Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 908
Filter
1.
Clin Transl Med ; 14(5): e1690, 2024 May.
Article in English | MEDLINE | ID: mdl-38760896

ABSTRACT

INTRODUCTION: Radiation-induced pulmonary fibrosis (RIPF) is a chronic, progressive, irreversible lung interstitial disease that develops after radiotherapy. Although several previous studies have focused on the mechanism of epithelial-mesenchymal transition (EMT) in lung epithelial cells, the essential factors involved in this process remain poorly understood. The DNA-dependent protein kinase catalytic subunit (DNA-PKcs) exhibits strong repair capacity when cells undergo radiation-induced damage; whether DNA-PKcs regulates EMT during RIPF remains unclear. OBJECTIVES: To investigate the role and molecular mechanism of DNA-PKcs in RIPF and provide an important theoretical basis for utilising DNA-PKcs-targeted drugs for preventing RIPF. METHODS: DNA-PKcs knockout (DPK-/-) mice were generated via the Cas9/sgRNA technique and subjected to whole chest ionizing radiation (IR) at a 20 Gy dose. Before whole chest IR, the mice were intragastrically administered the DNA-PKcs-targeted drug VND3207. Lung tissues were collected at 1 and 5 months after IR. RESULTS: The expression of DNA-PKcs is low in pulmonary fibrosis (PF) patients. DNA-PKcs deficiency significantly exacerbated RIPF by promoting EMT in lung epithelial cells. Mechanistically, DNA-PKcs deletion by shRNA or inhibitor NU7441 maintained the protein stability of Twist1. Furthermore, AKT1 mediated the interaction between DNA-PKcs and Twist1. High Twist1 expression and EMT-associated changes caused by DNA-PKcs deletion were blocked by insulin-like growth factor-1 (IGF-1), an AKT1 agonist. The radioprotective drug VND3207 prevented IR-induced EMT and alleviated RIPF in mice by stimulating the kinase activity of DNA-PKcs. CONCLUSION: Our study clarified the critical role and mechanism of DNA-PKcs in RIPF and showed that it could be a potential target for preventing RIPF.


Subject(s)
DNA-Activated Protein Kinase , Epithelial-Mesenchymal Transition , Nuclear Proteins , Proto-Oncogene Proteins c-akt , Pulmonary Fibrosis , Twist-Related Protein 1 , Epithelial-Mesenchymal Transition/drug effects , Animals , DNA-Activated Protein Kinase/metabolism , DNA-Activated Protein Kinase/genetics , Mice , Proto-Oncogene Proteins c-akt/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Twist-Related Protein 1/metabolism , Twist-Related Protein 1/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/etiology , Ubiquitination , Humans , Mice, Knockout , DNA-Binding Proteins
2.
Int J Mol Sci ; 25(10)2024 May 08.
Article in English | MEDLINE | ID: mdl-38791158

ABSTRACT

Triple-negative breast cancer (TNBC) remains the most lethal subtype of breast cancer, characterized by poor response rates to current chemotherapies and a lack of additional effective treatment options. While approximately 30% of patients respond well to anthracycline- and taxane-based standard-of-care chemotherapy regimens, the majority of patients experience limited improvements in clinical outcomes, highlighting the critical need for strategies to enhance the effectiveness of anthracycline/taxane-based chemotherapy in TNBC. In this study, we report on the potential of a DNA-PK inhibitor, peposertib, to improve the effectiveness of topoisomerase II (TOPO II) inhibitors, particularly anthracyclines, in TNBC. Our in vitro studies demonstrate the synergistic antiproliferative activity of peposertib in combination with doxorubicin, epirubicin and etoposide in multiple TNBC cell lines. Downstream analysis revealed the induction of ATM-dependent compensatory signaling and p53 pathway activation under combination treatment. These in vitro findings were substantiated by pronounced anti-tumor effects observed in mice bearing subcutaneously implanted tumors. We established a well-tolerated preclinical treatment regimen combining peposertib with pegylated liposomal doxorubicin (PLD) and demonstrated strong anti-tumor efficacy in cell-line-derived and patient-derived TNBC xenograft models in vivo. Taken together, our findings provide evidence that co-treatment with peposertib has the potential to enhance the efficacy of anthracycline/TOPO II-based chemotherapies, and it provides a promising strategy to improve treatment outcomes for TNBC patients.


Subject(s)
Doxorubicin , Drug Synergism , Topoisomerase II Inhibitors , Triple Negative Breast Neoplasms , Xenograft Model Antitumor Assays , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Humans , Animals , Female , Mice , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/therapeutic use , Cell Line, Tumor , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Doxorubicin/analogs & derivatives , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , Sulfones/pharmacology , Cell Proliferation/drug effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Polyethylene Glycols/pharmacology , Etoposide/pharmacology , Etoposide/therapeutic use , DNA Topoisomerases, Type II/metabolism , Epirubicin/pharmacology
3.
J Med Chem ; 67(8): 6253-6267, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38587857

ABSTRACT

In this work, a novel series of heterotricyclic DNA-PK inhibitors were rationally designed, synthesized, and assessed for their biological activity. In the DNA-PK biochemical assay, most compounds displayed potent enzymatic activity, with IC50 values between 0.11 and 71.5 nM. Among them, SK10 exhibited the most potent DNA-PK-inhibitory activity (IC50 = 0.11 nM). Studies of the mechanism of action indicated that SK10 could lower γH2A.X expression levels and demonstrate optimal synergistic antiproliferative activity against Jurkat cells (IC50 = 25 nM) when combined with doxorubicin. Importantly, in CT26 and B16-F10 tumor-bearing mouse models, the combination therapies of SK10 with chemotherapeutic drug doxorubicin, a PD-L1 antibody, and SWS1 (a potent PD-L1 small-molecule inhibitor) demonstrated superior synergistic anticancer and potential immunomodulatory effects. Furthermore, SK10 possessed favorable in vivo pharmacokinetic properties [e.g., oral bioavailability (F) = 31.8%]. Taken together, SK10 represents a novel heterotricyclic DNA-PK inhibitor with antitumor immune effects and favorable pharmacokinetics.


Subject(s)
Antineoplastic Agents , Biological Availability , DNA-Activated Protein Kinase , Protein Kinase Inhibitors , Humans , Animals , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Administration, Oral , Immunotherapy/methods , Doxorubicin/pharmacology , Structure-Activity Relationship , Cell Proliferation/drug effects , Mice, Inbred BALB C , Drug Discovery , Mice, Inbred C57BL , Cell Line, Tumor , Drug Synergism , Female
4.
Br J Cancer ; 130(10): 1621-1634, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38575732

ABSTRACT

BACKGROUND: DNA double-strand break (DSB) induction and repair are important events for determining cell survival and the outcome of cancer radiotherapy. The DNA-dependent protein kinase (DNA-PK) complex functions at the apex of DSBs repair, and its assembly and activity are strictly regulated by post-translation modifications (PTMs)-associated interactions. However, the PTMs of the catalytic subunit DNA-PKcs and how they affect DNA-PKcs's functions are not fully understood. METHODS: Mass spectrometry analyses were performed to identify the crotonylation sites of DNA-PKcs in response to γ-ray irradiation. Co-immunoprecipitation (Co-IP), western blotting, in vitro crotonylation assays, laser microirradiation assays, in vitro DNA binding assays, in vitro DNA-PK assembly assays and IF assays were employed to confirm the crotonylation, identify the crotonylase and decrotonylase, and elucidate how crotonylation regulates the activity and function of DNA-PKcs. Subcutaneous xenografts of human HeLa GCN5 WT or HeLa GCN5 siRNA cells in BALB/c nude mice were generated and utilized to assess tumor proliferation in vivo after radiotherapy. RESULTS: Here, we reveal that K525 is an important site of DNA-PKcs for crotonylation, and whose level is sharply increased by irradiation. The histone acetyltransferase GCN5 functions as the crotonylase for K525-Kcr, while HDAC3 serves as its dedicated decrotonylase. K525 crotonylation enhances DNA binding activity of DNA-PKcs, and facilitates assembly of the DNA-PK complex. Furthermore, GCN5-mediated K525 crotonylation is indispensable for DNA-PKcs autophosphorylation and the repair of double-strand breaks in the NHEJ pathway. GCN5 suppression significantly sensitizes xenograft tumors of mice to radiotherapy. CONCLUSIONS: Our study defines K525 crotonylation of DNA-PKcs is important for the DNA-PK complex assembly and DSBs repair activity via NHEJ pathway. Targeting GCN5-mediated K525 Kcr of DNA-PKcs may be a promising therapeutic strategy for improving the outcome of cancer radiotherapy.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-Activated Protein Kinase , Mice, Inbred BALB C , Radiation Tolerance , p300-CBP Transcription Factors , Humans , Animals , DNA-Activated Protein Kinase/metabolism , Mice , p300-CBP Transcription Factors/metabolism , HeLa Cells , Mice, Nude , Female , Protein Processing, Post-Translational , Neoplasms/radiotherapy , Neoplasms/metabolism , Neoplasms/genetics , Xenograft Model Antitumor Assays
5.
Cell Death Differ ; 31(5): 683-696, 2024 May.
Article in English | MEDLINE | ID: mdl-38589496

ABSTRACT

Protein phosphatase 1 catalytic subunit gamma (PPP1CC) promotes DNA repair and tumor development and progression, however, its underlying mechanisms remain unclear. This study investigated the molecular mechanism of PPP1CC's involvement in DNA repair and the potential clinical implications. High expression of PPP1CC was significantly correlated with radioresistance and poor prognosis in human nasopharyngeal carcinoma (NPC) patients. The mechanistic study revealed that PPP1CC bound to Ku70/Ku80 heterodimers and activated DNA-PKcs by promoting DNA-PK holoenzyme formation, which enhanced nonhomologous end junction (NHEJ) -mediated DNA repair and led to radioresistance. Importantly, BRCA1-BRCA2-containing complex subunit 3 (BRCC3) interacted with PPP1CC to enhance its stability by removing the K48-linked polyubiquitin chain at Lys234 to prevent PPP1CC degradation. Therefore, BRCC3 helped the overexpressed PPP1CC to maintain its high protein level, thereby sustaining the elevation of DNA repair capacity and radioresistance. Our study identified the molecular mechanism by which PPP1CC promotes NHEJ-mediated DNA repair and radioresistance, suggesting that the BRCC3-PPP1CC-Ku70 axis is a potential therapeutic target to improve the efficacy of radiotherapy.


Subject(s)
DNA End-Joining Repair , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Protein Phosphatase 1 , Radiation Tolerance , Humans , Nasopharyngeal Carcinoma/radiotherapy , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/metabolism , Protein Phosphatase 1/metabolism , Protein Phosphatase 1/genetics , Nasopharyngeal Neoplasms/radiotherapy , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/metabolism , Radiation Tolerance/genetics , Prognosis , Cell Line, Tumor , Ku Autoantigen/metabolism , Ku Autoantigen/genetics , Animals , DNA-Activated Protein Kinase/metabolism , DNA-Activated Protein Kinase/genetics , Mice, Nude , Female , Male , DNA Repair , Mice
6.
Cell Syst ; 15(4): 339-361.e8, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38593799

ABSTRACT

The DNA-dependent protein kinase, DNA-PK, is an essential regulator of DNA damage repair. DNA-PK-driven phosphorylation events and the activated DNA damage response (DDR) pathways are also components of antiviral intrinsic and innate immune responses. Yet, it is not clear whether and how the DNA-PK response differs between these two forms of nucleic acid stress-DNA damage and DNA virus infection. Here, we define DNA-PK substrates and the signature cellular phosphoproteome response to DNA damage or infection with the nuclear-replicating DNA herpesvirus, HSV-1. We establish that DNA-PK negatively regulates the ataxia-telangiectasia-mutated (ATM) DDR kinase during viral infection. In turn, ATM blocks the binding of DNA-PK and the nuclear DNA sensor IFI16 to viral DNA, thereby inhibiting cytokine responses. However, following DNA damage, DNA-PK enhances ATM activity, which is required for IFN-ß expression. These findings demonstrate that the DDR autoregulates cytokine expression through the opposing modulation of DDR kinases.


Subject(s)
Ataxia Telangiectasia , Herpesviridae Infections , Humans , Phosphorylation , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Cytokines/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Damage
7.
Mol Cancer Ther ; 23(6): 751-765, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38588408

ABSTRACT

A majority of patients with cancer receive radiotherapy as part of their treatment regimens whether using external beam therapy or locally-delivered radioisotopes. While often effective, some tumors are inadequately controlled with radiation and radiotherapy has significant short-term and long-term toxicities for cancer survivors. Insights into molecular mechanisms involved in cellular responses to DNA breaks introduced by radiation or other cancer therapies have been gained in recent years and approaches to manipulate these responses to enhance tumor cell killing or reduce normal tissue toxicity are of great interest. Here, we report the identification and initial characterization of XRD-0394, a potent and specific dual inhibitor of two DNA damage response kinases, ATM and DNA-PKcs. This orally bioavailable molecule demonstrates significantly enhanced tumor cell kill in the setting of therapeutic ionizing irradiation in vitro and in vivo. XRD-0394 also potentiates the effectiveness of topoisomerase I inhibitors in vitro. In addition, in cells lacking BRCA1/2 XRD-0394 shows single-agent activity and synergy in combination with PARP inhibitors. A phase Ia clinical trial (NCT05002140) with XRD-0394 in combination with radiotherapy has completed. These results provide a rationale for future clinical trials with XRD-0394 in combination with radiotherapy, PARP inhibitors, and targeted delivery of topoisomerase I inhibitors.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , DNA-Activated Protein Kinase , Poly(ADP-ribose) Polymerase Inhibitors , Radiation-Sensitizing Agents , Topoisomerase I Inhibitors , Humans , Animals , Topoisomerase I Inhibitors/pharmacology , Mice , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , Radiation-Sensitizing Agents/pharmacology , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , Xenograft Model Antitumor Assays , Cell Line, Tumor , Female , Drug Synergism
8.
Exp Cell Res ; 438(1): 114036, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38614421

ABSTRACT

Ovarian cancer is the leading cause of gynecologic cancer death. Among the most innovative anti-cancer approaches, the genetic concept of synthetic lethality is that mutations in multiple genes work synergistically to effect cell death. Previous studies found that although vaccinia-related kinase-1 (VRK1) associates with DNA damage repair proteins, its underlying mechanisms remain unclear. Here, we found high VRK1 expression in ovarian tumors, and that VRK1 depletion can significantly promote apoptosis and cell cycle arrest. The effect of VRK1 knockdown on apoptosis was manifested by increased DNA damage, genomic instability, and apoptosis, and also blocked non-homologous end joining (NHEJ) by destabilizing DNA-PK. Further, we verified that VRK1 depletion enhanced sensitivity to a PARP inhibitor (PARPi), olaparib, promoting apoptosis through DNA damage, especially in ovarian cancer cell lines with high VRK1 expression. Proteins implicated in DNA damage responses are suitable targets for the development of new anti-cancer therapeutic strategies, and their combination could represent an alternative form of synthetic lethality. Therefore, normal protective DNA damage responses are impaired by combining olaparib with elimination of VRK1 and could be used to reduce drug dose and its associated toxicity. In summary, VRK1 represents both a potential biomarker for PARPi sensitivity, and a new DDR-associated therapeutic target, in ovarian cancer.


Subject(s)
DNA Damage , DNA-Activated Protein Kinase , Intracellular Signaling Peptides and Proteins , Ovarian Neoplasms , Protein Serine-Threonine Kinases , Female , Humans , Apoptosis/drug effects , Cell Line, Tumor , DNA Damage/drug effects , DNA-Activated Protein Kinase/metabolism , DNA-Activated Protein Kinase/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genomic Instability/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics
9.
Phytother Res ; 38(5): 2496-2517, 2024 May.
Article in English | MEDLINE | ID: mdl-38447978

ABSTRACT

We investigated the mechanism by which quercetin preserves mitochondrial quality control (MQC) in cardiomyocytes subjected to ischemia-reperfusion stress. An enzyme-linked immunosorbent assay was employed in the in vivo experiments to assess myocardial injury markers, measure the transcript levels of SIRT5/DNAPK-cs/MLKL during various time intervals of ischemia-reperfusion, and observe structural changes in cardiomyocytes using transmission electron microscopy. In in vitro investigations, adenovirus transfection was employed to establish a gene-modified model of DNA-PKcs, and primary cardiomyocytes were obtained from a mouse model with modified SIRT5 gene. Reverse transcription polymerase chain reaction, laser confocal microscopy, immunofluorescence localization, JC-1 fluorescence assay, Seahorse energy analysis, and various other assays were applied to corroborate the regulatory influence of quercetin on the MQC network in cardiomyocytes after ischemia-reperfusion. In vitro experiments demonstrated that ischemia-reperfusion injury caused changes in the structure of the myocardium. It was seen that quercetin had a beneficial effect on the myocardial tissue, providing protection. As the ischemia-reperfusion process continued, the levels of DNA-PKcs/SIRT5/MLKL transcripts were also found to change. In vitro investigations revealed that quercetin mitigated cardiomyocyte injury caused by mitochondrial oxidative stress through DNA-PKcs, and regulated mitophagy and mitochondrial kinetics to sustain optimal mitochondrial energy metabolism levels. Quercetin, through SIRT5 desuccinylation, modulated the stability of DNA-PKcs, and together they regulated the "mitophagy-unfolded protein response." This preserved the integrity of mitochondrial membrane and genome, mitochondrial dynamics, and mitochondrial energy metabolism. Quercetin may operate synergistically to oversee the regulation of mitophagy and the unfolded protein response through DNA-PKcs-SIRT5 interaction.


Subject(s)
Myocytes, Cardiac , Quercetin , Sirtuins , Quercetin/pharmacology , Animals , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Mice , Sirtuins/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Oxidative Stress/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , DNA-Activated Protein Kinase/metabolism , Male , Mice, Inbred C57BL , Mitophagy/drug effects
10.
Int J Med Sci ; 21(4): 714-724, 2024.
Article in English | MEDLINE | ID: mdl-38464839

ABSTRACT

Sepsis-induced cardiomyopathy (SIC) represents a severe complication of systemic infection, characterized by significant cardiac dysfunction. This study examines the role of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) and Inverted Formin 2 (INF2) in the pathogenesis of SIC, focusing on their impact on mitochondrial homeostasis and dynamics. Our research demonstrates that silencing DNA-PKcs alleviates lipopolysaccharide (LPS)-induced cardiomyocyte death and dysfunction. Using HL-1 cardiomyocytes treated with LPS, we observed that DNA-PKcs knockdown notably reverses LPS-induced cytotoxicity, indicating a protective role against cellular damage. This effect is further substantiated by the reduction in caspase-3 and caspase-9 activation, key markers of apoptosis, upon DNA-PKcs knockdown. Besides, our data further reveal that DNA-PKcs knockdown attenuates LPS-induced mitochondrial dysfunction, evidenced by improved ATP production, enhanced activities of mitochondrial respiratory complexes, and preserved mitochondrial membrane potential. Moreover, DNA-PKcs deletion counteracts LPS-induced shifts towards mitochondrial fission, indicating its regulatory influence on mitochondrial dynamics. Conclusively, our research elucidates the intricate interplay between DNA-PKcs and INF2 in the modulation of mitochondrial function and dynamics during sepsis-induced cardiomyopathy. These findings offer new insights into the molecular mechanisms underpinning SIC and suggest potential therapeutic targets for mitigating mitochondrial dysfunction in this critical condition.


Subject(s)
Cardiomyopathies , Mitochondrial Diseases , Sepsis , Humans , DNA-Activated Protein Kinase/metabolism , Mitochondrial Dynamics , Lipopolysaccharides/toxicity , Lipopolysaccharides/metabolism , Catalytic Domain , Cardiomyopathies/genetics , Myocytes, Cardiac , Sepsis/complications , Sepsis/genetics , Mitochondrial Diseases/pathology , DNA/adverse effects , DNA/metabolism
11.
Oncogene ; 43(15): 1087-1097, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38383726

ABSTRACT

BRCA1-associated protein 1 (BAP1) has emerged as a major tumor suppressor gene in diverse cancer types, notably in malignant pleural mesothelioma (DPM), and has also been identified as a germline cancer predisposition gene for DPM and other select cancers. However, its role in the response to DNA damage has remained unclear. Here, we show that BAP1 inactivation is associated with increased DNA damage both in Met-5A human mesothelial cells and human DPM cell lines. Through proteomic analyses, we identified PRKDC as an interaction partner of BAP1 protein complexes in DPM cells and 293 T human embryonic kidney cells. PRKDC encodes the catalytic subunit of DNA protein kinase (DNA-PKcs) which functions in the nonhomologous end-joining (NHEJ) pathway of DNA repair. Double-stranded DNA damage resulted in prominent nuclear expression of BAP1 in DPM cells and phosphorylation of BAP1 at serine 395. A plasmid-based NHEJ assay confirmed a significant effect of BAP1 knockdown on cellular NHEJ activity. Combination treatment with X-ray irradiation and gemcitabine (as a radiosensitizer) strongly suppressed the growth of BAP1-deficient cells. Our results suggest reciprocal positive interactions between BAP1 and DNA-PKcs, based on phosphorylation of BAP1 by the latter and deubiquitination of DNA-PKcs by BAP1. Thus, functional interaction of BAP1 with DNA-PKcs supports a role for BAP1 in NHEJ DNA repair and may provide the basis for new therapeutic strategies and new insights into its role as a tumor suppressor.


Subject(s)
Neoplasms , Proteomics , Humans , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , DNA/genetics , DNA End-Joining Repair/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
12.
Nucleic Acids Res ; 52(9): 5048-5066, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38412274

ABSTRACT

Two DNA repair pathways, non-homologous end joining (NHEJ) and alternative end joining (A-EJ), are involved in V(D)J recombination and chromosome translocation. Previous studies reported distinct repair mechanisms for chromosome translocation, with NHEJ involved in humans and A-EJ in mice predominantly. NHEJ depends on DNA-PKcs, a critical partner in synapsis formation and downstream component activation. While DNA-PKcs inhibition promotes chromosome translocations harboring microhomologies in mice, its synonymous effect in humans is not known. We find partial DNA-PKcs inhibition in human cells leads to increased translocations and the continued involvement of a dampened NHEJ. In contrast, complete DNA-PKcs inhibition substantially increased microhomology-mediated end joining (MMEJ), thus bridging the two different translocation mechanisms between human and mice. Similar to a previous study on Ku70 deletion, DNA-PKcs deletion in G1/G0-phase mouse progenitor B cell lines, significantly impairs V(D)J recombination and generated higher rates of translocations as a consequence of dysregulated coding and signal end joining. Genetic DNA-PKcs inhibition suppresses NHEJ entirely, with repair phenotypically resembling Ku70-deficient A-EJ. In contrast, we find DNA-PKcs necessary in generating the near-exclusive MMEJ associated with Lig4 deficiency. Our study underscores DNA-PKcs in suppressing illegitimate chromosome rearrangement while also contributing to MMEJ in both species.


Subject(s)
DNA End-Joining Repair , DNA-Activated Protein Kinase , Translocation, Genetic , V(D)J Recombination , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Animals , Humans , Mice , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA Ligase ATP/genetics , DNA Ligase ATP/metabolism , Ku Autoantigen/genetics , Ku Autoantigen/metabolism
13.
Nucleic Acids Res ; 52(8): 4313-4327, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38407308

ABSTRACT

The complex formed by Ku70/80 and DNA-PKcs (DNA-PK) promotes the synapsis and the joining of double strand breaks (DSBs) during canonical non-homologous end joining (c-NHEJ). In c-NHEJ during V(D)J recombination, DNA-PK promotes the processing of the ends and the opening of the DNA hairpins by recruiting and/or activating the nuclease Artemis/DCLRE1C/SNM1C. Paradoxically, DNA-PK is also required to prevent the fusions of newly replicated leading-end telomeres. Here, we describe the role for DNA-PK in controlling Apollo/DCLRE1B/SNM1B, the nuclease that resects leading-end telomeres. We show that the telomeric function of Apollo requires DNA-PKcs's kinase activity and the binding of Apollo to DNA-PK. Furthermore, AlphaFold-Multimer predicts that Apollo's nuclease domain has extensive additional interactions with DNA-PKcs, and comparison to the cryo-EM structure of Artemis bound to DNA-PK phosphorylated on the ABCDE/Thr2609 cluster suggests that DNA-PK can similarly grant Apollo access to the DNA end. In agreement, the telomeric function of DNA-PK requires the ABCDE/Thr2609 cluster. These data reveal that resection of leading-end telomeres is regulated by DNA-PK through its binding to Apollo and its (auto)phosphorylation-dependent positioning of Apollo at the DNA end, analogous but not identical to DNA-PK dependent regulation of Artemis at hairpins.


Subject(s)
DNA-Activated Protein Kinase , DNA-Binding Proteins , Endonucleases , Telomere , DNA-Activated Protein Kinase/metabolism , DNA-Activated Protein Kinase/genetics , Telomere/metabolism , Telomere/genetics , Humans , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Endonucleases/metabolism , Endonucleases/genetics , DNA End-Joining Repair , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Ku Autoantigen/metabolism , Ku Autoantigen/genetics , Protein Binding , DNA Breaks, Double-Stranded , Phosphorylation , DNA/metabolism , DNA/chemistry , DNA/genetics
14.
J Cell Sci ; 137(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38240344

ABSTRACT

Anthracyclines, topoisomerase II enzyme poisons that cause DNA damage, are the mainstay of acute myeloid leukemia (AML) treatment. However, acquired resistance to anthracyclines leads to relapse, which currently lacks effective treatment and is the cause of poor survival in individuals with AML. Therefore, the identification of the mechanisms underlying anthracycline resistance remains an unmet clinical need. Here, using patient-derived primary cultures and clinically relevant cellular models that recapitulate acquired anthracycline resistance in AML, we have found that GCN5 (also known as KAT2A) mediates transcriptional upregulation of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) in AML relapse, independently of the DNA-damage response. We demonstrate that anthracyclines fail to induce DNA damage in resistant cells, owing to the loss of expression of their target enzyme, TOP2B; this was caused by DNA-PKcs directly binding to its promoter upstream region as a transcriptional repressor. Importantly, DNA-PKcs kinase activity inhibition re-sensitized AML relapse primary cultures and cells resistant to mitoxantrone, and abrogated their tumorigenic potential in a xenograft mouse model. Taken together, our findings identify a GCN5-DNA-PKcs-TOP2B transcriptional regulatory axis as the mechanism underlying anthracycline resistance, and demonstrate the therapeutic potential of DNA-PKcs inhibition to re-sensitize resistant AML relapse cells to anthracycline.


Subject(s)
DNA-Activated Protein Kinase , Leukemia, Myeloid, Acute , Humans , Mice , Animals , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Drug Resistance, Neoplasm/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type II/therapeutic use , Anthracyclines/pharmacology , Anthracyclines/therapeutic use , Antibiotics, Antineoplastic , Recurrence , DNA , Poly-ADP-Ribose Binding Proteins
15.
Cancer Treat Res ; 186: 299-312, 2023.
Article in English | MEDLINE | ID: mdl-37978142

ABSTRACT

This chapter explores the multifaceted roles of DNA-PK with particular focus on its functions in non-homologous end-joining (NHEJ) DNA repair. DNA-PK is the primary orchestrator of NHEJ but also regulates other biologic processes. The growing understanding of varied DNA-PK biologic roles highlights new avenues for cancer treatment. However, these multiple roles also imply challenges, particularly in combination therapies, with perhaps a higher risk of clinical toxicities than was previously envisioned. These considerations underscore the need for compelling and innovative strategies to accomplish effective clinical translation.


Subject(s)
Biological Products , DNA-Binding Proteins , Humans , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , DNA/genetics , DNA Repair , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism
16.
Int J Mol Sci ; 24(20)2023 Oct 18.
Article in English | MEDLINE | ID: mdl-37895013

ABSTRACT

The non-homologous end joining pathway is vital for repairing DNA double-strand breaks (DSB), with DNA-dependent protein kinase (DNA-PK) playing a critical role. Altered DNA damage response (DDR) in chronic (CML) and acute myeloid leukemia (AML) offers potential therapeutic opportunities. We studied the therapeutic potential of AZD-7648 (DNA-PK inhibitor) in CML and AML cell lines. This study used two CML (K-562 and LAMA-84) and five AML (HEL, HL-60, KG-1, NB-4, and THP-1) cell lines. DDR gene mutations were obtained from the COSMIC database. The copy number and methylation profile were evaluated using MS-MLPA and DDR genes, and telomere length using qPCR. p53 protein expression was assessed using Western Blot, chromosomal damage through cytokinesis-block micronucleus assay, and γH2AX levels and DSB repair kinetics using flow cytometry. Cell density and viability were analyzed using trypan blue assay after treatment with AZD-7648 in concentrations ranging from 10 to 200 µM. Cell death, cell cycle distribution, and cell proliferation rate were assessed using flow cytometry. The cells displayed different DNA baseline damage, DDR gene expressions, mutations, genetic/epigenetic changes, and p53 expression. Only HEL cells displayed inefficient DSB repair. The LAMA-84, HEL, and KG-1 cells were the most sensitive to AZD-7648, whereas HL-60 and K-562 showed a lower effect on density and viability. Besides the reduction in cell proliferation, AZD-7648 induced apoptosis, cell cycle arrest, and DNA damage. In conclusion, these results suggest that AZD-7648 holds promise as a potential therapy for myeloid leukemias, however, with variations in drug sensitivity among tested cell lines, thus supporting further investigation to identify the specific factors influencing sensitivity to this DNA-PK inhibitor.


Subject(s)
Leukemia, Myeloid, Acute , Tumor Suppressor Protein p53 , Humans , Apoptosis , Cell Cycle , Cell Cycle Checkpoints , DNA/metabolism , DNA Damage , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Nature ; 623(7985): 183-192, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37853125

ABSTRACT

The DNA damage response is essential to safeguard genome integrity. Although the contribution of chromatin in DNA repair has been investigated1,2, the contribution of chromosome folding to these processes remains unclear3. Here we report that, after the production of double-stranded breaks (DSBs) in mammalian cells, ATM drives the formation of a new chromatin compartment (D compartment) through the clustering of damaged topologically associating domains, decorated with γH2AX and 53BP1. This compartment forms by a mechanism that is consistent with polymer-polymer phase separation rather than liquid-liquid phase separation. The D compartment arises mostly in G1 phase, is independent of cohesin and is enhanced after pharmacological inhibition of DNA-dependent protein kinase (DNA-PK) or R-loop accumulation. Importantly, R-loop-enriched DNA-damage-responsive genes physically localize to the D compartment, and this contributes to their optimal activation, providing a function for DSB clustering in the DNA damage response. However, DSB-induced chromosome reorganization comes at the expense of an increased rate of translocations, also observed in cancer genomes. Overall, we characterize how DSB-induced compartmentalization orchestrates the DNA damage response and highlight the critical impact of chromosome architecture in genomic instability.


Subject(s)
Cell Compartmentation , Chromatin , DNA Damage , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line , Chromatin/genetics , Chromatin/metabolism , DNA Breaks, Double-Stranded , DNA Repair , DNA-Activated Protein Kinase/metabolism , G1 Phase , Histones/metabolism , Neoplasms/genetics , R-Loop Structures , Tumor Suppressor p53-Binding Protein 1/metabolism
18.
Redox Biol ; 67: 102893, 2023 11.
Article in English | MEDLINE | ID: mdl-37741045

ABSTRACT

BACKGROUND: DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a novel instigator for mitochondrial dysfunction, and plays an important role in the pathogenesis of cardiovascular diseases. However, the role and mechanism of DNA-PKcs in angiotensin II (Ang II)-induced vascular remodeling remains obscure. METHODS: Rat aortic smooth muscle cells (SMC) and VSMC-specific DNA-PKcs knockout (DNA-PKcsΔVSMC) mice were employed to examine the role of DNA-PKcs in vascular remodeling and the underlying mechanisms. Blood pressure of mice was monitored using the tail-cuff and telemetry methods. The role of DNA-PKcs in vascular function was evaluated using vascular relaxation assessment. RESULTS: In the tunica media of remodeled mouse thoracic aortas, and renal arteries from hypertensive patients, elevated DNA-PKcs expression was observed along with its cytoplasmic translocation from nucleus, suggesting a role for DNA-PKcs in vascular remodeling. We then infused wild-type (DNA-PKcsfl/fl) and DNA-PKcsΔVSMC mice with Ang II for 14 days to establish vascular remodeling, and demonstrated that DNA-PKcsΔVSMC mice displayed attenuated vascular remodeling through inhibition of dedifferentiation of VSMCs. Moreover, deletion of DNA-PKcs in VSMCs alleviated Ang II-induced vasodilation dysfunction and hypertension. Mechanistic investigations denoted that Ang II-evoked rises in cytoplasmic DNA-PKcs interacted with dynamin-related protein 1 (Drp1) at its TQ motif to phosphorylate Drp1S616, subsequently promoting mitochondrial fragmentation and dysfunction, as well as reactive oxygen species (ROS) production. Treatment of irbesartan, an Ang II type 1 receptor (AT1R) blocker, downregulated DNA-PKcs expression in VSMCs and aortic tissues following Ang II administration. CONCLUSION: Our data revealed that cytoplasmic DNA-PKcs in VSMCs accelerated Ang II-induced vascular remodeling by interacting with Drp1 at its TQ motif and phosphorylating Drp1S616 to provoke mitochondrial fragmentation. Maneuvers targeting DNA-PKcs might be a valuable therapeutic option for the treatment of vascular remodeling and hypertension.


Subject(s)
Angiotensin II , Hypertension , Humans , Mice , Rats , Animals , Angiotensin II/metabolism , Vascular Remodeling/physiology , Catalytic Domain , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Hypertension/metabolism , DNA/metabolism , Myocytes, Smooth Muscle/metabolism
19.
Int J Mol Sci ; 24(13)2023 Jun 26.
Article in English | MEDLINE | ID: mdl-37445835

ABSTRACT

Radioresistance remains a critical obstacle in the clinical management of glioblastoma (GBM) by radiotherapy. Therefore, it is necessary to explore the molecular mechanisms underlying radioresistance to improve patient response to radiotherapy and increase the treatment efficacy. The present study aimed to elucidate the role of specificity protein 1 (Sp1) in the radioresistance of GBM cells. Different human GBM cell lines and tumor-bearing mice were exposed to ionizing radiation (IR). Cell survival was determined by the colony formation assay. The expression of genes and proteins in the cells and tissues was analyzed by RT-PCR and western blotting, respectively. The γ-H2AX, p-Sp1 and dependent protein kinase catalytic subunit (DNA-PKcs phospho S2056) foci were analyzed by immunofluorescence. Apoptotic rates were measured by flow cytometry. Sp1 was upregulated after IR in vitro and in vivo and knocking down Sp1-sensitized GBM cells to IR. Sp1 activated the DNA-PKcs promoter and increased its expression and activity. Furthermore, the loss of Sp1 delayed double-strand breaks (DSB) repair and increased IR-induced apoptosis of GBM cells. Taken together, IR upregulates Sp1 expression in GBM cells, enhancing the activity of DNA-PKcs and promoting IR-induced DSB repair, thereby leading to increased radioresistance.


Subject(s)
Glioblastoma , Humans , Animals , Mice , Glioblastoma/genetics , Glioblastoma/radiotherapy , DNA Breaks, Double-Stranded , Up-Regulation , Radiation Tolerance/genetics , DNA Repair/genetics , DNA , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Cell Line, Tumor , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism
20.
Structure ; 31(8): 895-902.e3, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37311458

ABSTRACT

The ability of humans to maintain the integrity of the genome is imperative for cellular survival. DNA double-strand breaks (DSBs) are considered the most critical type of DNA lesion, which can ultimately lead to diseases including cancer. Non-homologous end joining (NHEJ) is one of two core mechanisms utilized to repair DSBs. DNA-PK is a key component in this process and has recently been shown to form alternate long-range synaptic dimers. This has led to the proposal that these complexes can be formed before transitioning to a short-range synaptic complex. Here we present cryo-EM data representing an NHEJ supercomplex consisting of a trimer of DNA-PK in complex with XLF, XRCC4, and DNA Ligase IV. This trimer represents a complex of both long-range synaptic dimers. We discuss the potential role of the trimeric structure, and possible higher order oligomers, as structural intermediates in the NHEJ mechanism, or as functional DNA repair centers.


Subject(s)
DNA Repair Enzymes , DNA Repair , Humans , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Cryoelectron Microscopy , DNA End-Joining Repair , DNA Ligase ATP , DNA-Activated Protein Kinase/metabolism , DNA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...