Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.305
Filtrar
1.
Dev Growth Differ ; 66(6): 357-368, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39072708

RESUMEN

In recent decades, developmental biologists have come to view vascular development as a series of progressive transitions. Mesoderm differentiates into endothelial cells; arteries, veins and lymphatic endothelial cells are specified from early endothelial cells; and vascular networks diversify and invade developing tissues and organs. Our understanding of this elaborate developmental process has benefitted from detailed studies using the zebrafish as a model system. Here, we review a number of key developmental transitions that occur in zebrafish during the formation of the blood and lymphatic vessel networks.


Asunto(s)
Células Endoteliales , Pez Cebra , Animales , Pez Cebra/embriología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Vasos Linfáticos/embriología , Vasos Linfáticos/metabolismo , Vasos Linfáticos/citología , Vasos Sanguíneos/citología , Vasos Sanguíneos/embriología , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/crecimiento & desarrollo , Neovascularización Fisiológica/fisiología , Diferenciación Celular
2.
Dev Biol ; 512: 26-34, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38705558

RESUMEN

The signals that regulate peripheral blood vessel formation during development are still under investigation. The hormone leptin promotes blood vessel formation, adipose tissue establishment and expansion, tumor growth, and wound healing, but the underlying mechanisms for these actions are currently unknown. We investigated whether leptin promotes angiogenesis in the developing tail fin using embryonic transgenic xflk-1:GFP Xenopus laevis, which express a green fluorescent protein on vascular endothelial cells to mark blood vessels. We found that leptin protein is expressed in endothelial cells of developing blood vessels and that leptin treatment via injection increased phosphorylated STAT3 signaling, which is indicative of leptin activation of its receptor, in blood vessels of the larval tail fin. Leptin administration via media increased vessel length, branching, and reconnection with the cardinal vein, while decreased leptin signaling via immunoneutralization had an opposing effect on vessel development. We also observed disorganization of major vessels and microvessels of the tail fin and muscle when leptin signaling was decreased. Reduced leptin signaling lowered mRNA expression of cenpk, gpx1, and mmp9, markers for cell proliferation, antioxidation, and extracellular matrix remodeling/cell migration, respectively, in the developing tail, providing insight into three possible mechanisms underlying leptin's promotion of angiogenesis. Together these results illustrate that leptin levels are correlated with embryonic angiogenesis and that leptin coordinates multiple aspects of blood vessel growth and development, showing that leptin is an important morphogen during embryonic development.


Asunto(s)
Larva , Leptina , Neovascularización Fisiológica , Transducción de Señal , Cola (estructura animal) , Xenopus laevis , Animales , Leptina/metabolismo , Cola (estructura animal)/irrigación sanguínea , Cola (estructura animal)/embriología , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Larva/metabolismo , Vasos Sanguíneos/embriología , Vasos Sanguíneos/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas de Xenopus/genética , Animales Modificados Genéticamente , Factor de Transcripción STAT3/metabolismo , Embrión no Mamífero/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Regulación del Desarrollo de la Expresión Génica
3.
Curr Top Dev Biol ; 159: 344-370, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38729681

RESUMEN

The development of the vascular system is crucial in supporting the growth and health of all other organs in the body, and vascular system dysfunction is the major cause of human morbidity and mortality. This chapter discusses three successive processes that govern vascular system development, starting with the differentiation of the primitive vascular system in early embryonic development, followed by its remodeling into a functional circulatory system composed of arteries and veins, and its final maturation and acquisition of an organ specific semi-permeable barrier that controls nutrient uptake into tissues and hence controls organ physiology. Along these steps, endothelial cells forming the inner lining of all blood vessels acquire extensive heterogeneity in terms of gene expression patterns and function, that we are only beginning to understand. These advances contribute to overall knowledge of vascular biology and are predicted to unlock the unprecedented therapeutic potential of the endothelium as an avenue for treatment of diseases associated with dysfunctional vasculature.


Asunto(s)
Remodelación Vascular , Humanos , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/embriología , Neovascularización Fisiológica , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Diferenciación Celular , Desarrollo Embrionario , Endotelio Vascular/citología
4.
Biol Open ; 13(5)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38639409

RESUMEN

Blood vessels serve as intermediate conduits for the extension of sympathetic axons towards target tissues, while also acting as crucial targets for their homeostatic processes encompassing the regulation of temperature, blood pressure, and oxygen availability. How sympathetic axons innervate not only blood vessels but also a wide array of target tissues is not clear. Here we show that in embryonic skin, after the establishment of co-branching between sensory nerves and blood vessels, sympathetic axons invade the skin alongside these sensory nerves and extend their branches towards these blood vessels covered by vascular smooth muscle cells (VSMCs). Our mosaic labeling technique for sympathetic axons shows that collateral branching predominantly mediates the innervation of VSMC-covered blood vessels by sympathetic axons. The expression of nerve growth factor (NGF), previously known to induce collateral axon branching in culture, can be detected in the vascular smooth muscle cell (VSMC)-covered blood vessels, as well as sensory nerves. Indeed, VSMC-specific Ngf knockout leads to a significant decrease of collateral branching of sympathetic axons innervating VSMC-covered blood vessels. These data suggest that VSMC-derived NGF serves as an inductive signal for collateral branching of sympathetic axons innervating blood vessels in the embryonic skin.


Asunto(s)
Músculo Liso Vascular , Factor de Crecimiento Nervioso , Piel , Animales , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/inervación , Factor de Crecimiento Nervioso/metabolismo , Ratones , Piel/inervación , Piel/irrigación sanguínea , Piel/metabolismo , Miocitos del Músculo Liso/metabolismo , Axones/metabolismo , Axones/fisiología , Vasos Sanguíneos/embriología , Vasos Sanguíneos/inervación , Vasos Sanguíneos/metabolismo , Sistema Nervioso Simpático/embriología , Sistema Nervioso Simpático/fisiología , Sistema Nervioso Simpático/metabolismo , Ratones Noqueados
5.
Dev Dyn ; 253(5): 519-541, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38112237

RESUMEN

BACKGROUND: Mural cells are an essential perivascular cell population that associate with blood vessels and contribute to vascular stabilization and tone. In the embryonic zebrafish vasculature, pdgfrb and tagln are commonly used as markers for identifying pericytes and vascular smooth muscle cells. However, the overlapping and distinct expression patterns of these markers in tandem have not been fully described. RESULTS: Here, we used the Tg(pdgfrb:Gal4FF; UAS:RFP) and Tg(tagln:NLS-EGFP) transgenic lines to identify single- and double-positive perivascular cell populations on the cranial, axial, and intersegmental vessels between 1 and 5 days postfertilization. From this comparative analysis, we discovered two novel regions of tagln-positive cell populations that have the potential to function as mural cell precursors. Specifically, we found that the hypochord-a reportedly transient structure-contributes to tagln-positive cells along the dorsal aorta. We also identified a unique mural cell progenitor population that resides along the midline between the neural tube and notochord and contributes to intersegmental vessel mural cell coverage. CONCLUSION: Together, our findings highlight the variability and versatility of tracking both pdgfrb and tagln expression in mural cells of the developing zebrafish embryo and reveal unexpected embryonic cell populations that express pdgfrb and tagln.


Asunto(s)
Animales Modificados Genéticamente , Pericitos , Proteínas de Pez Cebra , Pez Cebra , Animales , Vasos Sanguíneos/embriología , Vasos Sanguíneos/citología , Vasos Sanguíneos/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/fisiología , Pericitos/citología , Pericitos/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Pez Cebra/embriología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
6.
Circ Res ; 130(3): 366-383, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-34986653

RESUMEN

BACKGROUND: The chromatin-remodeling enzyme BRG1 (brahma-related gene 1) regulates gene expression in a variety of rapidly differentiating cells during embryonic development. However, the critical genes that BRG1 regulates during lymphatic vascular development are unknown. METHODS: We used genetic and imaging techniques to define the role of BRG1 in murine embryonic lymphatic development, although this approach inadvertently expanded our study to multiple interacting cell types. RESULTS: We found that omental macrophages fine-tune an unexpected developmental process by which erythrocytes escaping from naturally discontinuous omental blood vessels are collected by nearby lymphatic vessels. Our data indicate that circulating fibrin(ogen) leaking from gaps in omental blood vessels can trigger inflammasome-mediated IL-1ß (interleukin-1ß) production and secretion from nearby macrophages. IL-1ß destabilizes adherens junctions in omental blood and lymphatic vessels, contributing to both extravasation of erythrocytes and their uptake by lymphatics. BRG1 regulates IL-1ß production in omental macrophages by transcriptionally suppressing the inflammasome trigger RIPK3 (receptor interacting protein kinase 3). CONCLUSIONS: Genetic deletion of Brg1 in embryonic macrophages leads to excessive IL-1ß production, erythrocyte leakage from blood vessels, and blood-filled lymphatics in the developing omentum. Altogether, these results highlight a novel context for epigenetically regulated crosstalk between macrophages, blood vessels, and lymphatics.


Asunto(s)
Vasos Sanguíneos/metabolismo , ADN Helicasas/metabolismo , Interleucina-1beta/metabolismo , Vasos Linfáticos/metabolismo , Proteínas Nucleares/metabolismo , Epiplón/metabolismo , Factores de Transcripción/metabolismo , Uniones Adherentes/metabolismo , Animales , Vasos Sanguíneos/embriología , ADN Helicasas/genética , Eritrocitos/metabolismo , Inflamasomas/metabolismo , Vasos Linfáticos/embriología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Epiplón/irrigación sanguínea , Epiplón/embriología , Factores de Transcripción/genética
7.
Dev Biol ; 477: 70-84, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34015362

RESUMEN

The C-X-C chemokine receptor CXCR4 and its ligand CXCL12 play an important role in organ-specific vascular branching morphogenesis. CXCR4 is preferentially expressed by arterial endothelial cells, and local secretion of CXCL12 determines the organotypic pattern of CXCR4+ arterial branching. Previous loss-of-function studies clearly demonstrated that CXCL12-CXCR4 signaling is necessary for proper arterial branching in the developing organs such as the skin and heart. To further understand the role of CXCL12-CXCR4 signaling in organ-specific vascular development, we generated a mouse model carrying the Cre recombinase-inducible Cxcr4 transgene. Endothelial cell-specific Cxcr4 gain-of-function embryos exhibited defective vascular remodeling and formation of a hierarchical vascular branching network in the developing skin and heart. Ectopic expression of CXCR4 in venous endothelial cells, but not in lymphatic endothelial cells, caused blood-filled, enlarged lymphatic vascular phenotypes, accompanied by edema. These data suggest that CXCR4 expression is tightly regulated in endothelial cells for appropriate vascular development in an organ-specific manner.


Asunto(s)
Vasos Sanguíneos/embriología , Células Endoteliales/fisiología , Neovascularización Fisiológica/fisiología , Receptores CXCR4/fisiología , Animales , Vasos Sanguíneos/anatomía & histología , Células Endoteliales/metabolismo , Mutación con Ganancia de Función , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Receptores CXCR4/biosíntesis , Remodelación Vascular/fisiología
8.
Biochem Biophys Res Commun ; 557: 8-13, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33857842

RESUMEN

Precise quantification of vascular developments in Zebrafish requires continuous in-vivo 3D imaging. Here we employed a bi-directional light-sheet illumination microscope to characterize the development process of Zebrafish's intersegmental vessels. A Virtual Reality-based method was used to measure the lengths of intersegmental vessels (ISVs). The quantified growth rates of typical ISVs can be plotted, and unusual growth of some specific vessels was also observed.


Asunto(s)
Vasos Sanguíneos/embriología , Embrión no Mamífero/irrigación sanguínea , Microscopía/instrumentación , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Vasos Sanguíneos/crecimiento & desarrollo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Imagenología Tridimensional/métodos , Iluminación , Microscopía/métodos , Neovascularización Fisiológica , Pez Cebra/crecimiento & desarrollo
9.
Elife ; 102021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33929320

RESUMEN

Ras-responsive element-binding protein 1 (Rreb1) is a zinc-finger transcription factor acting downstream of RAS signaling. Rreb1 has been implicated in cancer and Noonan-like RASopathies. However, little is known about its role in mammalian non-disease states. Here, we show that Rreb1 is essential for mouse embryonic development. Loss of Rreb1 led to a reduction in the expression of vasculogenic factors, cardiovascular defects, and embryonic lethality. During gastrulation, the absence of Rreb1 also resulted in the upregulation of cytoskeleton-associated genes, a change in the organization of F-ACTIN and adherens junctions within the pluripotent epiblast, and perturbed epithelial architecture. Moreover, Rreb1 mutant cells ectopically exited the epiblast epithelium through the underlying basement membrane, paralleling cell behaviors observed during metastasis. Thus, disentangling the function of Rreb1 in development should shed light on its role in cancer and other diseases involving loss of epithelial integrity.


Asunto(s)
Vasos Sanguíneos/embriología , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/metabolismo , Ratones/embriología , Neovascularización Fisiológica , Factores de Transcripción/metabolismo , Actinas/genética , Actinas/metabolismo , Uniones Adherentes/genética , Uniones Adherentes/metabolismo , Animales , Vasos Sanguíneos/metabolismo , Proteínas de Unión al ADN/genética , Desarrollo Embrionario , Ratones/genética , Ratones/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción/genética
10.
Dev Dyn ; 250(8): 1173-1190, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33587326

RESUMEN

INTRODUCTION: The Drosophila dorsal vessel (DV) is comprised of two opposing rows of cardioblasts (CBs) that migrate toward the dorsal midline during development. While approaching the midline, CBs change shape, enabling dorsal and ventral attachments with their contralateral partners to create a linear tube with a central lumen. We previously demonstrated DV closure occurs via a "buttoning" mechanism where specific CBs advance ahead of their lateral neighbors, and attach creating transient holes, which eventually seal. RESULTS: Here, we investigate the role of the actin-regulatory protein enabled (Ena) in DV closure. Loss of Ena results in DV cell shape and alignment defects. Live analysis of DV formation in ena mutants shows a reduction in CB leading edge protrusion length and gaps in the DV between contralateral CB pairs. These gaps occur primarily between a specific genetic subtype of CBs, which express the transcription factor seven-up (Svp) and form the ostia inflow tracts of the heart. In WT embryos these gaps between Svp+ CBs are observed transiently during the final stages of DV closure. CONCLUSIONS: Our data suggest that Ena modulates the actin cytoskeleton in order to facilitate the complete sealing of the DV during the final stages of cardiac tube formation.


Asunto(s)
Vasos Sanguíneos/embriología , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Morfogénesis/fisiología , Animales , Vasos Sanguíneos/metabolismo , Movimiento Celular/fisiología , Proteínas de Unión al ADN/genética , Drosophila melanogaster
11.
Mol Neurobiol ; 58(4): 1755-1768, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33387302

RESUMEN

Ethanol consumption during pregnancy or lactation period can induce permanent damage to the development of the central nervous system (CNS), resulting in fetal alcohol spectrum disorders (FASD). CNS development depends on proper neural cells and blood vessel (BV) development and blood-brain barrier (BBB) establishment; however, little is known about how ethanol affects these events. Here, we investigated the impact of ethanol exposure to endothelial cells (ECs) function and to ECs interaction with astrocytes in the context of BBB establishment. Cerebral cortex of newborn mice exposed in utero to ethanol (FASD model) presented a hypervascularized phenotype, revealed by augmented vessel density, length, and branch points. Further, aberrant distribution of the tight junction ZO-1 protein along BVs and increased rates of perivascular astrocytic endfeet around BVs were observed. In vitro exposure of human brain microcapillary ECs (HBMEC) to ethanol significantly disrupted ZO-1 distribution, decreased Claudin-5 and GLUT-1 expression and impaired glucose uptake, and increased nitric oxide secretion. These events were accompanied by upregulation of angiogenesis-related secreted proteins by ECs in response to ethanol exposure. Treatment of cortical astrocytes with conditioned medium (CM) from ethanol exposed ECs, upregulated astrocyte's expression of GFAP, Cx43, and Lipocalin-2 genes, as well as the pro-inflammatory genes, IL-1beta, IL-6, and TNF-alpha, which was accompanied by NF-kappa B protein nuclear accumulation. Our findings suggest that ethanol triggers a dysfunctional phenotype in brain ECs, leading to impairment of cortical vascular network formation, and promotes ECs-induced astrocyte dysfunction, which could dramatically affect BBB establishment in the developing brain.


Asunto(s)
Astrocitos/patología , Vasos Sanguíneos/embriología , Barrera Hematoencefálica/patología , Corteza Cerebral/embriología , Células Endoteliales/patología , Etanol/efectos adversos , Efectos Tardíos de la Exposición Prenatal/patología , Animales , Animales Recién Nacidos , Células Endoteliales/metabolismo , Femenino , Trastornos del Espectro Alcohólico Fetal/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Ratones , Neovascularización Fisiológica , Fenotipo , Embarazo
12.
Cell Mol Life Sci ; 78(5): 2199-2212, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32910224

RESUMEN

It is critical to specify a signal that directly drives the transition that occurs between cell states. However, such inferences are often confounded by indirect intercellular communications or secondary transcriptomic changes due to primary transcription factors. Although FGF is known for its importance during mesoderm-to-endothelium differentiation, its specific role and signaling mechanisms are still unclear due to the confounding factors referenced above. Here, we attempted to minimize the secondary artifacts by manipulating FGF and its downstream mediators with a short incubation time before sampling and protein-synthesis blockage in a low-density angioblastic/endothelial differentiation system. In less than 8 h, FGF started the conversion of KDRlow/PDGFRAlow nascent mesoderm into KDRhigh/PDGFRAlow angioblasts, and the priming by FGF was necessary to endow endothelial formation 72 h later. Further, the angioblastic conversion was mediated by the FGFR1/BRAF/MEK/ERK pathway in mesodermal cells. Finally, two transcription factors, ETV2 and LMO2, were the early direct functional responders downstream of the FGF pathway, and ETV2 alone was enough to complement the absence of FGF. FGF's selective role in mediating the first-step, angioblastic conversion from mesoderm-to-endothelium thus allows for refined control over acquiring and manipulating angioblasts. The noise-minimized differentiation/analysis platform presented here is well-suited for studies on the signaling switches of other mesodermal-lineage fates as well.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Vasos Sanguíneos/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Proteínas con Dominio LIM/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Factores de Transcripción/metabolismo , Vasos Sanguíneos/citología , Vasos Sanguíneos/embriología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Mesodermo/citología , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología
13.
Blood ; 137(2): 258-268, 2021 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-32735640

RESUMEN

Tissue factor pathway inhibitor (TFPI) inhibits proteases in the blood coagulation cascade that lead to the production of thrombin, including prothrombinase (factor Xa [FXa]/FVa), the catalytic complex that directly generates thrombin. Thus, TFPI and FV are directly linked in regulating the procoagulant response. Studies using knockout mice indicate that TFPI and FV are necessary for embryogenesis, but their contributions to vascular development are unclear. We performed extensive histological analyses of Tfpi-/- and Tfpi-/-F5-/- mouse embryos to investigate the importance of the interplay between TFPI and FV in regulating hemostasis and vascular development during embryogenesis. We observed normal tissue development throughout Tfpi-/- embryos, except in the central nervous system (CNS). The CNS displayed stunted brain growth, delayed development of the meninges, and severe vascular pathology characterized by the formation of glomeruloid bodies surrounding areas of cellular death, fibrin deposition, and hemorrhage. Removing FV from Tfpi-/- embryos completely ameliorated their brain pathology, suggesting that TFPI dampens FV-dependent procoagulant activity in a manner that modulates cerebrovascular development. Thus, we have identified a previously unrecognized role for TFPI activity within the CNS. This TFPI activity likely diminishes an effect of excess thrombin activity on signaling pathways that control cerebral vascular development.


Asunto(s)
Vasos Sanguíneos/embriología , Encéfalo/irrigación sanguínea , Encéfalo/embriología , Desarrollo Embrionario/fisiología , Lipoproteínas/metabolismo , Animales , Factor V/metabolismo , Ratones , Ratones Noqueados
14.
Biochem Biophys Res Commun ; 529(3): 596-602, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32736679

RESUMEN

Glycine, a non-essential amino acid, exerts concentration-dependent biphasic effects on angiogenesis. Low-doses of glycine promote angiogenesis, whereas high-doses cause anti-angiogenesis. The phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling participates in angiogenesis of both physiological development, and pathological events including tumor and inflammation. We assessed the role of PI3K/Akt/mTOR signaling in vascular development, and the interaction with glycine, using transgenic zebrafish Tg(fli1a:Myr-mCherry)ncv1 embryos expressing fluorescent proteins in vascular endothelial cells. Treatment with inhibitors of mTORC1 (rapamycin and everolimus), mTORC1/mTORC2 (KU0063794), PI3K (LY29400), and Akt (Akt inhibitor) decreased the development of intersegmental vessels (ISVs). These inhibitors cancelled the angiogenic effects of a low-dose of glycine, while acted synergistically with a high-dose of glycine in anti-angiogenesis. mTOR signaling regulates the gene expression of vascular endothelial growth factor (VEGF), a major angiogenic factor, and nitric oxide (NO) synthase (NOS), an enzyme for the synthesis of an angiogenic mediator NO. Expressions of VEGF and NOS were consistent with the vascular features induced by glycine and an mTOR inhibitor. Our results suggest that PI3K/Akt/mTOR signaling may interact with dose-dependent biphasic effects of exogenous glycine on in vivo angiogenesis. mTOR signaling is a key target for cancer therapy, thus, the combining mTOR inhibitors with glycine may be a potential approach for controlling angiogenesis.


Asunto(s)
Vasos Sanguíneos/efectos de los fármacos , Glicina/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Vasos Sanguíneos/embriología , Cromonas/farmacología , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/embriología , Everolimus/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Morfolinas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Pirimidinas/farmacología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra/embriología
15.
PLoS One ; 15(7): e0235799, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32658897

RESUMEN

ATP-dependent chromatin-remodeling complexes epigenetically modulate transcription of target genes to impact a variety of developmental processes. Our lab previously demonstrated that CHD4-a central ATPase and catalytic enzyme of the NuRD chromatin-remodeling complex-plays an important role in murine embryonic endothelial cells by transcriptionally regulating vascular integrity at midgestation. Since NuRD complexes can incorporate the ATPase CHD3 as an alternative to CHD4, we questioned whether the CHD3 enzyme likewise modulates vascular development or integrity. We generated a floxed allele of Chd3 but saw no evidence of lethality or vascular anomalies when we deleted it in embryonic endothelial cells in vivo (Chd3ECKO). Furthermore, double-deletion of Chd3 and Chd4 in embryonic endothelial cells (Chd3/4ECKO) did not dramatically alter the timing and severity of embryonic phenotypes seen in Chd4ECKO mutants, indicating that CHD3 does not play a cooperative role with CHD4 in early vascular development. However, excision of Chd3 at the epiblast stage of development with a Sox2-Cre line allowed us to generate global heterozygous Chd3 mice (Chd3Δ/+), which were subsequently intercrossed and revealed partial lethality of Chd3Δ/Δ mutants prior to weaning. Tissues from surviving Chd3Δ/Δ mutants helped us confirm that CHD3 was efficiently deleted in these animals and that CHD3 is highly expressed in the gonads and brains of adult wildtype mice. Therefore, Chd3-flox mice will be beneficial for future studies about roles for this chromatin-remodeling enzyme in viable embryonic development and in gonadal and brain physiology.


Asunto(s)
Vasos Sanguíneos/embriología , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/embriología , Animales , Vasos Sanguíneos/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN/metabolismo , Pérdida del Embrión/genética , Pérdida del Embrión/metabolismo , Embrión de Mamíferos/metabolismo , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones
16.
Biochem Biophys Res Commun ; 527(2): 539-544, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32423801

RESUMEN

Glycine, a non-essential amino acid, is involved in both angiogenesis and anti-angiogenesis. We hypothesized that glycine would exert dose-dependent different effects on angiogenesis. In this study, we investigated the effects of a broad range of concentrations of glycine on vascular development using transgenic zebrafish Tg(fli1a:Myr-mCherry)ncv1 embryos. Effects of glycine transporter (GlyT) inhibitors (sarcosine and bitopertin) and a glycine receptor (GlyR) inhibitor (strychnine) were also examined in embryos in the absence or presence of glycine. After exposure to glycine and inhibitors, intersegmental vessels (ISVs) were observed by fluorescent microscopy. Low concentrations of glycine promoted the development of ISVs, whereas high concentrations reduced it. These effects of glycine could generally be reversed by treatment with GlyT and GlyR inhibitors. Furthermore, expressions of vascular endothelial growth factor (VEGF) (an angiogenic factor) and nitric oxide synthase (NOS) (an enzyme for nitric oxide synthesis) were associated with the dose-dependent effects of glycine. Our results suggest that glycine exerts dose-dependent biphasic effects on vascular development, which rely on GlyTs and GlyRs, and correlate with the expression of VEGF and NOS genes. At low concentrations, glycine acted as an angiogenic factor. In contrast, at high concentrations, glycine induced anti-angiogenesis. This evidence provides a novel insight into glycine as a unique target in angiogenic and anti-angiogenic therapy.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/farmacología , Vasos Sanguíneos/embriología , Embrión no Mamífero/efectos de los fármacos , Glicina/farmacología , Pez Cebra/embriología , Animales , Vasos Sanguíneos/efectos de los fármacos , Embrión no Mamífero/irrigación sanguínea , Neovascularización Fisiológica/efectos de los fármacos
17.
Int J Mol Sci ; 21(10)2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32429045

RESUMEN

The extracellular matrix (ECM) is critical in all aspects of vascular development and health: supporting cell anchorage, providing structure, organization and mechanical stability, and serving as a sink for growth factors and sustained survival signals. Abnormal changes in ECM protein expression, organization, and/or properties, and the ensuing changes in vascular compliance affect vasodilator responses, microvascular pressure transmission, and collateral perfusion. The changes in microvascular compliance are independent factors initiating, driving, and/or exacerbating a plethora of microvascular diseases of the eye including diabetic retinopathy (DR) and vitreoretinopathy, retinopathy of prematurity (ROP), wet age-related macular degeneration (AMD), and neovascular glaucoma. Congruently, one of the major challenges with most vascular regenerative therapies utilizing localized growth factor, endothelial progenitor, or genetically engineered cell delivery, is the regeneration of blood vessels with physiological compliance properties. Interestingly, vascular cells sense physical forces, including the stiffness of their ECM, through mechanosensitive integrins, their associated proteins and the actomyosin cytoskeleton, which generates biochemical signals that culminate in a rapid expression of matricellular proteins such as cellular communication network 1 (CCN1) and CCN2 (aka connective tissue growth factor or CTGF). Loss or gain of function of these proteins alters genetic programs of cell growth, ECM biosynthesis, and intercellular signaling, that culminate in changes in cell behavior, polarization, and barrier function. In particular, the function of the matricellular protein CCN2/CTGF is critical during retinal vessel development and regeneration wherein new blood vessels form and invest a preformed avascular neural retina following putative gradients of matrix stiffness. These observations underscore the need for further in-depth characterization of the ECM-derived cues that dictate structural and functional properties of the microvasculature, along with the development of new therapeutic strategies addressing the ECM-dependent regulation of pathophysiological stiffening of blood vessels in ischemic retinopathies.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/fisiopatología , Matriz Extracelular/metabolismo , Ojo/irrigación sanguínea , Ojo/patología , Microvasos/patología , Microvasos/fisiopatología , Animales , Fenómenos Biomecánicos , Vasos Sanguíneos/embriología , Ojo/embriología , Ojo/fisiopatología , Oftalmopatías/patología , Humanos
18.
Int J Mol Sci ; 21(10)2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32429094

RESUMEN

Diabetic retinopathy (DR) is the most frequent microvascular complication of long-term diabetes and the most common cause of blindness, increasing morbidity in the working-age population. The most effective therapies for these complications include laser photocoagulation and anti-vascular endothelial growth factor (VEGF) intravitreal injections. However, laser and anti-VEGF drugs are untenable as a final solution as they fail to address the underlying neurovascular degeneration and ischemia. Regenerative medicine may be a more promising approach, aimed at the repair of blood vessels and reversal of retinal ischemia. Stem cell therapy has introduced a novel way to reverse the underlying ischemia present in microvascular complications in diseases such as diabetes. The present review discusses current treatments, their side effects, and novel cell-based and tissue engineering approaches as a potential alternative therapeutic approach.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Retinopatía Diabética/terapia , Neovascularización Fisiológica , Ingeniería de Tejidos/métodos , Animales , Vasos Sanguíneos/embriología , Desarrollo Embrionario , Humanos
19.
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
20.
Clin Epigenetics ; 12(1): 30, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32070413

RESUMEN

The vasculature not only transports oxygenated blood, metabolites, and waste products but also serves as a conduit for hormonal communication between distant tissues. Therefore, it is important to maintain homeostasis within the vasculature. Recent studies have greatly expanded our understanding of the regulation of vasculature development and vascular-related diseases at the epigenetic level, including by protein posttranslational modifications, DNA methylation, and noncoding RNAs. Integrating epigenetic mechanisms into the pathophysiologic conceptualization of complex and multifactorial vascular-related diseases may provide promising therapeutic approaches. Several reviews have presented detailed discussions of epigenetic mechanisms not including histone methylation in vascular biology. In this review, we primarily discuss histone methylation in vascular development and maturity, and in vascular diseases.


Asunto(s)
Vasos Sanguíneos/metabolismo , Histonas/metabolismo , Enfermedades Vasculares/metabolismo , Disección Aórtica/metabolismo , Animales , Aorta Torácica , Aneurisma de la Aorta/metabolismo , Aterosclerosis/metabolismo , Vasos Sanguíneos/embriología , Angiopatías Diabéticas/metabolismo , Endotelio Vascular/metabolismo , Humanos , Metilación , Ratones , Neovascularización Patológica/metabolismo , Hipertensión Arterial Pulmonar/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA