Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.855
Filter
1.
Int J Mol Sci ; 25(10)2024 May 13.
Article in English | MEDLINE | ID: mdl-38791341

ABSTRACT

It is widely postulated that the majority of pathologically elevated extracellular or cell-free DNA (cfDNA) in cancer originates from tumor cells; however, evidence has emerged regarding the significant contributions of other cells from the tumor microenvironment. Here, the effect of cfDNA originating from murine B16 melanoma cells and L929 fibroblasts on B16 cells was investigated. It was found that cfDNAL929 increased the viability and migration properties of B16 cells in vitro and their invasiveness in vivo. In contrast, cfDNAB16 exhibited a negative effect on B16 cells, reducing their viability and migration in vitro, which in vivo led to decreased tumor size and metastasis number. It was shown that cell treatment with both cfDNAs resulted in an increase in the expression of genes encoding DNases and the oncogenes Braf, Kras, and Myc. cfDNAL929-treated cells were shown to experience oxidative stress. Gene expression changes in the case of cfDNAB16 treatment are well correlated with the observed decrease in proliferation and migration of B16 cells. The obtained data may indicate the possible involvement of fibroblast DNA in the tumor microenvironment in tumor progression and, potentially, in the formation of new tumor foci due to the transformation of normal cells.


Subject(s)
Cell Movement , Cell-Free Nucleic Acids , Fibroblasts , Melanoma, Experimental , Tumor Microenvironment , Animals , Mice , Fibroblasts/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/genetics , Tumor Microenvironment/genetics , Cell-Free Nucleic Acids/genetics , Cell Line, Tumor , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , DNA, Neoplasm/metabolism , DNA, Neoplasm/genetics , Cell Survival/drug effects , Oxidative Stress
2.
Signal Transduct Target Ther ; 7(1): 37, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35136034

ABSTRACT

DNA methylation is a reversible process catalyzed by the ten-eleven translocation (TET) family of enzymes (TET1, TET2, TET3) that convert 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC). Altered patterns of 5hmC and 5mC are widely reported in human cancers and loss of 5hmC correlates with poor prognosis. Understanding the mechanisms leading to 5hmC loss and its role in oncogenesis will advance the development of epigenetic-based therapeutics. We show that TET2 loss associates with glioblastoma (GBM) stem cells and correlates with poor survival of GBM patients. We further identify a SOX2:miR-10b-5p:TET2 axis that represses TET2 expression, represses 5hmC, increases 5mC levels, and induces GBM cell stemness and tumor-propagating potential. In vivo delivery of a miR-10b-5p inhibitor that normalizes TET2 expression and 5hmC levels inhibits tumor growth and prolongs survival of animals bearing pre-established orthotopic GBM xenografts. These findings highlight the importance of TET2 and 5hmC loss in Sox2-driven oncogenesis and their potential for therapeutic targeting.


Subject(s)
Brain Neoplasms/metabolism , DNA Methylation , DNA, Neoplasm/metabolism , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism , Glioblastoma/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cytidine/analogs & derivatives , Cytidine/genetics , Cytidine/metabolism , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Female , Glioblastoma/genetics , Humans , Mice , Mice, Nude , Neoplasm Proteins/genetics , SOXB1 Transcription Factors/genetics
3.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35110400

ABSTRACT

Oncogenic imbalance of DNA methylation is well recognized in cancer development. The ten-eleven translocation (TET) family of dioxygenases, which facilitates DNA demethylation, is frequently dysregulated in cancers. How such dysregulation contributes to tumorigenesis remains poorly understood, especially in solid tumors which present infrequent mutational incidence of TET genes. Here, we identify loss-of-function mutations of TET in 7.4% of human lung adenocarcinoma (LUAD), which frequently co-occur with oncogenic KRAS mutations, and this co-occurrence is predictive of poor survival in LUAD patients. Using an autochthonous mouse model of KrasG12D -driven LUAD, we show that individual or combinational loss of Tet genes markedly promotes tumor development. In this Kras-mutant and Tet-deficient model, the premalignant lung epithelium undergoes neoplastic reprogramming of DNA methylation and transcription, with a particular impact on Wnt signaling. Among the Wnt-associated components that undergo reprogramming, multiple canonical Wnt antagonizing genes present impaired expression arising from elevated DNA methylation, triggering aberrant activation of Wnt signaling. These impairments can be largely reversed upon the restoration of TET activity. Correspondingly, genetic depletion of ß-catenin, the transcriptional effector of Wnt signaling, substantially reverts the malignant progression of Tet-deficient LUAD. These findings reveal TET enzymes as critical epigenetic barriers against lung tumorigenesis and highlight the therapeutic vulnerability of TET-mutant lung cancer through targeting Wnt signaling.


Subject(s)
Adenocarcinoma of Lung/metabolism , DNA Methylation , DNA, Neoplasm/metabolism , Lung Neoplasms/metabolism , Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins/deficiency , Wnt Signaling Pathway , Adenocarcinoma of Lung/genetics , Animals , DNA, Neoplasm/genetics , Humans , Lung Neoplasms/genetics , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Proto-Oncogene Proteins/metabolism
4.
Int J Mol Sci ; 23(3)2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35163579

ABSTRACT

Mitochondrial DNA (mtDNA) has been identified as a significant genetic biomarker in disease, cancer and evolution. Mitochondria function as modulators for regulating cellular metabolism. In the clinic, mtDNA variations (mutations/single nucleotide polymorphisms) and dysregulation of mitochondria-encoded genes are associated with survival outcomes among cancer patients. On the other hand, nuclear-encoded genes have been found to regulate mitochondria-encoded gene expression, in turn regulating mitochondrial homeostasis. These observations suggest that the crosstalk between the nuclear genome and mitochondrial genome is important for cellular function. Therefore, this review summarizes the significant mechanisms and functional roles of mtDNA variations (DNA level) and mtDNA-encoded genes (RNA and protein levels) in cancers and discusses new mechanisms of crosstalk between mtDNA and the nuclear genome.


Subject(s)
DNA, Mitochondrial , DNA, Neoplasm , Mitochondria , Mutation , Neoplasms , Polymorphism, Single Nucleotide , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism
5.
Nat Commun ; 13(1): 208, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35017538

ABSTRACT

Cancer is often called a disease of aging. There are numerous ways in which cancer epidemiology and behaviour change with the age of the patient. The molecular bases for these relationships remain largely underexplored. To characterise them, we analyse age-associations in the nuclear and mitochondrial somatic mutational landscape of 20,033 tumours across 35 tumour-types. Age influences both the number of mutations in a tumour (0.077 mutations per megabase per year) and their evolutionary timing. Specific mutational signatures are associated with age, reflecting differences in exogenous and endogenous oncogenic processes such as a greater influence of tobacco use in the tumours of younger patients, but higher activity of DNA damage repair signatures in those of older patients. We find that known cancer driver genes such as CDKN2A and CREBBP are mutated in age-associated frequencies, and these alter the transcriptome and predict for clinical outcomes. These effects are most striking in brain cancers where alterations like SUFU loss and ATRX mutation are age-dependent prognostic biomarkers. Using three cancer datasets, we show that age shapes the somatic mutational landscape of cancer, with clinical implications.


Subject(s)
Aging/genetics , CREB-Binding Protein/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Repair , DNA, Neoplasm/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Age Factors , Aging/metabolism , CREB-Binding Protein/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA, Neoplasm/metabolism , Datasets as Topic , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mitochondria/metabolism , Mutation Rate , Neoplasm Proteins/metabolism , Neoplasms/classification , Neoplasms/metabolism , Neoplasms/pathology , Repressor Proteins/deficiency , Repressor Proteins/genetics , Smoking/genetics , Smoking/metabolism , Transcriptome , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism
6.
Pharmacogenomics ; 23(2): 119-134, 2022 01.
Article in English | MEDLINE | ID: mdl-35006002

ABSTRACT

Imaging and tissue biopsies represent the current gold standard for breast cancer diagnosis and patient management. However, these practices are time-consuming, expensive and require invasive procedures. Moreover, tissue biopsies do not capture spatial and temporal tumor heterogeneity. Conversely, liquid biopsy, which includes circulating tumor cells, circulating free nucleic acids and extracellular vesicles, is minimally invasive, easy to perform and can be repeated during a patient's follow-up. Increasing evidence also suggests that liquid biopsy can be used to efficiently screen and diagnose tumors at an early stage, and to monitor changes in the tumor molecular profile. In the present review, clinical applications and prospects are discussed.


Subject(s)
Breast Neoplasms/diagnosis , Liquid Biopsy/methods , Biomarkers, Tumor , Breast Neoplasms/pathology , Breast Neoplasms/therapy , DNA, Neoplasm/metabolism , Female , Humans , Neoplastic Cells, Circulating/pathology , Prognosis
7.
Nat Commun ; 13(1): 173, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013311

ABSTRACT

Mechanisms of drug-tolerance remain poorly understood and have been linked to genomic but also to non-genomic processes. 5-fluorouracil (5-FU), the most widely used chemotherapy in oncology is associated with resistance. While prescribed as an inhibitor of DNA replication, 5-FU alters all RNA pathways. Here, we show that 5-FU treatment leads to the production of fluorinated ribosomes exhibiting altered translational activities. 5-FU is incorporated into ribosomal RNAs of mature ribosomes in cancer cell lines, colorectal xenografts, and human tumors. Fluorinated ribosomes appear to be functional, yet, they display a selective translational activity towards mRNAs depending on the nature of their 5'-untranslated region. As a result, we find that sustained translation of IGF-1R mRNA, which encodes one of the most potent cell survival effectors, promotes the survival of 5-FU-treated colorectal cancer cells. Altogether, our results demonstrate that "man-made" fluorinated ribosomes favor the drug-tolerant cellular phenotype by promoting translation of survival genes.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Colorectal Neoplasms/drug therapy , DNA, Neoplasm/genetics , Drug Tolerance/genetics , Fluorouracil/pharmacology , Protein Biosynthesis/drug effects , Receptor, IGF Type 1/genetics , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Replication , DNA, Neoplasm/metabolism , Drug Resistance, Neoplasm/genetics , HCT116 Cells , Halogenation , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Receptor, IGF Type 1/agonists , Receptor, IGF Type 1/metabolism , Ribosomes/drug effects , Ribosomes/genetics , Ribosomes/metabolism , Xenograft Model Antitumor Assays
8.
Biomed Res Int ; 2022: 1987519, 2022.
Article in English | MEDLINE | ID: mdl-35059460

ABSTRACT

Radioresistance of breast cancer is a major reason for therapeutic failure and limits further increases in the dose of radiation due to severe adverse effects. Recently, long noncoding RNAs (lncRNAs) have been shown to regulate cancer proliferation, chemoresistance, and radioresistance. Among these lncRNAs, lncRNA GAS5 expression was shown to be downregulated in breast cancer and related to trastuzumab resistance. However, its role in the radiation response is unclear. In this study, we demonstrated that lncRNA GAS5 expression was reduced in irradiated cells and that overexpression of GAS5 reduced cell viability and promoted cell apoptosis after irradiation. Moreover, overexpression of GAS5 resulted in increased G2/M arrest and unrepaired DNA damage, indicating a radiosensitizing role of GAS5 in breast cancer cells. Finally, we found that a GAS5-interacting miRNA, miR-21, reversed the radiosensitizing effects of GAS5 by inhibiting the apoptotic pathway. In conclusion, we found that lncRNA GAS5 sensitized breast cancer cells to ionizing radiation by inhibiting DNA repair and suppressing miR-21, identifying novel targets for breast cancer radiosensitization.


Subject(s)
Breast Neoplasms/metabolism , DNA Repair , DNA, Neoplasm/metabolism , RNA, Long Noncoding/metabolism , RNA, Neoplasm/metabolism , X-Rays , Breast Neoplasms/genetics , DNA, Neoplasm/genetics , Female , Humans , MCF-7 Cells , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics
9.
Int J Cancer ; 150(4): 603-616, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34648658

ABSTRACT

Biomarkers with relevance for loco-regional therapy are needed in human papillomavirus negative aka HPV(-) head and neck squamous cell carcinoma (HNSCC). Based on the premise that DNA methylation pattern is highly conserved, we sought to develop a reliable and robust methylome-based classifier identifying HPV(-) HNSCC patients at risk for loco-regional recurrence (LR) and all-event progression after postoperative radiochemotherapy (PORT-C). The training cohort consisted of HPV-DNA negative HNSCC patients (n = 128) homogeneously treated with PORT-C in frame of the German Cancer Consortium-Radiation Oncology Group (DKTK-ROG) multicenter biomarker trial. DNA Methylation analysis was performed using Illumina 450 K and 850 K-EPIC microarray technology. The performance of the classifier was integrated with a series of biomarkers studied in the training set namely hypoxia-, 5-microRNA (5-miR), stem-cell gene-expression signatures and immunohistochemistry (IHC)-based immunological characterization of tumors (CD3/CD8/PD-L1/PD1). Validation occurred in an independent cohort of HPV(-) HNSCC patients, pooled from two German centers (n = 125). We identified a 38-methylation probe-based HPV(-) Independent Classifier of disease Recurrence (HICR) with high prognostic value for LR, distant metastasis and overall survival (P < 10-9 ). HICR remained significant after multivariate analysis adjusting for anatomical site, lymph node extracapsular extension (ECE) and size (T-stage). HICR high-risk tumors were enriched for younger patients with hypoxic tumors (15-gene signature) and elevated 5-miR score. After adjustment for hypoxia and 5-miR covariates, HICR maintained predicting all endpoints. HICR provides a novel mean for assessing the risk of LR in HPV(-) HNSCC patients treated with PORT-C and opens a new opportunity for biomarker-assisted stratification and therapy adaptation in these patients.


Subject(s)
Chemoradiotherapy , DNA Methylation , DNA, Neoplasm/metabolism , Head and Neck Neoplasms/genetics , Neoplasm Recurrence, Local/etiology , Squamous Cell Carcinoma of Head and Neck/genetics , Combined Modality Therapy , Female , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/therapy , Head and Neck Neoplasms/virology , Humans , Male , MicroRNAs/analysis , Middle Aged , Papillomaviridae/isolation & purification , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/therapy , Squamous Cell Carcinoma of Head and Neck/virology
10.
Leukemia ; 36(1): 33-41, 2022 01.
Article in English | MEDLINE | ID: mdl-34175901

ABSTRACT

Methotrexate/6-mercaptopurine maintenance therapy improves acute lymphoblastic leukemia (ALL) outcome. Cytotoxicity is mediated by DNA incorporation of thioguanine nucleotides (DNA-TG). We investigated the association of DNA-TG to relapse risk in 1 910 children and young adults with non-high risk ALL. In a cohort-stratified Cox regression analysis adjusted for sex, age, and white cell count at diagnosis, the relapse-specific hazard ratio (HRa) per 100 fmol/µg increase in weighted mean DNA-TG (wmDNA-TG) was 0.87 (95% CI 0.78-0.97; p = 0.013) in the 839 patients who were minimal residual disease (MRD) positive at end of induction therapy (EOI), whereas this was not the case in EOI MRD-negative patients (p = 0.76). Validation analysis excluding the previously published Nordic NOPHO ALL2008 pediatric cohort yielded a HRa of 0.92 (95% CI 0.82-1.03; p = 0.15) per 100 fmol/µg increase in wmDNA-TG in EOI MRD-positive patients. If also excluding the United Kingdom cohort, in which samples were taken non-randomly in selected patients, the HRa for the EOI MRD-positive patients was 0.82 (95% CI 0.68-0.99; p = 0.044) per 100 fmol/µg increase in wmDNA-TG. The importance of DNA-TG as a biomarker for maintenance therapy intensity calls for novel strategies to increase DNA-TG, although its clinical value may vary by protocol backbone.


Subject(s)
Clinical Trials as Topic/statistics & numerical data , DNA, Neoplasm/metabolism , Neoplasm Recurrence, Local/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Thioguanine/metabolism , Adult , Child , Cohort Studies , Female , Humans , Male , Neoplasm Recurrence, Local/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Prognosis , Young Adult
11.
Life Sci ; 289: 120242, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34922939

ABSTRACT

Bulky DNA damage inducing chemotherapeutic cancer drugs such as cisplatin (CIS) and doxorubicin (DOX) are commonly used in the treatment of a variety of cancers. However, they often cause multi-organ toxicity, and the mechanisms underlying are not clear. Using cellular model, the present study showed that persistent endogenous reactive oxygen species (ROS) were stimulated after a single dose short treatment with CIS and DOX. ROS level correlated with the formation of DNA double-strand breaks (DSBs). Knockdown BRCA1, a key player involved in homologous recombination (HR), enhanced ROS accumulation. Whereas knockdown DNA-PKcs and overexpress BRCA1 to inhibit nonhomologous end-joining (NHEJ) repair pathway and restore HR can partially suppress ROS levels. These data indicated that ROS production is associated with DSB formation and repair which is likely a downstream event of DNA repair. Further studies showed that knockdown DNA repair regulators PP2A but not ATM, could partially reduce ROS too. The induction of ROS affected the level of proinflammatory cytokines interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α). Collectively, the present study reveals that DNA repair associated metabolism change and oxidative stress may be a direct cause of the severe side effects associated with genotoxic chemotherapy cancer drugs.


Subject(s)
Antineoplastic Agents , DNA Breaks, Double-Stranded , DNA End-Joining Repair/drug effects , DNA, Neoplasm , Neoplasm Proteins , Neoplasms , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
12.
Mol Cell ; 82(1): 177-189.e4, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34936881

ABSTRACT

The DNA-dependent protein kinase (DNA-PK) initially protects broken DNA ends but then promotes their processing during non-homologous end joining (NHEJ). Before ligation by NHEJ, DNA hairpin ends generated during V(D)J recombination must be opened by the Artemis nuclease, together with autophosphorylated DNA-PK. Structures of DNA-PK bound to DNA before and after phosphorylation, and in complex with Artemis and a DNA hairpin, reveal an essential functional switch. When bound to open DNA ends in its protection mode, DNA-PK is inhibited for cis-autophosphorylation of the so-called ABCDE cluster but activated for phosphorylation of other targets. In contrast, DNA hairpin ends promote cis-autophosphorylation. Phosphorylation of four Thr residues in ABCDE leads to gross structural rearrangement of DNA-PK, widening the DNA binding groove for Artemis recruitment and hairpin cleavage. Meanwhile, Artemis locks DNA-PK into the kinase-inactive state. Kinase activity and autophosphorylation of DNA-PK are regulated by different DNA ends, feeding forward to coordinate NHEJ events.


Subject(s)
DNA Damage , DNA End-Joining Repair , DNA, Neoplasm/metabolism , DNA-Activated Protein Kinase/metabolism , Uterine Cervical Neoplasms/enzymology , DNA, Neoplasm/genetics , DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Enzyme Activation , Female , HEK293 Cells , HeLa Cells , Humans , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Nucleic Acid Conformation , Phosphorylation , Protein Binding , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
13.
J Alzheimers Dis ; 85(2): 573-585, 2022.
Article in English | MEDLINE | ID: mdl-34864677

ABSTRACT

BACKGROUND: 5-Hydroxymethylcytosine (5hmC) is an epigenetic DNA modification that is highly abundant in central nervous system. It has been reported that DNA 5hmC dysregulation play a critical role in Alzheimer's disease (AD) pathology. Changes in 5hmC signatures can be detected in circulating cell-free DNA (cfDNA), which has shown potential as a non-invasive liquid biopsy material. OBJECTIVE: However, the genome-wide profiling of 5hmC in cfDNA and its potential for the diagnosis of AD has not been reported to date. METHODS: We carried out a case-control study and used a genome-wide chemical capture followed by high-throughput sequencing to detect the genome-wide profiles of 5hmC in human cfDNA and identified differentially hydroxymethylated regions (DhMRs) in late-onset AD patients and the control. RESULTS: We discovered significant differences of 5hmC enrichment in gene bodies which were linked to multiple AD pathogenesis-associated signaling pathways in AD patients compared with cognitively normal controls, indicating they can be well distinguished from normal controls by DhMRs in cfDNA. Specially, we identified 7 distinct genes (RABEP1, CPNE4, DNAJC15, REEP3, ROR1, CAMK1D, and RBFOX1) with predicting diagnostic potential based on their significant correlations with MMSE and MoCA scores of subjects. CONCLUSION: The present results suggest that 5hmC markers derived from plasma cfDNA can served as an effective, minimally invasive biomarkers for clinical auxiliary diagnosis of late-onset AD.


Subject(s)
5-Methylcytosine/analogs & derivatives , Alzheimer Disease/diagnosis , Cell-Free Nucleic Acids/metabolism , DNA Methylation , Epigenesis, Genetic , 5-Methylcytosine/metabolism , Aged , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Biomarkers/metabolism , Case-Control Studies , DNA, Neoplasm/metabolism , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged
14.
Cell Rep ; 37(10): 110088, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34879271

ABSTRACT

Alternative lengthening of telomeres (ALT) is a telomere-elongation mechanism observed in ∼15% of cancer subtypes. Current models indicate that ALT is mediated by homology-directed repair mechanisms. By disrupting MSH6 gene expression, we show that the deficiency of MutSα (MSH2/MSH6) DNA mismatch repair complex causes striking telomere hyperextension. Mechanistically, we show MutSα is specifically recruited to telomeres in ALT cells by associating with the proliferating-cell nuclear antigen (PCNA) subunit of the ALT telomere replisome. We also provide evidence that MutSα counteracts Bloom (BLM) helicase, which adopts a crucial role in stabilizing hyper-extended telomeres and maintaining the survival of MutSα-deficient ALT cancer cells. Lastly, we propose a model in which MutSα deficiency impairs heteroduplex rejection, leading to premature initiation of telomere DNA synthesis that coincides with an accumulation of telomere variant repeats (TVRs). These findings provide evidence that the MutSα DNA mismatch repair complex acts to restrain unwarranted ALT.


Subject(s)
DNA, Neoplasm/metabolism , DNA-Binding Proteins/metabolism , MutS Homolog 2 Protein/metabolism , Neoplasms/enzymology , Nucleic Acid Heteroduplexes/metabolism , Telomere Homeostasis , Telomere/metabolism , Cell Line, Tumor , DNA Mismatch Repair , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Genomic Instability , HeLa Cells , Humans , Models, Genetic , MutS Homolog 2 Protein/genetics , Neoplasms/genetics , Neoplasms/pathology , Nucleic Acid Conformation , Nucleic Acid Heteroduplexes/genetics , RecQ Helicases/genetics , RecQ Helicases/metabolism , Telomere/genetics
15.
Cell Rep ; 37(10): 110080, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34879279

ABSTRACT

DNA-protein crosslinks (DPCs) are a specific type of DNA lesion in which proteins are covalently attached to DNA. Unrepaired DPCs lead to genomic instability, cancer, neurodegeneration, and accelerated aging. DPC proteolysis was recently identified as a specialized pathway for DPC repair. The DNA-dependent protease SPRTN and the 26S proteasome emerged as two independent proteolytic systems. DPCs are also repaired by homologous recombination (HR), a canonical DNA repair pathway. While studying the cellular response to DPC formation, we identify ubiquitylation and SUMOylation as two major signaling events in DNA replication-coupled DPC repair. DPC ubiquitylation recruits SPRTN to repair sites, promoting DPC removal. DPC SUMOylation prevents DNA double-strand break formation, HR activation, and potentially deleterious genomic rearrangements. In this way, SUMOylation channels DPC repair toward SPRTN proteolysis, which is a safer pathway choice for DPC repair and prevention of genomic instability.


Subject(s)
DNA Damage , DNA Repair , DNA, Neoplasm/metabolism , DNA-Binding Proteins/metabolism , Genomic Instability , Sumoylation , DNA Breaks, Double-Stranded , DNA Replication , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Female , HEK293 Cells , HeLa Cells , Homologous Recombination , Humans , Male , Proteolysis , Synthetic Lethal Mutations
16.
Molecules ; 26(24)2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34946681

ABSTRACT

Different types of DNA lesions forming in close vicinity, create clusters of damaged sites termed as "clustered/complex DNA damage" and they are considered to be a major challenge for DNA repair mechanisms resulting in significant repair delays and induction of genomic instability. Upon detection of DNA damage, the corresponding DNA damage response and repair (DDR/R) mechanisms are activated. The inability of cells to process clustered DNA lesions efficiently has a great impact on the normal function and survival of cells. If complex lesions are left unrepaired or misrepaired, they can lead to mutations and if persistent, they may lead to apoptotic cell death. In this in silico study, and through rigorous data mining, we have identified human genes that are activated upon complex DNA damage induction like in the case of ionizing radiation (IR) and beyond the standard DNA repair pathways, and are also involved in cancer pathways, by employing stringent bioinformatics and systems biology methodologies. Given that IR can cause repair resistant lesions within a short DNA segment (a few nm), thereby augmenting the hazardous and toxic effects of radiation, we also investigated the possible implication of the most biologically important of those genes in comorbid non-neoplastic diseases through network integration, as well as their potential for predicting survival in cancer patients.


Subject(s)
DNA Damage , DNA Repair , DNA, Neoplasm , Neoplasms , Systems Biology , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/radiotherapy , Radiation, Ionizing
17.
J Clin Invest ; 131(21)2021 11 01.
Article in English | MEDLINE | ID: mdl-34720085

ABSTRACT

Aberrant activation of telomerase in human cancer is achieved by various alterations within the TERT promoter, including cancer-specific DNA hypermethylation of the TERT hypermethylated oncological region (THOR). However, the impact of allele-specific DNA methylation within the TERT promoter on gene transcription remains incompletely understood. Using allele-specific next-generation sequencing, we screened a large cohort of normal and tumor tissues (n = 652) from 10 cancer types and identified that differential allelic methylation (DAM) of THOR is restricted to cancerous tissue and commonly observed in major cancer types. THOR-DAM was more common in adult cancers, which develop through multiple stages over time, than in childhood brain tumors. Furthermore, THOR-DAM was especially enriched in tumors harboring the activating TERT promoter mutations (TPMs). Functional studies revealed that allele-specific gene expression of TERT requires hypomethylation of the core promoter, both in TPM and TERT WT cancers. However, the expressing allele with hypomethylated core TERT promoter universally exhibits hypermethylation of THOR, while the nonexpressing alleles are either hypermethylated or hypomethylated throughout the promoter. Together, our findings suggest a dual role for allele-specific DNA methylation within the TERT promoter in the regulation of TERT expression in cancer.


Subject(s)
DNA Methylation , DNA, Neoplasm/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Neoplasms/metabolism , Promoter Regions, Genetic , Telomerase/biosynthesis , DNA, Neoplasm/genetics , Humans , Neoplasm Proteins/genetics , Neoplasms/genetics , Telomerase/genetics
18.
Sci Rep ; 11(1): 22314, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34785721

ABSTRACT

The comparison of chemical and histopathological data obtained from the analysis of excised tumor fragments oral squamous cell carcinoma (OSCC) with the demographic and clinical evolution data is an effective strategy scarcely explored in OSCC studies. The aim was to analyze OSCC tissues for protein expression of enzymes related to oxidative stress and DNA repair and trace elements as candidates as markers of tumor aggressiveness and prognosis. Tumor fragments from 78 OSCC patients that had undergone ablative surgery were qualitatively analyzed by synchrotron micro-X-ray fluorescence for trace elements. Protein expression of SOD-1, Trx, Ref-1 and OGG1/2 was performed by immunohistochemistry. Sociodemographic, clinical, and histopathological data were obtained from 4-year follow-up records. Disease relapse was highest in patients with the presence of chlorine and chromium and lowest in those with tumors with high OGG1/2 expression. High expression of SOD-1, Trx, and Ref-1 was determinant of the larger tumor. Presence of trace elements can be markers of disease prognosis. High expression of enzymes related to oxidative stress or to DNA repair can be either harmful by stimulating tumor growth or beneficial by diminishing relapse rates. Interference on these players may bring novel strategies for the therapeutic management of OSCC patients.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell , Chlorine/metabolism , Chromium/metabolism , DNA Repair , DNA, Neoplasm/metabolism , Mouth Neoplasms , Neoplasm Proteins/metabolism , Oxidative Stress , Aged , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Mouth Neoplasms/diagnosis , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Prognosis , Retrospective Studies
19.
Int J Mol Sci ; 22(21)2021 Oct 31.
Article in English | MEDLINE | ID: mdl-34769259

ABSTRACT

The gene TP53, which encodes the tumor suppressor protein p53, is mutated in about 50% of cancers. In response to cell stressors like DNA damage and after treatment with DNA-damaging therapeutic agents, p53 acts as a transcription factor to activate subsets of target genes which carry out cell fates such as apoptosis, cell cycle arrest, and DNA repair. Target gene selection by p53 is controlled by a complex regulatory network whose response varies across contexts including treatment type, cell type, and tissue type. The molecular basis of target selection across these contexts is not well understood. Knowledge gained from examining p53 regulatory network profiles across different DNA-damaging agents in different cell types and tissue types may inform logical ways to optimally manipulate the network to encourage p53-mediated tumor suppression and anti-tumor immunity in cancer patients. This may be achieved with combination therapies or with p53-reactivating targeted therapies. Here, we review the basics of the p53 regulatory network in the context of differential responses to DNA-damaging agents; discuss recent efforts to characterize differential p53 responses across treatment types, cell types, and tissue types; and examine the relevance of evaluating these responses in the tumor microenvironment. Finally, we address open questions including the potential relevance of alternative p53 transcriptional functions, p53 transcription-independent functions, and p53-independent functions in the response to DNA-damaging therapeutics.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , DNA Damage , DNA, Neoplasm/metabolism , Neoplasms/drug therapy , Tumor Suppressor Protein p53/metabolism , Apoptosis/genetics , DNA Repair/drug effects , DNA Repair/genetics , DNA, Neoplasm/genetics , Humans , Neoplasms/genetics , Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics
20.
Cell Rep ; 37(5): 109917, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731617

ABSTRACT

Assembly and disassembly of DNA repair protein complexes at DNA damage sites are essential for maintaining genomic integrity. Investigating factors coordinating assembly of the base excision repair (BER) proteins DNA polymerase ß (Polß) and XRCC1 to DNA lesion sites identifies a role for Polß in regulating XRCC1 disassembly from DNA repair complexes and, conversely, demonstrates Polß's dependence on XRCC1 for complex assembly. LivePAR, a genetically encoded probe for live-cell imaging of poly(ADP-ribose) (PAR), reveals that Polß and XRCC1 require PAR for repair-complex assembly, with PARP1 and PARP2 playing unique roles in complex dynamics. Further, BER complex assembly is modulated by attenuation/augmentation of NAD+ biosynthesis. Finally, SIRT6 does not modulate PARP1 or PARP2 activation but does regulate XRCC1 recruitment, leading to diminished Polß abundance at sites of DNA damage. These findings highlight coordinated yet independent roles for PARP1, PARP2, and SIRT6 and their regulation by NAD+ bioavailability to facilitate BER.


Subject(s)
DNA Breaks, Single-Stranded , DNA Repair , DNA, Neoplasm/metabolism , NAD/metabolism , Neoplasms/enzymology , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Sirtuins/metabolism , A549 Cells , DNA Polymerase beta/genetics , DNA Polymerase beta/metabolism , DNA, Neoplasm/genetics , Humans , Kinetics , Microscopy, Confocal , Neoplasms/genetics , Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerases/genetics , Sirtuins/genetics , X-ray Repair Cross Complementing Protein 1/genetics , X-ray Repair Cross Complementing Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...