs per case, assuming 200 cases per year, were US$ 4,209 and US$ 290, respectively. A minithoracotomy-AVR program performing 50 cases per year adds US$ 1,063,665 of operative costs over five years, compared to US$ 72,500 for a ministernotomy-AVR program. CONCLUSION: The present analysis suggested that the clinical benefits of ministernotomy-AVR are comparable or better than those of minithoracotomy-AVR, and at lower costs. Healthcare delivery organizations should consider the results of cost-benefit examinations when developing surgical valve replacement programs.


Asunto(s)
Válvula Aórtica/cirugía , Atención a la Salud/economía , Implantación de Prótesis de Válvulas Cardíacas/economía , Costos de Hospital , Evaluación de Procesos, Atención de Salud/economía , Esternotomía/economía , Toracotomía/economía , Adulto , Anciano , Anciano de 80 o más Años , Válvula Aórtica/fisiopatología , Pérdida de Sangre Quirúrgica/prevención & control , Transfusión Sanguínea/economía , Ahorro de Costo , Análisis Costo-Beneficio , Femenino , Implantación de Prótesis de Válvulas Cardíacas/efectos adversos , Implantación de Prótesis de Válvulas Cardíacas/métodos , Implantación de Prótesis de Válvulas Cardíacas/mortalidad , Humanos , Tiempo de Internación/economía , Masculino , Persona de Mediana Edad , Modelos Económicos , Tempo Operativo , Esternotomía/efectos adversos , Esternotomía/métodos , Esternotomía/mortalidad , Toracotomía/efectos adversos , Toracotomía/métodos , Toracotomía/mortalidad , Factores de Tiempo , Resultado del Tratamiento , Estados Unidos
14.
Hum Mol Genet ; 21(10): 2288-97, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22357654

RESUMEN

Aging is an intricate process that increases susceptibility to sarcopenia and cardiovascular diseases. The accumulation of mitochondrial DNA (mtDNA) mutations is believed to contribute to mitochondrial dysfunction, potentially shortening lifespan. The mtDNA mutator mouse, a mouse model with a proofreading-deficient mtDNA polymerase γ, was shown to develop a premature aging phenotype, including sarcopenia, cardiomyopathy and decreased lifespan. This phenotype was associated with an accumulation of mtDNA mutations and mitochondrial dysfunction. We found that increased expression of peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), a crucial regulator of mitochondrial biogenesis and function, in the muscle of mutator mice increased mitochondrial biogenesis and function and also improved the skeletal muscle and heart phenotypes of the mice. Deep sequencing analysis of their mtDNA showed that the increased mitochondrial biogenesis did not reduce the accumulation of mtDNA mutations but rather caused a small increase. These results indicate that increased muscle PGC-1α expression is able to improve some premature aging phenotypes in the mutator mice without reverting the accumulation of mtDNA mutations.


Asunto(s)
Envejecimiento/metabolismo , ADN Mitocondrial/metabolismo , Mitocondrias/metabolismo , Mutación , Fenotipo , Animales , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción
15.
Arterioscler Thromb Vasc Biol ; 33(2): 285-93, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23202364

RESUMEN

OBJECTIVE: Calcific aortic valve disease (CAVD) is a major public health problem with no effective treatment available other than surgery. We previously showed that mature heart valves calcify in response to retinoic acid (RA) treatment through downregulation of the SRY transcription factor Sox9. In this study, we investigated the effects of excess vitamin A and its metabolite RA on heart valve structure and function in vivo and examined the molecular mechanisms of RA signaling during the calcification process in vitro. METHODS AND RESULTS: Using a combination of approaches, we defined calcific aortic valve disease pathogenesis in mice fed 200 IU/g and 20 IU/g of retinyl palmitate for 12 months at molecular, cellular, and functional levels. We show that mice fed excess vitamin A develop aortic valve stenosis and leaflet calcification associated with increased expression of osteogenic genes and decreased expression of cartilaginous markers. Using a pharmacological approach, we show that RA-mediated Sox9 repression and calcification is regulated by classical RA signaling and requires both RA and retinoid X receptors. CONCLUSIONS: Our studies demonstrate that excess vitamin A dietary intake promotes heart valve calcification in vivo. Therefore suggesting that hypervitaminosis A could serve as a new risk factor of calcific aortic valve disease in the human population.


Asunto(s)
Válvula Aórtica/metabolismo , Calcinosis/etiología , Suplementos Dietéticos , Enfermedades de las Válvulas Cardíacas/etiología , Hipervitaminosis A/complicaciones , Vitamina A/análogos & derivados , Vitaminas , Animales , Válvula Aórtica/patología , Calcinosis/genética , Calcinosis/metabolismo , Calcinosis/patología , Línea Celular , Embrión de Pollo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Modelos Animales de Enfermedad , Diterpenos , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Enfermedades de las Válvulas Cardíacas/genética , Enfermedades de las Válvulas Cardíacas/metabolismo , Enfermedades de las Válvulas Cardíacas/patología , Hipervitaminosis A/inducido químicamente , Hipervitaminosis A/genética , Hipervitaminosis A/metabolismo , Hipervitaminosis A/patología , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteogénesis/genética , Osteopontina/genética , Osteopontina/metabolismo , Interferencia de ARN , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Ésteres de Retinilo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Transducción de Señal , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Transfección , Tretinoina/metabolismo , Vitamina A/metabolismo , Vitaminas/metabolismo
16.
Circ J ; 78(8): 1801-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24998280

RESUMEN

Valvular heart disease occurs as either a congenital or acquired condition and advances in medical care have resulted in valve disease becoming increasingly prevalent. Unfortunately, treatments remain inadequate because of our limited understanding of the genetic and molecular etiology of diseases affecting the heart valves. Therefore, surgical repair or replacement remains the most effective option, which comes with additional complications and no guarantee of life-long success. Over the past decade, there have been significant advances in our understanding of cardiac valve development and, not surprisingly, mutations in these developmental genes have been identified in humans with congenital valve malformations. Concurrently, there has been a greater realization that acquired valve disease is not simply a degenerative process. Molecular investigation of acquired valve disease has identified that numerous signaling pathways critical for normal valve development are re-expressed in diseased valves. This review will discuss recent advances in our understanding of the development of the heart valves, as well as the implications of these findings on the genetics of congenital and acquired valvular heart disease.


Asunto(s)
Enfermedades de las Válvulas Cardíacas , Válvulas Cardíacas , Transducción de Señal/genética , Enfermedades de las Válvulas Cardíacas/embriología , Enfermedades de las Válvulas Cardíacas/etiología , Enfermedades de las Válvulas Cardíacas/genética , Enfermedades de las Válvulas Cardíacas/metabolismo , Válvulas Cardíacas/embriología , Válvulas Cardíacas/metabolismo , Humanos
17.
Curr Cardiol Rep ; 16(6): 487, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24743897

RESUMEN

Valvular heart disease is associated with significant morbidity and mortality and often the result of congenital malformations. However, the prevalence is increasing in adults not only because of the growing aging population, but also because of improvements in the medical and surgical care of children with congenital heart valve defects. The success of the Human Genome Project and major advances in genetic technologies, in combination with our increased understanding of heart valve development, has led to the discovery of numerous genetic contributors to heart valve disease. These have been uncovered using a variety of approaches including the examination of familial valve disease and genome-wide association studies to investigate sporadic cases. This review will discuss these findings and their implications in the treatment of valvular heart disease.


Asunto(s)
Estudio de Asociación del Genoma Completo , Cardiopatías Congénitas/genética , Enfermedades de las Válvulas Cardíacas/epidemiología , Enfermedades de las Válvulas Cardíacas/genética , Adulto , Anticoagulantes/uso terapéutico , Válvula Aórtica/anomalías , Enfermedad de la Válvula Aórtica Bicúspide , Niño , Anomalía de Ebstein/epidemiología , Anomalía de Ebstein/genética , Cardiopatías Congénitas/epidemiología , Cardiopatías Congénitas/terapia , Enfermedades de las Válvulas Cardíacas/terapia , Corazón Auxiliar/estadística & datos numéricos , Corazón Auxiliar/tendencias , Humanos , Esperanza de Vida , Prolapso de la Válvula Mitral/epidemiología , Prolapso de la Válvula Mitral/genética , Prevalencia , Estenosis de la Válvula Pulmonar/epidemiología , Estenosis de la Válvula Pulmonar/genética
18.
Dev Dyn ; 242(6): 699-708, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23553854

RESUMEN

BACKGROUND: Formation of the epicardium requires several cellular processes including migration, transformation, invasion, and differentiation in order to give rise to fibroblast, smooth muscle, coronary endothelial and myocyte cell lineages within the developing myocardium. Snai1 is a zinc finger transcription factor that plays an important role in regulating cell survival and fate during embryonic development and under pathological conditions. However, its role in avian epicardial development has not been examined. RESULTS: Here we show that Snai1 is highly expressed in epicardial cells from as early as the proepicardial cell stage and its expression is maintained as proepicardial cells migrate and spread over the surface of the myocardium and undergo epicardial-to-mesenchymal transformation in the generation of epicardial-derived cells. Using multiple in vitro assays, we show that Snai1 overexpression in chick explants enhances proepicardial cell migration at Hamburger Hamilton Stage (HH St.) 16, and epicardial-to-mesenchymal transformation, cell migration, and invasion at HH St. 24. Further, we demonstrate that Snai1-mediated cell migration requires matrix metalloproteinase activity, and MMP15 is sufficient for this process. CONCLUSIONS: Together our data provide new insights into the multiple roles that Snai1 has in regulating avian epicardial development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Pericardio/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Embrión de Pollo , Corazón/embriología , Metaloproteinasa 15 de la Matriz/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Ratones , Miocardio/metabolismo , Pericardio/embriología , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción de la Familia Snail
19.
Ann Biomed Eng ; 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38874705

RESUMEN

Aortic valve (AV) disease is a common valvular lesion in the United States, present in about 5% of the population at age 65 with increasing prevalence with advancing age. While current replacement heart valves have extended life for many, their long-term use remains hampered by limited durability. Non-surgical treatments for AV disease do not yet exist, in large part because our understanding of AV disease etiology remains incomplete. The direct study of human AV disease remains hampered by the fact that clinical data is only available at the time of treatment, where the disease is at or near end stage and any time progression information has been lost. Large animal models, long used to assess replacement AV devices, cannot yet reproduce AV disease processes. As an important alternative mouse animal models are attractive for their ability to perform genetic studies of the AV disease processes and test potential pharmaceutical treatments. While mouse models have been used for cellular and genetic studies of AV disease, their small size and fast heart rates have hindered their use for tissue- and organ-level studies. We have recently developed a novel ex vivo micro-CT-based methodology to 3D reconstruct murine heart valves and estimate the leaflet mechanical behaviors (Feng et al. in Sci Rep 13(1):12852, 2023). In the present study, we extended our approach to 3D reconstruction of the in vivo functional murine AV (mAV) geometry using high-frequency four-dimensional ultrasound (4DUS). From the resulting 4DUS images we digitized the mAV mid-surface coordinates in the fully closed and fully opened states. We then utilized matched high-resolution µCT images of ex vivo mouse mAV to develop mAV NURBS-based geometric model. We then fitted the mAV geometric model to the in vivo data to reconstruct the 3D in vivo mAV geometry in the closed and open states in n = 3 mAV. Results demonstrated high fidelity geometric results. To our knowledge, this is the first time such reconstruction was ever achieved. This robust assessment of in vivo mAV leaflet kinematics in 3D opens up the possibility for longitudinal characterization of murine models that develop aortic valve disease.

20.
J Mol Cell Cardiol ; 65: 137-46, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24157418

RESUMEN

Mature heart valves are complex structures consisting of three highly organized extracellular matrix layers primarily composed of collagens, proteoglycans and elastin. Collectively, these diverse matrix components provide all the necessary biomechanical properties for valve function throughout life. In contrast to healthy valves, myxomatous valve disease is the most common cause of mitral valve prolapse in the human population and is characterized by an abnormal abundance of proteoglycans within the valve tri-laminar structure. Despite the clinical significance, the etiology of this phenotype is not known. Scleraxis (Scx) is a basic-helix-loop-helix transcription factor that we previously showed to be required for establishing heart valve structure during remodeling stages of valvulogenesis. In this study, we report that remodeling heart valves from Scx null mice express decreased levels of proteoglycans, particularly chondroitin sulfate proteoglycans (CSPGs), while overexpression in embryonic avian valve precursor cells and adult porcine valve interstitial cells increases CSPGs. Using these systems we further identify that Scx is positively regulated by canonical Tgfß2 signaling during this process and this is attenuated by MAPK activity. Finally, we show that Scx is increased in myxomatous valves from human patients and mouse models, and overexpression in human mitral valve interstitial cells modestly increases proteoglycan expression consistent with myxomatous mitral valve phenotypes. Together, these studies identify an important role for Scx in regulating proteoglycans in embryonic and mature valve cells and suggest that imbalanced regulation could influence myxomatous pathogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Válvulas Cardíacas/metabolismo , Sistema de Señalización de MAP Quinasas , Proteoglicanos/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Animales Recién Nacidos , Pollos , Modelos Animales de Enfermedad , Válvulas Cardíacas/embriología , Válvulas Cardíacas/patología , Humanos , Ratones , Válvula Mitral/embriología , Válvula Mitral/metabolismo , Válvula Mitral/patología , Células 3T3 NIH , Sus scrofa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA