Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Biochem Biophys Res Commun ; 461(1): 47-53, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25866182

RESUMEN

Wound healing is critical for normal development and pathological processes including cancer cell metastasis. MAPK, Rho-GTPases and NFκB are important regulators of wound healing, but mechanisms for their integration are incompletely understood. Annexin-A1 (ANXA1) is upregulated in invasive breast cancer cells resulting in constitutive activation of NFκB. We show here that silencing ANXA1 increases the formation of stress fibers and focal adhesions, which may inhibit wound healing. ANXA1 regulated wound healing is dependent on the activation of ERK1/2. ANXA1 increases the activation of RhoA, which is dependent on ERK activation. Furthermore, active RhoA is important in NF-κB activation, where constitutively active RhoA potentiates NFκB activation, while dominant negative RhoA inhibits NFκB activation in response to CXCL12 stimulation and active MEKK plasmids. These findings establish a central role for ANXA1 in the cell migration through the activation of NFκB, ERK1/2 and RhoA.


Asunto(s)
Anexina A1/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Cicatrización de Heridas/fisiología , Proteína de Unión al GTP rhoA/metabolismo , Movimiento Celular , Femenino , Humanos , Células MCF-7 , Células Tumorales Cultivadas
2.
Clin Transl Sci ; 13(1): 57-66, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31343094

RESUMEN

In the last decade, drug development has tackled substantial challenges to improve efficiency and facilitate access to innovative medicines. Integrated clinical protocols and the investigation of targeted oncology drugs in healthy volunteers (HVs) have emerged as modalities with an increase in scope and complexity of early clinical studies and first-in-human (FIH) studies in particular. However, limited work has been done to explore the impact of these two modalities, alone or in combination, on the scientific value and on the implementation of such articulated studies. We conducted an FIH study in HVs with an oncology targeted drug, an Mnk 1/2 small molecule inhibitor. In this article, we describe results, advantages, and limitations of an integrated clinical protocol with an oncology drug. We further discuss and indicate points to consider when designing and conducting similar scientifically and operationally demanding FIH studies.


Asunto(s)
Antineoplásicos/efectos adversos , Enfermedades Cardiovasculares/diagnóstico , Protocolos Clínicos , Inhibidores de Proteínas Quinasas/efectos adversos , Proyectos de Investigación , Administración Oral , Adulto , Antineoplásicos/administración & dosificación , Enfermedades Cardiovasculares/inducido químicamente , Electrocardiografía , Voluntarios Sanos , Neoplasias Hematológicas/tratamiento farmacológico , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Masculino , Oncología Médica/métodos , Persona de Mediana Edad , Monitoreo Ambulatorio/métodos , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Adulto Joven
3.
J Cell Physiol ; 219(2): 382-91, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19142852

RESUMEN

Previously, we reported the identification of a novel immunoglobulin-like cell adhesion molecule hepaCAM that promotes cell-extracellular matrix (ECM) interactions including cell adhesion and motility. Cell-ECM interactions are known to be directed by the actin cytoskeleton. In this study, we examined the association of hepaCAM with the actin cytoskeleton. We found that hepaCAM was partially insoluble in Triton X-100 and colocalized with the actin cytoskeleton on the plasma membrane. Disruption of F-actin decreased the detergent insolubility and disturbed the subcellular localization of hepaCAM. Coimmunoprecipitation and F-actin cosedimentation assays revealed that hepaCAM directly bound to F-actin. In addition, we constructed three N- and C-terminal domain-deleted mutants of hepaCAM to determine the actin-binding region as well as to evaluate the effect of the domains on the biological function of hepaCAM. Detergent solubility assays showed that the cytoplasmic domain of hepaCAM might be required for actin association. However, deletion of either the extracellular or the cytoplasmic domain of hepaCAM abolished actin coprecipitation as well as delayed cell-ECM adhesion and cell motility. The data suggest that an intact hepaCAM protein is critical for establishing a stable physical association with the actin cytoskeleton; and such association is important for modulating hepaCAM-mediated cell adhesion and motility.


Asunto(s)
Actinas/metabolismo , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Citoesqueleto/metabolismo , Proteínas/metabolismo , Actinas/genética , Animales , Proteínas de Ciclo Celular , Línea Celular , Detergentes/metabolismo , Matriz Extracelular/metabolismo , Humanos , Ratones , Proteínas/genética
4.
J Cell Biochem ; 107(6): 1129-38, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19507233

RESUMEN

Subsequent to our identification of a novel immunoglobulin-like cell adhesion molecule hepaCAM, we showed that hepaCAM is frequently lost in diverse human cancers and is capable of modulating cell motility and growth when re-expressed. Very recently, a molecule identical to hepaCAM (designated as GlialCAM) was found highly expressed in glial cells of the brain. Here, we demonstrate that hepaCAM is capable of inducing differentiation of the human glioblastoma U373-MG cells. Expression of hepaCAM resulted in a significant increase in the astrocyte differentiation marker glial fibrillary acid protein (GFAP), indicating that hepaCAM promotes glioblastoma cells to undergo differentiation. To determine the relationship between hepaCAM expression level and cell differentiation, we established two U373-MG cell lines expressing hepaCAM at different levels. The results revealed that high-level hepaCAM triggered a clear increase in GFAP expression as well as morphological changes characteristic of glioblastoma cell differentiation. Furthermore, high expression of hepaCAM significantly accelerated cell adhesion but inhibited cell proliferation and migration. Concomitantly, deregulation of cell cycle regulatory proteins was detected. Expectedly, the differentiation was noticeably less apparent in cells expressing low-level hepaCAM. Taken together, our findings suggest that hepaCAM induces differentiation of the glioblastoma U373-MG cells. The degree of cell differentiation is dependent on the expression level of hepaCAM.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Diferenciación Celular , Glioblastoma/patología , Proteínas/fisiología , Adhesión Celular , Moléculas de Adhesión Celular/análisis , Moléculas de Adhesión Celular/genética , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Expresión Génica , Humanos , Proteínas/análisis , Proteínas/genética
5.
Biochem Biophys Res Commun ; 378(4): 755-60, 2009 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-19059381

RESUMEN

Subsequent to our identification of the novel immunoglobulin-like cell adhesion molecule hepaCAM, we demonstrated that hepaCAM is capable of modulating cell growth and cell-extracellular matrix interactions. In this study, we examined the localization of hepaCAM in lipid rafts/caveolae as well as the interaction of hepaCAM with the caveolar structural protein caveolin-1 (Cav-1). Our results revealed that a portion of hepaCAM resided in detergent-resistant membranes and co-partitioned with Cav-1 to low buoyant density fractions characteristic of lipid rafts/caveolae. In addition, co-localization and coimmunoprecipitation assays confirmed the association of hepaCAM with Cav-1. Deletion analysis of hepaCAM showed that the extracellular first immunoglobulin domain of hepaCAM was required for binding Cav-1. Furthermore, when co-expressed, Cav-1 induced the expression of hepaCAM as well as distributed hepaCAM to intracellular Cav-1-positive caveolar structures. Taken together, our findings indicate that hepaCAM is partially localized in the lipid rafts/caveolae and interacts with Cav-1 through its first immunoglobulin domain.


Asunto(s)
Caveolas/metabolismo , Caveolina 1/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas/metabolismo , Animales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Humanos , Inmunoglobulinas/química , Ratones , Células 3T3 NIH , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas/genética , Eliminación de Secuencia , Transducción de Señal
6.
Carcinogenesis ; 29(12): 2298-305, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18845560

RESUMEN

Previously, we reported the identification of a novel immunoglobulin-like cell adhesion molecule hepaCAM that is frequently downregulated and inhibits cell growth in hepatocellular carcinoma. In this study, we show that the expression of hepaCAM is suppressed in diverse human cancers. Aiming to evaluate the biological role of hepaCAM in breast cancer, we stably transfected the MCF7 cell line with either wild-type hepaCAM or its mutant hCAM-tailless that lacked the cytoplasmic domain. We found that hepaCAM inhibited colony formation and cell proliferation and arrested cells in the G(2)/M phase. Intriguingly, hepaCAM was capable of inducing cellular senescence as defined by the enlarged cell morphology and increased beta-galactosidase activity. Furthermore, hepaCAM elevated the expression levels of senescence-associated proteins including p53, p21 and p27. In contrast, cell growth inhibition and senescence were less apparent in cells overexpressing hCAM-tailless mutant. To determine if the p53-mediated pathway was involved in hepaCAM-induced senescence, we used the small-interfering RNA system to knock down endogenous p53 expression. In the presence of hepaCAM, downregulation of p53 resulted in a clear reduction of p21, insignificant change in p27 and alleviated senescence. Together, the results suggest that the expression of hepaCAM in MCF7 cells not only inhibits cell growth but also induces cellular senescence through the p53/21 pathway.


Asunto(s)
Neoplasias de la Mama/metabolismo , Senescencia Celular/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Western Blotting , Neoplasias de la Mama/genética , Ciclo Celular/genética , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Regulación hacia Abajo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Transfección , Proteína p53 Supresora de Tumor/genética
7.
Int J Oncol ; 31(2): 293-301, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17611685

RESUMEN

Genetic alterations have been defined as the hallmark of cancers as they are responsible for the differences between normal and malignant phenotypes. A widely accepted approach to study genetic instability is to identify cancer-related genes, in particular, the two major groups of growth regulatory genes - oncogenes and tumour suppressor genes. Using the technique of suppression subtractive hybridisation, we identified a novel gene transcript, designated as HEPT3. RT-PCR demonstrated that HEPT3 was overexpressed in 87% (20/23) of HCC patients and in 4/5 HCC cell lines tested. Sequence analyses performed on the full-length cDNA revealed that HEPT3 is an intronless gene mapped to human chromosome 6q13-14. The gene transcript lacks an extensive open reading frame and contains an Alu sequence near the 5' terminus, indicating that HEPT3 encodes a noncoding RNA. Antisense studies on the HCC cell line HepG2 showed that, when HEPT3 expression level was reduced, cell proliferation rate was inhibited by approximately 5-fold and cell colony formation was reduced by at least 50%. Our data suggest that the novel gene HEPT3 may function through its noncoding RNA and its overexpression may play a role in hepatocarcinogenesis.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/tratamiento farmacológico , Proteínas de Neoplasias/biosíntesis , Secuencia de Bases , Línea Celular Tumoral , Cromosomas Humanos Par 6 , Cartilla de ADN/química , ADN Complementario/metabolismo , Humanos , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Hibridación de Ácido Nucleico , Oligonucleótidos Antisentido/química , ARN Neoplásico/metabolismo , ARN no Traducido , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Oncotarget ; 7(19): 27007-20, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27105503

RESUMEN

MiRNAs are endogenous ~22 nt RNAs which play critical regulatory roles in a wide range of biological and pathological processes, which can act as oncogenes or tumor suppressor genes depending on their target genes. We have recently shown that ANXA1 inhibits the expression of miRNAs including miR196a. Here, we show that miR196a was highly expressed in ER+ MCF-7 breast cancer cells when compared to normal mammary gland cells, with expression levels negatively correlating to ANXA1. ANXA1 inhibits the biogenesis of oncogenic miR-196a by suppressing primary-miR196a indirectly through the stimulation of c-myc and NFkB expression and activity in breast cancer cells. In a negative feedback loop, miR-196a directly inhibits ANXA1 and enhances breast cancer cell proliferation in vitro. Finally, miR196a promotes breast tumor growth in vivo. This study reports a novel regulatory circuit between ANXA1, NF-kB, c-myc and miR-196a which regulates breast cancer cell proliferation and tumor growth.


Asunto(s)
Anexina A1/genética , Neoplasias de la Mama/genética , Proliferación Celular/genética , MicroARNs/genética , FN-kappa B/genética , Proteínas Proto-Oncogénicas c-myc/genética , Animales , Anexina A1/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Retroalimentación Fisiológica , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Ratones Endogámicos BALB C , Ratones Desnudos , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , Trasplante Heterólogo , Carga Tumoral/genética
9.
Cell Stress Chaperones ; 20(4): 583-93, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25753354

RESUMEN

Febrile temperatures can induce stress responses which protect cells from damage and can reduce inflammation during infections and sepsis. However, the mechanisms behind the protective functions of heat in response to the bacterial endotoxin LPS are unclear. We have recently shown that Annexin-1 (ANXA1)-deficient macrophages exhibited higher TNFα levels after LPS stimulation. Moreover, we have previously reported that ANXA1 can function as a stress protein. Therefore in this study, we determined if ANXA1 is involved in the protective effects of heat on cytokine levels in macrophages after heat and LPS. Exposure of macrophages to 42 °C for 1 h prior to LPS results in an inhibition of TNFα production, which was not evident in ANXA1(-/-) macrophages. We show that this regulation involves primarily MYD88-independent pathways. ANXA1 regulates TNFα mRNA stability after heat and LPS, and this is dependent on endogenous ANXA1 expression and not exogenously secreted factors. Further mechanistic studies revealed the possible involvement of the heat shock protein HSP70 and JNK in the heat and inflammatory stress response regulated by ANXA1. This study shows that ANXA1, an immunomodulatory protein, is critical in the heat stress response induced after heat and endotoxin stimulation.


Asunto(s)
Anexina A1/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Lipopolisacáridos/toxicidad , Macrófagos/efectos de los fármacos , Factor de Necrosis Tumoral alfa/análisis , Animales , Anexina A1/deficiencia , Células de la Médula Ósea/citología , Células Cultivadas , Citocinas/análisis , Ensayo de Inmunoadsorción Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Estabilidad del ARN/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Temperatura , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/genética
10.
J Biol Chem ; 280(29): 27366-74, 2005 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-15917256

RESUMEN

We have recently identified a novel gene, hepaCAM, in liver that encodes a cell adhesion molecule of the immunoglobulin superfamily. In this study, we examined the characteristics of hepaCAM protein and the relationship between its structure and function, in particular its adhesive properties. The wild-type and the cytoplasmic domain-truncated mutants of hepaCAM were transfected into the human breast carcinoma MCF7 cells, and the physiological and biological properties were assessed. Biochemical analyses revealed that hepaCAM is an N-linked glycoprotein phosphorylated in the cytoplasmic domain and that it forms homodimers through cis-interaction on the cell surface. The subcellular localization of hepaCAM appears density-dependent; in well spread cells, hepaCAM is distributed to cell protrusions, whereas in confluent cells, hepaCAM is predominantly accumulated at the sites of cell-cell contacts on the cell membrane. In polarized cells, hepaCAM is recruited to the lateral and basal membranes, and lacking physical interaction, hepaCAM is shown to co-localize with E-cadherin at the lateral membrane. Cell adhesion and motility assays demonstrated that hepaCAM increased cell spreading on the matrices fibronectin and matrigel, delayed cell detachment, and enhanced wound healing. Furthermore, when the cytoplasmic domain was deleted, hepaCAM mutants did not affect cell surface localization and dimer formation. Cell-matrix adhesion, however, was less significantly increased, and cell motility was almost unchanged when compared with the effect of the wild-type hepaCAM. Taken together, the cytoplasmic domain of hepaCAM is essential to its function on cell-matrix interaction and cell motility.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas/metabolismo , Neoplasias de la Mama/química , Cadherinas/metabolismo , Adhesión Celular , Proteínas de Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Polaridad Celular , Extensiones de la Superficie Celular/química , Dimerización , Femenino , Glicoproteínas , Humanos , Uniones Intercelulares/química , Fosfoproteínas , Transporte de Proteínas , Proteínas/química , Proteínas/fisiología , Transfección
11.
J Hepatol ; 39(4): 580-6, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12971969

RESUMEN

BACKGROUND/AIMS: Examining genes associated with human hepatocellular carcinoma (HCC) by subtractive hybridisation, we identified a novel transcript, designated as HEPN1, in non-tumorous liver. In this study, we aimed to evaluate HEPN1 gene expression in HCC patients, to characterise and to explore the functional significance of HEPN1 in vitro. METHODS: One-step reverse transcription-polymerase chain reaction (RT-PCR) and real-time RT-PCR were employed to determine HEPN1 expression in 23 paired (HCC and the adjacent non-HCC) liver specimens. Sequence analyses were performed by bioinformatics. Transfection studies were carried out by expressing HEPN1, V5-fused HEPN1, and green fluorescent protein-fused HEPN1, individually, in HepG2 cells. RESULTS: Significant downregulation of HEPN1 (P<0.0001) was detected in 22/23 of HCC patients tested. Gene HEPN1 maps to chromosome 11q24.2; and the predicted gene product, a 10-kDa peptide with 88 amino acids, has no homology to known proteins. When transfected into HepG2 cells, HEPN1 reduced cell viability to 37.5+/-2.5% (P=0.001), and induced apoptosis with typical morphological changes as demonstrated by microscopy and Annexin V assay. CONCLUSIONS: Our data show that HEPN1 is frequently silenced in HCC, and that exogenous HEPN1 exhibits antiproliferative effect on HepG2 cells, suggesting that silencing of HEPN1 may be associated with carcinogenesis of hepatocytes.


Asunto(s)
Apoptosis , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Regulación hacia Abajo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Proteínas/genética , Adulto , Anciano , Secuencia de Aminoácidos , Secuencia de Bases , Carcinoma Hepatocelular/fisiopatología , Proteínas de Ciclo Celular , División Celular , Línea Celular Tumoral , ADN Complementario , Femenino , Humanos , Neoplasias Hepáticas/fisiopatología , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA