Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Biochem Biophys Res Commun ; 450(4): 1656-61, 2014 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-25044121

RESUMEN

Deregulation of nuclear factor-κB (NF-κB) and related pathways contribute to tumor cell proliferation and invasion. Mechanisms for constitutive NF-κB activation are not fully explained; however, the underlying defects appear to generate and maintain pro-oxidative conditions. In hepatocellular carcinoma (HCC) tissues, up-regulation of reactive oxygen species modulator 1 (Romo1) correlates positively with tumor size. In the present study, we showed that Romo1 expression is required to maintain constitutive nuclear DNA-binding activity of NF-κB and transcriptional activity through constitutive IκBα phosphorylation. Overexpression of Romo1 promoted p65 nuclear translocation and DNA-binding activity. We also show that Romo1 depletion suppressed anchorage-independent colony formation by HCC cells and suppressed tumor growth in vivo. Based on these findings, Romo1 may be a principal regulatory factor in the maintenance of constitutive NF-κB activation in tumor cells. In the interest of anti-proliferative treatments for cancer, Romo1 may also present a productive target for drug development.


Asunto(s)
División Celular/fisiología , Proteínas de la Membrana/fisiología , Proteínas Mitocondriales/fisiología , FN-kappa B/metabolismo , Neoplasias/patología , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Humanos , Neoplasias/metabolismo
2.
J Cell Sci ; 124(Pt 11): 1911-24, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21558421

RESUMEN

Degradation of Myc protein is mediated by E3 ubiquitin ligases, including SCF(Fbw7) and SCF(Skp2), but much remains unknown about the mechanism of S-phase kinase-associated protein (Skp2)-mediated Myc degradation. In the present study, we show that upregulated Myc protein, which triggers the G1-S phase progression in response to growth-stimulatory signals, induces reactive oxygen species modulator 1 (Romo1) expression. Romo1 subsequently triggers Skp2-mediated ubiquitylation and degradation of Myc by a mechanism not previously reported in normal lung fibroblasts. We also show that reactive oxygen species (ROS) derived from steady-state Romo1 expression are necessary for cell cycle entry of quiescent cells. From this study, we suggest that the generation of ROS mediated by pre-existing Romo1 protein is required for Myc induction. Meanwhile, Romo1 expression induced by Myc during G1 phase stimulates Skp2-mediated Myc degradation in a negative-feedback mechanism.


Asunto(s)
Retroalimentación Fisiológica , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Línea Celular , Medio de Cultivo Libre de Suero/metabolismo , Regulación hacia Abajo , Fase G1 , Humanos , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-myc/genética , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Transcripción Genética , Ubiquitinación
3.
Gastroenterology ; 143(4): 1084-94.e7, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22749933

RESUMEN

BACKGROUND & AIMS: Chronic oxidative stress from reactive oxygen species (ROS) produced by the mitochondria promotes hepatocarcinogenesis and tumor progression. However, the exact mechanism by which mitochondrial ROS contributes to tumor cell invasion is not known. We investigated the role of ROS modulator 1 (Romo1) in hepatocellular carcinoma (HCC) development and tumor cell invasiveness. METHODS: We performed real-time, semi-quantitative, reverse transcriptase polymerase chain reaction; invasion and luciferase assays; and immunofluorescence and immunohistochemical analyses. The formation of pulmonary metastatic nodules after tumor cell injection was tested in severe combined immunodeficient mice. We analyzed Romo1 expression in HCC cell lines and tissues (n = 95). RESULTS: Expression of Romo1 was increased in HCC cells, compared with normal human lung fibroblast cells. Exogenous expression of Romo1 in HCC cells increased their invasive activity, compared with control cells. Knockdown of Romo1 in Hep3B and Huh-7 HCC cells reduced their invasive activity in response to stimulation with 12-O-tetradecanoylphorbol-13-acetate. Levels of Romo1 were increased compared with normal liver tissues in 63 of 95 HCC samples from patients. In HCC samples from patients, there was an inverse correlation between Romo1 overexpression and patient survival times. Increased levels of Romo1 also correlated with vascular invasion by the tumors, reduced differentiation, and larger tumor size. CONCLUSIONS: Romo1 is a biomarker of HCC progression that might be used in diagnosis. Reagents that inhibit activity of Romo1 and suppress mitochondrial ROS production, rather than eliminate up-regulated intracellular ROS, might be developed as cancer therapies.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Línea Celular Tumoral , Distribución de Chi-Cuadrado , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones , Persona de Mediana Edad , Invasividad Neoplásica , Modelos de Riesgos Proporcionales , Ratas , Factores de Riesgo
4.
Biochem Biophys Res Commun ; 439(2): 315-20, 2013 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-23867822

RESUMEN

Oxidant-mediated death of lung epithelial cells due to cigarette smoking plays an important role in pathogenesis in lung diseases such as idiopathic pulmonary fibrosis (IPF). However, the exact mechanism by which oxidants induce epithelial cell death is not fully understood. Reactive oxygen species (ROS) modulator 1 (Romo1) is localized in the mitochondria and mediates mitochondrial ROS production through complex III of the mitochondrial electron transport chain. Here, we show that Romo1 mediates mitochondrial ROS production and apoptosis induced by oxidative stress in lung epithelial cells. Hydrogen peroxide (H2O2) treatment increased Romo1 expression, and Romo1 knockdown suppressed the cellular ROS levels and cell death triggered by H2O2 treatment. In immunohistochemical staining of lung tissues from patients with IPF, Romo1 was mainly localized in hyperplastic alveolar and bronchial epithelial cells. Romo1 overexpression was detected in 14 of 18 patients with IPF. TUNEL-positive alveolar epithelial cells were also detected in most patients with IPF but not in normal controls. These findings suggest that Romo1 mediates apoptosis induced by oxidative stress in lung epithelial cells.


Asunto(s)
Células Epiteliales/patología , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Muerte Celular , Línea Celular , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/citología , Proteínas de la Membrana/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Mitocondriales/genética
5.
Biochem Biophys Res Commun ; 408(1): 149-53, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21473856

RESUMEN

The mutant K-Ras elevates intracellular reactive oxygen species (ROS) levels and leads to oxidative DNA damage, resulting in malignant cell transformation. Ras association domain family 1 isoform A (RASSF1A) is known to play a role as a Ras effector. However, the suppressive effect of RASSF1A on K-RasV12-induced ROS increase and DNA damage has not been identified. Here, we show that RASSF1A blocks K-RasV12-triggered ROS production. RASSF1A expression also inhibits oxidative DNA damage and chromosomal damage. From the results obtained in this study, we suggest that RASSF1A regulates the cellular ROS levels enhanced by the Ras signaling pathway, and that it may function as a tumor suppressor by suppressing DNA damage caused by activated Ras.


Asunto(s)
Daño del ADN , Genes ras , Especies Reactivas de Oxígeno/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular Tumoral , Humanos , Ratones , Células 3T3 NIH , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética
6.
Int J Oncol ; 46(5): 2021-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25673177

RESUMEN

Reactive oxygen species (ROS) are important contributors to tumor cell invasion. ROS enhanced by reactive oxygen species modulator 1 (Romo1) expression has been reported to increase invasive potential and constitutive activation of nuclear factor-κB (NF-κB) in hepatocellular carcinoma (HCC). Therefore, we investigated whether constitutive NF-κB activation due to Romo1 expression is associated with breast cancer tumor cell invasion. In this study, we show that oxidative stress-induced invasion is mediated by Romo1 expression. The Romo1-induced increase of invasive activity was blocked by an inhibitor of κB kinase (IKK). These results demonstrate that tumor cell invasion in response to oxidative stress is associated with Romo1 expression and the NF-κB signaling pathway. Romo1 is therefore a promising therapeutic target for diseases characterized by NF-κB deregulation.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo , Western Blotting , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Ensayo de Cambio de Movilidad Electroforética , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Invasividad Neoplásica
7.
Free Radic Res ; 43(8): 729-37, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19513905

RESUMEN

Reactive oxygen species (ROS) steady-state levels are required for entry into the S phase of the cell cycle in normal cells, as well as in tumour cells. However, the contribution of mitochondrial ROS to normal cell proliferation has not been well investigated thus far. A previous report showed that Romo1 was responsible for the high ROS levels in tumour cells. Here, we show that endogenous ROS generated by Romo1 are indispensable for cell cycle transition from G1 to S phase in normal WI-38 human lung fibroblasts. The ROS level in these cells was down-regulated by Romo1 knockdown, resulting in cell cycle arrest in the G1 phase. This arrest was associated with an increase in the level of p27(Kip1). These results demonstrate that mitochondrial ROS generated by Romo1 expression is required for normal cell proliferation and it is suggested that Romo1 plays an important role in redox signalling during normal cell proliferation.


Asunto(s)
Ciclo Celular/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Fibroblastos/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas de la Membrana/fisiología , Mitocondrias/metabolismo , Proteínas Mitocondriales/fisiología , Especies Reactivas de Oxígeno/metabolismo , División Celular , Línea Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , ARN Interferente Pequeño/farmacología
8.
J Biol Chem ; 283(48): 33763-71, 2008 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-18836179

RESUMEN

Persistent accumulation of DNA damage induced by reactive oxygen species (ROS) is proposed to be a major contributor toward the aging process. Furthermore, an increase in age-associated ROS is strongly correlated with aging in various species, including humans. Here we showed that the enforced expression of the ROS modulator 1 (Romo1) triggered premature senescence by ROS production, and this also contributed toward induction of DNA damage. Romo1-derived ROS was found to originate in the mitochondrial electron transport chain. Romo1 expression gradually increased in proportion to population doublings of IMR-90 human fibroblasts. An increase in ROS production in these cells with high population doubling was blocked by the Romo1 knockdown using Romo1 small interfering RNA. Romo1 knockdown also inhibited the progression of replicative senescence. Based on these results, we suggest that age-related ROS levels increase, and this contributes to replicative senescence, which is directly associated with Romo1 expression.


Asunto(s)
Senescencia Celular/fisiología , Daño del ADN/fisiología , Fibroblastos/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas de la Membrana/biosíntesis , Proteínas Mitocondriales/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Línea Celular , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Fibroblastos/citología , Humanos , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , ARN Interferente Pequeño
9.
Biochem Biophys Res Commun ; 359(2): 304-10, 2007 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-17537404

RESUMEN

While acute oxidative stress triggers cell apoptosis or necrosis, persistent oxidative stress induces genomic instability and has been implicated in tumor progression and drug resistance. In a previous report, we demonstrated that reactive oxygen species modulator 1 (Romo1) expression was up-regulated in most cancer cell lines and suggested that increased Romo1 expression might confer chronic oxidative stress to tumor cells. In this study, we show that enforced Romo1 expression induces reactive oxygen species (ROS) production in the mitochondria leading to massive cell death. However, tumor cells that adapt to oxidative stress by increasing manganese superoxide dismutase (MnSOD), Prx I, and Bcl-2 showed drug resistance to 5-FU. To elucidate the relationship between 5-FU-induced ROS production and Romo1 expression, Romo1 siRNA was used to inhibit 5-FU-triggered Romo1 induction. Romo1 siRNA treatment efficiently blocked 5-FU-induced ROS generation, demonstrating that 5-FU treatment stimulated ROS production through Romo1 induction. Based on these results we suggest that cellular adaptive response to Romo1-induced ROS is another mechanism of drug resistance to 5-FU and Romo1 expression may provide a new clinical implication in drug resistance of cancer chemotherapy.


Asunto(s)
Resistencia a Antineoplásicos , Fluorouracilo/farmacología , Proteínas de la Membrana/biosíntesis , Proteínas Mitocondriales/biosíntesis , Especies Reactivas de Oxígeno , Antimetabolitos Antineoplásicos/farmacología , Ciclo Celular , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Peróxido de Hidrógeno/farmacología , Potencial de la Membrana Mitocondrial , Estrés Oxidativo , ARN Interferente Pequeño/metabolismo , Espectrometría de Fluorescencia , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda