Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.124
Filter
1.
Cell Mol Life Sci ; 81(1): 339, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39120648

ABSTRACT

Senataxin is an evolutionarily conserved DNA/RNA helicase, whose dysfunctions are linked to neurodegeneration and cancer. A main activity of this protein is the removal of R-loops, which are nucleic acid structures capable to promote DNA damage and replication stress. Here we found that Senataxin deficiency causes the release of damaged DNA into extranuclear bodies, called micronuclei, triggering the massive recruitment of cGAS, the apical sensor of the innate immunity pathway, and the downstream stimulation of interferon genes. Such cGAS-positive micronuclei are characterized by defective membrane envelope and are particularly abundant in cycling cells lacking Senataxin, but not after exposure to a DNA breaking agent or in absence of the tumor suppressor BRCA1 protein, a partner of Senataxin in R-loop removal. Micronuclei with a discontinuous membrane are normally cleared by autophagy, a process that we show is impaired in Senataxin-deficient cells. The formation of Senataxin-dependent inflamed micronuclei is promoted by the persistence of nuclear R-loops stimulated by the DSIF transcription elongation complex and the engagement of EXO1 nuclease activity on nuclear DNA. Coherently, high levels of EXO1 result in poor prognosis in a subset of tumors lacking Senataxin expression. Hence, R-loop homeostasis impairment, together with autophagy failure and unscheduled EXO1 activity, elicits innate immune response through micronuclei formation in cells lacking Senataxin.


Subject(s)
Autophagy , DNA Damage , DNA Helicases , Inflammation , Multifunctional Enzymes , Nucleotidyltransferases , R-Loop Structures , RNA Helicases , Humans , Autophagy/genetics , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/deficiency , DNA Helicases/metabolism , DNA Helicases/genetics , DNA Helicases/deficiency , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/deficiency , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics , Immunity, Innate , Inflammation/pathology , Inflammation/metabolism , Inflammation/genetics , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Phosphoproteins , RNA Helicases/metabolism , RNA Helicases/genetics
2.
Sci Adv ; 10(32): eado0636, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39121215

ABSTRACT

Ubiquitination is a crucial posttranslational modification required for the proper repair of DNA double-strand breaks (DSBs) induced by ionizing radiation (IR). DSBs are mainly repaired through homologous recombination (HR) when template DNA is present and nonhomologous end joining (NHEJ) in its absence. In addition, microhomology-mediated end joining (MMEJ) and single-strand annealing (SSA) provide backup DSBs repair pathways. However, the mechanisms controlling their use remain poorly understood. By using a high-resolution CRISPR screen of the ubiquitin system after IR, we systematically uncover genes required for cell survival and elucidate a critical role of the E3 ubiquitin ligase SCFcyclin F in cell cycle-dependent DSB repair. We show that SCFcyclin F-mediated EXO1 degradation prevents DNA end resection in mitosis, allowing MMEJ to take place. Moreover, we identify a conserved cyclin F recognition motif, distinct from the one used by other cyclins, with broad implications in cyclin specificity for cell cycle control.


Subject(s)
Cell Cycle , Cyclins , DNA Breaks, Double-Stranded , DNA Repair , Exodeoxyribonucleases , Humans , Cell Cycle/genetics , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics , Cyclins/metabolism , Cyclins/genetics , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , DNA End-Joining Repair , Ubiquitination , Radiation, Ionizing
3.
DNA Repair (Amst) ; 141: 103726, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39096697

ABSTRACT

Trypanosoma cruzi is the etiological agent of Chagas disease and a peculiar eukaryote with unique biological characteristics. DNA damage can block RNA polymerase, activating transcription-coupled nucleotide excision repair (TC-NER), a DNA repair pathway specialized in lesions that compromise transcription. If transcriptional stress is unresolved, arrested RNA polymerase can activate programmed cell death. Nonetheless, how this parasite modulates these processes is unknown. Here, we demonstrate that T. cruzi cell death after UV irradiation, a genotoxic agent that generates lesions resolved by TC-NER, depends on active transcription and is signaled mainly by an apoptotic-like pathway. Pre-treated parasites with α-amanitin, a selective RNA polymerase II inhibitor, become resistant to such cell death. Similarly, the gamma pre-irradiated cells are more resistant to UV when the transcription processes are absent. The Cockayne Syndrome B protein (CSB) recognizes blocked RNA polymerase and can initiate TC-NER. Curiously, CSB overexpression increases parasites' cell death shortly after UV exposure. On the other hand, at the same time after irradiation, the single-knockout CSB cells show resistance to the same treatment. UV-induced fast death is signalized by the exposition of phosphatidylserine to the outer layer of the membrane, indicating a cell death mainly by an apoptotic-like pathway. Furthermore, such death is suppressed in WT parasites pre-treated with inhibitors of ataxia telangiectasia and Rad3-related (ATR), a key DDR kinase. Signaling for UV radiation death may be related to R-loops since the overexpression of genes associated with the resolution of these structures suppress it. Together, results suggest that transcription blockage triggered by UV radiation activates an ATR-dependent apoptosis-like mechanism in T. cruzi, with the participation of CSB protein in this process.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , DNA Damage , DNA Repair , R-Loop Structures , Transcription, Genetic , Trypanosoma cruzi , Ultraviolet Rays , Trypanosoma cruzi/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , Protozoan Proteins/metabolism , DNA Helicases/metabolism , DNA Helicases/genetics , Cell Death , Apoptosis , Humans
4.
Clin Exp Med ; 24(1): 187, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39136841

ABSTRACT

Glioblastoma (GBM) is a highly heterogeneous disease with poor clinical outcomes. To comprehensively dissect the molecular landscape of GBM and heterogeneous macrophage clusters in the progression of GBM, this study integrates single-cell and bulk transcriptome data to recognize a distinct pro-tumor macrophage cluster significantly associated with the prognosis of GBM and develop a GBM prognostic signature to facilitate prior subtypes. Leveraging glioma single-cell sequencing data, we identified a novel pro-tumor macrophage subgroup, marked by S100A9, which might interact with endothelial cells to facilitate tumor progression via angiogenesis. To further benefit clinical application, a prognostic signature was established with the genes associated with pro-tumor macrophages. Patients classified within the high-risk group characterized with enrichment in functions related to tumor progression, including epithelial-mesenchymal transition and hypoxia, displays elevated mutations in the TERT promoter region, reduced methylation in the MGMT promoter region, poorer prognoses, and diminished responses to temozolomide therapy, thus effectively discriminating between the prognostic outcomes of GBM patients. Our research sheds light on the intricate microenvironment of gliomas and identifies potential molecular targets for the development of novel therapeutic approaches.


Subject(s)
Gene Expression Profiling , Glioblastoma , Single-Cell Analysis , Humans , Glioblastoma/genetics , Glioblastoma/pathology , Prognosis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Tumor Microenvironment/genetics , Temozolomide/therapeutic use , Macrophages/metabolism , Transcriptome , Telomerase/genetics , Tumor Suppressor Proteins/genetics , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , DNA Methylation , Gene Expression Regulation, Neoplastic , DNA Modification Methylases/genetics , DNA Repair Enzymes
5.
J Am Chem Soc ; 146(32): 22553-22562, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39101269

ABSTRACT

N2-Alkyl-2'-deoxyguanosine (N2-alkyl-dG) is a major type of minor-groove DNA lesions arising from endogenous metabolic processes and exogenous exposure to environmental contaminants. The N2-alkyl-dG lesions, if left unrepaired, can block DNA replication and transcription and induce mutations in these processes. Nevertheless, the repair pathways for N2-alkyl-dG lesions remain incompletely elucidated. By utilizing a photo-cross-linking coupled with mass spectrometry-based quantitative proteomic analysis, we identified a series of candidate N2-alkyl-dG-binding proteins. We found that two of these proteins, i.e., high-mobility group protein B3 (HMGB3) and SUB1, could bind directly to N2-nBu-dG-containing duplex DNA in vitro and promote the repair of this lesion in cultured human cells. In addition, HMGB3 and SUB1 protected cells against benzo[a]pyrene-7,8-diol-9,10-epoxide (BPDE). SUB1 exhibits preferential binding to both the cis and trans diastereomers of N2-BPDE-dG over unmodified dG. On the other hand, HMGB3 binds favorably to trans-N2-BPDE-dG; the protein, however, does not distinguish cis-N2-BPDE-dG from unmodified dG. Consistently, genetic ablation of HMGB3 conferred diminished repair of trans-N2-BPDE-dG, but not its cis counterpart, whereas loss of SUB1 conferred attenuated repair of both diastereomers. Together, we identified proteins involved in the cellular sensing and repair of minor-groove N2-alkyl-dG lesions and documented a unique role of HMGB3 in the stereospecific recognition and repair of N2-BPDE-dG.


Subject(s)
DNA Repair , DNA , HMGB3 Protein , Humans , DNA/chemistry , DNA/metabolism , DNA Damage , DNA Repair Enzymes , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry , Guanine/chemistry , Guanine/metabolism , HMGB3 Protein/metabolism , HMGB3 Protein/chemistry , Protein Binding
7.
Cancer Med ; 13(15): e7456, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39118481

ABSTRACT

OBJECTIVES: High-grade gliomas (HGGs) are highly malignant, aggressive, and have a high incidence and mortality rate. The aim of this study was to investigate survival outcomes and prognostic factors in patients with HGGs. METHODS: In this retrospective study, a total of 159 patients with histologically confirmed HGGs were included. The recruitment period was from January 2011 to December 2019. We evaluated patient demographic data, tumor characteristics, treatment methods, immunocytochemistry results, overall survival (OS) time, and progression-free survival (PFS) time using Kaplan-<>Meier survival analysis with log-rank testing. Additionally, we employed Cox regression analysis to identify independent factors associated with survival outcomes. RESULTS: Kaplan-Meier survival analysis revealed that the 1-, 2-, and 5-years OS rates were 81.8%, 50.3%, and 12.6%, respectively. Similarly, the 1-, 2-, and 5-years PFS rates were 50.9%, 22.4%, and 3.1%, respectively. The median OS duration was 35.0 months. The univariate analysis indicated that postoperative pathological classification, grade, and age were significantly associated with patient outcomes (p < 0.01). Among the patients, 147 received concurrent chemoradiotherapy, while 12 did not. The immunohistochemical markers of ki-67, MGMT, IDH1R132H, and p53 demonstrated statistically significant differences in their prognostic impact (p = 0.001, p = 0.020, p = 0.003, and p = 0.021, respectively). In conclusion, we found that grades, age, pathological classification, ki-67, MGMT, and IDH1R132H expression were statistically significantly associated with PFS (p < 0.01, p = 0.004, p = 0.003, p = 0.001, p = 0.036, and p = 0.028). Additionally, immunohistochemical expressions of TRIB3 and AURKA were significantly higher in patients with shorter survival (p = 0.015 and p = 0.023). CONCLUSIONS: Tumor grade and the use of concurrent chemoradiotherapy after surgery were independent prognostic factors that significantly influenced patient survival. Additionally, tumor grade and MGMT expression were found to be independent factors affecting progression-free survival (PFS). Notably, the expression of TRIB3 and AURKA was higher in patients with poor survival outcomes.


Subject(s)
Brain Neoplasms , Glioma , Neoplasm Grading , Humans , Female , Male , Glioma/mortality , Glioma/pathology , Glioma/therapy , Glioma/metabolism , Retrospective Studies , Middle Aged , Adult , Prognosis , Aged , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Chemoradiotherapy , Young Adult , Kaplan-Meier Estimate , Biomarkers, Tumor/metabolism , Progression-Free Survival , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Multivariate Analysis , Tumor Suppressor Proteins/metabolism , Survival Rate , Adolescent , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/analysis
8.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2136-2149, 2024 Jul 25.
Article in Chinese | MEDLINE | ID: mdl-39044580

ABSTRACT

African swine fever virus (ASFV), as a contagious viral pathogen, is responsible for the occurrence of African swine fever (ASF), a rapidly spreading and highly lethal disease. Since ASFV was introduced into China in 2018, it has been quickly spread to many provinces, which brought great challenges to the pig industry in China. Due to the limited knowledge about the pathogenesis of ASFV, neither vaccines nor antiviral drugs are available. We have found that ASFV infection can induce oxidative stress responses in cells, and DNA repair enzymes play a key role in this process. This study employed RNA interference, RT-qPCR, Western blotting, Hemadsorption (HAD), and flow cytometry to investigate the effects of the inhibitors of DNA repair enzymes OGG1 and MTH1 on ASFV replication and evaluated the anti-ASFV effects of the inhibitors. This study provides reference for the development of anti-viral drugs.


Subject(s)
African Swine Fever Virus , DNA Glycosylases , Phosphoric Monoester Hydrolases , Virus Replication , African Swine Fever Virus/genetics , African Swine Fever Virus/drug effects , Animals , Virus Replication/drug effects , Swine , DNA Glycosylases/metabolism , DNA Glycosylases/genetics , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , African Swine Fever/virology , Antiviral Agents/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Enzyme Inhibitors/pharmacology , Oxidative Stress/drug effects , Vero Cells
9.
Medicine (Baltimore) ; 103(27): e38794, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38968484

ABSTRACT

BACKGROUND: Extracranial metastases occur in <2% of cases of glioblastoma (GBM). When metastases do occur, bone is the most common destination. Herein, we review clinical characteristics of GBM patients with osseous metastases and evaluate both potential risk factors and prognostic significance. METHODS: Using an institutional database, we identified and retrospectively analyzed 6 patients with both GBM and osseous metastases. We collected data on patient demographics, tumor genetics, clinical courses, and outcomes. Given the rarity of metastatic GBM, we conducted historical comparisons using previously published literature. RESULTS: Five patients with osseous metastases (83%) were male, with a median age of 46 years at GBM diagnosis (range: 20-84). All patients had IDH-wildtype, MGMT promoter unmethylated GBM and 5 (83%) had alterations in TP53. All patients underwent surgical resection for GBM followed by radiation with concurrent and adjuvant temozolomide. Four patients (67%) received bevacizumab prior to bone metastasis diagnosis. Bone metastases were discovered at a median of 12.2 months (range: 5.3-35.2) after GBM diagnosis and 4.8 months after starting bevacizumab (range: 3.5-13.2). Three patients (50%) received immunotherapy. After osseous metastasis diagnosis, the median survival was 25 days (range: 13-225). CONCLUSION: In our cohort, most patients were male and young at the time of GBM diagnosis. All patients had IDH-wildtype, MGMT promoter unmethylated GBM, and most had alterations in TP53, which may be important for osseous metastasis. Most patients received bevacizumab, which has been associated with earlier metastasis. Osseous metastases of GBM occur and portend a dismal prognosis in an already aggressive malignancy.


Subject(s)
Bone Neoplasms , Brain Neoplasms , Glioblastoma , Humans , Male , Glioblastoma/genetics , Glioblastoma/secondary , Glioblastoma/pathology , Glioblastoma/therapy , Middle Aged , Female , Adult , Retrospective Studies , Bone Neoplasms/secondary , Bone Neoplasms/genetics , Brain Neoplasms/secondary , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Aged , Aged, 80 and over , Young Adult , Prognosis , Bevacizumab/therapeutic use , Tumor Suppressor Protein p53/genetics , DNA Repair Enzymes/genetics , DNA Modification Methylases , Tumor Suppressor Proteins
10.
BMC Gastroenterol ; 24(1): 239, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075342

ABSTRACT

BACKGROUND: MutT homolog 1 (MTH1) sanitizes oxidized dNTP pools to promote the survival of cancer cells and its expression is frequently upregulated in cancers. Polyubiquitination stabilizes MTH1 to facilitate the proliferation of melanoma cells, suggesting the ubiquitin system controls the stability and function of MTH1. However, whether ubiquitination regulates MTH1 in gastric cancers has not been well defined. This study aims to investigate the interaction between MTH1 and a deubiquitinase, USP9X, in regulating the proliferation, survival, migration, and invasion of gastric cancer cells. METHODS: The interaction between USP9X and MTH1 was evaluated by co-immunoprecipitation (co-IP) in HGC-27 gastric cancer cells. siRNAs were used to interfere with USP9X expression in gastric cancer cell lines HGC-27 and MKN-45. MTT assays were carried out to examine the proliferation, propidium iodide (PI) and 7-AAD staining assays were performed to assess the cell cycle, Annexin V/PI staining assays were conducted to examine the apoptosis, and transwell assays were used to determine the migration and invasion of control, USP9X-deficient, and USP9X-deficient plus MTH1-overexpressing HGC-27 and MKN-45 gastric cancer cells. RESULTS: Co-IP data show that USP9X interacts with and deubiquitinates MTH1. Overexpression of USP9X elevates MTH1 protein level by downregulating its ubiquitination, while knockdown of USP9X has the opposite effect on MTH1. USP9X deficiency in HGC-27 and MKN-45 cells causes decreased proliferation, cell cycle arrest, extra apoptosis, and defective migration and invasion, which could be rescued by excessive MTH1. CONCLUSION: USP9X interacts with and stabilizes MTH1 to promote the proliferation, survival, migration and invasion of gastric cancer cells.


Subject(s)
Cell Movement , Cell Proliferation , DNA Repair Enzymes , Neoplasm Invasiveness , Phosphoric Monoester Hydrolases , Stomach Neoplasms , Ubiquitin Thiolesterase , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Cell Proliferation/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Cell Line, Tumor , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/genetics , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , Ubiquitination , Cell Survival , Apoptosis , RNA, Small Interfering
11.
Biomed Phys Eng Express ; 10(5)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39029475

ABSTRACT

Background.Glioblastoma Multiforme (GBM) is an aggressive form of malignant brain tumor with a generally poor prognosis.O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation has been shown to be a predictive bio-marker for resistance to treatment of GBM, but it is invasive and time-consuming to determine methylation status. There has been effort to predict the MGMT methylation status through analyzing MRI scans using machine learning, which only requires pre-operative scans that are already part of standard-of-care for GBM patients.Purpose.To improve the performance of conventional transfer learning in the identification of MGMT promoter methylation status, we developed a 3D SpotTune network with adaptive fine-tuning capability. Using the pretrained weights of MedicalNet with the SpotTune network, we compared its performance with a randomly initialized network for different combinations of MR modalities.Methods.Using a ResNet50 as the base network, three categories of networks are created: (1) A 3D SpotTune network to process volumetric MR images, (2) a network with randomly initialized weights, and (3) a network pre-trained on MedicalNet. These three networks are trained and evaluated using a public GBM dataset provided by the University of Pennsylvania. The MRI scans from 240 patients are used, with 11 different modalities corresponding to a set of perfusion, diffusion, and structural scans. The performance is evaluated using 5-fold cross validation with a hold-out testing dataset.Results.The SpotTune network showed better performance than the randomly initialized network. The best performing SpotTune model achieved an area under the Receiver Operating Characteristic curve (AUC), average precision of the precision-recall curve (AP), sensitivity, and specificity values of 0.6604, 0.6179, 0.6667, and 0.6061 respectively.Conclusions.SpotTune enables transfer learning to be adaptive to individual patients, resulting in improved performance in predicting MGMT promoter methylation status in GBM using equivalent MRI modalities as compared to a randomly initialized network.


Subject(s)
Brain Neoplasms , DNA Methylation , DNA Modification Methylases , DNA Repair Enzymes , Glioblastoma , Magnetic Resonance Imaging , Promoter Regions, Genetic , Tumor Suppressor Proteins , Humans , Glioblastoma/genetics , Glioblastoma/diagnostic imaging , Magnetic Resonance Imaging/methods , Brain Neoplasms/genetics , Brain Neoplasms/diagnostic imaging , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Machine Learning , ROC Curve , Male , Female , Neural Networks, Computer , Adult , Algorithms
12.
Nat Commun ; 15(1): 6374, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075067

ABSTRACT

Transcription-blocking DNA lesions are specifically targeted by transcription-coupled nucleotide excision repair (TC-NER), which removes a broad spectrum of DNA lesions to preserve transcriptional output and thereby cellular homeostasis to counteract aging. TC-NER is initiated by the stalling of RNA polymerase II at DNA lesions, which triggers the assembly of the TC-NER-specific proteins CSA, CSB and UVSSA. CSA, a WD40-repeat containing protein, is the substrate receptor subunit of a cullin-RING ubiquitin ligase complex composed of DDB1, CUL4A/B and RBX1 (CRL4CSA). Although ubiquitination of several TC-NER proteins by CRL4CSA has been reported, it is still unknown how this complex is regulated. To unravel the dynamic molecular interactions and the regulation of this complex, we apply a single-step protein-complex isolation coupled to mass spectrometry analysis and identified DDA1 as a CSA interacting protein. Cryo-EM analysis shows that DDA1 is an integral component of the CRL4CSA complex. Functional analysis reveals that DDA1 coordinates ubiquitination dynamics during TC-NER and is required for efficient turnover and progression of this process.


Subject(s)
DNA Repair , DNA-Binding Proteins , Transcription, Genetic , Ubiquitination , Humans , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Cullin Proteins/metabolism , Cullin Proteins/genetics , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , Cryoelectron Microscopy , HEK293 Cells , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , DNA Damage , RNA Polymerase II/metabolism , Protein Binding , Excision Repair , Carrier Proteins , DNA Helicases , Transcription Factors , Receptors, Interleukin-17
13.
DNA Repair (Amst) ; 141: 103715, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39029375

ABSTRACT

Glioblastoma (GBM) is a highly aggressive brain tumor associated with poor patient survival. The current standard treatment involves invasive surgery, radiotherapy, and chemotherapy employing temozolomide (TMZ). Resistance to TMZ is, however, a major challenge. Previous work from our group has identified candidate genes linked to TMZ resistance, including genes encoding translesion synthesis (TLS) DNA polymerases iota (PolÉ©) and kappa (Polκ). These specialized enzymes are known for bypassing lesions and tolerating DNA damage. Here, we investigated the roles of PolÉ© and Polκ in TMZ resistance, employing MGMT-deficient U251-MG glioblastoma cells, with knockout of either POLI or POLK genes encoding PolÉ© and Polκ, respectively, and assess their viability and genotoxic stress responses upon subsequent TMZ treatment. Cells lacking either of these polymerases exhibited a significant decrease in viability following TMZ treatment compared to parental counterparts. The restoration of the missing polymerase led to a recovery of cell viability. Furthermore, knockout cells displayed increased cell cycle arrest, mainly in late S-phase, and lower levels of genotoxic stress after TMZ treatment, as assessed by a reduction of γH2AX foci and flow cytometry data. This implies that TMZ treatment does not trigger a significant H2AX phosphorylation response in the absence of these proteins. Interestingly, combining TMZ with Mirin (double-strand break repair pathway inhibitor) further reduced the cell viability and increased DNA damage and γH2AX positive cells in TLS KO cells, but not in parental cells. These findings underscore the crucial roles of PolÉ© and Polκ in conferring TMZ resistance and the potential backup role of homologous recombination in the absence of these TLS polymerases. Targeting these TLS enzymes, along with double-strand break DNA repair inhibition, could, therefore, provide a promising strategy to enhance TMZ's effectiveness in treating GBM.


Subject(s)
DNA Modification Methylases , DNA Polymerase iota , DNA Repair Enzymes , DNA-Directed DNA Polymerase , Drug Resistance, Neoplasm , Glioblastoma , Temozolomide , Temozolomide/pharmacology , Humans , Glioblastoma/genetics , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , Cell Line, Tumor , DNA Modification Methylases/metabolism , DNA Modification Methylases/genetics , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/deficiency , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , DNA Damage , Cell Survival/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , DNA Repair , Gene Knockout Techniques
14.
Drug Resist Updat ; 76: 101122, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39079407

ABSTRACT

O6-methylguanine DNA methyltransferase (MGMT) is a crucial determinant of temozolomide (TMZ) sensitivity in patients with glioblastoma (GBM). The therapeutic potential of small interfering RNA (siRNA) targeting MGMT to enhance TMZ sensitivity has been hampered by serum nuclease degradation, off-target effects, poor accumulation at tumor sites, and low circulation in blood stream. In this study, we developed a framework nucleic acid-based nanoparticles (FNN), which is constructed from a six-helix DNA bundle, to encapsulate and protect siMGMT for improving TMZ sensitivity in GBM treatment. For better blood-brain barrier (BBB) penetration and GBM targeting, we conjugated Angiopep-2 (ANG) targeting modules to each end of the FNN. Nucleolin (NCL)-responsive locks were engineered along the sides of the six-helix DNA bundle, which safeguard siMGMT before tumor entry. Upon interaction with tumor-overexpressed NCL, these locks unlock, exposing siMGMT, this allows for effective suppression of MGMT, resulting in a significant improvement of TMZ therapeutic efficacy in GBM. This innovative strategy has the potential to transform the current treatment landscape for GBM.


Subject(s)
Antineoplastic Agents, Alkylating , Blood-Brain Barrier , Brain Neoplasms , Glioblastoma , Nanoparticles , Temozolomide , Glioblastoma/drug therapy , Glioblastoma/pathology , Temozolomide/pharmacology , Temozolomide/administration & dosage , Temozolomide/therapeutic use , Humans , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Nanoparticles/chemistry , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , RNA-Binding Proteins/metabolism , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , DNA Modification Methylases/metabolism , Nucleolin , Phosphoproteins/metabolism , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , RNA, Small Interfering/administration & dosage , Nucleic Acids , Peptides
15.
Nat Commun ; 15(1): 6031, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39019869

ABSTRACT

Mutations in the Cockayne Syndrome group B (CSB) gene cause cancer in mice, but premature aging and severe neurodevelopmental defects in humans. CSB, a member of the SWI/SNF family of chromatin remodelers, plays diverse roles in regulating gene expression and transcription-coupled nucleotide excision repair (TC-NER); however, these functions do not explain the distinct phenotypic differences observed between CSB-deficient mice and humans. During investigating Cockayne Syndrome-associated genome instability, we uncover an intrinsic mechanism that involves elongating RNA polymerase II (RNAPII) undergoing transient pauses at internal T-runs where CSB is required to propel RNAPII forward. Consequently, CSB deficiency retards RNAPII elongation in these regions, and when coupled with G-rich sequences upstream, exacerbates genome instability by promoting R-loop formation. These R-loop prone motifs are notably abundant in relatively long genes related to neuronal functions in the human genome, but less prevalent in the mouse genome. These findings provide mechanistic insights into differential impacts of CSB deficiency on mice versus humans and suggest that the manifestation of the Cockayne Syndrome phenotype in humans results from the progressive evolution of mammalian genomes.


Subject(s)
Cockayne Syndrome , DNA Helicases , DNA Repair Enzymes , Genomic Instability , Poly-ADP-Ribose Binding Proteins , R-Loop Structures , RNA Polymerase II , Cockayne Syndrome/genetics , Cockayne Syndrome/pathology , Cockayne Syndrome/metabolism , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Animals , Humans , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , Mice , DNA Helicases/metabolism , DNA Helicases/genetics , R-Loop Structures/genetics , DNA Repair , Transcription Elongation, Genetic , Mice, Knockout
16.
Article in Russian | MEDLINE | ID: mdl-39072567

ABSTRACT

OBJECTIVE: Analysis of long-lived patients from the group of patients with glioblastomas after using photodynamic therapy in the structure of their complex treatment in order to assess the influence of various factors on their life expectancy. MATERIAL AND METHODS: In total, a single-center, retrospective categorical study analyzed the long-term results of treatment of 63 patients with glioblastoma in the structure of complex treatment including photodynamic therapy. Clinical factors (age, sex, number of cases, preoperative Karnofsky index, location and size of the tumor, radicality of the operation), histological (nuclear polymorphism, mitosis, vascular proliferation, necrosis), immunohistochemical (Ki-67, p53 index) molecular-genetic factors (expression of VEGF, MGMT, IDH, CD34), amount of radiation and chemotherapy were analyzed. RESULTS: In the entire group of patients, there was a direct correlation of life expectancy with MGMT status, IDH status, the number of courses of chemotherapy, the age of the patient, and the severity of the first surgical intervention. CONCLUSION: Clinical features such as age at diagnosis and extent of surgical resection and amount of chemotherapy have predictive value in assessing their effect on life expectancy. Mutations in IDH and MGMT promoter methylation were the most important molecular factors determining long-term patient survival.


Subject(s)
Brain Neoplasms , Photochemotherapy , Humans , Male , Female , Middle Aged , Photochemotherapy/methods , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Retrospective Studies , Adult , Aged , Glioblastoma/drug therapy , Glioblastoma/mortality , Glioblastoma/genetics , Tumor Suppressor Proteins/genetics , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Isocitrate Dehydrogenase/genetics , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Glioma/drug therapy , Glioma/mortality , Glioma/genetics , Survival Rate , Life Expectancy
17.
Sci Rep ; 14(1): 16031, 2024 07 11.
Article in English | MEDLINE | ID: mdl-38992201

ABSTRACT

O6-methylguanine-DNA methyltransferase (MGMT) has been demonstrated to be an important prognostic and predictive marker in glioblastoma (GBM). To establish a reliable radiomics model based on MRI data to predict the MGMT promoter methylation status of GBM. A total of 183 patients with glioblastoma were included in this retrospective study. The visually accessible Rembrandt images (VASARI) features were extracted for each patient, and a total of 14676 multi-region features were extracted from enhanced, necrotic, "non-enhanced, and edematous" areas on their multiparametric MRI. Twelve individual radiomics models were constructed based on the radiomics features from different subregions and different sequences. Four single-sequence models, three single-region models and the combined radiomics model combining all individual models were constructed. Finally, the predictive performance of adding clinical factors and VASARI characteristics was evaluated. The ComRad model combining all individual radiomics models exhibited the best performance in test set 1 and test set 2, with the area under the receiver operating characteristic curve (AUC) of 0.839 (0.709-0.963) and 0.739 (0.581-0.897), respectively. The results indicated that the radiomics model combining multi-region and multi-parametric MRI features has exhibited promising performance in predicting MGMT methylation status in GBM. The Modeling scheme that combining all individual radiomics models showed best performance among all constructed moels.


Subject(s)
Brain Neoplasms , DNA Methylation , DNA Modification Methylases , DNA Repair Enzymes , Glioblastoma , Tumor Suppressor Proteins , Adult , Aged , Female , Humans , Male , Middle Aged , Brain Neoplasms/genetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Glioblastoma/genetics , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Magnetic Resonance Imaging/methods , Prognosis , Promoter Regions, Genetic , Radiomics , Retrospective Studies , ROC Curve , Tumor Suppressor Proteins/genetics
18.
Cancer Lett ; 598: 217107, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-38992489

ABSTRACT

Glioblastoma (GBM) presents a daunting challenge due to its resistance to temozolomide (TMZ), a hurdle exacerbated by the proneural-to-mesenchymal transition (PMT) from a proneural (PN) to a mesenchymal (MES) phenotype. TAGLN2 is prominently expressed in GBM, particularly in the MES subtype compared to low-grade glioma (LGG) and the PN subtype. Our research reveals TAGLN2's involvement in PMT and TMZ resistance through a series of in vitro and in vivo experiments. TAGLN2 knockdown can restrain proliferation and invasion, trigger DNA damage and apoptosis, and heighten TMZ sensitivity in GBM cells. Conversely, elevating TAGLN2 levels amplifies resistance to TMZ in cellular and intracranial xenograft mouse models. We demonstrate the interaction relationship between TAGLN2 and ERK1/2 through co-immunoprecipitation (Co-IP) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) spectrometry analysis. Knockdown of TAGLN2 results in a decrease in the expression of p-ERK1/2, whereas overexpression of TAGLN2 leads to an increase in p-ERK1/2 expression within the nucleus. Subsequently, the regulatory role of TAGLN2 in the expression and control of MGMT has been demonstrated. Finally, the regulation of TAGLN2 by NF-κB has been validated through chromatin immunoprecipitation and ChIP-PCR assays. In conclusion, our results confirm that TAGLN2 exerts its biological functions by interacting with the ERK/MGMT axis and being regulated by NF-κB, thereby facilitating the acquisition of promoting PMT and increased resistance to TMZ therapy in glioblastoma. These results provide valuable insights for the advancement of targeted therapeutic approaches to overcome TMZ resistance in clinical treatments.


Subject(s)
Antineoplastic Agents, Alkylating , Brain Neoplasms , Drug Resistance, Neoplasm , Glioblastoma , Temozolomide , Temozolomide/pharmacology , Humans , Glioblastoma/pathology , Glioblastoma/genetics , Glioblastoma/drug therapy , Glioblastoma/metabolism , Animals , Cell Line, Tumor , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Mice , Antineoplastic Agents, Alkylating/pharmacology , Cell Proliferation/drug effects , Xenograft Model Antitumor Assays , Apoptosis/drug effects , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Mice, Nude , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA Modification Methylases/metabolism , DNA Modification Methylases/genetics
19.
ESMO Open ; 9(6): 103485, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38833969

ABSTRACT

BACKGROUND: The World Health Organization (WHO) 2021 classification of central nervous system (CNS) tumors classified astrocytoma isocitrate dehydrogenase-mutant (A IDHm) with either microvascular proliferation and/or necrosis or homozygous deletion of CDKN2A/B as CNS grade 4 (CNS WHO G4), introducing a distinct entity and posing new challenges to physicians for appropriate management and prognostication. PATIENTS AND METHODS: We retrospectively collected information about patients diagnosed with A IDHm CNS WHO G4 at three reference neuro-oncological Italian centers and correlated them with survival. RESULTS: A total of 133 patients were included. Patients were young (median age 41 years) and most received post-operative treatment including chemo-radiation (n = 101) and/or temozolomide maintenance (n = 112). With a median follow-up of 51 months, the median overall survival (mOS) was 31.2 months, with a 5-year survival probability of 26%. In the univariate analysis, complete resection (mOS: 40.2 versus 26.3 months, P = 0.03), methyl-guaninemethyltransferase (MGMT) promoter methylation (mOS: 40.7 versus 18 months, P = 0.0136), and absence of telomerase reverse transcriptase (TERT) promoter mutation (mOS: 40.7 versus 18 months, P = 0.0003) correlated with better prognosis. In the multivariate models, lack of TERT promoter mutation [hazard ratio (HR) 0.23, 95% confidence interval (CI) 0.07-0.82, P = 0.024] and MGMT methylation (HR 0.40, 95% CI 0.20-0.81, P = 0.01) remained associated with improved survival. CONCLUSIONS: This is the largest experience in Western countries exploring the prognostic signature of patients with A IDHm CNS G4. Our results show that MGMT promoter methylation and TERT promoter mutation may impact clinical outcomes. This may support physicians in prognostication, clinical management, and design of future studies of this distinct diagnostic entity.


Subject(s)
Astrocytoma , Isocitrate Dehydrogenase , Mutation , Humans , Retrospective Studies , Isocitrate Dehydrogenase/genetics , Astrocytoma/genetics , Astrocytoma/mortality , Astrocytoma/therapy , Male , Female , Adult , Prognosis , Middle Aged , Young Adult , Brain Neoplasms/genetics , DNA Repair Enzymes/genetics , DNA Modification Methylases/genetics , Aged , Telomerase/genetics , Adolescent , Neoplasm Grading , DNA Methylation , Tumor Suppressor Proteins/genetics
20.
Proc Natl Acad Sci U S A ; 121(24): e2404383121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38843184

ABSTRACT

Transcription is extremely important for cellular processes but can be hindered by RNA polymerase II (RNAPII) pausing and stalling. Cockayne syndrome protein B (CSB) promotes the progression of paused RNAPII or initiates transcription-coupled nucleotide excision repair (TC-NER) to remove stalled RNAPII. However, the specific mechanism by which CSB initiates TC-NER upon damage remains unclear. In this study, we identified the indispensable role of the ARK2N-CK2 complex in the CSB-mediated initiation of TC-NER. The ARK2N-CK2 complex is recruited to damage sites through CSB and then phosphorylates CSB. Phosphorylation of CSB enhances its binding to stalled RNAPII, prolonging the association of CSB with chromatin and promoting CSA-mediated ubiquitination of stalled RNAPII. Consistent with this finding, Ark2n-/- mice exhibit a phenotype resembling Cockayne syndrome. These findings shed light on the pivotal role of the ARK2N-CK2 complex in governing the fate of RNAPII through CSB, bridging a critical gap necessary for initiating TC-NER.


Subject(s)
Cockayne Syndrome , DNA Helicases , DNA Repair Enzymes , DNA Repair , Poly-ADP-Ribose Binding Proteins , RNA Polymerase II , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , Humans , Animals , Mice , DNA Helicases/metabolism , DNA Helicases/genetics , Cockayne Syndrome/genetics , Cockayne Syndrome/metabolism , Transcription, Genetic , Phosphorylation , Casein Kinase II/metabolism , Casein Kinase II/genetics , Mice, Knockout , DNA Damage , ATPases Associated with Diverse Cellular Activities/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Chromatin/metabolism , Ubiquitination , Excision Repair
SELECTION OF CITATIONS
SEARCH DETAIL