Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 1188, 2024 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-38216643

RESUMO

Ku70 is a multifunctional protein with pivotal roles in DNA repair via non-homologous end-joining, V(D)J recombination, telomere maintenance, and neuronal apoptosis control. Nonetheless, its regulatory mechanisms remain elusive. Chicken Ku70 (GdKu70) cDNA has been previously cloned, and DT40 cells expressing it have significantly contributed to critical biological discoveries. GdKu70 features an additional 18 amino acids at its N-terminus compared to mammalian Ku70, the biological significance of which remains uncertain. Here, we show that the 5' flanking sequence of GdKu70 cDNA is not nearly encoded in the chicken genome. Notably, these 18 amino acids result from fusion events involving the NFE2L1 gene on chromosome 27 and the Ku70 gene on chromosome 1. Through experiments using newly cloned chicken Ku70 cDNA and specific antibodies, we demonstrated that Ku70 localizes within the cell nucleus as a heterodimer with Ku80 and promptly accumulates at DNA damage sites following injury. This suggests that the functions and spatiotemporal regulatory mechanisms of Ku70 in chickens closely resemble those in mammals. The insights and resources acquired will contribute to elucidate the various mechanisms by which Ku functions. Meanwhile, caution is advised when interpreting the previous numerous key studies that relied on GdKu70 cDNA and its expressing cells.


Assuntos
Antígenos Nucleares , Galinhas , Dano ao DNA , Autoantígeno Ku , Animais , Aminoácidos/genética , Antígenos Nucleares/metabolismo , Galinhas/genética , Galinhas/metabolismo , Clonagem Molecular , Dano ao DNA/genética , Reparo do DNA , DNA Complementar , Proteínas de Ligação a DNA/metabolismo , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Mamíferos/metabolismo
2.
Phytomedicine ; 116: 154876, 2023 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-37210962

RESUMO

BACKGROUND: Targeting DNA damage response and DNA repair proficiency of cancers is an important anticancer strategy. Kaempferol (Kae), a natural flavonoid, displays potent antitumor properties in some cancers. However, the precise underlying mechanism of Kae regulates DNA repair system are poorly understood. PURPOSE: We aim to evaluate the efficacy of Kae in the treatment of human glioma as well as the molecular mechanism regarding DNA repair. STUDY DESIGN: Effects of Kae on glioma cells were detected using CCK-8 and EdU labeling assays. The molecular mechanism of Kae on glioma was determined using RNAseq. The inhibition effects of Kae on DNA repair were verified using Immunoprecipitation, immunofluorescence, and pimEJ5-GFP report assays. For in vivo study, orthotopic xenograft models were established and treated with Kae or vehicle. Glioma development was monitored by bioluminescence imaging, Magnetic Resonance Imaging (MRI), and brain sections Hematoxylin/Eosin (HE) staining. Immunohistochemical (IHC) analysis was used to detect expression of Ku80, Ki67 and γH2AX in engrafted glioma tissue. RESULTS: We found that Kae remarkably inhibits viability of glioma cells and decreases its proliferation. Mechanistically, Kae regulates multiple functional pathways associated with cancer, including non-homologous end joining (NHEJ) repair. Further studies revealed that Kae inhibits release of Ku80 from the double-strand breaks (DSBs) sites via reducing ubiquitylation and degradation of Ku80. Therefore, Kae significantly suppresses NHEJ repair and induces accumulation of DSBs in glioma cells. Moreover, Kae displays a dramatic inhibition effects on glioma growth in an orthotopic transplantation model. These data demonstrate that Kae can induce deubiquitination of Ku80, suppress NHEJ repair and inhibit glioma growth. CONCLUSION: Our findings indicate that inhibiting release of Ku80 from the DSBs by Kae may be a potential effective approach for glioma treatment.


Assuntos
Quebras de DNA de Cadeia Dupla , Glioma , Humanos , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Quempferóis/farmacologia , Reparo do DNA por Junção de Extremidades , Glioma/tratamento farmacológico
3.
Nutrients ; 14(19)2022 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-36235593

RESUMO

Oxidative stress induces DNA damage which can be repaired by DNA repair proteins, such as Ku70/80. Excess reactive oxygen species (ROS) stimulate the activation of caspase-3, which degrades Ku 70/80. Cells with decreased Ku protein levels undergo apoptosis. Astaxanthin exerts antioxidant activity by inducing the expression of catalase, an antioxidant enzyme, in gastric epithelial cells. Therefore, astaxanthin may inhibit oxidative stress-induced DNA damage by preventing Ku protein degradation and thereby suppressing apoptosis. Ku proteins can be degraded via ubiquitination and neddylation which adds ubiquitin-like protein to substrate proteins. We aimed to determine whether oxidative stress decreases Ku70/80 expression through the ubiquitin-proteasome pathway to induce apoptosis and whether astaxanthin inhibits oxidative stress-induced changes in gastric epithelial AGS cells. We induced oxidative stress caused by the treatment of ß-D-glucose (G) and glucose oxidase (GO) in the cells. As a result, the G/GO treatment increased ROS levels, decreased nuclear Ku protein levels and Ku-DNA-binding activity, and induced the ubiquitination of Ku80. G/GO increased the DNA damage marker levels (γ-H2AX; DNA fragmentation) and apoptosis marker annexin V-positive cells and cell death. Astaxanthin inhibited G/GO-induced alterations, including Ku degradation in AGS cells. MLN4924, a neddylation inhibitor, and MG132, a proteasome inhibitor, suppressed G/GO-mediated DNA fragmentation and decreased cell viability. These results indicated that G/GO-induced oxidative stress causes Ku protein loss through the ubiquitin-proteasome pathway, resulting in DNA fragmentation and apoptotic cell death. Astaxanthin inhibited oxidative stress-mediated apoptosis via the reduction of ROS levels and inhibition of Ku protein degradation. In conclusion, dietary astaxanthin supplementation or astaxanthin-rich food consumption may be effective for preventing or delaying oxidative stress-mediated cell damage by suppressing Ku protein loss and apoptosis in gastric epithelial cells.


Assuntos
Antioxidantes , Complexo de Endopeptidases do Proteassoma , Anexina A5/metabolismo , Anexina A5/farmacologia , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Apoptose , Caspase 3/metabolismo , Catalase/metabolismo , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Células Epiteliais/metabolismo , Glucose/metabolismo , Glucose Oxidase/metabolismo , Glucose Oxidase/farmacologia , Autoantígeno Ku/metabolismo , Estresse Oxidativo , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Proteólise , Espécies Reativas de Oxigênio/metabolismo , Ubiquitinas/metabolismo , Ubiquitinas/farmacologia , Xantofilas
4.
Biomed Pharmacother ; 129: 110427, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32574974

RESUMO

Triptolide is a multi-functional natural small molecular compound extracted from a traditional Chinese medicinal herb. Triptolide and its derivatives exhibit cytotoxicity through inducing DNA damage, therefore increasing sensitivity to DNA-damage based chemotherapy or radiotherapy in different types of cells. However, the regulatory mechanism of genotoxicity by triptolide, and the loss of genome integrity induced by triptolide are not fully understood. Here, we measured the effects of triptolide on genome integrity in a human fibroblast line HCA2-hTERT using the neutral comet assay. We demonstrated that treating cells with triptolide induced genomic instability in HCA2-hTERT cells. Furthermore, we observed the accumulation of γH2AX foci in triptolide treated cells than control cells at 24 h post ionizing radiation. Further mechanistic studies indicated that triptolide inhibited the enzymatic activity of DNA-PKcs, the critical nonhomologous end joining factor. In vitro kinase activity assays showed that triptolide suppressed the kinase activity of DNA-PKcs and molecular docking also predicted a potential interaction between triptolide and DNA-PKcs. As a consequence, we found that triptolide treatment enhanced the interaction between DNA-PKcs and KU80 and hampered the following recruitment of 53BP1. Altogether, our finding provides a new perspective about the toxicity of triptolide in non-cancer cells and highlights the necessity of taking genome effects of triptolide and its derivatives into consideration in the future clinical and research applications.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Diterpenos/toxicidade , Fibroblastos/efeitos dos fármacos , Instabilidade Genômica/efeitos dos fármacos , Fenantrenos/toxicidade , Inibidores de Proteínas Quinases/farmacologia , Linhagem Celular , Proteína Quinase Ativada por DNA/genética , Proteína Quinase Ativada por DNA/metabolismo , Compostos de Epóxi/toxicidade , Fibroblastos/enzimologia , Fibroblastos/patologia , Histonas/metabolismo , Humanos , Autoantígeno Ku/metabolismo , Fosforilação , Telomerase/genética , Telomerase/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo
5.
J Trace Elem Med Biol ; 55: 89-95, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31345372

RESUMO

Gastric cancer is one of the most prevalent cancers in northern Iran. The DNA repair genes X-ray repair cross-complementing (XRCC) group 5, XRCC6, which are important members of non-homologous end-joining repair system, play an important role in repairing the DNA double-strand breaks. Chronic exposure to heavy metals has long been recognized as being capable of augmenting gastric cancer incidence among exposed human populations. Since trace elements could directly or indirectly damage DNA, and polymorphism in DNA DSBs-repair genes can alter the capacity of system repair, we assumed that XRCC5 VNTR and XRCC6-61C >G polymorphism also impress the DSBs-repair system ability and contribute to gastric cancer. Therefore, the objective of this research was to evaluate the tissue accumulation of Selenium (Se), Cadmium (Cd) and Arsenic (As), and XRCC5 VNTR, XRCC6-61C >G polymorphisms in cancerous and non-cancerous tissues in Golestan province. The study population included 46 gastric cancer patients and 43 cancer-free controls. Two polymorphisms of XRCC5, XRCC6 were genotyped using polymerase chain reaction (PCR) or polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. Further employed was atomic absorption spectroscopy so as to determine the levels of Se, Cd and As. Finally, the data were analyzed by SPSS (version 16) statistical software. The Se level was significantly higher in tumors as compared to non-tumor tissues, but there was no significant correlation between As and Cd in cancerous and noncancerous tissues. Allele frequencies of the selected genes were not statistically different between groups regarding XRCC6 (-61C>G). XRCC5 0R/0R, 0R/1R, 1R/1R, and 0R/2R genotypes were more common in cancerous group. High levels of Se in cancerous tissues vs. non-cancerous tissues may be one of the carcinogenic factors; in Golestan province, unlike other regions of Iran and the world, the level of Se is high, hence the higher risks of gastric cancer.


Assuntos
Arsênio/análise , Cádmio/análise , Reparo do DNA/genética , Autoantígeno Ku/genética , Polimorfismo de Nucleotídeo Único/genética , Selênio/análise , Neoplasias Gástricas/genética , DNA de Neoplasias/genética , DNA de Neoplasias/isolamento & purificação , Feminino , Perfilação da Expressão Gênica , Genótipo , Humanos , Irã (Geográfico) , Masculino , Pessoa de Meia-Idade
6.
Anticancer Res ; 38(1): 131-136, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29277765

RESUMO

BACKGROUND/AIM: The aim of the present study was to investigate the radio-sensitizing efficacy of curcumin, a traditional Chinese medicine (TCM) on colon cancer cells in vitro and in vivo. MATERIALS AND METHODS: Human colon cancer HT-29 cells were treated with curcumin (2.5 µM), irradiation (10 Gy) and the combination of irradiation and curcumin. Cell proliferation was assessed using the MTT assay. Apoptotic cells were detected by Annexin V-PE/7-AAD analysis. PCR was performed to determine differential-expression profiling of 95 DNA-repair genes in irradiated cells and cells treated with both irradiation and curcumin. Differentially-expressed genes were confirmed by Western blotting. In vivo radio-sensitizing efficacy of curcumin was assessed in a xenograft mouse model of HT-29 colon cancer. Curcumin was administrated daily by intraperitoneal injection at 20 mg/kg/dose. Mice received irradiation (10 Gy) twice weekly. Apoptosis of the cancer cells following treatment was determined by TUNEL staining. RESULTS: Irradiation induced proliferation inhibition and apoptosis of HT-29 cells in vitro. Concurrent curcumin treatment sensitized the HT-29 tumor to irradiation (p<0.01). DNA repair-related genes CCNH and XRCC5 were upregulated and LIG4 and PNKP downregulated by the combination of curcumin and irradiation compared with irradiation alone (p<0.05). Combined treatment of curcumin and irradiation resulted in a significantly greater tumor-growth inhibition and apoptosis compared to irradiation treatment alone (p<0.01). CONCLUSION: Curcumin sensitizes human colon cancer in vitro and in vivo to radiation. Downregulation of LIG4 and PNKP and upregulation of XRCC5 and CCNH DNA-repair-related genes were involved in the radio-sensitizing efficacy of curcumin in colon cancer.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/radioterapia , Curcumina/farmacologia , Curcumina/uso terapêutico , Radiossensibilizantes/farmacologia , Radiossensibilizantes/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Ciclina H/genética , Ciclina H/metabolismo , DNA Ligase Dependente de ATP/genética , DNA Ligase Dependente de ATP/metabolismo , Reparo do DNA/genética , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Células HT29 , Humanos , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Medicina Tradicional Chinesa , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Carga Tumoral/efeitos dos fármacos
7.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(7): 926-30, 2016 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-27363274

RESUMO

Objective To investigate the effect of insulin in combination with selenium on p38-mitogen-activated protein kinase/CREB-binding protein (p38MAPK/CBP) pathway in rats with diabetic cardiomyopathy. Methods Fifty SD rats were randomly grouped into control group, diabetic cardiomyopathy (DCM) group, diabetic cardiomyopathy with insulin treatment (DCM-In) group, diabetic cardiomyopathy with selenium treatment (DCM-Se) group, and diabetic cardiomyopathy with insulin and selenium combination treatment (DCM-In-Se) group. Flow cytometry was used to analyze cell cycle. TUNEL staining was used to detect cardiomyocyte apoptosis. Western blotting was used to examine the levels of cyclin D1, cyclin E, Bax, Bcl-2, p38MAPK, p-p38MAPK, CBP and Ku70. Co-immunoprecipitation was used to examine the acetylation status of Ku70. Results Insulin in combination with selenium significantly inhibited cardiomyocyte apoptosis, increased Bcl-2 levels and decreased Bax, cyclin D1, cyclin E, p38MAPK, p-p38MAPK, CBP, Ku70 and acetylated Ku70 levels. Conclusion The combined treatment of insulin and selenium suppresses cardiomyocyte apoptosis by inhibiting p38MAPK/CBP pathway.


Assuntos
Apoptose/efeitos dos fármacos , Proteína de Ligação a CREB/metabolismo , Cardiomiopatias Diabéticas/tratamento farmacológico , Insulina/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Selênio/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Acetilação/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Western Blotting , Ciclina D1/metabolismo , Ciclina E/metabolismo , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/patologia , Citometria de Fluxo , Hipoglicemiantes/farmacologia , Autoantígeno Ku/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Distribuição Aleatória , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
8.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(7): 940-4, 2016 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-27363277

RESUMO

Objective To investigate the effect of evodiamine on the radiosensitivity of esophageal squamous cell cancer Eca-109 cells. Methods Eca-109 cells were treated with various concentrations of evodiamine [(10, 20, 40, 60, 80, 100, 120) µg/mL], and then cell proliferation was examined by MTT assay. After the optimal evodiamine concentration was determined, the cells were divided into radiation group (0, 2, 4, 6, 8 Gy X-ray radiation) and radiation combined with evodiamine group (80 µg/mL evodiamine and 0, 2, 4, 6, 8 Gy X-ray radiation) .The radiosensitivity of Eca-109 cells was detected using colony formation assay. Flow cytometry was used to determine cell cycle of Eca-109 cells. The protein expressions of Ku70, Ku80, DNA-PKcs and Rad51 were examined by Western blotting. Results MTT assay showed that evodiamine decreased the proliferation of Eca-109 cells in a concentration-dependent manner. The inhibition reached the maximal level at 80 µg/mL. Compared with radiotherapy alone, the combination of 80 µg/mL evodiamine and radiotherapy improved survival curve and decreased the values of D0 and Dq. Sensitizer enhancement ratio was 1.86±0.06. Furthermore, cell cycle analysis revealed that evodiamine suppressed radiotherapy-induced the G2/M arrest. Additionally, evodiamine treatment also significantly inhibited radiotherapy-induced increase in Ku70, Ku80, DNA-PKcs and Rad51 expressions. Conclusion Evodiamine enhances radiosensitivity of Eca-109 cells during radiotherapy. The effect may be associated with the inhibition of G2/M arrest and the attenuation of Ku70, Ku80, DNA-PKcs and Rad51 expressions.


Assuntos
Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Quinazolinas/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Proteína Quinase Ativada por DNA/metabolismo , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Humanos , Autoantígeno Ku/metabolismo , Proteínas Nucleares/metabolismo , Extratos Vegetais/farmacologia , Rad51 Recombinase/metabolismo , Raios X
9.
PLoS One ; 10(4): e0122977, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25902193

RESUMO

Hyperthermia as an anticancer method has been paid increasing attention in recent years. Several studies have shown that hyperthermia can kill tumor cells by inducing apoptosis. However, the underlying molecular mechanisms of hyperthermia-induced apoptosis are largely unknown. To investigate the effects and molecular mechanism of hyperthermia on the apoptosis in renal carcinoma 786-O cells, we firstly examined apoptosis and Ku expression in 786-O cell line treated with heat exposure (42°C for 0-4 h). The results showed that hyperthermia induced apoptosis of 786-O cells, and suppressed significantly Ku80 expression, but not Ku70 expression. Next, we knock-down Ku80 in 786-O cells, generating stable cell line 786-O-shKu80, and detected apoptosis, cell survival and cell cycle distribution. Our data showed higher apoptotic rate and lower surviving fraction in the stable cell line 786-O-shKu80 compared with those in control cells, exposed to the same heat stress (42°C for 0-4 h). Moreover, the results also showed suppression of Ku80 led to G2/M phase arrest in the stable cell line 786-O-shKu80 following heat treatment. Together, these findings indicate that Ku80 may play an important role in hyperthermia-induced apoptosis and heat-sensitivity of renal carcinoma cells through influencing the cell cycle distribution.


Assuntos
Antígenos Nucleares/genética , Apoptose , Carcinoma de Células Renais/patologia , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Hipertermia Induzida , Neoplasias Renais/patologia , Ciclo Celular , Linhagem Celular Tumoral , Temperatura Alta , Humanos , Autoantígeno Ku
10.
Mol Endocrinol ; 28(9): 1571-84, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25000480

RESUMO

The mineralocorticoid receptor (MR) plays a central role in salt and water homeostasis via the kidney; however, inappropriate activation of the MR in the heart can lead to heart failure. A selective MR modulator that antagonizes MR signaling in the heart but not the kidney would provide the cardiovascular protection of current MR antagonists but allow for normal electrolyte balance. The development of such a pharmaceutical requires an understanding of coregulators and their tissue-selective interactions with the MR, which is currently limited by the small repertoire of MR coregulators described in the literature. To identify potential novel MR coregulators, we used T7 phage display to screen tissue-selective cDNA libraries for MR-interacting proteins. Thirty MR binding peptides were identified, from which three were chosen for further characterization based on their nuclear localization and their interaction with other MR-interacting proteins or, in the case of x-ray repair cross-complementing protein 6, its known status as an androgen receptor coregulator. Eukaryotic elongation factor 1A1, structure-specific recognition protein 1, and x-ray repair cross-complementing protein 6 modulated MR-mediated transcription in a ligand-, cell- and/or promoter-specific manner and colocalized with the MR upon agonist treatment when imaged using immunofluorescence microscopy. These results highlight the utility of phage display for rapid and sensitive screening of MR binding proteins and suggest that eukaryotic elongation factor 1A1, structure-specific recognition protein 1, and x-ray repair cross-complementing protein 6 may be potential MR coactivators whose activity is dependent on the ligand, cellular context, and target gene promoter.


Assuntos
Biblioteca de Peptídeos , Receptores de Mineralocorticoides/metabolismo , Antígenos Nucleares/metabolismo , Bacteriófago T7/metabolismo , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/metabolismo , Biblioteca Gênica , Células HEK293 , Proteínas de Grupo de Alta Mobilidade/metabolismo , Humanos , Autoantígeno Ku , Ligantes , Microscopia de Fluorescência , Fator 1 de Elongação de Peptídeos/metabolismo , Receptores Androgênicos/metabolismo , Transcrição Gênica , Ativação Transcricional , Fatores de Elongação da Transcrição/metabolismo
11.
J Tradit Chin Med ; 34(3): 329-37, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24992761

RESUMO

OBJECTIVE: To explore the role and mechanism of a radiation protection cream (Rp) in the treatment of radiation dermatitis, and to accumulate necessary technical information for a new drug report on Rp. METHODS: High-performance liquid chromatography was used to establish the method of measuring the main effective ingredients of sovereign and adjuvant herbs of Rp drugs, and to formulate the draft quality standards of Rp. A total of 48 Sprague-Dawley male rats were randomly divided into the Model, Trolamine cream (Tc), Rp and Blank groups according to a random number table method. The skin of each rat's buttocks was irradiated using an electron linear accelerator to establish an acute radiation dermatitis model. The histological changes were observed under light microscopy and electron microscopy during wound healing and the effect of Rp on rat fibroblast Ku70/80 gene expression was detected at the transcriptional level. RESULTS: Pathological examination revealed that Rp protected the cellular and subcellular structures of skin after irradiation, promoting the proliferation and restoration of collagen fibers. Ku70/80 mRNA expression levels in the Rp and Tc groups were higher than that in the model group (P < 0.05). Moreover, The majority of grade radiation dermatitis relative to the Model, Rp and Tc groups for reducing grade III and IV dermatitis efficiency were 85.7% and 69.2% (P < 0.05), respectively. The efficacy of Rp group in treating radiation dermatitis was better than the Trolamine cream group by 16.5% (P < 0.05). CONCLUSION: Compared with Tc, Rp had certain advantages in the efficacy and performance to price ratio. Thus, Rp is considered an effective alternative formulation for the prevention and treatment of radiation dermatitis.


Assuntos
Medicamentos de Ervas Chinesas/administração & dosagem , Protetores contra Radiação/administração & dosagem , Radiodermite/tratamento farmacológico , Creme para a Pele/administração & dosagem , Animais , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Avaliação de Medicamentos , Humanos , Autoantígeno Ku , Masculino , Radiodermite/genética , Radiodermite/metabolismo , Ratos , Ratos Sprague-Dawley
12.
PLoS One ; 9(4): e94213, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24728004

RESUMO

This study describes the sensitization mechanism to thermal stress by histone deacetylase inhibitors (HDACIs) in lung cancer cells and shows that Ku70, based on its acetylation status, mediates the protection of lung cancer from hyperthermia (42.5°C, 1-6 hrs). Ku70 regulates apoptosis by sequestering pro-apoptotic Bax. However, its role in thermal stress is not fully understood. The findings showed that, pre-treating lung cancer cells with HDACIs, nicotinamide (NM) or Trichostatin A (TsA) or both significantly enhanced hyperthermia-induced Bax-dependent apoptosis in PC-10 cells. We found that hyperthermia induces SirT-1, Sirtuin, upregulation but not HDAC6 or SirT-3, therefore transfection with dominant negative SirT-1 (Y/H) also eliminated the protection and resulted in more cell death by hyperthermia, in H1299 cells through Bax activation. Hyperthermia alone primed lung cancer cells to apoptosis without prominent death. After hyperthermia Bax was upregulated, Bcl-2 was downregulated, the Bax/Bcl-2 ratio was inversed and Bax/Bcl-2 heterodimer was dissociated. Although hyperthermia did not affect total Ku70 expression level, it stimulated Ku70 deacetylation, which in turn could bind more Bax in the PC-10 cells. These findings suggest an escape mechanism from hyperthermia-induced Bax activation. To verify the role of Ku70 in this protection mechanism, Ku70 was silenced by siRNA. Ku70 silencing significantly sensitized the lung cancer cells to hyperthermia. The Ku70 KD cells underwent cytotoxic G1 arrest and caspase-dependant apoptosis when compared to scrambled transfectants which showed only G2/M cytostatic arrest in the cell lines investigated, suggesting an additional cell cycle-dependent, novel, role of Ku70 in protection from hyperthermia. Taken together, our data show a Ku70-dependent protection mechanism from hyperthermia. Targeting Ku70 and/or its acetylation during hyperthermia may represent a promising therapeutic approach for lung cancer.


Assuntos
Inibidores de Histona Desacetilases/farmacologia , Hipertermia Induzida/métodos , Antígenos Nucleares/metabolismo , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Temperatura Alta , Humanos , Ácidos Hidroxâmicos/farmacologia , Autoantígeno Ku , Neoplasias Pulmonares/metabolismo , Niacinamida/farmacologia , Proteína X Associada a bcl-2/metabolismo
13.
Environ Mol Mutagen ; 55(5): 436-48, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24500925

RESUMO

Radiation exposure is a serious threat to biomolecules, particularly DNA, proteins and lipids. Various exogenous substances have been reported to protect these biomolecules. In this study we explored the effect of pre-treatment with G-002M, a mixture of three active derivatives isolated from the rhizomes of Podophyllum hexandrum, on DNA damage response in irradiated human blood leukocytes. Blood was collected from healthy male volunteers, preincubated with G-002M and then irradiated with various doses of radiation. Samples were analyzed using flow cytometry to quantify DNA double strand break (DSB) biomarkers including γ-H2AX, P53BP1 and levels of ligase IV. Blood samples were irradiated in vitro and processed to determine time and dose-dependent kinetics. Semiquantitative RT-PCR was performed at various time points to measure gene expression of DNA-PKcs, Ku80, ATM, and 53BP1; each of these genes is involved in DNA repair signaling. Pre-treatment of blood with G-002M resulted in reduction of γ-H2AX and P53BP1 biomarkers levels and elevated ligase IV levels relative to non-G-002M-treated irradiated cells. These results confirm suppression in radiation-induced DNA DSBs. Samples pre-treated with G-002M and then irradiated also showed significant up-regulation of DNA-PKcs and Ku80 and downregulation of ATM and 53BP1 gene expressions, suggesting that G-002M plays a protective role against DNA damage. The protective effect of G-002M may be due to its ability to scavange radiation-induced free radicals or assist in DNA repair. Further studies are needed to decipher the role of G-002M on signaling molecules involved in radiation-induced DNA damage repair pathways.


Assuntos
Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Flavonoides/farmacologia , Raios gama/efeitos adversos , Leucócitos/efeitos dos fármacos , Podophyllum/química , Protetores contra Radiação/farmacologia , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Berberidaceae , Células Cultivadas , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/genética , Reparo do DNA/efeitos da radiação , Proteína Quinase Ativada por DNA/genética , Proteína Quinase Ativada por DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Flavonoides/química , Histonas/genética , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Autoantígeno Ku , Leucócitos/metabolismo , Leucócitos/efeitos da radiação , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Linfócitos/efeitos da radiação , Masculino , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Monócitos/efeitos da radiação , Fosforilação/efeitos dos fármacos , Fosforilação/efeitos da radiação , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
14.
PLoS One ; 8(7): e70428, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23922990

RESUMO

We report that Imetelstat, a telomerase inhibitor that binds to the RNA component of telomerase (hTR), can sensitize primary CLL lymphocytes to fludarabine in vitro. This effect was observed in lymphocytes from clinically resistant cases and with cytogenetic abnormalities associated with bad prognosis. Imetelstat mediated-sensitization to fludarabine was not associated with telomerase activity, but with the basal expression of Ku80. Since both Imetelstat and Ku80 bind hTR, we assessed 1) if Ku80 and Imetelstat alter each other's binding to hTR in vitro and 2) the effect of an oligonucleotide complementary to the Ku binding site in hTR (Ku oligo) on the survival of primary CLL lymphocytes exposed to fludarabine. We show that Imetelstat interferes with the binding of Ku70/80 (Ku) to hTR and that the Ku oligo can sensitize CLL lymphocytes to FLU. Our results suggest that Ku binding to hTR may contribute to fludarabine resistance in CLL lmphocytes. This is the first report highlighting the potentially broad effectiveness of Imetelstat in CLL, and the potential biological and clinical implications of a functional interaction between Ku and hTR in primary human cancer cells.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Leucemia Linfoide/genética , Telomerase/genética , Vidarabina/análogos & derivados , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/uso terapêutico , Domínio Catalítico/efeitos dos fármacos , Deleção Cromossômica , Cromossomos Humanos Par 11 , Cromossomos Humanos Par 17 , DNA Helicases/genética , DNA Helicases/metabolismo , Ativação Enzimática , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Humanos , Indóis/farmacologia , Autoantígeno Ku , Leucemia Linfoide/tratamento farmacológico , Leucemia Linfoide/metabolismo , Pessoa de Meia-Idade , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Oligonucleotídeos , Fosforilação , Ligação Proteica/efeitos dos fármacos , Telomerase/química , Telomerase/metabolismo , Vidarabina/farmacologia , Vidarabina/uso terapêutico
15.
Int J Mol Med ; 30(5): 1166-72, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22895634

RESUMO

Berberine, an isoquinoline derivative alkaloid, has recently been shown to have antitumor activity. The present study aimed to investigate the effects of the concomitant administration of berberine and radiation on breast cancer. The effects of berberine on the radiosensitivity of MCF-7 and MDA-MB-468 cells were evaluated by using cell clonogenic assays. Cells pre-treated with berberine or dimethyl sulfoxide (DMSO) for 24 h were irradiated using a Faxitron Cabinet X-ray System to deliver the indicated doses (0, 1, 2, 3 and 4 Gy). Changes in cell cycle distribution were determined by flow cytometry. γ-H2AX foci were detected by immunofluorescence staining. The levels of Ku70, Ku86 and RAD51 proteins were evaluated by western blot analysis. We observed that berberine increased the MCF-7 and MDA-MB-468 cell radiosensitivity with cell clonogenic assays. the radiation-induced G2/M cell cycle delay was reduced in the MCF-7 cells pre-teated with berberine. Berberine pre-treatment prolonged the persistence of DNA double-strand breaks in the MCF-7 cell line. In comparison with the control cells, the protein levels of RAD51 were decreased in the MCF-7 and MDA-MB-468 cells treated with berberine, and in the cells pre-treated with 15 µM berberine for 24 h, the level of RAD51 protein decreased significantly at the indicated time-points (0, 2, 6 and 24 h) following X-ray exposure. In conclusion, berberine sensitizes human breast cancer cells to ionizing radiation by inducing cell cycle arrest and the downregulation of the homologous recombination repair protein, RAD51. Berberine may be a promising radiosensitizer for the treatment of breast cancer.


Assuntos
Berberina/farmacologia , Radiossensibilizantes/farmacologia , Antígenos Nucleares/metabolismo , Neoplasias da Mama , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Histonas/metabolismo , Humanos , Autoantígeno Ku , Células MCF-7 , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo
16.
Nucleic Acids Res ; 40(11): e81, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22362741

RESUMO

A chemistry-based artificial restriction DNA cutter (ARCUT) was recently prepared from Ce(IV)/EDTA complex and a pair of pseudo-complementary peptide nucleic acids. This cutter has freely tunable scission-site and site specificity. In this article, homologous recombination (HR) in human cells was promoted by cutting a substrate DNA with ARCUT, and the efficiency of this bioprocess was optimized by various chemical and biological approaches. Of two kinds of terminal structure formed by ARCUT, 3'-overhang termini provided by 1.7-fold higher efficiency than 5'-overhang termini. A longer homology length (e.g. 698 bp) was about 2-fold more favorable than shorter one (e.g. 100 bp). When the cell cycle was synchronized to G2/M phase with nocodazole, the HR was promoted by about 2-fold. Repression of the NHEJ-relevant proteins Ku70 and Ku80 by siRNA increased the efficiency by 2- to 3-fold. It was indicated that appropriate combination of all these chemical and biological approaches should be very effective to promote ARCUT-mediated HR in human cells.


Assuntos
Cério/química , Ácido Edético/química , Recombinação Homóloga , Ácidos Nucleicos Peptídicos/química , Antígenos Nucleares/genética , Ciclo Celular , Linhagem Celular , DNA/química , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Humanos , Autoantígeno Ku , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Interferência de RNA , Homologia de Sequência do Ácido Nucleico
17.
Methods Enzymol ; 504: 3-28, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22264527

RESUMO

The use of nano- and microbeam techniques to induce and identify subcellular localized energy deposition within a region of a living cell provides a means to investigate the effects of low radiation doses. Particularly within the nucleus where the propagation and processing of deoxyribonucleic acid (DNA) damage (and repair) in both targeted and nontargeted cells, the latter being able to study cell-cell (bystander) effects. We have pioneered a near infrared (NIR) femtosecond laser microbeam to mimic ionizing radiation through multiphoton absorption within a 3D femtoliter volume of a highly focused Gaussian laser beam. The novel optical microbeam mimics both complex ionizing and UV-radiation-type cell damage including double strand breaks (DSBs). Using the microbeam technology, we have been able to investigate the formation of DNA DSB and subsequent recruitment of repair proteins to the submicrometer size site of damage introduced in viable cells. The use of a phosphorylated H2AX (γ-H2AX a marker for DSBs, visualized by immunofluorescent staining) and real-time imaging of fluorescently labeling proteins, the dynamics of recruitment of repair proteins in viable mammalian cells can be observed. Here we show the recruitment of ATM, p53 binding protein 1 (53BP1), and RAD51, an integral protein of the homologous recombination process in the DNA repair pathway and Ku-80-GFP involved in the nonhomologous end joining (NHEJ) pathway as exemplar repair process to show differences in the repair kinetics of DNA DSBs. The laser NIR multiphoton microbeam technology shows persistent DSBs at later times post laser irradiation which are indicative of DSBs arising at replication presumably from UV photoproducts or clustered damage containing single strand breaks (SSBs) that are also observed. Effects of the cell cycle may also be investigated in real time. Postirradiation and fixed cells studies show that in G1 cells a fraction of multiphoton laser-induced DSBs is persistent for >6h in addition to those induced at replication demonstrating the broad range of timescales taken to repair DNA damage.


Assuntos
Células/citologia , Quebras de DNA de Cadeia Dupla , Histonas/análise , Lasers , Terapia com Luz de Baixa Intensidade/métodos , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Animais , Antígenos Nucleares/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Efeito Espectador , Ciclo Celular/genética , Ciclo Celular/efeitos da radiação , Proteínas de Ciclo Celular/metabolismo , Células/efeitos da radiação , Reparo do DNA/efeitos da radiação , Replicação do DNA/genética , Replicação do DNA/efeitos da radiação , Proteínas de Ligação a DNA/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Autoantígeno Ku , Mamíferos , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Rad51 Recombinase/metabolismo , Radiação Ionizante , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Proteínas Supressoras de Tumor/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
18.
J Biomol Struct Dyn ; 28(6): 895-906, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21469750

RESUMO

Non-homologous end joining (NHEJ) is a major DNA double strand breaks (DSBs) repair pathway that maintains genome integrity. However, this pathway may reduce radiotherapy efficacy by repairing DSBs on cancer cells. This research reported a computer-aided drug design (CADD) method to identify novel inhibitors from traditional Chinese medicine (TCM) that disrupt NHEJ. We aim to inhibit Ku86, the initiator of NHEJ. By integrating binding energy evaluation and molecular dynamics simulation methods, we reported glycyrrhizic acid, macedonoside C, lithospermic acid, and salvianolic acid B as potential Ku86 inhibitors. All four TCM compounds show low binding energy and stable binding poses to Ku86. The carboxyl groups on a ligand are the major binding region by forming salt bridges at Ku86 binding sites. Additional features were defined by a carbonyl group or a dihydroxyphenyl group that form additional hydrogen bond or pi-cation respectively with the ligand binding site on Ku86. These features strengthen the binding affinity between Ku86 and the potential TCM ligand. We reported all four TCM compounds are potential Ku86 inhibitors and may be used to enhance radiotherapy for cancer treatment.


Assuntos
Quebras de DNA de Cadeia Dupla , DNA Helicases/antagonistas & inibidores , Enzimas Reparadoras do DNA/antagonistas & inibidores , Reparo do DNA/efeitos dos fármacos , Medicamentos de Ervas Chinesas/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Sítios de Ligação , DNA/metabolismo , DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Metabolismo Energético , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Autoantígeno Ku , Ligantes , Modelos Moleculares , Simulação de Dinâmica Molecular , Ligação Proteica , Estrutura Terciária de Proteína
19.
Immunohematology ; 26(3): 119-23, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21214298

RESUMO

Anti-Ku seen in K(o) (Kell-null) individuals has previously been shown to cause severe hemolytic transfusion reactions. Maternal anti-Ku can cause none or moderate to severe hemolytic disease of the fetus and newborn (HDFN). In two of four previously described HDFN cases, intrauterine transfusions were required because of severe anemia. We report a case in which maternal anti-Ku did not cause HDFN. Standard serologic methods were used for RBC antibody screening and identification, adsorption and elution of RBC antibodies, and antigen typing. A gravida 3, para 3 (G3P3) woman was first evaluated in 2006 and was found to have an IgG RBC antibody that reacted against all panel RBCs in the anti-human globulin phase. A panel of RBCs treated with DTT did not react with the antibody. The antibody failed to react with one example of K(o) RBCs. The patient's RBCs typed negative for the following Kell blood group antigens: KEL1, KEL2, KEL3, KEL4, KEL6, KEL7, KEL11, KEL13, and KEL18. These results established the presence of anti-Ku in maternal serum. The newborn was group A, D+ and required phototherapy for hyperbilirubinemia, but did not require transfusion. The woman was seen again in January 2010 during the third trimester (G4P3). At this time, anti-Ku titer was 256. She delivered a healthy group O, D+ baby boy at 37 weeks' gestation. Cord RBCs were 4+ for IgG by DAT. An eluate reacted with all RBCs tested, but did not react when tested against a panel of DTT-treated RBCs. K(o) phenotype is rare to begin with, and the maternal anti-Ku formation may require more than one pregnancy. Therefore, cases that can be evaluated for anti-Ku­related HDFN are rare. Our case contributes to serologic and clinical aspects of such rare cases.


Assuntos
Antígenos Nucleares/imunologia , Proteínas de Ligação a DNA/imunologia , Eritroblastose Fetal , Imunoglobulina G/imunologia , Antígenos Nucleares/sangue , Proteínas de Ligação a DNA/sangue , Eritroblastose Fetal/etiologia , Feminino , Humanos , Imunoglobulina G/sangue , Recém-Nascido , Sistema do Grupo Sanguíneo de Kell/análise , Autoantígeno Ku , Masculino , Gravidez , Sensibilidade e Especificidade
20.
Cancer Sci ; 99(7): 1461-70, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18422750

RESUMO

Triterpenes are the main components with cytotoxicity in Ganoderma lucidum, which is used popularly as a complementary treatment for cancer therapy in traditional Chinese medicine. To investigate the possible interaction between chemotherapeutic agents and triterpenes extracted from G. lucidum, the cytotoxicity of doxorubicin (DOX) combined with Ganoderma triterpenes (GTS) or lucidenic acid N (LCN), a purified compound, was examined in HeLa cells. The combinations targeting DOX with GTS or LCN resulted in a synergistic interaction in HeLa cells. Moreover, to identify the molecular targets of GTS, two-dimensional gel electrophoresis-based comparative proteomics was carried out and proteins with altered expression levels after GTS treatment in HeLa cells were identified by matrix-assisted laser desorption/ionization time-of-flight tandem mass spectrometry. The results of our proteomic study indicated that the GTS treatment caused regulated expression of 14 proteins, which play important roles in cell proliferation, the cell cycle, apoptosis, and oxidative stress. Flow cytometric analysis confirmed that GTS could induce weak G(0)-G(1) phase arrest and combined use of GTS with DOX could induce apoptosis in cells. Furthermore, GTS enhanced the reactive oxygen species (ROS)-producing effect of DOX, and a ROS scavenger could affect the synergism between GTS and DOX. In cells with high Ku80 protein expression, the synergism between GTS and DOX was also partly affected. Importantly, in cells with high Ku80 expression that were treated with a ROS scavenger, the synergism between GTS and DOX totally disappeared. These results suggest that the synergism between GTS and DOX might be based on GTS-induced sensitization of cells to chemotherapeutics through enhanced oxidative stress, DNA damage, and apoptosis.


Assuntos
Doxorrubicina/farmacologia , Ganoderma/química , Proteínas de Neoplasias/análise , Proteômica , Triterpenos/farmacologia , Antígenos Nucleares/análise , Western Blotting , Ciclo Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/análise , Sinergismo Farmacológico , Células HeLa , Humanos , Autoantígeno Ku , Proteína Fosfatase 2/fisiologia , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA