Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 361
Filtrar
1.
J Cell Mol Med ; 24(18): 10792-10802, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32803867

RESUMEN

Brain-derived neurotrophic factor (BDNF) has been reported to participate in fracture healing, whereas the mechanism is still unclear. Since osteoblast migration is important for fracture healing, investigating effects of BDNF on osteoblasts migration may help to reveal its mechanism. Here, MC3T3-E1 cells were used in vitro while closed femur fracture mice were applied in vivo. Cells migration was assessed with Transwell assay. The protein expression was analysed by immunoblotting. X-ray and Micro-CT were performed at different time after fracture. Our results showed that BDNF promoted MC3T3-E1 cells migration, integrin ß1 expression and ERK1/2 and AKT phosphorylation. K252a, a specific inhibitor for TrkB, suppressed BDNF-induced migration, integrin ß1 expression and activation of ERK1/2 and AKT. PD98059 (an ERK1/2 inhibitor) and LY294002 (an AKT inhibitor) both inhibited BDNF-induced migration and integrin ß1 expression while integrin ß1 blocking antibody only suppressed cell migration. X-ray and Micro-CT analyses showed that the adenoviral carried integrin ß1 shRNA group had slower fracture healing at 7 and 21 days, but not 35 days compared to the control group. Thus, we proposed that BDNF stimulated MC3T3-E1 cells migration by up-regulating integrin ß1 via TrkB mediated ERK1/2 and AKT signalling, and this may help to enhance the fracture healing.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Curación de Fractura/efectos de los fármacos , Integrina beta1/biosíntesis , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Glicoproteínas de Membrana/fisiología , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Línea Celular Transformada , Movimiento Celular/efectos de los fármacos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/análisis , Fracturas del Fémur/diagnóstico por imagen , Fracturas del Fémur/tratamiento farmacológico , Fracturas del Fémur/fisiopatología , Integrina beta1/genética , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/química , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Regulación hacia Arriba/efectos de los fármacos , Microtomografía por Rayos X
2.
Stem Cells ; 37(12): 1528-1541, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31574184

RESUMEN

Nearly 1 in every 120 children born has a congenital heart defect. Although surgical therapy has improved survival, many of these children go on to develop right ventricular heart failure (RVHF). The emergence of cardiovascular regenerative medicine as a potential therapeutic strategy for pediatric HF has provided new avenues for treatment with a focus on repairing or regenerating the diseased myocardium to restore cardiac function. Although primarily tried using adult cells and adult disease models, stem cell therapy is relatively untested in the pediatric population. Here, we investigate the ability of electrical stimulation (ES) to enhance the retention and therapeutic function of pediatric cardiac-derived c-kit+ progenitor cells (CPCs) in an animal model of RVHF. Human CPCs isolated from pediatric patients were exposed to chronic ES and implanted into the RV myocardium of rats. Cardiac function and cellular retention analysis showed electrically stimulated CPCs (ES-CPCs) were retained in the heart at a significantly higher level and longer time than control CPCs and also significantly improved right ventricular functional parameters. ES also induced upregulation of extracellular matrix and adhesion genes and increased in vitro survival and adhesion of cells. Specifically, upregulation of ß1 and ß5 integrins contributed to the increased retention of ES-CPCs. Lastly, we show that ES induces CPCs to release higher levels of pro-reparative factors in vitro. These findings suggest that ES can be used to increase the retention, survival, and therapeutic effect of human c-kit+ progenitor cells and can have implications on a variety of cell-based therapies. Stem Cells 2019;37:1528-1541.


Asunto(s)
Estimulación Eléctrica/métodos , Insuficiencia Cardíaca/terapia , Miocitos Cardíacos/citología , Trasplante de Células Madre/métodos , Función Ventricular Derecha/fisiología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Preescolar , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Cardiopatías Congénitas/cirugía , Humanos , Lactante , Recién Nacido , Integrina beta1/biosíntesis , Masculino , Proteínas Proto-Oncogénicas c-kit/metabolismo , Ratas , Medicina Regenerativa/métodos , Células Madre/citología
3.
Int J Cancer ; 145(3): 830-841, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30719702

RESUMEN

Breast cancer remains the second cause of tumor-related mortality in women worldwide mainly due to chemoresistance and metastasis. The chemoresistance and metastasis are attributed to a rare subpopulation with enriched stem-like characteristics, thus called Cancer Stem Cells (CSCs). We have previously reported aberrant expression of the actin-bundling protein (fascin) in breast cancer cells, which enhances their chemoresistance, metastasis and enriches CSC population. The intracellular mechanisms that link fascin with its downstream effectors are not fully elucidated. Here, loss and gain of function approaches in two different breast cancer models were used to understand how fascin promotes disease progression. Importantly, findings were aligned with expression data from actual breast cancer patients. Expression profiling of a large breast cancer dataset (TCGA, 530 patients) showed statistically significant correlation between fascin expression and a key adherence molecule, ß1 integrin (ITGB1). In vitro manipulation of fascin expression in breast cancer cells exhibited its direct effect on ITGB1 expression. Fascin-mediated regulation of ITGB1 was critical for several breast cancer cell functions including adhesion to different extracellular matrix, self-renewability and chemoresistance. Importantly, there was a significant relationship between fascin and ITGB1 co-expression and short disease-free as well as overall survival in chemo-treated breast cancer patients. This novel role of fascin effect on ITGB1 expression and its outcome on cell self-renewability and chemoresistance strongly encourages for dual targeting of fascin-ITGB1 axis as a therapeutic approach to halt breast cancer progression and eradicate it from the root.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas Portadoras/biosíntesis , Integrina beta1/biosíntesis , Proteínas de Microfilamentos/biosíntesis , Células Madre Neoplásicas/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Adhesión Celular/fisiología , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Expresión Génica , Humanos , Inmunohistoquímica , Integrina beta1/genética , Integrina beta1/metabolismo , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Células Madre Neoplásicas/metabolismo , Análisis de Supervivencia , Regulación hacia Arriba
4.
Biochem Biophys Res Commun ; 515(1): 119-124, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31128921

RESUMEN

Skin epidermis is a stratified epithelium that composed of interfollicular epidermis (IFE) and hair follicles (HFs). Integrins are cell-cell and cell-matrix adhesive ligands that play important roles in epidermal cell proliferation, migration and differentiation behaviors. Here, we analyzed the expression of both α6 and ß1 integrins. In vitro epidermal cell culture, both α6 and ß1 integrins displayed downregulation upon high Ca2+ induced differentiation. During wound healing (WH), α6 integrin showed dynamic expression, first greatly upregulated in unclosed wounds and then downregulated upon re-epithelialization. Further analysis of different wound regions confirmed α6 integrin significantly increased in migratory cells and migration was coupled with differentiation. However, expression level of ß1 integrin did not show significant correlation with migration. We discovered that α6 integrin directly indicates epidermal cell differentiation and wound directed migration behaviors with its expression level.


Asunto(s)
Células Epidérmicas/metabolismo , Epidermis/metabolismo , Folículo Piloso/metabolismo , Integrina alfa6/biosíntesis , Animales , Calcio/metabolismo , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Células Epidérmicas/citología , Femenino , Folículo Piloso/citología , Inmunohistoquímica , Integrina beta1/biosíntesis , Masculino , Ratones Endogámicos C57BL , Cicatrización de Heridas
5.
Biochem Biophys Res Commun ; 514(4): 1115-1121, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31101337

RESUMEN

Cancer cells can invade as a population in various cancer tissues. This phenomenon is called collective invasion, which is associated with the metastatic potential and prognosis of cancer patients. The collectiveness of cancer cells is necessary for collective invasion. However, the mechanism underlying the generation of collectiveness by cancer cells is not well known. In this study, the phenomenon of contact following, where neighboring cells move in the same direction via intercellular adhesion, was investigated. An experimental system was created to observe the two-dimensional invasion using a collagen gel overlay to study contact following in collective invasion. The role of integrin-ß1, one of the major extracellular matrix (ECM) receptors, in contact following was examined through the experimental system. Integrin-ß1 was localized to the intercellular site in squamous carcinoma cells. Moreover, the intercellular adhesion and contact following were suppressed by treatment of an integrin-ß1 inhibitory antibody. ECM proteins such as laminin-332 and type-XVII collagen were also localized to the intercellular site and critical for contact following. Collectively, it was demonstrated that the activity of integrin-ß1 and expression of ECM proteins in the intercellular site promote contact following in the collective invasion of a cancer cell population.


Asunto(s)
Autoantígenos/metabolismo , Carcinoma de Células Escamosas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Integrina beta1/metabolismo , Colágenos no Fibrilares/metabolismo , Neoplasias Cutáneas/metabolismo , Autoantígenos/biosíntesis , Carcinoma de Células Escamosas/patología , Adhesión Celular , Moléculas de Adhesión Celular/biosíntesis , Humanos , Integrina beta1/biosíntesis , Colágenos no Fibrilares/biosíntesis , Neoplasias Cutáneas/patología , Células Tumorales Cultivadas , Kalinina , Colágeno Tipo XVII
6.
Blood ; 128(1): 55-9, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27207790

RESUMEN

Krüppel-like factor 5 (Klf5) encodes a zinc-finger transcription factor and has been reported to be a direct target of C/EBPα, a master transcription factor critical for formation of granulocyte-macrophage progenitors (GMP) and leukemic GMP. Using an in vivo hematopoietic-specific gene ablation model, we demonstrate that loss of Klf5 function leads to a progressive increase in peripheral white blood cells, associated with increasing splenomegaly. Long-term hematopoietic stem cells (HSCs), short-term HSCs (ST-HSCs), and multipotent progenitors (MPPs) were all significantly reduced in Klf5(Δ/Δ) mice, and knockdown of KLF5 in human CD34(+) cells suppressed colony-forming potential. ST-HSCs, MPPs, and total numbers of committed progenitors were increased in the spleen of Klf5(Δ/Δ) mice, and reduced ß1- and ß2-integrin expression on hematopoietic progenitors suggests that increased splenic hematopoiesis results from increased stem and progenitor mobilization. Klf5(Δ/Δ) mice show a significant reduction in the fraction of Gr1(+)Mac1(+) cells (neutrophils) in peripheral blood and bone marrow and increased frequency of eosinophils in the peripheral blood, bone marrow, and lung. Thus, these studies demonstrate dual functions of Klf5 in regulating hematopoietic stem and progenitor proliferation and localization in the bone marrow, as well as lineage choice after GMP, promoting increased neutrophil output at the expense of eosinophil production.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Células Progenitoras de Granulocitos y Macrófagos/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Células Madre Multipotentes/metabolismo , Animales , Antígenos CD18/biosíntesis , Antígenos CD18/genética , Eosinófilos/citología , Eosinófilos/metabolismo , Células Progenitoras de Granulocitos y Macrófagos/citología , Integrina beta1/biosíntesis , Integrina beta1/genética , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Noqueados , Células Madre Multipotentes/citología , Neutrófilos/citología , Neutrófilos/metabolismo
7.
Exp Cell Res ; 353(2): 88-99, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28284838

RESUMEN

In a previous transcriptomic analysis of 63 ocular melanomas of the uvea, we found that expression of the PRL-3/PTP4A3 gene, encoding a phosphatase that is anchored to the plasma membrane, was associated with the risk of metastasis, and a poor prognosis. We also showed that PRL-3 overexpression in OCM-1 ocular melanoma cells significantly increased cell migration in vitro and invasiveness in vivo, suggesting a direct role for PRL-3 in the metastatic spreading of uveal melanoma. Here, we aimed to identify PRL-3 substrates at the plasma membrane involved in adhesion to the extracellular matrix. We focused on integrin ß1, which is the most highly expressed integrin in our cohort of uveal melanomas. We show that preventing PRL-3 anchorage to the plasma membrane i) abolishes PRL-3-induced migration in OCM-1 cells, ii) specifically enhances the spreading of OCM-1 cells overexpressing PRL-3, and iii) favors the maturation of large focal adhesions (FAs) containing integrin ß1 on collagen I. Knockdown experiments confirmed integrin ß1 involvement in PRL3-induced migration. We identified interactions between PRL-3 and integrin ß1, as well as with FAK P-Y397, an auto-activated form of Focal Adhesion Kinase found in FAs. We also show that integrin ß1 may be dephosphorylated by PRL-3 in its intracytoplasmic S/T region, an important motif for integrin-mediated cell adhesion. Finally, we observed that PRL-3 regulated the clustering of integrin ß1 in FAs on collagen I but not on fibronectin. This work identifies PRL-3 as a new regulator of cell adhesion structures to the extracellular matrix, and further supports PRL-3 as a key actor of metastasis in uveal melanoma, of which molecular mechanisms are still poorly understood.


Asunto(s)
Movimiento Celular/genética , Integrina beta1/biosíntesis , Melanoma/genética , Proteínas de Neoplasias/biosíntesis , Proteínas Tirosina Fosfatasas/biosíntesis , Neoplasias de la Úvea/genética , Adhesión Celular/genética , Línea Celular Tumoral , Membrana Celular/genética , Membrana Celular/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/patología , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Integrina beta1/genética , Melanoma/patología , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Proteínas Tirosina Fosfatasas/genética , Neoplasias de la Úvea/patología
8.
J Cell Biochem ; 118(9): 2693-2699, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28112417

RESUMEN

Breast cancer is a complex and multifactorial disease. Tumors have a heterogeneous microenvironment, which have multiple interactions with other cell types, greatly influencing the behavior of tumor cells and response to therapy. The 3D culture mimics the microenvironment better found in vivo and is more appropriated than the traditional 2D culture made from plastic to test the cellular response to drugs. To investigate the effects of [10]-gingerol on breast tumor cells, we used physiologically relevant three-dimensional (3D) cultures of malignant and non-malignant human breast cells grown in laminin-rich extracellular matrix gels (lr-ECM). Our results showed selective cytotoxicity of [10]-gingerol against the malignant T4-2 breast cancer cell line compared to non-malignant S1 cells. The compound reverted the malignant phenotype of the cancer cells, downregulating the expression of epidermal growth factor receptor (EGFR) and ß1-integrin. Moreover, [10]-gingerol induced apoptosis in this cell line. These results suggest that [10]-gingerol may be an effective compound to use as adjuvant therapy in breast cancer treatment. J. Cell. Biochem. 118: 2693-2699, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Alcoholes Grasos/farmacología , Guayacol/análogos & derivados , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Receptores ErbB/biosíntesis , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Guayacol/farmacología , Humanos , Integrina beta1/biosíntesis , Ratones Desnudos , Proteínas de Neoplasias/biosíntesis
9.
Cancer Sci ; 107(7): 916-23, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27094578

RESUMEN

Increasing evidence has revealed that miR-199a-5p is actively involved in tumor invasion and metastasis as well as in the decline of breast cancer tissues. In this research, overexpression of miR-199a-5p weakened motility and invasion of breast cancer cells MCF-7 and MDA-MB-231. Upregulation of Ets-1 increased breast cancer cell invasion, but the mechanism by which miR-199a-5p modulates activation of Ets-1 in breast cancer was not clarified. We investigated the relationship between miR-199a-5p and Ets-1 on the basis of 158 primary breast cancer case specimens, and the results showed that Ets-1 expression was inversely correlated with endogenous miR-199a-5p. Overexpression of miR-199a-5p reduced the mRNA and protein levels of Ets-1 in MCF-7 and MDA-MB-231 cells, whereas anti-miR-199a-5p elevated Ets-1. siRNA-mediated Ets-1 knockdown phenocopied the inhibition invasion of miR-199a-5p in vitro. Moreover, luciferase reporter assay revealed that miR-199a-5p directly targeted 3'-UTR of Ets-1 mRNA. This research revealed that miR-199a-5p could descend the levels of ß1 integrin by targeting 3'-UTR of Ets-1 to alleviate the invasion of breast cancer via FAK/Src/Akt/mTOR signaling pathway. Our results provide insight into the regulation of ß1 integrin through miR-199a-5p-mediated Ets-1 silence and will help in designing new therapeutic strategies to inhibit signal pathways induced by miR-199a-5p in breast cancer invasion.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Integrina beta1/genética , MicroARNs/genética , Invasividad Neoplásica/genética , Proteína Proto-Oncogénica c-ets-1/genética , Proteína Proto-Oncogénica c-ets-1/metabolismo , Regiones no Traducidas 3'/genética , Línea Celular Tumoral , Movimiento Celular/genética , Regulación hacia Abajo/genética , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Integrina beta1/biosíntesis , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteína Proto-Oncogénica c-ets-1/deficiencia , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo , Transcripción Genética/genética , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
10.
Am J Pathol ; 185(5): 1304-23, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25797647

RESUMEN

Cytomegalovirus (CMV) is a prevalent pathogen in intrauterine infections that causes congenital anomalies such as CMV encephalitis, which is characterized by the focal areas of reactive gliosis, reactive mononuclear cells, microglial nodules, and ventriculoencephalitis. To elucidate the mechanisms of CMV susceptibility in the developing brain, cell tropism and the infectious dynamics of CMV infection were investigated. We evaluated intraventricular and intravascular infections from the perspective of the distribution of CMV and its receptor (ß1 integrin) in the earliest phase of infection. Murine CMV (MCMV) immediate early 1-positive cells were colocalized mainly with meninges and choroid plexus (after intraventricular infection) or with endothelial cells and pericytes (after intravascular infection). Using green fluorescent protein-expressing recombinant MCMV particles and fluorescent microbeads (100 to 300 nm), we revealed that CMV particle size is the primary factor determining the initial CMV distribution. ß1 Integrin inhibition using a shRNA and functional blocking antibody significantly reduced MCMV infection. IHC analysis, flow cytometric, and brain slice analyses strongly support that high-level ß1 integrin-expressing cells (eg, endothelial cells, pericytes, meninges, choroid plexus, and neural stem progenitor cells) are the first targets of MCMV. Therefore, our data demonstrate that the initial distributions of MCMV particles and ß1 integrin determine the distinct pattern of infection in the brain in the acute phase.


Asunto(s)
Encefalitis/virología , Infecciones por Herpesviridae/virología , Integrina beta1/biosíntesis , Animales , Animales Recién Nacidos , Western Blotting , Modelos Animales de Enfermedad , Encefalitis/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Infecciones por Herpesviridae/metabolismo , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos ICR , Muromegalovirus/patogenicidad
11.
J Immunol ; 193(4): 1636-44, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25024385

RESUMEN

Lymph node (LN) stromal cells (LNSCs) form the functional structure of LNs and play an important role in lymphocyte survival and the maintenance of immune tolerance. Despite their broad spectrum of function, little is known about LNSC responses during microbial infection. In this study, we demonstrate that LNSC subsets display distinct kinetics following vaccinia virus infection. In particular, compared with the expansion of other LNSC subsets and the total LN cell population, the expansion of fibroblastic reticular cells (FRCs) was delayed and sustained by noncirculating progenitor cells. Notably, newly generated FRCs were preferentially located in perivascular areas. Viral clearance in reactive LNs preceded the onset of FRC expansion, raising the possibility that viral infection in LNs may have a negative impact on the differentiation of FRCs. We also found that MHC class II expression was upregulated in all LNSC subsets until day 10 postinfection. Genetic ablation of radioresistant stromal cell-mediated Ag presentation resulted in slower contraction of Ag-specific CD4(+) T cells. We propose that activated LNSCs acquire enhanced Ag-presentation capacity, serving as an extrinsic brake system for CD4(+) T cell responses. Disrupted function and homeostasis of LNSCs may contribute to immune deregulation in the context of chronic viral infection, autoimmunity, and graft-versus-host disease.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Ganglios Linfáticos/citología , Células del Estroma/inmunología , Virus Vaccinia/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno/inmunología , Antígeno B7-1/biosíntesis , Linfocitos T CD4-Positivos/trasplante , Diferenciación Celular/inmunología , Antígenos de Histocompatibilidad Clase I/biosíntesis , Antígenos de Histocompatibilidad Clase II/biosíntesis , Receptores de Hialuranos/biosíntesis , Tolerancia Inmunológica , Integrina beta1/biosíntesis , Ganglios Linfáticos/virología , Receptor beta de Linfotoxina/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Parabiosis , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Transducción de Señal/inmunología , Vaccinia/inmunología , Molécula 1 de Adhesión Celular Vascular/biosíntesis
12.
Eur Arch Otorhinolaryngol ; 273(4): 845-51, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25864182

RESUMEN

E-cadherin, ß-catenin, and ß1 integrin are important cell adhesion molecules to maintain epithelial structure and function. We investigated the expression of these cell adhesion molecules in cholesteatomas to understand the role of cell-cell and cell-extracellular matrix interaction in cholesteatomas. An immunohistochemical investigation was carried out on 35 cholesteatoma tissue samples (14 congenital, 21 acquired cholesteatomas) and 10 normal retroauricular skin (RAS) tissues which are obtained during middle ear surgery. The expression rate was measured to find out differences between retroauricular skin and cholesteatoma, as well as between congenital and acquired cholesteatoma. E-cadherin expression rate was significantly lower in the cholesteatoma (spinous layer 88.7 ± 17.9 %, granular layer 54.6 ± 22.6 %) than in the RAS (100 %, 74.4 ± 7.4 %) and in the acquired (83.3 ± 19.4 %, 48.1 ± 22.9 %) than in the congenital (96.7 ± 12.0 %, 64.4 ± 18.8 %). ß-catenin expression rate was significantly lower in the cholesteatoma (spinous layer 84.1 ± 17.2 %, granular layer 28.7 ± 30.8 %) than in the RAS (100 %, 75.9 ± 6.1 %) and in the acquired (78.1 ± 17.0 %, 17.1 ± 22.3 %) than in the congenital (93.2 ± 13.5 %, 46.1 ± 34.2 %). The expression pattern of ß-catenin is similar to that of E-cadherin. In ß1 integrin, there was no significant difference of the expression rate between RAS and cholesteatoma, as well as between congenital and acquired cholesteatoma. In conclusion, the expression of E-cadherin and ß-catenin is reduced in cholesteatoma, and the reduction is more pronounced in acquired cholesteatoma than in congenital cholesteatoma. Acquired cholesteatomas showed more aggressive characteristics than congenital cholesteatomas in terms of cell-cell adhesion.


Asunto(s)
Cadherinas/genética , Colesteatoma del Oído Medio/genética , Regulación de la Expresión Génica , Integrina beta1/genética , ARN/genética , beta Catenina/genética , Adolescente , Adulto , Anciano , Cadherinas/biosíntesis , Moléculas de Adhesión Celular , Niño , Preescolar , Colesteatoma/congénito , Colesteatoma/genética , Colesteatoma/metabolismo , Colesteatoma/patología , Colesteatoma del Oído Medio/metabolismo , Colesteatoma del Oído Medio/patología , Femenino , Humanos , Inmunohistoquímica , Lactante , Integrina beta1/biosíntesis , Masculino , Persona de Mediana Edad , Adulto Joven , beta Catenina/biosíntesis
13.
Carcinogenesis ; 36(9): 1071-83, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26153023

RESUMEN

Rhabdomyosarcoma represents about 50% of soft-tissue sarcomas and 10% of malignant solid tumors in childhood. Embryonal rhabdomyosarcoma (ERMS) is the most frequent subtype, suggested to have an origin in muscle precursor cells that fail to exit the cell cycle and terminally differentiate mainly because of overexpression of the transcription factor, PAX7, which sustains proliferation, migration and invasiveness in ERMS cells. Artesunate (ARS) is a semi-synthetic derivative of artemisinin (ART), a natural compound well known as an antimalarial drug. However, ART and its derivatives have been found efficacious even as anticancer drugs that induce cell cycle arrest and/or apoptosis in several kinds of cancer. Here, we show that ARS dose-dependently induces DNA damage and apoptosis in ERMS cell lines. Production of reactive oxygen species (ROS) and activation of p38 MAPK have a central role in triggering ARS-mediated apoptosis in ERMS cells; indeed either the antioxidant, N-acetylcysteine or the p38 MAPK inhibitor, SB203580, protects ERMS cells from ARS-induced apoptosis. Moreover, ARS treatment in ERMS cells ROS-dependently induces the expression of the myo-miRs, miR-133a and miR-206, which are down-regulated in RMS, and reduces PAX7 protein levels. Finally, ARS upregulates the expression of the adhesion molecules, NCAM and integrin ß1, and reduces migration and invasiveness of ERMS cells in vitro, and ARS treatment reduces of about 50% the growth of ERMS xenografts in vivo. Our results are the first evidence of efficacy of ART derivatives in restraining ERMS growth in vivo, and suggest ARS as a potential candidate for therapeutic treatment of ERMS.


Asunto(s)
Apoptosis/efectos de los fármacos , Artemisininas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Rabdomiosarcoma Embrionario/tratamiento farmacológico , Rabdomiosarcoma Embrionario/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Acetilcisteína/farmacología , Animales , Antineoplásicos/farmacología , Apoptosis/fisiología , Artemisininas/uso terapéutico , Artesunato , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Daño del ADN/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Imidazoles/farmacología , Integrina beta1/biosíntesis , Ratones , MicroARNs/biosíntesis , Invasividad Neoplásica , Trasplante de Neoplasias , Moléculas de Adhesión de Célula Nerviosa/biosíntesis , Factor de Transcripción PAX7/biosíntesis , Piridinas/farmacología , Trasplante Heterólogo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
14.
J Biol Chem ; 289(14): 10029-44, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24550382

RESUMEN

Elevated macrophage infiltration in tumor tissues is associated with breast cancer metastasis. Cancer cell migration/invasion toward angiogenic microvasculature is a key step in metastatic spread. We therefore studied how macrophages stimulated breast cancer cell interactions with endothelial cells. Macrophages produced cytokines, such as interleukin-8 and tumor necrosis factor-α, to stimulate endothelin (ET) and ET receptor (ETR) expression in breast cancer cells and human umbilical vascular endothelial cells (HUVECs). ET-1 was induced to a greater extent from HUVECs than from breast cancer cells, resulting in a density difference that facilitated cancer cell chemotaxis toward HUVECs. Macrophages also stimulated breast cancer cell adhesion to HUVECs and transendothelial migration, which were repressed by ET-1 antibody or ETR inhibitors. The ET axis induced integrins, such as αV and ß1, and their counterligands, such as intercellular adhesion molecule-2 and P-selectin, in breast cancer cells and HUVECs, and antibodies against these integrins efficiently suppressed macrophage-stimulated breast cancer cell interactions with HUVECs. ET-1 induced Ets-like kinase-1 (Elk-1), signal transducer and activator of transcription-3 (STAT-3), and nuclear factor-κB (NF-κB) phosphorylation in breast cancer cells. The use of inhibitors to prevent their phosphorylation or ectopic overexpression of dominant-negative IκBα perturbed ET-1-induced integrin αV and integrin ß1 expression. The physical associations of these three transcriptional factors with the gene promoters of the two integrins were furthermore evidenced by a chromatin immunoprecipitation assay. Finally, our mouse orthotopic tumor model revealed an ET axis-mediated lung metastasis of macrophage-stimulated breast cancer cells, suggesting that the ET axis was involved in macrophage-enhanced breast cancer cell endothelial interactions.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular , Endotelina-1/metabolismo , Integrina alfaV/biosíntesis , Integrina beta1/biosíntesis , Macrófagos/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Endotelina-1/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Xenoinjertos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfaV/genética , Integrina beta1/genética , Macrófagos/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Selectina-P/genética , Selectina-P/metabolismo , Proteína Elk-1 con Dominio ets/genética , Proteína Elk-1 con Dominio ets/metabolismo
15.
Circ J ; 79(1): 201-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25373595

RESUMEN

BACKGROUND: Endothelial-mesenchymal transition (EndMT) plays a pivotal role in cardiac fibrosis. However, it is unclear whether EndMT is involved in dyssynchronous heart failure (DHF). METHODS AND RESULTS: Twelve dogs received 3-week rapid right ventricular pacing (RVP) to develop DHF and then were randomly divided into a RVP group (n=6; RVP for another 3 weeks) and a biventricular pacing (BiVP) group (n=6; BiVP for 3 weeks), and another 6 dogs were in the control group. Contractile function in BiVP group was a little better than that in RVP group (P<0.05), but significant heart failure remained in 2 groups. RVP induced more significant cardiac fibrosis and higher collagen 1A2 expression in the left ventricular lateral wall (late-contracting and high-stress) than that in the anterior wall, and for those in the BiVP group, it was much lower. CD31, S100A4, α-smooth muscle actin and collagen 1A2 were used to evaluate EndMT. EndMT levels, transforming growth factor-ß (TGF-ß)/snail signaling, collagen 1A2 and integrin ß1 expression were much higher in the endothelial cells from the RVP lateral wall than that from BiVP. In this in vitro study, cyclic stretch could independently induce EndMT and enhance the pro-EndMT effect of TGF-ß in HUVECs, which could be partly blocked by integrin ß1 siRNA. CONCLUSIONS: RVP-induced DHF could aggravate fibrosis due to regional heterogeneity of mechanical stress, and it was better in the BiVP group where mechanical stress-induced EndMT might play a pivotal role through the integrin ß1 pathway.


Asunto(s)
Estimulación Cardíaca Artificial/efectos adversos , Transdiferenciación Celular/fisiología , Endotelio/patología , Insuficiencia Cardíaca/fisiopatología , Mesodermo/patología , Miocardio/patología , Animales , Terapia de Resincronización Cardíaca/efectos adversos , Colágeno Tipo I/biosíntesis , Perros , Células Endoteliales/metabolismo , Fibrosis , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Ventrículos Cardíacos/fisiopatología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina beta1/biosíntesis , Integrina beta1/genética , Masculino , Contracción Miocárdica , Miocardio/metabolismo , Distribución Aleatoria , Transducción de Señal/fisiología , Factores de Transcripción de la Familia Snail , Estrés Mecánico , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/biosíntesis
16.
Biosci Biotechnol Biochem ; 79(11): 1794-801, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26057585

RESUMEN

MUC5B mucin is a principal component of airway mucus and plays a key role in biodefense. We investigated the regulation of MUC5B production using the signals from extracellular matrix (ECM) components in NCI-H292 human lung epithelial cells. We found that MUC5B production in NCI-H292 cells cultured on fibronectin or laminin increased by 4-5-fold, with the increase occurring in a dose- and time-dependent manner. In contrast, MUC5B production was unchanged on type-IV collagen. Inhibition of integrin ß1 induced upregulation of MUC5B and MUC5AC; however, inhibition of p38 MAPK did not show any remarkable change in overproduced MUC5B. Inhibition of extracellular signal-regulated kinase (ERK) pathway or the transcription factor NF-κB induced the recovery of overproduced MUC5B on fibronectin and laminin. These results suggest that MUC5B production can be regulated by ECM components and that MUC5B is upregulated by fibronectin and laminin via the integrin, ERK, and NF-κB dependent pathway.


Asunto(s)
Fibronectinas/biosíntesis , Laminina/biosíntesis , Mucina 5B/biosíntesis , Moco/metabolismo , Colágeno Tipo IV/administración & dosificación , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Integrina beta1/biosíntesis , Pulmón/metabolismo , Pulmón/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Mucina 5B/genética , Sistema Respiratorio/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis
17.
J Cell Mol Med ; 18(11): 2235-51, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25215932

RESUMEN

Pancreatic ductal adenocarcinoma is one of the lethal cancers with extensive local tumour invasion, metastasis, early systemic dissemination and poorest prognosis. Thus, understanding the mechanisms regulating invasion/metastasis and epithelial-mesenchymal transition (EMT), is the key for developing effective therapeutic strategies for pancreatic cancer (PaCa). Eukaryotic elongation factor-2 kinase (eEF-2K) is an atypical kinase that we found to be highly up-regulated in PaCa cells. However, its role in PaCa invasion/progression remains unknown. Here, we investigated the role of eEF-2K in cellular invasion, and we found that down-regulation of eEF-2K, by siRNA or rottlerin, displays impairment of PaCa cells invasion/migration, with significant decreases in the expression of tissue transglutaminase (TG2), the multifunctional enzyme implicated in regulation of cell attachment, motility and survival. These events were associated with reductions in ß1 integrin/uPAR/MMP-2 expressions as well as decrease in Src activity. Furthermore, inhibition of eEF-2K/TG2 axis suppresses the EMT, as demonstrated by the modulation of the zinc finger transcription factors, ZEB1/Snail, and the tight junction proteins, claudins. Importantly, while eEF-2K silencing recapitulates the rottlerin-induced inhibition of invasion and correlated events, eEF-2K overexpression, by lentivirus-based expression system, suppresses such rottlerin effects and potentiates PaCa cells invasion/migration capability. Collectively, our results show, for the first time, that eEF-2K is involved in regulation of the invasive phenotype of PaCa cells through promoting a new signalling pathway, which is mediated by TG2/ß1 integrin/Src/uPAR/MMP-2, and the induction of EMT biomarkers which enhance cancer cell motility and metastatic potential. Thus, eEF-2K could represent a novel potential therapeutic target in pancreatic cancer.


Asunto(s)
Quinasa del Factor 2 de Elongación/genética , Integrina beta1/biosíntesis , Neoplasias Pancreáticas/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Transglutaminasas/biosíntesis , Acetofenonas/administración & dosificación , Benzopiranos/administración & dosificación , Línea Celular Tumoral , Quinasa del Factor 2 de Elongación/metabolismo , Transición Epitelial-Mesenquimal/genética , Proteínas de Unión al GTP , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Integrina beta1/genética , Invasividad Neoplásica/genética , Neoplasias Pancreáticas/patología , Proteína Glutamina Gamma Glutamiltransferasa 2 , ARN Interferente Pequeño , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Transducción de Señal/genética , Familia-src Quinasas/biosíntesis , Familia-src Quinasas/genética , Neoplasias Pancreáticas
18.
Eur J Immunol ; 43(1): 209-18, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23065740

RESUMEN

Carbon monoxide (CO) treatment improves pathogenic outcome of autoimmune diseases by promoting tolerance. However, the mechanism behind this protective tolerance is not yet defined. Here, we show in a transgenic mouse model for autoimmune diabetes that ex vivo gaseous CO (gCO)-treated DCs loaded with pancreatic ß-cell peptides protect mice from disease. This protection is peptide-restricted, independent of IL-10 secretion by DCs and of CD4(+) T cells. Although no differences were observed in autoreactive CD8(+) T-cell function from gCO-treated versus untreated DC-immunized groups, gCO-treated DCs strongly inhibited accumulation of autoreactive CD8(+) T cells in the pancreas. Interestingly, induction of ß1-integrin was curtailed when CD8(+) T cells were primed with gCO-treated DCs, and the capacity of these CD8(+) T cells to lyse isolated islet was dramatically impaired. Thus, immunotherapy using CO-treated DCs appears to be an original strategy to control autoimmune disease.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Monóxido de Carbono/farmacología , Células Dendríticas/efectos de los fármacos , Diabetes Mellitus Tipo 1/terapia , Células Secretoras de Insulina/inmunología , Integrina beta1/biosíntesis , Páncreas/inmunología , Animales , Autoantígenos/inmunología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Humanos , Tolerancia Inmunológica , Integrina beta1/genética , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/inmunología
19.
J Biol Chem ; 287(49): 41324-33, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23066031

RESUMEN

Rad9 as part of the Rad9-Hus1-Rad1 complex is known to participate in cell cycle checkpoint activation and DNA repair. However, Rad9 can act as a sequence-specific transcription factor, modulating expression of a number of genes. Importantly, Rad9 is up-regulated in prostate cancer cell lines and clinical specimens. Its expression correlates positively with advanced stage tumors and its down-regulation reduces tumor burden in mice. We show here that transient down-regulation of Rad9 by RNA interference reduces DU145 and PC3 prostate cancer cell proliferation and survival in vitro. In addition, transient or stable down-regulation of Rad9 impairs migration and invasion of the cells. Moreover, stable reduction of Rad9 renders DU145 cell growth anchorage-dependent. It also decreases expression of integrin ß1 protein and sensitizes DU145 and LNCaP cells to anoikis and impairs Akt activation. On the other hand, stable expression of Mrad9, the mouse homolog, in DU145/shRNA Rad9 cells restores migration, invasion, anchorage-independent growth, integrin ß1 expression, and anoikis resistance with a concomitant elevation of Akt activation. We thus demonstrate for the first time that Rad9 contributes to prostate tumorigenesis by increasing not only tumor proliferation and survival but also tumor migration and invasion, anoikis resistance, and anchorage-independent growth.


Asunto(s)
Anoicis , Proteínas de Ciclo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/patología , Adhesión Celular , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Reparación del ADN , Progresión de la Enfermedad , Regulación hacia Abajo , Silenciador del Gen , Humanos , Integrina beta1/biosíntesis , Masculino , Invasividad Neoplásica , Neoplasias de la Próstata/metabolismo , Interferencia de ARN , Transducción de Señal
20.
J Biol Chem ; 287(39): 32861-73, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22854957

RESUMEN

Intracellular vesicle transport pathways are critical for neuronal survival and central nervous system development. The Vps-C complex regulates multiple vesicle transport pathways to the lysosome in lower organisms. However, little is known regarding its physiological function in mammals. We deleted Vps18, a central member of Vps-C core complex, in neural cells by generating Vps18(F/F); Nestin-Cre mice (Vps18 conditional knock-out mice). These mice displayed severe neurodegeneration and neuronal migration defects. Mechanistic studies revealed that Vps18 deficiency caused neurodegeneration by blocking multiple vesicle transport pathways to the lysosome, including autophagy, endocytosis, and biosynthetic pathways. Our study also showed that ablation of Vps18 resulted in up-regulation of ß1 integrin in mouse brain probably due to lysosome dysfunction but had no effects on the reelin pathway, expression of N-cadherin, or activation of JNK, which are implicated in the regulation of neuronal migration. Finally, we demonstrated that knocking down ß1 integrin partially rescued the migration defects, suggesting that Vps18 deficiency-mediated up-regulation of ß1 integrin may contribute to the defect of neuronal migration in the Vps18-deficient brain. Our results demonstrate important roles of Vps18 in neuron survival and migration, which are disrupted in multiple neural disorders.


Asunto(s)
Encéfalo/metabolismo , Movimiento Celular , Lisosomas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Transporte Biológico Activo/genética , Encéfalo/patología , Cadherinas/genética , Cadherinas/metabolismo , Regulación de la Expresión Génica/genética , Integrina beta1/biosíntesis , Integrina beta1/genética , Lisosomas/genética , Lisosomas/patología , Ratones , Ratones Mutantes , Proteínas del Tejido Nervioso/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Proteína Reelina , Proteínas de Transporte Vesicular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA