Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Anat Rec (Hoboken) ; 302(1): 117-124, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30288957

RESUMEN

Mitral valve prolapse (MVP) affects 2.4% of the population and has poorly understood etiology. Recent genetic studies have begun to unravel the complexities of MVP and through these efforts, mutations in the FLNA (Filamin-A) gene were identified as disease causing. Our in vivo and in vitro studies have validated these genetic findings and have revealed FLNA as a central regulator of valve morphogenesis. The mechanisms by which FLNA mutations result in myxomatous mitral valve disease are currently unknown, but may involve proteins previously associated with mutated regions of the FLNA protein, such as the small GTPase signaling protein, R-Ras. Herein, we report that Filamin-A is required for R-Ras expression and activation of the Ras-Mek-Erk pathway. Loss of the Ras/Erk pathway correlated with hyperactivation of pSmad2/3, increased extracellular matrix (ECM) production and enlarged mitral valves. Analyses of integrin receptors in the mitral valve revealed that Filamin-A was required for ß1-integrin expression and provided a potential mechanism for impaired ECM compaction and valve enlargement. Our data support Filamin-A as a protein that regulates the balance between Erk and Smad activation and an inability of Filamin-A deficient valve interstitial cells to effectively remodel the increased ECM production through a ß1-integrin mechanism. As a consequence, loss of Filamin-A function results in increased ECM production and generation of a myxomatous phenotype characterized by improperly compacted mitral valve tissue. Anat Rec, 302:117-124, 2019. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Matriz Extracelular/metabolismo , Filaminas/fisiología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Válvula Mitral/metabolismo , Organogénesis , Proteína smad3/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Noqueados , Válvula Mitral/citología , Fenotipo
2.
Cell Rep ; 22(7): 1913-1922, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29444441

RESUMEN

Direct reprogramming of fibroblasts into cardiomyocyte-like cells (iCM) holds great potential for heart regeneration and disease modeling and may lead to future therapeutic applications. Currently, application of this technology is limited by our lack of understanding of the molecular mechanisms that drive direct iCM reprogramming. Using a quantitative mass spectrometry-based proteomic approach, we identified the temporal global changes in protein abundance that occur during initial phases of iCM reprogramming. Collectively, our results show systematic and temporally distinct alterations in levels of specific functional classes of proteins during the initiating steps of reprogramming including extracellular matrix proteins, translation factors, and chromatin-binding proteins. We have constructed protein relational networks associated with the initial transition of a fibroblast into an iCM. These findings demonstrate the presence of an orchestrated series of temporal steps associated with dynamic changes in protein abundance in a defined group of protein pathways during the initiating events of direct reprogramming.


Asunto(s)
Reprogramación Celular , Miocitos Cardíacos/citología , Animales , Cromatina/metabolismo , Regulación hacia Abajo , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Unión Proteica , Proteómica , Factores de Tiempo , Factores de Transcripción/metabolismo , Regulación hacia Arriba
3.
Development ; 143(5): 774-9, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26811383

RESUMEN

Congenital heart defects are the most common birth defects in humans, and those that affect the proper alignment of the outflow tracts and septation of the ventricles are a highly significant cause of morbidity and mortality in infants. A late differentiating population of cardiac progenitors, referred to as the anterior second heart field (AHF), gives rise to the outflow tract and the majority of the right ventricle and provides an embryological context for understanding cardiac outflow tract alignment and membranous ventricular septal defects. However, the transcriptional pathways controlling AHF development and their roles in congenital heart defects remain incompletely elucidated. Here, we inactivated the gene encoding the transcription factor MEF2C in the AHF in mice. Loss of Mef2c function in the AHF results in a spectrum of outflow tract alignment defects ranging from overriding aorta to double-outlet right ventricle and dextro-transposition of the great arteries. We identify Tdgf1, which encodes a Nodal co-receptor (also known as Cripto), as a direct transcriptional target of MEF2C in the outflow tract via an AHF-restricted Tdgf1 enhancer. Importantly, both the MEF2C and TDGF1 genes are associated with congenital heart defects in humans. Thus, these studies establish a direct transcriptional pathway between the core cardiac transcription factor MEF2C and the human congenital heart disease gene TDGF1. Moreover, we found a range of outflow tract alignment defects resulting from a single genetic lesion, supporting the idea that AHF-derived outflow tract alignment defects may constitute an embryological spectrum rather than distinct anomalies.


Asunto(s)
Factor de Crecimiento Epidérmico/fisiología , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas de Membrana/fisiología , Proteínas de Neoplasias/fisiología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/genética , Femenino , Eliminación de Gen , Corazón/embriología , Cardiopatías Congénitas/genética , Defectos del Tabique Interventricular/genética , Ventrículos Cardíacos , Humanos , Hibridación in Situ , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/fisiología , Masculino , Glicoproteínas de Membrana/genética , Ratones , Morfogénesis/genética , Proteínas de Neoplasias/genética , Organogénesis , Análisis de Secuencia de ARN , Distribución Tisular , Transcripción Genética , Transposición de los Grandes Vasos/genética
4.
Curr Biol ; 26(1): 27-37, 2016 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-26725196

RESUMEN

During valvulogenesis, globular endocardial cushions elongate and remodel into highly organized thin fibrous leaflets. Proper regulation of this dynamic process is essential to maintain unidirectional blood flow as the embryonic heart matures. In this study, we tested how mechanosensitive small GTPases, RhoA and Rac1, coordinate atrioventricular valve (AV) differentiation and morphogenesis. RhoA activity and its regulated GTPase-activating protein FilGAP are elevated during early cushion formation but decreased considerably during valve remodeling. In contrast, Rac1 activity was nearly absent in the early cushions but increased substantially as the valve matured. Using gain- and loss-of-function assays, we determined that the RhoA pathway was essential for the contractile myofibroblastic phenotype present in early cushion formation but was surprisingly insufficient to drive matrix compaction during valve maturation. The Rac1 pathway was necessary to induce matrix compaction in vitro through increased cell adhesion, elongation, and stress fiber alignment. Facilitating this process, we found that acute cyclic stretch was a potent activator of RhoA and subsequently downregulated Rac1 activity via FilGAP. On the other hand, chronic cyclic stretch reduced active RhoA and downstream FilGAP, which enabled Rac1 activation. Finally, we used partial atrial ligation experiments to confirm in vivo that altered cyclic mechanical loading augmented or restricted cushion elongation and thinning, directly through potentiation of active Rac1 and active RhoA, respectively. Together, these results demonstrate that cyclic mechanical signaling coordinates the RhoA to Rac1 signaling transition essential for proper embryonic mitral valve remodeling.


Asunto(s)
Válvula Mitral/embriología , Válvula Mitral/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Diferenciación Celular/fisiología , Embrión de Pollo , Matriz Extracelular/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Mecanotransducción Celular/fisiología , Morfogénesis , Transducción de Señal , Fibras de Estrés/metabolismo , Estrés Mecánico
5.
J Cardiovasc Dev Dis ; 2(3): 214-232, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26527432

RESUMEN

The heart valve interstitial cell (VIC) population is dynamic and thought to mediate lay down and maintenance of the tri-laminar extracellular matrix (ECM) structure within the developing and mature valve throughout life. Disturbances in the contribution and distribution of valve ECM components are detrimental to biomechanical function and associated with disease. This pathological process is associated with activation of resident VICs that in the absence of disease reside as quiescent cells. While these paradigms have been long standing, characterization of this abundant and ever-changing valve cell population is incomplete. Here we examine the expression pattern of Smooth muscle α-actin, Periostin, Twist1 and Vimentin in cultured VICs, heart valves from healthy embryonic, postnatal and adult mice, as well as mature valves from human patients and established mouse models of disease. We show that the VIC population is highly heterogeneous and phenotypes are dependent on age, species, location, and disease state. Furthermore, we identify phenotypic diversity across common models of mitral valve disease. These studies significantly contribute to characterizing the VIC population in health and disease and provide insights into the cellular dynamics that maintain valve structure in healthy adults and mediate pathologic remodeling in disease states.

6.
J Cardiovasc Dev Dis ; 2(3): 200-213, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26473162

RESUMEN

Mutations in the actin-binding gene Filamin-A have been linked to non-syndromic myxomatous valvular dystrophy and associated mitral valve prolapse. Previous studies by our group traced the adult valve defects back to developmental errors in valve interstitial cell-mediated extracellular matrix remodeling during fetal valve gestation. Mice deficient in Filamin-A exhibit enlarged mitral leaflets at E17.5, and subsequent progression to a myxomatous phenotype is observed by two months. For this study, we sought to define mechanisms that contribute to myxomatous degeneration in the adult Filamin-A-deficient mouse. In vivo experiments demonstrate increased infiltration of hematopoietic-derived cells and macrophages in adolescent Filamin-A conditional knockout mice. Concurrent with this infiltration of hematopoietic cells, we show an increase in Erk activity, which localizes to regions of MMP2 expression. Additionally, increases in cell proliferation are observed at two months, when hematopoietic cell engraftment and signaling are pronounced. Similar changes are observed in human myxomatous mitral valve tissue, suggesting that infiltration of hematopoietic-derived cells and/or increased Erk signaling may contribute to myxomatous valvular dystrophy. Consequently, immune cell targeting and/or suppression of pErk activities may represent an effective therapeutic option for mitral valve prolapse patients.

7.
Nature ; 525(7567): 109-13, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26258302

RESUMEN

Mitral valve prolapse (MVP) is a common cardiac valve disease that affects nearly 1 in 40 individuals. It can manifest as mitral regurgitation and is the leading indication for mitral valve surgery. Despite a clear heritable component, the genetic aetiology leading to non-syndromic MVP has remained elusive. Four affected individuals from a large multigenerational family segregating non-syndromic MVP underwent capture sequencing of the linked interval on chromosome 11. We report a missense mutation in the DCHS1 gene, the human homologue of the Drosophila cell polarity gene dachsous (ds), that segregates with MVP in the family. Morpholino knockdown of the zebrafish homologue dachsous1b resulted in a cardiac atrioventricular canal defect that could be rescued by wild-type human DCHS1, but not by DCHS1 messenger RNA with the familial mutation. Further genetic studies identified two additional families in which a second deleterious DCHS1 mutation segregates with MVP. Both DCHS1 mutations reduce protein stability as demonstrated in zebrafish, cultured cells and, notably, in mitral valve interstitial cells (MVICs) obtained during mitral valve repair surgery of a proband. Dchs1(+/-) mice had prolapse of thickened mitral leaflets, which could be traced back to developmental errors in valve morphogenesis. DCHS1 deficiency in MVP patient MVICs, as well as in Dchs1(+/-) mouse MVICs, result in altered migration and cellular patterning, supporting these processes as aetiological underpinnings for the disease. Understanding the role of DCHS1 in mitral valve development and MVP pathogenesis holds potential for therapeutic insights for this very common disease.


Asunto(s)
Cadherinas/genética , Cadherinas/metabolismo , Prolapso de la Válvula Mitral/genética , Prolapso de la Válvula Mitral/patología , Mutación/genética , Animales , Tipificación del Cuerpo/genética , Proteínas Relacionadas con las Cadherinas , Cadherinas/deficiencia , Movimiento Celular/genética , Cromosomas Humanos Par 11/genética , Femenino , Humanos , Masculino , Ratones , Válvula Mitral/anomalías , Válvula Mitral/embriología , Válvula Mitral/patología , Válvula Mitral/cirugía , Linaje , Fenotipo , Estabilidad Proteica , ARN Mensajero/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
8.
J Clin Invest ; 124(1): 448-60, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24355923

RESUMEN

Loeys-Dietz syndrome (LDS) is a connective tissue disorder that is characterized by a high risk for aneurysm and dissection throughout the arterial tree and phenotypically resembles Marfan syndrome. LDS is caused by heterozygous missense mutations in either TGF-ß receptor gene (TGFBR1 or TGFBR2), which are predicted to result in diminished TGF-ß signaling; however, aortic surgical samples from patients show evidence of paradoxically increased TGF-ß signaling. We generated 2 knockin mouse strains with LDS mutations in either Tgfbr1 or Tgfbr2 and a transgenic mouse overexpressing mutant Tgfbr2. Knockin and transgenic mice, but not haploinsufficient animals, recapitulated the LDS phenotype. While heterozygous mutant cells had diminished signaling in response to exogenous TGF-ß in vitro, they maintained normal levels of Smad2 phosphorylation under steady-state culture conditions, suggesting a chronic compensation. Analysis of TGF-ß signaling in the aortic wall in vivo revealed progressive upregulation of Smad2 phosphorylation and TGF-ß target gene output, which paralleled worsening of aneurysm pathology and coincided with upregulation of TGF-ß1 ligand expression. Importantly, suppression of Smad2 phosphorylation and TGF-ß1 expression correlated with the therapeutic efficacy of the angiotensin II type 1 receptor antagonist losartan. Together, these data suggest that increased TGF-ß signaling contributes to postnatal aneurysm progression in LDS.


Asunto(s)
Angiotensina II/fisiología , Aneurisma de la Aorta/metabolismo , Síndrome de Loeys-Dietz/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Animales , Aorta/patología , Aneurisma de la Aorta/prevención & control , Células Cultivadas , Progresión de la Enfermedad , Femenino , Haploinsuficiencia , Humanos , Síndrome de Loeys-Dietz/tratamiento farmacológico , Síndrome de Loeys-Dietz/patología , Losartán/uso terapéutico , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense , Miocitos del Músculo Liso/metabolismo , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Proteína Smad2/metabolismo
9.
Nat Genet ; 44(11): 1249-54, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23023332

RESUMEN

Elevated transforming growth factor (TGF)-ß signaling has been implicated in the pathogenesis of syndromic presentations of aortic aneurysm, including Marfan syndrome (MFS) and Loeys-Dietz syndrome (LDS). However, the location and character of many of the causal mutations in LDS intuitively imply diminished TGF-ß signaling. Taken together, these data have engendered controversy regarding the specific role of TGF-ß in disease pathogenesis. Shprintzen-Goldberg syndrome (SGS) has considerable phenotypic overlap with MFS and LDS, including aortic aneurysm. We identified causative variation in ten individuals with SGS in the proto-oncogene SKI, a known repressor of TGF-ß activity. Cultured dermal fibroblasts from affected individuals showed enhanced activation of TGF-ß signaling cascades and higher expression of TGF-ß-responsive genes relative to control cells. Morpholino-induced silencing of SKI paralogs in zebrafish recapitulated abnormalities seen in humans with SGS. These data support the conclusions that increased TGF-ß signaling is the mechanism underlying SGS and that high signaling contributes to multiple syndromic presentations of aortic aneurysm.


Asunto(s)
Aneurisma de la Aorta/genética , Aracnodactilia/genética , Craneosinostosis/genética , Proteínas de Unión al ADN , Síndrome de Marfan/genética , Proteínas Proto-Oncogénicas , Factor de Crecimiento Transformador beta , Animales , Aracnodactilia/metabolismo , Células Cultivadas , Craneosinostosis/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibroblastos , Humanos , Síndrome de Loeys-Dietz/genética , Síndrome de Marfan/metabolismo , Ratones , Mutación , Fenotipo , Fosforilación , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/genética , Pez Cebra
10.
Cardiovasc Res ; 96(1): 109-19, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22843703

RESUMEN

AIMS: We hypothesized that the structure and function of the mature valves is largely dependent upon how these tissues are built during development, and defects in how the valves are built can lead to the pathological progression of a disease phenotype. Thus, we sought to uncover potential developmental origins and mechanistic underpinnings causal to myxomatous mitral valve disease. We focus on how filamin-A, a cytoskeletal binding protein with strong links to human myxomatous valve disease, can function as a regulatory interface to control proper mitral valve development. METHODS AND RESULTS: Filamin-A-deficient mice exhibit abnormally enlarged mitral valves during foetal life, which progresses to a myxomatous phenotype by 2 months of age. Through expression studies, in silico modelling, 3D morphometry, biochemical studies, and 3D matrix assays, we demonstrate that the inception of the valve disease occurs during foetal life and can be attributed, in part, to a deficiency of interstitial cells to efficiently organize the extracellular matrix (ECM). This ECM organization during foetal valve gestation is due, in part, to molecular interactions between filamin-A, serotonin, and the cross-linking enzyme, transglutaminase-2 (TG2). Pharmacological and genetic perturbations that inhibit serotonin-TG2-filamin-A interactions lead to impaired ECM remodelling and engender progression to a myxomatous valve phenotype. CONCLUSIONS: These findings illustrate a molecular mechanism by which valve interstitial cells, through a serotonin, TG, and filamin-A pathway, regulate matrix organization during foetal valve development. Additionally, these data indicate that disrupting key regulatory interactions during valve development can set the stage for the generation of postnatal myxomatous valve disease.


Asunto(s)
Proteínas Contráctiles/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/embriología , Cardiopatías Congénitas/embriología , Proteínas de Microfilamentos/metabolismo , Prolapso de la Válvula Mitral/embriología , Válvula Mitral/embriología , Mixoma/embriología , Animales , Proteínas Contráctiles/genética , Filaminas , Proteínas de Unión al GTP/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Cardiopatías Congénitas/genética , Masculino , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Prolapso de la Válvula Mitral/genética , Mixoma/genética , Proteína Glutamina Gamma Glutamiltransferasa 2 , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Transglutaminasas/metabolismo , Triptófano Hidroxilasa/metabolismo
11.
Differentiation ; 84(1): 103-16, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22579502

RESUMEN

Atrioventricular valve development commences with an EMT event whereby endocardial cells transform into mesenchyme. The molecular events that induce this phenotypic change are well understood and include many growth factors, signaling components, and transcription factors. Besides their clear importance in valve development, the role of these transformed mesenchyme and the function they serve in the developing prevalve leaflets is less understood. Indeed, we know that these cells migrate, but how and why do they migrate? We also know that they undergo a transition to a mature, committed cell, largely defined as an interstitial fibroblast due to their ability to secrete various matrix components including collagen type I. However, we have yet to uncover mechanisms by which the matrix is synthesized, how it is secreted, and how it is organized. As valve disease is largely characterized by altered cell number, cell activation, and matrix disorganization, answering questions of how the valves are built will likely provide us with information of real clinical relevance. Although expression profiling and descriptive or correlative analyses are insightful, to advance the field, we must now move past the simplicity of these assays and ask fundamental, mechanistic based questions aimed at understanding how valves are "built". Herein we review current understandings of atrioventricular valve development and present what is known and what isn't known. In most cases, basic, biological questions and hypotheses that were presented decades ago on valve development still are yet to be answered but likely hold keys to uncovering new discoveries with relevance to both embryonic development and the developmental basis of adult heart valve diseases. Thus, the goal of this review is to remind us of these questions and provide new perspectives on an old theme of valve development.


Asunto(s)
Válvulas Cardíacas/embriología , Animales , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Embrión de Pollo , Colágeno Tipo I/metabolismo , Cojinetes Endocárdicos/citología , Endocardio/citología , Células Endoteliales/citología , Matriz Extracelular/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Cardiopatías Congénitas/embriología , Enfermedades de las Válvulas Cardíacas/embriología , Enfermedades de las Válvulas Cardíacas/etiología , Humanos , Mesodermo/citología , Ratones , Válvula Mitral/embriología , Válvula Mitral/patología , Válvula Tricúspide/embriología , Válvula Tricúspide/patología
12.
Dev Biol ; 366(2): 111-24, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22546693

RESUMEN

The importance of the epicardium for myocardial and valvuloseptal development has been well established; perturbation of epicardial development results in cardiac abnormalities, including thinning of the ventricular myocardial wall and malformations of the atrioventricular valvuloseptal complex. To determine the spatiotemporal contribution of epicardially derived cells to the developing fibroblast population in the heart, we have used a mWt1/IRES/GFP-Cre mouse to trace the fate of EPDCs from embryonic day (ED)10 until birth. EPDCs begin to populate the compact ventricular myocardium around ED12. The migration of epicardially derived fibroblasts toward the interface between compact and trabecular myocardium is completed around ED14. Remarkably, epicardially derived fibroblasts do not migrate into the trabecular myocardium until after ED17. Migration of EPDCs into the atrioventricular cushion mesenchyme commences around ED12. As development progresses, the number of EPDCs increases significantly, specifically in the leaflets which derive from the lateral atrioventricular cushions. In these developing leaflets the epicardially derived fibroblasts eventually largely replace the endocardially derived cells. Importantly, the contribution of EPDCs to the leaflets derived from the major AV cushions is very limited. The differential contribution of EPDCs to the various leaflets of the atrioventricular valves provides a new paradigm in valve development and could lead to new insights into the pathogenesis of abnormalities that preferentially affect individual components of this region of the heart. The notion that there is a significant difference in the contribution of epicardially and endocardially derived cells to the individual leaflets of the atrioventricular valves has also important pragmatic consequences for the use of endocardial and epicardial cre-mouse models in studies of heart development.


Asunto(s)
Fibroblastos/citología , Válvulas Cardíacas/embriología , Corazón/embriología , Pericardio/citología , Animales , Desarrollo Embrionario , Válvulas Cardíacas/citología , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/embriología , Ratones , Organogénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...