Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 335
Filtrar
1.
J Cell Biol ; 221(6)2022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35482005

RESUMEN

Tissue-resident macrophages play essential functions in the maintenance of tissue homeostasis and repair. Recently, the endocardium has been reported as a de novo hemogenic site for the contribution of hematopoietic cells, including cardiac macrophages, during embryogenesis. These observations challenge the current consensus that hematopoiesis originates from the hemogenic endothelium within the yolk sac and dorsal aorta. Whether the developing endocardium has such a hemogenic potential requires further investigation. Here, we generated new genetic tools to trace endocardial cells and reassessed their potential contribution to hematopoietic cells in the developing heart. Fate-mapping analyses revealed that the endocardium contributed minimally to cardiac macrophages and circulating blood cells. Instead, cardiac macrophages were mainly derived from the endothelium during primitive/transient definitive (yolk sac) and definitive (dorsal aorta) hematopoiesis. Our findings refute the concept of endocardial hematopoiesis, suggesting that the developing endocardium gives rise minimally to hematopoietic cells, including cardiac macrophages.


Asunto(s)
Linaje de la Célula , Corazón , Macrófagos , Miocardio , Animales , Aorta/citología , Endocardio/citología , Corazón/embriología , Hematopoyesis/genética , Miocardio/citología , Saco Vitelino/citología
2.
Cell Rep ; 37(1): 109782, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34610316

RESUMEN

In the zebrafish embryo, the onset of blood flow generates fluid shear stress on endocardial cells, which are specialized endothelial cells that line the interior of the heart. High levels of fluid shear stress activate both Notch and Klf2 signaling, which play crucial roles in atrioventricular valvulogenesis. However, it remains unclear why only individual endocardial cells ingress into the cardiac jelly and initiate valvulogenesis. Here, we show that lateral inhibition between endocardial cells, mediated by Notch, singles out Delta-like-4-positive endocardial cells. These cells ingress into the cardiac jelly, where they form an abluminal cell population. Delta-like-4-positive cells ingress in response to Wnt9a, which is produced in parallel through an Erk5-Klf2-Wnt9a signaling cascade also activated by blood flow. Hence, mechanical stimulation activates parallel mechanosensitive signaling pathways that produce binary effects by driving endocardial cells toward either luminal or abluminal fates. Ultimately, these cell fate decisions sculpt cardiac valve leaflets.


Asunto(s)
Endocardio/metabolismo , Mecanotransducción Celular , Transducción de Señal , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente/metabolismo , Embrión no Mamífero/metabolismo , Embrión no Mamífero/patología , Desarrollo Embrionario , Endocardio/citología , Válvulas Cardíacas/crecimiento & desarrollo , Válvulas Cardíacas/metabolismo , Válvulas Cardíacas/patología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Morfolinos/metabolismo , Receptores de Neurotransmisores/antagonistas & inhibidores , Receptores de Neurotransmisores/genética , Receptores de Neurotransmisores/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética
3.
Clin Sci (Lond) ; 135(6): 829-846, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33720353

RESUMEN

The endothelial-to-mesenchymal transition (EndMT) is a critical process that occurs during the development of the outflow tract (OFT). Malformations of the OFT can lead to the occurrence of conotruncal defect (CTD). SOX7 duplication has been reported in patients with congenital CTD, but its specific role in OFT development remains poorly understood. To decipher this, histological analysis showed that SRY-related HMG-box 7 (SOX7) was regionally expressed in the endocardial endothelial cells and in the mesenchymal cells of the OFT, where EndMT occurs. Experiments, using in vitro collagen gel culture system, revealed that SOX7 was a negative regulator of EndMT that inhibited endocardial cell (EC) migration and resulted in decreased number of mesenchymal cells. Forced expression of SOX7 in endothelial cells blocked further migration and improved the expression of the adhesion protein vascular endothelial (VE)-cadherin (VE-cadherin). Moreover, a VE-cadherin knockdown could partly reverse the SOX7-mediated repression of cell migration. Luciferase and electrophoretic mobility shift assay (EMSA) demonstrated that SOX7 up-regulated VE-cadherin by directly binding to the gene's promoter in endothelial cells. The coding exons and splicing regions of the SOX7 gene were also scanned in the 536 sporadic CTD patients and in 300 unaffected controls, which revealed four heterozygous SOX7 mutations. Luciferase assays revealed that two SOX7 variants weakened the transactivation of the VE-cadherin promoter. In conclusion, SOX7 inhibited EndMT during OFT development by directly up-regulating the endothelial-specific adhesion molecule VE-cadherin. SOX7 mutations can lead to impaired EndMT by regulating VE-cadherin, which may give rise to the molecular mechanisms associated with SOX7 in CTD pathogenesis.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Endocardio/embriología , Cardiopatías Congénitas/embriología , Factores de Transcripción SOXF/metabolismo , Animales , Antígenos CD/genética , Cadherinas/genética , Movimiento Celular , Embrión de Mamíferos , Endocardio/citología , Endotelio/crecimiento & desarrollo , Transición Epitelial-Mesenquimal/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Ratas , Factores de Transcripción SOXF/genética
4.
Gene Expr Patterns ; 39: 119165, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33373688

RESUMEN

Endoglin (ENG) is essential for cardiovascular development and is expressed in the heart from its earliest developmental stages. ENG expression has been reported in the cardiac crescent, endocardium, valve mesenchyme and coronary vascular endothelial cells. However, its expression in these cell types is non-uniform and the dynamic changes in ENG expression during heart development have not been systematically studied. Using immunofluorescent staining we tracked ENG protein expression in mouse embryonic hearts aged from 11.5 to 17.5 days, and in postnatal and adult hearts. ENG is expressed in the endocardium and in venous endothelial cells throughout these developmental stages. ENG protein is down-regulated by approximately two-fold as a subset of early coronary veins reprogram to form arteries within the developing myocardium from E13.5. This two-fold higher ratio of ENG protein in veins versus arteries is maintained throughout cardiac development and in the adult heart. ENG is also down-regulated two-fold following mesenchymal transition of endocardial cells to form cardiac valve mesenchyme, whilst expression of the pan-endothelial marker CD31 is completely lost. A subset of epicardial cells (which do not express ENG protein) delaminate and undergo a similar mesenchymal transition to form epicardially derived cells (EPDCs). This transient intra-myocardial mesenchymal cell population expresses low levels of ENG protein, similar to valve mesenchyme. In conclusion, ENG shows dynamic changes of expression in vascular endothelial cells, endocardial cells and mesenchymal cells in the developing heart that vary according to cardiovascular cell type.


Asunto(s)
Endoglina/genética , Corazón/embriología , Miocitos Cardíacos/metabolismo , Animales , Vasos Coronarios/embriología , Vasos Coronarios/metabolismo , Endocardio/citología , Endocardio/embriología , Endocardio/metabolismo , Endoglina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL
5.
Development ; 147(12)2020 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-32439760

RESUMEN

Physical forces are important participants in the cellular dynamics that shape developing organs. During heart formation, for example, contractility and blood flow generate biomechanical cues that influence patterns of cell behavior. Here, we address the interplay between function and form during the assembly of the cardiac outflow tract (OFT), a crucial connection between the heart and vasculature that develops while circulation is under way. In zebrafish, we find that the OFT expands via accrual of both endocardial and myocardial cells. However, when cardiac function is disrupted, OFT endocardial growth ceases, accompanied by reduced proliferation and reduced addition of cells from adjacent vessels. The flow-responsive TGFß receptor Acvrl1 is required for addition of endocardial cells, but not for their proliferation, indicating distinct modes of function-dependent regulation for each of these essential cell behaviors. Together, our results indicate that cardiac function modulates OFT morphogenesis by triggering endocardial cell accumulation that induces OFT lumen expansion and shapes OFT dimensions. Moreover, these morphogenetic mechanisms provide new perspectives regarding the potential causes of cardiac birth defects.


Asunto(s)
Endocardio/metabolismo , Corazón/fisiología , Pez Cebra/metabolismo , Receptores de Activinas/antagonistas & inhibidores , Receptores de Activinas/genética , Receptores de Activinas/metabolismo , Animales , Animales Modificados Genéticamente/crecimiento & desarrollo , Animales Modificados Genéticamente/metabolismo , Proliferación Celular , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Endocardio/citología , Corazón/anatomía & histología , Corazón/crecimiento & desarrollo , Morfolinos/metabolismo , Troponina T/antagonistas & inhibidores , Troponina T/genética , Troponina T/metabolismo , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
6.
J Vis Exp ; (158)2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32364545

RESUMEN

It has been shown that endocardial endothelial cells (EECs) and coronary endothelial cells (CECs) differ in origin, development, markers, and functions. Consequently, these two cell populations play unique roles in cardiac diseases. Current studies involving isolated endothelial cells investigate cell populations consisting of both EECs and CECs. This protocol outlines a method to independently isolate these two cell populations for cell-specific characterization. Following the collection of the left and right ventricular free wall, endothelial cells from the outer surface and inner surface are separately liberated using a digestion buffer solution. The sequential digestion of the outer surface and the inner endocardial layer retained separation of the two endothelial cell populations. The separate isolation of EECs and CECs is further verified through the identification of markers specific to each population. Based on previously published single cell RNA profiling in the mouse heart, the Npr3, Hapln1, and Cdh11 gene expression is unique to EECs; while Fabp4, Mgll, and Cd36 gene expression is unique to CECs. qPCR data revealed enriched expression of these characteristic markers in their respective samples, indicating successful EEC and CEC isolation, as well as maintenance of cell phenotype, enabling further cell-specific functional analysis.


Asunto(s)
Vasos Coronarios/citología , Endocardio/citología , Endotelio Vascular/citología , Ventrículos Cardíacos/citología , Corazón/fisiología , Animales , Biomarcadores/metabolismo , Células Cultivadas , Vasos Coronarios/metabolismo , Endocardio/metabolismo , Endotelio Vascular/metabolismo , Perfilación de la Expresión Génica , Ventrículos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley
7.
Anat Histol Embryol ; 49(5): 643-655, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32319704

RESUMEN

The initial intraembryonic vasculogenesis occurs in the cardiogenic mesoderm. Here, a cell population of proendocardial cells detaches from the mesoderm that subsequently generates the single endocardial tube by forming vascular plexuses. In the course of embryogenesis, the endocardium retains vasculogenic, angiogenic and haematopoietic potential. The coronary blood vessels that sustain the rapidly expanding myocardium develop in the course of the formation of the cardiac loop by vasculogenesis and angiogenesis from progenitor cells of the proepicardial serosa at the venous pole of the heart as well as from the endocardium and endothelial cells of the sinus venosus. Prospective coronary endothelial cells and progenitor cells of the coronary blood vessel walls (smooth muscle cells, perivascular cells) originate from different cell populations that are in close spatial as well as regulatory connection with each other. Vasculo- and angiogenesis of the coronary blood vessels are for a large part regulated by the epicardium and epicardium-derived cells. Vasculogenic and angiogenic signalling pathways include the vascular endothelial growth factors, the angiopoietins and the fibroblast growth factors and their receptors.


Asunto(s)
Vasos Sanguíneos/embriología , Vasos Sanguíneos/crecimiento & desarrollo , Corazón/embriología , Corazón/crecimiento & desarrollo , Animales , Vasos Coronarios/embriología , Vasos Coronarios/crecimiento & desarrollo , Endocardio/citología , Endocardio/embriología , Endocardio/crecimiento & desarrollo , Endotelio/citología , Humanos
8.
Sci Rep ; 10(1): 2617, 2020 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-32054938

RESUMEN

Cardiac pathologies associated with arrhythmic activity are often accompanied by inflammation. The contribution of inflammatory cells to the electrophysiological properties of injured myocardium is unknown. Myocardial scar cell types and intercellular contacts were analyzed using a three-dimensional reconstruction from serial blockface scanning electron microscopy data. Three distinct cell populations were identified: inflammatory, fibroblastic and endocardial cells. While individual fibroblastic cells interface with a greater number of cells, inflammatory cells have the largest contact area suggesting a role in establishing intercellular electrical connections in scar tissue. Optical mapping was used to study the electrophysiological properties of scars in fetal liver chimeric mice generated using connexin43 knockout donors (bmpKO). Voltage changes were elicited in response to applied current pulses. Isopotential maps showed a steeper pattern of decay with distance from the electrode in scars compared with uninjured regions, suggesting reduced electrical coupling. The tissue decay constant, defined as the distance voltage reaches 37% of the amplitude at the edge of the scar, was 0.48 ± 0.04 mm (n = 11) in the scar of the bmpCTL group and decreased 37.5% in the bmpKO group (n = 10). Together these data demonstrate inflammatory cells significantly contribute to scar electrophysiology through coupling mediated at least partially by connexin43 expression.


Asunto(s)
Células de la Médula Ósea/patología , Conexina 43/análisis , Lesiones Cardíacas/patología , Miocardio/patología , Animales , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Endocardio/citología , Endocardio/patología , Endocardio/fisiopatología , Fibroblastos/patología , Lesiones Cardíacas/fisiopatología , Imagenología Tridimensional , Inflamación/patología , Inflamación/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Rastreo , Miocardio/citología
9.
Cell Immunol ; 347: 104019, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31744596

RESUMEN

Quantitative gene expression profiling of cardiac allografts characterizes the phenotype of the alloimmune response, yields information regarding differential effects that may be associated with various anti-rejection drug regimens, and generates testable hypotheses regarding the pathogenesis of the chronic rejection lesions typically observed in non-human primate heart transplant models. The goal of this study was to assess interplatform performance and variability between the relatively novel NanoString nCounter Analysis System, ΔΔCT (relative) RT-qPCR, and standard curve (absolute) RT-qPCR utilizing cynomolgus monkey cardiac allografts. Methods for RNA isolation and preamplification were also systematically evaluated and effective methods are proposed. In this study, we demonstrate strong correlation between the two RT-qPCR methods, but variable and, at times, weak correlation between RT-qPCR and NanoString. NanoString fold change results demonstrate less sensitivity to small changes in gene expression than RT-qPCR. These findings appear to be driven by technical aspects of each platform that influence the conditions under which each technique is ideal. Collectively, our data contribute to the general effort to optimally utilize gene expression profiling techniques, not only for transplanted tissues, but for many other applications where accurate rank-order of gene expression versus precise quantification of absolute gene transcript number may be relatively valuable.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Rechazo de Injerto/genética , Trasplante de Corazón/métodos , Ventrículos Cardíacos/citología , Aloinjertos , Animales , Endocardio/citología , Expresión Génica/fisiología , Genes Esenciales/genética , Terapia de Inmunosupresión , Macaca fascicularis , Técnicas de Amplificación de Ácido Nucleico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Trasplantes
10.
Stem Cell Rev Rep ; 16(1): 181-185, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31758373

RESUMEN

Very Small Embryonic-Like (VSEL) stem cells are a proposed pluripotent population, residing in adult tissues. VSELs have been described in multiple tissues including bone marrow, cord blood, and gonads. They exhibit multiple characteristics of embryonic stem cells including the ability to differentiate into cellular lineages of all three germ layers, including cardiomyocytes and vascular endothelial cells. However, their presence in adult solid organs such as heart in humans has not been established. VSELs are valuable source of stem cells for tissue regeneration and replacement of cells for turnover and usual wear-and-tear. The purpose of our study was to explore the existence of human VSELs (huVSELs) in human heart tissue and examine the changes in their prevalence with aging and cardiac disease. Human heart tissue, collected from healthy and ischemic heart disease subjects was examined for the prevalence of VSELS, defined as CD45-/CD133+/SSEA4+. Both epicardial and endocardial tissues were examined comparing VSEL numbers across different age groups. Our data confirm the existence of huVSELs in adult hearts with decreasing prevalence during aging. This is the first evidence of huVSELs in adult cardiac tissue. Cardiac huVSELs could be further explored in future studies to characterize their primitive potential and therapeutic potential in regenerative studies.


Asunto(s)
Endocardio/crecimiento & desarrollo , Células Madre Embrionarias Humanas/citología , Miocardio/citología , Pericardio/crecimiento & desarrollo , Adolescente , Adulto , Factores de Edad , Anciano , Diferenciación Celular/genética , Linaje de la Célula/genética , Niño , Endocardio/citología , Células Endoteliales/citología , Femenino , Sangre Fetal/citología , Humanos , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/citología , Pericardio/citología , Células Madre Pluripotentes/citología , Adulto Joven
11.
Nat Commun ; 10(1): 4113, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31511517

RESUMEN

Intra-organ communication guides morphogenetic processes that are essential for an organ to carry out complex physiological functions. In the heart, the growth of the myocardium is tightly coupled to that of the endocardium, a specialized endothelial tissue that lines its interior. Several molecular pathways have been implicated in the communication between these tissues including secreted factors, components of the extracellular matrix, or proteins involved in cell-cell communication. Yet, it is unknown how the growth of the endocardium is coordinated with that of the myocardium. Here, we show that an increased expansion of the myocardial atrial chamber volume generates higher junctional forces within endocardial cells. This leads to biomechanical signaling involving VE-cadherin, triggering nuclear localization of the Hippo pathway transcriptional regulator Yap1 and endocardial proliferation. Our work suggests that the growth of the endocardium results from myocardial chamber volume expansion and ends when the tension on the tissue is relaxed.


Asunto(s)
Endocardio/crecimiento & desarrollo , Miocardio/metabolismo , Transducción de Señal , Pez Cebra/embriología , Animales , Antígenos CD/metabolismo , Fenómenos Biomecánicos , Cadherinas/metabolismo , Núcleo Celular/metabolismo , Proliferación Celular , Tamaño de la Célula , Proteínas del Citoesqueleto/metabolismo , Endocardio/citología , Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , Proteína Homeótica Nkx-2.5/metabolismo , Uniones Intercelulares/metabolismo , Modelos Biológicos , Mutación/genética , Transactivadores/metabolismo , Proteínas Wnt/metabolismo , Proteínas Señalizadoras YAP , Proteínas de Pez Cebra/metabolismo
12.
Sci Rep ; 9(1): 11953, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31420575

RESUMEN

The endocardium is the endothelial component of the vertebrate heart and plays a key role in heart development. Where, when, and how the endocardium segregates during embryogenesis have remained largely unknown, however. We now show that Nkx2-5+ cardiac progenitor cells (CPCs) that express the Sry-type HMG box gene Sox17 from embryonic day (E) 7.5 to E8.5 specifically differentiate into the endocardium in mouse embryos. Although Sox17 is not essential or sufficient for endocardium fate, it can bias the fate of CPCs toward the endocardium. On the other hand, Sox17 expression in the endocardium is required for heart development. Deletion of Sox17 specifically in the mesoderm markedly impaired endocardium development with regard to cell proliferation and behavior. The proliferation of cardiomyocytes, ventricular trabeculation, and myocardium thickening were also impaired in a non-cell-autonomous manner in the Sox17 mutant, likely as a consequence of down-regulation of NOTCH signaling. An unknown signal, regulated by Sox17 and required for nurturing of the myocardium, is responsible for the reduction in NOTCH-related genes in the mutant embryos. Our results thus provide insight into differentiation of the endocardium and its role in heart development.


Asunto(s)
Diferenciación Celular , Embrión de Mamíferos/embriología , Endocardio/embriología , Regulación del Desarrollo de la Expresión Génica , Proteínas HMGB/biosíntesis , Factores de Transcripción SOXF/biosíntesis , Transducción de Señal , Células Madre/metabolismo , Animales , Embrión de Mamíferos/citología , Endocardio/citología , Proteínas HMGB/genética , Mesodermo/citología , Mesodermo/embriología , Ratones , Ratones Transgénicos , Receptores Notch/genética , Receptores Notch/metabolismo , Factores de Transcripción SOXF/genética , Células Madre/citología
13.
Mol Cell Proteomics ; 18(9): 1782-1795, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31249105

RESUMEN

The endocardium is a specialized endothelium that lines the inner surface of the heart. Functional studies in mice and zebrafish have established that the endocardium is a source of instructive signals for the development of cardiac structures, including the heart valves and chambers. Here, we characterized the NOTCH-dependent endocardial secretome by manipulating NOTCH activity in mouse embryonic endocardial cells (MEEC) followed by mass spectrometry-based proteomics. We profiled different sets of soluble factors whose secretion not only responds to NOTCH activation but also shows differential ligand specificity, suggesting that ligand-specific inputs may regulate the expression of secreted proteins involved in different cardiac development processes. NOTCH signaling activation correlates with a transforming growth factor-ß2 (TGFß2)-rich secretome and the delivery of paracrine signals involved in focal adhesion and extracellular matrix (ECM) deposition and remodeling. In contrast, NOTCH inhibition is accompanied by the up-regulation of specific semaphorins that may modulate cell migration. The secretome protein expression data showed a good correlation with gene profiling of RNA expression in embryonic endocardial cells. Additional characterization by in situ hybridization in mouse embryos revealed expression of various NOTCH candidate effector genes (Tgfß2, Loxl2, Ptx3, Timp3, Fbln2, and Dcn) in heart valve endocardium and/or mesenchyme. Validating these results, mice with conditional Dll4 or Jag1 loss-of-function mutations showed gene expression alterations similar to those observed at the protein level in vitro These results provide the first description of the NOTCH-dependent endocardial secretome and validate MEEC as a tool for assaying the endocardial secretome response to a variety of stimuli and the potential use of this system for drug screening.


Asunto(s)
Endocardio/embriología , Endocardio/metabolismo , Válvulas Cardíacas/embriología , Receptores Notch/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Benzazepinas/farmacología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Células Cultivadas , Endocardio/citología , Endocardio/efectos de los fármacos , Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Válvulas Cardíacas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Ratones Mutantes , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores Notch/genética , Reproducibilidad de los Resultados
14.
Development ; 146(9)2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-31023876

RESUMEN

The endocardium interacts with the myocardium to promote proliferation and morphogenesis during the later stages of heart development. However, the role of the endocardium in early cardiac ontogeny remains under-explored. Given the shared origin, subsequent juxtaposition, and essential cell-cell interactions of endocardial and myocardial cells throughout heart development, we hypothesized that paracrine signaling from the endocardium to the myocardium is crucial for initiating early differentiation of myocardial cells. To test this, we generated an in vitro, endocardial-specific ablation model using the diphtheria toxin receptor under the regulatory elements of the Nfatc1 genomic locus (NFATc1-DTR). Early treatment of NFATc1-DTR mouse embryoid bodies with diphtheria toxin efficiently ablated endocardial cells, which significantly attenuated the percentage of beating EBs in culture and expression of early and late myocardial differentiation markers. The addition of Bmp2 during endocardial ablation partially rescued myocyte differentiation, maturation and function. Therefore, we conclude that early stages of myocardial differentiation rely on endocardial paracrine signaling mediated in part by Bmp2. Our findings provide novel insight into early endocardial-myocardial interactions that can be explored to promote early myocardial development and growth.


Asunto(s)
Diferenciación Celular/fisiología , Endocardio/citología , Endocardio/metabolismo , Miocardio/citología , Miocardio/metabolismo , Animales , Diferenciación Celular/genética , Femenino , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Organogénesis/genética , Organogénesis/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
15.
J Biol Chem ; 294(22): 8894-8906, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31010826

RESUMEN

Early embryonic endocardium undergoes endothelial-to-mesenchymal transition to form cardiac cushion mesenchymal cells (MCs). Embryonic endocardium also gives rise to fibroblasts, intramyocardial adipocytes, and coronary mural cells, including smooth muscle cells and pericytes, in development. Whether endocardial cells directly differentiate into fibroblasts, coronary mural cells, and adipocytes or indirectly via an intermediate stage of endocardial-derived cushion MCs remains unknown. In addition to endocardium, epicardium and neural crest also contribute to cardiac cushion MCs. Given the developmental heterogeneity of cushion MCs and the lack of specific markers for endocardial-derived cushion MCs, conventional genetic lineage tracing utilizing Cre recombinase driven by one specific regulatory element is not sufficient to examine the fates of endocardial-derived cushion MCs. Intersectional genetic targeting approaches, which combine regulatory elements from two or more genes, have been employed to increase the specificity of cell targeting. Here, we developed a dual-recombinase intersectional targeting approach using Nfatc1-Dre, Sox9-CreER, and Cre/Dre double-dependent reporter Ai66 to specifically label endocardial-derived cushion MCs. Taking advantage of intersectional lineage tracing, we found that a subset of cardiac cells including fibroblasts, coronary mural cells, and intramyocardial adipocytes in adult hearts were derived from endocardial-derived cushion MCs. Our study suggests that embryonic endocardium contributes to cushion MCs first, and then endocardial-derived cushion MCs migrate into myocardium and differentiate into fibroblasts, coronary mural cells, and adipocytes in development. Understanding developmental origins of cardiac cell lineages will provide us more insights into cardiac development, regeneration, and diseases.


Asunto(s)
Adipocitos/citología , Linaje de la Célula , Endocardio/citología , Células Endoteliales/citología , Fibroblastos/citología , Células Madre Mesenquimatosas/citología , Adipocitos/metabolismo , Animales , Diferenciación Celular , Movimiento Celular , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Miocardio/metabolismo , Miocardio/patología , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo
16.
Can J Physiol Pharmacol ; 97(9): 820-828, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30897335

RESUMEN

Tumor necrosis factor alpha (TNFα) and its type 1 receptor (TNFR1) are implicated in several autoimmune diseases, including rheumatoid arthritis, and are associated with complications at the cardiovascular level. Using human cardiomyocytes, vascular smooth muscle, vascular endothelial, and endocardial endothelial cells coupled to indirect immunofluorescence, our results showed the presence of TNFR1 at the levels of the plasma membrane (including the cytosol) and mostly at the level of the nuclear membranes (including the nucleoplasm). The distribution of the receptor is different between cell types; however, the density is significantly higher at the nuclear level in all 4 cell types. The density of the receptor was the highest in contractile cells including the cardiomyocytes and vascular smooth muscle cells, compared with endothelial cells including endocardial endothelial and vascular endothelial cells. Using the Ca2+ probe Fluo-3 coupled to quantitative confocal microscopy, our results showed that the cytokine induced a sustained Ca2+ increase in both the cytosol and nucleoplasm of all 4 cell types. This increase was more significant at the nuclear level, mainly in endothelial cells. Our results demonstrated the presence of TNFR1 at both the cell and nuclear membranes of cardiovascular cells, and that its activation modulated both cytosolic and nuclear Ca2+.


Asunto(s)
Calcio/metabolismo , Sistema Cardiovascular/citología , Núcleo Celular/metabolismo , Citosol/metabolismo , Espacio Extracelular/metabolismo , Espacio Intracelular/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Adolescente , Adulto , Endocardio/citología , Células Endoteliales/citología , Femenino , Homeostasis , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
17.
Curr Top Dev Biol ; 132: 395-416, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30797515

RESUMEN

Heart formation involves a complex series of tissue rearrangements, during which regions of the developing organ expand, bend, converge, and protrude in order to create the specific shapes of important cardiac components. Much of this morphogenesis takes place while cardiac function is underway, with blood flowing through the rapidly contracting chambers. Fluid forces are therefore likely to influence the regulation of cardiac morphogenesis, but it is not yet clear how these biomechanical cues direct specific cellular behaviors. In recent years, the optical accessibility and genetic amenability of zebrafish embryos have facilitated unique opportunities to integrate the analysis of flow parameters with the molecular and cellular dynamics underlying cardiogenesis. Consequently, we are making progress toward a comprehensive view of the biomechanical regulation of cardiac chamber emergence, atrioventricular canal differentiation, and ventricular trabeculation. In this review, we highlight a series of studies in zebrafish that have provided new insight into how cardiac function can shape cardiac morphology, with a particular focus on how hemodynamics can impact cardiac cell behavior. Over the long-term, this knowledge will undoubtedly guide our consideration of the potential causes of congenital heart disease.


Asunto(s)
Líquidos Corporales/fisiología , Corazón/embriología , Corazón/fisiología , Morfogénesis , Pez Cebra/embriología , Animales , Fenómenos Biomecánicos , Diferenciación Celular/genética , Cojinetes Endocárdicos/citología , Cojinetes Endocárdicos/embriología , Cojinetes Endocárdicos/metabolismo , Endocardio/citología , Endocardio/embriología , Endocardio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/anatomía & histología , Pez Cebra/genética
18.
Mol Cell Biochem ; 451(1-2): 1-10, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29934862

RESUMEN

Endocardial endothelium, which lines the chambers of the heart, is distinct in its origin, structure, and function. Characterization studies using genomics and proteomics have reported molecular signatures supporting the structural and functional heterogeneity of various endothelial cells. However, though functionally very important, no studies at protein level have been conducted so far characterizing endocardial endothelium. In this study, we used endothelial cells from pig heart to investigate if endocardial endothelial cells are distinct at the proteome level. Using a high-throughput liquid chromatography-tandem mass spectrometry for proteome profiling and expression, we identified sets of proteins that belong to specific biological processes and metabolic pathways in endocardial endothelial cells supporting its specific structural and functional roles. The study also identified several transcription factors and cell surface markers, which may have roles in the specificity of endocardial endothelium. The detection of sets proteins preferentially expressed in endocardial endothelium offers new insights into its role in the regulation of cardiac function. Data are made available through ProteomeXchange with identifier PXD009194.


Asunto(s)
Biomarcadores/metabolismo , Endocardio/metabolismo , Endotelio Vascular/metabolismo , Proteoma/análisis , Proteómica/métodos , Animales , Endocardio/citología , Endotelio Vascular/citología , Masculino , Porcinos
19.
Artículo en Inglés | MEDLINE | ID: mdl-30580044

RESUMEN

INTRODUCTION: The use of in silico cardiac action potential simulations is one of the pillars of the CiPA initiative (Comprehensive in vitro Proarrhythmia Assay) currently under evaluation designed to detect more accurately proarrhythmic liabilities of new drug candidate. In order to take into account the variability of clinical situations, we propose to improve this method by studying the impact of various disease states on arrhythmic events induced by 30 torsadogenic or non-torsadogenic compounds. METHOD: In silico modelling was done on the human myocytes using the Dutta revised O'Hara-Rudy algorithm. Results were analysed using a new metric based on the compound IC50s against the seven cardiac ionic currents considered to be the most important by the CiPA initiative (IKr, IKs, INa, INaL, IK1, Ito, ICaL) and the minimal rate of action potential voltage decrease calculated at the early-afterdepolarization (EAD) take-off membrane voltage (Vmin). RESULTS: The specific threshold at which each torsadogenic compounds induced EAD, was exacerbated by the presence of cardiac risk factors ranked as follows: congestive heart failure > hypertrophic cardiomyopathy > cardiac pause > no risk factor. Non-torsadogenic compounds induced no EAD even in the presence of cardiac risk factors. DISCUSSION: The present study highlighted the impact of pre-existing cardiovascular disease on arrhythmic event detection suggesting that disease state modelling may need to be incorporated in order to fully realize the goal of the CiPA paradigm in a more accurate predictability of proarrhythmic liabilities of new drug candidate.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Miocitos Cardíacos/efectos de los fármacos , Torsades de Pointes/inducido químicamente , Potenciales de Acción/efectos de los fármacos , Algoritmos , Simulación por Computador , Endocardio/citología , Endocardio/efectos de los fármacos , Humanos , Modelos Cardiovasculares , Medición de Riesgo
20.
Biophys J ; 115(12): 2486-2498, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30503533

RESUMEN

BACKGROUND: Understanding the biophysical processes by which electrical stimuli applied to cardiac tissue may result in local activation is important in both the experimental and clinical electrophysiology laboratory environments, as well as for gaining a more in-depth knowledge of the mechanisms of focal-trigger-induced arrhythmias. Previous computational models have predicted that local myocardial tissue architecture alone may significantly modulate tissue excitability, affecting both the local stimulus current required to excite the tissue and the local effective refractory period (ERP). In this work, we present experimental validation of this structural modulation of local tissue excitability on the endocardial tissue surface, use computational models to provide mechanistic understanding of this phenomena in relation to localized changes in electrotonic loading, and demonstrate its implications for the capture of afterdepolarizations. METHODS AND RESULTS: Experiments on rabbit ventricular wedge preparations showed that endocardial ridges (surfaces of negative mean curvature) had a stimulus capture threshold that was 0.21 ± 0.03 V less than endocardial grooves (surfaces of positive mean curvature) for pairwise comparison (24% reduction, corresponding to 56.2 ± 6.4% of the energy). When stimulated at the minimal stimulus strength for capture, ridge locations showed a shorter ERP than grooves (n = 6, mean pairwise difference 7.4 ± 4.2 ms). When each site was stimulated with identical-strength stimuli, the difference in ERP was further increased (mean pairwise difference 15.8 ± 5.3 ms). Computational bidomain models of highly idealized cylindrical endocardial structures qualitatively agreed with these findings, showing that such changes in excitability are driven by structural modulation in electrotonic loading, quantifying this relationship as a function of surface curvature. Simulations further showed that capture of delayed afterdepolarizations was more likely in trabecular ridges than grooves, driven by this difference in loading. CONCLUSIONS: We have demonstrated experimentally and explained mechanistically in computer simulations that the ability to capture tissue on the endocardial surface depends upon the local tissue architecture. These findings have important implications for deepening our understanding of excitability differences related to anatomical structure during stimulus application that may have important applications in the translation of novel experimental optogenetics pacing strategies. The uncovered preferential vulnerability to capture of afterdepolarizations of endocardial ridges, compared to grooves, provides important insight for understanding the mechanisms of focal-trigger-induced arrhythmias.


Asunto(s)
Endocardio/citología , Endocardio/fisiología , Ventrículos Cardíacos/citología , Modelos Cardiovasculares , Periodo Refractario Electrofisiológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...