Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.586
Filtrer
1.
Biol Pharm Bull ; 47(7): 1275-1281, 2024.
Article de Anglais | MEDLINE | ID: mdl-38987176

RÉSUMÉ

The generation of DNA damage causes mutations and consequently cancer. Reactive oxygen species are important sources of DNA damage and some mutation signatures found in human cancers. 8-Oxo-7,8-dihydroguanine (GO, 8-hydroxyguanine) is one of the most abundant oxidized bases and induces a G→T transversion mutation at the modified site. The damaged G base also causes untargeted base substitution mutations at the G bases of 5'-GpA-3' dinucleotides (action-at-a-distance mutations) in human cells, and the cytosine deaminase apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3 (APOBEC3) is involved in the mutation process. The deaminated cytosine, i.e., uracil, bases are expected to be removed by uracil DNA glycosylase. Most of the substitution mutations at the G bases of 5'-GpA-3' might be caused by abasic sites formed by the glycosylase. In this study, we expressed the uracil DNA glycosylase inhibitor from Bacillus subtilis bacteriophage PBS2 in human U2OS cells and examined the effects on the GO-induced action-at-a-distance mutations. The inhibition of uracil DNA glycosylase increased the mutation frequency, and in particular, the frequency of G→A transitions. These results indicated that uracil DNA glycosylase, in addition to APOBEC3, is involved in the untargeted mutation process induced by GO.


Sujet(s)
Guanine , Mutation , Uracil-DNA glycosidase , Humains , Guanine/analogues et dérivés , Guanine/métabolisme , Uracil-DNA glycosidase/métabolisme , Uracil-DNA glycosidase/génétique , Lignée cellulaire tumorale , Altération de l'ADN , Bacillus subtilis/génétique , Bactériophages/génétique
2.
Nat Commun ; 15(1): 4897, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38851742

RÉSUMÉ

DNA base editors enable direct editing of adenine (A), cytosine (C), or guanine (G), but there is no base editor for direct thymine (T) editing currently. Here we develop two deaminase-free glycosylase-based base editors for direct T editing (gTBE) and C editing (gCBE) by fusing Cas9 nickase (nCas9) with engineered human uracil DNA glycosylase (UNG) variants. By several rounds of structure-informed rational mutagenesis on UNG in cultured human cells, we obtain gTBE and gCBE with high activity of T-to-S (i.e., T-to-C or T-to-G) and C-to-G conversions, respectively. Furthermore, we conduct parallel comparison of gTBE/gCBE with those recently developed using other protein engineering strategies, and find gTBE/gCBE show the outperformance. Thus, we provide several base editors, gTBEs and gCBEs, with corresponding engineered UNG variants, broadening the targeting scope of base editors.


Sujet(s)
Protéine-9 associée à CRISPR , Édition de gène , Ingénierie des protéines , Uracil-DNA glycosidase , Humains , Édition de gène/méthodes , Uracil-DNA glycosidase/métabolisme , Uracil-DNA glycosidase/génétique , Ingénierie des protéines/méthodes , Protéine-9 associée à CRISPR/métabolisme , Protéine-9 associée à CRISPR/génétique , Cytosine/métabolisme , Thymine/métabolisme , Systèmes CRISPR-Cas , Cellules HEK293 , Mutagenèse , Guanine/métabolisme , ADN/métabolisme , ADN/génétique
3.
PLoS One ; 19(6): e0305590, 2024.
Article de Anglais | MEDLINE | ID: mdl-38875261

RÉSUMÉ

The analysis of nucleic acids is one of the fundamental parts of modern molecular biology and molecular diagnostics. The information collected predominantly depends on the condition of the genetic material. All potential damage induced by oxidative stress may affect the final results of the analysis of genetic material obtained using commonly used techniques such as polymerase chain reaction or sequencing. The aim of this work was to evaluate the effects of high temperature and pH on DNA structure in the context of the occurrence of oxidative damage, using square-wave voltammetry and two independent research protocols. We resulted in visible oxidation damage registered in acidic conditions after the thermal denaturation process (pH 4.7) with changes in the intensity of guanine and adenine signals. However, using phosphate buffer (pH 7.0) for DNA denaturation negatively affected the DNA structure, but without any oxidized derivatives present. This leads to the conclusion that oxidation occurring in the DNA melting process results in the formation of various derivatives of nucleobases, both electrochemically active and inactive. These derivatives may distort the results of molecular tests due to the possibility of forming complementary bonds with various nucleobases. For example, 8-oxoguanine can form pairs with both cytosine and adenine.


Sujet(s)
ADN , Dénaturation d'acide nucléique , Stress oxydatif , Température , ADN/composition chimique , ADN/métabolisme , Oxydoréduction , Altération de l'ADN , Concentration en ions d'hydrogène , Guanine/composition chimique , Guanine/analogues et dérivés , Guanine/métabolisme , Techniques électrochimiques/méthodes , Adénine/composition chimique
4.
Org Biomol Chem ; 22(27): 5629-5635, 2024 07 10.
Article de Anglais | MEDLINE | ID: mdl-38912549

RÉSUMÉ

8-oxoguanine (o8G), a prevalent oxidative modification in RNA induced by reactive oxygen species (ROS), plays a pivotal role in regulating RNA functions. Accurate detection and quantification of o8G modifications is critical to understanding their biological significance and potential as disease biomarkers, but effective detection methods remain limited. Here, we have developed a highly specific T3 DNA ligase-dependent qPCR assay that exploits the enzyme's ability to discriminate o8G from guanine (G) with single-nucleotide resolution. This method can detect o8G in RNA at levels as low as 500 fM, with an up to 18-fold higher selectivity for discriminating o8G from G. By simulating oxidative stress conditions in SH-SY5Y and HS683 cell lines treated with rotenone, we successfully identified site-specific o8G modifications in key miRNAs associated with neuroprotective responses, including miR-124, let-7a and miR-29a. The developed assay holds significant promise for the practical identification of o8G, facilitating its potential for detailed studies of o8G dynamics in various biological contexts and diseases.


Sujet(s)
Guanine , Guanine/analogues et dérivés , Guanine/composition chimique , Guanine/métabolisme , Humains , ARN/métabolisme , ARN/analyse , microARN/analyse , microARN/métabolisme , DNA ligases/métabolisme , Lignée cellulaire tumorale , Stress oxydatif , Réaction de polymérisation en chaine en temps réel
5.
Mol Biol Rep ; 51(1): 745, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38874758

RÉSUMÉ

BACKGROUND: Sn1-type alkylating agents methylate the oxygen atom on guanine bases thereby producing O6-methylguanine. This modified base could pair with thymine and cytosine, resulting in the formation of O6-methylguanine/thymine mismatch during DNA replication, recognized by the mismatch repair (MMR) complex, which then initiates the DNA damage response and subsequent apoptotic processes. In our investigation of the molecular mechanisms underlying MMR-dependent apoptosis, we observed FANCD2 modification upon the activity of alkylating agent N-methyl-N-nitrosourea (MNU). This observation led us to hypothesize a relevant role for FANCD2 in the apoptosis induction process. METHODS AND RESULTS: We generated FANCD2 knockout cells using the CRISPR/Cas9 method in the human cervical cancer cell line HeLa MR. FANCD2-deficient cells exhibited MNU hypersensitivity. Upon MNU exposure, FANCD2 colocalized with the MMR complex. MNU-treated FANCD2 knockout cells displayed severe S phase delay followed by increased G2/M arrest and MMR-dependent apoptotic cell death. Moreover, FANCD2 knockout cells exhibited impaired CtIP and RAD51 recruitment to the damaged chromatin and DNA double-strand break accumulation, indicated by simultaneously observed increased γH2AX signal and 53BP1 foci. CONCLUSIONS: Our data suggest that FANCD2 is crucial for recruiting homologous recombination factors to the sites of the MMR-dependent replication stress to resolve the arrested replication fork and counteract O6-methylguanine-triggered MMR-dependent apoptosis.


Sujet(s)
Apoptose , Réparation de mésappariement de l'ADN , Protéine du groupe de complémentation D2 de l'anémie de Fanconi , Guanine , Humains , Réparation de mésappariement de l'ADN/génétique , Protéine du groupe de complémentation D2 de l'anémie de Fanconi/métabolisme , Protéine du groupe de complémentation D2 de l'anémie de Fanconi/génétique , Apoptose/génétique , Apoptose/effets des médicaments et des substances chimiques , Guanine/métabolisme , Guanine/analogues et dérivés , Cellules HeLa , Altération de l'ADN , 1-Méthyl-1-nitroso-urée/toxicité , Systèmes CRISPR-Cas , Techniques de knock-out de gènes , Rad51 Recombinase/métabolisme , Rad51 Recombinase/génétique , Réplication de l'ADN/effets des médicaments et des substances chimiques , Réplication de l'ADN/génétique
6.
PLoS One ; 19(5): e0284642, 2024.
Article de Anglais | MEDLINE | ID: mdl-38718041

RÉSUMÉ

The GO DNA repair system protects against GC → TA mutations by finding and removing oxidized guanine. The system is mechanistically well understood but its origins are unknown. We searched metagenomes and abundantly found the genes encoding GO DNA repair at the Lost City Hydrothermal Field (LCHF). We recombinantly expressed the final enzyme in the system to show MutY homologs function to suppress mutations. Microbes at the LCHF thrive without sunlight, fueled by the products of geochemical transformations of seafloor rocks, under conditions believed to resemble a young Earth. High levels of the reductant H2 and low levels of O2 in this environment raise the question, why are resident microbes equipped to repair damage caused by oxidative stress? MutY genes could be assigned to metagenome-assembled genomes (MAGs), and thereby associate GO DNA repair with metabolic pathways that generate reactive oxygen, nitrogen and sulfur species. Our results indicate that cell-based life was under evolutionary pressure to cope with oxidized guanine well before O2 levels rose following the great oxidation event.


Sujet(s)
Réparation de l'ADN , Guanine , Métagénome , Oxydoréduction , Guanine/métabolisme , Cheminées hydrothermales/microbiologie
7.
Circ Res ; 135(1): 76-92, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38747146

RÉSUMÉ

BACKGROUND: Hypoxia and oxidative stress contribute to the development of pulmonary hypertension (PH). tRNA-derived fragments play important roles in RNA interference and cell proliferation, but their epitranscriptional roles in PH development have not been investigated. We aimed to gain insight into the mechanistic contribution of oxidative stress-induced 8-oxoguanine in pulmonary vascular remodeling. METHODS: Through small RNA modification array analysis and quantitative polymerase chain reaction, a significant upregulation of the 8-oxoguanine -modified tRF-1-AspGTC was found in the lung tissues and the serum of patients with PH. RESULTS: This modification occurs at the position 5 of the tRF-1-AspGTC (5o8G tRF). Inhibition of the 5o8G tRF reversed hypoxia-induced proliferation and apoptosis resistance in pulmonary artery smooth muscle cells. Further investigation unveiled that the 5o8G tRF retargeted mRNA of WNT5A (Wingless-type MMTV integration site family, member 5A) and CASP3 (Caspase3) and inhibited their expression. Ultimately, BMPR2 (Bone morphogenetic protein receptor 2) -reactive oxygen species/5o8G tRF/WNT5A signaling pathway exacerbated the progression of PH. CONCLUSIONS: Our study highlights the role of site-specific 8-oxoguanine-modified tRF in promoting the development of PH. Our findings present a promising therapeutic avenue for managing PH and propose 5o8G tRF as a potential innovative marker for diagnosing this disease.


Sujet(s)
Marqueurs biologiques , Récepteurs de la protéine morphogénique osseuse de type II , Hypertension pulmonaire , Artère pulmonaire , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/génétique , Hypertension pulmonaire/étiologie , Humains , Récepteurs de la protéine morphogénique osseuse de type II/métabolisme , Récepteurs de la protéine morphogénique osseuse de type II/génétique , Animaux , Marqueurs biologiques/métabolisme , Marqueurs biologiques/sang , Artère pulmonaire/métabolisme , Protéine Wnt-5a/métabolisme , Protéine Wnt-5a/génétique , Guanine/analogues et dérivés , Guanine/métabolisme , Mâle , Stress oxydatif , Caspase-3/métabolisme , Myocytes du muscle lisse/métabolisme , Prolifération cellulaire , Apoptose , Cellules cultivées , Remodelage vasculaire , Femelle , Rats , Espèces réactives de l'oxygène/métabolisme , Muscles lisses vasculaires/métabolisme
8.
Pharmacol Res ; 204: 107187, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38657843

RÉSUMÉ

Cardiovascular diseases (CVD) persist as a prominent cause of mortality worldwide, with oxidative stress constituting a pivotal contributory element. The oxidative modification of guanosine, specifically 8-oxoguanine, has emerged as a crucial biomarker for oxidative stress, providing novel insights into the molecular underpinnings of CVD. 8-Oxoguanine can be directly generated at the DNA (8-oxo-dG) and RNA (8-oxo-G) levels, as well as at the free nucleotide level (8-oxo-dGTP or 8-oxo-GTP), which are produced and can be integrated through DNA replication or RNA transcription. When exposed to oxidative stress, guanine is more readily produced in RNA than in DNA. A burgeoning body of research surrounds 8-oxoguanine, exhibits its accumulation playing a pivotal role in the development of CVD. Therapeutic approaches targeting oxidative 8-Oxoguanine damage to DNA and RNA, encompassing the modulation of repair enzymes and the development of small molecule inhibitors, are anticipated to enhance CVD management. In conclusion, we explore the noteworthy elevation of 8-oxoguanine levels in patients with various cardiac conditions and deliberate upon the formation and regulation of 8-oxo-dG and 8-oxo-G under oxidative stress, as well as their function in CVD.


Sujet(s)
Maladies cardiovasculaires , ADN , Guanine , Guanosine , Oxydoréduction , Stress oxydatif , ARN , Humains , Maladies cardiovasculaires/métabolisme , Maladies cardiovasculaires/génétique , ARN/métabolisme , ARN/génétique , Guanosine/analogues et dérivés , Guanosine/métabolisme , ADN/métabolisme , Animaux , Guanine/analogues et dérivés , Guanine/métabolisme , Altération de l'ADN
9.
RNA ; 30(7): 901-919, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38670632

RÉSUMÉ

A key to understanding the roles of RNA in regulating gene expression is knowing their structures in vivo. One way to obtain this information is through probing the structures of RNA with chemicals. To probe RNA structure directly in cells, membrane-permeable reagents that modify the Watson-Crick (WC) face of unpaired nucleotides can be used. Although dimethyl sulfate (DMS) has led to substantial insight into RNA structure, it has limited nucleotide specificity in vivo, with WC face reactivity only at adenine (A) and cytosine (C) at neutral pH. The reagent 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) was recently shown to modify the WC face of guanine (G) and uracil (U). Although useful at lower concentrations in experiments that measure chemical modifications by reverse transcription (RT) stops, at higher concentrations necessary for detection by mutational profiling (MaP), EDC treatment leads to degradation of RNA. Here, we demonstrate EDC-stimulated degradation of RNA in Gram-negative and Gram-positive bacteria. In an attempt to overcome these limitations, we developed a new carbodiimide reagent, 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide methiodide (ETC), which we show specifically modifies unpaired Gs and Us in vivo without substantial degradation of RNA. We establish ETC as a probe for MaP and optimize the RT conditions and computational analysis in Escherichia coli Importantly, we demonstrate the utility of ETC as a probe for improving RNA structure prediction both alone and with DMS.


Sujet(s)
Guanine , Conformation d'acide nucléique , Sulfates organiques , Uracile , Sulfates organiques/composition chimique , Uracile/composition chimique , Uracile/analogues et dérivés , Uracile/métabolisme , Guanine/composition chimique , Guanine/métabolisme , ARN/composition chimique , ARN/génétique , Escherichia coli/génétique , Escherichia coli/effets des médicaments et des substances chimiques , Carbodiimides/composition chimique , ARN bactérien/composition chimique , ARN bactérien/génétique , Stabilité de l'ARN , Indicateurs et réactifs/composition chimique
10.
Arch Toxicol ; 98(4): 1081-1093, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38436695

RÉSUMÉ

Large interspecies differences between rats and mice concerning the hepatotoxicity and carcinogenicity of aflatoxin B1 (AFB1) are known, with mice being more resistant. However, a comprehensive interspecies comparison including subcellular liver tissue compartments has not yet been performed. In this study, we performed spatio-temporal intravital analysis of AFB1 kinetics in the livers of anesthetized mice and rats. This was supported by time-dependent analysis of the parent compound as well as metabolites and adducts in blood, urine, and bile of both species by HPLC-MS/MS. The integrated data from intravital imaging and HPLC-MS/MS analysis revealed major interspecies differences between rats and mice: (1) AFB1-associated fluorescence persisted much longer in the nuclei of rat than mouse hepatocytes; (2) in the sinusoidal blood, AFB1-associated fluorescence was rapidly cleared in mice, while a time-dependent increase was observed in rats in the first three hours after injection followed by a plateau that lasted until the end of the observation period of six hours; (3) this coincided with a far stronger increase of AFB1-lysine adducts in the blood of rats compared to mice; (4) the AFB1-guanine adduct was detected at much higher concentrations in bile and urine of rats than mice. In both species, the AFB1-glutathione conjugate was efficiently excreted via bile, where it reached concentrations at least three orders of magnitude higher compared to blood. In conclusion, major differences between mice and rats were observed, concerning the nuclear persistence, formation of AFB1-lysine adducts, and the AFB1-guanine adducts.


Sujet(s)
Aflatoxines , Rats , Souris , Animaux , Aflatoxines/métabolisme , Aflatoxines/toxicité , Lysine/métabolisme , , Spectrométrie de masse en tandem , Foie/métabolisme , Aflatoxine B1/toxicité , Guanine/métabolisme , Microscopie intravitale
11.
Mol Biochem Parasitol ; 258: 111616, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38401850

RÉSUMÉ

Trypanosoma cruzi is a protozoan parasite and the etiological agent of Chagas disease, a debilitating and sometimes fatal disease that continues to spread to new areas. Yet, Chagas disease is still only treated with two related nitro compounds that are insufficiently effective and cause severe side effects. Nucleotide metabolism is one of the known vulnerabilities of T. cruzi, as they are auxotrophic for purines, and nucleoside analogues have been shown to have genuine promise against this parasite in vitro and in vivo. Since purine antimetabolites require efficient uptake through transporters, we here report a detailed characterisation of the T. cruzi NB1 nucleobase transporter with the aim of elucidating the interactions between TcrNB1 and its substrates and finding the positions that can be altered in the design of novel antimetabolites without losing transportability. Systematically determining the inhibition constants (Ki) of purine analogues for TcrNB1 yielded their Gibbs free energy of interaction, ΔG0. Pairwise comparisons of substrate (hypoxanthine, guanine, adenine) and analogues allowed us to determine that optimal binding affinity by TcrNB1 requires interactions with all four nitrogen residues of the purine ring, with N1 and N9, in protonation state, functioning as presumed hydrogen bond donors and unprotonated N3 and N7 as hydrogen bond acceptors. This is the same interaction pattern as we previously described for the main nucleobase transporters of Trypanosoma brucei spp. and Leishmania major and makes it the first of the ENT-family genes that is functionally as well as genetically conserved between the three main kinetoplast pathogens.


Sujet(s)
Guanine , Hypoxanthine , Trypanosoma cruzi , Trypanosoma cruzi/métabolisme , Trypanosoma cruzi/génétique , Trypanosoma cruzi/composition chimique , Guanine/métabolisme , Hypoxanthine/métabolisme , Protéines de protozoaire/métabolisme , Protéines de protozoaire/génétique , Protéines de protozoaire/composition chimique , Transporteurs de nucléobases/métabolisme , Transporteurs de nucléobases/génétique , Transporteurs de nucléobases/composition chimique , Transport biologique , Spécificité du substrat , Liaison aux protéines , Nucléosides/métabolisme
12.
Sci Rep ; 14(1): 4509, 2024 02 24.
Article de Anglais | MEDLINE | ID: mdl-38402266

RÉSUMÉ

The 5'-mRNA-cap formation is a conserved process in protection of mRNA in eukaryotic cells, resulting in mRNA stability and efficient translation. In humans, two methyltransferases, RNA cap guanine-N7 methyltransferase (hRNMT) and cap-specific nucleoside-2'-O-methyltransferase 1 (hCMTr1) methylate the mRNA resulting in cap0 (N7mGpppN-RNA) and cap1 (N7mGpppN2'-Om-RNA) formation, respectively. Coronaviruses mimic this process by capping their RNA to evade human immune systems. The coronaviral nonstructural proteins, nsp14 and nsp10-nsp16, catalyze the same reactions as hRNMT and hCMTr1, respectively. These two viral enzymes are important targets for development of inhibitor-based antiviral therapeutics. However, assessing the selectivity of such inhibitors against human corresponding proteins is crucial. Human RNMTs have been implicated in proliferation of cancer cells and are also potential targets for development of anticancer therapeutics. Here, we report the development and optimization of a radiometric assay for hRNMT, full kinetic characterization of its activity, and optimization of the assay for high-throughput screening with a Z-factor of 0.79. This enables selectivity determination for a large number of hits from various screening of coronaviral methyltransferases, and also screening hRNMT for discovery of inhibitors and chemical probes that potentially could be used to further investigate the roles RNMTs play in cancers.


Sujet(s)
Infections à coronavirus , Coronavirus , Humains , Coronavirus/génétique , Guanine/métabolisme , Methyltransferases/métabolisme , Coiffes des ARN/métabolisme , ARN messager/génétique , ARN viral/génétique , Protéines virales non structurales/génétique
13.
DNA Repair (Amst) ; 134: 103625, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38237481

RÉSUMÉ

By observing one molecule at a time, single-molecule studies can offer detailed insights about biomolecular processes including on rates, off rates, and diffusivity of molecules on strands of DNA. A recent technological advance (Single-molecule Analysis of DNA-binding proteins from Nuclear Extracts, SMADNE) has lowered the barrier to entry for single-molecule studies, and single-molecule dynamics can now be determined directly out of nuclear extracts, providing information in an intermediate environment between purified proteins in isolation and the heterogeneity of a nucleus. To compare and contrast the single-molecule DNA binding dynamics in nuclear extracts versus purified proteins, combined optical tweezers and fluorescence microscopy experiments were performed with purified GFP-tagged 8-oxoguanine glycosylase 1 (OGG1), purified GFP-OGG1 spiked into nuclear extracts, and nuclear extracts from human cells overexpressing GFP-OGG1. We observed differences in undamaged DNA binding during DNA damage search in each of the three conditions. Purified GFP-OGG1 engaged undamaged DNA for a weighted average lifetime of 5.7 s and 21% of these events underwent DNA diffusion after binding. However, unlike other glycosylases studied by SMADNE, OGG1 does not bind non-damaged DNA efficiently in nuclear extracts. In contrast, GFP-OGG1 binding dynamics on DNA substrates containing oxidative damage were relatively similar in all three conditions, with the weighted average binding lifetimes varying from 2.2 s in nuclear extracts to 7.8 s with purified GFP-OGG1 in isolation. Finally, we compared the purified protein and nuclear extract approaches for a catalytically dead OGG1 variant (GFP-OGG1-K249Q). This variant greatly increased the binding lifetime for oxidative DNA damage, with the weighted average lifetime for GFP-OGG1-249Q in nuclear extracts at 15.4 s vs 10.7 s for the purified protein. SMADNE will provide a new window of observation into the behavior of nucleic acid binding proteins only accessible by biophysicists trained in protein purification and protein labeling.


Sujet(s)
DNA Glycosylases , Réparation de l'ADN , Guanine , Humains , ADN , Altération de l'ADN , DNA Glycosylases/métabolisme , Guanine/analogues et dérivés , Guanine/métabolisme
14.
Int J Biol Macromol ; 261(Pt 2): 129629, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38266843

RÉSUMÉ

The existing DNA damage detection technology cannot meet the current detection requirements. It is critical to build new methods and discover novel biomarkers. In this study, alkaline comet and 8-OHDG ELISA assays were used to identify DNA damage in HT-1080 cells exposed to K2Cr2O7, and electrochemical behaviors of HT-1080 cells with DNA damage was studied. With an increase in K2Cr2O7 exposure time, two electrochemical signals from HT-1080 cells at 0.69 and 1.01 V steadily grew before decreasing after reaching their highest values. The electrochemical signal's initial response time and peak time decreased as the concentration of K2Cr2O7 increased. The duration of the high dose group was 0.5 and 1 h, while the low dose group was 1.5 and 6 h. Western blotting analysis revealed that DNA damage increased the expression of proteins involved in catabolism and de novo purine synthesis, particularly de novo purine synthesis. Expressions of PRPP amidotransferase, IMPDH, and ADA were all higher than those of ADSS, XOD, and GDA, which resulted in larger concentrations of hypoxanthine, guanine, and xanthine, and in turn improved electrochemical signaling. These findings suggest that intracellular purine identified by linear scan voltammetry is predicted to evolve as a marker of early DNA damage.


Sujet(s)
Guanine , Purines , Purines/métabolisme , Hypoxanthine , Guanine/métabolisme , Xanthine/métabolisme , Altération de l'ADN
15.
Mutagenesis ; 39(1): 24-31, 2024 Feb 08.
Article de Anglais | MEDLINE | ID: mdl-37471265

RÉSUMÉ

DNA oxidation is a serious threat to genome integrity and is involved in mutations and cancer initiation. The G base is most frequently damaged, and 8-oxo-7,8-dihydroguanine (GO, 8-hydroxyguanine) is one of the predominant damaged bases. In human cells, GO causes a G:C→T:A transversion mutation at the modified site, and also induces untargeted substitution mutations at the G bases of 5'-GpA-3' dinucleotides (action-at-a-distance mutations). The 5'-GpA-3' sequences are complementary to the 5'-TpC-3' sequences, the preferred substrates for apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3 (APOBEC3) cytosine deaminases, and thus their contribution to mutagenesis has been considered. In this study, APOBEC3B, the most abundant APOBEC3 protein in human U2OS cells, was knocked down in human U2OS cells, and a GO-shuttle plasmid was then transfected into the cells. The action-at-a-distance mutations were reduced to ~25% by the knockdown, indicating that GO-induced action-at-a-distance mutations are highly dependent on APOBEC3B in this cell line.


Sujet(s)
ADN , Guanine , Guanine/analogues et dérivés , Humains , Mutation , Mutagenèse , Guanine/métabolisme , Cytidine deaminase/génétique , Antigènes mineurs d'histocompatibilité/génétique
16.
Free Radic Biol Med ; 210: 65-74, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37977212

RÉSUMÉ

Exercise-induced adaptation is achieved by altering the epigenetic landscape of the entire genome leading to the expression of genes involved in various processes including regulatory, metabolic, adaptive, immune, and myogenic functions. Clinical and experimental data suggest that the methylation pattern/levels of promoter/enhancer is not linearly correlated with gene expression and proteome levels during physical activity implying a level of complexity and interplay with other regulatory modulators. It has been shown that a higher level of physical fitness is associated with a slower DNA methylation-based aging clock. There is strong evidence supporting exercise-induced ROS being a key regulatory mediator through overlapping events, both as signaling entities and through oxidative modifications to various protein mediators and DNA molecules. ROS generated by physical activity shapes epigenome both directly and indirectly, a complexity we are beginning to unravel within the epigenetic arrangement. Oxidative modification of guanine to 8-oxoguanine is a non-genotoxic alteration, does not distort DNA helix and serves as an epigenetic-like mark. The reader and eraser of oxidized guanine is the 8-oxoguanine DNA glycosylase 1, contributing to changes in gene expression. In fact, it can modulate methylation patterns of promoters/enhancers consequently leading to multiple phenotypic changes. Here, we provide evidence and discuss the potential roles of exercise-induced ROS in altering cytosine methylation patterns during muscle adaptation processes.


Sujet(s)
Méthylation de l'ADN , Épigenèse génétique , Espèces réactives de l'oxygène/métabolisme , Exercice physique , ADN/métabolisme , Guanine/métabolisme
17.
Biochimie ; 220: 39-47, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38128776

RÉSUMÉ

Many prokaryotic Argonaute (pAgo) proteins act as programmable nucleases that use small guide DNAs for recognition and cleavage of complementary target DNA. Recent studies suggested that pAgos participate in cell defense against invader DNA and may also be involved in other genetic processes, including DNA replication and repair. The ability of pAgos to recognize specific targets potentially make them an invaluable tool for DNA manipulations. Here, we demonstrate that DNA-guided DNA-targeting pAgo nucleases from three bacterial species, DloAgo from Dorea longicatena, CbAgo from Clostridium butyricum and KmAgo from Kurthia massiliensis, can sense site-specific modifications in the target DNA, including 8-oxoguanine, thymine glycol, ethenoadenine and pyrimidine dimers. The effects of DNA modifications on the activity of pAgos strongly depend on their positions relative to the site of cleavage and are comparable to or exceed the effects of guide-target mismatches at corresponding positions. For all tested pAgos, the strongest effects are observed when DNA lesions are located at the cleavage position. The results demonstrate that DNA cleavage by pAgos is strongly affected by DNA modifications, thus making possible their use as sensors of DNA damage.


Sujet(s)
Protéines Argonaute , Protéines bactériennes , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/composition chimique , Protéines Argonaute/métabolisme , Protéines Argonaute/génétique , ADN/métabolisme , Altération de l'ADN , Guanine/métabolisme , Guanine/composition chimique , Guanine/analogues et dérivés , Clostridium butyricum/métabolisme , Clostridium butyricum/génétique , Thymine/métabolisme , Thymine/composition chimique , Thymine/analogues et dérivés
18.
Bioelectrochemistry ; 156: 108634, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38160510

RÉSUMÉ

The estrogenic effect of plant growth regulators has been received little attention, which leads to the lack of relevant toxicity data. In this study, the estrogenic effect induced by gibberellin with ERα-dependent manner was found by E-screen and western blot methods, and the electrochemical signals of MCF-7 cells regulated by gibberellin and fulvestrant were investigated. The results showed that the electrochemical signals of MCF-7 cells were increased by gibberellin, while reduced by fulvestrant significantly, and displayed an extremely sensitive response to the effects of estrogenic effect induced by ERα agonist and antagonist. Western blot results showed that the expressions of phosphoribosyl pyrophosphate amidotransferase and hypoxanthine nucleotide dehydrogenase in de novo purine synthesis and adenine deaminase in catabolism were more effective regulated by gibberellin and fulvestrant, resulting in significant changes of the levels of guanine, hypoxanthine and xanthine in cells, and then electrochemical signals. The results provide a theoretical basis for the establishment of new electrochemical detection method of the estrogenic effect of plant regulators.


Sujet(s)
Récepteur alpha des oestrogènes , Gibbérellines , Fulvestrant , Gibbérellines/pharmacologie , Oestrogènes , Électrochimie , Purines/pharmacologie , Purines/métabolisme , Guanine/métabolisme
19.
J Biol Phys ; 50(1): 71-87, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38150168

RÉSUMÉ

Methyl damage to DNA bases is common in the cell nucleus. O6-alkylguanine-DNA alkyl transferase (AGT) may be a promising candidate for direct damage reversal in methylated DNA (mDNA) at the O6 point of the guanine. Indeed, atomic-level investigations in the contact region of AGT-DNA complex can provide an in-depth understanding of their binding mechanism, allowing to evaluate the silico-drug nature of AGT and its utility in removing methyl damage in DNA. In this study, molecular dynamics (MD) simulation was utilized to examine the flipping of methylated nucleotide, the binding mechanism between mDNA and AGT, and the comparison of binding strength prior and post methyl transfer to AGT. The study reveals that methylation at the O6 atom of guanine weakens the hydrogen bond (H-bond) between guanine and cytosine, permitting for the flipping of such nucleotide. The formation of a H-bond between the base pair of methylated nucleotide (i.e., cytosine) and the intercalated arginine of AGT also forces the nucleotide to rotate. Following that, electrostatics and van der Waals contacts as well as hydrogen bonding contribute to form the complex of DNA and protein. The stronger binding of AGT with DNA before methyl transfer creates the suitable condition to transfer methyl adduct from DNA to AGT.


Sujet(s)
Réparation de l'ADN , O(6)-methylguanine-DNA methyltransferase , O(6)-methylguanine-DNA methyltransferase/composition chimique , O(6)-methylguanine-DNA methyltransferase/génétique , O(6)-methylguanine-DNA methyltransferase/métabolisme , Nucléotides/composition chimique , ADN/composition chimique , Guanine/composition chimique , Guanine/métabolisme , Cytosine
20.
DNA Repair (Amst) ; 129: 103550, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37542751

RÉSUMÉ

OGG1 is the DNA glycosylase responsible for the removal of the oxidative lesion 8-oxoguanine (8-oxoG) from DNA. The recognition of this lesion by OGG1 is a complex process that involves scanning the DNA for the presence of 8-oxoG, followed by recognition and lesion removal. Structural data have shown that OGG1 evolves through different stages of conformation onto the DNA, corresponding to elementary steps of the 8-oxoG recognition and extrusion from the double helix. Single-molecule studies of OGG1 on naked DNA have shown that OGG1 slides in persistent contact with the DNA, displaying different binding states probably corresponding to the different conformation stages. However, in cells, the DNA is not naked and OGG1 has to navigate into a complex and highly crowded environment within the nucleus. To ensure rapid detection of 8-oxoG, OGG1 alternates between 3D diffusion and sliding along the DNA. This process is regulated by the local chromatin state but also by protein co-factors that could facilitate the detection of oxidized lesions. We will review here the different methods that have been used over the last years to better understand how OGG1 detects and process 8-oxoG lesions.


Sujet(s)
DNA Glycosylases , DNA Glycosylases/métabolisme , Réparation de l'ADN , Guanine/métabolisme , ADN/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...