Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
2.
Mol Biol Cell ; 31(8): 741-752, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32049581

RESUMEN

Collective cell migration plays crucial roles in tissue remodeling, wound healing, and cancer cell invasion. However, its underlying mechanism remains unknown. Previously, we showed that the RhoA-targeting guanine nucleotide exchange factor Solo (ARHGEF40) is required for tensile force-induced RhoA activation and proper organization of keratin-8/keratin-18 (K8/K18) networks. Here, we demonstrate that Solo knockdown significantly increases the rate at which Madin-Darby canine kidney cells collectively migrate on collagen gels. However, it has no apparent effect on the migratory speed of solitary cultured cells. Therefore, Solo decelerates collective cell migration. Moreover, Solo localized to the anteroposterior regions of cell-cell contact sites in collectively migrating cells and was required for the local accumulation of K8/K18 filaments in the forward areas of the cells. Partial Rho-associated protein kinase (ROCK) inhibition or K18 or plakoglobin knockdown also increased collective cell migration velocity. These results suggest that Solo acts as a brake for collective cell migration by generating pullback force at cell-cell contact sites via the RhoA-ROCK pathway. It may also promote the formation of desmosomal cell-cell junctions related to K8/K18 filaments and plakoglobin.


Asunto(s)
Movimiento Celular/fisiología , Transducción de Señal/fisiología , Proteínas de Unión al GTP rho/fisiología , Quinasas Asociadas a rho/fisiología , Amidas/farmacología , Animales , Polaridad Celular , Colágeno , Citoesqueleto/fisiología , Desmosomas/fisiología , Perros , Geles , Técnicas de Silenciamiento del Gen , Queratina-18/antagonistas & inhibidores , Queratina-18/genética , Queratina-18/fisiología , Queratina-8/antagonistas & inhibidores , Queratina-8/genética , Queratina-8/fisiología , Células de Riñón Canino Madin Darby , Piridinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Estrés Mecánico , Imagen de Lapso de Tiempo , gamma Catenina/antagonistas & inhibidores , gamma Catenina/genética , gamma Catenina/fisiología , Proteína de Unión al GTP rac1/fisiología , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/fisiología
3.
Nat Med ; 21(9): 1076-84, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26280121

RESUMEN

Tumor necrosis factor-α (TNF-α), one of the major stress-induced proinflammatory cytokines, is upregulated in the heart after tissue injury, and its sustained expression can contribute to the development of heart failure. Whether TNF-α also exerts cytoprotective effects in heart failure is not known. Here we provide evidence for a cardioprotective function of TNF-α in a genetic heart failure model, desmin-deficient mice. The cardioprotective effects of TNF-α are a consequence of nuclear factor-κB (NF-κB)-mediated ectopic expression in cardiomyocytes of keratin 8 (K8) and keratin 18 (K18), two epithelial-specific intermediate filament proteins. In cardiomyocytes, K8 and K18 (K8/K18) formed an alternative cytoskeletal network that localized mainly at intercalated discs (IDs) and conferred cardioprotection by maintaining normal ID structure and mitochondrial integrity and function. Ectopic induction of K8/K18 expression in cardiomyocytes also occurred in other genetic and experimental models of heart failure. Loss of the K8/K18 network resulted in a maladaptive cardiac phenotype following transverse aortic constriction. In human failing myocardium, where TNF-α expression is upregulated, K8/K18 were also ectopically expressed and localized primarily at IDs, which did not contain detectable amounts of desmin. Thus, TNF-α- and NF-κB-mediated formation of an alternative, stress-induced intermediate filament cytoskeleton has cardioprotective function in mice and potentially in humans.


Asunto(s)
Queratina-18/fisiología , Queratina-8/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Cardiomegalia/prevención & control , Desmina/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/fisiología
4.
Infect Immun ; 82(4): 1719-24, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24516112

RESUMEN

Fimbria-mediated adherence to the intestinal epithelia is a key step in enteroaggregative Escherichia coli (EAEC) pathogenesis. To date, four fimbriae have been described for EAEC; aggregative adherence fimbria II (AAF/II) is the most important adherence factor for EAEC prototype strain 042. Previously, we described results showing that extracellular matrix (ECM) components might be involved in the recognition of AAF/II fimbriae by intestinal cells. In this study, we sought to identify novel potential receptors on intestinal epithelial cells recognized by the AAF/II fimbriae. Purified AafA-dsc protein, the major subunit of AAF/II fimbriae, was incubated with a monolayer of T84 cells, cross-linked to the surface-exposed T84 cell proteins, and immunoprecipitated by using anti-AafA antibodies. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis of cellular proteins bound to AafA-dsc protein identified laminin (previously recognized as a potential receptor for AAF/II) and cytokeratin 8 (CK8). Involvement of the major subunit of AAF/II fimbriae (AafA protein) in the binding to recombinant CK8 was confirmed by adherence assays with purified AAF/II fimbriae, AafA-dsc protein, and strain 042. Moreover, HEp-2 cells transfected with CK8 small interfering RNA (siRNA) showed reduced 042 adherence compared with cells transfected with scrambled siRNA as a control. Adherence of 042 to HEp-2 cells preincubated with antibodies against ECM proteins or CK8 was substantially reduced. Altogether, our results supported the idea of a role of CK8 as a potential receptor for EAEC.


Asunto(s)
Adhesión Bacteriana/fisiología , Células Epiteliales/microbiología , Escherichia coli/fisiología , Fimbrias Bacterianas/fisiología , Queratina-8/fisiología , Laminina/fisiología , Adhesinas de Escherichia coli , Línea Celular , Células Epiteliales/fisiología , Fibronectinas/inmunología , Humanos , Mucosa Intestinal/citología , Queratina-8/metabolismo , Laminina/inmunología , Proteínas de la Membrana
5.
Exp Cell Res ; 319(4): 474-86, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23164509

RESUMEN

As differentiated cells, hepatocytes primarily metabolize glucose for ATP production through oxidative phosphorylation of glycolytic pyruvate, whereas proliferative hepatocellular carcinoma (HCC) cells undergo a metabolic shift to aerobic glycolysis despite oxygen availability. Keratins, the intermediate filament (IF) proteins of epithelial cells, are expressed as pairs in a lineage/differentiation manner. Hepatocyte and HCC (hepatoma) cell IFs are made solely of keratins 8/18 (K8/K18), thus providing models of choice to address K8/K18 IF functions in normal and cancerous epithelial cells. Here, we demonstrate distinctive increases in glucose uptake, glucose-6-phosphate formation, lactate release, and glycogen formation in K8/K18 IF-lacking hepatocytes and/or hepatoma cells versus their respective IF-containing counterparts. We also show that the K8/K18-dependent glucose uptake/G6P formation is linked to alterations in hexokinase I/II/IV content and localization at mitochondria, with little effect on GLUT1 status. In addition, we find that the insulin-stimulated glycogen formation in normal hepatocytes involves the main PI-3 kinase-dependent signaling pathway and that the K8/K18 IF loss makes them more efficient glycogen producers. In comparison, the higher insulin-dependent glycogen formation in K8/K18 IF-lacking hepatoma cells is associated with a signaling occurring through a mTOR-dependent pathway, along with an augmentation in cell proliferative activity. Together, the results uncover a key K8/K18 regulation of glucose metabolism in normal and cancerous hepatic cells through differential modulations of mitochondrial HK status and insulin-mediated signaling.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Glucosa/metabolismo , Hepatocitos/metabolismo , Hexoquinasa/metabolismo , Insulina/metabolismo , Queratina-18/fisiología , Queratina-8/fisiología , Neoplasias Hepáticas/metabolismo , Animales , Carcinoma Hepatocelular/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Hepatocitos/patología , Humanos , Insulina/farmacología , Queratina-18/metabolismo , Queratina-8/metabolismo , Neoplasias Hepáticas/patología , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
6.
J Cancer Res Clin Oncol ; 136(10): 1477-88, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20700687

RESUMEN

PURPOSE: This study aimed to explore the mechanism of multi-drug resistance (MDR) in 5-fluorouracil (5-FU)-induced breast cancer cell MCF-7. METHODS: MCF-7 cells were exposed in stepwise escalating concentration of 5-FU to develop the resistant cell line, MCF-7/5-FU. Biological and molecular characteristics of the cells were studied through MTT, flow cytometry, real-time PCR, western-blot, and the global protein profiles between MCF-7/5-FU and parental MCF-7 were compared using proteomic approach. Then some of the differentially expressed proteins were validated by western-blot. In addition, the role of 14-3-3sigma was validated using gene transfection. RESULTS: Drug resistance of MCF-7/5-FU cells to 5-FU, MX, cDDP, ADM, TAXOL all increased significantly compared with MCF-7 cells and that maybe related to BCRP, but not MDR1 and MRP1. Differentially expressed proteins between MCF-7/5-FU and MCF-7 cells were identified; 12 proteins were up-regulated and 18 proteins were down-regulated in MCF-7/5-FU cells. Expressive levels of some proteins in western-blot validation were consistent with the results in proteomic analysis. Enforced 14-3-3sigma expression can increase the sensitivity of MCF-7/5-FU cells to 5-FU and cDDP. CONCLUSION: MDR of MCF-7/5-FU likely associated with differentially expressed proteins and 14-3-3sigma may play a positive role in chemotherapy. These findings may provide theoretical support for the prediction of chemotherapeutic response and reverse of MDR.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Fluorouracilo/farmacología , Proteínas de Neoplasias/análisis , Proteómica/métodos , Proteínas 14-3-3/fisiología , Secuencia de Aminoácidos , Biomarcadores de Tumor/fisiología , Neoplasias de la Mama/química , Neoplasias de la Mama/patología , Línea Celular Tumoral , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Exonucleasas/fisiología , Exorribonucleasas , Femenino , Proteínas del Choque Térmico HSC70/fisiología , Humanos , Queratina-8/fisiología , Datos de Secuencia Molecular , Proteínas de Neoplasias/fisiología , Factor Tu de Elongación Peptídica/fisiología , Superóxido Dismutasa/fisiología
7.
Cancer Lett ; 265(2): 188-96, 2008 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-18353540

RESUMEN

By comparing protein profiles of nasopharyngeal carcinoma HONE1 cells to transformed nasopharyngeal epithelial NP 69 cells, several clusters of differentially expressed proteins were identified. The increased expression of cytokeratin 8 (CK8) and pyruvate kinase M2 was a common feature in four NPC cell lines compared to the two transformed epithelial cell lines. Suppression of CK8 was associated with the sensitivity to cisplatin in HONE1 cells; while overexpression of CK8 provided resistance to cisplatin-mediated apoptosis; and this protection occurred through an enhanced phosphorylation of c-Jun NH(2)-terminal kinase (JNK). Our findings implicate an underlying molecular mechanism in which CK8 is required for cisplatin resistance.


Asunto(s)
Cisplatino/farmacología , Resistencia a Antineoplásicos/fisiología , Queratina-8/fisiología , Neoplasias Nasofaríngeas/metabolismo , Apoptosis , Línea Celular Transformada , Línea Celular Tumoral , Silenciador del Gen , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Fosforilación , Análisis por Matrices de Proteínas , Transfección
8.
Hepatology ; 46(5): 1639-49, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17969036

RESUMEN

Keratin polypeptides 8 and 18 (K8/K18) are the cytoskeletal intermediate filament proteins of hepatocytes while K8/K18/K19 are the keratins of hepatobiliary ductal cells. Hepatocyte K8/K18 are highly abundant and behave as stress proteins with injury-inducible expression. Human association studies show that K8/K18 germline heterozygous mutations predispose to end-stage liver disease of multiple etiologies ( approximately 3 fold increased risk), and to liver disease progression in patients with chronic hepatitis C infection. These findings are supported by extensive transgenic mouse and ex vivo primary hepatocyte culture studies showing that K8 or K18 mutations predispose the liver to acute or subacute injury and promote apoptosis and fibrosis. Mutation-associated predisposition to liver injury is likely related to mechanical and nonmechanical keratin functions including maintenance of cell integrity, protection from apoptosis and oxidative injury, serving as a phosphate sponge, regulation of mitochondrial organization/function and protein targeting. These functions are altered by mutation-induced changes in keratin phosphorylation, solubility and filament organization/reorganization. Keratins are also the major constituents of Mallory-Denk bodies (MDBs). A toxin-induced K8>K18 ratio, and keratin crosslinking by transglutaminase-2 play essential roles in MDB formation. Furthermore, intracellular or cell-released K18 fragments, generated by caspase-mediated proteolysis during apoptosis serve as markers of liver injury. Therefore, K8 and K18 are cytoprotective stress proteins that play a central role in guarding hepatocytes from apoptosis. Keratin involvement in liver disease is multi-faceted and includes modulating disease progression upon mutation, formation of MDBs in response to unique forms of injury, and serving as markers of epithelial cell death.


Asunto(s)
Queratina-18/fisiología , Queratina-8/fisiología , Hepatopatías/fisiopatología , Biomarcadores/sangre , Progresión de la Enfermedad , Predisposición Genética a la Enfermedad , Hepatocitos/fisiología , Humanos , Queratina-18/genética , Queratina-18/metabolismo , Queratina-8/genética , Queratina-8/metabolismo , Hígado/patología , Hepatopatías/genética , Hepatopatías/patología , Mutación
9.
Cancer Res ; 67(5): 2107-13, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17332340

RESUMEN

Chronic exposures to cadmium compounds are carcinogenic. It was hypothesized that the development of resistance to cadmium may drive carcinogenesis. This is achieved by selection of resistant cells in which the apoptotic response is significantly attenuated. The induction of cadmium resistance in rat lung epithelial cells (LEC) was used to explore the mechanisms of cadmium-induced adaptation and carcinogenesis. Our previous results showed that LECs developed resistance to apoptosis during cadmium adaptation possibly due to perturbation of the c-Jun NH(2)-terminal kinase pathway. Here, we further study these cells by comparative proteomics. Interestingly, we showed that two intermediate filament proteins, cytokeratin 8 (CK8) and cytokeratin 14 (CK14), were increased significantly and stably maintained only in the adapted cells but not in cadmium-treated parental cells. It has been documented that CK8/cytokeratin 18 provided resistance to tumor necrosis factor (TNF)-induced apoptosis and CK14 may function as an inhibitor of TNF-TNF receptor 1 (TNFR1) signaling through an association with TNFR1-associated death domain protein, suggesting that up-regulation of CK8 and CK14 may be responsible for apoptotic resistance. Finally, we showed that small interfering RNA-specific knockdown of CK8 in cadmium-adapted cells attenuated the cadmium resistance, indicating the potential role of CK8 in cadmium resistance. This acquired self-resistance to apoptosis could account for cadmium-induced carcinogenesis, as this promotes neoplastic cell survival as well as subsequent clonal expansion and then progression of tumor development. Thus, increased expression of these cytokeratins represents an adaptive survival mechanism that resists cadmium-induced apoptosis and it is unprecedented that cells respond to long-term cadmium exposure by modulating keratin dynamics.


Asunto(s)
Cadmio/farmacología , Transformación Celular Neoplásica , Resistencia a Medicamentos , Queratina-8/fisiología , Animales , Transformación Celular Neoplásica/efectos de los fármacos , Células Cultivadas , Silenciador del Gen , Queratina-14/metabolismo , Queratina-8/antagonistas & inhibidores , Queratina-8/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Modelos Biológicos , Proteoma/análisis , Ratas , Tiempo
10.
J Biol Chem ; 282(11): 8219-27, 2007 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-17213200

RESUMEN

Keratin 8 (K8) and keratin-18 (K18) are the major intermediate filament proteins in the intestinal epithelia. The regulation and function of keratin in the intestinal epithelia is largely unknown. In this study we addressed the role and regulation of K8 and K18 expression by interleukin 6 (IL-6). Caco2-BBE cell line and IL-6 null mice were used to study the effect of IL-6 on keratin expression. Keratin expression was studied by Northern blot, Western blot, and confocal microscopy. Paracellular permeability was assessed by apical-to-basal transport of a fluorescein isothiocyanate dextran probe (FD-4). K8 was silenced using the small interfering RNA approach. IL-6 significantly up-regulated mRNA and protein levels of K8 and K18. Confocal microscopy showed a reticular pattern of intracellular keratin localized to the subapical region after IL-6 treatment. IL-6 also induced serine phosphorylation of K8. IL-6 decreased paracellular flux of FD-4 compared with vehicle-treated monolayers. K8 silencing abolished the decrease in paracellular permeability induced by IL-6. Administration of dextran sodium sulfate (DSS) significantly increased intestinal permeability in IL-6-/- mice compared with wild type mice given DSS. Collectively, our data demonstrate that IL-6 regulates the colonic expression of K8 and K18, and K8/K18 mediates barrier protection by IL-6 under conditions where intestinal barrier is compromised. Thus, our data uncover a novel function of these abundant cytoskeletal proteins, which may have implications in intestinal disorders such as inflammatory bowel disease wherein barrier dysfunction underlies the inflammatory response.


Asunto(s)
Células Epiteliales/citología , Interleucina-6/fisiología , Intestinos/citología , Queratina-8/fisiología , Queratinas/biosíntesis , Animales , Células CACO-2 , Colon/metabolismo , Colorantes Fluorescentes/farmacología , Humanos , Inflamación/patología , Interleucina-6/metabolismo , Ratones , Ratones Transgénicos , Microscopía Confocal , Permeabilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA