Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 41(8): 2277-2292, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34162228

RESUMEN

OBJECTIVE: Healing processes, particularly reendothelialization, are essential for vascular homeostasis after plain old balloon angioplasty and stent implantation. Drug-eluting stents (DES) are commonly used for percutaneous coronary intervention because restenosis rates are reduced as compared with bare metal stents (BMS). However, in addition to understanding the nature of regenerated endothelial cells, concerns over incomplete stent healing persist, and the molecular effects of antiproliferative drug coatings on endothelium remain poorly understood. APPROACH AND RESULTS: We used the rabbit iliac artery model to analyze differences in stent endothelialization in BMS and DES. Histology and immunohistochemistry confirmed that stent coverage was significantly greater in BMS than in DES at 30 days after stent implantation. Single-cell RNA sequencing revealed a more immature transcriptomic signature of neointimal endothelial cell harvested from stented arteries in comparison with native and plain old balloon angioplasty­ treated arteries. Whereas the genetic signature of BMS was overall proangiogenic with enrichment of genes involved in endothelial proliferation, sprouting, and migration, as well as extracellular matrix assembly, DES-derived endothelial cell showed upregulation of genes associated with angiogenesis inhibition and endothelial activation. CONCLUSIONS: Single-cell RNA sequencing analysis identified unique transcriptional changes within regenerated endothelium after plain old balloon angioplasty and stent implantation. These data suggest unique endothelial transcriptional differences, which characterize the different response of the endothelium to vascular injury and may help explain why long-term responses in DES remain suboptimal.


Asunto(s)
Stents Liberadores de Fármacos , Células Endoteliales/ultraestructura , Procedimientos Endovasculares/instrumentación , Arteria Ilíaca/ultraestructura , Neointima , Repitelización , Análisis de la Célula Individual , Animales , Proliferación Celular , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Células Endoteliales/metabolismo , Procedimientos Endovasculares/efectos adversos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Arteria Ilíaca/metabolismo , Masculino , Microscopía Confocal , Microscopía Electrónica de Rastreo , Modelos Animales , Intervención Coronaria Percutánea/efectos adversos , Intervención Coronaria Percutánea/instrumentación , RNA-Seq , Conejos , Factores de Tiempo , Transcriptoma
2.
Microvasc Res ; 133: 104073, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32949575

RESUMEN

In this study, the angiogenic capacity of human endothelial cells was studied after being plated on the surface of polyurethane-poly caprolactone (PU/PCL) scaffolds for 72 h. In this study, cells were designated into five different groups, including PU, PU/PCL (2:1), PU/PCL (1:1); PU/PCL (1:2); and PCL. Data revealed that the PU/PCL (2:1) composition had a higher modulus and breakpoint in comparison with the other groups (p < 0.05). Compared to the other groups, the PU/PCL scaffold with a molar ratio of 2:1 had lower the contact angle θ and higher tensile stress (p < 0.05). The mean size of the PU nanofibers was reduced after the addition of PCL (p < 0.05). Based on our data, the culture of endothelial cells on the surface of PU/PCL (2:1) did not cause nitrosative stress and cytotoxic effects under static conditions compared to cells plated on a conventional plastic surface (p > 0.05). Based on data from the static condition, we fabricated a tubular PU/PCL (2:1) construct for six-day dynamic cell culture inside loop air-lift bioreactors. Scanning electron microscopy showed the attachment of endothelial cells to the luminal surface of the PU/PCL scaffold. Cells were flattened and aligned under the culture medium flow. Immunofluorescence imaging showed the attachment of cells to the luminal surface indicated by blue nuclei on the luminal surface. These data demonstrated that the application of PU/PCL substrate could stimulate endothelial cells activity under static and dynamic conditions.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/fisiología , Nanofibras , Poliésteres/química , Poliuretanos/química , Andamios del Tejido , Reactores Biológicos , Adhesión Celular , Técnicas de Cultivo de Célula , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Módulo de Elasticidad , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Resistencia a la Tracción , Factores de Tiempo
3.
EMBO Rep ; 19(8)2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29967223

RESUMEN

The fate of mesenchymal stem cells (MSCs) in the perivascular niche, as well as factors controlling their fate, is poorly understood. Here, we study MSCs in the perivascular microenvironment of endothelial capillaries by modifying a synthetic 3D biomimetic poly(ethylene glycol) (PEG)-hydrogel system in vitro We show that MSCs together with endothelial cells form micro-capillary networks specifically in soft PEG hydrogels. Transcriptome analysis of human MSCs isolated from engineered capillaries shows a prominent switch in extracellular matrix (ECM) production. We demonstrate that the ECM phenotypic switch of MSCs can be recapitulated in the absence of endothelial cells by functionalizing PEG hydrogels with the Notch-activator Jagged1. Moreover, transient culture of MSCs in Notch-inducing microenvironments reveals the reversibility of this ECM switch. These findings provide insight into the perivascular commitment of MSCs by use of engineered niche-mimicking synthetic hydrogels.


Asunto(s)
Linaje de la Célula , Hidrogeles/farmacología , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica/efectos de los fármacos , Receptores Notch/metabolismo , Células de la Médula Ósea/citología , Capilares/efectos de los fármacos , Capilares/fisiología , Capilares/ultraestructura , Linaje de la Célula/efectos de los fármacos , Microambiente Celular/efectos de los fármacos , Técnicas de Cocultivo , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestructura , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/ultraestructura , Polietilenglicoles/farmacología
4.
Arterioscler Thromb Vasc Biol ; 39(12): 2492-2504, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31597449

RESUMEN

OBJECTIVE: Understanding message delivery among vascular cells is essential for deciphering the intercellular communications in cardiovascular diseases. MicroRNA (miR)-92a is enriched in endothelial cells (ECs) and circulation under atheroprone conditions. Macrophages are the primary immune cells in atherosclerotic lesions that modulate lesion development. Therefore, we hypothesize that, in response to atheroprone stimuli, ECs export miR-92a to macrophages to regulate their functions and enhance atherosclerotic progression. Approach and Results: We investigated the macrophage functions that are regulated by EC miR-92a under atheroprone microenvironments. We first determined the distributions of functional extracellular miR-92a by fractionating the intravesicular and extravesicular compartments from endothelial conditioned media and mice serum. The results indicate that extracellular vesicles are the primary vehicles for EC miR-92a transportation. Overexpression of miR-92a in ECs enhanced the proinflammatory responses and low-density lipoprotein uptake, while impaired the migration, of cocultured macrophage. Opposite effects were found in macrophages cocultured with ECs with miR-92a knockdown. Further analyses demonstrated that intravesicular miR-92a suppressed the expression of target gene KLF4 (Krüppel-like factor 4) in macrophages, suggesting a mechanism by which intravesicular miR-92a regulates recipient cell functions. Indeed, the overexpression of KLF4 rescued the EC miR-92a-induced macrophage atheroprone phenotypes. Furthermore, an inverse correlation of intravesicular miR-92a in blood serum and KLF4 expression in lesions was observed in atherosclerotic animals, indicating the potential function of extracellular miR-92a in regulating vascular diseases. CONCLUSIONS: EC miR-92a can be transported to macrophages through extracellular vesicles to regulate KLF4 levels, thus leading to the atheroprone phenotypes of macrophage and, hence, atherosclerotic lesion formation.


Asunto(s)
Aterosclerosis/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Macrófagos/metabolismo , MicroARNs/genética , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Comunicación Celular , Células Cultivadas , Líquido Extracelular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/biosíntesis , Macrófagos/ultraestructura , Ratones , MicroARNs/biosíntesis , Microscopía Electrónica de Transmisión
5.
Lipids Health Dis ; 19(1): 128, 2020 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-32505182

RESUMEN

BACKGROUND: Inhalation of common air pollutants such as diesel and biodiesel combustion products can induce vascular changes in humans which may contribute to increased mortality and morbidity associated with fine particulate matter exposures. Diesel, biodiesel, and other combustion byproducts contain fatty acid components capable of entering the body through particulate matter inhalation. Fatty acids can also be endogenously released into circulation following a systemic stress response to some inhaled pollutants such as ozone. When in the circulation, bioactive fatty acids may interact with cells lining the blood vessels, potentially inducing endothelial dysfunction. To examine whether fatty acids could potentially be involved in human vascular responses to air pollutants, we determined the effects of fatty acids and derivatives on important vascular cell functions. METHODS: Human umbilical vein endothelial cells (HUVEC) were exposed in vitro to oleic acid (OA) or OA metabolites for 4-48 h. Cytotoxicity, vasodilator production (by ELISA measurement), mitochondrial function (using Sea Horse assays), and iron metabolism (inferred by ICP-OES measurements) were examined, with standard statistical testing (ANOVA, t-tests) employed. RESULTS: Dose-dependent cytotoxicity was noted at 24 h, with 12-hydroxy OA more potent than OA. Mitochondrial stress testing showed that 12-hydroxy OA and OA induce mitochondrial dysfunction. Analysis of soluble mediator release from HUVEC showed a dose-dependent increase in prostaglandin F2α, a lipid involved in control of vascular tone, at 24 h (85% above controls) after OA-BSA exposure. RT-PCR analysis revealed OA did not induce changes in gene expression at noncytotoxic concentrations in exposed HUVEC, but 12-OH OA did alter ICAM and COX2 gene expression. CONCLUSIONS: Together, these data demonstrate that FA may be capable of inducing cytotoxic effects and altering expression of mediators of vascular function following inhalation exposure, and may be implicated in air pollutant-induced deaths and hospitalizations. (267 of max 350 words).


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Ácido Oléico/toxicidad , Sistema Vasomotor/efectos de los fármacos , Contaminantes Atmosféricos/toxicidad , Ciclooxigenasa 2/genética , Dinoprost/biosíntesis , Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Molécula 1 de Adhesión Intercelular/genética , Hierro/metabolismo , Ácidos Ricinoleicos/toxicidad , Sistema Vasomotor/fisiología
6.
Biophys J ; 117(10): 1795-1804, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31706566

RESUMEN

Cells in vivo encounter and exert forces as they interact with the extracellular matrix (ECM) and neighboring cells during migration. These mechanical forces play crucial roles in regulating cell migratory behaviors. Although a variety of studies have focused on describing single-cell or the collective cell migration behaviors, a fully mechanistic understanding of how the cell-cell (intercellular) and cell-ECM (extracellular) traction forces individually and cooperatively regulate single-cell migration and coordinate multicellular movement in a cellular monolayer is still lacking. Here, we developed an integrated experimental and analytical system to examine both the intercellular and extracellular traction forces acting on individual cells within an endothelial cell colony as well as their roles in guiding cell migratory behaviors (i.e., cell translation and rotation). Combined with force, multipole, and moment analysis, our results revealed that traction force dominates in regulating cell active translation, whereas intercellular force actively modulates cell rotation. Our findings advance the understanding of the intricacies of cell-cell and cell-ECM forces in regulating cellular migratory behaviors that occur during the monolayer development and may yield deeper insights into the single-cell dynamic behaviors during tissue development, embryogenesis, and wound healing.


Asunto(s)
Movimiento Celular/fisiología , Uniones Célula-Matriz/fisiología , Animales , Fenómenos Biomecánicos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Ratones , Rotación , Torque
7.
J Cell Physiol ; 234(6): 9525-9534, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30370660

RESUMEN

Sprouting angiogenesis is a major form of neovascularization of tissues suffering from hypoxia and other related stress. Endothelial cells (ECs) undergo proliferation, differentiation, programmed death, and migration during angiogenic sprouting, but the underlying molecular mechanisms regulating ECs in angiogenesis have been incompletely elucidated. Here we report that the transmembrane protein 215 (TMEM215) is involved in angiogenesis by regulating EC survival. The murine TMEM215 gene, which possesses two transcriptional starting sites as determined by 5'-rapid amplification of complementary DNA (cDNA) ends (RACE), encodes a two-pass TMEM. The TMEM215 transcripts were detected in ECs in addition to other tissues by quantitative reverse transcription-polymerase chain reaction. Immunofluorescence showed that TMEM215 was expressed in the vasculature in retina, liver, and tumor, and colocalized with EC markers. We show that knockdown of TMEM215 in ECs induced strong cell death of ECs in vitro without affecting cell proliferation and migration, suggesting that TMEM215 was required for EC survival. Downregulation of TMEM215 expression compromised lumen formation and sprouting capacities of ECs in vitro. Moreover, intravitreous injection of TMEM215 small interfering RNA resulted in delayed and abnormal development of retinal vasculature with poor perfusion. These results identified TMEM215 as a novel molecule involved in angiogenesis by regulating the survival of ECs.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/metabolismo , Proteínas de la Membrana/metabolismo , Neovascularización Fisiológica , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Supervivencia Celular , Células Endoteliales/ultraestructura , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Retina/metabolismo
8.
Microcirculation ; 26(1): e12515, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30431204

RESUMEN

OBJECTIVE: To investigate the effects of TNF-α-induced exosomes release on the biological behavior, metabolism, and bioenergetics of HUVECs. METHODS: Exosomes were isolated from conditioned media of HUVECs by ultracentrifugation after treatment with or without TNF-α. HUVECs were treated with or without TNF-α, or different concentrations of exosomes isolated from conditioned media with or without TNF-α induction (TExo and CExo , respectively). RESULTS: The results showed that TNF-α significantly inhibited migration, tube formation, and increased apoptosis rate of HUVECs compared with controls. Furthermore, TNF-α-induced exosomes (TExo ) rather than CExo , indicated similar effects to inhibit migration, tube formation and promote endothelial apoptosis. Although TNF-α treatment did not show a statistical difference, TExo significantly inhibited extracellular OCR compared with controls. TExo could significantly inhibited intracellular OCR in a hypoxia condition. TNF-α significantly increased L-ECA compared with control cells, and TExo showed similar stimulative effect on L-ECA. CONCLUSIONS: TNF-α-induced exosomes could significantly (a) change migration, tube formation, and apoptosis; (b) inhibit endothelial extracellular OCR and intracellular OCR (hypoxia); (c) increase glycolysis rate of the endothelial cells. These data provide new evidence for exploring endothelial behavior regulation using exosomes and their effects on endothelial metabolism and bioenergetics.


Asunto(s)
Exosomas/fisiología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Apoptosis , Movimiento Celular , Células Cultivadas , Metabolismo Energético , Exosomas/efectos de los fármacos , Glucólisis , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Hipoxia , Ácido Láctico/metabolismo , Consumo de Oxígeno
9.
J Cell Sci ; 129(4): 743-56, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26769900

RESUMEN

During angiogenesis, endothelial cells must coordinate matrix proteolysis with migration. Here, we tested whether the focal adhesion scaffold protein Hic-5 (also known as TGFB1I1) regulated endothelial sprouting in three dimensions. Hic-5 silencing reduced endothelial sprouting and lumen formation, and sprouting defects were rescued by the return of Hic-5 expression. Pro-angiogenic factors enhanced colocalization and complex formation between membrane type-1 matrix metalloproteinase (MT1-MMP, also known as MMP14) and Hic-5, but not between paxillin and MT1-MMP. The LIM2 and LIM3 domains of Hic-5 were necessary and sufficient for Hic-5 to form a complex with MT1-MMP. The degree of interaction between MT1-MMP and Hic-5 and the localization of the complex within detergent-resistant membrane fractions were enhanced during endothelial sprouting, and Hic-5 depletion lowered the surface levels of MT1-MMP. In addition, we observed that loss of Hic-5 partially reduced complex formation between MT1-MMP and focal adhesion kinase (FAK, also known as PTK2), suggesting that Hic-5 bridges MT1-MMP and FAK. Finally, Hic-5 LIM2-LIM3 deletion mutants reduced sprout initiation. Hic-5, MT1-MMP and FAK colocalized in angiogenic vessels during porcine pregnancy, supporting that this complex assembles during angiogenesis in vivo. Collectively, Hic-5 appears to enhance complex formation between MT1-MMP and FAK in activated endothelial cells, which likely coordinates matrix proteolysis and cell motility.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/enzimología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas con Dominio LIM/fisiología , Metaloproteinasa 14 de la Matriz/metabolismo , Animales , Movimiento Celular , Extensiones de la Superficie Celular/enzimología , Células Cultivadas , Femenino , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Neovascularización Fisiológica , Embarazo , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Sus scrofa
10.
J Cell Sci ; 129(21): 4091-4104, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27656109

RESUMEN

Endocytosis plays a crucial role in receptor signalling. VEGFR2 (also known as KDR) and its ligand VEGFA are fundamental in neovascularisation. However, our understanding of the role of endocytosis in VEGFR2 signalling remains limited. Despite the existence of diverse internalisation routes, the only known endocytic pathway for VEGFR2 is the clathrin-mediated pathway. Here, we show that this pathway is the predominant internalisation route for VEGFR2 only in the absence of ligand. Intriguingly, VEGFA induces a new internalisation itinerary for VEGFR2, the pathway of macropinocytosis, which becomes the prevalent endocytic route for the receptor in the presence of ligand, whereas the contribution of the clathrin-mediated route becomes minor. Macropinocytic internalisation of VEGFR2, which mechanistically is mediated through the small GTPase CDC42, takes place through macropinosomes generated at ruffling areas of the membrane. Interestingly, macropinocytosis plays a crucial role in VEGFA-induced signalling, endothelial cell functions in vitro and angiogenesis in vivo, whereas clathrin-mediated endocytosis is not essential for VEGFA signalling. These findings expand our knowledge on the endocytic pathways of VEGFR2 and suggest that VEGFA-driven internalisation of VEGFR2 through macropinocytosis is essential for endothelial cell signalling and angiogenesis.


Asunto(s)
Neovascularización Fisiológica , Pinocitosis , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Clatrina/metabolismo , Dinaminas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Modelos Biológicos , Proteína de Unión al GTP cdc42/metabolismo
11.
J Mol Recognit ; 31(9): e2723, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29748974

RESUMEN

Endothelial cells, due to their location, are interesting objects for atomic force spectroscopy study. They constitute a barrier between blood and vessel tissues located deeper, and therefore they are the first line of contact with various substances present in blood, eg, drugs or nanoparticles. This work intends to verify whether the mechanical response of immortalized human umbilical vein endothelial cells (EA.hy926), when exposed to silver nanoparticles, as measured using force spectroscopy, could be effectively used as a bio-indicator of the physiological state of the cells. Silver nanoparticles were characterized with transmission electron microscopy and dynamic light scattering techniques. Tetrazolium salt reduction test was used to determine cell viability after treatment with silver nanoparticles. An elasticity of native cells was examined in the Hanks' buffer whereas fixed cells were softly fixed with formaldehyde. Additional aspect of the work is the comparative force spectroscopy utilizing AFM probes of ball-shape and conical geometries, in order to understand what changes in cell elasticity, caused by SNPs, were detectable with each probe. As a supplement to elasticity studies, cell morphology observation by atomic force microscopy and detection of silver nanoparticles inside cells using transmission electron microscopy were also performed. Cells exposed to silver nanoparticles at the highest selected concentrations (3.6 µg/mL, 16 µg/mL) are less elastic. It may be associated with the reorganization of the cellular cytoskeleton and the "strengthening" of the cell cortex caused by presence of silver nanoparticles. This observation does not depend on cell fixation. Agglomerates of silver nanoparticles were observed on the cell membrane as well as inside the cells.


Asunto(s)
Células Endoteliales/química , Fenómenos Mecánicos , Nanopartículas del Metal/química , Supervivencia Celular/efectos de los fármacos , Citoesqueleto/química , Citoesqueleto/efectos de los fármacos , Dispersión Dinámica de Luz , Células Endoteliales/efectos de los fármacos , Células Endoteliales/ultraestructura , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Nanopartículas del Metal/administración & dosificación , Nanopartículas del Metal/ultraestructura , Microscopía Electrónica de Transmisión , Plata/química , Espectrofotometría Atómica
12.
Microvasc Res ; 118: 101-112, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29550275

RESUMEN

Small-diameter vascular grafts are needed for the treatment of coronary artery diseases in the case of limited accessibility of the autologous vessels. Synthetic scaffolds have many disadvantages so in recent years vascular constructs (VCs) made from cellularized natural scaffolds was seen to be very promising but number of studies comprising this area is very limited. In our study, our aim is to generate fully natural triple-layered VC that constitutes all the layers of blood vessel with vascular cells. CD146+ perivascular cells (PCs) were isolated from human umbilical cord vein (HUCV) and differentiated into smooth muscle cells (SMCs) and fibroblasts. They were then combined with collagen type I/elastin/dermatan sulfate and collagen type I/fibrin to form tunica media and tunica adventitia respectively. HUCV endothelial cells (ECs) were seeded on the construct by cell sheet engineering method after fibronectin and heparin coating. Characterization of the VC was performed by immunolabeling, histochemical staining and electron microscopy (SEM and TEM). Differentiated cells were identified by means of immunofluorescent (IF) labeling. SEM and TEM analysis of VCs revealed the presence of three histologic tunicae. Collagen and elastic fibers were observed within the ECM by histochemical staining. The vascular endothelial growth factor receptor expressing ECs in tunica intima; α-SMA expressing SMCs in tunica media and; the tenascin expressing fibroblasts in tunica adventitia were detected by IF labeling. In conclusion, by combining natural scaffolds and vascular cells differentiated from CD146+ PCs, VCs can be generated layer by layer. This study will provide a preliminary blood vessel model for generation of fully natural small-diameter vascular grafts.


Asunto(s)
Prótesis Vascular , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Miocitos del Músculo Liso/metabolismo , Ingeniería de Tejidos/métodos , Andamios del Tejido , Implantación de Prótesis Vascular , Antígeno CD146/metabolismo , Comunicación Celular , Transdiferenciación Celular , Células Cultivadas , Colágeno/metabolismo , Dermatán Sulfato/metabolismo , Elastina/metabolismo , Matriz Extracelular/trasplante , Matriz Extracelular/ultraestructura , Fibroblastos/trasplante , Fibroblastos/ultraestructura , Células Endoteliales de la Vena Umbilical Humana/trasplante , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Miocitos del Músculo Liso/trasplante , Miocitos del Músculo Liso/ultraestructura , Fenotipo
13.
J Biol Chem ; 291(37): 19245-56, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27435676

RESUMEN

Endorepellin, the C-terminal domain of perlecan, is an angiostatic molecule that acts as a potent inducer of autophagy via its interaction with VEGFR2. In this study, we examined the effect of endorepellin on endothelial cells using atomic force microscopy. Soluble endorepellin caused morphological and biophysical changes such as an increase in cell surface roughness and cell height. Surprisingly, these changes were not accompanied by alterations in the endothelial cell elastic modulus. We discovered that endorepellin-induced autophagic flux led to co-localization of mammalian target of rapamycin with LC3-positive autophagosomes. Endorepellin functioned upstream of AMP-activated kinase α, as compound C, an inhibitor of AMP-activated kinase α, abrogated endorepellin-mediated activation and co-localization of Beclin 1 and LC3, thereby reducing autophagic progression. Functionally, we discovered that both endorepellin and Torin 1, a canonical autophagic inducer, blunted ex vivo angiogenesis. We conclude that autophagy is a novel mechanism by which endorepellin promotes angiostasis independent of nutrient deprivation.


Asunto(s)
Autofagia , Proteoglicanos de Heparán Sulfato/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Fragmentos de Péptidos/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adenilato Quinasa/metabolismo , Beclina-1/metabolismo , Módulo de Elasticidad , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Lactosilceramidos/metabolismo
14.
Blood ; 125(22): 3509-16, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-25716207

RESUMEN

Weibel-Palade bodies (WPBs) comprise an on-demand storage organelle within vascular endothelial cells. It's major component, the hemostatic protein von Willebrand factor (VWF), is known to assemble into long helical tubules and is hypothesized to drive WPB biogenesis. However, electron micrographs of WPBs at the Golgi apparatus show that these forming WPBs contain very little tubular VWF compared with mature peripheral WPBs, which raises questions on the mechanisms that increase the VWF content and facilitate vesicle growth. Using correlative light and electron microscopy and electron tomography, we investigated WPB biogenesis in time. We reveal that forming WPBs maintain multiple connections to the Golgi apparatus throughout their biogenesis. Also by volume scanning electron microscopy, we confirmed the presence of these connections linking WPBs and the Golgi apparatus. From electron tomograms, we provided evidence that nontubular VWF is added to WPBs, which suggested that tubule formation occurs in the WPB lumen. During this process, the Golgi membrane and clathrin seem to provide a scaffold to align forming VWF tubules. Overall, our data show that multiple connections with the Golgi facilitate content delivery and indicate that the Golgi appears to provide a framework to determine the overall size and dimensions of newly forming WPBs.


Asunto(s)
Aparato de Golgi/metabolismo , Cuerpos de Weibel-Palade/metabolismo , Transporte Biológico/efectos de los fármacos , Células Cultivadas , Aparato de Golgi/ultraestructura , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Microscopía Electrónica , Microscopía Electrónica de Rastreo , Microscopía de Polarización , Acetato de Tetradecanoilforbol/farmacología , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/ultraestructura , Cuerpos de Weibel-Palade/ultraestructura , Factor de von Willebrand/metabolismo
15.
Circ Res ; 116(11): 1753-64, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25801897

RESUMEN

RATIONALE: The miR-143/145 cluster is highly expressed in smooth muscle cells (SMCs), where it regulates phenotypic switch and vascular homeostasis. Whether it plays a role in neighboring endothelial cells (ECs) is still unknown. OBJECTIVE: To determine whether SMCs control EC functions through passage of miR-143 and miR-145. METHODS AND RESULTS: We used cocultures of SMCs and ECs under different conditions, as well as intact vessels to assess the transfer of miR-143 and miR-145 from one cell type to another. Imaging of cocultured cells transduced with fluorescent miRNAs suggested that miRNA transfer involves membrane protrusions known as tunneling nanotubes. Furthermore, we show that miRNA passage is modulated by the transforming growth factor (TGF) ß pathway because both a specific transforming growth factor-ß (TGFß) inhibitor (SB431542) and an shRNA against TGFßRII suppressed the passage of miR-143/145 from SMCs to ECs. Moreover, miR-143 and miR-145 modulated angiogenesis by reducing the proliferation index of ECs and their capacity to form vessel-like structures when cultured on matrigel. We also identified hexokinase II (HKII) and integrin ß 8 (ITGß8)-2 genes essential for the angiogenic potential of ECs-as targets of miR-143 and miR-145, respectively. The inhibition of these genes modulated EC phenotype, similarly to miR-143 and miR-145 overexpression in ECs. These findings were confirmed by ex vivo and in vivo approaches, in which it was shown that TGFß and vessel stress, respectively, triggered miR-143/145 transfer from SMCs to ECs. CONCLUSIONS: Our results demonstrate that miR-143 and miR-145 act as communication molecules between SMCs and ECs to modulate the angiogenic and vessel stabilization properties of ECs.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , MicroARNs/metabolismo , Miocitos del Músculo Liso/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Animales , Benzamidas/farmacología , Transporte Biológico/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiología , Western Blotting , Comunicación Celular/efectos de los fármacos , Comunicación Celular/genética , Extensiones de la Superficie Celular/efectos de los fármacos , Extensiones de la Superficie Celular/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Dioxoles/farmacología , Regulación de la Expresión Génica , Hexoquinasa/genética , Hexoquinasa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Cadenas beta de Integrinas/genética , Cadenas beta de Integrinas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Microscopía Confocal , Microscopía Electrónica de Rastreo , Miocitos del Músculo Liso/ultraestructura , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 36(9): 1847-53, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27386939

RESUMEN

OBJECTIVE: It is widely accepted that the presence of a glycosaminoglycan-rich glycocalyx is essential for endothelialized vasculature health; in fact, a damaged or impaired glycocalyx has been demonstrated in many vascular diseases. Currently, there are no methods that characterize glycocalyx functionality, thus limiting investigators' ability to assess the role of the glycocalyx in vascular health. APPROACH AND RESULTS: We have developed novel, easy-to-use, in vitro assays that directly quantify live endothelialized surface's functional heparin weights and their anticoagulant capacity to inactivate Factor Xa and thrombin. Using our assays, we characterized 2 commonly used vascular models: native rat aorta and cultured human umbilical vein endothelial cell monolayer. We determined heparin contents to be ≈10 000 ng/cm(2) on the native aorta and ≈10-fold lower on cultured human umbilical vein endothelial cells. Interestingly, human umbilical vein endothelial cells demonstrated a 5-fold lower anticoagulation capacity in inactivating both Factor Xa and thrombin relative to native aortas. We verified the validity and accuracy of the novel assays developed in this work using liquid chromatography-mass spectrometry analysis. CONCLUSIONS: Our assays are of high relevance in the vascular community because they can be used to establish the antithrombogenic capacity of many different types of surfaces such as vascular grafts and transplants. This work will also advance the capacity for glycocalyx-targeting therapeutics development to treat damaged vasculatures.


Asunto(s)
Aorta Torácica/metabolismo , Bioensayo/métodos , Coagulación Sanguínea , Factor Xa/metabolismo , Glicocálix/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Trombina/metabolismo , Animales , Antitrombinas/metabolismo , Aorta Torácica/ultraestructura , Células Cultivadas , Cromatografía Liquida , Glicocálix/ultraestructura , Heparina/metabolismo , Heparitina Sulfato/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Masculino , Espectrometría de Masas , Microscopía Electrónica de Transmisión , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Factores de Tiempo
17.
Methods ; 98: 150-157, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26564237

RESUMEN

Nascent transcripts being copied from specific human genes can be detected using RNA FISH (fluorescence in situ hybridization) with intronic probes, and the distance between two different nascent transcripts is often measured when studying structure-function relationships. Such distance measurements are limited by the resolution of the light microscope. Here we describe methods for measuring these distances in cultured cells with a precision of a few tens of nanometers, using equipment found in most laboratories (i.e., a wide-field fluorescence microscope equipped with a charged-coupled-device camera). Using images of pairs of transcripts that are often co-transcribed, we discuss how selection of cell type, design of FISH probes, image acquisition, and image processing affect the precision that can be achieved.


Asunto(s)
Colorantes Fluorescentes/química , Hibridación Fluorescente in Situ/métodos , Sondas Moleculares/química , ARN Mensajero/genética , Proteínas Represoras/genética , Transcripción Genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Procesamiento de Imagen Asistido por Computador , Intrones , Microscopía Fluorescente , ARN Mensajero/metabolismo , Proteínas Represoras/metabolismo , Imagen Individual de Molécula/métodos , Programas Informáticos , Fijación del Tejido/métodos
18.
Cell Biochem Funct ; 35(2): 83-97, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28211084

RESUMEN

To date, many studies have been conducted to find out the underlying mechanisms of hyperglycemia-induced complications in diabetes mellitus, attributed to the cellular pathologies of different cells-especially endothelial cells. However, there are still many ambiguities and unresolved issues to be clarified. Here, we investigated the alteration in biophysical and biochemical properties in human umbilical vein endothelial cells exposed to a high-glucose concentration (30mM), comparable to glucose content in type 2 diabetes mellitus, over a course of 120 hours. In addition to a reduction in the rate of cell viability and induction of oxidative stress orchestrated by the high-glucose condition, the dynamic of the fatty acid profile-including polyunsaturated, monounsaturated, and saturated fatty acids-was also altered in favor of saturated fatty acids. Genetic imbalances were also detected at chromosomal level in the cells exposed to the abnormal concentration of glucose after 120 hours. Moreover, the number of tip cells (CD31+ /CD34+ ) and in vitro tubulogenesis capability negatively diminished in comparison to parallel control groups. We found that diabetic hyperglycemia was associated with a decrease in the cell-cell tight junction and upregulation in vascular endothelial cadherin and zonula occludens (ZO)-1 molecules after 72 and 120 hours of exposure to the abnormal glucose concentration, which resulted in a profound reduction in transendothelial electrical resistance. The surface plasmon resonance analysis of the human umbilical vein endothelial cells immobilized on gold-coated sensor chips confirmed the loosening of the cell to cell intercellular junction as well as stable attachment of each cell to the basal surface. Our findings highlighted the disturbing effects of a diabetic hyperglycemia on either biochemical or biophysical properties of endothelial cells.


Asunto(s)
Aberraciones Cromosómicas/efectos de los fármacos , Ácidos Grasos Insaturados/antagonistas & inhibidores , Ácidos Grasos/agonistas , Glucosa/toxicidad , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Uniones Estrechas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Impedancia Eléctrica , Ácidos Grasos/metabolismo , Ácidos Grasos Insaturados/metabolismo , Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Hiperglucemia/metabolismo , Hiperglucemia/patología , Cariotipificación , Modelos Biológicos , Necrosis/inducido químicamente , Necrosis/patología , Estrés Oxidativo , Uniones Estrechas/metabolismo , Uniones Estrechas/ultraestructura , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
19.
Toxicol Mech Methods ; 27(3): 191-200, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27997269

RESUMEN

It was recently shown that ZnO nanoparticles (NPs) could induce endoplasmic reticulum (ER) stress in human umbilical vein endothelial cells (HUVECs). If ER stress is associated the toxicity of ZnO NPs, the presence of ER stress inducer thapsigargin (TG) should alter the response of HUVECs to ZnO NP exposure. In this study, we addressed this issue by assessing cytotoxicity, oxidative stress and inflammatory responses in ZnO NP exposed HUVECs with or without the presence of TG. Moreover, TiO2 NPs were used to compare the effects. Exposure to 32 µg/mL ZnO NPs (p < 0.05), but not TiO2 NPs (p > 0.05), significantly induced cytotoxicity as assessed by WST-1 and neutral red uptake assay, as well as intracellular ROS. ZnO NPs dose-dependently increased the accumulation of intracellular Zn ions, and ZnSO4 induced similar cytotoxic effects as ZnO NPs, which indicated a role of Zn ions. The release of inflammatory proteins tumor necrosis factor α (TNFα) and interleukin-6 (IL-6) or the adhesion of THP-1 monocytes to HUVECs was not significantly affected by ZnO or TiO2 NP exposure (p > 0.05). The presence of 250 nM TG significantly induced cytotoxicity, release of IL-6 and THP-1 monocyte adhesion (p < 0.01), but did not significantly affect intracellular ROS or release of TNFα (p > 0.05). ANOVA analysis indicated no interaction between exposure to ZnO NPs and the presence of TG on almost all the endpoints (p > 0.05) except neutral red uptake assay (p < 0.01). We concluded ER stress is probably not associated with ZnO NP exposure induced oxidative stress and inflammatory responses in HUVECs.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Nanopartículas/toxicidad , Tapsigargina/toxicidad , Titanio/toxicidad , Óxido de Zinc/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Nanopartículas/química , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Especies Reactivas de Oxígeno/metabolismo , Propiedades de Superficie , Tapsigargina/química , Titanio/química , Óxido de Zinc/química
20.
Biochem Biophys Res Commun ; 470(4): 961-6, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26797282

RESUMEN

Carvedilol, a third-generation and nonselective ß-adrenoceptor antagonist, is a licensed drug for treating patients suffering from heart failure in clinics. It has been shown that Carvedilol protects cells against mitochondrial dysfunction. However, it's unknown whether Carvedilol affects mitochondrial biogenesis. In this study, we found that treatment with Carvedilol in HUVECs resulted in a significant increase of PGC-1α, NRF1, and TFAM. Notably, Carvedilol significantly increased mtDNA contents and the two mitochondrial proteins, cytochrome C and COX IV. In addition, MitoTracker Red staining results indicated that treatment with Carvedilol increased mitochondria mass. Mechanistically, we found that the effect of Carvedilol on the expression of PGC-1α is mediated by the PKA-CREB pathway. Importantly, our results revealed that stimulation of mitochondrial biogenesis by carvedilol resulted in functional gain of the mitochondria by showing increased oxygen consumption and mitochondrial respiratory rate. The increased expression of PGC-1α and mitochondrial biogenesis induced by Carvedilol might suggest a new mechanism of the therapeutic effects of Carvedilol in heart failure.


Asunto(s)
Carbazoles/administración & dosificación , Proteínas de Unión al ADN/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Mitocondrias/fisiología , Proteínas Mitocondriales/metabolismo , Propanolaminas/administración & dosificación , Factores de Transcripción/metabolismo , Antagonistas Adrenérgicos beta/administración & dosificación , Carvedilol , Aumento de la Célula/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Mitocondrias/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA