scholarly journals Identification of New Markers of Alcohol-Derived DNA Damage in Humans

Biomolecules ◽  
2021 ◽  
Vol 11 (3) ◽  
pp. 366
Author(s):  
Valeria Guidolin ◽  
Erik S. Carlson ◽  
Andrea Carrà ◽  
Peter W. Villalta ◽  
Laura A. Maertens ◽  
...  

Alcohol consumption is a risk factor for the development of several cancers, including those of the head and neck and the esophagus. The underlying mechanisms of alcohol-induced carcinogenesis remain unclear; however, at these sites, alcohol-derived acetaldehyde seems to play a major role. By reacting with DNA, acetaldehyde generates covalent modifications (adducts) that can lead to mutations. Previous studies have shown a dose dependence between levels of a major acetaldehyde-derived DNA adduct and alcohol exposure in oral-cell DNA. The goal of this study was to optimize a mass spectrometry (MS)-based DNA adductomic approach to screen for all acetaldehyde-derived DNA adducts to more comprehensively characterize the genotoxic effects of acetaldehyde in humans. A high-resolution/-accurate-mass data-dependent constant-neutral-loss-MS3 methodology was developed to profile acetaldehyde-DNA adducts in purified DNA. This resulted in the identification of 22 DNA adducts. In addition to the expected N2-ethyldeoxyguanosine (after NaBH3CN reduction), two previously unreported adducts showed prominent signals in the mass spectra. MSn fragmentation spectra and accurate mass were used to hypothesize the structure of the two new adducts, which were then identified as N6-ethyldeoxyadenosine and N4-ethyldeoxycytidine by comparison with synthesized standards. These adducts were quantified in DNA isolated from oral cells collected from volunteers exposed to alcohol, revealing a significant increase after the exposure. In addition, 17 of the adducts identified in vitro were detected in these samples confirming our ability to more comprehensively characterize the DNA damage deriving from alcohol exposures.

Author(s):  
Sarah McCarrick ◽  
Valentin Romanovski ◽  
Zheng Wei ◽  
Elin M. Westin ◽  
Kjell-Arne Persson ◽  
...  

AbstractWelders are daily exposed to various levels of welding fumes containing several metals. This exposure can lead to an increased risk for different health effects which serves as a driving force to develop new methods that generate less toxic fumes. The aim of this study was to explore the role of released metals for welding particle-induced toxicity and to test the hypothesis that a reduction of Cr(VI) in welding fumes results in less toxicity by comparing the welding fume particles of optimized Cr(VI)-reduced flux-cored wires (FCWs) to standard FCWs. The welding particles were thoroughly characterized, and toxicity (cell viability, DNA damage and inflammation) was assessed following exposure to welding particles as well as their released metal fraction using cultured human bronchial epithelial cells (HBEC-3kt, 5–100 µg/mL) and human monocyte-derived macrophages (THP-1, 10–50 µg/mL). The results showed that all Cr was released as Cr(VI) for welding particles generated using standard FCWs whereas only minor levels (< 3% of total Cr) were released from the newly developed FCWs. Furthermore, the new FCWs were considerably less cytotoxic and did not cause any DNA damage in the doses tested. For the standard FCWs, the Cr(VI) released in cell media seemed to explain a large part of the cytotoxicity and DNA damage. In contrast, all particles caused rather similar inflammatory effects suggesting different underlying mechanisms. Taken together, this study suggests a potential benefit of substituting standard FCWs with Cr(VI)-reduced wires to achieve less toxic welding fumes and thus reduced risks for welders.


2012 ◽  
Vol 13 (3) ◽  
pp. 2846-2862 ◽  
Author(s):  
Samuel Saito ◽  
Givaldo Silva ◽  
Regineide Xavier Santos ◽  
Grace Gosmann ◽  
Cristina Pungartnik ◽  
...  

Blood ◽  
2007 ◽  
Vol 109 (11) ◽  
pp. 5016-5026 ◽  
Author(s):  
Tsukasa Oda ◽  
Toshiya Hayano ◽  
Hidenobu Miyaso ◽  
Nobuhiro Takahashi ◽  
Takayuki Yamashita

Abstract Heat shock protein 90 (Hsp90) regulates diverse signaling pathways. Emerging evidence suggests that Hsp90 inhibitors, such as 17-allylamino-17-demethoxygeldanamycin (17-AAG), enhance DNA damage-induced cell death, suggesting that Hsp90 may regulate cellular responses to genotoxic stress. However, the underlying mechanisms are poorly understood. Here, we show that the Fanconi anemia (FA) pathway is involved in the Hsp90-mediated regulation of genotoxic stress response. In the FA pathway, assembly of 8 FA proteins including FANCA into a nuclear multiprotein complex, and the complex-dependent activation of FANCD2 are critical events for cellular tolerance against DNA cross-linkers. Hsp90 associates with FANCA, in vivo and in vitro, in a 17-AAG–sensitive manner. Disruption of the FANCA/Hsp90 association by cellular treatment with 17-AAG induces rapid proteasomal degradation and cytoplasmic relocalization of FANCA, leading to impaired activation of FANCD2. Furthermore, 17-AAG promotes DNA cross-linker–induced cytotoxicity, but this effect is much less pronounced in FA pathway-defective cells. Notably, 17-AAG enhances DNA cross-linker–induced chromosome aberrations. In conclusion, our results identify FANCA as a novel client of Hsp90, suggesting that Hsp90 promotes activation of the FA pathway through regulation of intracellular turnover and trafficking of FANCA, which is critical for cellular tolerance against genotoxic stress.


2005 ◽  
Vol 25 (9) ◽  
pp. 3596-3607 ◽  
Author(s):  
Elizabeth Peterson-Roth ◽  
Mindy Reynolds ◽  
George Quievryn ◽  
Anatoly Zhitkovich

ABSTRACT Chromium(VI) is a toxic and carcinogenic metal that causes the formation of DNA phosphate-based adducts. Cr-DNA adducts are genotoxic in human cells, although they do not block replication in vitro. Here, we report that induction of cytotoxicity in Cr(VI)-treated human colon cells and mouse embryonic fibroblasts requires the presence of all major mismatch repair (MMR) proteins. Cr-DNA adducts lost their ability to block replication of Cr-modified plasmids in human colon cells lacking MLH1 protein. The presence of functional mismatch repair caused induction of p53-independent apoptosis associated with activation of caspases 2 and 7. Processing of Cr-DNA damage by mismatch repair resulted in the extensive formation of γ-H2AX foci in G2 phase, indicating generation of double-stranded breaks as secondary toxic lesions. Induction of γ-H2AX foci was observed at 6 to 12 h postexposure, which was followed by activation of apoptosis in the absence of significant G2 arrest. Our results demonstrate that mismatch repair system triggers toxic responses to Cr-DNA backbone modifications through stress mechanisms that are significantly different from those for other forms of DNA damage. Selection for Cr(VI) resistant, MMR-deficient cells may explain the very high frequency of lung cancers with microsatellite instability among chromate workers.


Animals ◽  
2021 ◽  
Vol 11 (9) ◽  
pp. 2512
Author(s):  
Daniela-Alejandra Medina-Chávez ◽  
Irene Sánchez-Ajofrín ◽  
Patricia Peris-Frau ◽  
Carolina Maside ◽  
Vidal Montoro ◽  
...  

To date, the underlying mechanisms by which cAMP modulators act during in vitro maturation to improve oocyte developmental competence are poorly understood. Here, we sought to fill this knowledge gap by evaluating the use of phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine (IBMX) and adenylyl cyclase activator forskolin during a culture period of 2 h before in vitro maturation (pre-IVM) on the nuclear and cytoplasmic maturation features in essential organelles, cumulus cells activity, and in vitro developmental potential of sheep oocytes. Results showed that pre-IVM treatment significantly decreased (p < 0.05) the DNA damage of mature oocytes (pre-IVM = 2.08% ± 3.51% vs. control = 20.58% ± 3.51%) and increased (p ≤ 0.05) expanded blastocyst rates compared to the control (from the total of oocytes: pre-IVM = 23.89% ± 1.47% vs. control = 18.22% ± 1.47%, and from the cleaved embryos: pre-IVM = 45.16% ± 1.73% vs. control = 32.88% ± 1.73%). Considering that oocytes are highly vulnerable to the accumulation of DNA damage because of exposure to in vitro culture conditions, our results suggest that the modulation of intra-oocyte cAMP levels with forskolin and IBMX before IVM might afford oocytes a more effective DNA repair mechanism to overcome damage obstacles and ultimately improve developmental competence. This previously unappreciated action of cAMP modulators could help to develop improved methods for assisted reproduction technologies in animal and clinical research.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1167-1167
Author(s):  
Meletios A. Dimopoulos ◽  
V. L. Souliotis ◽  
A. Anagnostopoulos ◽  
A. Pouli ◽  
I. Baltadakis ◽  
...  

Abstract Introduction: Melphalan reacts extensively with cellular DNA. DNA monoadducts (MA) and interstrand cross-links (ICL) are the main events occurring after drug exposure. Subsequently, complex pathways contribute to repair of DNA lesions. We have previously showed that individual repairing capacity in p53 gene in vivo varied up to 16-fold among pts with MM receiving HDM, while increased DNA damage and slower repairing capacity correlated with improved clinical outcome (Dimopoulos et al, JCO 2005). We examine here if measurement of gene-specific DNA damage formation and repair after in vitro exposure of peripheral blood lymphocytes (PBL) to melphalan correlates with in vivo DNA damage and repair after exposure to HDM and if in vitro findings correlate with subsequent clinical outcome. Methods: Gene-specific MA and ICL formation and repair in the p53 gene were measured in PBL from MM pts, candidates for HDM (200mg/m2) and ASCT following in vitro exposure to 10 μg/ml melphalan for 1 h at 37°C. Measurements were performed 0, 2, 8 and 24 hours after in vitro exposure to melphalan. The same measurements were performed in vivo at 0, 2, 8 and 24 hours after treatment with HDM as previously described. Individual amounts of each type of DNA adducts over time (0–24h) were assessed by the area under the curve (AUC) during the whole experiment. Response after HDM was assessed according to the EBMT criteria. Measurements of DNA adducts after in vivo and in vitro treatment were correlated by the correlation-coefficient method. Results: So far, in 25 pts in vitro measurements have been performed and in 15 pts in vivo measurements and correlations with clinical outcome were made as well. One pt was treated in relapse, 3 patients had primary refractory disease and 11 were in remission. Individual kinetics of melphalan-induced DNA damage formation and repair varied remarkably among patients both for the in vivo and in vitro measurements. A strong correlation between in vivo and in vitro measurements was found (p≤0.02 for all measurements). Patients were separated into 2 groups. Responders i.e. patients who achieved CR or PR (n=10) after HDM and non responders i.e. pts who were rated as SD or PD (n=5). A significant correlation of clinical response with p53 gene-specific damage formation and repair was found in both in vitro and in vivo data. AUC (adducts/106 nucleotides x h) Total adducts (mean) Interstrand crosslinks (mean) Monoadducts (mean) Responders 919 +/− 215 303,9 +/− 108 616 +/− 154 In vitro Non responders 495 +/− 220 161,3 +/− 80,8 336 +/− 154 p-value 0,003 0,02 0,006 Responders 257 +/− 76 29,4 +/− 8,5 229 +/− 68 In vivo Non responders 122 +/− 91 13,8 +/− 4,6 112,8 +/− 82 p-value 0,009 0,002 0,01 Conclusion: Our results suggests that individuals with slower repairing capacity of the in vitro melphalan-induced p53 damage in peripheral blood lymphocytes have improved clinical outcome following subsequent treatment with HDM. We believe that our ongoing study may help select patients with MM who are more likely to benefit from HDM.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1879-1879
Author(s):  
Meletios A. Dimopoulos ◽  
Christine Liakos ◽  
Hara G. Episkopou ◽  
Dimitra T. Stefanou ◽  
Soterios A. Kyrtopoulos ◽  
...  

Abstract Abstract 1879 Poster Board I-903 DNA repair plays an important role in the protection of cells and tissues after exposure to genotoxic agents including chemotherapeutics. We have previously shown that, in peripheral blood mononuclear cells (PBMC) of multiple myeloma (MM) patients treated with melphalan, accumulation of DNA adducts in the p53 gene correlates with better therapeutic response, and that repair in different genes correlated with the gene transcriptional activity and the degree of local chromatin condensation (Dimopoulos et al, J Clin Oncol 2005;23:4381–9; Souliotis et al, DNA Repair 2006;5:972–85; Dimopoulos et al, Haematologica 2007;92:1505–12). However, the assays used are fairly time-consuming, and require complex procedures such as Southern transfer and hybridization. Thus, we now present the development and clinical application in MM of a gene-specific, quantitative method for measuring DNA damage formation/repair following exposure to anticancer drugs inducing bulky adducts. Cell line (HepG2) as well as human whole blood and PBMC from eighteen patients (13M/5F) with MM were in vitro treated with melphalan. These patients underwent high dose melphalan with autologous stem cell support (ASCT) as part of their first line therapy and the whole blood was collected on the day of stem cell mobilization. Ten (55.5%) patients achieved further myeloma reduction after ASCT; 3 patients achieved a stringent complete response (CR), 2 a CR, 2 a very good partial response (vgPR) and 3 a PR. Among 8 non-responders post-ASCT, 6 had a stable disease while 2 experienced disease progression, according to the IMWG criteria. None of the patients had previously received alkylating agent therapy (melphalan-naive patients). Moreover, cell line (HepG2) and PBMC from five healthy volunteers (all females) were treated with platinum-based drugs (cisplatin, carboplatin). Following DNA isolation, gene-specific damage formation/repair was examined using Southern blot as well as a multiplex long quantitative PCR (Q-PCR). The extent of PCR amplification was conversely proportional to the treatment concentrations of all anticancer drugs examined, implying dose-related inhibition by the DNA adducts formed. In the case of melphalan, the adduct levels measured by Q-PCR were identical to the levels of interstrand cross-links (ICL) measured by Southern blot analysis. In addition, monoadducts induced by monofunctional melphalan could not be measured by Q-PCR, suggesting that this assay measures only melphalan-induced ICLs. Application of the Q-PCR assay to in vitro-treated human blood samples from MM patients taken prior to ASCT showed that the levels of DNA damage varied up to 12-fold, which probably reflects inter-individual DNA repair differences. Interestingly, significantly greater gene-specific damage was found in the responders group compared to non-responders [176.8±67.3 adducts/106 nucleotides (range 41.0 to 273.0) for responders and 65.1±39.4 adducts/106 nucleotides (range 22.0 to 135.0) for non-responders, p=0.002]. Similar results were obtained using whole blood from the same MM patients, but differences did not reach statistical significance [84.3±63.0 adducts/106 nucleotides (range 15.0 to 165.0) for responders and 46.5±2.1 adducts/106 nucleotides (range 45.0 to 48.0) for non-responders, p=0.5]. As for the platinum-based drugs, cisplatin-induced intrastrand cross-links levels measured by Southern blot analysis, reached a plateau within ∼3h of treatment, while peak interstrand cross-links was obtained at ∼24h of exposure. Carboplatin-induced maximal levels of both intra- and interstrand cross-links were obtained within 24h of drug incubation. Parallel analysis of the same samples using both Southern blot and Q-PCR showed that the DNA adducts measured by Q-PCR correspond to total platinum-induced lesions. In conclusion, our study suggest that by using the current Q-PCR methodology, it is feasible to measure gene-specific damage formation/repair in a readily accessible biological material such as PBMC from humans exposed to anticancer drugs inducing bulky adducts and to examine, at the level of individual patient, the relationship between the induction/repair of cytotoxic DNA damage and the clinical outcome. Patient accrual is ongoing and updated results will be presented during the meeting. Disclosures: No relevant conflicts of interest to declare.


Biomedicines ◽  
2019 ◽  
Vol 7 (4) ◽  
pp. 90 ◽  
Author(s):  
Bernd Kaina

Temozolomide, a DNA methylating drug, is currently being used first-line in glioblastoma therapy. Although the mode of action of this so-called SN1 alkylating agent is well described, including the types of induced DNA damage triggering the DNA damage response and survival and death pathways, some researchers expressed doubt that data mostly obtained by in vitro models can be translated into the in vivo situation. In experimental settings, high doses of the agent are often used, which are likely to activate responses triggered by base N-alkylations instead of O6-methylguanine (O6MeG), which is the primary cytotoxic lesion induced by low doses of temozolomide and other methylating drugs in O6-methylguanine-DNA methyltransferase (MGMT) repair incompetent cells. However, numerous studies provided compelling evidence that O6MeG is not only a mutagenic, but also a powerful toxic lesion inducing DNA double-strand breaks, apoptosis, autophagy and cellular senescence. MGMT, repairing the lesion through methyl group transfer, is a key node in protecting cells against all these effects and has a significant impact on patient’s survival following temozolomide therapy, supporting the notion that findings obtained on a molecular and cellular level can be translated to the therapeutic setting in vivo. This comment summarizes the current knowledge on O6MeG-triggered pathways, including dose dependence and the question of thresholds, and comes up with the conclusion that data obtained on cell lines using low dose protocols are relevant and apoptosis, autophagy and senescence are therapeutically important endpoints.


2018 ◽  
Vol 2018 ◽  
pp. 1-10 ◽  
Author(s):  
Dongshao Chen ◽  
Xiaoting Lin ◽  
Jing Gao ◽  
Lin Shen ◽  
Zhongwu Li ◽  
...  

Based on the mechanisms by which Wee1 inhibitor and cisplatin played their own role, a promising strategy of Wee1 inhibitor combined with cisplatin was proposed, which was investigated in gastric cancer (GC). Either Wee1 inhibitor AZD1775 or cisplatin alone had a certain inhibitory effect on in vitro cell proliferation; however, the inhibitory effect was more significant when AZD1775 combined with cisplatin in vitro and in vivo. The underlying mechanisms unveiled that the increased DNA damage indicated by increased γH2AX protein, as well as augmented cell apoptosis indicated by upregulated proapoptotic proteins, was responsible for the significant inhibitory effect of AZD1775 plus cisplatin. Moreover, compared to any single drug, in vitro cell migration and invasion abilities were further attenuated by AZD1775 combined with cisplatin. There were suggestive results that the potentiated cytotoxicity between AZD1775 and cisplatin deserved a deep exploration in the future.


Sign in / Sign up

Export Citation Format

Share Document