Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Cancer Res ; 67(2): 634-42, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17234773

RESUMO

Ku70 is one component of a protein complex, the Ku70/Ku80 heterodimer, which binds to DNA double-strand breaks and activates DNA-dependent protein kinase (DNA-PK), leading to DNA damage repair. Our previous work has confirmed that Ku70 is important for DNA damage repair in that Ku70 deficiency compromises the ability of cells to repair DNA double-strand breaks, increases the radiosensitivity of cells, and enhances radiation-induced apoptosis. Because of the radioresistance of some human cancers, particularly glioblastoma, we examined the use of a radio-gene therapy paradigm to sensitize cells to ionizing radiation. Based on the analysis of the structure-function of Ku70 and the crystal structure of Ku70/Ku80 heterodimer, we designed and identified a candidate dominant negative fragment involving an NH(2)-terminal deletion, and designated it as DNKu70. We generated this mutant construct, stably overexpressed it in Rat-1 cells, and showed that it has a dominant negative effect (i.e., DNKu70 overexpression results in decreased Ku-DNA end-binding activity, and increases radiosensitivity). We then constructed and generated recombinant replication-defective adenovirus, with DNKu70 controlled by the cytomegalovirus promoter, and infected human glioma U-87 MG cells and human colorectal tumor HCT-8 cells. We show that the infected cells significantly express DNKu70 and are greatly radiosensitized under both aerobic and hypoxic conditions. The functional ramification of DNKu70 was further shown in vivo: expression of DNKu70 inhibits radiation-induced DNA-PK catalytic subunit autophosphorylation and prolongs the persistence of gamma-H2AX foci. If radiation-resistant tumor cells could be sensitized by down-regulating the cellular level/activity of Ku/DNA-PK, this approach could be evaluated as an adjuvant to radiation therapy.


Assuntos
Antígenos Nucleares/fisiologia , Neoplasias Colorretais/radioterapia , Proteínas de Ligação a DNA/fisiologia , Glioma/radioterapia , Adenoviridae/genética , Aerobiose , Animais , Antígenos Nucleares/biossíntese , Antígenos Nucleares/genética , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Terapia Combinada , Proteína Quinase Ativada por DNA/metabolismo , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Terapia Genética/métodos , Glioma/genética , Glioma/metabolismo , Histonas/biossíntese , Humanos , Autoantígeno Ku , Camundongos , Fosforilação/efeitos da radiação , Tolerância a Radiação/genética , Ratos , Transfecção
2.
Int J Radiat Oncol Biol Phys ; 71(2): 533-41, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18374502

RESUMO

PURPOSE: To evaluate the response of cells over-expressing dominant negative (DN) Ku70 to single and multiple small radiation doses. METHODS AND MATERIALS: Clones of fibroblasts over-expressing DNKu70, DNKu70-7, DNKu70-11, and parental Rat-1 cells were irradiated under oxic or hypoxic conditions with single or multiple doses. Cells were trypsinized 0 or 6 h after irradiation to determine surviving fraction (SF). RESULTS: Oxic DNKu70-7 or -11 cells trypsinized 6 h after irradiation were 1.52 or 1.25 and 1.28 or 1.15 times more sensitive than oxic Rat-1 at SF of 0.5 and 0.1, respectively. Hypoxic DNKu70-7 or -11 cells trypsinized 6 h after irradiation were 1.44 or 1.70 and 1.33 or 1.51 times more sensitive than hypoxic Rat-1 at SF of 0.5 and 0.1, respectively. To the multiple doses, oxic and hypoxic DNKu70-7 or -11 cells were 1.35 or 1.37 and 2.23 or 4.61 times more sensitive than oxic and hypoxic Rat-1, respectively, resulting in very small oxygen enhancement ratios. Namely, enhancement caused by DNKu70 under hypoxia after multiple doses was greater than that under oxic conditions and greater than that after single dose. CONCLUSIONS: Over-expression of DNKu70 enhances cells' response to radiation given as a single dose and as multiple small doses. The enhancement after multiple doses was stronger under hypoxic than under oxic conditions. These results encourage the use of DNKu70 fragment in a gene-radiotherapy.


Assuntos
Antígenos Nucleares/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/efeitos da radiação , Tolerância a Radiação/fisiologia , Animais , Antígenos Nucleares/genética , Antígenos Nucleares/uso terapêutico , Hipóxia Celular , Linhagem Celular , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Dano ao DNA , Reparo do DNA , Proteína Quinase Ativada por DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/uso terapêutico , Relação Dose-Resposta à Radiação , Fibroblastos/metabolismo , Autoantígeno Ku , Modelos Lineares , Doses de Radiação , Ratos , Fatores de Tempo
3.
Oncol Rep ; 39(3): 912-920, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29344644

RESUMO

The DNA-dependent protein kinase (DNA-PK) complex plays a pivotal role in non-homologous end-joining (NHEJ) repair. We investigated the mechanism of NU7441, a highly selective DNA-PK inhibitor, in NHEJ-competent mouse embryonic fibroblast (MEF) cells and NHEJ-deficient cells and explored the feasibility of its application in radiosensitizing nasopharyngeal carcinoma (NPC) cells. We generated wild-type and DNA-PKcs-/- MEF cells. Clonogenic survival assays, flow cytometry, and immunoblotting were performed to study the effect of NU7441 on survival, cell cycle, and DNA repair. NU7441 profoundly radiosensitized wild-type MEF cells and SUNE-1 cells, but not DNA-PKcs-/- MEF cells. NU7441 significantly suppressed radiation-induced DSB repair post-irradiation through unrepaired and lethal DNA damage, the cell cycle arrest. The effect was associated with the activation of cell cycle checkpoints. The present study revealed a mechanism by which inhibition of DNA-PK sensitizes cells to irradiation suggesting that radiotherapy in combination with DNA-PK inhibitor is a promising paradigm for the management of NPC which merits further investigation.


Assuntos
Carcinoma/patologia , Cromonas/farmacologia , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , Proteína Quinase Ativada por DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Raios gama/efeitos adversos , Morfolinas/farmacologia , Neoplasias Nasofaríngeas/patologia , Proteínas Nucleares/fisiologia , Radiossensibilizantes/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Carcinoma/genética , Carcinoma/terapia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Células Cultivadas , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA por Junção de Extremidades/efeitos da radiação , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Fibroblastos/efeitos da radiação , Humanos , Camundongos , Camundongos Knockout , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/terapia
4.
Oncotarget ; 8(14): 22662-22673, 2017 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-28186989

RESUMO

DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a distinct factor in the non-homologous end-joining (NHEJ) pathway involved in DNA double-strand break (DSB) repair. We examined the crosstalk between key proteins in the DSB NHEJ repair pathway and cell cycle regulation and found that mouse embryonic fibroblast (MEF) cells deficient in DNA-PKcs or Ku70 were more vulnerable to ionizing radiation (IR) compared with wild-type cells and that DSB repair was delayed. γH2AX was associated with phospho-Ataxia-telangiectasia mutated kinase (Ser1987) and phospho-checkpoint effector kinase 1 (Ser345) foci for the arrest of cell cycle through the G2/M phase. Inhibition of DNA-PKcs prolonged IR-induced G2/M phase arrest because of sequential activation of cell cycle checkpoints. DSBs were introduced, and cell cycle checkpoints were recruited after exposure to IR in nasopharyngeal carcinoma SUNE-1 cells. NU7441 radiosensitized MEF cells and SUNE-1 cells by interfering with DSB repair. Together, these results reveal a mechanism in which coupling of DSB repair with the cell cycle radiosensitizes NHEJ repair-deficient cells, justifying further development of DNA-PK inhibitors in cancer therapy.


Assuntos
Carcinoma/genética , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA por Junção de Extremidades/genética , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/antagonistas & inibidores , Autoantígeno Ku/fisiologia , Neoplasias Nasofaríngeas/genética , Proteínas Nucleares/antagonistas & inibidores , Tolerância a Radiação/genética , Animais , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Carcinoma/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Proliferação de Células , Quinase 1 do Ponto de Checagem/metabolismo , Cromonas/farmacologia , Proteína Quinase Ativada por DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/efeitos da radiação , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/efeitos da radiação , Camundongos , Camundongos Knockout , Morfolinas/farmacologia , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/patologia , Proteínas Nucleares/fisiologia , Radiação Ionizante , Radiossensibilizantes/farmacologia , Células Tumorais Cultivadas
5.
Oncotarget ; 6(33): 34732-44, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26416246

RESUMO

Bortezomib, a novel proteasome inhibitor, has been approved for treating multiple myeloma and mantle cell lymphoma and studied pre-clinically and clinically for solid tumors. Preferential cytotoxicity of bortezomib was found toward hypoxic tumor cells and endothelial cells in vitro. The purpose of this study is to investigate the role of a pretreatment hypoxic tumor microenvironment on the effects of bortezomib in vitro and ex vivo, and explore the feasibility of dynamic contrast enhanced magnetic resonance imaging (DCE MRI) to noninvasively evaluate the biological effects of bortezomib. It was shown in vitro by Western blot, flow cytometry, and ELISA that bortezomib accumulated HIF-1α in non-functional forms and blocks its hypoxia response in human colorectal cancer cell lines. Ex vivo experiments were performed with fluorescent immunohistochemical staining techniques using multiple endogenous and exogenous markers to identify hypoxia (pimonidazole, HRE-TKeGFP), blood flow/permeability (Hoechst 33342), micro-vessels (CD31 and SMA), apoptosis (cleaved caspase 3) and hypoxia response (CA9). After bortezomib administration, overall apoptosis index was significantly increased and blood perfusion was dramatically decreased in tumor xenografts. More importantly, apoptosis signals were found preferentially located in moderate and severe pretreatment hypoxic regions in both tumor and endothelial cells. Meanwhile, DCE MRI examinations showed that the tumor blood flow and permeability decreased significantly after bortezomib administration. The present study revealed that bortezomib reduces tumor hypoxia response and blood perfusion, thus, presenting antivascular properties. It will be important to determine the hypoxic/perfusion status pre- and during treatment at further translational studies.


Assuntos
Antineoplásicos/farmacologia , Bortezomib/farmacologia , Hipóxia Celular/efeitos dos fármacos , Neoplasias Experimentais/irrigação sanguínea , Microambiente Tumoral/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Meios de Contraste , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Aumento da Imagem , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Imageamento por Ressonância Magnética/métodos , Camundongos , Camundongos Nus , Neoplasias Experimentais/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Int J Radiat Biol ; 88(7): 515-22, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22617044

RESUMO

PURPOSE: To study the effect of DN (dominant-negative) Ku70 and reoxygenation on the hypoxia-induced cell-kill. MATERIALS AND METHODS: Cell lines were human colorectal carcinoma HCT8 and HT29 cells and their respective derivatives, v-HCT8 and v-HT29 infected with DNKu70-containing adenovirus. Cells were plated in glass tubes and made hypoxic by flushing N(2) gas containing 0, 0.1 or 0.5% O(2). Cell survival was determined by colony formation assay immediately after 0-96 h hypoxia. To reoxygenate medium were replaced fresh following 48 or 72 h in hypoxia and cells were incubated in aerobic environment for 2-24 h before survival assay. RESULTS: When incubated in hypoxia, cells lost reproductive capability ∼ exponentially as a function of time in hypoxia, and depending on the O(2) concentration. DNKu70 rendered cells more prone to hypoxia-induced cell-kill. Following reoxygenation cell survival increased rapidly but without detectable cell proliferation during first 24 hours. This evinced hypoxia-induced potentially lethal damage (PLD) that was repairable upon reoxygenation. DNKu70 did not significantly inhibit this repair. CONCLUSION: Hypoxia-induced cell lethality was facilitated by DNKu70, but substantially repaired upon reoxygenation. This may have negative impact on the effect of reoxygenation in cancer therapy.


Assuntos
Antígenos Nucleares/metabolismo , Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Neoplasias Experimentais/patologia , Neoplasias Experimentais/fisiopatologia , Consumo de Oxigênio , Oxigênio/metabolismo , Antígenos Nucleares/genética , Apoptose , Hipóxia Celular , Proliferação de Células , Sobrevivência Celular , Proteínas de Ligação a DNA/genética , Células HT29 , Humanos , Autoantígeno Ku , Regulação para Cima
7.
Int J Radiat Oncol Biol Phys ; 77(3): 877-85, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20510198

RESUMO

PURPOSE: To investigate the effect of recombinant replication-defective adenovirus containing dominant-negative Ku70 fragment on the response of tumor cells to multiple small radiation doses. Our ultimate goal is to demonstrate the feasibility of using this virus in gene-radiotherapy to enhance the radiation response of tumor cells. METHODS AND MATERIALS: Human colorectal HCT8 and HT29 carcinoma cells were plated in glass tubes, infected with virus (25 multiplicity of infection), and irradiated with a single dose or zero to five doses of 3 Gy each at 6-h intervals. Hypoxia was induced by flushing with 100% nitrogen gas. The cells were trypsinized 0 or 6 h after the final irradiation, and cell survival was determined by colony formation. The survival data were fitted to linear-quadratic model or exponential line. RESULTS: Virus infection enhanced the radiation response of the HCT8 and HT29 cells. The virus enhancement ratio for single-dose irradiation at a surviving fraction of 0.1 was approximately 1.3 for oxic and hypoxic HCT8 and 1.4 and 1.1 for oxic and hypoxic HT29, respectively. A similar virus enhancement ratio of 1.2-1.3 was observed for both oxic and hypoxic cells irradiated with multiple doses; however, these values were smaller than the values found for dominant-negative Ku70-transfected Rat-1 cells. This difference has been discussed. The oxygen enhancement ratio for HCT8 and HT29 receiving fractionated doses was 1.2 and 2.0, respectively, and virus infection altered them slightly. CONCLUSION: Infection of recombinant replication-defective adenovirus containing dominant-negative Ku70 fragment enhanced the response of human colorectal cancer cells to single and multiple radiation doses.


Assuntos
Adenoviridae/fisiologia , Antígenos Nucleares/metabolismo , Neoplasias Colorretais , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Vírus Defeituosos/fisiologia , Tolerância a Radiação/fisiologia , Adenoviridae/genética , Animais , Antígenos Nucleares/genética , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral/efeitos da radiação , Linhagem Celular Tumoral/virologia , Sobrevivência Celular , Ensaio de Unidades Formadoras de Colônias/métodos , Neoplasias Colorretais/patologia , Neoplasias Colorretais/radioterapia , Neoplasias Colorretais/virologia , Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Vírus Defeituosos/genética , Terapia Genética/métodos , Células HT29/efeitos da radiação , Células HT29/virologia , Humanos , Autoantígeno Ku , Modelos Lineares , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Doses de Radiação , Ratos , Transfecção/métodos
8.
Cancer Res ; 68(20): 8597-606, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18922936

RESUMO

Tumor hypoxia is important in the development and treatment of human cancers. We have developed a novel xenograft model for studying and imaging of hypoxia-induced gene expression. A hypoxia-inducible dual reporter herpes simplex virus type 1 thymidine kinase and enhanced green fluorescence protein (HSV1-TKeGFP), under the control of hypoxia response element (9HRE), was stably transfected into human colorectal HT29 cancer cells. Selected clones were further enriched by repeated live cell sorting gated for hypoxia-induced eGFP expression. Fluorescent microscopy, fluorescence-activated cell sorting, and radioactive substrate trapping assays showed strong hypoxia-induced expression of eGFP and HSV1-tk enzyme in the HT29-9HRE cells in vitro. Sequential micropositron emission tomography (PET) imaging of tumor-bearing animals, using the hypoxic cell tracer (18)F-FMISO and the reporter substrate (124)I-FIAU, yielded similar tumor hypoxia images for the HT29-9HRE xenograft but not in the parental HT29 tumor. Using autoradiography and IHC, detailed spatial distributions in tumor sections were obtained and compared for the following hypoxia-associated biomarkers in the HT29-9HRE xenograft: (124)I-FIAU, (18)F-FMISO, Hoechst (perfusion), lectin-TRITC (functional blood vessels), eGFP, pimonidazole, EF5, and CA9. Intratumoral distributions of (124)I-FIAU and (18)F-FMISO were similar, and eGFP, pimonidazole, EF5, and CA9 colocalized in the same areas but not in well-perfused regions that were positive for Hoechst and lectin-TRITC. In enabling the detection of hypoxia-induced molecular events and mapping their distribution in vivo with serial noninvasive positron emission tomography imaging, and multiple variable analysis with immunohistochemistry and fluorescence microscopy, this human xenograft model provides a valuable tool for studying tumor hypoxia and in validating existing and future exogenous markers for tumor hypoxia.


Assuntos
Hipóxia Celular , Neoplasias/metabolismo , Animais , Antígenos de Neoplasias/metabolismo , Arabinofuranosiluracila/análogos & derivados , Autorradiografia , Biomarcadores , Anidrase Carbônica IX , Anidrases Carbônicas/metabolismo , Etanidazol/análogos & derivados , Etanidazol/metabolismo , Feminino , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HT29 , Herpesvirus Humano 1/enzimologia , Humanos , Hidrocarbonetos Fluorados/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Nus , Misonidazol/análogos & derivados , Transplante de Neoplasias , Nitroimidazóis/metabolismo , Tomografia por Emissão de Pósitrons , Timidina Quinase/genética , Distribuição Tecidual , Transplante Heterólogo
9.
Int J Hyperthermia ; 22(3): 215-21, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16754341

RESUMO

In recent years, both hyperthermia and gene-therapy have been evaluated as approaches to improve cancer radiotherapy. In addition, potential exists to combine these approaches to increase the overall therapeutic efficacy. For example, it has been reported that adenovirus-mediated heat-inducible gene expression may reduce the normal tissue toxicity associated with constitutively controlled expression of therapeutic genes. In our laboratory, we have shown that adenovirus-mediated, heat-activated antisense Ku70 expression radiosensitizes tumor cells in vitro and in vivo, suggesting a novel approach to use heat-activated gene-radiotherapy to radiosensitize human tumors.However, to optimize the application of heat-activated gene-radiotherapy in the clinic, we need to develop techniques to improve the delivery of the therapeutic adenovirus and to verify/monitor the delivery non-invasively. In an ongoing study we test the effect of mild hyperthermia in improving adenovirus-medicated vector delivery in a mouse tumor model. In addition, we evaluate the use of non-invasive microPET imaging to monitor the spread of the adenoviral vector. Our preliminary results show that (1) microPET imaging can be used to monitor non-invasively the viral vector delivery and dissemination, and (2) mild heat shock leads to significantly improved viral vector distribution, in other words, a wider spatial spread, in vivo.Here, we will present a short review on the current status of hyperthermia and heat-activated gene-radiotherapy, and the potential use of PET imaging in gene therapy.


Assuntos
Terapia Genética/métodos , Hipertermia Induzida/métodos , Neoplasias/terapia , Tomografia por Emissão de Pósitrons/métodos , Adenoviridae/genética , Animais , Terapia Combinada/métodos , Reparo do DNA/efeitos da radiação , Vetores Genéticos/administração & dosagem , Humanos , Camundongos , Neoplasias/diagnóstico por imagem , Neoplasias/radioterapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA