Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Int J Oncol ; 51(3): 859-866, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28713989

RESUMO

Proton beam therapy has recently been used to improve local control of tumor growth and reduce side-effects by decreasing the global dose to normal tissue. However, the regulatory mechanisms underlying the physiological role of proton beam radiation are not well understood, and many studies are still being conducted regarding these mechanisms. To determine the effects of proton beams on mitochondrial biogenesis, we investigated: mitochondrial DNA (mtDNA) mass; the gene expression of mitochondrial transcription factors, functional regulators, and dynamic-related regulators; and the phosphorylation of the signaling molecules that participate in mitochondrial biogenesis. Both the mtDNA/nuclear DNA (nDNA) ratio and the mitochondria staining assays showed that proton beam irradiation increases mitochondrial biogenesis in 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced aggressive HT-29 cells. Simultaneously, proton beam irradiation increases the gene expression of the mitochondrial transcription factors PGC-1α, NRF1, ERRα, and mtTFA, the dynamic regulators DRP1, OPA1, TIMM44, and TOM40, and the functional regulators CytC, ATP5B and CPT1-α. Furthermore, proton beam irradiation increases the phosphorylation of AMPK, an important molecule involved in mitochondrial biogenesis that is an energy sensor and is regulated by the AMP/ATP ratio. Based on these findings, we suggest that proton beam irradiation inhibits metastatic potential by increasing mitochondrial biogenesis and function in TPA-induced aggressive HT-29 cells.


Assuntos
Adenocarcinoma/radioterapia , Neoplasias Colorretais/radioterapia , DNA Mitocondrial/efeitos da radiação , Biogênese de Organelas , Adenocarcinoma/genética , Adenocarcinoma/patologia , Monofosfato de Adenosina/metabolismo , Monofosfato de Adenosina/efeitos da radiação , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/efeitos da radiação , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Células HT29 , Humanos , Proteínas de Neoplasias/efeitos da radiação , Fosforilação/efeitos da radiação , Terapia com Prótons , Acetato de Tetradecanoilforbol/toxicidade
2.
Oncotarget ; 7(38): 62340-62351, 2016 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-27694690

RESUMO

Radiotherapy is promising and effective for treating prostate cancer but the addition of a tumor cell radiosensitizer would improve therapeutic outcomes. PC-1/PrLZ, a TPD52 protein family member is frequently upregulated in advanced prostate cancer cells and may be a biomarker of aggressive prostate cancer. Therefore, we investigated the potential role of PC-1/PrLZ for increasing radioresistance in human prostate cancer cell lines. Growth curves and survival assays after g-ray irradiation confirmed that depletion of endogenous PC-1/PrLZ significantly increased prostate cancer cell radiosensitivity. Irradiation (IR) increased PC-1/PrLZ expression in a dose- and time-dependent manner and increased radiosensitivity in PC-1/PrLZ-suppressed cells was partially due to decreased DNA double strand break (DBS) repair which was measured with comet and gH2AX foci assays. Furthermore, depletion of PC-1/PrLZ impaired the IR-induced G2/M checkpoint, which has been reported to be correlate with radioresistance in cancer cells. PC-1/PrLZ-deficient cells exhibited higher level of autophagy when compared with control cells. Thus, specific inhibition of PC-1/PrLZ might provide a novel therapeutic strategy for radiosensitizing prostate cancer cells.


Assuntos
Autofagia/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/efeitos da radiação , Proteínas de Neoplasias/efeitos da radiação , Neoplasias da Próstata/radioterapia , Tolerância a Radiação , Linhagem Celular Tumoral , Relação Dose-Resposta à Radiação , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos da radiação , Raios gama , Histonas/metabolismo , Histonas/efeitos da radiação , Humanos , Masculino , Microscopia de Fluorescência , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fosforilação , Próstata/citologia , Próstata/efeitos da radiação , Neoplasias da Próstata/patologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Regulação para Cima
3.
Adv Exp Med Biol ; 937: 207-28, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27573902

RESUMO

Despite recent progress in understanding the cancer signaling pathways and in developing new therapeutic strategies, however, the resistance of colorectal cancer (CRC) cells to chemo- and radiotherapy represents the main hurdle to the successful treatment, leading to tumor recurrence and, consequently, a poor prognosis. Therefore, overcoming drug and radiation resistance, enhancing drug and radiation sensitivity of CRC cells, and improving the efficacy of chemo- and radiotherapy have an important significance in the treatment of CRC. The identification of new molecular biomarkers which can predict therapy response and prognosis is one of the most significant aims in pharmacogenomics and cancer research.Recent studies showed that non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), may play important roles in the regulation of chemo- and radioresistance of CRC, by controlling several signaling pathways, including cell cycle, proliferation, apoptosis and DNA damage repair. Recent data have demonstrated that selective modulation of the ncRNA activity can improve the response to chemo- and radiotherapy, providing an innovative anti-tumor approach based on a ncRNA-related gene therapy. Therefore, ncRNAs could not only be useful as predictive and prognostic biomarkers but also serve as targets for the development of novel therapeutic strategies to overcome drug and radiation resistance in CRC. In this chapter, we discuss the involvement of ncRNAs in chemo- and radiotherapy resistance of CRC, highlighting the impact of these molecules in prediction of the treatment response and modification of the therapy, and describing possible intracellular pathways involved in these processes.


Assuntos
Neoplasias Colorretais/genética , Resistencia a Medicamentos Antineoplásicos/genética , RNA Neoplásico/genética , RNA não Traduzido/genética , Tolerância a Radiação/genética , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/radioterapia , Previsões , Regulação Neoplásica da Expressão Gênica , Humanos , Terapia de Alvo Molecular , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/fisiologia , Proteínas de Neoplasias/efeitos da radiação , Prognóstico , Transdução de Sinais , Terapias em Estudo
4.
Tumour Biol ; 36(8): 6019-28, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25750034

RESUMO

This study aimed to to evaluate the stability of commonly used endogenous control genes for messenger RNA (mRNA) (N = 16) and miRNAs (N = 3) expression studies in prostate cell lines following irradiation. The stability of endogenous control genes expression in irradiated (6 Gy) versus unirradiated controls was quantified using NormFinder and coefficient of variation analyses. HPRT1 and 18S were identified as most and least stable endogenous controls, respectively, for mRNA expression studies in irradiated prostate cells. SNORD48 and miR16 miRNA endogenous controls tested were associated with low coefficient of variations following irradiation (6 Gy). This study highlights that commonly used endogenous controls can be responsive to radiation and validation is required prior to gene/miRNAs expression studies.


Assuntos
Perfilação da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Neoplasias da Próstata/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Masculino , MicroRNAs/biossíntese , MicroRNAs/efeitos da radiação , Proteínas de Neoplasias/efeitos da radiação , Neoplasias da Próstata/patologia , Neoplasias da Próstata/radioterapia , RNA Mensageiro/biossíntese , Radiação
5.
Mutagenesis ; 30(2): 287-96, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25392149

RESUMO

To investigate polypeptide from Chlamy Farreri (PCF)'s protective effect against skin cancer, we used a cellular model of ultraviolet B (UVB)-induced malignant transformation. The human keratinocyte cell line HaCaT was repeatly exposed to UVB (10 mJ/cm(2), 20 times) and malignant transformation was confirmed by Gimesa staining, cell cycle analysis and various assays [anchorage independent growth, matrix metalloproteinase-9 (MMP9) activity, plating efficiency]. The malignant transformation was found to be effectively prevented by PCF pretreatment (2.84mM for 2h prior to each UVB exposure). We investigated the mechanism of PCF-mediated action by determining its effect on DNA methylation status of the tumour suppressor genes [P16 and ras association domain family 1 A (RASSF1A)] in the UVB-transformed cells. Both genes were found to be hypermethylated by chronic UVB exposure. The expression levels of P16, RASSF1A, DNA methyltransferases (DNMTs) and DNA damage inducible protein a (GADD45a) were measured by reverse transcriptase-polymerase chain reaction and western blotting. While chronic UVB exposure was found to suppress the expression of P16 and RASSF1A, it enhanced the expression of DNMT3b. In the early phase of UVB-induced malignant transformation, the GADD45a expression was increased, however, it declined with a continued irradiation of the cells. The UVB-induced DNA hypermethylation of P16 and RASSF1A and subsequent gene silencing was reversed by PCF treatment. The inhibition of DNMTs expression suggested that PCF blocked DNA methylation and thereby the silencing of tumour suppressor genes. Furthermore, the PCF-mediated substantial increase in GADD45a expression indicated that PCF promoted demethylation of tumour suppressor genes via GADD45a induction.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Queratinócitos/efeitos da radiação , Neoplasias Induzidas por Radiação/prevenção & controle , Pectinidae , Peptídeos/farmacologia , Neoplasias Cutâneas/prevenção & controle , Raios Ultravioleta , Animais , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/efeitos da radiação , Linhagem Celular , Transformação Celular Neoplásica/genética , Inibidor p16 de Quinase Dependente de Ciclina , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/efeitos da radiação , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/efeitos da radiação , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/efeitos da radiação , Humanos , Queratinócitos/efeitos dos fármacos , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/efeitos da radiação , Neoplasias Induzidas por Radiação/genética , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/genética , Proteínas Nucleares/efeitos da radiação , Substâncias Protetoras/farmacologia , Neoplasias Cutâneas/genética , Proteínas Supressoras de Tumor/efeitos dos fármacos , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/efeitos da radiação , DNA Metiltransferase 3B
6.
Hum Pathol ; 45(10): 2029-36, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25150747

RESUMO

Immunohistochemistry (IHC) testing for mismatch repair proteins (MMRP) is currently being used primarily in colorectal cancer resection specimens. We aimed to compare the results of IHC staining performed on biopsy specimens obtained at endoscopy with that performed on surgical specimens after neoadjuvant therapy. Thirty-two rectal cancer subjects had paired preneoadjuvant and postneoadjuvant tissue available for IHC staining (MLH1, MSH2, MSH6, and PMS2), whereas 39 rectosigmoid cancer patients who did not receive neoadjuvant treatment served as controls. Each slide received a qualitative (absent, focal, and strong) and quantitative score (immunoreactivity [0-3] × percent positivity [0-4]). The quantitative scores of MMRP from the operative material were significantly lower in the neoadjuvant group than in the control (P < .05 for all).The scores of all MMRP from endoscopic biopsies were not significantly different between the neoadjuvant and the control groups. Disagreement between the endoscopic biopsy and the operative material was evident in 23 of 128 stains (18.5%) in the neoadjuvant group and in 12 of 156 stains (7.7%) in the control group (P = .009). In the neoadjuvant group, a disagreement pattern of "endoscopic strong operative focal" was observed in 28.1% for PMS2, 12.5% for MSH6, 12.5% for MLH1, and 6.3% for MSH2, and in the control group, this same disagreement pattern was found in 12.8% for PMS2, 7.7% for MSH6, 7.7% for MLH1, and 0% for MSH2. Based on our findings, we suggest that for rectal cancer, the endoscopic material rather than the operative material should serve as the primary material for IHC staining.


Assuntos
Neoplasias Colorretais/genética , Reparo de Erro de Pareamento de DNA , Imuno-Histoquímica/normas , Terapia Neoadjuvante , Proteínas de Neoplasias/análise , Proteínas Adaptadoras de Transdução de Sinal/análise , Proteínas Adaptadoras de Transdução de Sinal/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/efeitos da radiação , Adenosina Trifosfatases/análise , Adenosina Trifosfatases/efeitos dos fármacos , Adenosina Trifosfatases/efeitos da radiação , Idoso , Quimiorradioterapia , Neoplasias Colorretais/terapia , Enzimas Reparadoras do DNA/análise , Enzimas Reparadoras do DNA/efeitos dos fármacos , Enzimas Reparadoras do DNA/efeitos da radiação , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/efeitos da radiação , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Endonuclease PMS2 de Reparo de Erro de Pareamento , Proteína 1 Homóloga a MutL , Proteína 2 Homóloga a MutS/análise , Proteína 2 Homóloga a MutS/efeitos dos fármacos , Proteína 2 Homóloga a MutS/efeitos da radiação , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/efeitos da radiação , Proteínas Nucleares/análise , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/efeitos da radiação , Reprodutibilidade dos Testes
7.
Radiother Oncol ; 103(3): 415-20, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22633816

RESUMO

BACKGROUND AND PURPOSE: Ionizing radiation (IR)-induced DNA damage causes the accumulation of DNA damage response (DDR) proteins as visible foci in cell nuclei. Despite the identified functional roles in DNA repair, the spatial relationships of different DDR proteins at foci have not been explicitly examined. This study aims to systematically compare the distribution of DDR proteins at IR-induced foci. MATERIALS AND METHODS: MCF-7 cells were treated with IR, stained for γH2AX, MDC1, RNF8, RNF168, 53BP1, Abraxas (CCDC98), BRCA1, BRCC36, Merit40 (NBA1) and RAP80, and then imaged using high-resolution three-dimensional (3-D) confocal microscopy to assess the relative localization of proteins at foci. RESULTS: All BRCA1-A complex components displayed strong co-localization, which overlapped significantly with RNF8 and RNF168, but not with γH2AX and MDC1. Intriguingly, 53BP1 co-located well with γH2AX and MDC1, but remained separate from RNF8 and RNF168. These co-localization patterns were consistent for at least 3h after IR. CONCLUSIONS: The foci formations of γH2AX-MDC1-53BP1 and RNF8-RNF168-BRCA1-A complexes are spatially independent. Such divergence was not anticipated from prior studies on the recruitment of these proteins to foci. This information indicates that individual foci may represent distinct sites of DNA repair facilitated by a specific subset of DDR proteins.


Assuntos
Neoplasias da Mama/metabolismo , Dano ao DNA , Complexos Multiproteicos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Células Cultivadas , Reparo do DNA por Junção de Extremidades , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Feminino , Imunofluorescência , Histonas/metabolismo , Humanos , Imageamento Tridimensional , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Microscopia Confocal , Proteínas de Neoplasias/efeitos da radiação , Proteínas Nucleares/efeitos da radiação , Radiação Ionizante , Reparo de DNA por Recombinação , Transativadores/metabolismo , Transfecção , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Ubiquitina-Proteína Ligases/metabolismo
8.
Int J Radiat Oncol Biol Phys ; 83(1): 394-9, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22056068

RESUMO

PURPOSE: To investigate the effect of carbon ion irradiation on glioma cell migration. METHODS AND MATERIALS: U87 and Ln229 glioma cells were irradiated with photons and carbon ions. Migration was analyzed 24 h after irradiation. Fluorescence-activated cell sorting analysis was performed in order to quantify surface expression of integrins. RESULTS: Single photon doses of 2 Gy and 10 Gy enhanced α(ν)ß(3) and α(ν)ß(5) integrin expression and caused tumor cell hypermigration on both vitronectin (Vn) and fibronectin (Fn). Compared to integrin expression in unirradiated cells, carbon ion irradiation caused decreased integrin expression and inhibited cell migration on both Vn and Fn. CONCLUSION: Photon radiotherapy (RT) enhances the risk of tumor cell migration and subsequently promotes locoregional spread via photon induction of integrin expression. In contrast to photon RT, carbon ion RT causes decreased integrin expression and suppresses glioma cell migration on both Vn and Fn, thus promising improved local control.


Assuntos
Carbono/uso terapêutico , Movimento Celular/efeitos da radiação , Regulação para Baixo/efeitos da radiação , Glioma/radioterapia , Integrina alfaVbeta3/efeitos da radiação , Proteínas de Neoplasias/efeitos da radiação , Receptores de Vitronectina/efeitos da radiação , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Regulação para Baixo/fisiologia , Fibronectinas/fisiologia , Glioma/metabolismo , Glioma/patologia , Humanos , Integrina alfaVbeta3/metabolismo , Proteínas de Neoplasias/metabolismo , Fótons/efeitos adversos , Fótons/uso terapêutico , Doses de Radiação , Receptores de Vitronectina/metabolismo , Vitronectina/fisiologia
9.
Radiother Oncol ; 99(2): 231-4, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21620500
10.
Chin J Cancer ; 29(10): 900-6, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20868560

RESUMO

BACKGROUND AND OBJECTIVE: The mRNA levels of 59 genes, detected by cDNA microarray, were up-regulated in the radioresistant human esophageal cacinoma cell line TE13R120 as compared with its parental cell line TE13 before and after radiation, and the expression of NRAGE gene showed a gradually up-regulating tendency. This study aimed to further detect the differences of NRAGE gene and protein expression and apoptosis between TE13R120 and TE13 cells, and to investigate the relationship between the NRAGE and the radioresistance of TE13R120 cells and its mechanism. METHODS: The two cell lines were irradiated by 6°Co γ-ray at different conditions. Reverse transcription-polymerase chain reaction (RT-PCR), Western blot, and immunocytochemistry were used to detect the expression of NRAGE. Flow cytometry (FCM) was used to detect the cell apoptosis before and after irradiation. RESULTS: The mRNA level of NRAGE was higher in TE13R120 cells than in TE13 cells before and after irradiation (before radiation: 0.25 ± 0.03 vs. 0.49 ± 0.03; 4 Gy 4 h: 0.31 ± 0.03 vs. 0.53 ± 0.02; 4 Gy 16 h: 0.32 ± 0.04 vs. 0.59 ± 0.04; 4 Gy 24 h: 0.36 ± 0.05 vs. 0.72 ± 0.04; 2 Gy 12 h: 0.32 ± 0.02 vs. 0.64 ± 0.04; 6 Gy 12 h: 0.36 ± 0.02 vs. 0.79 ± 0.05; 10 Gy 12 h: 0.46 ± 0.04 vs. 0.85 ± 0.01; P < 0.01), and the mRNA level of NRAGE was increased gradually with the increase of radiation dose and time in the two cell lines (P < 0.05 and P < 0.01). Western blot results showed no difference of NRAGE protein level in cytoplasm between TE13R120 cells and TE13 cells before and after irradiation, but its level in nuclei was higher in TE13R120 cells than in TE13 cells at different radiation time and dosages. Immunocytochemistry showed similar results as Western blot. FCM showed no significant difference in apoptosis rate between TE13R120 and TE13 cells before and after radiation. CONCLUSION: NRAGE may play an important role in the radiation responses of the two cell lines, and may participate in the formation of radioresistance of TE13R120 cells by changing its subcellular localization, but its relationship with cell apoptosis has not been confirmed.


Assuntos
Antígenos de Neoplasias/metabolismo , Neoplasias Esofágicas/metabolismo , Proteínas de Neoplasias/metabolismo , Tolerância a Radiação , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/efeitos da radiação , Apoptose/efeitos da radiação , Linhagem Celular Tumoral/efeitos da radiação , Radioisótopos de Cobalto , Neoplasias Esofágicas/patologia , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/efeitos da radiação , RNA Mensageiro/metabolismo , RNA Mensageiro/efeitos da radiação , Dosagem Radioterapêutica , Fatores de Tempo , Regulação para Cima
11.
Ir J Med Sci ; 179(3): 327-35, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19921309

RESUMO

OBJECTIVE: The aim of our study was to explore the expression of survivin gene in colorectal cancer (CRC) and its possibility as a molecular target for CRC chemoradiotherapy. MATERIALS AND METHODS: Immunohistochemistry was performed to detect the expression of survivin in 68 CRC specimens. The correlations between survivin expression and clinicopathological factors and prognosis were evaluated. RNA interference was employed to downregulate survivin expression. The effects of survivin downregulation on chemoradiotherapy of CRC cells were evaluated. RESULTS: The staining of survivin protein was strongly positive in the cytoplasm of CRC cells. Its expression was significantly correlated with tumor differentiation, Duke's stage, lymph node metastasis. Moreover, the elevated survivin expression was an independent factor for predicting the prognosis of CRC patients. Survivin downregulation could also enhance chemosensitivity or radiosensitivity of colorectal cells. CONCLUSIONS: Survivin might be an independent prognostic factor and a potential target for the chemoradiotherapy of CRC patients.


Assuntos
Neoplasias do Colo/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Retais/metabolismo , Adulto , Idoso , Apoptose/fisiologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Colo/mortalidade , Neoplasias do Colo/terapia , Regulação para Baixo/fisiologia , Feminino , Humanos , Imuno-Histoquímica , Proteínas Inibidoras de Apoptose , Estimativa de Kaplan-Meier , Masculino , Proteínas Associadas aos Microtúbulos/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/efeitos da radiação , Pessoa de Meia-Idade , Análise Multivariada , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/efeitos da radiação , Prognóstico , RNA Interferente Pequeno , Dosagem Radioterapêutica , Neoplasias Retais/mortalidade , Neoplasias Retais/terapia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Survivina , Ensaio Tumoral de Célula-Tronco
12.
Int J Radiat Oncol Biol Phys ; 75(2): 534-42, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19735878

RESUMO

PURPOSE: Radiation therapy is an integral part of the preoperative treatment of rectal cancers. However, only a minority of patients achieve a complete pathologic response to therapy because of resistance of these tumors to radiation therapy. This resistance may be mediated by constitutively active pro-survival signaling pathways or by inducible/acquired mechanisms in response to radiation therapy. Simultaneous inhibition of these pathways can sensitize these tumors to radiation therapy. METHODS AND MATERIALS: Human colorectal cancer cells were exposed to clinically relevant doses of gamma rays, and the mechanism of their radioresistance was investigated. We characterized the transcription factor nuclear factor-kappaB (NF-kappaB) activation as a mechanism of inducible radioresistance in colorectal cancer and used curcumin, the active ingredient in the yellow spice turmeric, to overcome this resistance. RESULTS: Curcumin inhibited the proliferation and the post-irradiation clonogenic survival of multiple colorectal cancer cell lines. Radiation stimulated NF-kappaB activity in a dose- and time-dependent manner, whereas curcumin suppressed this radiation-induced NF-kappaB activation via inhibition of radiation-induced phosphorylation and degradation of inhibitor of kappaB alpha, inhibition of inhibitor of kappaB kinase activity, and inhibition of Akt phosphorylation. Curcumin also suppressed NF-kappaB-regulated gene products (Bcl-2, Bcl-x(L), inhibitor of apoptosis protein-2, cyclooxygenase-2, and cyclin D1). CONCLUSIONS: Our results suggest that transient inducible NF-kappaB activation provides a prosurvival response to radiation that may account for development of radioresistance. Curcumin blocks this signaling pathway and potentiates the antitumor effects of radiation therapy.


Assuntos
Neoplasias Colorretais/radioterapia , Curcumina/farmacologia , NF-kappa B/antagonistas & inibidores , Proteínas de Neoplasias/antagonistas & inibidores , Tolerância a Radiação/fisiologia , Radiossensibilizantes/farmacologia , Apoptose , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Neoplasias do Colo/radioterapia , Neoplasias Colorretais/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células HT29 , Humanos , NF-kappa B/metabolismo , NF-kappa B/efeitos da radiação , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/efeitos da radiação , Fosforilação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Retais/metabolismo , Neoplasias Retais/radioterapia
13.
J Proteome Res ; 8(8): 3977-86, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19545154

RESUMO

Evading apoptosis is pivotal in both of carcinogenesis and resistance to anticancer therapy. We investigated the molecules and pathways of apoptosis evasion in human hepatoma cells by irradiating hepatoma cells with optimized UV (so-called "hormetic responses"). Proteins and pathways related to hormetic responses were identified via proteomic approaches followed by reconstruction of function-networks. Of the 2326 defined protein spots, 42 distinct proteins significantly changed their expression. Eleven hormetic response proteins (HINT1, PHB, CTSD, ANXA1, LGASL1, TPT1, NPM, PRDX2, UCHL1, CERK, and C1QBP) were involved in 5 death-regulatory pathways, including the p53-dependent apoptotic pathway, protein ubiquinization, cellular redox, calcium-mediated signaling pathway, and sphingomyelin-metabolism pathway. Knockdown of HINT1 expression via RNA interference increased tumor cell resistance to apoptosis induction, while silencing NPM, UCHL1, or CERK greatly sensitized tumor cells to apoptosis induction. In conclusion, NPM, UCHL1, and CERK act as apoptosis-evasion proteins that may serve as therapeutic targets for hepatoma. Silencing their expression would increase therapeutic efficacy, thereby reducing the corresponding doses and side-effects of anticancer therapy. This model of induction of cellular hormetic responses to identify apoptosis-evasion molecules/pathways via proteomic approaches can be applied to other modalities of anticancer therapy.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/efeitos da radiação , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/metabolismo , Proteômica/métodos , Sequência de Aminoácidos , Análise de Variância , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/efeitos da radiação , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/radioterapia , Linhagem Celular Tumoral , Eletroforese em Gel Bidimensional , Humanos , Immunoblotting , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/radioterapia , Dados de Sequência Molecular , Proteínas de Neoplasias/efeitos da radiação , Proibitinas , Proteoma/metabolismo , Proteoma/efeitos da radiação , Interferência de RNA , Reprodutibilidade dos Testes , Transdução de Sinais/efeitos da radiação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Proteína Tumoral 1 Controlada por Tradução , Raios Ultravioleta
14.
Exp Biol Med (Maywood) ; 234(1): 112-22, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18997097

RESUMO

While the pre-treatment status of cancer is generally correlated with outcome, little is known about microenvironmental change caused by anti-cancer treatment and how it may affect outcome. For example, treatment may lead to induction of gene expression that promotes resistance to therapy. In the present study, we attempted to find a gene that was both induced by irradiation and associated with radioresistance in tumors. Using single-color oligo-microarrays, we analyzed the gene expression profiles of two murine squamous cell carcinomas, NR-S1, which is highly radioresistant, and SCCVII, which is radiosensitive, after irradiation with 137-Cs gamma rays or carbon ions. Candidate genes were those differentially regulated between NR-S1 and SCCVII after any kind of irradiation. Four genes, Efna1 (Ephrin-A1), Sprr1a (small proline-rich protein 1A), Srgap3 (SLIT-ROBO Rho GTPase activating protein 3) and Xrra1 [RIKEN 2 days neonate thymus thymic cells (NOD) cDNA clone E430023D08 3'], were selected as candidate genes associated with radiotherapy-induced radioresistance. We focused on Efna1, which encodes a ligand for the Eph receptor tyrosine kinase known to be involved in the vascular endothelial growth factor (VEGF) pathway. We used immunohistochemical methods to detect expression of Ephrin-A1, VEGF, and the microvascular marker CD31 in radioresistant NR-S1 tumor cells. Ephrin-A1 was detected in the cytoplasm of NR-S1 tumor cells after irradiation, but not in SCCVII tumor cells. Irradiation of NR-S1 tumor cells also led to significant increases in microvascular density, and up-regulation of VEGF expression. Our results suggest that radiotherapy-induced changes in gene expression related with angiogenesis might also modulate microenvironment and influence responsiveness of tumors.


Assuntos
Efrina-A1/genética , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Neoplasias do Timo/radioterapia , Animais , Raios gama , Masculino , Camundongos , Camundongos Endogâmicos C3H , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/efeitos da radiação , Análise de Sequência com Séries de Oligonucleotídeos , RNA Neoplásico/genética , RNA Neoplásico/efeitos da radiação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias do Timo/genética , Regulação para Cima
15.
Int J Radiat Oncol Biol Phys ; 72(2): 543-52, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18793956

RESUMO

PURPOSE: To investigate the potential of irradiation in combination with drugs targeting the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptor (DR)4 and DR5 and their mechanism of action in a cervical cancer cell line. METHODS AND MATERIALS: Recombinant human TRAIL (rhTRAIL) and the agonistic antibodies against DR4 and DR5 were added to irradiated HeLa cells. The effect was evaluated with apoptosis and cytotoxicity assays and at the protein level. Membrane receptor expression was measured with flow cytometry. Small-interfering RNA against p53, DR4, and DR5 was used to investigate their function on the combined effect. RESULTS: rhTRAIL and the agonistic DR4 and DR5 antibodies strongly enhanced 10-Gy-induced apoptosis. This extra effect was 22%, 23%, and 29% for rhTRAIL, DR4, and DR5, respectively. Irradiation increased p53 expression and increased the membrane expression of DR5 and DR4. p53 suppression, as well as small-interfering RNA against DR5, resulted in a significant downregulation of DR5 membrane expression but did not affect apoptosis induced by irradiation and rhTRAIL. After small-interfering RNA against DR4, rhTRAIL-induced apoptosis and the additive effect of irradiation on rhTRAIL-induced apoptosis were abrogated, implicating an important role for DR4 in apoptosis induced through irradiation in combination with rhTRAIL. CONCLUSION: Irradiation-induced apoptosis is strongly enhanced by targeting the pro-apoptotic TRAIL receptors DR4 or DR5. Irradiation results in a p53-dependent increase in DR5 membrane expression. The sensitizing effect of rhTRAIL on irradiation in the HeLa cell line is, however especially mediated through the DR4 receptor.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Neoplasias/efeitos da radiação , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Neoplasias do Colo do Útero/radioterapia , Apoptose/efeitos da radiação , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Colágeno Tipo XI/metabolismo , Terapia Combinada/métodos , Ensaios de Seleção de Medicamentos Antitumorais , Ativação Enzimática , Feminino , Células HeLa/efeitos dos fármacos , Células HeLa/efeitos da radiação , Humanos , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , RNA Interferente Pequeno/metabolismo , Doses de Radiação , Tolerância a Radiação , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/efeitos da radiação , Receptores do Fator de Necrose Tumoral/efeitos dos fármacos , Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/efeitos da radiação , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/metabolismo
16.
J Cell Sci ; 121(11): 1825-31, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18460583

RESUMO

Securin is a chaperone protein with bifunctional properties. It binds to separase to inhibit premature sister chromatid separation until the onset of anaphase, and it also takes part in cell-cycle arrest after UV irradiation. At metaphase-to-anaphase transition, securin is targeted for proteasomal destruction by the anaphase-promoting complex or cyclosome (APC/C), allowing activation of separase. However, although securin is reported to undergo proteasome-dependent degradation after UV irradiation, the ubiquitin ligase responsible for securin ubiquitylation has not been well characterized. In this study, we show that UV radiation induced a marked reduction of securin in both the nucleus and cytoplasm. Moreover, we show that GSK-3beta inhibitors prevent securin degradation, and that CUL1 and betaTrCP are involved in this depletion. We also confirmed that SKP1-CUL1-betaTrCP (SCF(betaTrCP)) ubiquitylates securin in vivo, and identified a conserved and unconventional betaTrCP recognition motif (DDAYPE) in the securin primary amino acid sequence of humans, nonhuman primates and rodents. Furthermore, downregulation of betaTrCP caused an accumulation of securin in non-irradiated cells. We conclude that SCF(betaTrCP) is the E3 ubiquitin ligase responsible for securin degradation after UV irradiation, and that it is involved in securin turnover in nonstressed cells.


Assuntos
Proteínas de Neoplasias/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/efeitos da radiação , Raios Ultravioleta , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Motivos de Aminoácidos/fisiologia , Animais , Células COS , Ciclo Celular/fisiologia , Ciclo Celular/efeitos da radiação , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/efeitos da radiação , Chlorocebus aethiops , Proteínas Culina/metabolismo , Citoplasma/metabolismo , Citoplasma/efeitos da radiação , Regulação para Baixo/fisiologia , Inibidores Enzimáticos/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Células HeLa , Humanos , Proteínas de Neoplasias/efeitos da radiação , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Securina , Ubiquitinação/fisiologia
17.
Brain Res ; 1188: 25-34, 2008 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-18048012

RESUMO

Although meningiomas represent the most common class of tumors of the central nervous system, the molecular events underlying their genesis and development are still not well defined, and therapeutic approaches based on the genetics of these tumors are currently lacking. In the present study we have used the immunoblotting technique to show that the p16(INK4A), Cdk6 and pRB proteins are differentially expressed in primary meningioma cells with 20-, 30- and 36-fold difference between the lowest and the highest levels of each protein, respectively. In addition, we present evidence that the level of the anti-apoptosis survivin protein is high in these benign tumors. Moreover, the annexin V-associated flow cytometry technique was used to show that 60% of meningioma cell cultures underwent apoptosis in response to both gamma-rays and cisplatin, and 50% of these cells exhibited significant sensitivity to hydroxyurea. These agents triggered apoptosis through the mitochondrial pathway, by increasing the Bax/Bcl-2 ratio. Interestingly, the induction of apoptosis following radiation and cisplatin was significant in all cells that expressed low levels of p16(INK4A), Cdk6 and pRB proteins. These data shed more light on the molecular biology of meningioma cells and suggest that survivin and proteins of the RB pathway could play a determinant role in the development and the treatment of meningiomas.


Assuntos
Apoptose/fisiologia , Quinase 6 Dependente de Ciclina/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína do Retinoblastoma/metabolismo , Adolescente , Adulto , Idoso , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Quinase 6 Dependente de Ciclina/efeitos dos fármacos , Quinase 6 Dependente de Ciclina/efeitos da radiação , Inibidor p16 de Quinase Dependente de Ciclina/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/efeitos da radiação , Feminino , Citometria de Fluxo , Humanos , Hidroxiureia/farmacologia , Immunoblotting , Proteínas Inibidoras de Apoptose , Masculino , Neoplasias Meníngeas/tratamento farmacológico , Neoplasias Meníngeas/radioterapia , Meningioma/tratamento farmacológico , Meningioma/radioterapia , Proteínas Associadas aos Microtúbulos/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/efeitos da radiação , Pessoa de Meia-Idade , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/efeitos da radiação , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/efeitos da radiação , Radioterapia , Proteína do Retinoblastoma/efeitos dos fármacos , Proteína do Retinoblastoma/efeitos da radiação , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Survivina , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , Regulação para Cima/efeitos da radiação , Proteína X Associada a bcl-2/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo , Proteína X Associada a bcl-2/efeitos da radiação
18.
Int J Radiat Oncol Biol Phys ; 67(5): 1519-25, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17394948

RESUMO

PURPOSE: Survivin, a member of the inhibitor of apoptosis gene family, has also been shown to regulate mitosis. It binds Aurora B kinase and the inner centromere protein to form the chromosome passenger complex. Both Aurora B and survivin are overexpressed in many tumors. In this study, we examined whether irradiation affected survivin and Aurora B expression in mesothelioma cells, and how inhibition of these molecules affected radiosensitivity. METHODS AND MATERIALS: ZM447439 and survivin antisense oligonucleotides were used to inhibit survivin and Aurora B kinase respectively. Western blot was performed to determine the expression of survivin, Aurora B, phosphorylated-histone H3 (Ser 10), and caspase cleavage. Multinucleated cells were counted using flow cytometry, and cell survival after treatment was determined using clonogenic assay. RESULTS: At 3-Gy irradiation an increase was observed in levels of survivin and Aurora B as well as the kinase activity of Aurora B, with an increase in G2/M phase. The radiation-induced upregulation of these molecules was effectively attenuated by antisense oligonucleotides against survivin and a small-molecule inhibitor of Aurora B, ZM447439. Dual inhibition of survivin and Aurora B synergistically radiosensitized mesothelioma cells with a dose enhancement ratio of 2.55. This treatment resulted in increased formation of multinucleated cells after irradiation but did not increase levels of cleaved caspase 3. CONCLUSION: Inhibition of survivin and Aurora B induces mitotic cell arrest in mesothelioma cells after irradiation. These two proteins may be potential therapeutic targets for the enhancement of radiotherapy in malignant pleural mesothelioma.


Assuntos
Histonas/metabolismo , Mesotelioma/radioterapia , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Mitose/efeitos dos fármacos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Aurora Quinase B , Aurora Quinases , Benzamidas , Caspase 3/metabolismo , Caspase 3/efeitos da radiação , Sobrevivência Celular , Fase G2/efeitos da radiação , Histonas/efeitos da radiação , Humanos , Proteínas Inibidoras de Apoptose , Mesotelioma/metabolismo , Mesotelioma/patologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/efeitos da radiação , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/efeitos da radiação , Oligonucleotídeos Antissenso/farmacologia , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/efeitos da radiação , Quinazolinas , Tolerância a Radiação/efeitos dos fármacos , Survivina , Regulação para Cima/efeitos da radiação
19.
Cancer Lett ; 248(2): 292-8, 2007 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-16959403

RESUMO

Deregulation of survivin expression is implicated in tumorigenesis. To examine the regulation of survivin expression in response to DNA damage, we exposed A549 human lung cancer cells to ultraviolet C (UVC) radiation, which induces DNA single-strand breakage. UVC irradiation induced G(2)-M arrest that was accompanied by accumulation of p53 and subsequent down-regulation of survivin. Depletion of p53 by RNA interference prevented the UVC-induced down-regulation of survivin. Furthermore, depletion of survivin resulted in G(2)-M arrest, suggesting that down-regulation of survivin by p53 contributes to the p53-dependent G(2)-M checkpoint triggered by DNA damage.


Assuntos
Ciclo Celular/efeitos da radiação , Neoplasias Pulmonares/metabolismo , Proteínas Associadas aos Microtúbulos/efeitos da radiação , Proteínas de Neoplasias/efeitos da radiação , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta/efeitos adversos , Linhagem Celular Tumoral , Regulação para Baixo , Citometria de Fluxo , Humanos , Immunoblotting , Proteínas Inibidoras de Apoptose , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Interferência de RNA , Survivina , Proteína Supressora de Tumor p53/genética
20.
Bioelectromagnetics ; 28(2): 99-108, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17004241

RESUMO

An in vitro study focusing on the effects of low-level radiofrequency (RF) fields from mobile radio base stations employing the International Mobile Telecommunication 2000 (IMT-2000) cellular system was conducted to test the hypothesis that modulated RF fields act to induce phosphorylation and overexpression of heat shock protein hsp27. First, we evaluated the responses of human cells to microwave exposure at a specific absorption rate (SAR) of 80 mW/kg, which corresponds to the limit of the average whole-body SAR for general public exposure defined as a basic restriction in the International Commission on Non-Ionizing Radiation Protection (ICNIRP) guidelines. Second, we investigated whether continuous wave (CW) and Wideband Code Division Multiple Access (W-CDMA) modulated signal RF fields at 2.1425 GHz induced activation or gene expression of hsp27 and other heat shock proteins (hsps). Human glioblastoma A172 cells were exposed to W-CDMA radiation at SARs of 80 and 800 mW/kg for 2-48 h, and CW radiation at 80 mW/kg for 24 h. Human IMR-90 fibroblasts from fetal lungs were exposed to W-CDMA at 80 and 800 mW/kg for 2 or 28 h, and CW at 80 mW/kg for 28 h. Under the RF field exposure conditions described above, no significant differences in the expression levels of phosphorylated hsp27 at serine 82 (hsp27[pS82]) were observed between the test groups exposed to W-CDMA or CW signal and the sham-exposed negative controls, as evaluated immediately after the exposure periods by bead-based multiplex assays. Moreover, no noticeable differences in the gene expression of hsps were observed between the test groups and the negative controls by DNA Chip analysis. Our results confirm that exposure to low-level RF field up to 800 mW/kg does not induce phosphorylation of hsp27 or expression of hsp gene family.


Assuntos
Telefone Celular , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/efeitos da radiação , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/efeitos da radiação , Fosforilação/efeitos da radiação , Neoplasias Encefálicas , Linhagem Celular Tumoral , Exposição Ambiental , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Glioblastoma , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/genética , Humanos , Chaperonas Moleculares , Proteínas de Neoplasias/genética , Fosfosserina/metabolismo , Fosfosserina/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...