Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 268
Filtrar
1.
Circ Res ; 130(3): 352-365, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-34995101

RESUMEN

BACKGROUND: Unraveling how new coronary arteries develop may provide critical information for establishing novel therapeutic approaches to treating ischemic cardiac diseases. There are 2 distinct coronary vascular populations derived from different origins in the developing heart. Understanding the formation of coronary arteries may provide insights into new ways of promoting coronary artery formation after myocardial infarction. METHODS: To understand how intramyocardial coronary arteries are generated to connect these 2 coronary vascular populations, we combined genetic lineage tracing, light sheet microscopy, fluorescence micro-optical sectioning tomography, and tissue-specific gene knockout approaches to understand their cellular and molecular mechanisms. RESULTS: We show that a subset of intramyocardial coronary arteries form by angiogenic extension of endocardium-derived vascular tunnels in the neonatal heart. Three-dimensional whole-mount fluorescence imaging showed that these endocardium-derived vascular tunnels or tubes adopt an arterial fate in neonates. Mechanistically, we implicate Mettl3 (methyltransferase-like protein 3) and Notch signaling in regulating endocardium-derived intramyocardial coronary artery formation. Functionally, these intramyocardial arteries persist into adulthood and play a protective role after myocardial infarction. CONCLUSIONS: A subset of intramyocardial coronary arteries form by extension of endocardium-derived vascular tunnels in the neonatal heart.


Asunto(s)
Vasos Coronarios/embriología , Endocardio/embriología , Animales , Vasos Coronarios/crecimiento & desarrollo , Vasos Coronarios/metabolismo , Endocardio/crecimiento & desarrollo , Endocardio/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Organogénesis
2.
Development ; 149(4)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35088848

RESUMEN

Endothelial cells emerge from the atrioventricular canal to form coronary blood vessels in juvenile zebrafish hearts. We find that pdgfrb is first expressed in the epicardium around the atrioventricular canal and later becomes localized mainly in the mural cells. pdgfrb mutant fish show severe defects in mural cell recruitment and coronary vessel development. Single-cell RNA sequencing analyses identified pdgfrb+ cells as epicardium-derived cells (EPDCs) and mural cells. Mural cells associated with coronary arteries also express cxcl12b and smooth muscle cell markers. Interestingly, these mural cells remain associated with coronary arteries even in the absence of Pdgfrß, although smooth muscle gene expression is downregulated. We find that pdgfrb expression dynamically changes in EPDCs of regenerating hearts. Differential gene expression analyses of pdgfrb+ EPDCs and mural cells suggest that they express genes that are important for regeneration after heart injuries. mdka was identified as a highly upregulated gene in pdgfrb+ cells during heart regeneration. However, pdgfrb but not mdka mutants show defects in heart regeneration after amputation. Our results demonstrate that heterogeneous pdgfrb+ cells are essential for coronary development and heart regeneration.


Asunto(s)
Vasos Coronarios/crecimiento & desarrollo , Vasos Coronarios/metabolismo , Corazón/fisiología , Organogénesis/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Regeneración/fisiología , Animales , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Miocitos del Músculo Liso/metabolismo , Pericardio/metabolismo , Pez Cebra/metabolismo , Pez Cebra/fisiología
3.
Nat Cell Biol ; 23(9): 967-977, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34497373

RESUMEN

New coronary vessels are added to the heart around birth to support postnatal cardiac growth. Here we show that, in late fetal development, the embryonic coronary plexus at the inner myocardium of the ventricles expresses the angiogenic signalling factors VEGFR3 and DLL4 and generates new coronary vessels in neonates. Contrary to a previous model in which the formation of new coronary vessels in neonates from ventricular endocardial cells was proposed, we find that late fetal and neonatal ventricular endocardial cells lack angiogenic potential and do not contribute to new coronary vessels. Instead, we show using lineage-tracing as well as gain- and loss-of-function experiments that the pre-existing embryonic coronary plexus at the inner myocardium undergoes angiogenic expansion through the DLL4-NOTCH1 signalling pathway to vascularize the expanding myocardium. We also show that the pre-existing coronary plexus revascularizes the regenerating neonatal heart through a similar mechanism. These findings provide a different model of neonatal coronary angiogenesis and regeneration, potentially informing cardiovascular medicine.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al Calcio/metabolismo , Vasos Coronarios/crecimiento & desarrollo , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica/fisiología , Receptor Notch1/metabolismo , Animales , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Corazón/crecimiento & desarrollo , Ventrículos Cardíacos/metabolismo , Ratones Transgénicos , Morfogénesis/fisiología , Miocardio/metabolismo , Transducción de Señal/fisiología
4.
Am J Physiol Lung Cell Mol Physiol ; 321(4): L675-L685, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34346780

RESUMEN

Humans and animals with pulmonary hypertension (PH) show right ventricular (RV) capillary growth, which positively correlates with overall RV hypertrophy. However, molecular drivers of RV vascular augmentation in PH are unknown. Prolyl hydroxylase (PHD2) is a regulator of hypoxia-inducible factors (HIFs), which transcriptionally activates several proangiogenic genes, including the glycolytic enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3). We hypothesized that a signaling axis of PHD2-HIF1α-PFKFB3 contributes to adaptive coupling between the RV vasculature and tissue volume to maintain appropriate vascular density in PH. We used design-based stereology to analyze endothelial cell (EC) proliferation and the absolute length of the vascular network in the RV free wall, relative to the tissue volume in mice challenged with hypoxic PH. We observed increased RV EC proliferation starting after 6 h of hypoxia challenge. Using parabiotic mice, we found no evidence for a contribution of circulating EC precursors to the RV vascular network. Mice with transgenic deletion or pharmacological inhibition of PHD2, HIF1α, or PFKFB3 all had evidence of impaired RV vascular adaptation following hypoxia PH challenge. PHD2-HIF1α-PFKFB3 contributes to structural coupling between the RV vascular length and tissue volume in hypoxic mice, consistent with homeostatic mechanisms that maintain appropriate vascular density. Activating this pathway could help augment the RV vasculature and preserve RV substrate delivery in PH, as an approach to promote RV function.


Asunto(s)
Vasos Coronarios/crecimiento & desarrollo , Ventrículos Cardíacos/patología , Hipertensión Pulmonar/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Fosfofructoquinasa-2/metabolismo , Anaerobiosis/fisiología , Animales , Células Endoteliales/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología
5.
Int J Mol Sci ; 22(16)2021 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-34445494

RESUMEN

Despite significant advances in treatment of acute coronary syndromes (ACS) many subjects still develop heart failure due to significantly reduced ejection fraction. Currently, there are no commonly available treatment strategies that replace the infarcted/dysfunctional myocardium. Therefore, understanding the mechanisms that control the regeneration of the heart muscle is important. The development of new coronary vessels plays a pivotal role in cardiac regeneration. Employing microarray expression assays and RT-qPCR validation expression pattern of genes in long-term primary cultured cells isolated form the right atrial appendage (RAA) and right atrium (RA) was evaluated. After using DAVID software, it indicated the analysis expression profiles of genes involved in ontological groups such as: "angiogenesis", "blood vessel morphogenesis", "circulatory system development", "regulation of vasculature development", and "vasculature development" associated with the process of creation new blood vessels. The performed transcriptomic comparative analysis between two different compartments of the heart muscle allowed us to indicate the presence of differences in the expression of key transcripts depending on the cell source. Increases in culture intervals significantly increased expression of SFRP2, PRRX1 genes and some other genes involved in inflammatory process, such as: CCL2, IL6, and ROBO1. Moreover, the right atrial appendage gene encoding lysyl oxidase (LOX) showed much higher expression compared to the pre-cultivation state.


Asunto(s)
Vasos Coronarios/crecimiento & desarrollo , Perfilación de la Expresión Génica/métodos , Desarrollo de Músculos , Miocardio/citología , Animales , Células Cultivadas , Vasos Coronarios/química , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Miocardio/química , Análisis de Secuencia por Matrices de Oligonucleótidos , Cultivo Primario de Células , Porcinos , Secuenciación del Exoma
6.
FASEB J ; 34(8): 10041-10055, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32573852

RESUMEN

As loss of contractile function in heart disease could often be mitigated by increased cardiomyocyte number, expansion of cardiomyocyte endowment paired with increased vascular supply is a desirable therapeutic goal. Insulin-like growth factor 1 (IGF-1) administration increases fetal cardiomyocyte proliferation and heart mass, but how fetal IGF-1 treatment affects coronary growth and function is unknown. Near-term fetal sheep underwent surgical instrumentation and were studied from 127 to 134 d gestation (term = 147 d), receiving either IGF-1 LR3 or vehicle. Coronary growth and function were interrogated using pressure-flow relationships, an episode of acute hypoxia with progressive blockade of adenosine receptors and nitric oxide synthase, and by modeling the determinants of coronary flow. The main findings were that coronary conductance was preserved on a per-gram basis following IGF-1 treatment, adenosine and nitric oxide contributed to hypoxia-mediated coronary vasodilation similarly in IGF-1-treated and Control fetuses, and the relationships between coronary flow and blood oxygen contents were similar between groups. We conclude that IGF-1-stimulated fetal myocardial growth is accompanied by appropriate expansion and function of the coronary vasculature. These findings support IGF-1 as a potential strategy to increase cardiac myocyte and coronary vascular endowment at birth.


Asunto(s)
Vasos Coronarios/crecimiento & desarrollo , Feto/fisiología , Factor I del Crecimiento Similar a la Insulina/farmacología , Miocitos Cardíacos/fisiología , Animales , Vasos Coronarios/citología , Vasos Coronarios/efectos de los fármacos , Femenino , Feto/efectos de los fármacos , Hipoxia/fisiopatología , Masculino , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Ovinos
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.
Biosci Trends ; 14(2): 115-122, 2020 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-32238672

RESUMEN

Dimethylarginine dimethylaminohydrolase-1 (DDAH1) maintains nitric oxide (NO) bioavailability by degrading asymmetric dimethylarginine (ADMA), which is an endogenous inhibitor of nitric oxide synthase (NOS). It has been well established that DDAH1 and exercise play crucial roles in promoting cardiac angiogenesis under pathological conditions. However, the role of DDAH1 in exercise-induced cardiac angiogenesis remains unclear. In this study, we focused on the change in DDAH1 in response to moderate exercise and the underlying mechanism of exercise-induced cardiac angiogenesis. Eight-week-old male DDAH1 global knockout (KO) mice and DDAH1flox/flox mice (wild-type) were randomly divided into sedentary groups (control) and swimming groups (exercise). After eight weeks of swimming at five days per week, all the mice were anesthetized and sacrificed. Histological examination and Western blot analysis were performed. There were low levels of myocardial capillaries in DDAH1 KO mice under control and exercise conditions. Notably, exercise elevated DDAH1 protein expression, as observed by Western blot analysis. The common cardiac angiogenesis biomarkers vascular endothelial growth factor (VEGF) and Caveolin-1 were increased during exercise. A significant difference in VEGF was observed between the DDAH1 KO and wild-type groups. Similarly, increased Caveolin-1 expression was abrogated in DDAH1 KO mice. Furthermore, we tested the R-Ras/AKT/GSK3ß signaling pathway to study the underlying molecular mechanism. DDAH1 may regulate the R-Ras/AKT/GSK3ß pathway due to distinct protein changes in this pathway in the DDAH1 KO and wild-type groups. Our findings suggest that DDAH1 plays an important role in exercise-induced cardiac angiogenesis by regulating the R-Ras/AKT/GSK3ßsignaling pathway.


Asunto(s)
Amidohidrolasas/metabolismo , Vasos Coronarios/crecimiento & desarrollo , Corazón/fisiología , Neovascularización Fisiológica , Condicionamiento Físico Animal/fisiología , Amidohidrolasas/genética , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Conducta Animal , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Óxido Nítrico/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Conducta Sedentaria , Transducción de Señal/fisiología , Proteínas ras/metabolismo
9.
Elife ; 82019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31789590

RESUMEN

Coronaries are essential for myocardial growth and heart function. Notch is crucial for mouse embryonic angiogenesis, but its role in coronary development remains uncertain. We show Jag1, Dll4 and activated Notch1 receptor expression in sinus venosus (SV) endocardium. Endocardial Jag1 removal blocks SV capillary sprouting, while Dll4 inactivation stimulates excessive capillary growth, suggesting that ligand antagonism regulates coronary primary plexus formation. Later endothelial ligand removal, or forced expression of Dll4 or the glycosyltransferase Mfng, blocks coronary plexus remodeling, arterial differentiation, and perivascular cell maturation. Endocardial deletion of Efnb2 phenocopies the coronary arterial defects of Notch mutants. Angiogenic rescue experiments in ventricular explants, or in primary human endothelial cells, indicate that EphrinB2 is a critical effector of antagonistic Dll4 and Jag1 functions in arterial morphogenesis. Thus, coronary arterial precursors are specified in the SV prior to primary coronary plexus formation and subsequent arterial differentiation depends on a Dll4-Jag1-EphrinB2 signaling cascade.


Asunto(s)
Vasos Coronarios/crecimiento & desarrollo , Vasos Coronarios/metabolismo , Efrina-B2/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Jagged-1/metabolismo , Proteínas de la Membrana/metabolismo , Transducción de Señal , Animales , Endocardio/metabolismo , Endotelio Vascular/metabolismo , Ventrículos Cardíacos/crecimiento & desarrollo , Ventrículos Cardíacos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hipoxia/metabolismo , Hipoxia/fisiopatología , Ligandos , Ratones , Morfogénesis , Mutación/genética , Factores de Transcripción NFATC/metabolismo , Neovascularización Fisiológica , Receptores Notch/metabolismo , Estrés Fisiológico , Transcriptoma/genética , Remodelación Vascular
10.
Mech Ageing Dev ; 184: 111161, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31647940

RESUMEN

Aging results in functional and structural changes in the cardiovascular system, translating into a progressive increase of mechanical vessel stiffness, due to a combination of changes in micro-RNA expression patterns, autophagy, arterial calcification, smooth muscle cell migration and proliferation. The two pivotal mechanisms of aging-related endothelial dysfunction are oxidative stress and inflammation, even in the absence of clinical disease. A comprehensive understanding of the aging process is emerging as a primary concern in literature, as vascular aging has recently become a target for prevention and treatment of cardiovascular disease. Change of life-style, diet, antioxidant regimens, anti-inflammatory treatments, senolytic drugs counteract the pro-aging pathways or target senescent cells modulating their detrimental effects. Such therapies aim to reduce the ineluctable burden of age and contrast aging-associated cardiovascular dysfunction. This narrative review intends to summarize the macrovascular and microvascular changes related with aging, as a better understanding of the pathways leading to arterial aging may contribute to design new mechanism-based therapeutic approaches to attenuate the features of vascular senescence and its clinical impact on the cardiovascular system.


Asunto(s)
Capilares/crecimiento & desarrollo , Capilares/ultraestructura , Vasos Coronarios/crecimiento & desarrollo , Vasos Coronarios/ultraestructura , Corazón/crecimiento & desarrollo , Miocardio/ultraestructura , Anciano , Anciano de 80 o más Años , Animales , Humanos , Investigación Biomédica Traslacional
11.
Biomed Res Int ; 2019: 6595437, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31317035

RESUMEN

Endothelial-to-mesenchymal transition (EndMT) is an essential mechanism in myocardial fibrosis (MF). Tongxinluo (TXL) has been confirmed to protect the endothelium against reperfusion injury after acute myocardial infarction (AMI). However, whether TXL can inhibit MF after AMI via inhibiting EndMT remained unknown. This study aims to identify the role of EndMT in MF after AMI as well as the protective effects and underlying mechanisms of TXL on MF. The AMI model was established in rats by ligating left anterior descending coronary artery. Then, rats were administered with high- (0.8 g·kg-1·d-1), mid- (0.4 g·kg-1·d-1), and low- (0.2 g·kg-1·d-1) dose Tongxinluo and benazepril for 4 weeks, respectively. Cardiac function, infarct size, MF, and related indicators of EndMT were measured. In vitro, human cardiac microvascular endothelial cells (HCMECs) were pretreated with TXL for 4 h and then incubated in hypoxia conditions for 3 days to induce EndMT. Under this hypoxic condition, neuregulin-1 (NRG-1) siRNA were further applied to silence NRG-1 expression. Immunofluorescence microscopy was used to assess expression of endothelial marker of vWF and fibrotic marker of Vimentin. Related factors of EndMT were determined by Western blot analysis. TXL treatment significantly improved cardiac function, ameliorated MF, reduced collagen of fibrosis area (types I and III collagen) and limited excessive extracellular matrix deposition (mmp2 and mmp9). In addition, TXL inhibited EndMT in cardiac tissue and hypoxia-induced HCMECs. In hypoxia-induced HCMECs, TXL increased the expression of endothelial markers, whereas decreasing the expression of fibrotic markers, partially through enhanced expressions of NRG-1, phosphorylation of ErbB2, ErbB4, AKT, and downregulated expressions of hypoxia inducible factor-1a and transcription factor snail. After NRG-1 knockdown, the protective effect of TXL on HCMEC was partially abolished. In conclusion, TXL attenuates MF after AMI by inhibiting EndMT and through activating the NRG-1/ErbB- PI3K/AKT signalling cascade.


Asunto(s)
Cardiomiopatías/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Fibrosis/tratamiento farmacológico , Infarto del Miocardio/tratamiento farmacológico , Animales , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Hipoxia de la Célula/efectos de los fármacos , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/crecimiento & desarrollo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Endotelio/efectos de los fármacos , Endotelio/metabolismo , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Ratas
12.
Circ J ; 83(3): 556-566, 2019 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-30700665

RESUMEN

BACKGROUND: Little is known about serial changes in lumen and device dimensions after bioresorbable scaffold implantation in a growing animal model. Methods and Results: ABSORB (n=14) or bare metal stents (ICROS amg [Abbott Vascular, Santa Clara, CA, USA], Winsen-Luhe, Germany; n=15) were implanted in the coronary arteries of domestic swine (a hybrid of Finnish-Norwegian Landrace swine) weighing 30-35 kg. Angiography and optical coherence tomography (OCT) were performed immediately after implantation and repeated at 7 days, 1, 3, 6 and 12 months after the index procedure. One month after implantation, mean lumen area decreased relative to baseline in both groups (relative area change from baseline, -41.4±15.6% for ABSORB vs. -20.9±18.6% for ICROS) while mean device area decreased only in the ABSORB group (relative area change: -11.1±9.4% vs. +0.14±7.95%, respectively). At 12 months, mean lumen area increased relative to baseline in both groups (relative area change from baseline, +55.6±22.4% vs. +32.3±83.6%, respectively) in accordance with the swine growth weighing up to 260-300 kg. Mean device area in the ICROS group remained stable whereas that in the ABSORB group began to increase between 3 and 6 months along with the vessel growth (relative area change: +107.8±25.7% vs. +0.14±7.95%). CONCLUSIONS: In the growing porcine model, ABSORB was associated with greater extent of recoil 1 month after implantation compared with ICROS but demonstrated substantial adaptability to vessel growth in late phase.


Asunto(s)
Implantes Absorbibles/normas , Vasos Coronarios/diagnóstico por imagen , Stents/normas , Tomografía de Coherencia Óptica/métodos , Animales , Angiografía Coronaria/métodos , Vasos Coronarios/crecimiento & desarrollo , Vasos Coronarios/cirugía , Modelos Animales , Diseño de Prótesis/normas , Porcinos , Factores de Tiempo
13.
Cell ; 176(5): 1128-1142.e18, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30686582

RESUMEN

Collateral arteries are an uncommon vessel subtype that can provide alternate blood flow to preserve tissue following vascular occlusion. Some patients with heart disease develop collateral coronary arteries, and this correlates with increased survival. However, it is not known how these collaterals develop or how to stimulate them. We demonstrate that neonatal mouse hearts use a novel mechanism to build collateral arteries in response to injury. Arterial endothelial cells (ECs) migrated away from arteries along existing capillaries and reassembled into collateral arteries, which we termed "artery reassembly". Artery ECs expressed CXCR4, and following injury, capillary ECs induced its ligand, CXCL12. CXCL12 or CXCR4 deletion impaired collateral artery formation and neonatal heart regeneration. Artery reassembly was nearly absent in adults but was induced by exogenous CXCL12. Thus, understanding neonatal regenerative mechanisms can identify pathways that restore these processes in adults and identify potentially translatable therapeutic strategies for ischemic heart disease.


Asunto(s)
Circulación Colateral/fisiología , Corazón/crecimiento & desarrollo , Regeneración/fisiología , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Quimiocina CXCL12/metabolismo , Vasos Coronarios/crecimiento & desarrollo , Células Endoteliales/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/fisiología , Receptores CXCR4/metabolismo , Transducción de Señal
14.
Angiogenesis ; 22(2): 237-250, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30446855

RESUMEN

The coronary vasculature is crucial for normal heart function, yet much remains to be learned about its development, especially the maturation of coronary arterial endothelium. Here, we show that endothelial inactivation of ADAM10, a key regulator of Notch signaling, leads to defects in coronary arterial differentiation, as evidenced by dysregulated genes related to Notch signaling and arterial identity. Moreover, transcriptome analysis indicated reduced EGFR signaling in A10ΔEC coronary endothelium. Further analysis revealed that A10ΔEC mice have enlarged dysfunctional hearts with abnormal myocardial compaction, and increased expression of venous and immature endothelium markers. These findings provide the first evidence for a potential role for endothelial ADAM10 in cardioprotective homeostatic EGFR signaling and implicate ADAM10/Notch signaling in coronary arterial cell specification, which is vital for normal heart development and function. The ADAM10/Notch signaling pathway thus emerges as a potential therapeutic target for improving the regenerative capacity and maturation of the coronary vasculature.


Asunto(s)
Proteína ADAM10/fisiología , Secretasas de la Proteína Precursora del Amiloide/fisiología , Diferenciación Celular/genética , Vasos Coronarios/fisiología , Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Proteínas de la Membrana/fisiología , Animales , Vasos Coronarios/citología , Vasos Coronarios/crecimiento & desarrollo , Endotelio Vascular/crecimiento & desarrollo , Femenino , Corazón/crecimiento & desarrollo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal/genética
15.
Int J Mol Sci ; 19(12)2018 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-30567345

RESUMEN

The shortage of donors for transplantation therapy is a serious issue worldwide. Tissue engineering is considered a potential solution to this problem. Connection and perfusion in engineered tissues after transplantation is vital for the survival of the transplanted tissue, especially for tissues requiring blood perfusion to receive nutrients, such as the heart. A myocardial cell sheet containing an endothelial cell network structure was fabricated in vitro using cell sheet technology. Transplantation of the three-dimensional (3D) tissue by layering myocardial sheets could ameliorate ischemic heart disease in a rat model. The endothelial cell network in the 3D tissue was able to rapidly connect to host vasculature and begin perfusion within 24 h after transplantation. In this review, we compare and discuss the engineered tissue⁻host vasculature connection process between tissue engineered constructs with hydrogels and cell sheets by histological analysis. This review provides information that may be useful for further improvements of in vivo engineered tissue vascularization techniques.


Asunto(s)
Trasplante de Corazón/tendencias , Miocitos Cardíacos/trasplante , Neovascularización Fisiológica , Ingeniería de Tejidos , Animales , Vasos Coronarios/crecimiento & desarrollo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Humanos , Hidrogeles/uso terapéutico , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/terapia , Miocitos Cardíacos/fisiología , Ratas
16.
Dev Cell ; 47(6): 680-681, 2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30562506

RESUMEN

The leading cause of death worldwide is disease of the coronary arteries, the vessels that nourish the heart muscle. However, mechanisms that control their development and possible regeneration remain unknown. Recent work is challenging current dogma of coronary artery origins and illuminating key programs that govern coronary artery formation.


Asunto(s)
Vasos Coronarios/crecimiento & desarrollo , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiología , Animales , Diferenciación Celular/fisiología , Corazón/crecimiento & desarrollo , Humanos , Miocardio/citología , Miocardio/metabolismo , Neovascularización Fisiológica/fisiología , Organogénesis/fisiología , Venas/crecimiento & desarrollo
17.
Dev Dyn ; 247(12): 1286-1296, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30376688

RESUMEN

BACKGROUND: Proper development of the great vessels of the heart and septation of the cardiac outflow tract requires cardiac neural crest cells. These cells give rise to the parasympathetic cardiac ganglia, the smooth muscle layer of the great vessels, some cardiomyocytes, and the conotruncal cushions and aorticopulmonary septum of the outflow tract. Ablation of cardiac neural crest cells results in defective patterning of each of these structures. Previous studies have shown that targeted deletion of the forkhead transcription factor C2 (Foxc2), results in cardiac phenotypes similar to that derived from cardiac neural crest cell ablation. RESULTS: We report that Foxc2-/- embryos on the 129s6/SvEv inbred genetic background display persistent truncus arteriosus and hypoplastic ventricles before embryonic lethality. Foxc2 loss-of-function resulted in perturbed cardiac neural crest cell migration and their reduced contribution to the outflow tract as evidenced by lineage tracing analyses together with perturbed expression of the neural crest cell markers Sox10 and Crabp1. Foxc2 loss-of-function also resulted in alterations in PlexinD1, Twist1, PECAM1, and Hand1/2 expression in association with vascular and ventricular defects. CONCLUSIONS: Our data indicate Foxc2 is required for proper migration of cardiac neural crest cells, septation of the outflow tract, and development of the ventricles. Developmental Dynamics 247:1286-1296, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Embrión de Mamíferos , Factores de Transcripción Forkhead/fisiología , Cresta Neural/citología , Animales , Movimiento Celular , Vasos Coronarios/embriología , Vasos Coronarios/crecimiento & desarrollo , Corazón/inervación , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/crecimiento & desarrollo , Ratones , Miocardio/citología , Cresta Neural/embriología , Organogénesis
18.
Dev Dyn ; 247(10): 1135-1145, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30204931

RESUMEN

BACKGROUND: Proper coronary vasculature development is essential for late-embryonic and adult heart function. The developmental regulation of coronary embryogenesis is complex and includes the coordinated activity of multiple signaling pathways. CCBE1 plays an important role during lymphangiogenesis, enhancing VEGF-C signaling, which is also required for coronary vasculature formation. However, whether CCBE1 plays a similar role during coronary vasculature development is still unknown. Here, we investigate the coronary vasculature development in Ccbe1 mutant embryos. RESULTS: We show that Ccbe1 is expressed in the epicardium, like Vegf-c, and also in the sinus venosus (SV) at the stages of its contribution to coronary vasculature formation. We also report that absence of CCBE1 in cardiac tissue inhibited coronary growth that sprouts from the SV endocardium at the dorsal cardiac wall. This disruption of coronary formation correlates with abnormal processing of VEGF-C propeptides, suggesting VEGF-C-dependent signaling alteration. Moreover, Ccbe1 loss-of-function leads to the development of defective dorsal and ventral intramyocardial vessels. We also demonstrate that Ccbe1 mutants display delayed and mispatterned coronary artery (CA) stem formation. CONCLUSIONS: CCBE1 is essential for coronary vessel formation, independent of their embryonic origin, and is also necessary for peritruncal vessel growth and proper CA stem patterning. Developmental Dynamics 247:1135-1145, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Vasos Coronarios/crecimiento & desarrollo , Proteínas Supresoras de Tumor/fisiología , Animales , Proteínas de Unión al Calcio/metabolismo , Vasos Coronarios/embriología , Corazón/embriología , Corazón/crecimiento & desarrollo , Linfangiogénesis , Ratones , Pericardio/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo
19.
Dev Dyn ; 247(8): 1018-1027, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29770532

RESUMEN

BACKGROUND: Coronary artery development is an intensely studied field. Mice are a popular genetic model for developmental studies, but there is no widely accepted protocol for high-throughput, high-resolution imaging of their developmental and adult coronary artery anatomy. RESULTS: Using tissue-clearing protocols and confocal microscopy, we have analyzed embryonic and juvenile mouse hearts in Cx40:GFP knock-in models with a special focus on septal artery development. We found that the septal artery, which supplies the interventricular septum, was initially formed as an arterial plexus that connected to both the left and right coronary arteries. During development, the plexus was remodeled into a single tube, which then remained connected only to the right coronary artery. Since optical imaging became limited at postnatal stages, it was supplemented with injection techniques using India ink and Microfil; the latter was subsequently analyzed with micro-CT to visualize the anatomy of coronary vessels in 3D. CONCLUSIONS: The techniques described here enable us to study the finer details of coronary artery development in mice and can, therefore, be implemented to study the pathogenesis of coronary malformations in various mouse models. Developmental Dynamics 247:1018-1027, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Vasos Coronarios/crecimiento & desarrollo , Animales , Vasos Coronarios/anatomía & histología , Embrión de Mamíferos , Tabiques Cardíacos , Imagenología Tridimensional/métodos , Ratones , Microscopía Confocal/métodos
20.
Dev Dyn ; 247(8): 976-991, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29806219

RESUMEN

BACKGROUND: During the final stages of heart development the myocardium grows and becomes vascularized by means of paracrine factors and cell progenitors derived from the epicardium. There is evidence to suggest that retinoic acid (RA), a metabolite of vitamin A, plays an important role in epicardial-based developmental programming. However, the consequences of altered RA-signaling in coronary development have not been systematically investigated. RESULTS: We explored the developmental consequences of altered RA-signaling in late cardiogenic events that involve the epicardium. For this, we used a model of embryonic RA excess based on mouse embryos deficient in the retinaldehyde reductase DHRS3, and a complementary model of embryonic RA deficiency based on pharmacological inhibition of RA synthesis. We found that alterations in embryonic RA signaling led to a thin myocardium and aberrant coronary vessel formation and remodeling. Both excess, and deficient RA-signaling are associated with reductions in ventricular coverage and density of coronary vessels, altered vessel morphology, and impaired recruitment of epicardial-derived mural cells. Using a combined transcriptome and proteome profiling approach, we found that RA treatment of epicardial cells influenced key signaling pathways relevant for cardiac development. CONCLUSIONS: Epicardial RA-signaling plays critical roles in the development of the coronary vasculature needed to support myocardial growth. Developmental Dynamics 247:976-991, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Vasos Coronarios/crecimiento & desarrollo , Transducción de Señal/fisiología , Tretinoina/farmacología , Animales , Vasos Coronarios/embriología , Corazón/crecimiento & desarrollo , Ratones , Pericardio/citología , Proteoma , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA