Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Radiat Biol ; 100(2): 220-235, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37812149

RESUMEN

PURPOSE: Due to the expanding repertoire of treatment devices that use radiation, the possibility of exposure to both low-dose and high-dose radiation continues to increase. Skin is the outermost part of the body and thus directly exposed to radiation-induced damage. In particular, the skin of diabetes patients is fragile and easily damaged by external stimuli, such as radiation. However, damage and cellular responses induced by ionizing irradiation in diabetic skin have not been explored in detail. In this study, we investigated the effects of several irradiation dose on normal keratinocytes and those from type II diabetes patients, with particular focus on DNA damage. MATERIALS AND METHODS: Cellular responses to low-dose radiation (0.1 Gy) and high-dose radiation (0.5 and 2 Gy) were evaluated. Cell cycle analysis was conducted via flow cytometry and cell viability analyzed using the 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay. Proteins related to the DNA damage response (DDR) and repair signaling pathways and apoptosis were detected via immunoblot analysis. Apoptosis and cell differentiation were additionally examined in 3D skin organoids using immunohistochemistry. RESULTS: Compared to respective control groups, no significant changes were observed in cell cycle, DDR and repair mechanisms, cell survival, and differentiation in response to 0.1 Gy irradiation in both normal and diabetes type II keratinocytes. On the other hand, the cell cycle showed an increase in the G2/M phase in both cell types following exposure to 2 Gy irradiation. At radiation doses 2 Gy, activation of the DDR and repair signaling pathways, apoptosis, and cell differentiation were increased and viability was decreased in both cell types. Notably, these differences were more pronounced in normal than diabetes type II keratinocytes. CONCLUSIONS: Normal keratinocytes respond more strongly to radiation-induced damage and recovery than diabetes type II keratinocytes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Humanos , Diabetes Mellitus Tipo 2/radioterapia , Queratinocitos/efectos de la radiación , Radiación Ionizante , Dosis de Radiación , Supervivencia Celular/efectos de la radiación , Daño del ADN , Apoptosis/efectos de la radiación , Relación Dosis-Respuesta en la Radiación
2.
Exp Dermatol ; 32(10): 1706-1716, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37421206

RESUMEN

Melanoma is a notoriously radioresistant type of skin cancer. Elucidation of the specific mechanisms underlying radioresistance is necessary to improve the clinical efficacy of radiation therapy. To identify the key factors contributing to radioresistance, five melanoma cell lines were selected for study and genes that were upregulated in relatively radioresistant melanomas compared with radiosensitive melanoma cells determined via RNA sequencing technology. In particular, we focused on cyclin D1 (CCND1), a well known cell cycle regulatory molecule. In radiosensitive melanoma, overexpression of cyclin D1 reduced apoptosis. In radioresistant melanoma cell lines, suppression of cyclin D1 with a specific inhibitor or siRNA increased apoptosis and decreased cell proliferation in 2D and 3D spheroid cultures. In addition, we observed increased expression of γ-H2AX, a molecular marker of DNA damage, even at a later time after γ-irradiation, under conditions of inhibition of cyclin D1, with a response pattern similar to that of radiosensitive SK-Mel5. In the same context, expression and nuclear foci formation of RAD51, a key enzyme for homologous recombination (HR), were reduced upon inhibition of cyclin D1. Downregulation of RAD51 also reduced cell survival to irradiation. Overall, suppression of cyclin D1 expression or function led to reduced radiation-induced DNA damage response (DDR) and triggered cell death. Our collective findings indicate that the presence of increased cyclin D1 potentially contributes to the development of radioresistance through effects on RAD51 in melanoma and could therefore serve as a therapeutic target for improving the efficacy of radiation therapy.


Asunto(s)
Ciclina D1 , Reparación del ADN , Melanoma , Recombinasa Rad51 , Humanos , Apoptosis , Línea Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Melanoma/genética , Melanoma/radioterapia , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Tolerancia a Radiación/genética
3.
Anim Cells Syst (Seoul) ; 26(5): 203-213, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36275445

RESUMEN

Brain type of creatine kinase (CKB) regulates energy homeostasis by reversibly transferring phosphate groups between phosphocreatine and ATP at sites of high energy demand. Several types of cancer cells exhibit upregulated CKB expression, but the function of CKB in cancer cells remains unclear. In this study, we investigated the function of CKB in breast cancer by overexpressing CKB in MDA-MB-231 cells. The overexpression of CKB did not affect cell growth rate, cell cycle distribution, ATP level or key mediators of aerobic glycolysis and lactate dehydrogenase isoform levels. Meanwhile, CKB overexpression did increase resistance to doxorubicin. TGF-ß-induced Smad phosphorylation and Smad-dependent transcriptional activity were significantly up-regulated by CKB expression without changes in inhibitory Smad protein levels. Moreover, treatment with TGF-ß considerably enhanced cell viability during doxorubicin treatment and decreased doxorubicin-induced apoptosis in CKB-expressing MDA-MB-231 cells compared to control cells. These results suggest that CKB attenuates doxorubicin-induced apoptosis and potentiates resistance to doxorubicin by enhancing TGF-ß signaling in MDA-MB-231 cells.

4.
Exp Dermatol ; 29(6): 562-569, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32298492

RESUMEN

Melanoma is a deadly type of skin cancer that is particularly difficult to treat owing to its resistance to radiation therapy. Here, we attempted to determine the key proteins responsible for melanoma radioresistance, with the aim of improving disease response to radiation therapy. Two melanoma cell lines, SK-Mel5 and SK-Mel28, with different radiosensitivities were analysed via RNA-Seq (Quant-Seq) and target proteins with higher abundance in the more radioresistant cell line, SK-Mel28, identified. Among these proteins, integrin αvß3, a well-known molecule in cell adhesion, was selected for analysis. Treatment of SK-Mel28 cells with cilengitide, an integrin αvß3 inhibitor, as well as γ-irradiation resulted in more significant cell death than γ-irradiation alone. In addition, Akt, a downstream signal transducer of integrin αvß3, showed high basic activation in SK-Mel28 and was significantly decreased upon co-treatment with cilengitide and γ-irradiation. MK-2206, an Akt inhibitor, exerted similar effects on the SK-Mel28 cell line following γ-irradiation. Our results collectively demonstrate that the integrin αvß3-Akt signalling pathway contributes to radioresistance in SK-Mel28 cells, which may be manipulated to improve therapeutic options for melanoma.


Asunto(s)
Integrina alfaVbeta3/metabolismo , Melanoma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tolerancia a Radiación , Neoplasias Cutáneas/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Rayos gamma , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Integrina alfaVbeta3/antagonistas & inhibidores , Melanoma/radioterapia , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal , Neoplasias Cutáneas/radioterapia , Venenos de Serpiente/farmacología
5.
Cells ; 8(9)2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31438652

RESUMEN

An understanding of cellular processes that determine the response to ionizing radiation exposure is essential for improving radiotherapy and assessing risks to human health after accidental radiation exposure. Radiation exposure leads to many biological effects, but the mechanisms underlying the metabolic effects of radiation are not well known. Here, we investigated the effects of radiation exposure on the metabolic rate and mitochondrial bioenergetics in skeletal muscle. We show that ionizing radiation increased mitochondrial protein and mass and enhanced proton leak and mitochondrial maximal respiratory capacity, causing an increase in the fraction of mitochondrial respiration devoted to uncoupling reactions. Thus, mice and cells treated with radiation became energetically efficient and displayed increased fatty acid and amino acid oxidation metabolism through the citric acid cycle. Finally, we demonstrate that radiation-induced alterations in mitochondrial energy metabolism involved adenosine monophosphate-activated kinase signaling in skeletal muscle. Together, these results demonstrate that alterations in mitochondrial mass and function are important adaptive responses of skeletal muscle to radiation.


Asunto(s)
Metabolismo Energético , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Rayos X , Animales , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos ICR , Mitocondrias/efectos de la radiación , Músculo Esquelético/efectos de la radiación , Exposición a la Radiación , Irradiación Corporal Total
6.
Biochem Biophys Res Commun ; 506(4): 939-943, 2018 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-30396570

RESUMEN

Imatinib mesylate, commercially known as Gleevec/Glivec, is the first targeted anticancer drug that inhibits activity of the tyrosine kinases, c-ABL, c-KIT, and PDGFR. A number of studies have shown that treatment with imatinib mesylate elicits extracellular signal-related kinase (ERK) activation, which, in turn, has been shown to confer radioresistance. Here, we investigated whether treatment with imatinib mesylate protects skin-derived epithelial cells, including normal keratinocytes, immortalized HaCaT and A431 cancer cell lines, from the effects of γ-radiation. ERK activation was detected 30 min after imatinib mesylate treatment in all three cell lines. In cells exposed to γ-irradiation in the presence of imatinib mesylate, this activation of ERK was associated with a reduction in radiation-induced apoptosis and enhanced cell survival. Similar effects of imatinib mesylate treatment were observed following γ-irradiation of a three-dimensional human skin culture system that reproduces a fully differentiated epithelium. Collectively, our findings provide the evidence of a protective effect of imatinib mesylate against the effects of γ-irradiation on epithelial-derived cells, regardless of their malignancy status.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/enzimología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Rayos gamma , Mesilato de Imatinib/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Citoprotección/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Células Epiteliales/efectos de los fármacos , Células Epiteliales/efectos de la radiación , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Queratinocitos/efectos de la radiación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Piel/citología
7.
J Dermatol Sci ; 90(1): 13-20, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29290529

RESUMEN

BACKGROUND: Histone deacetylase (HDAC) is an enzyme that regulates gene expression, cell cycle arrest, apoptosis and modulation of various pathways. HDAC inhibitors (HDACis) can modulate these pathways by hyper-acetylating target proteins, thereby acting as cancer chemotherapeutic agents. OBJECTIVE: One of HDACis, suberoylanilide hydroxamic acid (SAHA), has been found to regulate the Smad signaling pathway, by an as yet unclear mechanism. This study therefore investigated the mechanism by which SAHA regulates Smad signaling in the melanoma cell line SK-Mel-5. METHODS: Cell proliferation was assessed by MTT assays and fluorescence activated cell sorter (FACS) analysis. The activation of Smad signaling pathway was assessed by western blots analysis. The transcriptions of target genes were checked by RT-PCR and dual luciferase assay. RESULTS: Treatment with SAHA inhibited the proliferation of SK-Mel-5 cells, enhanced the phosphorylation of R-Smad, and up-regulated p21 protein. Surprisingly, R-Smad was also activated by conditioned medium from SAHA-treated SK-Mel-5 cells. An analysis of the conditioned medium showed that activin A was responsible for the activation of R-Smad. SAHA treatment enhanced the level of activin A mRNA, increasing the level of activin A in the secretome. The activity of the SAHA-treated secretome could be eliminated by pre-incubation with antibody to activin A. In addition, activin A supplemented medium could mimic the effect of the SAHA-treated secretome. CONCLUSION: These findings indicate that the anti-cancer function of SAHA is mediated, at least in part, by the upregulation of activin A.


Asunto(s)
Activinas/metabolismo , Antineoplásicos/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Transducción de Señal/efectos de los fármacos , Acetilación , Activinas/genética , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Separación Celular/métodos , Medios de Cultivo Condicionados/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citometría de Flujo/métodos , Inhibidores de Histona Desacetilasas/uso terapéutico , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Melanoma/tratamiento farmacológico , ARN Mensajero/metabolismo , Proteínas Smad Reguladas por Receptores , Regulación hacia Arriba , Vorinostat
8.
Biochem Biophys Res Commun ; 482(2): 270-276, 2017 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-27845041

RESUMEN

Studies have shown that γ-irradiation induces various biological responses, including oxidative stress and apoptosis, as well as cellular repair and immune system responses. However, most such studies have been performed using traditional two-dimensional cell culture systems, which are limited in their ability to faithfully represent in vivo conditions. A three-dimensional (3D) environment composed of properly interconnected and differentiated cells that allow communication and cooperation among cells via secreted molecules would be expected to more accurately reflect cellular responses. Here, we investigated γ-irradiation-induced changes in the secretome of 3D-cultured keratinocytes. An analysis of keratinocyte secretome profiles following fractionated-dose γ-irradiation revealed changes in genes involved in cell adhesion, angiogenesis, and the immune system. Notably, peroxisome proliferator-activated receptor-α (PPARα) was upregulated in response to fractionated-dose γ-irradiation. This upregulation was associated with an increase in the transcription of known PPARα target genes in secretome, including angiopoietin-like protein 4, dermokine and kallikrein-related peptide 12, which were differentially regulated by fractionated-dose γ-irradiation. Collectively, our data imply a mechanism linking γ-irradiation and secretome changes, and suggest that these changes could play a significant role in the coordinated cellular responses to harmful ionizing radiation, such as those associated with radiation therapy. This extension of our understanding of γ-irradiation-induced secretome changes has the potential to improve radiation therapy strategies.


Asunto(s)
Rayos gamma , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , PPAR gamma/metabolismo , Impresión Tridimensional , Proteoma/metabolismo , Adhesión Celular/fisiología , Adhesión Celular/efectos de la radiación , Células Cultivadas , Fraccionamiento de la Dosis de Radiación , Relación Dosis-Respuesta a Droga , Humanos , Neovascularización Fisiológica/fisiología , Neovascularización Fisiológica/efectos de la radiación , Regulación hacia Arriba/fisiología , Regulación hacia Arriba/efectos de la radiación
9.
Biochem Biophys Res Commun ; 476(4): 420-425, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27237972

RESUMEN

The evidence suggests that transforming growth factor-beta (TGF-ß) regulates the DNA-damage response (DDR) upon irradiation, and we previously reported that TGF-ß1 induced DNA ligase IV (Lig4) expression and enhanced the nonhomologous end-joining repair pathway in irradiated cells. In the present study, we investigated the effects of TGF-ß1 on the irradiation-induced DDRs of A431 and HaCaT cells. Cells were pretreated with or without TGF-ß1 and irradiated. At 30 min post-irradiation, DDRs were detected by immunoblotting of phospho-ATM, phospho-Chk2, and the presence of histone foci (γH2AX). The levels of all three factors were similar right after irradiation regardless of TGF-ß1 pretreatment. However, they soon thereafter exhibited downregulation in TGF-ß1-pretreated cells, indicating the acceleration of the DDR. Treatment with a TGF-ß type I receptor inhibitor (SB431542) or transfections with siRNAs against Smad2/3 or DNA ligase IV (Lig4) reversed this acceleration of the DDR. Furthermore, the frequency of irradiation-induced apoptosis was decreased by TGF-ß1 pretreatment in vivo, but this effect was abrogated by SB431542. These results collectively suggest that TGF-ß1 could enhance cell survival by accelerating the DDR via Smad signaling and Lig4 expression.


Asunto(s)
Daño del ADN , Células Epiteliales/efectos de la radiación , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Animales , Línea Celular , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , ADN Ligasa (ATP)/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Rayos gamma , Humanos , Ratones Desnudos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Proteínas Smad/genética , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Cell Biochem ; 117(1): 230-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26104915

RESUMEN

Heat shock protein 90 (HSP90) regulates the stability of various proteins and plays an essential role in cellular homeostasis. Many client proteins of HSP90 are involved in cell growth, survival, and migration; processes that are generally accepted as participants in tumorigenesis. HSP90 is also up-regulated in certain tumors. Indeed, the inhibition of HSP90 is known to be effective in cancer treatment. Recently, studies showed that HSP90 regulates transforming growth factor ß1 (TGF-ß1)-induced transcription by increasing the stability of the TGF-ß receptor. TGF-ß signaling also has been implicated in cancer, suggesting the possibility that TGF-ß1 and HSP90 function cooperatively during the cancer cell progression. Here in this paper, we investigated the role of HSP90 in TGF-ß1-stimulated Mv1Lu cells. Treatment of Mv1Lu cells with the HSP90 inhibitor, 17-allylamino-demethoxy-geldanamycin (17AAG), or transfection with truncated HSP90 (ΔHSP90) significantly reduced TGF-ß1-induced cell migration. Pretreatment with 17AAG or transfection with ΔHSP90 also reduced the levels of phosphorylated Smad2 and Smad3. In addition, the HSP90 inhibition interfered the nuclear localization of Smads induced by constitutively active Smad2 (S2EE) or Smad3 (S3EE). We also found that the HSP90 inhibition decreased the protein level of importin-ß1 which is known to regulate R-Smad nuclear translocation. These data clearly demonstrate a novel function of HSP90; HSP90 modulates TGF-ß signaling by regulating Smads localization. Overall, our data could provide a detailed mechanism linking HSP90 and TGF-ß signaling. The extension of our understanding of HSP90 would offer a better strategy for treating cancer.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Animales , Benzoquinonas/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/genética , Lactamas Macrocíclicas/farmacología , Fosforilación/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
11.
Oncol Rep ; 34(5): 2731-7, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26351897

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive cancer with a poor prognosis due to its epithelial­to-mesenchymal transition (EMT) phenotype. Cancer patients often experience several detrimental effects of cancer treatment, such as chemoresistance, radioresistance and the maintenance of cancer stem cells due to EMT. Thus, EMT signaling is considered to be a valuable therapeutic target for cancer treatment, and its inhibition is being attempted as a new treatment option for TNBC patients. Previously, we showed that 3-(2-chlorobenzyl)-1,7-dimethyl-1H-imidazo[2,1-f]purine­2,4(3H,8H)-dione (IM-412) inhibits transforming growth factor-ß (TGF-ß)-induced differentiation of human lung fibroblasts through both Smad-dependent and -independent pathways. In the present study, we examined the inhibitory effect of IM-412 on EMT pathways and invasiveness in TNBC cells since the TGF-ß signaling pathway is a typical signaling pathway that functions in EMT. IM-412 not only potently suppressed the migration and invasion of MDA-MB-231 cells, but also lowered the expression of mesenchymal markers and EMT-activating transcription factors in these cells. IM-412 inhibited the activation of several signaling proteins, including Smad2/Smad3, p38MAPK, Akt and JNK, and it also attenuated the phosphorylation of FGFR1 and FGFR3. Collectively, our findings suggest that the synthetic compound IM-412 suppressed the EMT process in MDA-MB-231 cells and thereby effectively inhibited the migration and invasion of these cancer cells. Thus, IM-412 could serve as a novel therapeutic agent for malignant cancers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Imidazoles/farmacología , Purinas/farmacología , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , Fosforilación/efectos de los fármacos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Neoplasias de la Mama Triple Negativas/patología
12.
Oncotarget ; 6(28): 26528-40, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-25605253

RESUMEN

In the present study, we found that selective inhibition of histone deacetylase 2 (HDAC2) with small inhibitory RNA (siRNA) induced survivin downregulation in a p53-dependent manner. Interestingly, suberoylanilide hydroxamic acid (SAHA) or knockdown of HDAC2 induced downregulation of Mdm2, a negative regulator of p53, at the protein level. SAHA and/or HDAC2 siRNA increased Mdm2 ubiquitination, and MG132, an inhibitor of proteosome function, prevented HDAC2 inhibition-induced degradation of Mdm2. Clinically, the mRNA levels of HDAC2 and survivin were prominently overexpressed in lung cancer patients compared to normal lung tissues. Silencing of HDAC2 enhanced the cell death caused by ionizing radiation in lung cancer cells. Collectively, our results indicate that selective inhibition of HDAC2 causes survivin downregulation through activation of p53, which is mediated by downregulation of Mdm2. They further suggest that HDAC2 may exert a dominant effect on lung cancer cell survival by sustaining Mdm2-survivin levels.


Asunto(s)
Histona Desacetilasa 2/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Proteínas Inhibidoras de la Apoptosis/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Línea Celular Tumoral , Regulación hacia Abajo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Complejo de la Endopetidasa Proteasomal/genética , Inhibidores de Proteasoma/farmacología , Proteolisis , Proteínas Proto-Oncogénicas c-mdm2/genética , Interferencia de ARN , Survivin , Transfección , Proteína p53 Supresora de Tumor/genética , Ubiquitinación
13.
Mol Cancer Res ; 13(2): 319-29, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25319009

RESUMEN

UNLABELLED: Several groups have reported that TGFß1 regulates cellular responses to γ-irradiation; however, the exact mechanism has not been fully elucidated. In the current study, the role of TGFß1 in cellular responses to γ-irradiation was investigated in detail. The data indicate that TGFß1 pretreatment decreased the aftermath of ionizing radiation (IR)-induced DNA damage in a SMAD-dependent manner. To determine the underlying mechanism for these effects, the extent of IR-induced DNA repair activity in the presence or absence of TGFß1 was examined. Studies reveal that TGFß1 upregulated DNA ligase IV (Lig4), augmented IR-induced nuclear retention of the DNA ligase, and enhanced nonhomologous end-joining (NHEJ) repair activity. In addition, knockdown of Lig4 reduced the TGFß1-induced protection against IR. Overall, these data indicate that TGFß1 facilitates the NHEJ repair process upon γ-irradiation and thereby enhances long-term survival. IMPLICATIONS: These findings provide new insight and a possible approach to controlling genotoxic stress by the TGFß signaling pathway.


Asunto(s)
Reparación del ADN por Unión de Extremidades/efectos de los fármacos , ADN Ligasas/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Benzamidas/farmacología , Técnicas de Cultivo de Célula , Línea Celular , Supervivencia Celular/efectos de la radiación , Daño del ADN , Reparación del ADN por Unión de Extremidades/efectos de la radiación , ADN Ligasa (ATP) , ADN Ligasas/genética , Dioxoles/farmacología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación
14.
Exp Cell Res ; 326(1): 125-35, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24928277

RESUMEN

Fibrosis is one of the most serious side effects in cancer patients undergoing radio-/ chemo-therapy, especially of the lung, pancreas or kidney. Based on our previous finding that galectin-1 (Gal-1) was significantly increased during radiation-induced lung fibrosis in areas of pulmonary fibrosis, we herein clarified the roles and action mechanisms of Gal-1 during fibrosis. Our results revealed that treatment with TGF-ß1 induced the differentiation of fibroblast cell lines (NIH3T3 and IMR-90) to myofibroblasts, as evidenced by increased expression of the fibrotic markers smooth muscle actin-alpha (α-SMA), fibronectin, and collagen (Col-1). We also observed marked and time-dependent increases in the expression level and nuclear accumulation of Gal-1. The TGF-ß1-induced increases in Gal-1, α-SMA and Col-1 were decreased by inhibitors of PI3-kinase and p38 MAPK, but not ERK. Gal-1 knockdown using shRNA decreased the phosphorylation and nuclear retention of Smad2, preventing the differentiation of fibroblasts. Gal-1 interacted with Smad2 and phosphorylated Smad2, which may accelerate fibrotic processes. In addition, up-regulation of Gal-1 expression was demonstrated in a bleomycin (BLM)-induced mouse model of lung fibrosis in vivo. Together, our results indicate that Gal-1 may promote the TGF-ß1-induced differentiation of fibroblasts by sustaining nuclear localization of Smad2, and could be a potential target for the treatment of pulmonary fibrotic diseases.


Asunto(s)
Núcleo Celular/metabolismo , Fibroblastos/patología , Galectina 1/metabolismo , Fibrosis Pulmonar/patología , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Animales , Apoptosis , Western Blotting , Diferenciación Celular , Núcleo Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Galectina 1/antagonistas & inhibidores , Galectina 1/genética , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Ratones , Fosforilación/efectos de los fármacos , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo , ARN Interferente Pequeño/genética
15.
Biochem Biophys Res Commun ; 449(4): 490-5, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24845567

RESUMEN

TWIST1 is implicated in the process of epithelial mesenchymal transition, metastasis, stemness, and drug resistance in cancer cells, and therefore is a potential target for cancer therapy. In the present study, we found that knockdown of TWIST1 by small interfering RNA (siRNA) enhanced arsenic trioxide (ATO)- and ionizing radiation (IR)-induced cell death in non-small-cell lung cancer cells. Interestingly, intracellular reactive oxygen species levels were increased in cells treated with TWIST1 siRNA and further increased by co-treatment with ATO or IR. Pretreatment of lung cancer cells with the antioxidant N-acetyl-cysteine markedly suppressed the cell death induced by combined treatment with TWIST1 siRNA and ATO or IR. Moreover, treatment of cells with TWIST1 siRNA induced mitochondrial membrane depolarization and significantly increased mitochondrial fragmentation (fission) and upregulated the fission-related proteins FIS1 and DRP1. Collectively, our results demonstrate that siRNA-mediated TWIST1 knockdown induces mitochondrial dysfunction and enhances IR- and ATO-induced cell death in lung cancer cells.


Asunto(s)
Proteínas Nucleares/genética , Proteína 1 Relacionada con Twist/genética , Acetilcisteína/farmacología , Trióxido de Arsénico , Arsenicales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Técnicas de Silenciamiento del Gen , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas Nucleares/biosíntesis , Óxidos , ARN Interferente Pequeño/farmacología , Radiación Ionizante , Especies Reactivas de Oxígeno/metabolismo , Proteína 1 Relacionada con Twist/biosíntesis
16.
Exp Cell Res ; 319(4): 466-73, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23262026

RESUMEN

Transforming growth factor-ß1 (TGF-ß1) regulates various biological processes, including differentiation, bone remodeling and angiogenesis, and is particularly important as a regulator of homeostasis and cell growth in normal tissue. Interestingly, some studies have reported that TGF-ß1 induces apoptosis through induction of specific genes, whereas others suggest that TGF-ß1 inhibits apoptosis and facilitates cell survival. Resolving these discrepancies, which may reflect differences in cellular context, is an important research priority. Here, using the parental mink lung epithelial cell line, Mv1Lu, and its derivatives, R1B and DR26, lacking TGF-ß receptors, we investigated the involvement of TGF-ß signaling in the effects of γ-irradiation. We found that canonical TGF-ß signaling played an important role in protecting cells from γ-irradiation. Introduction of functional TGF-ß receptors or constitutively active Smads into R1B and DR26 cell lines reduced DNA fragmentation, Caspase-3 cleavage and γ-H2AX foci formation in γ-irradiated cells. Notably, we also found that de novo protein synthesis was required for the radio-resistant effects of TGF-ß1. Our data thus indicate that TGF-ß1 protected against γ-irradiation, decreasing DNA damage and reducing apoptosis, and thereby enhanced cell survival.


Asunto(s)
Rayos gamma , Tolerancia a Radiación , Factor de Crecimiento Transformador beta/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Apoptosis/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Células Epiteliales/efectos de la radiación , Pulmón/citología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/efectos de la radiación , Visón , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética , Tolerancia a Radiación/fisiología , Protectores contra Radiación/metabolismo , Protectores contra Radiación/farmacología , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Transfección , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
17.
Cell Signal ; 24(11): 2132-42, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22800863

RESUMEN

To determine the role of CD24 in breast cancer cells, we knocked down CD24 in MCF-7 human breast cancer cells by retroviral delivery of shRNA. MCF-7 cells with knocked down CD24 (MCF-7 hCD24 shRNA) exhibited decreased cell proliferation and cell adhesion as compared to control MCF-7 mCD24 shRNA cells. Decreased proliferation of MCF-7 hCD24 shRNA cells resulted from the inhibition of cell cycle progression from G1 to S phase. The specific inhibition of MEK/ERK signaling by CD24 ablation might be responsible for the inhibition of cell proliferation. Phosphorylation of Src/FAK and TGF-ß1-mediated epithelial to mesenchymal transition was also down-regulated in MCF-7 hCD24 shRNA cells. Reduced Src/FAK activity was caused by a decrease in integrin ß1 bound with CD24 and subsequent destabilization of integrin ß1. Our results suggest that down-regulation of Raf/MEK/ERK signaling via Src/FAK may be dependent on integrin ß1 function and that this mechanism is largely responsible for the CD24 ablation-induced decreases in cell proliferation and epithelial to mesenchymal transition.


Asunto(s)
Antígeno CD24/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Integrina beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Antígeno CD24/química , Antígeno CD24/genética , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Células HEK293 , Humanos , Células MCF-7 , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación , Unión Proteica , Estabilidad Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Quinasas raf/metabolismo
18.
FEBS Lett ; 586(16): 2207-11, 2012 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-22659189

RESUMEN

Syndecan are a family of cell surface heparan sulfate proteoglycans that act as cell surface receptors. Most cell surface receptors have a limited number and type of ligand interactions, responding only to the binding of (a) specific ligand(s). In contrast, syndecans can interact with various numbers and types of ligands, and thus play more diverse roles than others. Various syndecan functions have not yet been fully classified and categorized, but we herein review previous studies suggesting that syndecans play dual function as cell surface receptors by acting as both adhesion receptors and docking receptors. Through this dual regulatory function, syndecans are capable of regulating both intra- and extracellular activities, potentially altering a variety of cell behaviors.


Asunto(s)
Regulación de la Expresión Génica , Receptores de Superficie Celular/metabolismo , Sindecanos/fisiología , Animales , Adhesión Celular , Comunicación Celular , Membrana Celular/metabolismo , Humanos , Ligandos , Glicoproteínas de Membrana/metabolismo , Modelos Biológicos , Complejo GPIb-IX de Glicoproteína Plaquetaria , Proteoglicanos/metabolismo , Sindecanos/metabolismo
19.
Biochem Pharmacol ; 83(7): 849-57, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22266356

RESUMEN

ß-Galactoside α2,6-sialyltransferase (ST6Gal-I) has been shown to catalyze α2,6 sialylation of N-glycan, an action that is highly correlated with colon cancer progression and metastasis. We have recently demonstrated that ST6Gal-I-induced α2,6 sialylation is critical for adhesion and migration of colon cancer cells. Increase of α2,6 sialylation also contributes to radioresistance of colon cancer. A number of studies have focused on the involvement of sialylation in tumorigenesis, but the mechanism underlying ST6Gal-I-induced cancer progression and the identity of enzyme substrates has received scant research attention. To provide further support for the relevance of ST6Gal-I in the malignancy of colon cancer, we prepared and characterized a ST6Gal-I-knockdown SW480 colorectal carcinoma cell line. We found that inhibition of ST6Gal-I expression increased cell proliferation and tumor growth in vitro and in vivo. An examination of the effect of sialylation on epidermal growth factor receptor (EGFR) activity and downstream signaling, which are highly correlated with cell proliferation, showed that the loss of ST6Gal-I augmented EGF-induced EGFR phosphorylation and activation of extracellular signal-regulated kinase (ERK) in colon cancer cells. Moreover, ST6Gal-I induced sialylation of both wild type and mutant EGFR. These studies provide the first demonstration that ST6Gal-I induces EGFR sialylation in human colon cancer cell lines. Importantly, the anticancer effect of the EGFR kinase inhibitor, gefitinib, was increased in ST6Gal-I-deficient colon cancer cells. In contrast, overexpression of ST6Gal I decreased the cytotoxic effect of gefitinib. These results suggest that sialylation of the EGFR affects EGF-mediated cell growth and induces chemoresistance to gefitinib in colon cancer cells.


Asunto(s)
Antígenos CD/metabolismo , Antineoplásicos/farmacología , Neoplasias del Colon/metabolismo , Receptores ErbB/antagonistas & inhibidores , Quinazolinas/farmacología , Sialiltransferasas/metabolismo , Animales , Antígenos CD/genética , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Resistencia a Antineoplásicos , Receptores ErbB/metabolismo , Citometría de Flujo , Gefitinib , Células HT29 , Humanos , Immunoblotting , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Ratones , Ratones Desnudos , Fosforilación , Quinazolinas/uso terapéutico , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ácidos Siálicos/metabolismo , Sialiltransferasas/genética , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Carcinogenesis ; 32(10): 1474-83, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21798852

RESUMEN

Cluster of differentiation 24 (CD24) is a small glycosylphosphatidylinositol-linked cell surface molecule that is expressed in a variety of human carcinomas, including breast cancer. To determine the role of CD24 in breast cancer cells, we expressed CD24 in CD24-negative/low and cluster of differentiation 44 (CD44)-positive MDA-MB-231 metastatic breast cancer cells. Forced expression of CD24 resulted in a decrease in c-Raf/mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK)/mitogen-activated protein kinase signaling and reduced cell proliferation. Apoptosis induced by DNA damage was greatly enhanced in MDA-MB-231 CD24 cells as compared with MDA-MB-231 vec cells. CD24 expression efficiently attenuated DNA damage-induced nuclear factor-kappaB (NF-κB) signaling in MDA-MB-231 cells. However, in CD24-positive and CD44-negative/low MCF-7 cells, knockdown of CD24 did not significantly affect DNA damage-induced apoptosis nor NF-κB signaling. Silencing of CD24 in CD24/CD44-double-positive MDA-MB-468 cells partially rescued DNA damage-induced apoptosis. Transient transfection studies with 293T cells also revealed that CD24 attenuated cell viability and NF-κB signaling only when CD44 was cotransfected. These data indicate that CD24 expression potentiated DNA-induced apoptosis by suppressing antiapoptotic NF-κB signaling in CD44-expressing cells.


Asunto(s)
Apoptosis , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Antígeno CD24/metabolismo , Daño del ADN , FN-kappa B/metabolismo , Antibióticos Antineoplásicos/farmacología , Western Blotting , Neoplasias de la Mama/genética , Antígeno CD24/química , Antígeno CD24/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Doxorrubicina/farmacología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Citometría de Flujo , Humanos , Luciferasas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Invasividad Neoplásica , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Radiación Ionizante , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...