Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 268
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 176(5): 1128-1142.e18, 2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30686582

RESUMO

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.


Assuntos
Circulação Colateral/fisiologia , Coração/crescimento & desenvolvimento , Regeneração/fisiologia , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Quimiocina CXCL12/metabolismo , Vasos Coronários/crescimento & desenvolvimento , Células Endoteliais/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/fisiologia , Receptores CXCR4/metabolismo , Transdução de Sinais
2.
Development ; 149(4)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35088848

RESUMO

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.


Assuntos
Vasos Coronários/crescimento & desenvolvimento , Vasos Coronários/metabolismo , Coração/fisiologia , Organogênese/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Regeneração/fisiologia , Animais , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Miócitos de Músculo Liso/metabolismo , Pericárdio/metabolismo , Peixe-Zebra/metabolismo , Peixe-Zebra/fisiologia
3.
Genes Dev ; 31(13): 1308-1324, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28779009

RESUMO

Sufficient blood flow to tissues relies on arterial blood vessels, but the mechanisms regulating their development are poorly understood. Many arteries, including coronary arteries of the heart, form through remodeling of an immature vascular plexus in a process triggered and shaped by blood flow. However, little is known about how cues from fluid shear stress are translated into responses that pattern artery development. Here, we show that mice lacking endothelial Dach1 had small coronary arteries, decreased endothelial cell polarization, and reduced expression of the chemokine Cxcl12 Under shear stress in culture, Dach1 overexpression stimulated endothelial cell polarization and migration against flow, which was reversed upon CXCL12/CXCR4 inhibition. In vivo, DACH1 was expressed during early arteriogenesis but was down in mature arteries. Mature artery-type shear stress (high, uniform laminar) specifically down-regulated DACH1, while the remodeling artery-type flow (low, variable) maintained DACH1 expression. Together, our data support a model in which DACH1 stimulates coronary artery growth by activating Cxcl12 expression and endothelial cell migration against blood flow into developing arteries. This activity is suppressed once arteries reach a mature morphology and acquire high, laminar flow that down-regulates DACH1. Thus, we identified a mechanism by which blood flow quality balances artery growth and maturation.


Assuntos
Vasos Coronários/crescimento & desenvolvimento , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Neovascularização Fisiológica/genética , Transdução de Sinais/genética , Animais , Velocidade do Fluxo Sanguíneo/fisiologia , Movimento Celular/genética , Células Cultivadas , Quimiocina CXCL12/genética , Vasos Coronários/fisiopatologia , Células Endoteliais/citologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Técnicas de Cultura de Órgãos , Receptores CXCR4/genética , Estresse Mecânico
4.
Circ Res ; 130(3): 352-365, 2022 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-34995101

RESUMO

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.


Assuntos
Vasos Coronários/embriologia , Endocárdio/embriologia , Animais , Vasos Coronários/crescimento & desenvolvimento , Vasos Coronários/metabolismo , Endocárdio/crescimento & desenvolvimento , Endocárdio/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Organogênese
5.
Am J Physiol Lung Cell Mol Physiol ; 321(4): L675-L685, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34346780

RESUMO

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.


Assuntos
Vasos Coronários/crescimento & desenvolvimento , Ventrículos do Coração/patologia , Hipertensão Pulmonar/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Fosfofrutoquinase-2/metabolismo , Anaerobiose/fisiologia , Animais , Células Endoteliais/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica/fisiologia , Transdução de Sinais/fisiologia
6.
FASEB J ; 34(8): 10041-10055, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32573852

RESUMO

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.


Assuntos
Vasos Coronários/crescimento & desenvolvimento , Feto/fisiologia , Fator de Crescimento Insulin-Like I/farmacologia , Miócitos Cardíacos/fisiologia , Animais , Vasos Coronários/citologia , Vasos Coronários/efeitos dos fármacos , Feminino , Feto/efeitos dos fármacos , Hipóxia/fisiopatologia , Masculino , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Ovinos
7.
Annu Rev Physiol ; 79: 1-19, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-27959616

RESUMO

Coronary artery disease (CAD) is the number one cause of death worldwide and involves the accumulation of plaques within the artery wall that can occlude blood flow to the heart and cause myocardial infarction. The high mortality associated with CAD makes the development of medical interventions that repair and replace diseased arteries a high priority for the cardiovascular research community. Advancements in arterial regenerative medicine could benefit from a detailed understanding of coronary artery development during embryogenesis and of how these pathways might be reignited during disease. Recent research has advanced our knowledge on how the coronary vasculature is built and revealed unexpected features of progenitor cell deployment that may have implications for organogenesis in general. Here, we highlight these recent findings and discuss how they set the stage to interrogate developmental pathways during injury and disease.


Assuntos
Diferenciação Celular/fisiologia , Doença da Artéria Coronariana/patologia , Vasos Coronários/crescimento & desenvolvimento , Vasos Coronários/fisiologia , Organogênese/fisiologia , Células-Tronco/fisiologia , Animais , Coração/fisiologia , Humanos
8.
Int J Mol Sci ; 22(16)2021 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-34445494

RESUMO

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.


Assuntos
Vasos Coronários/crescimento & desenvolvimento , Perfilação da Expressão Gênica/métodos , Desenvolvimento Muscular , Miocárdio/citologia , Animais , Células Cultivadas , Vasos Coronários/química , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Miocárdio/química , Análise de Sequência com Séries de Oligonucleotídeos , Cultura Primária de Células , Suínos , Sequenciamento do Exoma
9.
Nature ; 514(7524): 585-90, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25317562

RESUMO

Endothelial cells contribute to a subset of cardiac fibroblasts by undergoing endothelial-to-mesenchymal transition, but whether cardiac fibroblasts can adopt an endothelial cell fate and directly contribute to neovascularization after cardiac injury is not known. Here, using genetic fate map techniques, we demonstrate that cardiac fibroblasts rapidly adopt an endothelial-cell-like phenotype after acute ischaemic cardiac injury. Fibroblast-derived endothelial cells exhibit anatomical and functional characteristics of native endothelial cells. We show that the transcription factor p53 regulates such a switch in cardiac fibroblast fate. Loss of p53 in cardiac fibroblasts severely decreases the formation of fibroblast-derived endothelial cells, reduces post-infarct vascular density and worsens cardiac function. Conversely, stimulation of the p53 pathway in cardiac fibroblasts augments mesenchymal-to-endothelial transition, enhances vascularity and improves cardiac function. These observations demonstrate that mesenchymal-to-endothelial transition contributes to neovascularization of the injured heart and represents a potential therapeutic target for enhancing cardiac repair.


Assuntos
Transdiferenciação Celular , Vasos Coronários/citologia , Vasos Coronários/crescimento & desenvolvimento , Células Endoteliais/citologia , Mesoderma/citologia , Isquemia Miocárdica/patologia , Neovascularização Fisiológica , Animais , Feminino , Fibroblastos/citologia , Técnicas In Vitro , Masculino , Camundongos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
10.
PLoS Genet ; 13(10): e1007068, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29084269

RESUMO

The coronary vasculature is an essential vessel network providing the blood supply to the heart. Disruptions in coronary blood flow contribute to cardiac disease, a major cause of premature death worldwide. The generation of treatments for cardiovascular disease will be aided by a deeper understanding of the developmental processes that underpin coronary vessel formation. From an ENU mutagenesis screen, we have isolated a mouse mutant displaying embryonic hydrocephalus and cardiac defects (EHC). Positional cloning and candidate gene analysis revealed that the EHC phenotype results from a point mutation in a splice donor site of the Myh10 gene, which encodes NMHC IIB. Complementation testing confirmed that the Myh10 mutation causes the EHC phenotype. Characterisation of the EHC cardiac defects revealed abnormalities in myocardial development, consistent with observations from previously generated NMHC IIB null mouse lines. Analysis of the EHC mutant hearts also identified defects in the formation of the coronary vasculature. We attribute the coronary vessel abnormalities to defective epicardial cell function, as the EHC epicardium displays an abnormal cell morphology, reduced capacity to undergo epithelial-mesenchymal transition (EMT), and impaired migration of epicardial-derived cells (EPDCs) into the myocardium. Our studies on the EHC mutant demonstrate a requirement for NMHC IIB in epicardial function and coronary vessel formation, highlighting the importance of this protein in cardiac development and ultimately, embryonic survival.


Assuntos
Vasos Coronários/crescimento & desenvolvimento , Desenvolvimento Embrionário/genética , Cadeias Pesadas de Miosina/genética , Miosina não Muscular Tipo IIB/genética , Pericárdio/crescimento & desenvolvimento , Animais , Diferenciação Celular/genética , Vasos Coronários/metabolismo , Embrião de Mamíferos , Transição Epitelial-Mesenquimal/genética , Humanos , Hidrocefalia/genética , Hidrocefalia/metabolismo , Hidrocefalia/patologia , Camundongos , Camundongos Knockout , Mutação , Miocárdio/metabolismo , Pericárdio/metabolismo
11.
Angiogenesis ; 22(2): 237-250, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30446855

RESUMO

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.


Assuntos
Proteína ADAM10/fisiologia , Secretases da Proteína Precursora do Amiloide/fisiologia , Diferenciação Celular/genética , Vasos Coronários/fisiologia , Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Proteínas de Membrana/fisiologia , Animais , Vasos Coronários/citologia , Vasos Coronários/crescimento & desenvolvimento , Endotélio Vascular/crescimento & desenvolvimento , Feminino , Coração/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais/genética
12.
Circ J ; 83(3): 556-566, 2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30700665

RESUMO

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.


Assuntos
Implantes Absorvíveis/normas , Vasos Coronários/diagnóstico por imagem , Stents/normas , Tomografia de Coerência Óptica/métodos , Animais , Angiografia Coronária/métodos , Vasos Coronários/crescimento & desenvolvimento , Vasos Coronários/cirurgia , Modelos Animais , Desenho de Prótese/normas , Suínos , Fatores de Tempo
13.
Dev Dyn ; 247(8): 1018-1027, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29770532

RESUMO

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.


Assuntos
Vasos Coronários/crescimento & desenvolvimento , Animais , Vasos Coronários/anatomia & histologia , Embrião de Mamíferos , Septos Cardíacos , Imageamento Tridimensional/métodos , Camundongos , Microscopia Confocal/métodos
14.
Dev Dyn ; 247(8): 976-991, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29806219

RESUMO

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.


Assuntos
Vasos Coronários/crescimento & desenvolvimento , Transdução de Sinais/fisiologia , Tretinoína/farmacologia , Animais , Vasos Coronários/embriologia , Coração/crescimento & desenvolvimento , Camundongos , Pericárdio/citologia , Proteoma , Transcriptoma
15.
Dev Dyn ; 247(12): 1286-1296, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30376688

RESUMO

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.


Assuntos
Embrião de Mamíferos , Fatores de Transcrição Forkhead/fisiologia , Crista Neural/citologia , Animais , Movimento Celular , Vasos Coronários/embriologia , Vasos Coronários/crescimento & desenvolvimento , Coração/inervação , Ventrículos do Coração/embriologia , Ventrículos do Coração/crescimento & desenvolvimento , Camundongos , Miocárdio/citologia , Crista Neural/embriologia , Organogênese
16.
Dev Dyn ; 247(10): 1135-1145, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30204931

RESUMO

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.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Vasos Coronários/crescimento & desenvolvimento , Proteínas Supressoras de Tumor/fisiologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Vasos Coronários/embriologia , Coração/embriologia , Coração/crescimento & desenvolvimento , Linfangiogênese , Camundongos , Pericárdio/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo
17.
J Mol Cell Cardiol ; 119: 51-63, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29680681

RESUMO

AIMS: Following an acute myocardial infarction (MI) the extracellular matrix (ECM) undergoes remodeling in order to prevent dilation of the infarct area and maintain cardiac output. Excessive and prolonged inflammation following an MI exacerbates adverse ventricular remodeling. Macrophages are an integral part of the inflammatory response that contribute to this remodeling. Treatment with histone deacetylase (HDAC) inhibitors preserves LV function and myocardial remodeling in the post-MI heart. This study tested whether inhibition of HDAC activity resulted in preserving post-MI LV function through the regulation of macrophage phenotype and early resolution of inflammation. METHODS AND RESULTS: HDAC inhibition does not affect the recruitment of CD45+ leukocytes, CD45+/CD11b+ inflammatory monocytes or CD45+/CD11b+CD86+ inflammatory macrophages for the first 3 days following infarct. Further, HDAC inhibition does not change the high expression level of the inflammatory cytokines in the first days following MI. However, by day 7, there was a significant reduction in the levels of CD45+/Cd11b+ and CD45+/CD11b+/CD86+ cells with HDAC inhibition. Remarkably, HDAC inhibition resulted in the dramatic increase in the recruitment of CD45+/CD11b+/CD206+ alternatively activated macrophages as early as 1 day which remained significantly elevated until 5 days post-MI. qRT-PCR revealed that HDAC inhibitor treatment shifts the cytokine and chemokine environment towards an M2 phenotype with upregulation of M2 markers at 1 and 5 days post-MI. Importantly, HDAC inhibition correlates with significant preservation of both LV ejection fraction and end-diastolic volume and is associated with a significant increase in micro-vessel density in the border zone at 14 days post-MI. CONCLUSION: Inhibition of HDAC activity result in the early recruitment of reparative CD45+/CD11b+/CD206+ macrophages in the post-MI heart and correlates with improved ventricular function and remodeling. This work identifies a very promising therapeutic opportunity to manage macrophage phenotype and enhance resolution of inflammation in the post-MI heart.


Assuntos
Histona Desacetilase 1/genética , Inibidores de Histona Desacetilases/administração & dosagem , Inflamação/tratamento farmacológico , Infarto do Miocárdio/tratamento farmacológico , Cicatrização/genética , Animais , Antígeno B7-2/metabolismo , Antígeno CD11b/metabolismo , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Coração/crescimento & desenvolvimento , Coração/fisiopatologia , Histona Desacetilase 1/antagonistas & inibidores , Humanos , Inflamação/genética , Inflamação/fisiopatologia , Antígenos Comuns de Leucócito/metabolismo , Leucócitos/metabolismo , Macrófagos/metabolismo , Camundongos , Monócitos/efeitos dos fármacos , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Neovascularização Fisiológica/genética , Remodelação Ventricular/efeitos dos fármacos , Remodelação Ventricular/genética , Cicatrização/efeitos dos fármacos
18.
Biochem Biophys Res Commun ; 495(1): 884-891, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29158084

RESUMO

The coronary vascular system is critical for myocardial growth and cardiomyocyte survival. However, the molecular mechanism regulating coronary angiogenesis remains elusive. Vascular endothelial growth factor (VEGF) regulates angiogenesis by binding to the specific receptors Flk1 and Flt1, which results in different functions. Despite the importance of Flk1 and Flt1, their expression in the coronary vasculature remains largely unknown due to the lack of appropriate antibodies for immunostaining. Here, we analyzed multiple reporter mice including Flk1-GFP BAC transgenic (Tg), Flk1-LacZ knock-in, Flt1-DsRed BAC Tg, and Flk1-GFP/Flt1-DsRed double Tg animals to determine expression patterns in mouse hearts during cardiac growth and after myocardial infarction (MI). We found that Flk1 was expressed in endothelial cells (ECs) with a pattern of epicardial-to-endocardial transmural gradients in the neonatal mouse ventricle, which was downregulated in adult coronary vessels with development. In contrast, Flt1 was homogeneously expressed in the ECs of neonatal mouse hearts and expression was maintained until adulthood. After MI, expression of both Flk1 and Flt1 was induced in the regenerating coronary vessels at day 7. Intriguingly, Flk1 expression was downregulated thereafter, whereas Flt1 expression was maintained in the newly formed coronary vessels until 30 days post-MI, recapitulating their expression kinetics during development. This is the first report demonstrating the spatiotemporal expression patterns of Flk1 and Flt1 in the coronary vascular system during development and after MI; thus, this study suggests that these factors have distinct and important functions in coronary angiogenesis.


Assuntos
Envelhecimento/metabolismo , Vasos Coronários/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Vasos Coronários/crescimento & desenvolvimento , Progressão da Doença , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Neovascularização Fisiológica/fisiologia
19.
Development ; 142(1): 21-30, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25516967

RESUMO

An important pool of cardiovascular progenitor cells arises from the epicardium, a single layer of mesothelium lining the heart. Epicardium-derived progenitor cell (EPDC) formation requires epithelial-to-mesenchymal transition (EMT) and the subsequent migration of these cells into the sub-epicardial space. Although some of the physiological signals that promote EMT are understood, the functional mediators of EPDC motility and differentiation are not known. Here, we identify a novel regulatory mechanism of EPDC mobilization. Myocardin-related transcription factor (MRTF)-A and MRTF-B (MKL1 and MKL2, respectively) are enriched in the perinuclear space of epicardial cells during development. Transforming growth factor (TGF)-ß signaling and disassembly of cell contacts leads to nuclear accumulation of MRTFs and the activation of the motile gene expression program. Conditional ablation of Mrtfa and Mrtfb specifically in the epicardium disrupts cell migration and leads to sub-epicardial hemorrhage, partially stemming from the depletion of coronary pericytes. Using lineage-tracing analyses, we demonstrate that sub-epicardial pericytes arise from EPDCs in a process that requires the MRTF-dependent motile gene expression program. These findings provide novel mechanisms linking EPDC motility and differentiation, shed light on the transcriptional control of coronary microvascular maturation and suggest novel therapeutic strategies to manipulate epicardium-derived progenitor cells for cardiac repair.


Assuntos
Movimento Celular , Vasos Coronários/crescimento & desenvolvimento , Pericárdio/citologia , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células COS , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Chlorocebus aethiops , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/metabolismo , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Pericárdio/metabolismo , Pericárdio/ultraestrutura , Pericitos/citologia , Pericitos/efeitos dos fármacos , Fator de Resposta Sérica/metabolismo , Transativadores/genética , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta1/farmacologia
20.
Circ Res ; 118(10): 1577-92, 2016 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-27174951

RESUMO

Aging-related cardiovascular diseases are a rapidly increasing problem worldwide. Cardiac aging demonstrates progressive decline of diastolic dysfunction of ventricle and increase in ventricular and arterial stiffness accompanied by increased fibrosis stimulated by angiotensin II and proinflammatory cytokines. Reactive oxygen species and multiple signaling pathways on cellular senescence play major roles in the process. Aging is also associated with an alteration in steady state of macromolecular dynamics including a dysfunction of protein synthesis and degradation. Currently, impaired macromolecular degradation is considered to be closely related to enhanced inflammation and be involved in the process and mechanism of cardiac aging. Herein, we review the role and mechanisms of the degradation system of intracellular macromolecules in the process and pathophysiology of cardiovascular aging.


Assuntos
Envelhecimento/metabolismo , Miocárdio/metabolismo , Proteólise , Envelhecimento/patologia , Animais , Autofagia , Vasos Coronários/crescimento & desenvolvimento , Vasos Coronários/metabolismo , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA