Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Mol Med (Berl) ; 97(1): 37-47, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30406363

RESUMEN

In radiotherapy, radiation (IR)-induced lung fibrosis has severe and dose-limiting side effects. To elucidate the molecular effects of IR fibrosis, we examined the fibrosis process in irradiated mouse lung tissues. High focal IR (90 Gy) was exposed to a 3-mm volume of the left lung in C57BL6 mice. In the diffused irradiation, 20 Gy dose delivered with a 7-mm collimator almost covered the entire left lung. Histological examination for lung tissues of both irradiated and neighboring regions was done for 4 weeks after irradiation. Long-term effects (12 months) of 20Gy IR were compared on a diffuse region of the left lung and non-irradiated right lung. Fibrosis was initiated as early as 2 weeks after IR in the irradiated lung region and neighboring region. Upregulation of gtse1 in both 90Gy-irradiated and neighboring regions was observed. Upregulation of fgl1 in both 20Gy diffused irradiated and non-irradiated lungs was identified. When gtse1 or flg1 was knock-downed, TGFß or IR-induced epithelial-mesenchymal transition was inhibited, accompanied with the inhibition of cellular migration, suggesting fibrosis responsible genes. Immunofluorescence analysis using mouse fibrotic lung tissues suggested that fibrotic regions showed increased expressions of Gtse1 and Fgl1, indicating novel molecular signatures of gtse1and fgl1 for IR-induced lung fibrosis. Even though their molecular mechanisms and IR doses or irradiated volumes for lung fibrosis may be different, these genes may be novel targets for understanding IR-induced lung fibrosis and in treatment strategies. KEY MESSAGES: Upregulation of gtse1 by 90Gy focal irradiation and upregulation of fgl1 by 20Gy diffused irradiation are identified in mouse lung fibrosis model. Gtse1 and Fgl1 are involved in radiation or TGFß-induced epithelial-mesenchymal transition. Radiation-induced fibrotic regions of mouse lungs showed increased expressions of Gtse1 and Fgl1. Gtse1 and Fgl1 are suggested to be novel targets for radiation-induced lung fibrosis.


Asunto(s)
Fibrinógeno/genética , Proteínas Asociadas a Microtúbulos/genética , Fibrosis Pulmonar/genética , Neumonitis por Radiación/genética , Células A549 , Animales , Línea Celular , Transición Epitelial-Mesenquimal/efectos de la radiación , Humanos , Pulmón/metabolismo , Pulmón/patología , Pulmón/efectos de la radiación , Masculino , Ratones Endogámicos C57BL , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/patología , Neumonitis por Radiación/etiología , Neumonitis por Radiación/patología , Radioterapia/efectos adversos , Transcriptoma/efectos de la radiación , Regulación hacia Arriba/efectos de la radiación
2.
Int J Radiat Biol ; 93(2): 184-193, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27645719

RESUMEN

PURPOSE: The molecular effects of focal exposure of limited lung volumes to high-dose per fraction irradiation (HDFR) such as stereotactic body radiotherapy (SBRT) have not been fully characterized. In this study, we used such an irradiation system and identified the genes and proteins after HDFR to mouse lung, similar to those associated with human therapy. METHODS AND MATERIALS: High focal radiation (90 Gy) was applied to a 3-mm volume of the left lung of C57BL6 mice using a small-animal stereotactic irradiator. As well as histological examination for lungs, a cDNA micro array using irradiated lung tissues and a protein array of sera were performed until 4 weeks after irradiation, and radiation-responsive genes and proteins were identified. For comparison, the long-term effects (12 months) of 20 Gy radiation wide-field dose to the left lung were also investigated. RESULTS: The genes ermap, epb4.2, cd200r3 (up regulation) and krt15, hoxc4, gdf2, cst9, cidec, and bnc1 (down-regulation) and the proteins of AIF, laminin, bNOS, HSP27, ß-amyloid (upregulation), and calponin (downregulation) were identified as being responsive to 90 Gy HDFR. The gdf2, cst9, and cidec genes also responded to 20 Gy, suggesting that they are universal responsive genes in irradiated lungs. No universal proteins were identified in both 90 Gy and 20 Gy. Calponin, which was downregulated in protein antibody array analysis, showed a similar pattern in microarray data, suggesting a possible HDFR responsive serum biomarker that reflects gene alteration of irradiated lung tissue. These genes and proteins also responded to the lower doses of 20 Gy and 50 Gy HDFR. CONCLUSIONS: These results suggest that identified candidate genes and proteins are HDFR-specifically expressed in lung damage induced by HDFR relevant to SBRT in humans.


Asunto(s)
Regulación de la Expresión Génica/efectos de la radiación , Pulmón/metabolismo , Pulmón/efectos de la radiación , Proteoma/metabolismo , Neumonitis por Radiación/metabolismo , Radiocirugia/efectos adversos , Animales , Relación Dosis-Respuesta en la Radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Hipofraccionamiento de la Dosis de Radiación , Neumonitis por Radiación/etiología
3.
BMC Genet ; 17: 29, 2016 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-26818610

RESUMEN

BACKGROUND: Despite the emergence of stereotactic body radiotherapy (SBRT) for treatment of medically inoperable early-stage non-small-cell lung cancer patients, the molecular effects of focal exposure of limited lung volumes to high-dose radiation have not been fully characterized. This study was designed to identify molecular changes induced by focal high-dose irradiation using a mouse model of SBRT. RESULTS: Central areas of the mouse left lung were focally-irradiated (3 mm in diameter) with a single high-dose of radiation (90 Gy). Temporal changes in gene expression in the irradiated and non-irradiated neighboring lung regions were analyzed by microarray. For comparison, the long-term effect (12 months) of 20 Gy radiation on a diffuse region of lung was also measured. The majority of genes were down-regulated in the focally-irradiated lung areas at 2 to 3 weeks after irradiation. This pattern of gene expression was clearly different than gene expression in the diffuse region of lungs exposed to low-dose radiation. Ontological and pathway analyses indicated these down-regulated genes were mainly associated with organ development. Although the number was small, genes that were up-regulated after focal irradiation were associated with immune-related functions. The temporal patterns of gene expression and the associated biological functions were also similar in non-irradiated neighboring lung regions, although statistical significance was greatly reduced when compared with those from focally-irradiated areas of the lung. From network analysis of temporally regulated genes, we identified inter-related modules associated with diverse functions, including organ development and the immune response, in both the focally-irradiated regions and non-irradiated neighboring lung regions. CONCLUSIONS: Focal exposure of lung tissue to high-dose radiation induced expression of genes associated with organ development and the immune response. This pattern of gene expression was also observed in non-irradiated neighboring areas of lung tissue, indicating a global lung response to focal high-dose irradiation.


Asunto(s)
Regulación de la Expresión Génica/efectos de la radiación , Pulmón/efectos de la radiación , Animales , Relación Dosis-Respuesta en la Radiación , Regulación hacia Abajo , Perfilación de la Expresión Génica , Ontología de Genes , Inmunidad/genética , Inmunidad/efectos de la radiación , Pulmón/crecimiento & desarrollo , Pulmón/inmunología , Masculino , Ratones , Fibrosis Pulmonar , Radiocirugia , Regulación hacia Arriba
4.
Oncotarget ; 6(30): 29712-24, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26359349

RESUMEN

A novel role for HSF1 as an inhibitor of non-homologous end joining (NHEJ) repair activity was identified. HSF1 interacted directly with both of the N-terminal sequences of the Ku70 and Ku86 proteins, which inhibited the endogenous heterodimeric interaction between Ku70 and Ku86. The blocking of the Ku70 and Ku86 interaction by HSF1 induced defective NHEJ repair activity and ultimately activated genomic instability after ionizing radiation (IR), which was similar to effects seen in Ku70 or Ku80 knockout cells. The binding activity between HSF1 and Ku70 or Ku86 was dependent on DNA damage response such as IR exposure, but not on the heat shock mediated transcriptional activation of HSF1. Moreover, the posttranslational modification such as phosphorylation, acetylation and sumoylation of HSF1 did not alter the binding activities of HSF1-Ku70 or HSF1-Ku86. Furthermore, the defect in DNA repair activity by HSF1 was observed regardless of p53 status. Rat mammary tumors derived using dimethylbenz(a)anthracence revealed that high levels of HSF1 expression which correlate with aggressive malignancy, interfered with the binding of Ku70-Ku80. This data suggests that HSF1 interacts with both Ku70 and Ku86 to induce defective NHEJ repair activity and genomic instability, which in turn suggests a novel mechanism of HSF1-mediated cellular carcinogenesis.


Asunto(s)
Antígenos Nucleares/metabolismo , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Factores de Transcripción/metabolismo , Animales , Antígenos Nucleares/genética , Línea Celular Tumoral , Células Cultivadas , Daño del ADN , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/citología , Femenino , Fibroblastos/citología , Fibroblastos/efectos de la radiación , Células HEK293 , Factores de Transcripción del Choque Térmico , Humanos , Immunoblotting , Inmunohistoquímica , Autoantígeno Ku , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Ratones Noqueados , Radiación Ionizante , Ratas Sprague-Dawley , Factores de Transcripción/genética
5.
Curr Alzheimer Res ; 12(5): 481-92, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26017559

RESUMEN

The involvement of radiofrequency electromagnetic fields (RF-EMF) in the neurodegenerative disease, especially Alzheimer's disease (AD), has received wide consideration, however, outcomes from several researches have not shown consistency. In this study, we determined whether RF-EMF influenced AD pathology in vivo using Tg-5xFAD mice as a model of AD-like amyloid ß (Aß) pathology. The transgenic (Tg)-5xFAD and wild type (WT) mice were chronically exposed to RF-EMF for 8 months (1950 MHz, SAR 5W/kg, 2 hrs/day, 5 days/week). Notably, chronic RFEMF exposure significantly reduced not only Aß plaques, APP, and APP carboxyl-terminal fragments (CTFs) in whole brain including hippocampus and entorhinal cortex but also the ratio of Aß42 and Aß40 peptide in the hippocampus of Tg-5xFAD mice. We also found that parenchymal expression of ß-amyloid precursor protein cleaving enzyme 1(BACE1) and neuroinflammation were inhibited by RF-EMF exposure in Tg-5xFAD. In addition, RF-EMF was shown to rescue memory impairment in Tg-5xFAD. Moreover, gene profiling from microarray data using hippocampus of WT and Tg- 5xFAD following RF-EMF exposure revealed that 5 genes (Tshz2, Gm12695, St3gal1, Isx and Tll1), which are involved in Aß, are significantly altered inTg-5xFAD mice, exhibiting different responses to RF-EMF in WT or Tg-5xFAD mice; RF-EMF exposure in WT mice showed similar patterns to control Tg-5xFAD mice, however, RF-EMF exposure in Tg- 5xFAD mice showed opposite expression patterns. These findings indicate that chronic RF-EMF exposure directly affects Aß pathology in AD but not in normal brain. Therefore, RF-EMF has preventive effects against AD-like pathology in advanced AD mice with a high expression of Aß, which suggests that RF-EMF can have a beneficial influence on AD.


Asunto(s)
Enfermedad de Alzheimer/terapia , Magnetoterapia/métodos , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Astrocitos/patología , Astrocitos/fisiología , Reacción de Prevención/fisiología , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Trastornos de la Memoria/terapia , Ratones Transgénicos , Microglía/patología , Microglía/fisiología , Actividad Motora/fisiología , Presenilina-1/genética , Presenilina-1/metabolismo
6.
J Nat Prod ; 77(5): 1123-9, 2014 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-24746225

RESUMEN

Heat shock factor 1 (HSF1) is a transcription factor that regulates expression of heat shock protein (HSP) genes in response to stress. HSPs are expressed at high levels in a wide range of tumors. It has been reported that HSF1 and HSPs are associated closely in tumorigenesis. In the present study, a screen was performed using a luciferase reporter under the control of a heat shock element to find inhibitors of HSF1 activity, and 2,4-bis(4-hydroxybenzyl)phenol (1), isolated from the rhizomes of Gastrodia elata, was identified as an active compound. This substance effectively inhibited HSF1 activity and decreased levels of HSP27 and HSP70. Compound 1 induced the degradation of HSF1 protein through dephosphorylation of HSF1 on S326, which decreases HSF1 protein stability. In addition, 1 also induced growth arrest and apoptosis of NCI-H460 human lung cancer cells. Markers of apoptosis, such as cleaved PARP and cleaved caspase-3, were detected after treatment with 1. Furthermore, cotreatment with 1 and conventional anticancer modalities such as paclitaxel, cisplatin, or ionizing radiation potentiated their effects on lung cancer cells. These results suggest that inhibition of HSF1 by 1 may help overcome resistance to conventional anticancer modalities in HSF1-overexpressed cancer cells.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Fenoles/farmacología , Factores de Transcripción/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Compuestos de Bencidrilo/química , Caspasa 3/metabolismo , Proteínas de Choque Térmico HSP27/análisis , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas HSP70 de Choque Térmico/análisis , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Factores de Transcripción del Choque Térmico , Humanos , Neoplasias Pulmonares , Estructura Molecular , Fenoles/química , Reacción en Cadena de la Polimerasa
7.
PLoS One ; 9(1): e85509, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24465581

RESUMEN

Matrix metalloproteinases regulate pathophysiological events by processing matrix proteins and secreted proteins. Previously, we demonstrated that soluble heat shock protein B1 (HSPB1) is released primarily from endothelial cells (ECs) and regulates angiogenesis via direct interaction with vascular endothelial growth factor (VEGF). Here we report that MMP9 can cleave HSPB1 and release anti-angiogenic fragments, which play a key role in tumorprogression. We mapped the cleavage sites and explored their physiological relevance during these processing events. HSPB1 cleavage by MMP9 inhibited VEGF-induced ECs activation and the C-terminal HSPB1 fragment exhibited more interaction with VEGF than did full-length HSPB1. HSPB1 cleavage occurs during B16F10 lung progression in wild-type mice. Also, intact HSPB1 was more detected on tumor endothelium of MMP9 null mice than wild type mice. Finally, we confirmed that secretion of C-terminal HSPB1 fragment was significantly inhibited lung and liver tumor progression of B16F10 melanoma cells and lung tumor progression of CT26 colon carcinoma cells, compared to full-length HSPB1. These data suggest that in vivo MMP9-mediated processing of HSPB1 acts to regulate VEGF-induced ECs activation for tumor progression, releasing anti-angiogenic HSPB1 fragments. Moreover, these findings potentially explain an anti-target effect for the failure of MMP inhibitors in clinical trials, suggesting that MMP inhibitors may have pro-tumorigenic effects by reducing HSPB1 fragmentation.


Asunto(s)
Carcinogénesis/patología , Proteínas de Choque Térmico HSP27/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Metaloproteinasa 9 de la Matriz/genética , Ratones , Chaperonas Moleculares , Fosforilación , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Sarcoma/genética , Sarcoma/metabolismo , Sarcoma/patología , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/metabolismo , Neoplasias de los Tejidos Blandos/patología
8.
J Basic Microbiol ; 54(1): 72-80, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23456650

RESUMEN

The Schizosaccharomyces pombe msrB(+) gene encoding methionine-R-sulfoxide reductase (MsrB) was cloned into the shuttle vector pRS316 to generate the recombinant plasmid pFMetSO. The msrB(+) mRNA level was significantly increased in the S. pombe cells harboring pFMetSO, indicating that the cloned msrB(+) gene is functioning. In the presence of 0.1 mM L-methionine-(R,S)-sulfoxide, the S. pombe cells harboring pFMetSO could grow normally but the growth of the vector control cells was almost arrested. The S. pombe cells harboring pFMetSO exhibited the enhanced growth on the minimal medium plates with stress-inducing agents, such as hydrogen peroxide, superoxide radical-generating menadione (MD), nitric oxide (NO)-generating sodium nitroprusside (SNP), and cadmium (Cd), when compared with the vector control cells. They also gave rise to the enhanced growth at the high incubation temperature of 37 °C than the vector control cells. The S. pombe cells harboring pFMetSO contained lower reactive oxygen species (ROS) and higher total glutathione (GSH) levels than the vector control cells. In brief, the S. pombe MsrB plays a protective role against oxidative, nitrosative, and thermal stresses, and is involved in diminishing intracellular ROS level.


Asunto(s)
Metionina Sulfóxido Reductasas/metabolismo , Schizosaccharomyces/fisiología , Secuencia de Aminoácidos , Secuencia de Bases , Glutatión/metabolismo , Metionina Sulfóxido Reductasas/genética , Datos de Secuencia Molecular , Especies Reactivas de Oxígeno/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/crecimiento & desarrollo , Estrés Fisiológico
9.
J Biol Chem ; 288(45): 32506-32516, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24085291

RESUMEN

Our previous study suggested that the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) interacts with Snail1, which affects genomic instability, sensitivity to DNA-damaging agents, and migration of tumor cells by reciprocal regulation between DNA-PKcs and Snail1. Here, we further investigate that a peptide containing 7-amino acid sequences (amino acids 15-21) of Snail1 (KPNYSEL, SP) inhibits the endogenous interaction between DNA-PKcs and Snail1 through primary interaction with DNA-PKcs. SP restored the inhibited DNA-PKcs repair activity and downstream pathways. On the other hand, DNA-PKcs-mediated phosphorylation of Snail1 was inhibited by SP, which resulted in decreased Snail1 stability and Snail1 functions. However, these phenomena were only shown in p53 wild-type cells, not in p53-defective cells. From these results, it is suggested that interfering with the protein interaction between DNA-PKcs and Snail1 might be an effective strategy for sensitizing cancer cells and inhibiting tumor migration, especially in both Snail1-overexpressing and DNA-PKcs-overexpressing cancer cells with functional p53.


Asunto(s)
Proteína Quinasa Activada por ADN/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Proteínas de Neoplasias/biosíntesis , Neoplasias/tratamiento farmacológico , Proteínas Nucleares/biosíntesis , Péptidos/farmacología , Factores de Transcripción/biosíntesis , Animales , Línea Celular Tumoral , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Ratones , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/genética , Estabilidad Proteica/efectos de los fármacos , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
10.
Int J Radiat Biol ; 88(4): 374-80, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22191540

RESUMEN

PURPOSE: Epidemiological studies have demonstrated a possible correlation between exposure to extremely low-frequency magnetic fields (ELF-MF) and cancer. However, this correlation has yet to be definitively confirmed by epidemiological studies. The principal objective of this study was to assess the effects of 60 Hz magnetic fields in a normal cell line system, and particularly in combination with various external factors, via micronucleus (MN) assays. MATERIALS AND METHODS: Mouse embryonic fibroblast NIH3T3 cells and human lung fibroblast WI-38 cells were exposed for 4 h to a 60 Hz, 1 mT uniform magnetic field with or without ionizing radiation (IR, 2 Gy), H(2)O(2) (100 µM) and cellular myelocytomatosis oncogene (c-Myc) activation. RESULTS: The results obtained showed no significant differences between the cells exposed to ELF-MF alone and the unexposed cells. Moreover, no synergistic effects were observed when ELF-MF was combined with IR, H(2)O(2), and c-Myc activation. CONCLUSIONS: Our results demonstrate that ELF-MF did not enhance MN frequency by IR, H(2)O(2) and c-Myc activation.


Asunto(s)
Campos Electromagnéticos/efectos adversos , Peróxido de Hidrógeno/farmacología , Proteínas Proto-Oncogénicas c-myc/genética , Activación Transcripcional , Animales , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Humanos , Ratones , Pruebas de Micronúcleos , Células 3T3 NIH
11.
Mol Cancer Ther ; 9(4): 825-32, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20354123

RESUMEN

Peroxiredoxins (PRDX) are a family of thiol-dependent peroxidases. Among the six mammalian members of this family, PRDX6 is the only protein that additionally exhibits phospholipase A(2) (PLA(2)) activity. The physiologic role of this interesting PRDX6 feature is largely unknown at present. In this study, we show that PRDX6 increases the metastatic potential of lung cancer cells. Functional analyses of the enzymatic activities of PRDX6, using specific pharmacologic inhibitors and mutagenesis studies, reveal that both peroxidase and PLA(2) activities are required for metastasis. Specifically, peroxidase activity facilitates the growth of cancer cells, and PLA(2) activity promotes invasiveness. Further investigation of the latter event discloses that PLA(2) activity promotes accumulation of arachidonic acid, which, in turn, induces the invasive pathway involving p38 kinase, phosphoinositide 3-kinase, Akt, and urokinase-type plasminogen activator. This study is the first to define the functions of the enzymatic activities of PRDX6 in metastasis and to show the involvement of arachidonic acid in PRDX6 action in intact cells. These novel findings provide a significant step toward elucidating the role of PRDX6 in cancer and the mechanism of its action. Mol Cancer Ther; 9(4); 825-32. (c)2010 AACR.


Asunto(s)
Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Peroxiredoxina VI/metabolismo , Fosfolipasas A2/metabolismo , Animales , Ácido Araquidónico/metabolismo , Peso Corporal , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Metástasis de la Neoplasia , Tamaño de los Órganos , Transfección
12.
Cancer Sci ; 101(6): 1417-23, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20331635

RESUMEN

Previous reports suggest that, in addition to its therapeutic effects, ionizing radiation (IR) increases the invasiveness of surviving cancer cells. Here, we demonstrate that this activity of IR in lung cancer cells is mediated by a signaling pathway involving p38 kinase, phosphoinositide 3-kinase, Akt, and matrix metalloproteinase (MMP-2). The invasion-promoting doses of IR also increased and reduced the levels of vimentin and E-cadherin, respectively, both of which are markers for the epithelial-mesenchymal transition (EMT). Interestingly, all of these malignant actions of IR were mimicked by the overexpression of Bcl-X(L), a pro-survival member of the Bcl-2 family, in lung cancer cells. Moreover, both RNA and protein levels of Bcl-X(L) were elevated upon irradiation of the cells, and the prevention of this event using small-interfering RNAs of Bcl-X(L) reduced the ability of IR to promote invasion signals and EMT-associated events. This suggests that Bcl-X(L) functions as a signaling mediator of the malignant effects of IR. It was also demonstrated that IR enhances signal transducer and activator of transcription 3 (STAT3) phosphorylation, and the reduction of STAT3 levels via RNA interference prevented IR-induced Bcl-X(L) accumulation, and thus all the tested Bcl-X(L)-dependent events. Overall, the data suggest that IR induces Bcl-X(L) accumulation via STAT3, which then promotes cancer cell invasion and EMT-associated markers. Our findings demonstrate a novel function of Bcl-X(L) in cancer, and also advance our understanding of the malignant actions of IR significantly.


Asunto(s)
Rayos gamma/efectos adversos , Neoplasias Pulmonares/patología , Factor de Transcripción STAT3/fisiología , Proteína bcl-X/fisiología , Línea Celular Tumoral , Células Epiteliales/patología , Humanos , Mesodermo/patología , Invasividad Neoplásica
13.
Mol Cells ; 28(6): 583-8, 2009 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-19937138

RESUMEN

The peroxiredoxin family of peroxidase has six mammalian members (Prx 1-6). Considering their frequent up-regulation in cancer cells, Prxs may contribute to cancer cells' survival in face of oxidative stress. Here, we show that Prx 6 promotes the invasiveness of lung cancer cells, accompanied by an increase in the activity of phosphoinositide 3-kinase (PI3K), the phosphorylation of p38 kinase and Akt, and the protein levels of uPA. Functional studies reveal that these components support Prx 6-induced invasion in the sequence p38 kinase/PI3K, Akt, and uPA. The findings provide a new understanding of the action of Prx 6 in cancer.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Peroxiredoxina VI/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Neoplasias Pulmonares/genética , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Estrés Oxidativo/genética , Peroxiredoxina VI/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Transgenes/genética , Activador de Plasminógeno de Tipo Uroquinasa/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
J Microbiol ; 47(1): 60-7, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19229492

RESUMEN

The structural gene encoding bacterioferritin comigratory protein (Bcp) was amplified using PCR from the genomic DNA of Schizosaccharomyces pombe, and transferred into the shuttle vector pRS316 to generate the recombinant plasmid pBCPlO. The bcp(+) mRNA level in the pBCPlO-containing yeast cells was significantly higher than that in the control yeast cells, indicating that the cloned gene is functioning. The S. pombe cells harboring the plasmid pBCPIO exhibited higher survival on the solid minimal media with hydrogen peroxide, tert-BOOH or cadmium than the control yeast cells. They also exhibited enhanced cellular viability in the liquid media containing the stressful agents. The increased viabilities of the fission yeast cells harboring the plasmid pBCP10 were also obtained with 0.4% glucose or 0.4% sucrose as a sole carbon source, and nitrogen starvation, compared with those of the control yeast cells. The total glutathione (GSH) content and total GSH/GSSG ratio were significantly higher in the yeast cells harboring the plasmid pBCP10 than in the control yeast cells. In brief, the S. pombe Bcp plays a protective role in the defensive response to oxidative stress possibly via up-regulation of total and reduced glutathione levels.


Asunto(s)
Glutatión/biosíntesis , Viabilidad Microbiana/genética , Estrés Oxidativo/genética , Peroxirredoxinas/biosíntesis , Peroxirredoxinas/genética , Proteínas de Schizosaccharomyces pombe/biosíntesis , Proteínas de Schizosaccharomyces pombe/genética , Schizosaccharomyces/enzimología , Schizosaccharomyces/genética , Secuencia de Aminoácidos , Secuencia de Bases , Secuencia de Consenso/genética , ADN de Hongos/aislamiento & purificación , ADN Recombinante/biosíntesis , Datos de Secuencia Molecular , Especies Reactivas de Oxígeno/metabolismo , Schizosaccharomyces/química , Proteínas de Schizosaccharomyces pombe/química , Análisis de Secuencia de ADN , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA