Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Circ Res ; 129(12): 1158-1174, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34747636

RESUMEN

RATIONALE: Disturbed flow occurring in arterial branches and curvatures induces vascular endothelial cell (EC) dysfunction and atherosclerosis. We postulated that disturbed flow plays important role in modulating phosphoprotein expression profiles to regulate endothelial functions and atherogenesis. OBJECTIVE: The goal of this study is to discover novel site-specific phosphorylation alterations induced by disturbed flow in ECs to contribute to atherosclerosis. METHODS AND RESULTS: Quantitative phosphoproteomics analysis of ECs exposed to disturbed flow with low and oscillatory shear stress (0.5±4 dynes/cm2) versus pulsatile shear stress (12±4 dynes/cm2) revealed that oscillatory shear stress induces phospho-YY1S118 (serine [S]118 phosphorylation of Yin Yang 1) in ECs. Elevated phospho-YY1S118 level in ECs was further confirmed to be present in the disturbed flow regions in experimental animals and human atherosclerotic arteries. This disturbed flow-induced EC phospho-YY1S118 is mediated by CK2α (casein kinase 2α) through its direct interaction with YY1. Yeast 2-hybrid library screening and in situ proximity ligation assays demonstrate that phospho-YY1S118 directly binds ZKSCAN4 (zinc finger with KRAB [krüppel-associated box] and SCAN [SRE-ZBP, CTfin51, AW-1 and Number 18 cDNA] domains 4) to induce promoter activity and gene expression of HDM2 (human double minute 2), which consequently induces EC proliferation through downregulation of p53 and p21CIP1. Administration of apoE-deficient (ApoE-/-) mice with CK2-specific inhibitor tetrabromocinnamic acid or atorvastatin inhibits atherosclerosis formation through downregulations of EC phospho-YY1S118 and HDM2. Generation of novel transgenic mice bearing EC-specific overexpression of S118-nonphosphorylatable mutant of YY1 in ApoE-/- mice confirms the critical role of phospho-YY1S118 in promoting atherosclerosis through EC HDM2. CONCLUSIONS: Our findings provide new insights into the mechanisms by which disturbed flow induces endothelial phospho-YY1S118 to promote atherosclerosis, thus indicating phospho-YY1S118 as a potential molecular target for atherosclerosis treatment.


Asunto(s)
Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Aterosclerosis/fisiopatología , Sitios de Unión , Circulación Sanguínea , Quinasa de la Caseína II/metabolismo , Línea Celular , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción YY1/química , Factor de Transcripción YY1/genética , Dedos de Zinc
2.
Proc Natl Acad Sci U S A ; 114(8): 2072-2077, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28167758

RESUMEN

Histone deacetylases (HDACs) and microRNAs (miRs) have emerged as two important epigenetic factors in the regulation of vascular physiology. This study aimed to elucidate the relationship between HDACs and miRs in the hemodynamic modulation of endothelial cell (EC) dysfunction. We found that miR-10a has the lowest expression among all examined shear-responsive miRs in ECs under oscillatory shear stress (OS), and a relatively high expression under pulsatile shear stress (PS). PS and OS alter EC miR-10a expression to regulate the expression of its direct target GATA6 and downstream vascular cell adhesion molecule (VCAM)-1. PS induces the expression, nuclear accumulation, and association of retinoid acid receptor-α (RARα) and retinoid X receptor-α (RXRα). RARα and RXRα serve as a "director" and an "enhancer," respectively, to enhance RARα binding to RA-responsive element (RARE) and hence miR-10a expression, thus down-regulating GATA6/VCAM-1 signaling in ECs. In contrast, OS induces associations of "repressors" HDAC-3/5/7 with RARα to inhibit the RARα-directed miR-10a signaling. The flow-mediated miR-10a expression is regulated by Krüppel-like factor 2 through modulation in RARα-RARE binding, with the consequent regulation in GATA6/VCAM-1 in ECs. These results are confirmed in vivo by en face staining on the aortic arch vs. the straight thoracic aorta of rats. Our findings identify a mechanism by which HDACs and RXRα modulate the hormone receptor RARα to switch miR-10a expression and hence the proinflammatory vs. anti-inflammatory responses of vascular endothelium under different hemodynamic forces.


Asunto(s)
Endotelio Vascular/fisiología , Factor de Transcripción GATA6/genética , Histona Desacetilasas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , MicroARNs/metabolismo , Receptor alfa de Ácido Retinoico/metabolismo , Receptor alfa X Retinoide/metabolismo , Animales , Aorta/citología , Aorta/fisiología , Aterosclerosis/fisiopatología , Células Cultivadas , Regulación hacia Abajo , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Factor de Transcripción GATA6/metabolismo , Humanos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Receptor alfa de Ácido Retinoico/genética , Receptor alfa X Retinoide/genética , Transducción de Señal/genética , Estrés Mecánico , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
3.
J Biomed Sci ; 25(1): 1, 2018 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-29295709

RESUMEN

BACKGROUND: Atherosclerosis occurs in arterial curvatures and branches, where the flow is disturbed with low and oscillatory shear stress (OSS). The remodeling and alterations of extracellular matrices (ECMs) and their composition is the critical step in atherogenesis. In this study, we investigated the effects of different ECM proteins on the regulation of mechanotransduction in vascular endothelial cells (ECs) in response to OSS. METHODS: Through the experiments ranging from in vitro cell culture studies on effects of OSS on molecular signaling to in vivo examinations on clinical specimens from patients with coronary artery disease (CAD), we elucidated the roles of integrins and different ECMs, i.e., fibronectin (FN) and laminin (LM), in transforming growth factor (TGF)-ß receptor (TßR)-mediated Smad2 activation and nuclear factor-κB (NF-κB) signaling in ECs in response to OSS and hence atherogenesis. RESULTS: OSS at 0.5±12 dynes/cm2 induces sustained increases in the association of types I and II TßRs with ß1 and ß3 integrins in ECs grown on FN, but it only transient increases in ECs grown on LM. OSS induces a sustained activation of Smad2 in ECs on FN, but only a transient activation of Smad2 in ECs on LM. OSS-activation of Smad2 in ECs on FN regulates downstream NF-κB signaling and pro-inflammatory gene expression through the activation of ß1 integrin and its association with TßRs. In contrast, OSS induces transient activations of ß1 and ß3 integrins in ECs on LM, which associate with type I TßR to regulate Smad2 phosphorylation, resulting in transient induction of NF-κB and pro-inflammatory gene expression. In vivo investigations on diseased human coronary arteries from CAD patients revealed that Smad2 is highly activated in ECs of atherosclerotic lesions, which is accompanied by the concomitant increase of FN rather than LM in the EC layer and neointimal region of atherosclerotic lesions. CONCLUSIONS: Our findings provide new insights into the mechanisms of how OSS regulates Smad2 signaling and pro-inflammatory genes through the complex signaling networks of integrins, TßRs, and ECMs, thus illustrating the molecular basis of regional pro-inflammatory activation within disturbed flow regions in the arterial tree.


Asunto(s)
Células Endoteliales/fisiología , Mecanotransducción Celular , Proteína Smad2/genética , Fenómenos Biomecánicos , Células Cultivadas , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Humanos , Laminina/genética , Laminina/metabolismo , Proteína Smad2/metabolismo , Estrés Mecánico
4.
Proc Natl Acad Sci U S A ; 111(5): 1855-60, 2014 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-24449884

RESUMEN

ß-Catenin phosphorylation plays important roles in modulating its functions, but the effects of different phosphorylated forms of ß-catenin in response to heterocellular interaction are unclear. Here we investigated whether distinct modes of phosphorylation on ß-catenin could be triggered through heterocellular interactions between endothelial cells (ECs) and smooth muscle cells (SMCs), and the consequent modulation of EC functions. ECs were cocultured with SMCs to initiate direct contact and paracrine interaction. EC-SMC coculture induced EC ß-catenin phosphorylations simultaneously at tyrosine 142 (Tyr142) and serine 45/threonine 41 (Ser45/Thr41) at the cytoplasm/nuclei and the membrane, respectively. Treating ECs with SMC-conditional medium induced ß-catenin phosphorylation only at Ser45/Thr41. These findings indicate that different phosphorylation effects of EC-SMC coculture were induced through heterocellular direct contact and paracrine effects, respectively. Using specific blocking peptides, antagonists, and siRNAs, we found that the ß-catenin Tyr142-phosphorylation was mediated by connexin 43/Fer and that the ß-catenin Ser45/Thr41-phosphorylation was mediated by SMC-released bone morphogenetic proteins through VE-cadherin and bone morphogenetic protein receptor-II/Smad5. Transfecting ECs with ß-catenin-Tyr142 or -Ser45 mutants showed that these two phosphorylated forms of ß-catenin modulate differential EC function: The Tyr142-phosphorylated ß-catenin stimulates vascular cell-adhesion molecule-1 expression to increase EC-monocytic adhesion, but the Ser45/Thr41-phosphorylated ß-catenin attenuates VE-cadherin-dependent junction structures to increase EC permeability. Our findings provide new insights into the understanding of regulatory complexities of distinct modes of ß-catenin phosphorylations under EC-SMC interactions and suggest that different phosphorylated forms of ß-catenin play important roles in modulating vascular pathophysiology through different heterocellular interactions.


Asunto(s)
Comunicación Celular , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Miocitos del Músculo Liso/citología , beta Catenina/metabolismo , Animales , Antígenos CD/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Cadherinas/metabolismo , Bovinos , Adhesión Celular , Compartimento Celular , Permeabilidad de la Membrana Celular , Conexina 43/metabolismo , Modelos Biológicos , Monocitos/citología , Monocitos/metabolismo , Miocitos del Músculo Liso/metabolismo , Comunicación Paracrina , Fosforilación , Fosfotreonina/metabolismo , Fosfotirosina/metabolismo , Unión Proteica , Proteínas Tirosina Quinasas/metabolismo , Proteolisis , Proteína Smad5/metabolismo , Ubiquitina/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
5.
Proc Natl Acad Sci U S A ; 109(20): 7770-5, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22550179

RESUMEN

Vascular endothelial cells (ECs) are constantly exposed to blood flow-induced shear stress, but the mechanism of force-specific activation of their signaling to modulate cellular function remains unclear. We have demonstrated that bone morphogenetic protein receptor (BMPR)-specific Smad1/5 can be force-specifically activated by oscillatory shear stress (OSS) in ECs to cause cell cycle progression. Smad1/5 is highly activated in ECs of atherosclerotic lesions in diseased human coronary arteries from patients with end-stage heart failure undergoing heart transplantation and from apolipoprotein E-deficient mice. Application of OSS (0.5 ± 4 dyn/cm(2)) causes the sustained activation of Smad1/5 in ECs through activations of mammalian target of rapamycin and p70S6 kinase, leading to up-regulation of cyclin A and down-regulations of p21(CIP1) and p27(KIP1) and, hence, EC cycle progression. En face examination of rat aortas reveals high levels of phospho-Smad1/5 in ECs of the inner, but not the outer, curvature of aortic arch, nor the straight segment of thoracic aorta [corrected]. Immunohistochemical and en face examinations of the experimentally stenosed abdominal aorta in rats show high levels of phospho-Smad1/5 in ECs at poststenotic sites, where OSS occurs. These OSS activations of EC Smad1/5 in vitro and in vivo are not inhibited by the BMP-specific antagonist Noggin and, hence, are independent of BMP ligand. Transfecting ECs with Smad1/5-specific small interfering RNAs inhibits the OSS-induced EC cycle progression. Our findings demonstrate the force-specificity of the activation of Smad1/5 and its contribution to cell cycle progression in ECs induced by disturbed flow.


Asunto(s)
Aterosclerosis/fisiopatología , Ciclo Celular/fisiología , Células Endoteliales/fisiología , Regulación de la Expresión Génica/fisiología , Flujo Sanguíneo Regional/fisiología , Proteína Smad1/metabolismo , Estrés Mecánico , Animales , Aorta Abdominal/citología , Aorta Abdominal/patología , Apolipoproteínas E/genética , Fenómenos Biomecánicos , Vasos Coronarios/citología , Vasos Coronarios/patología , Ciclina A/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Ratas , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Taiwán
6.
Biochim Biophys Acta ; 1833(12): 3124-3133, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24021264

RESUMEN

Mechanical forces induced by interstitial fluid flow in and surrounding tissues and by blood/lymphatic flow in vessels may modulate cancer cell invasion and metastasis and anticancer drug delivery. Our previous study demonstrated that laminar flow-induced shear stress induces G2/M arrest in tumor cells. However, whether shear stress modulates final cell fate remains unclear. In this study, we investigated the role of flow-induced shear stress in modulating the survival of four human tumor cell lines, i.e., Hep3B hepatocarcinoma cells, MG63 osteosarcoma cells, SCC25 oral squamous carcinoma cells, and A549 carcinomic alveolar basal epithelial cells. Laminar shear stress (LSS) ranging from 0.5 to 12dyn/cm(2) induced death of these four tumor cell lines. In contrast to LSS at 0.5dyn/cm(2), oscillatory shear stress (OSS) at 0.5±4dyn/cm(2) cannot induce cancer cell death. Both LSS and OSS had no effect on human normal hepatocyte, lung epithelial, and endothelial cells. Application of LSS to these four cell lines increased the percentage of cells stained positively for annexin V-FITC, with up-regulations of cleaved caspase-8, -9, and -3, and PARP. In addition, LSS also induced Hep3B cell autophagy, as detected by acidic vesicular organelle formation, LC3B transformation, and p62/SQSTM1 degradation. By transfecting with small interfering RNA, we found that the shear-induced apoptosis and autophagy are mediated by bone morphogenetic protein receptor type (BMPR)-IB, BMPR-specific Smad1 and Smad5, and p38 mitogen-activated protein kinase in Hep3B cells. Our findings provide insights into the molecular mechanisms by which shear stress induces apoptosis and autophagy in tumor cells.


Asunto(s)
Apoptosis , Autofagia , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Neoplasias/patología , Proteínas Smad/metabolismo , Estrés Mecánico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Línea Celular Tumoral , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Modelos Biológicos , Neoplasias/enzimología , Transducción de Señal , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo
7.
Biomaterials ; 309: 122600, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38718614

RESUMEN

Engineering vascularized tissues remains a promising approach for treating ischemic cardiovascular diseases. The availability of 3D-bioprinted vascular grafts that induce therapeutic angiogenesis can help avoid necrosis and excision of ischemic tissues. Here, using a combination of living cells and biodegradable hydrogels, we fabricated 3D-printed biocompatible proangiogenic patches from endothelial cell-laden photo-crosslinked gelatin (EC-PCG) bioink and smooth muscle cell-encapsulated polyurethane (SMC-PU) bioink. Implantation of 3D-bioprinted proangiogenic patches in a mouse model showed that EC-PCG served as an angiogenic capillary bed, whereas patterned SMC-PU increased the density of microvessels. Moreover, the assembled patterns between EC-PCG and SMC-PU induced the geometrically guided generation of microvessels with blood perfusion. In a rodent model of hindlimb ischemia, the vascular patches rescued blood flow to distal tissues, prevented toe/foot necrosis, promoted muscle remodeling, and increased the capillary density, thereby improving the heat-escape behavior of ischemic animals. Thus, our 3D-printed vascular cell-laden bioinks constitute efficient and scalable biomaterials that facilitate the engineering of vascular patches capable of directing therapeutic angiogenesis for treating ischemic vascular diseases.


Asunto(s)
Gelatina , Hidrogeles , Isquemia , Neovascularización Fisiológica , Poliuretanos , Impresión Tridimensional , Animales , Gelatina/química , Poliuretanos/química , Hidrogeles/química , Isquemia/terapia , Neovascularización Fisiológica/efectos de los fármacos , Ratones , Humanos , Miocitos del Músculo Liso/citología , Reactivos de Enlaces Cruzados/química , Células Endoteliales de la Vena Umbilical Humana , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Masculino , Ingeniería de Tejidos/métodos , Bioimpresión/métodos
8.
Proc Natl Acad Sci U S A ; 105(10): 3927-32, 2008 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-18310319

RESUMEN

Interstitial flow in and around tumor tissue affects the mechanical microenvironment to modulate tumor cell growth and metastasis. We investigated the roles of flow-induced shear stress in modulating cell cycle distribution in four tumor cell lines and the underlying mechanisms. In all four cell lines, incubation under static conditions for 24 or 48 h led to G(0)/G(1) arrest; in contrast, shear stress (12 dynes/cm(2)) induced G(2)/M arrest. The molecular basis of the shear effect was analyzed, and the presentation on molecular mechanism is focused on human MG63 osteosarcoma cells. Shear stress induced increased expressions of cyclin B1 and p21(CIP1) and decreased expressions of cyclins A, D1, and E, cyclin-dependent protein kinases (Cdk)-1, -2, -4, and -6, and p27(KIP1) as well as a decrease in Cdk1 activity. Using specific antibodies and small interfering RNA, we found that the shear-induced G(2)/M arrest and corresponding changes in G(2)/M regulatory protein expression and activity were mediated by alpha(v)beta(3) and beta(1) integrins through bone morphogenetic protein receptor type IA-specific Smad1 and Smad5. Shear stress also down-regulated runt-related transcription factor 2 (Runx2) binding activity and osteocalcin and alkaline phosphatase expressions in MG63 cells; these responses were mediated by alpha(v)beta(3) and beta(1) integrins through Smad5. Our findings provide insights into the mechanism by which shear stress induces G(2)/M arrest in tumor cells and inhibits cell differentiation and demonstrate the importance of mechanical microenvironment in modulating molecular signaling, gene expression, cell cycle, and functions in tumor cells.


Asunto(s)
Ciclo Celular , Integrinas/metabolismo , Neoplasias/patología , Proteínas Smad/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Fase G2 , Humanos , Integrina alfaVbeta3/metabolismo , Integrina beta1/metabolismo , Mitosis , Modelos Biológicos , Fosforilación , Unión Proteica , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Estrés Mecánico
9.
Cardiol Res Pract ; 2021: 1452917, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34336268

RESUMEN

BACKGROUND: MicroRNA-10a (miR-10a) inhibits transcriptional factor GATA6 to repress inflammatory GATA6/VCAM-1 signaling, which is regulated by blood flow to affect endothelial function/dysfunction. This study aimed to identify the expression patterns of miR-10a/GATA6/VCAM-1 in vivo and study their implications in the pathophysiology of human coronary artery disease (CAD), i.e., atherosclerosis. METHODS: Human atherosclerotic coronary arteries and nondiseased arteries were used to detect the expressions of miR-10a/GATA6/VCAM-1 in pathogenic vs. normal conditions. In addition, sera from CAD patients and healthy subjects were collected to detect the level of circulating miR-10a. RESULTS: The comparison of human atherosclerotic coronary arteries with nondiseased arteries demonstrated that lower levels of endothelial miR-10a are related to human atherogenesis. Moreover, GATA6/VCAM-1 (a downstream target of miR-10a) was highly expressed in the endothelium, accompanied by the reduced levels of miR-10a during the development of human atherosclerosis. In addition, CAD patients had a significantly lower concentration of miR-10a in their serum compared to healthy subjects. CONCLUSIONS: Our findings suggest that low miR-10a and high GATA6/VCAM-1 in the cardiovascular endothelium correlates to the development of human atherosclerotic lesions, suggesting that miR-10a signaling has the potential to be developed as a biomarker for human atherosclerosis.

10.
Front Cell Dev Biol ; 9: 697539, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34262908

RESUMEN

BACKGROUND: Pathophysiological vascular remodeling in response to disturbed flow with low and oscillatory shear stress (OSS) plays important roles in atherosclerosis progression. Pomegranate extraction (PE) was reported having anti-atherogenic effects. However, whether it can exert a beneficial effect against disturbed flow-induced pathophysiological vascular remodeling to inhibit atherosclerosis remains unclear. The present study aims at investigating the anti-atherogenic effects of pomegranate peel polyphenols (PPP) extraction and its purified compound punicalagin (PU), as well as their protective effects on disturbed flow-induced vascular dysfunction and their underlying molecular mechanisms. METHODS: The anti-atherogenic effects of PPP/PU were examined on low-density lipoprotein receptor knockout mice fed with a high fat diet. The vaso-protective effects of PPP/PU were examined in rat aortas using myograph assay. A combination of in vivo experiments on rats and in vitro flow system with human endothelial cells (ECs) was used to investigate the pharmacological actions of PPP/PU on EC dysfunction induced by disturbed flow. In addition, the effects of PPP/PU on vascular smooth muscle cell (VSMC) dysfunction were also examined. RESULTS: PU is the effective component in PPP against atherosclerosis. PPP/PU evoked endothelium-dependent relaxation in rat aortas. PPP/PU inhibited the activation of Smad1/5 in the EC layers at post-stenotic regions of rat aortas exposed to disturbed flow with OSS. PPP/PU suppressed OSS-induced expression of cell cycle regulatory and pro-inflammatory genes in ECs. Moreover, PPP/PU inhibited inflammation-induced VSMC dysfunction. CONCLUSION: PPP/PU protect against OSS-induced vascular remodeling through inhibiting force-specific activation of Smad1/5 in ECs and this mechanism contributes to their anti-atherogenic effects.

11.
Biomaterials ; 28(7): 1355-66, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17126899

RESUMEN

Chitooligosaccharides (COS) have been shown to regulate various cellular and biological functions. However, the effect of COS on inflammatory responses of the cells remains unclear. We investigated the regulatory effect of highly N-acetylated COS (NACOS) on tumor necrosis factor-alpha (TNF-alpha)-induced endothelial cell (EC) E-selectin expression, which is crucial for leukocyte recruitment. ECs were kept as controls or pre-treated with NACOS for different times, and then stimulated with TNF-alpha for 4h. The results show that pre-treating ECs with NACOS inhibited the TNF-alpha-induced E-selectin expression in a dose- and time-dependent manner. This NACOS-mediated inhibition in E-selectin expression was regulated at the transcriptional level, but not due to changes in mRNA stability. Stimulation of ECs with TNF-alpha-induced rapid increases in the phosphorylation of their mitogen-activated protein kinases (MAPKs) [extracellular signal-regulated kinase (ERK), c-Jun-NH2-terminal kinase (JNK), and p38 MAPK]; the inhibitor for JNK (i.e., SP600125), but not those for ERK (i.e., PD98059) and p38 MAPK (i.e., SB203580), attenuated this TNF-alpha-induced E-selectin expression. Pre-treating ECs with NACOS inhibited the TNF-alpha-induced JNK activation, suggesting that JNK was involved in the inhibitory effect of NACOS on TNF-alpha-induced E-selectin expression. Pre-treating ECs with NACOS inhibited the TNF-alpha-induced p65 and p50 mRNA expressions. Gel shifting and chromatin immunoprecipitation assays showed that NACOS blocked the TNF-alpha-induced increases in the binding activity and in vivo promoter binding of nuclear factor-kappaB (NF-kappaB) in ECs. Our findings provide a molecular mechanism by which NACOS inhibit TNF-alpha-induced E-selectin expression in ECs, and a basis for using NACOS in pharmaceutical therapy against inflammation.


Asunto(s)
Selectina E/genética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Oligosacáridos/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Secuencia de Bases , Materiales Biocompatibles , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , ADN/genética , Células Endoteliales/citología , Expresión Génica/efectos de los fármacos , Humanos , Ensayo de Materiales , Oligosacáridos/química , Oligosacáridos/toxicidad , Fosforilación , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células U937
12.
Arterioscler Thromb Vasc Biol ; 25(5): 963-9, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15718492

RESUMEN

OBJECTIVES: Vascular endothelial cells (ECs) are influenced by shear stress and neighboring smooth muscle cells (SMCs). We investigated the inflammation-relevant gene expression in EC/SMC cocultures under static condition and in response to shear stress. MATERIALS AND METHODS: Under static condition, DNA microarrays and reverse-transcription polymerase chain reaction identified 23 inflammation-relevant genes in ECs whose expression was significantly affected by coculture with SMCs, with 18 upregulated and 5 downregulated. Application of shear stress (12 dynes/cm2) to the EC side of the coculture for 6 hours inhibited most of the proinflammatory gene expressions in ECs induced by coculture with SMCs. Inhibition of nuclear factor-kappaB (NF-kappaB) activation by the p65-antisense, lactacystin, and N-acetyl-cysteine blocked the coculture-induced EC expression of proinflammatory genes, indicating that the NF-kappaB binding sites in the promoters of these genes play a significant role in their expression as a result of coculture with SMCs. Chromatin immunoprecipitation assays demonstrated the in vivo regulation of NF-kappaB recruitment to selected target promoters. Shear stress inhibited the SMC coculture-induced NF-kappaB activation in ECs and monocytic THP-1 cell adhesion to ECs. CONCLUSIONS: Our findings suggest that shear stress plays an inhibitory role in the proinflammatory gene expression in ECs located in close proximity to SMCs.


Asunto(s)
Endotelio Vascular/fisiología , Músculo Liso Vascular/fisiología , Subunidad p50 de NF-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo , Vasculitis/fisiopatología , Adhesión Celular/inmunología , Comunicación Celular/fisiología , Células Cultivadas , Quimiocina CCL2/genética , Cromatina/fisiología , Técnicas de Cocultivo , Endotelio Vascular/citología , Regulación de la Expresión Génica/inmunología , Humanos , Inmunoprecipitación , Molécula 1 de Adhesión Intercelular/genética , Monocitos/citología , Músculo Liso Vascular/citología , Subunidad p50 de NF-kappa B/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/análisis , Estrés Mecánico , Factor de Transcripción ReIA/genética , Vasculitis/genética , Vasculitis/inmunología
13.
Arterioscler Thromb Vasc Biol ; 24(1): 73-9, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14615388

RESUMEN

OBJECTIVE: Vascular endothelial cells (ECs) are subjected to shear stress and cytokine stimulation. We studied the interplay between shear stress and cytokine in modulating the expression of adhesion molecule genes in ECs. METHODS AND RESULTS: Shear stress (20 dynes/cm2) was applied to ECs prior to and/or following the addition of tumor necrosis factor (TNF)-alpha. Shear stress increased the TNF-alpha-induced expression of intercellular adhesion molecule-1 (ICAM-1) at both mRNA and surface protein levels, but decreased the TNF-alpha-induced expression of vascular adhesion molecule-1 (VCAM-1) and E-selectin. Transfection studies using promoter reporter gene constructs of ICAM-1, VCAM-1, and E-selectin demonstrated that these shear stress modulations of gene expression occur at the transcriptional levels. After 24-hour preshearing followed by 1 hour of static incubation, the effect of preshearing on TNF-alpha-induced ICAM-1 mRNA expression vanished. The recovery of the TNF-alpha-induced VCAM-1 and E-selectin mRNA expressions following preshearing, however, required a static incubation time of >6 hours (complete recovery at 24 hours). Pre- and postshearing caused a reduction in the nuclear factor-kappaB-DNA binding activity induced by TNF-alpha in the EC nucleus. CONCLUSIONS: Our findings suggest that shear stress plays differential roles in modulating the TNF-alpha-induced expressions of ICAM-1 versus VCAM-1 and E-selectin genes in ECs.


Asunto(s)
Selectina E/biosíntesis , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Regulación de la Expresión Génica , Molécula 1 de Adhesión Intercelular/biosíntesis , Estrés Mecánico , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Núcleo Celular/metabolismo , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , ADN/genética , ADN/metabolismo , Selectina E/genética , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Molécula 1 de Adhesión Intercelular/genética , FN-kappa B/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética
14.
Microb Drug Resist ; 21(6): 610-21, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26167865

RESUMEN

A growing tendency for community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) to be involved in nosocomial infections was reported. The predominance of SCCmec type IV or V CA-MRSA in soft tissue infection has also been indicated in Northern Taiwan. To establish basic information about the molecular characteristics of MRSA in our region, a total of 102 MRSA isolates were collected and characterized by an array of typing methods. Healthcare-associated MRSA (HA-MRSA) were found to be more resistant to levofloxacin (p=0.016) and moxifloxacin (p=0.015) than CA-MRSA. However, no difference was found in each and overall SCCmec type distribution between the two MRSA groups. Type I (8.7% vs. 2.6%) was more frequently found in CA-MRSA, whereas type V was more often observed in HA-MRSA (24.4% vs. 8.7%). No difference was found in the dichotomous group of PVL, SCCmec type IV, V, and IV/V between the two MRSA groups. Twenty-seven distinct spa types were identified; t437 and t1081 were the predominant types in our isolates. Moreover, 12 novel spa types with extremely low global frequency were detected in our isolates. SCCmec type III and IV were the major subtypes in the MRSA we collected. The t1081 clones all belonged to HA-MRSA and mostly to SCCmec type V (71.4%). CA-MRSA t437 clones were mostly SCCmec type IV strains (71.4%), but HA-MRSA t437 clones were predominantly SCCmec type IV (42.1%) and III (36.8%). Our findings support a difference in the molecular characteristics of CA-MRSA and HA-MRSA that may reflect various clonal origins in our isolates.


Asunto(s)
Antibacterianos/farmacología , Infección Hospitalaria/epidemiología , Farmacorresistencia Bacteriana Múltiple/genética , Regulación Bacteriana de la Expresión Génica , Staphylococcus aureus Resistente a Meticilina/genética , Infecciones Estafilocócicas/epidemiología , Técnicas de Tipificación Bacteriana , Secuencia de Bases , Infecciones Comunitarias Adquiridas , Infección Hospitalaria/tratamiento farmacológico , Infección Hospitalaria/microbiología , Fluoroquinolonas/farmacología , Humanos , Levofloxacino/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Pruebas de Sensibilidad Microbiana , Epidemiología Molecular , Datos de Secuencia Molecular , Moxifloxacino , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Taiwán/epidemiología
15.
J Biomech ; 37(4): 531-9, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14996565

RESUMEN

Vascular endothelial cells (ECs) are constantly subjected to blood flow-induced shear stress and the influences of neighboring smooth muscle cells (SMCs). In the present study, a coculture flow system was developed to study the effect of shear stress on EC-SMC interactions. ECs and SMCs were separated by a porous membrane with only the EC side subjected to the flow condition. When ECs were exposed to a shear stress of 12 dynes/cm2 for 24 h, the cocultured SMCs tended to orient perpendicularly to the flow direction. This perpendicular orientation of the cocultured SMCs to flow direction was not observed when ECs were exposed to a shear stress of 2 dynes/cm2. Under the static condition, long and parallel actin bundles were observed in the central regions of the cocultured SMCs, whereas the actin filaments localized mainly at the periphery of the cocultured ECs. After 24 h of flow application, the cocultured ECs displayed very long, well-organized, parallel actin stress fibers aligned with the flow direction in the central regions of the cells. Immunostaining of platelet endothelial cell adhesion molecule-1 confirmed the elongation and alignment of the cocultured ECs with the flow direction. Coculture with SMCs under static condition induced EC gene expressions of growth-related oncogene-alpha and monocyte chemotactic protein-1, and shear stress was found to abolish these SMC-induced gene expressions. Our results suggest that shear stress may serve as a down-regulator for the pathophysiologically relevant gene expression in ECs cocultured with SMCs.


Asunto(s)
Células Endoteliales/fisiología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , Citoesqueleto de Actina/fisiología , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/genética , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Humanos , Técnicas Inmunológicas , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Oncogenes/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Coloración y Etiquetado , Estrés Mecánico , Factores de Tiempo
16.
J Biomech ; 36(12): 1883-95, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14614942

RESUMEN

The preferential adhesion of monocytes to vascular endothelial cells (ECs) at regions near branches and curvatures of the arterial tree, where flow is disturbed, suggests that hemodynamic conditions play significant roles in monocyte adhesion. The present study aims to elucidate the effects of disturbed flow on monocyte adhesion to ECs and the adhesive properties of ECs. We applied, for the first time, the micron-resolution particle image velocimetry (microPIV) technique to analyze the characteristics of the disturbed flow produced in our vertical-step flow (VSF) chamber. The results demonstrated the existence of a higher near-wall concentration and a longer residence time of the monocytic analog THP-1 cells near the step and the reattachment point. THP-1 cells showed prominent adhesion to ECs pretreated with TNFalpha in the regions near the step and the reattachment point, but they showed virtually no adhesion to un-stimulated ECs. Pre-incubation of the TNFalpha-treated ECs with antibodies against intercellular adhesion molecule-1 (ICAM-1), vascular adhesion molecule-1 (VCAM-1), and E-selectin inhibited the THP-1 adhesion; the maximal inhibition was observed with a combination of these antibodies. Pre-exposure of ECs to disturbed flow in VSF for 24 h led to significant increases in their surface expressions of ICAM-1 and E-selectin, but not VCAM-1, and in the adhesion of THP-1 cells. Our findings demonstrate the importance of complex flow environment in modulating the adhesive properties of vascular endothelium and consequently monocyte adhesion in regions of prevalence of atherosclerotic lesions.


Asunto(s)
Movimiento Celular/fisiología , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Citometría de Flujo/métodos , Microfluídica/métodos , Monocitos/citología , Monocitos/fisiología , Reología/métodos , Velocidad del Flujo Sanguíneo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Técnicas de Cultivo de Célula/métodos , Movimiento Celular/efectos de los fármacos , Selectina E , Endotelio Vascular/efectos de los fármacos , Citometría de Flujo/instrumentación , Hemorreología/instrumentación , Hemorreología/métodos , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Microfluídica/instrumentación , Reología/instrumentación , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/metabolismo
17.
Chin J Physiol ; 45(4): 169-76, 2002 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-12817708

RESUMEN

The interplay between shear stress and cytokines in regulating vascular endothelial function remains largely unexplored. In the present study, the potential role of shear stress in regulating tumor necrosis factor-alpha (TNF-alpha)-induced gene expression in endothelial cells (ECs) was investigated. The TNF-alpha-induced monocyte chemotactic protein-1 (MCP-1) mRNA expressions were significantly attenuated in ECs subjected to a high level of shear stress (20 dynes/cm2) for 4 or 24 h prior to the addition of TNF-alpha in the presence of flow. Less inhibition of TNF-alpha-induced MCP-1 mRNA expression was found in ECs pre-exposed to a low level of shear stress (1.2 dynes/cm2) for 24 h as compared with the cells presheared (pre-exposed to shear stress) for 4 h. Simultaneous exposure of ECs to TNF-alpha and a high or low level of shear stress down-regulated TNF-alpha-induced MCP-1 gene expressions, suggesting that the post-flow condition modulates endothelial responses to cytokine stimulation. Individually or combined, an endothelial nitric oxide synthase (eNOS) inhibitor and a glutathione (GSH) biosynthesis inhibitor had no effect on this shear stress-mediated inhibition. Moreover, in ECs either presheared or remained in a static condition prior to stimulation by TNF-alpha while under shear flow, the ability of TNF-alpha to induce AP-1-DNA binding activity in the nucleus was reduced. Our findings suggest that shear stress plays a protective role in vascular homeostasis by inhibiting endothelial responses to cytokine stimulation.


Asunto(s)
Antineoplásicos/farmacología , Quimiocina CCL2/genética , Endotelio Vascular/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Núcleo Celular/metabolismo , Endotelio Vascular/citología , Inhibidores Enzimáticos/farmacología , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Glutatión/metabolismo , Humanos , Monocitos/fisiología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo III , Oxidación-Reducción , Estrés Mecánico , Factor de Transcripción AP-1/metabolismo , Cordón Umbilical/citología , omega-N-Metilarginina/farmacología
18.
J Mol Cell Biol ; 6(6): 458-72, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25548371

RESUMEN

T-lymphocyte migration under flow is critical for immune responses, but the mechanisms by which flow modulates the migratory behaviors of T-lymphocytes remain unclear. Human peripheral blood T-lymphocytes (PBTLs), when stimulated with phorbol 12-myristate 13-acetate (PMA), stretched their cell bodies dramatically and moved along the flow direction. In contrast, stromal cell-derived factor-1α-stimulated PBTLs deformed and migrated in a random manner. Here we elucidated the molecular mechanisms underlying flow-induced directionality and deformation of PMA-stimulated PBTLs. PMA primed PBTLs for polarization under flow, with protein kinase C (PKC)-δ enriched in the leading edge, PKC-ßI in the microtubule organizing center, and PKC-ßII in the uropod and peripheral region. PKC-δ regulated cell protrusions in the leading edge through Tiam1/Rac1/calmodulin, whereas PKC-ß regulated RhoA/Rho-associated kinase activity and microtubule stability to modulate uropod contractility and detachment. Our findings indicate that PKC-δ and -ß coordinate in the cell leading edge and uropod, respectively, to modulate the directionality and deformability of migratory T-lymphocytes under flow.


Asunto(s)
Movimiento Celular/fisiología , Proteína Quinasa C beta/metabolismo , Proteína Quinasa C-delta/metabolismo , Linfocitos T/enzimología , Carcinógenos/farmacología , Movimiento Celular/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Linfocitos T/citología , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Acetato de Tetradecanoilforbol/farmacología , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
19.
J Hazard Mater ; 277: 3-12, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-24731914

RESUMEN

Zinc oxide nanoparticles (ZnO NPs) exhibit novel physiochemical properties and have found increasing use in sunscreen products and cosmetics. The potential toxicity is of increasing concern due to their close association with human skin. A time-of-flight secondary ion mass spectrometry (TOF-SIMS) and confocal laser scanning microscopy (CLSM) imaging method was developed and validated for rapid and sensitive cytotoxicity study of ZnO NPs using human skin equivalent HaCaT cells as a model system. Assorted material, chemical, and toxicological analysis methods were used to confirm their shape, size, crystalline structure, and aggregation properties as well as dissolution behavior and effect on HaCaT cell viability in the presence of various concentrations of ZnO NPs in aqueous media. Comparative and correlative analyses of aforementioned results with TOF-SIMS and CLSM imaging results exhibit reasonable and acceptable outcome. A marked drop in survival rate was observed with 50µg/ml ZnO NPs. The CLSM images reveal the absorption and localization of ZnO NPs in cytoplasm and nuclei. The TOF-SIMS images demonstrate elevated levels of intracellular ZnO concentration and associated Zn concentration-dependent (40)Ca/(39)K ratio, presumably caused by the dissolution behavior of ZnO NPs. Additional validation by using stable isotope-labeled (68)ZnO NPs as tracers under the same experimental conditions yields similar cytotoxicity effect. The imaging results demonstrate spatially-resolved cytotoxicity relationship between intracellular ZnO NPs, (40)Ca/(39)K ratio, phosphocholine fragments, and glutathione fragments. The trend of change in TOF-SIMS spectra and images of ZnO NPs treated HaCaT cells demonstrate the possible mode of actions by ZnO NP involves cell membrane disruption, cytotoxic response, and ROS mediated apoptosis.


Asunto(s)
Portadores de Fármacos/química , Microscopía Confocal/métodos , Nanopartículas/química , Piel/efectos de los fármacos , Espectrometría de Masa de Ion Secundario/métodos , Óxido de Zinc/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Microscopía Confocal/instrumentación , Reproducibilidad de los Resultados , Piel/citología , Solubilidad , Espectrometría de Masa de Ion Secundario/instrumentación , Propiedades de Superficie , Pruebas de Toxicidad/instrumentación , Pruebas de Toxicidad/métodos , Isótopos de Zinc , Óxido de Zinc/administración & dosificación
20.
Cardiovasc Res ; 96(2): 296-307, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22865639

RESUMEN

AIMS: The implication of circulating haematopoietic CD34(+) progenitors in the vasculature is unclear due to the lack of understanding of their characteristics and plasticity mediated by their cellular microenvironment. We investigated how vascular smooth muscle cells (SMCs) and their interactions with endothelial cells (ECs) affect the behaviour and plasticity of CD34(+)CD31(+) progenitors and the underlying mechanisms. METHODS AND RESULTS: Human peripheral blood-derived CD34(+)CD31(+) cells were directly transplanted into injured arteries in vivo and co-cultured with ECs and SMCs in vitro. CD34(+)CD31(+) progenitors injected into wire-injured mouse arteries differentiate into ECs and macrophages in the neoendothelial layer and neointima, respectively. SMC-co-culture increases CD34(+)CD31(+) cell mobility and adhesion to and transmigration across ECs. Sorted CD34(+)CD31(+) progenitors that adhered to ECs co-cultured with SMCs have the capacity to form capillary-like structures in Matrigel and chimeric blood vessels in vivo. Sorted transmigrated progenitors give rise to macrophages with increased pro-angiogenic activity. These differentiations of CD34(+)CD31(+) progenitors into ECs and macrophages are mediated by ß(2)-integrin and Notch-1, respectively. ß(2)-Integrin and Notch-1 are activated by their counterligands, intercellular adhesion molecule-1 (ICAM-1) and jagged-1, which are highly expressed in the neoendothelium and neointima in injured arteries. Intra-arterial injection of ß(2)-integrin-activated CD34(+)CD31(+) progenitors into wire-injured mouse arteries inhibits neointima formation. CONCLUSION: Our findings indicate that the peripheral vascular niches composed of ECs and SMCs may predispose haematopoietic CD34(+)CD31(+) progenitors to differentiate into ECs and macrophages through the activations of the ICAM-1/ß(2)-integrin and jagged-1/Notch-1 cascades, respectively.


Asunto(s)
Antígenos CD18/metabolismo , Células Endoteliales/fisiología , Células Madre Hematopoyéticas/fisiología , Miocitos del Músculo Liso/fisiología , Receptor Notch1/metabolismo , Animales , Apolipoproteínas E/genética , Proteínas de Unión al Calcio/metabolismo , Adhesión Celular , Movimiento Celular , Células Endoteliales/citología , Endotelio Vascular/fisiología , Arteria Femoral/lesiones , Células Endoteliales de la Vena Umbilical Humana , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Macrófagos/citología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones SCID , Músculo Liso Vascular/fisiología , Neointima/prevención & control , Neovascularización Fisiológica , Proteínas Serrate-Jagged , Migración Transendotelial y Transepitelial
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA