Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Biomed Pharmacother ; 129: 110427, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32574974

ABSTRACT

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.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , Diterpenes/toxicity , Fibroblasts/drug effects , Genomic Instability/drug effects , Phenanthrenes/toxicity , Protein Kinase Inhibitors/pharmacology , Cell Line , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Epoxy Compounds/toxicity , Fibroblasts/enzymology , Fibroblasts/pathology , Histones/metabolism , Humans , Ku Autoantigen/metabolism , Phosphorylation , Telomerase/genetics , Telomerase/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism
2.
Nat Commun ; 9(1): 4342, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30337542

ABSTRACT

Oncolytic virotherapy is a promising therapeutic strategy that uses replication-competent viruses to selectively destroy malignancies. However, the therapeutic effect of certain oncolytic viruses (OVs) varies among cancer patients. Thus, it is necessary to overcome resistance to OVs through rationally designed combination strategies. Here, through an anticancer drug screening, we show that DNA-dependent protein kinase (DNA-PK) inhibition sensitizes cancer cells to OV M1 and improves therapeutic effects in refractory cancer models in vivo and in patient tumour samples. Infection of M1 virus triggers the transcription of interferons (IFNs) and the activation of the antiviral response, which can be abolished by pretreatment of DNA-PK inhibitor (DNA-PKI), resulting in selectively enhanced replication of OV M1 within malignancies. Furthermore, DNA-PK inhibition promotes the DNA damage response induced by M1 virus, leading to increased tumour cell apoptosis. Together, our study identifies the combination of DNA-PKI and OV M1 as a potential treatment for cancers.


Subject(s)
Antiviral Agents/pharmacology , DNA Damage , DNA-Activated Protein Kinase/antagonists & inhibitors , Oncolytic Viruses/physiology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Combined Modality Therapy , DNA-Activated Protein Kinase/metabolism , Drug Evaluation, Preclinical , Endoplasmic Reticulum Stress/drug effects , Humans , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Oncolytic Virotherapy , Protein Kinase Inhibitors/pharmacology , Rats
3.
Oncotarget ; 7(40): 65504-65513, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27602767

ABSTRACT

Radiotherapy is based on the induction of lethal DNA damage, primarily DNA double-strand breaks (DSB). Efficient DSB repair via Non-Homologous End Joining or Homologous Recombination can therefore undermine the efficacy of radiotherapy. By suppressing DNA-DSB repair with hyperthermia (HT) and DNA-PKcs inhibitor NU7441 (DNA-PKcsi), we aim to enhance the effect of radiation.The sensitizing effect of HT for 1 hour at 42°C and DNA-PKcsi [1 µM] to radiation treatment was investigated in cervical and breast cancer cells, primary breast cancer sphere cells (BCSCs) enriched for cancer stem cells, and in an in vivo human tumor model. A significant radio-enhancement effect was observed for all cell types when DNA-PKcsi and HT were applied separately, and when both were combined, HT and DNA-PKcsi enhanced radio-sensitivity to an even greater extent. Strikingly, combined treatment resulted in significantly lower survival rates, 2 to 2.5 fold increase in apoptosis, more residual DNA-DSB 6 h post treatment and a G2-phase arrest. In addition, tumor growth analysis in vivo showed significant reduction in tumor growth and elevated caspase-3 activity when radiation was combined with HT and DNA-PKcsi compared to radiation alone. Importantly, no toxic side effects of HT or DNA-PKcsi were found.In conclusion, inhibiting DNA-DSB repair using HT and DNA-PKcsi before radiotherapy leads to enhanced cytotoxicity in cancer cells. This effect was even noticed in the more radio-resistant BCSCs, which are clearly sensitized by combined treatment. Therefore, the addition of HT and DNA-PKcsi to conventional radiotherapy is promising and might contribute to more efficient tumor control and patient outcome.


Subject(s)
Breast Neoplasms/therapy , Chromones/pharmacology , DNA-Activated Protein Kinase/antagonists & inhibitors , Hyperthermia, Induced , Morpholines/pharmacology , Neoplastic Stem Cells/radiation effects , Radiation-Sensitizing Agents/pharmacology , Uterine Cervical Neoplasms/therapy , Animals , Breast Neoplasms/pathology , DNA Breaks, Double-Stranded , DNA Damage , DNA End-Joining Repair , DNA Repair , Female , Homologous Recombination , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Radiation Tolerance , Radiotherapy , Tumor Cells, Cultured , Uterine Cervical Neoplasms/pathology
4.
Biomed Pharmacother ; 83: 693-703, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27470570

ABSTRACT

DNA-dependent protein kinase (DNA-PK) is a key enzyme in non-homologous DNA end joining (NHEJ) repair pathway. The targeted inhibition of such enzyme would furnish a valuable option for cancer treatment. In this study we report the development of validation of enzyme homology model, and the subsequent use of this model to perform docking-based virtual screening against a database of FDA-approved drugs. The nominated highest ranking hits (Praziquantel and Dutasteride) were subjected to biological investigation. Additionally, molecular dynamic study was carried-out for binding mode exploration. Results of the biological evaluation revealed that both compounds inhibit the DNA-PK enzymatic activity at relatively high concentration levels with an IC50 of 17.3µM for praziquantel and >20µM for dutasteride. Furthermore, both agents enhanced the anti-proliferative effects of doxorubicin and cisplatin on breast cancer (MCF7) and lung cancer (A549) cell lines. This result indicates that these two hits are good candidate as DNA-PK inhibitors and worth further structural modifications to enhance their enzyme inhibitory effects.


Subject(s)
Computer Simulation , DNA-Activated Protein Kinase/antagonists & inhibitors , Drug Evaluation, Preclinical , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Kinase Inhibitors/analysis , Protein Kinase Inhibitors/chemistry , Structural Homology, Protein , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catalytic Domain , DNA-Activated Protein Kinase/chemistry , DNA-Activated Protein Kinase/metabolism , Dutasteride/chemistry , Dutasteride/pharmacology , Humans , Ligands , Praziquantel/chemistry , Praziquantel/pharmacology , Protein Kinase Inhibitors/pharmacology , ROC Curve
5.
J Nutr Biochem ; 24(5): 781-7, 2013 May.
Article in English | MEDLINE | ID: mdl-22841545

ABSTRACT

Selenium induces a senescence response in cells through induction of ataxia-telangiectasia mutated (ATM) and reactive oxygen species (ROS). Although a role of the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) in DNA double-strand break repair is established, it is unclear how these proteins function in response to selenium-induced oxidative stress and senescence induction. In this study, we demonstrated that pretreating normal human diploid fibroblasts with DNA-PK kinase inhibitor NU 7026 suppressed selenium-induced senescence response. Selenium treatment induced phosphorylation of DNA-PKcs on Thr-2647 and Ser-2056, the extent of which was decreased in the presence of ATM kinase inhibitor KU 55933 or the antioxidants N-acetylcysteine or 2,2,6,6-tetramethylpiperidine-1-oxyl. In contrast, the selenium-induced phosphorylation of ATM on Ser-1981 was not affected by NU 7026. Cells deficient in DNA-PKcs or pretreated with NU 7026 or N-acetylcysteine were defective in selenite-induced ROS formation. Taken together, these results indicate a distinct role of DNA-PKcs, in which this kinase can respond to and feed forward selenium-induced ROS formation and is placed downstream of ATM in the resultant senescence response.


Subject(s)
Catalytic Domain , Cellular Senescence/drug effects , DNA-Activated Protein Kinase/metabolism , Oxidative Stress/drug effects , Selenium/pharmacology , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cells, Cultured , Chromones/pharmacology , DNA Repair/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/genetics , Dextrans/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Morpholines/pharmacology , Mutation , Phosphorylation , Pyrones/pharmacology , Reactive Oxygen Species/metabolism , Selenious Acid/pharmacology
6.
Br J Haematol ; 152(1): 61-71, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21083655

ABSTRACT

Defects in the DNA damage response pathway [e.g. del(17p)] are associated with drug-resistant B-cell chronic lymphocytic leukaemia (CLL). We previously demonstrated that over-expression of DNA-dependent protein kinase (DNA-PK) correlates with chemo-resistance and that inhibition of DNA-PK sensitizes CLL cells to chemotherapeutics. Here, we investigated expression of DNA-PK and other proteins that impact on drug resistance, and evaluated the effects of a DNA-PK inhibitor (NU7441) on mitoxantrone-induced cytotoxicity in CLL cells. NU7441 sensitized cells from 42/49 CLL samples to mitoxantrone, with sensitization ranging from 2- to 200-fold Co-culture of CLL cells in conditioned stromal medium increased chemoresistance but did not reduce sensitization by NU7441. Mitoxantrone treatment induced γH2AX foci and NU7441 increased their longevity (24 h). NU7441 prevented mitoxantrone-induced autophosphorylation of the DNA-PK catalytic subunit (DNA-PKcs) at Ser 2056, confirming that DNA-PK participates in repair of mitoxantrone-induced DNA damage. del(17p) cases were more resistant to mitoxantrone than del(13q) cases, but were resensitized (7-16 fold) by co-incubation with NU7441. Expression of DNA-PKcs, Ku80, P-glycoprotein and topoisomerase IIß were significantly higher in del(17p) cases. PRKDC mRNA levels correlated with DNA-PKcs protein expression, which predicted shorter survival. These data confirm the potential of DNA-PK as a therapeutic target in poor prognosis CLL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Cell Death/drug effects , Chromones/pharmacology , Culture Media, Conditioned , DNA Repair/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/drug effects , DNA-Activated Protein Kinase/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mitoxantrone/pharmacology , Morpholines/pharmacology , Neoplasm Proteins/metabolism , Phosphorylation/drug effects , Tumor Cells, Cultured
7.
Clin Cancer Res ; 14(12): 3984-92, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18559621

ABSTRACT

PURPOSE: del(17p), del(11q), and associated p53 dysfunction predict for short survival and chemoresistance in B-cell chronic lymphocytic leukemia (CLL). DNA-dependent protein kinase (DNA-PK) is activated by DNA damage and mediates DNA double-strand break repair. We hypothesized that inhibiting DNA-PK would sensitize CLL cells to drug-induced DNA damage and that this approach could increase the therapeutic index of agents used to treat CLL. EXPERIMENTAL DESIGN: Fifty-four CLL cases were characterized for poor prognosis markers [del(17p), del(11q), CD38, and ZAP-70]. In selected cases, DNA-PK catalytic subunit (DNA-PKcs) expression and activity and p53 function were also measured. Ex vivo viability assays established sensitivity to fludarabine and chlorambucil and also tested the ability of a novel DNA-PK inhibitor (NU7441) to sensitize CLL cells to these drugs. The effects of NU7441 on fludarabine-induced DNA damage repair were also assessed (Comet assays and detection of gammaH2AX). RESULTS: DNA-PKcs levels correlated with DNA-PK activity and varied 50-fold between cases but were consistently higher in del(17p) (P = 0.01) and del(11q) cases. NU7441 sensitized CLL cells to chlorambucil and fludarabine, including cases with del(17p), del(11q), p53 dysfunction, or high levels of DNA-PKcs. NU7441 increased fludarabine-induced double-strand breaks and abrogated drug-induced autophosphorylation of DNA-PKcs at Ser2056. High DNA-PK levels predicted for reduced treatment-free interval. CONCLUSIONS: These data validate the concept of targeting DNA-PKcs in poor risk CLL, and demonstrate a mechanistic rationale for use of a DNA-PK inhibitor. The novel observation that DNA-PKcs is overexpressed in del(17p) and del(11q) cases indicates that DNA-PK may contribute to disease progression in CLL.


Subject(s)
Chromones/therapeutic use , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/physiology , Drug Delivery Systems , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Morpholines/therapeutic use , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/physiology , DNA-Activated Protein Kinase/metabolism , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/drug effects , Female , Humans , Male , Middle Aged , Prognosis , Tumor Cells, Cultured
8.
Br J Cancer ; 93(9): 1011-8, 2005 Oct 31.
Article in English | MEDLINE | ID: mdl-16249792

ABSTRACT

In this study we investigated the in vitro time dependence of radiosensitisation, pharmacokinetics and metabolism of NU7026, a novel inhibitor of the DNA repair enzyme DNA-dependent protein kinase (DNA-PK). At a dose of 10 muM, which is nontoxic to cells per se, a minimum NU7026 exposure of 4 h in combination with 3 Gy radiation is required for a significant radiosensitisation effect in CH1 human ovarian cancer cells. Following intravenous administration to mice at 5 mg kg(-1), NU7026 underwent rapid plasma clearance (0.108 l h(-1)) and this was largely attributed to extensive metabolism. Bioavailability following interperitoneal (i.p.) and p.o. administration at 20 mg kg(-1) was 20 and 15%, respectively. Investigation of NU7026 metabolism profiles in plasma and urine indicated that the compound undergoes multiple hydroxylations. A glucuronide conjugate of a bis-hydroxylated metabolite represented the major excretion product in urine. Identification of the major oxidation site as C-2 of the morpholine ring was confirmed by the fact that the plasma clearance of NU7107 (an analogue of NU7026 methylated at C-2 and C-6 of the morpholine ring) was four-fold slower than that of NU7026. The pharmacokinetic simulations performed predict that NU7026 will have to be administered four times per day at 100 mg kg(-1) i.p. in order to obtain the drug exposure required for radiosensitisation.


Subject(s)
Chromones/metabolism , Chromones/pharmacokinetics , DNA-Activated Protein Kinase/antagonists & inhibitors , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Morpholines/metabolism , Morpholines/pharmacokinetics , Ovarian Neoplasms/metabolism , Animals , Biological Availability , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Drug Evaluation, Preclinical , Female , Gamma Rays , Humans , Mice , Mice, Inbred BALB C , Ovarian Neoplasms/radiotherapy , Radiation Tolerance , Tumor Stem Cell Assay
SELECTION OF CITATIONS
SEARCH DETAIL