Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Dalton Trans ; 51(8): 3188-3197, 2022 Feb 22.
Article in English | MEDLINE | ID: mdl-35113100

ABSTRACT

Ku70 protein and topoisomerase IIα (Topo IIα) are promising targets of anticancer drugs, which play critical roles in DNA repair and replication processes. Three platinum(II) complexes, [PtCl(NH3)2(9-(pyridin-2-ylmethyl)-9H-carbazole)]NO3 (OPPC), [PtCl(NH3)2(9-(pyridin-3-ylmethyl)-9H-carbazole)]NO3 (MPPC), and [PtCl(NH3)2(9-(pyridin-4-ylmethyl)-9H-carbazole)]NO3 (PPPC), were designed as inhibitors of Ku70 and Topo IIα. Their antitumor activity and inhibitory efficacy on Ku70 and Topo IIα were investigated on cellular and molecular levels. OPPC exhibited high antiproliferative activity against various cancer cell lines, with acute toxicity to mice being lower than that of cisplatin. Moreover, OPPC could enter cancer cells effectively and cause DNA damage, which was evidenced by the enhanced expression of γ-H2AX, Chk1/2 phosphorylation, p53 and cell cycle arrest. OPPC also downregulated the DNA damage repair protein Ku70 and inhibited the formation of Ku70 foci-the central points or loci of Ku70, which would suppress DNA repair and induce a nonhomologous end joining response in cancer cells. More importantly, these complexes showed inhibition towards Topo IIα; in particular, OPPC was more effective than MPPC and PPPC. In the Topo IIα knockdown cells, Ku70 and Topo IIα were directly associated with the DNA damage and apoptotic response. The molecular docking provided detailed structural insights into the interactions of the complexes with Topo IIα. This study demonstrates that the cytotoxicity of these complexes is associated with the DNA damage and repair pathways mediated by Ku70 and Topo IIα; OPPC is an effective inhibitor of Ku70 and Topo IIα and restrains cancer cells via a mechanism utterly distinct from that of cisplatin.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Ku Autoantigen/antagonists & inhibitors , Platinum Compounds/chemical synthesis , Platinum Compounds/pharmacology , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Cell Line, Tumor , DNA Topoisomerases, Type II , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Structure , Platinum Compounds/chemistry
2.
J Am Chem Soc ; 143(48): 20332-20342, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34817176

ABSTRACT

Chemoproteomic profiling of cysteines has emerged as a powerful method for screening the proteome-wide targets of cysteine-reactive fragments, drugs, and natural products. Herein, we report the development and an in-depth evaluation of a tetrafluoroalkyl benziodoxole (TFBX) as a cysteine-selective chemoproteomic probe. We show that this probe features numerous key improvements compared to the traditionally used cysteine-reactive probes, including a superior target occupancy, faster labeling kinetics, and broader proteomic coverage, thus enabling profiling of cysteines directly in live cells. In addition, the fluorine "signature" of probe 7 constitutes an additional advantage resulting in a more confident adduct-amino acid site assignment in mass-spectrometry-based identification workflows. We demonstrate the utility of our new probe for proteome-wide target profiling by identifying the cellular targets of (-)-myrocin G, an antiproliferative fungal natural product with a to-date unknown mechanism of action. We show that this natural product and a simplified analogue target the X-ray repair cross-complementing protein 5 (XRCC5), an ATP-dependent DNA helicase that primes DNA repair machinery for nonhomologous end joining (NHEJ) upon DNA double-strand breaks, making them the first reported inhibitors of this biomedically highly important protein. We further demonstrate that myrocins disrupt the interaction of XRCC5 with DNA leading to sensitization of cancer cells to the chemotherapeutic agent etoposide as well as UV-light-induced DNA damage. Altogether, our next-generation cysteine-reactive probe enables broader and deeper profiling of the cysteinome, rendering it a highly attractive tool for elucidation of targets of electrophilic small molecules.


Subject(s)
Cysteine/chemistry , Heterocyclic Compounds, 2-Ring/chemistry , Hydrocarbons, Fluorinated/chemistry , Molecular Probes/chemistry , Proteomics/methods , Alkylation , DNA End-Joining Repair/drug effects , Diterpenes/pharmacology , Enzyme Inhibitors/pharmacology , HEK293 Cells , HeLa Cells , Humans , Ku Autoantigen/antagonists & inhibitors , Ku Autoantigen/chemistry
3.
Gene ; 787: 145647, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-33845136

ABSTRACT

RecQ4, a member of the RecQ helicase family, is required for the maintenance of genome integrity. RecQ4 has been shown to promote the following two DNA double-strand break (DSB) repair pathways: non-homologous end joining (NHEJ) and homologous recombination (HR). However, its molecular function has not been fully elucidated. In the present study, we aimed to investigate the role of RecQ4 in NHEJ using Xenopus egg extracts. The N-terminal 598 amino acid region of Xenopus RecQ4 (N598), which lacks a central helicase domain and a downstream C-terminal region, was added to the extracts and its effect on the joining of DNA ends was analyzed. We found that N598 inhibited the joining of linearized DNA ends in the extracts. In addition, N598 inhibited DSB-induced chromatin binding of Ku70, which is essential for NHEJ, while the DSB-induced chromatin binding of the HR-associated proteins, replication protein A (RPA) and Rad51, increased upon the addition of N598. These results suggest that RecQ4 possibly influences the choice of the DSB repair pathway by influencing the association of the Ku heterodimer with the DNA ends.


Subject(s)
DNA End-Joining Repair , Ku Autoantigen/metabolism , RecQ Helicases/metabolism , Xenopus Proteins/physiology , Animals , Chromatin , DNA/metabolism , Ku Autoantigen/antagonists & inhibitors , Protein Binding , RecQ Helicases/genetics , Xenopus laevis
4.
Med Mycol ; 59(8): 763-772, 2021 Jul 14.
Article in English | MEDLINE | ID: mdl-33550403

ABSTRACT

Aspergillus fumigatus is the main cause of invasive aspergillosis, for which azole drugs are the first-line therapy. Emergence of pan-azole resistance among A. fumigatus is concerning and has been mainly attributed to mutations in the target gene (cyp51A). However, azole resistance may also result from other mutations (hmg1, hapE) or other adaptive mechanisms. We performed microevolution experiment exposing an A. fumigatus azole-susceptible strain (Ku80) to sub-minimal inhibitory concentration of voriconazole to analyze emergence of azole resistance. We obtained a strain with pan-azole resistance (Ku80R), which was partially reversible after drug relief, and without mutations in cyp51A, hmg1, and hapE. Transcriptomic analyses revealed overexpression of the transcription factor asg1, several ATP-binding cassette (ABC) and major facilitator superfamily transporters and genes of the ergosterol biosynthesis pathway in Ku80R. Sterol analysis showed a significant decrease of the ergosterol mass under voriconazole exposure in Ku80, but not in Ku80R. However, the proportion of the sterol compounds was similar between both strains. To further assess the role of transporters, we used the ABC transporter inhibitor milbemycine oxime (MLB). MLB inhibited transporter activity in both Ku80 and Ku80R and demonstrated some potentiating effect on azole activity. Criteria for synergism were reached for MLB and posaconazole against Ku80. Finally, deletion of asg1 revealed some role of this transcription factor in controlling drug transporter expression, but had no impact on azole susceptibility.This work provides further insight in mechanisms of azole stress adaptation and suggests that drug transporters inhibition may represent a novel therapeutic target. LAY SUMMARY: A pan-azole-resistant strain was generated in vitro, in which drug transporter overexpression was a major trait. Analyses suggested a role of the transporter inhibitor milbemycin oxime in inhibiting drug transporters and potentiating azole activity.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus fumigatus/genetics , Azoles/pharmacology , ATP-Binding Cassette Transporters/metabolism , Aspergillus fumigatus/drug effects , CCAAT-Binding Factor/genetics , Cell Membrane/chemistry , Cell Membrane/metabolism , Cytochrome P-450 Enzyme System/genetics , Drug Resistance, Fungal , Fungal Proteins/genetics , Gas Chromatography-Mass Spectrometry , HMGB1 Protein/genetics , Ku Autoantigen/antagonists & inhibitors , Ku Autoantigen/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sterols/analysis , Transcriptome , Voriconazole/pharmacology
5.
Nucleic Acids Res ; 48(20): 11536-11550, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33119767

ABSTRACT

DNA-dependent protein kinase (DNA-PK) plays a critical role in the non-homologous end joining (NHEJ) repair pathway and the DNA damage response (DDR). DNA-PK has therefore been pursued for the development of anti-cancer therapeutics in combination with ionizing radiation (IR). We report the discovery of a new class of DNA-PK inhibitors that act via a novel mechanism of action, inhibition of the Ku-DNA interaction. We have developed a series of highly potent and specific Ku-DNA binding inhibitors (Ku-DBi's) that block the Ku-DNA interaction and inhibit DNA-PK kinase activity. Ku-DBi's directly interact with the Ku and inhibit in vitro NHEJ, cellular NHEJ, and potentiate the cellular activity of radiomimetic agents and IR. Analysis of Ku-null cells demonstrates that Ku-DBi's cellular activity is a direct result of Ku inhibition, as Ku-null cells are insensitive to Ku-DBi's. The utility of Ku-DBi's was also revealed in a CRISPR gene-editing model where we demonstrate that the efficiency of gene insertion events was increased in cells pre-treated with Ku-DBi's, consistent with inhibition of NHEJ and activation of homologous recombination to facilitate gene insertion. These data demonstrate the discovery and application of new series of compounds that modulate DNA repair pathways via a unique mechanism of action.


Subject(s)
DNA End-Joining Repair/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , Ku Autoantigen/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Cells, Cultured , DNA/chemistry , DNA Breaks, Double-Stranded , Gene Editing , Humans , Ku Autoantigen/chemistry , Mice , Protein Kinase Inhibitors/chemistry
6.
Bioorg Chem ; 104: 104210, 2020 11.
Article in English | MEDLINE | ID: mdl-32920356

ABSTRACT

Cisplatin, a representative of platinum-based drug, is clinically and widely used in the treatment of various types of malignant cancer. However, its non-selectivity to almost all the cell lines and resistance in long-term use severely limit its scope of use. As biotin-specific uptake systems are overexpressed in many types of tumors but rarely occur in normal tissues, making biotin a promising target for cancer treatment. In the study, we synthesized the Pt(II) complex C2 and determined its biological activities. The existence of biotin enhanced the ability of the complex to target tumors, while the introduction of a naphthalimide compound makes it possible to diagnose tumors and monitor their progress. We have also introduced a known Pt(II) complex DN604, which not only retains the excellent cytotoxicity of platinum drugs, but also inhibits the expression of DNA double-strand breaks (DSBs) repair-related NHEJ protein Ku70 and HR protein Rad51. In summary, we report a novel trifunctional Pt(II) complex that could target tumor cells, monitor tumor progression, and reverse DSBs repair-induced cisplatin-resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/drug effects , Ku Autoantigen/antagonists & inhibitors , Lung Neoplasms/drug therapy , Organoplatinum Compounds/pharmacology , Rad51 Recombinase/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cisplatin/chemistry , Cisplatin/pharmacology , DNA Breaks, Double-Stranded/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Ku Autoantigen/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Molecular Structure , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/chemistry , Rad51 Recombinase/metabolism , Structure-Activity Relationship
7.
Mol Cancer Res ; 18(6): 873-882, 2020 06.
Article in English | MEDLINE | ID: mdl-32098827

ABSTRACT

The development of therapeutic agents that specifically target cancer cells while sparing healthy tissue could be used to enhance the efficacy of cancer therapy without increasing its toxicity. Specific targeting of cancer cells can be achieved through the use of pH-low insertion peptides (pHLIP), which take advantage of the acidity of the tumor microenvironment to deliver cargoes selectively to tumor cells. We developed a pHLIP-peptide nucleic acid (PNA) conjugate as an antisense reagent to reduce expression of the otherwise undruggable DNA double-strand break repair factor, KU80, and thereby radiosensitize tumor cells. Increased antisense activity of the pHLIP-PNA conjugate was achieved by partial mini-PEG sidechain substitution of the PNA at the gamma position, designated pHLIP-αKu80(γ). We evaluated selective effects of pHLIP-αKu80(γ) in cancer cells in acidic culture conditions as well as in two subcutaneous mouse tumor models. Fluorescently labeled pHLIP-αKu80(γ) delivers specifically to acidic cancer cells and accumulates preferentially in tumors when injected i.v. in mice. Furthermore, pHLIP-αKu80(γ) selectively reduced KU80 expression in cells under acidic conditions and in tumors in vivo. When pHLIP-αKu80(γ) was administered to mice prior to local tumor irradiation, tumor growth was substantially reduced compared with radiation treatment alone. Furthermore, there was no evidence of acute toxicity associated with pHLIP-αKu80(γ) administration to the mice. These results establish pHLIP-αKu80(γ) as a tumor-selective radiosensitizing agent. IMPLICATIONS: This study describes a novel agent, pHLIP-αKu80(γ), which combines PNA antisense and pHLIP technologies to selectively reduce the expression of the DNA repair factor KU80 in tumors and confer tumor-selective radiosensitization.


Subject(s)
Drug Delivery Systems , Gene Expression Regulation, Neoplastic/radiation effects , Ku Autoantigen/antagonists & inhibitors , Lung Neoplasms/radiotherapy , Membrane Proteins/chemistry , Radiation, Ionizing , Animals , Apoptosis , Cell Proliferation , Humans , Hydrogen-Ion Concentration , Ku Autoantigen/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Toxicol Lett ; 318: 104-113, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31672611

ABSTRACT

BACKGROUND: Bupivacaine (BP) is commonly used as a local anaesthetic(LA) in the clinic, but it can also cause neurotoxicity, especially in patients with diabetes. Previous studies have found that high-glucose environments can aggravate BP-induced DNA damage in nerve cells. Ku70 is subunit of the DNA damage repair enzyme DNA-PK. This study was designed to determine whether high-glucose conditions enhance BP neurotoxicity and DNA damage by inhibiting Ku70 expression. METHODS: We examined the effect of BP on apoptosis and DNA damage in murine dorsal root ganglion (DRG) neurons under hyperglycaemic conditions. Untreated DRG cells and DRG cells pretreated with NU7441, a DNA-PK inhibitor, were cultured for 3 days under normal culture conditions or with 50 mM glucose, and the cells were then treated with BP for 3 h. DNA damage was investigated via comet assays, the ratio of early to late apoptotic cells was assessed by Annexin V-FITC/PI staining, and cell viability was measured by CCK-8 assays. The protein expression levels of DNA-PK, Ku70, Bax, Bcl-2 and γH2ax were measured by immunofluorescence or Western blotting. RESULTS: Compared to its effect under normal culture conditions, BP treatment led to decreased cell viability and increased DNA damage in DRG cells grown under high-glucose conditions. The rate of DRG cell apoptosis and the expression of γH2ax, the ratio of Bax to Bcl-2 also increased under the high-glucose conditions. Furthermore, Ku70 expression was inhibited. The DNA-PK inhibitor, NU7441, could significantly inhibit DNA-PK and Ku70 expression, simultaneously further aggravating BP-induced apoptosis and DNA damage under high-glucose conditions. CONCLUSION: These data indicate that hyperglycaemia may enhance BP-induced neurotoxicity and DNA damage by inhibiting the DNA repair protein Ku70.


Subject(s)
Anesthetics, Local/toxicity , Apoptosis/drug effects , Bupivacaine/toxicity , Chromones/toxicity , Enzyme Inhibitors/toxicity , Ganglia, Spinal/drug effects , Glucose/toxicity , Ku Autoantigen/antagonists & inhibitors , Morpholines/toxicity , Neurotoxicity Syndromes/etiology , Animals , Cells, Cultured , DNA Damage , Ganglia, Spinal/enzymology , Ganglia, Spinal/pathology , Ku Autoantigen/metabolism , Mice , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/pathology , Signal Transduction/drug effects
9.
Lab Med ; 50(4): 421-425, 2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31089725

ABSTRACT

BACKGROUND: Management of pregnancy in patients with Kell-null phenotype can be challenging. The immune systems of these patients form an antibody that is universally reactive against the Kell Blood Group System and can cause hemolytic disease of the fetus and newborn. METHODS: A 29-year-old woman, pregnant for the first time, developed anti-D and anti-Ku. The mother had to have labor induced when her fetus showed signs of severe anemia, but no compatible blood was available for transfusion. The induction was delayed so that a unit of blood could be collected from the mother. RESULTS: Due to delayed cord clamping at delivery, the newborn did not have anemia and did not require a transfusion. The remaining blood was frozen for future needs. CONCLUSION: Blood donation by a pregnant woman for potential transfusion to a newborn with anemia is safe for the mother and fetus, and is possibly the only option in hemolytic disease of the newborn due to a rare antibody.


Subject(s)
Anemia/therapy , Autoimmune Diseases/therapy , Blood Donors , Ku Autoantigen/antagonists & inhibitors , Pregnancy Complications/therapy , Adult , Female , Humans , Infant, Newborn , Labor, Induced , Pregnancy
10.
Cell Physiol Biochem ; 49(6): 2111-2123, 2018.
Article in English | MEDLINE | ID: mdl-30273928

ABSTRACT

BACKGROUND/AIMS: T-Cell Acute Lymphoblastic Leukemia (T-ALL) [corrected] is an aggressive disease which is highly resistant to chemotherapy. Studies show that enhanced ability of DNA damage repair (DDR) in cancer cells plays a key role in chemotherapy resistance. Here, we suggest that defect in DDR related genes might be a promising target to destroy the genome stability of tumor cells. METHODS: Since KU70 is highly expressed in Jurkat cells, one of the most representative cell lines of ATL, we knocked down KU70 by shRNA and analyzed the impact of KU70 deficiency in Jurkat cells as well as in NOD-SCID animal models by western blot, immunofluorescence, flow cytometry and measuring DNA repair efficiency. RESULTS: It is observed that silencing of KU70 resulted in accumulated DNA damage and impaired DDR in Jurkat cells, resulting in more apoptosis, decreased cell proliferation and cell cycle arrest. DNA damage leads to DNA double-strand breaks (DSBs), which are processed by either non-homologous end joining(NHEJ) or homologous recombination(HR). In our study, both NHEJ and HR are impaired because of KU70 defect, accompanied with increased protein level of SHP-1, a dephosphorylation enzyme. In turn, SHP-1 led to dephosphorylation of SIRT1, which further impaired HR repair efficiency. Moreover, KU70 deficiency prolonged survival of Jurkat-xenografted mice. CONCLUSION: These findings suggest that targeting KU70 is a promising target for ATL and might overcome the existing difficulties in chemotherapy.


Subject(s)
DNA End-Joining Repair , Ku Autoantigen/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Recombinational DNA Repair , Sirtuin 1/metabolism , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Checkpoints , DNA Breaks, Double-Stranded , Humans , Jurkat Cells , Ku Autoantigen/antagonists & inhibitors , Ku Autoantigen/genetics , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , RNA Interference , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , Rad51 Recombinase/metabolism
11.
Int J Biochem Cell Biol ; 99: 154-160, 2018 06.
Article in English | MEDLINE | ID: mdl-29655920

ABSTRACT

The main DNA repair pathways, nonhomologous end joining (NHEJ) and homology-directed repair (HDR), are complementary to each other; hence, interruptions of the NHEJ pathway can favor HDR. Improving HDR efficiency in animal primary fibroblasts can facilitate the generation of gene knock-in animals with agricultural and biomedical values by somatic cell nuclear transfer. In this study, we used siRNA to suppress the expression of Ku70 and Ku80, which are the key factors in NHEJ pathway, to investigate the effect of Ku silencing on the HDR efficiency in pig fetal fibroblasts. Down-regulation of Ku70 and Ku80 resulted in the promotion of the frequencies of multiple HDR pathways, including homologous recombination, single strand annealing, and single-stranded oligonucleotide-mediated DNA repair. We further evaluated the effects of Ku70 and Ku80 silencing on promoting HR-mediated knock-in efficiency in two porcine endogenous genes and found a significant increase in the amount of knock-in cells in Ku-silenced fibroblasts compared with control. The RNA interference strategy will benefit the generation of cell lines and organisms with precise genetic modifications.


Subject(s)
DNA End-Joining Repair , Fetus/metabolism , Fibroblasts/metabolism , Homologous Recombination , Ku Autoantigen/metabolism , Animals , Cells, Cultured , Fetus/cytology , Fibroblasts/cytology , Ku Autoantigen/antagonists & inhibitors , Ku Autoantigen/genetics , Swine
12.
PLoS One ; 12(1): e0168165, 2017.
Article in English | MEDLINE | ID: mdl-28099440

ABSTRACT

An involvement of components of DNA-break repair (DBR) complex including DNA-dependent protein kinase (DNA-PK) and poly-ADP-ribose polymerase 1 (PARP-1) in transcription regulation in response to distinct cellular signalling has been revealed by different laboratories. Here, we explored the involvement of DNA-PK and PARP-1 in the heat shock induced transcription of Hsp70A1A. We find that inhibition of both the catalytic subunit of DNA-PK (DNA-PKc), and Ku70, a regulatory subunit of DNA-PK holo-enzyme compromises transcription of Hsp70A1A under heat shock treatment. In immunoprecipitation based experiments we find that Ku70 or DNA-PK holoenzyme associates with NFκB. This NFκB associated complex also carries PARP-1. Downregulation of both NFκB and PARP-1 compromises Hsp70A1A transcription induced by heat shock treatment. Alteration of three bases by site directed mutagenesis within the consensus κB sequence motif identified on the promoter affected inducibility of Hsp70A1A transcription by heat shock treatment. These results suggest that NFκB engaged with the κB motif on the promoter cooperates in Hsp70A1A activation under heat shock in human cells as part of a DBR complex including DNA-PK and PARP-1.


Subject(s)
DNA Repair/genetics , DNA Topoisomerases, Type II/genetics , DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/genetics , HSP70 Heat-Shock Proteins/genetics , NF-kappa B p50 Subunit/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Transcription Factor RelA/genetics , Catalytic Domain/genetics , Cell Line, Tumor , DNA Breaks , DNA-Activated Protein Kinase/metabolism , HSP70 Heat-Shock Proteins/biosynthesis , HeLa Cells , Humans , Ku Autoantigen/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/metabolism , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering/genetics , Transcription, Genetic/genetics , Up-Regulation/genetics
13.
Tumour Biol ; 37(10): 13903-13914, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27488115

ABSTRACT

The first known function of Ku70 is as a DNA repair factor in the nucleus. Using neuronal neuroblastoma cells as a model, we have established that cytosolic Ku70 binds to the pro-apoptotic protein Bax in the cytosol and blocks Bax's cell death activity. Ku70-Bax binding is regulated by Ku70 acetylation in that when Ku70 is acetylated Bax dissociates from Ku70, triggering cell death. We propose that Ku70 may act as a survival factor in these cells such that Ku70 depletion triggers Bax-dependent cell death. Here, we addressed two fundamental questions about this model: (1) Does all Bax, which is a cytosolic protein, bind to all cytosolic Ku70? and (2) Is Ku70 a survival factor in cells types other than neuronal neuroblastoma cells? We show here that, in neuronal neuroblastoma cells, only a small fraction of Ku70 binds to a small fraction of Bax; most Bax is monomeric. Interestingly, there is no free or monomeric Ku70 in the cytosol; most cytosolic Ku70 is in complex with other factors forming several high molecular weight complexes. A fraction of cytosolic Ku70 also binds to cytosolic Ku80, Ku70's binding partner in the nucleus. Ku70 may not be a survival factor in some cell types (Ku70-depletion less sensitive) because Ku70 depletion does not affect survival of these cells. These results indicate that, in addition to Ku70 acetylation, other factors may be involved in regulating Ku70-Bax binding in the Ku70-depletion less sensitive cells because Ku70 acetylation in these cells is not sufficient to dissociate Bax from Ku70 or to activate Bax.


Subject(s)
Apoptosis , Cytosol/metabolism , Gene Expression Regulation, Neoplastic , Ku Autoantigen/metabolism , Neuroblastoma/pathology , Ovarian Neoplasms/pathology , bcl-2-Associated X Protein/metabolism , Acetylation , Blotting, Western , Cell Proliferation , Female , Humans , Immunoprecipitation , Ku Autoantigen/antagonists & inhibitors , Ku Autoantigen/genetics , Neuroblastoma/metabolism , Ovarian Neoplasms/metabolism , RNA, Small Interfering/genetics , Tumor Cells, Cultured
14.
DNA Repair (Amst) ; 43: 98-106, 2016 07.
Article in English | MEDLINE | ID: mdl-27130816

ABSTRACT

Non-Homologous End-Joining (NHEJ) is the predominant pathway for the repair of DNA double strand breaks (DSBs) in human cells. The NHEJ pathway is frequently upregulated in several solid cancers as a compensatory mechanism for a separate DSB repair defect or for innate genomic instability, making this pathway a powerful target for synthetic lethality approaches. In addition, NHEJ reduces the efficacy of cancer treatment modalities which rely on the introduction of DSBs, like radiation therapy or genotoxic chemotherapy. Consequently, inhibition of the NHEJ pathway can modulate a radiation- or chemo-refractory disease presentation. The Ku70/80 heterodimer protein plays a pivotal role in the NHEJ process. It possesses a ring-shaped structure with high affinity for DSBs and serves as the first responder and central scaffold around which the rest of the repair complex is assembled. Because of this central position, the Ku70/80 dimer is a logical target for the disruption of the entire NHEJ pathway. Surprisingly, specific inhibitors of the Ku70/80 heterodimer are currently not available. We here describe an in silico, pocket-based drug discovery methodology utilizing the crystal structure of the Ku70/80 heterodimer. We identified a novel putative small molecule binding pocket and selected several potential inhibitors by computational screening. Subsequent biological screening resulted in the first identification of a compound with confirmed Ku-inhibitory activity in the low micro-molar range, capable of disrupting the binding of Ku70/80 to DNA substrates and impairing Ku-dependent activation of another NHEJ factor, the DNA-PKCS kinase. Importantly, this compound synergistically sensitized human cell lines to radiation treatment, indicating a clear potential to diminish DSB repair. The chemical scaffold we here describe can be utilized as a lead-generating platform for the design and development of a novel class of anti-cancer agents.


Subject(s)
DNA End-Joining Repair/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA/genetics , Ku Autoantigen/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Pyrimidines/pharmacology , Small Molecule Libraries/pharmacology , Cell Survival/drug effects , Cell Survival/radiation effects , Crystallography, X-Ray , DNA/metabolism , DNA Breaks, Double-Stranded , DNA-Activated Protein Kinase/chemistry , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Gamma Rays , Gene Expression , HeLa Cells , Humans , Ku Autoantigen/chemistry , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Molecular Docking Simulation , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Domains , Protein Multimerization , Protein Structure, Secondary , Pyrimidines/chemical synthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Small Molecule Libraries/chemical synthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...