Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
1.
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
2.
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
3.
Biosci Trends ; 14(2): 115-122, 2020 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-32238672

RESUMO

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.


Assuntos
Amidoidrolases/metabolismo , Vasos Coronários/crescimento & desenvolvimento , Coração/fisiologia , Neovascularização Fisiológica , Condicionamento Físico Animal/fisiologia , Amidoidrolases/genética , Animais , Arginina/análogos & derivados , Arginina/metabolismo , Comportamento Animal , Glicogênio Sintase Quinase 3 beta/metabolismo , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Óxido Nítrico/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Comportamento Sedentário , Transdução de Sinais/fisiologia , Proteínas ras/metabolismo
4.
Elife ; 82019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31789590

RESUMO

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.


Assuntos
Vasos Coronários/crescimento & desenvolvimento , Vasos Coronários/metabolismo , Efrina-B2/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Jagged-1/metabolismo , Proteínas de Membrana/metabolismo , Transdução de Sinais , Animais , Endocárdio/metabolismo , Endotélio Vascular/metabolismo , Ventrículos do Coração/crescimento & desenvolvimento , Ventrículos do Coração/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Ligantes , Camundongos , Morfogênese , Mutação/genética , Fatores de Transcrição NFATC/metabolismo , Neovascularização Fisiológica , Receptores Notch/metabolismo , Estresse Fisiológico , Transcriptoma/genética , Remodelação Vascular
5.
Biomed Res Int ; 2019: 6595437, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31317035

RESUMO

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.


Assuntos
Cardiomiopatias/tratamento farmacológico , Medicamentos de Ervas Chinesas/farmacologia , Fibrose/tratamento farmacológico , Infarto do Miocárdio/tratamento farmacológico , Animais , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Hipóxia Celular/efeitos dos fármacos , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/crescimento & desenvolvimento , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Endotélio/efeitos dos fármacos , Endotélio/metabolismo , Fibrose/metabolismo , Fibrose/patologia , Humanos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Ratos
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Sci Rep ; 6: 35366, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27752085

RESUMO

Myc is an essential regulator of cell growth and proliferation. Myc overexpression promotes the homeostatic expansion of cardiomyocyte populations by cell competition, however whether this applies to other cardiac lineages remains unknown. The epicardium contributes signals and cells to the developing and adult injured heart and exploring strategies for modulating its activity is of great interest. Using inducible genetic mosaics, we overexpressed Myc in the epicardium and determined the differential expansion of Myc-overexpressing cells with respect to their wild type counterparts. Myc-overexpressing cells overcolonized all epicardial-derived lineages and showed increased ability to invade the myocardium and populate the vasculature. We also found massive colonization of the myocardium by Wt1Cre-derived Myc-overexpressing cells, with preservation of cardiac development. Detailed analyses showed that this contribution is unlikely to derive from Cre activity in early cardiomyocytes but does not either derive from established epicardial cells, suggesting that early precursors expressing Wt1Cre originate the recombined cardiomyocytes. Myc overexpression does not modify the initial distribution of Wt1Cre-recombined cardiomyocytes, indicating that it does not stimulate the incorporation of early expressing Wt1Cre lineages to the myocardium, but differentially expands this initial population. We propose that strategies using epicardial lineages for heart repair may benefit from promoting cell competitive ability.


Assuntos
Coração/crescimento & desenvolvimento , Miocárdio/metabolismo , Organogênese/genética , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Proliferação de Células/genética , Vasos Coronários/crescimento & desenvolvimento , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Regulação da Expressão Gênica no Desenvolvimento , Integrases/genética , Camundongos , Miócitos Cardíacos/metabolismo , Pericárdio/crescimento & desenvolvimento , Pericárdio/metabolismo
12.
Nat Commun ; 7: 12088, 2016 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-27357444

RESUMO

Increasing angiogenesis has long been considered a therapeutic target for improving heart function after injury such as acute myocardial infarction. However, gene, protein and cell therapies to increase microvascularization have not been successful, most likely because the studies failed to achieve regulated and concerted expression of pro-angiogenic and angiostatic factors needed to produce functional microvasculature. Here, we report that the transcription factor RBPJ is a homoeostatic repressor of multiple pro-angiogenic and angiostatic factor genes in cardiomyocytes. RBPJ controls angiogenic factor gene expression independently of Notch by antagonizing the activity of hypoxia-inducible factors (HIFs). In contrast to previous strategies, the cardiomyocyte-specific deletion of Rbpj increased microvascularization of the heart without adversely affecting cardiac structure or function even into old age. Furthermore, the loss of RBPJ in cardiomyocytes increased hypoxia tolerance, improved heart function and decreased pathological remodelling after myocardial infarction, suggesting that inhibiting RBPJ might be therapeutic for ischaemic injury.


Assuntos
Vasos Coronários/crescimento & desenvolvimento , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/fisiologia , Miócitos Cardíacos/metabolismo , Neovascularização Fisiológica , Animais , Feminino , Regulação da Expressão Gênica , Células HEK293 , Humanos , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Camundongos , Microvasos/crescimento & desenvolvimento , Comunicação Parácrina
13.
Cardiovasc Res ; 109(2): 204-16, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26811390

RESUMO

Congenital coronary artery anomalies are of major significance in clinical cardiology and cardiac surgery due to their association with myocardial ischaemia and sudden death. Such anomalies are detectable by imaging modalities and, according to various definitions, their prevalence ranges from 0.21 to 5.79%. This consensus document from the Development, Anatomy and Pathology Working Group of the European Society of Cardiology aims to provide: (i) a definition of normality that refers to essential anatomical and embryological features of coronary vessels, based on the integrated analysis of studies of normal and abnormal coronary embryogenesis and pathophysiology; (ii) an animal model-based systematic survey of the molecular and cellular mechanisms that regulate coronary blood vessel development; (iii) an organization of the wide spectrum of coronary artery anomalies, according to a comprehensive anatomical and embryological classification scheme; (iv) current knowledge of the pathophysiological mechanisms underlying symptoms and signs of coronary artery anomalies, with diagnostic and therapeutic implications. This document identifies the mosaic-like embryonic development of the coronary vascular system, as coronary cell types differentiate from multiple cell sources through an intricate network of molecular signals and haemodynamic cues, as the necessary framework for understanding the complex spectrum of coronary artery anomalies observed in human patients.


Assuntos
Doença da Artéria Coronariana/congênito , Anomalias dos Vasos Coronários , Vasos Coronários , Coração/anatomia & histologia , Isquemia Miocárdica/complicações , Isquemia Miocárdica/patologia , Animais , Cardiologia/métodos , Doença da Artéria Coronariana/patologia , Anomalias dos Vasos Coronários/embriologia , Anomalias dos Vasos Coronários/patologia , Anomalias dos Vasos Coronários/fisiopatologia , Vasos Coronários/anatomia & histologia , Vasos Coronários/crescimento & desenvolvimento , Vasos Coronários/patologia , Coração/fisiologia , Humanos
14.
J Mol Cell Cardiol ; 87: 113-25, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26255251

RESUMO

Repairing defective cardiac cells is important towards improving heart function. Due to the frequency and severity of ischemic heart disease, management of patients featuring this type of cardiac failure receives significant interest. Previously we discovered that Thymosin ß4 (TB4), a 43 amino-acid secreted actin sequestering peptide, is beneficial for myocardial cell survival and coronary re-growth after infarction in adult mammals. Considering the regenerative potential of full-length TB4 in the heart, and that minimal structural variations alter TB4's influence on actin assembly and cell movement, we investigated how various TB4 domains affect cardiac cell behavior and post-ischemic mammalian heart function. We synthesized 17 domain combinations of full-length TB4 and analyzed their impact on embryonic cardiac cells in vitro, and after cardiac infarction in vivo. We discovered the domains of TB4 affect cardiac cell behavior distinctly. We revealed TB4 specific C-terminal tetrapeptide, AGES, increases embryonic cardiac cell migration and myocyte beating in culture, and improves adult mammalian heart function following ischemia. Investigating the molecular background and mechanism we discovered systemic injection of AGES enhances early myocyte survival by activating Akt-mediated signaling mechanisms, increases coronary vessel growth and inhibits inflammation in mice and pigs. Biodistribution analyses revealed cardiomyocytes uptake AGES efficiently in vitro and in vivo projecting a potential independent clinical utilization for the tetrapeptide. Our comprehensive domain investigations also suggest, preservation and/or restoration of cardiomyocyte communication is a target of TB4 and AGES, and critical to improve post-ischemic heart function in pigs. In summary, we identified the C-terminal four amino-acid variable end of TB4 as the essential and responsible domain for the molecule's full benefits in the hypoxic heart. Additionally, we introduced AGES as a novel, systemically applicable drug candidate to aid cardiac infarction in adult mammals.


Assuntos
Infarto do Miocárdio/genética , Isquemia Miocárdica/genética , Miócitos Cardíacos/metabolismo , Timosina/genética , Motivos de Aminoácidos/genética , Animais , Proliferação de Células/genética , Sobrevivência Celular/genética , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Infarto do Miocárdio/embriologia , Infarto do Miocárdio/patologia , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/patologia , Peptídeos/administração & dosagem , Suínos , Timosina/antagonistas & inibidores , Timosina/metabolismo
15.
J Mol Cell Cardiol ; 86: 199-207, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26241844

RESUMO

RATIONALE: During embryogenesis, hematopoietic cells appear in the myocardium prior to the initiation of coronary formation. However, their role is unknown. OBJECTIVE: Here we investigate whether pre-existing hematopoietic cells are required for the formation of coronary vasculature. METHODS AND RESULTS: As a model of for hematopoietic cell deficient animals, we used Runx1 knockout embryos and Vav1-cre; R26-DTA embryos, latter of which genetically ablates 2/3 of CD45(+) hematopoietic cells. Both Runx1 knockout embryos and Vav1-cre; R26-DTA embryos revealed disorganized, hypoplastic microvasculature of coronary vessels on section and whole-mount stainings. Furthermore, coronary explant experiments showed that the mouse heart explants from Runx1 and Vav1-cre; R26-DTA embryos exhibited impaired coronary formation ex vivo. Interestingly, in both models it appears that epicardial to mesenchymal transition is adversely affected in the absence of hematopoietic progenitors. CONCLUSION: Hematopoietic cells are not merely passively transported via coronary vessel, but substantially involved in the induction of the coronary growth. Our findings suggest a novel mechanism of coronary growth.


Assuntos
Diferenciação Celular/genética , Vasos Coronários/crescimento & desenvolvimento , Desenvolvimento Embrionário/genética , Coração/crescimento & desenvolvimento , Animais , Linhagem da Célula/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Vasos Coronários/embriologia , Vasos Coronários/metabolismo , Embrião de Mamíferos , Transição Epitelial-Mesenquimal/genética , Camundongos , Camundongos Knockout
17.
J Atheroscler Thromb ; 22(6): 570-81, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25735431

RESUMO

AIMS: High mobility group box 1 (HMGB1) is a DNA-binding protein secreted into the extracellular space from necrotic cells that acts as a cytokine. We examined the role of HMGB1 in angiogenesis from bone marrow-derived cells in the heart using transgenic mice exhibiting the cardiac-specific overexpression of HMGB1 (HMGB1-TG). METHODS: HMGB1-TG mice and wild-type littermate (WT) mice were lethally irradiated and injected with bone marrow cells from green fluorescent protein mice through the tail vein. After bone marrow transplantation, the left anterior descending artery was ligated to induce myocardial infarction (MI). RESULTS: Flow cytometry revealed that the levels of circulating endothelial progenitor cells (EPCs) mobilized from the bone marrow increased after MI in the HMGB-TG mice versus the WT mice. In addition, the size of MI was smaller in the HMGB1-TG mice than in the WT mice, and immunofluorescence staining demonstrated that the number of engrafted vascular endothelial cells derived from bone marrow in the border zones of the MI areas was increased in the HMGB1-TG mice compared to that observed in the WT mice. Moreover, the levels of cardiac vascular endothelial growth factor after MI were higher in the HMGB1-TG mice than in the WT mice. CONCLUSIONS: The present study demonstrated that HMGB1 promotes angiogenesis and reduces the MI size by enhancing the mobilization and differentiation of bone marrow cells to EPCs as well as their migration to the border zones of the MI areas and engraftment as vascular endothelial cells in new capillaries or arterioles in the infarcted heart.


Assuntos
Medula Óssea/irrigação sanguínea , Vasos Coronários/crescimento & desenvolvimento , Células Progenitoras Endoteliais/fisiologia , Proteína HMGB1/fisiologia , Coração/fisiologia , Infarto do Miocárdio/fisiopatologia , Neovascularização Fisiológica , Animais , Proliferação de Células , Células Cultivadas , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Remodelação Ventricular
18.
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
19.
PLoS One ; 9(10): e109493, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25299188

RESUMO

ATP-dependent SWI/SNF chromatin remodeling complexes utilize ATP hydrolysis to non-covalently change nucleosome-DNA interactions and are essential in stem cell development, organogenesis, and tumorigenesis. Biochemical studies show that SWI/SNF in mammalian cells can be divided into two subcomplexes BAF and PBAF based on the subunit composition. ARID2 or BAF200 has been defined as an intrinsic subunit of PBAF complex. However, the function of BAF200 in vivo is not clear. To dissect the possible role of BAF200 in regulating embryogenesis and organ development, we generated BAF200 mutant mice and found they were embryonic lethal. BAF200 mutant embryos exhibited multiple cardiac defects including thin myocardium, ventricular septum defect, common atrioventricular valve, and double outlet right ventricle around E14.5. Moreover, we also detected reduced intramyocardial coronary arteries in BAF200 mutants, suggesting that BAF200 is required for proper migration and differentiation of subepicardial venous cells into arterial endothelial cells. Our work revealed that PBAF complex plays a critical role in heart morphogenesis and coronary artery angiogenesis.


Assuntos
Diferenciação Celular/fisiologia , Proteínas Cromossômicas não Histona/metabolismo , Vasos Coronários/crescimento & desenvolvimento , Vasos Coronários/fisiologia , Desenvolvimento Embrionário/fisiologia , Coração/crescimento & desenvolvimento , Coração/fisiologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/genética , Movimento Celular/genética , Movimento Celular/fisiologia , Proteínas Cromossômicas não Histona/genética , Vasos Coronários/metabolismo , Desenvolvimento Embrionário/genética , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Miocárdio/metabolismo , Neovascularização Fisiológica/fisiologia , Organogênese/genética , Organogênese/fisiologia , Fatores de Transcrição/genética
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA