1,324 results on '"DNA Alkylation"'
Search Results
2. Dual-Action Therapeutics: DNA Alkylation and Antimicrobial Peptides for Cancer Therapy.
- Author
-
Andrés, Celia María Curieses, Pérez de la Lastra, José Manuel, Munguira, Elena Bustamante, Andrés Juan, Celia, and Pérez-Lebeña, Eduardo
- Subjects
- *
THERAPEUTIC use of antineoplastic agents , *CELL proliferation , *IMMUNOTHERAPY , *MECHLORETHAMINE , *ANTIMICROBIAL peptides , *DNA , *IMMUNODIAGNOSIS , *CANCER chemotherapy , *MONOCLONAL antibodies , *NITROGEN mustards , *CELL lines , *DNA damage , *CELL death , *MOLECULAR structure , *CYTOCHROME P-450 , *TUMORS , *GENETIC mutation , *CELL surface antigens - Abstract
Simple Summary: Conventional cancer treatments, based on chemotherapy and radiotherapy, are often effective but suffer from serious side effects and a potential risk of resistance. Dual therapies, combining DNA alkylating agents and antimicrobial peptides, are generating great interest. Within chemotherapies, a frequently used mechanism is DNA alkylation, inducing DNA damage and subsequent cell death. Antimicrobial peptides, in turn, have demonstrated their efficacy as anticancer agents due to their ability to selectively alter cancer cell membranes. In this review, our aim has been to explore the synergistic potential of these two therapeutic modalities when used together. Cancer remains one of the most difficult diseases to treat, requiring continuous research into innovative therapeutic strategies. Conventional treatments such as chemotherapy and radiotherapy are effective to a certain extent but often have significant side effects and carry the risk of resistance. In recent years, the concept of dual-acting therapeutics has attracted considerable attention, particularly the combination of DNA alkylating agents and antimicrobial peptides. DNA alkylation, a well-known mechanism in cancer therapy, involves the attachment of alkyl groups to DNA, leading to DNA damage and subsequent cell death. Antimicrobial peptides, on the other hand, have been shown to be effective anticancer agents due to their ability to selectively disrupt cancer cell membranes and modulate immune responses. This review aims to explore the synergistic potential of these two therapeutic modalities. It examines their mechanisms of action, current research findings, and the promise they offer to improve the efficacy and specificity of cancer treatments. By combining the cytotoxic power of DNA alkylation with the unique properties of antimicrobial peptides, dual-action therapeutics may offer a new and more effective approach to fighting cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Development and Application of a Slot-Blot Assay Using the Damage Sensing Protein Atl1 to Detect and Quantify O 6 -Alkylated Guanine Bases in DNA.
- Author
-
Yaakub, Hanum, Howell, Anthony, Margison, Geoffrey P., and Povey, Andrew C.
- Subjects
DNA alkylation ,HUMAN DNA ,ALKYLATING agents ,SCHIZOSACCHAROMYCES pombe ,TEMOZOLOMIDE ,METHYLGUANINE - Abstract
Humans are unavoidably exposed to numerous different mutagenic DNA alkylating agents (AAs), but their role in the initiation of cancers is uncertain, in part due to difficulties in assessing human exposure. To address this, we have developed a screening method that measures promutagenic O
6 -alkylguanines (O6 -AlkGs) in DNA and applied it to human DNA samples. The method exploits the ability of the Schizosaccharomyces pombe alkyltransferase-like protein (Atl1) to recognise and bind to a wide range of O6 -AlkGs in DNA. We established an Atl1-based slot-blot (ASB) assay and validated it using calf thymus DNA alkylated in vitro with a range of alkylating agents and both calf thymus and human placental DNA methylated in vitro with temozolomide (TMZ). ASB signals were directly proportional to the levels of O6 -meG in these controls. Pre-treatment of DNA with the DNA repair protein O6 -methylguanine–DNA methyltransferase (MGMT) reduced binding of Atl1, confirming its specificity. In addition, MCF 10A cells were treated with 500 μM TMZ and the extracted DNA, analysed using the ASB, was found to contain 1.34 fmoles O6 -meG/μg DNA. Of six human breast tumour DNA samples assessed, five had detectable O6 -AlkG levels (mean ± SD 1.24 ± 0.25 O6 -meG equivalents/μg DNA. This study shows the potential usefulness of the ASB assay to detect and quantify total O6 -AlkGs in human DNA samples. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
4. Evaluation and comparison of DNA alkylation and oxidative damage in e-cigarette and heated tobacco users.
- Author
-
Morgil, Göksel Koç and Çok, İsmet
- Subjects
- *
DNA alkylation , *TOBACCO products , *ELECTRONIC cigarettes , *CIGARETTE smokers , *DNA damage , *DNA adducts , *LIQUID chromatography-mass spectrometry - Abstract
AbstractObjectivesMethodsResultsConclusionThis study, aimed to determine and compare DNA damage in e-cigarette and HTP (IQOS) users by assessing DNA-adducts, which are biomarkers of various DNA alkylation and oxidation.For the evaluation of DNA alkylation, N3-Ethyladenine (N3-EtA) and N3-Methyladenine (N3-MeA) adducts were used. DNA oxidation was assessed using, 8-hydroxy-2’-deoxyguanosine(8-OHdG). The urinary cotinine, N3-MeA, N3-EtA, and 8-OHdG concentrations of the cigarette smokers (n:39), e-cigarette users (n:28), IQOS users (n:20), passive smokers (n:32), and nonsmokers(n:41) who lived Ankara, Turkiye were determined using, liquid chromatography–tandem mass spectrometry (LC–MS/MS).In light of the detected 8-OHdG levels, e-cigarette (3.19 ng/g creatinine) and IQOS (4.38 ng/g creatinine) users had higher oxidative DNA damage than healthy nonsmokers (2.51 ng/g creatinine). Alkylated DNA-adducts were identified in the urine of e-cigarette (N3-MeA: 3.92 ng/g creatinine; N3-EtA: 0.23 ng/g creatinine) and IQOS (N3-MeA: 7.54 ng/g creatinine; N3-EtA: 0.29 ng/g creatinine) users. In the generation of N3-MeA adducts, a significant difference was found between IQOS users and e-cigarette users (
p < 0.05). Also, DNA alkylation in flavored e-cigarette users (N3-MeA: 4.51 ng/g creatinine; N3-EtA: 0.27 ng/g creatinine) was higher than in non-flavored e-cigarette users (N3-MeA: 2.27 ng/g creatinine; N3-EtA: 0.06 ng/g creatinine). The highest cotinine levels were found in cigarette smokers (16.1316 ng/g creatinine). No significant difference was found when e-cigarette (1163.02 ng/g creatinine) and IQOS smokers were compared (1088.3 ng/g creatinine).People who use e-cigarettes and IQOS may be at higher risk of genotoxicity than those who do not use and are not exposed to any tobacco products. Furthermore, the usage of flavoring additives in e-cigarettes contributed to additional genotoxic damage risks. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
5. Accidental Encounter of Repair Intermediates in Alkylated DNA May Lead to Double-Strand Breaks in Resting Cells.
- Author
-
Fujii, Shingo and Fuchs, Robert P.
- Subjects
- *
DNA alkylation , *DOUBLE-strand DNA breaks , *EXCISION repair , *ALKYLATING agents , *CELL proliferation , *DNA damage , *DNA repair - Abstract
In clinics, chemotherapy is often combined with surgery and radiation to increase the chances of curing cancers. In the case of glioblastoma (GBM), patients are treated with a combination of radiotherapy and TMZ over several weeks. Despite its common use, the mechanism of action of the alkylating agent TMZ has not been well understood when it comes to its cytotoxic effects in tumor cells that are mostly non-dividing. The cellular response to alkylating DNA damage is operated by an intricate protein network involving multiple DNA repair pathways and numerous checkpoint proteins that are dependent on the type of DNA lesion, the cell type, and the cellular proliferation state. Among the various alkylating damages, researchers have placed a special on O6-methylguanine (O6-mG). Indeed, this lesion is efficiently removed via direct reversal by O6-methylguanine-DNA methyltransferase (MGMT). As the level of MGMT expression was found to be directly correlated with TMZ efficiency, O6-mG was identified as the critical lesion for TMZ mode of action. Initially, the mode of action of TMZ was proposed as follows: when left on the genome, O6-mG lesions form O6-mG: T mispairs during replication as T is preferentially mis-inserted across O6-mG. These O6-mG: T mispairs are recognized and tentatively repaired by a post-replicative mismatched DNA correction system (i.e., the MMR system). There are two models (futile cycle and direct signaling models) to account for the cytotoxic effects of the O6-mG lesions, both depending upon the functional MMR system in replicating cells. Alternatively, to explain the cytotoxic effects of alkylating agents in non-replicating cells, we have proposed a "repair accident model" whose molecular mechanism is dependent upon crosstalk between the MMR and the base excision repair (BER) systems. The accidental encounter between these two repair systems will cause the formation of cytotoxic DNA double-strand breaks (DSBs). In this review, we summarize these non-exclusive models to explain the cytotoxic effects of alkylating agents and discuss potential strategies to improve the clinical use of alkylating agents. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. A systematic review on multipotent carcinogenic agent, N‐nitrosodiethylamine (NDEA), its major risk assessment, and precautions.
- Author
-
Janmeda, Pracheta, Jain, Divya, Chaudhary, Priya, Meena, Mukesh, and Singh, Devendra
- Subjects
CARCINOGENS ,FREE radical scavengers ,DNA alkylation ,MUTAGENS ,TOBACCO smoke ,ORAL drug administration - Abstract
The International Agency for Research on Cancer has classified N‐nitrosodiethylamine (NDEA) as a possible carcinogen and mutagenic substances, placing it in category 2A of compounds that are probably harmful to humans. It is found in nature and tobacco smoke, along with its precursors, and is also synthesized endogenously in the human body. The oral or parenteral administration of a minimal quantity of NDEA results in severe liver and kidney organ damage. The NDEA required bioactivation by CYP450 enzyme to form DNA adduct in the alkylation mechanism. Thus, this bioactivation directs oxidative stress and injury to cells due to the higher formation of reactive oxygen species and alters antioxidant system in tissues, whereas free radical scavengers guard the membranes from NDEA‐directed injury in many enzymes. This might be one of the reasons in the etiology of cancer that is not limited to a certain target organ but can affect various organs and organ systems. Although there are various possible approaches for the treatment of NDEA‐induced cancer, their therapeutic outcomes are still very dismal. However, several precautions were considered to be taken during handling or working with NDEA, as it considered being the best way to lower down the occurrence of NDEA‐directed cancers. The present review was designed to enlighten the general guidelines for working with NDEA, possible mechanism, to alter the antioxidant line to cause malignancy in different parts of animal body along with its protective agents. Thus, revelation to constant, unpredictable stress situations even in common life may remarkably augment the toxic potential through the rise in the oxidative stress and damage of DNA. N‐Nitrosodiethylamine (NDEA) is a potential carcinogen and mutagen. Their carcinogenic action is not limited to a certain target organ but can affect various organs and organ systems. It induces oxidative stress and cellular injury due to higher formation of reactive oxygen species and alters antioxidant system in the tissues. The present review was designed to enlighten the general guidelines for working with NDEA, possible mechanism, to alter antioxidant line to cause malignancy in different parts of animal body along with its protective agents. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. REV1 coordinates a multi-faceted tolerance response to DNA alkylation damage and prevents chromosome shattering in Drosophila melanogaster.
- Author
-
Khodaverdian, Varandt, Sano, Tokio, Maggs, Lara, Tomarchio, Gina, Dias, Ana, Tran, Mai, Clairmont, Connor, and McVey, Mitch
- Subjects
- *
DNA repair , *DROSOPHILA melanogaster , *DNA replication , *DNA polymerases , *POLYMERASES , *HOMOLOGOUS recombination , *DNA alkylation , *CHROMOSOMES - Abstract
When replication forks encounter damaged DNA, cells utilize damage tolerance mechanisms to allow replication to proceed. These include translesion synthesis at the fork, postreplication gap filling, and template switching via fork reversal or homologous recombination. The extent to which these different damage tolerance mechanisms are utilized depends on cell, tissue, and developmental context-specific cues, the last two of which are poorly understood. To address this gap, we have investigated damage tolerance responses in Drosophila melanogaster. We report that tolerance of DNA alkylation damage in rapidly dividing larval tissues depends heavily on translesion synthesis. Furthermore, we show that the REV1 protein plays a multi-faceted role in damage tolerance in Drosophila. Larvae lacking REV1 are hypersensitive to methyl methanesulfonate (MMS) and have highly elevated levels of γ-H2Av (Drosophila γ-H2AX) foci and chromosome aberrations in MMS-treated tissues. Loss of the REV1 C-terminal domain (CTD), which recruits multiple translesion polymerases to damage sites, sensitizes flies to MMS. In the absence of the REV1 CTD, DNA polymerases eta and zeta become critical for MMS tolerance. In addition, flies lacking REV3, the catalytic subunit of polymerase zeta, require the deoxycytidyl transferase activity of REV1 to tolerate MMS. Together, our results demonstrate that Drosophila prioritize the use of multiple translesion polymerases to tolerate alkylation damage and highlight the critical role of REV1 in the coordination of this response to prevent genome instability. Author summary: Organisms have evolved several ways to continue copying their DNA when it is damaged, grouped into the categories of translesion synthesis and template switching. These damage tolerance mechanisms prevent replication forks from collapsing when they encounter DNA damage and prevent catastrophic genome instability and cell death. While the proteins and pathways involved in damage tolerance are beginning to be understood at the single cell level, how they are regulated in multicellular organisms remains an intriguing question. In this study, we investigated the mechanisms by which Drosophila tolerate alkylation damage. We discovered that tissues containing rapidly dividing diploid cells favor translesion synthesis over recombination-based mechanisms of damage tolerance, preferentially utilizing different translesion polymerases in a context-dependent manner. Furthermore, we showed that the REV1 protein, best known for its role in recruiting translesion DNA polymerases to damage sites, performs multiple functions during damage tolerance. Together, our results demonstrate that damage tolerance preferences for multicellular organisms may differ from those observed in cultured cells and establish Drosophila as a useful model system for studying tolerance mechanisms. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Quantum chemical calculations of nitrosamine activation and deactivation pathways for carcinogenicity risk assessment.
- Author
-
Göller, Andreas H., Johanssen, Sandra, Zalewski, Adam, and Ziegler, Verena
- Subjects
GIBBS' energy diagram ,SMALL molecules ,DNA alkylation ,AMES test ,DNA adducts ,NITROSOAMINES - Abstract
N-nitrosamines and nitrosamine drug substance related impurities (NDSRIs) became a critical topic for the development and safety of small molecule medicines following the withdrawal of various pharmaceutical products from the market. To assess the mutagenic and carcinogenic potential of different N-nitrosamines lacking robust carcinogenicity data, several approaches are in use including the published carcinogenic potency categorization approach (CPCA), the Enhanced Ames Test (EAT), in vivo mutagenicity studies as well as read-across to analogue molecules with robust carcinogenicity data. We employ quantum chemical calculations as a pivotal tool providing insights into the likelihood of reactive ion formation and subsequent DNA alkylation for a selection of molecules including e.g., carcinogenic N-nitrosopiperazine (NPZ), N-nitrosopiperidine (NPIP), together with N-nitrosodimethylamine (NDMA) as well as non-carcinogenic N-nitrosomethyl-tert-butylamine (NTBA) and bis (butan-2-yl) (nitros)amine (BBNA). In addition, a series of nitrosomethylaminopyridines is compared side-by-side. We draw comparisons between calculated reaction profiles for structures representing motifs common to NDSRIs and those of confirmed carcinogenic and noncarcinogenic molecules with in vivo data from cancer bioassays. Furthermore, our approach enables insights into reactivity and relative stability of intermediate species that can be formed upon activation of several nitrosamines. Most notably, we reveal consistent differences between the free energy profiles of carcinogenic and non-carcinogenic molecules. For the former, the intermediate diazonium ions mostly react, kinetically controlled, to the more stable DNA adducts and less to the water adducts via transition-states of similar heights. Non-carcinogenic molecules yield stable carbocations as intermediates that, thermodynamically controlled, more likely form the statistically preferred water adducts. In conclusion, our data confirm that quantum chemical calculations can contribute to a weight of evidence approach for the risk assessment of nitrosamines. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. ALKBH5-mediated m6A demethylation of Runx2 mRNA promotes extracellular matrix degradation and intervertebral disc degeneration.
- Author
-
Lei, Yu, Zhan, Enyu, Chen, Chao, Hu, Yaoquan, Lv, Zhengpin, He, Qicong, Wang, Xuenan, Li, Xingguo, and Zhang, Fan
- Subjects
- *
INTERVERTEBRAL disk , *RNA-binding proteins , *DNA alkylation , *DEMETHYLATION , *EXTRACELLULAR matrix - Abstract
Background: N6-methyladenosine (m6A) methylation is a prevalent RNA modification implicated in various diseases. However, its role in intervertebral disc degeneration (IDD), a common cause of low back pain, remains unclear. Results: In this investigation, we explored the involvement of m6A demethylation in the pathogenesis of IDD. Our findings revealed that ALKBH5 (alkylated DNA repair protein AlkB homolog 5), an m6A demethylase, exhibited upregulation in degenerative discs upon mild inflammatory stimulation. ALKBH5 facilitated m6A demethylation within the three prime untranslated region (3′-UTR) of Runx2 mRNA, consequently enhancing its mRNA stability in a YTHDF1 (YTH N6-methyladenosine RNA binding protein F1)-dependent manner. The subsequent elevation in Runx2 expression instigated the upregulation of ADAMTSs and MMPs, pivotal proteases implicated in extracellular matrix (ECM) degradation and IDD progression. In murine models, subcutaneous administration of recombinant Runx2 protein proximal to the lumbar disc in mice elicited complete degradation of intervertebral discs (IVDs). Injection of recombinant MMP1a and ADAMTS10 proteins individually induced mild to moderate degeneration of the IVDs, while co-administration of MMP1a and ADAMTS10 resulted in moderate to severe degeneration. Notably, concurrent injection of the Runx2 inhibitor CADD522 with recombinant Runx2 protein did not result in IVD degeneration in mice. Furthermore, genetic knockout of ALKBH5 and overexpression of YTHDF1 in mice, along with lipopolysaccharide (LPS) treatment to induce inflammation, did not alter the expression of Runx2, MMPs, and ADAMTSs, and no degeneration of the IVDs was observed. Conclusion: Our study elucidates the role of ALKBH5-mediated m6A demethylation of Runx2 mRNA in activating MMPs and ADAMTSs, thereby facilitating ECM degradation and promoting the occurrence of IDD. Our findings suggest that targeting the ALKBH5/Runx2/MMPs/ADAMTSs axis may represent a promising therapeutic strategy for preventing IDD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Duocarmycin SA Reduces Proliferation and Increases Apoptosis in Acute Myeloid Leukemia Cells In Vitro.
- Author
-
Chen, William A., Williams, Terry G., So, Leena, Drew, Natalie, Fang, Jie, Ochoa, Pedro, Nguyen, Nhi, Jawhar, Yasmeen, Otiji, Jide, Duerksen-Hughes, Penelope J., Reeves, Mark E., Casiano, Carlos A., Jin, Hongjian, Dovat, Sinisa, Yang, Jun, Boyle, Kristopher E., and Francis-Boyle, Olivia L.
- Subjects
- *
ACUTE myeloid leukemia , *MYELOID cells , *DOUBLE-strand DNA breaks , *DNA damage , *APOPTOSIS , *DNA repair , *HEMATOLOGIC malignancies - Abstract
Acute myeloid leukemia (AML) is a hematological malignancy that is characterized by an expansion of immature myeloid precursors. Despite therapeutic advances, the prognosis of AML patients remains poor and there is a need for the evaluation of promising therapeutic candidates to treat the disease. The objective of this study was to evaluate the efficacy of duocarmycin Stable A (DSA) in AML cells in vitro. We hypothesized that DSA would induce DNA damage in the form of DNA double-strand breaks (DSBs) and exert cytotoxic effects on AML cells within the picomolar range. Human AML cell lines Molm-14 and HL-60 were used to perform 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT), DNA DSBs, cell cycle, 5-ethynyl-2-deoxyuridine (EdU), colony formation unit (CFU), Annexin V, RNA sequencing and other assays described in this study. Our results showed that DSA induced DNA DSBs, induced cell cycle arrest at the G2M phase, reduced proliferation and increased apoptosis in AML cells. Additionally, RNA sequencing results showed that DSA regulates genes that are associated with cellular processes such as DNA repair, G2M checkpoint and apoptosis. These results suggest that DSA is efficacious in AML cells and is therefore a promising potential therapeutic candidate that can be further evaluated for the treatment of AML. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Insights on cyclophosphamide metabolism and anticancer mechanism of action: A computational study.
- Author
-
Dabbish, Eslam, Scoditti, Stefano, Shehata, Mohammed N. I., Ritacco, Ida, Ibrahim, Mahmoud A. A., Shoeib, Tamer, and Sicilia, Emilia
- Subjects
- *
CYCLOPHOSPHAMIDE , *DNA alkylation , *CANCER chemotherapy , *PRODRUGS , *ANTINEOPLASTIC agents , *METABOLISM - Abstract
The oxazaphosphorine cyclophosphamide (CP) is a DNA-alkylating agent commonly used in cancer chemotherapy. This anticancer agent is administered as a prodrug activated by a liver cytochrome P450-catalyzed 4-hydroxylation reaction that yields the active, cytotoxic metabolite. The primary metabolite, 4-hydroxycyclophosphamide, equilibrates with the ring-open aldophosphamide that undergoes ß-elimination to yield the therapeutically active DNA cross-linking phosphoramide mustard and the byproduct acrolein. The present paper presents a DFT investigation of the different metabolic phases and an insight into the mechanism by which CP exerts its cytotoxic action. A detailed computational analysis of the energy profiles describing all the involved transformations and the mechanism of DNA alkylation is given with the aim to contribute to an increase of knowledge that, after more than 60 years of unsuccessful attempts, can lead to the design and development of a new generation of oxazaphosphorines. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Size- and Stereochemistry-Dependent Transcriptional Bypass of DNA Alkyl Phosphotriester Adducts in Mammalian Cells.
- Author
-
Tan, Ying, Wu, Jiabin, Clabaugh, Garrit, Li, Lin, Du, Hua, and Wang, Yinsheng
- Subjects
DNA alkylation ,DNA damage ,transcriptional mutagenesis - Abstract
Environmental, endogenous and therapeutic alkylating agents can react with internucleotide phosphate groups in DNA to yield alkyl phosphotriester (PTE) adducts. Alkyl-PTEs are induced at relatively high frequencies and are persistent in mammalian tissues; however, their biological consequences in mammalian cells have not been examined. Herein, we assessed how alkyl-PTEs with different alkyl group sizes and stereochemical configurations (S P and R P diastereomers of Me and nPr) affect the efficiency and fidelity of transcription in mammalian cells. We found that, while the R P diastereomer of Me- and nPr-PTEs constituted moderate and strong blockages to transcription, respectively, the S P diastereomer of the two lesions did not appreciably perturb transcription efficiency. In addition, none of the four alkyl-PTEs induced mutant transcripts. Furthermore, polymerase η assumed an important role in promoting transcription across the S P-Me-PTE, but not any of other three lesions. Loss of other translesion synthesis (TLS) polymerases tested, including Pol κ, Pol ι, Pol ξ and REV1, did not alter the transcription bypass efficiency or mutation frequency for any of the alkyl-PTE lesions. Together, our study provided important new knowledge about the impact of alkyl-PTE lesions on transcription and expanded the substrate pool of Pol η in transcriptional bypass.
- Published
- 2022
13. RNA polymerase tracking along damaged DNA: Impact on DNA repair and mutagenesis.
- Author
-
Yi-Ying Chiou and Kemp, Michael G.
- Subjects
- *
DNA repair , *RNA polymerases , *MUTAGENESIS , *DNA alkylation , *RNA synthesis , *AUTOMOBILE repair - Abstract
This article explores the relationship between DNA damage, DNA repair, and mutagenesis. It specifically focuses on the behavior of RNA polymerase (RNAP) at sites of DNA damage and its impact on gene transcription. The research, conducted using a mutagen-induced mouse tumor model and mathematical modeling, suggests that RNAP often bypasses DNA lesions during transcription and does not resume transcription at the site of stalling after DNA repair. Additionally, the article discusses a study on the genetic changes in liver tumors caused by a specific chemical treatment. Whole genome sequencing revealed that lesions not repaired before DNA replication led to mutations in the tumor cells. The study also found that highly expressed genes had fewer mutations. These findings have implications for understanding mutagenesis, DNA repair, and the development of cancer. [Extracted from the article]
- Published
- 2024
- Full Text
- View/download PDF
14. Ada protein- and sequence context-dependent mutagenesis of alkyl phosphotriester lesions in Escherichia coli cells.
- Author
-
Wu, Jiabin, Yuan, Jun, Price, Nathan, and Wang, Yinsheng
- Subjects
Ada ,DNA alkylation ,DNA damage ,DNA polymerase ,DNA repair ,DNA replication ,alkyl phosphotriester ,diastereomer ,mutagenesis ,mutagenesis mechanism ,replication bypass ,sequence context ,translesion synthesis ,Alkylation ,DNA Damage ,DNA Replication ,DNA ,Bacterial ,Escherichia coli ,Escherichia coli Proteins ,Gene Deletion ,Mutagenesis ,O(6)-Methylguanine-DNA Methyltransferase ,Transcription Factors - Abstract
Alkyl phosphotriester (alkyl-PTE) lesions are frequently induced in DNA and are resistant to repair. Here, we synthesized and characterized methyl (Me)- and n-butyl (nBu)-PTEs in two diastereomeric configurations (Sp and Rp) at six different flanking dinucleotide sites, i.e. XT and TX (X = A, C, or G), and assessed how these lesions impact DNA replication in Escherichia coli cells. When single-stranded vectors contained an Sp-Me-PTE in the sequence contexts of 5-AT-3, 5-CT-3, or 5-GT-3, DNA replication was highly efficient and the replication products for all three sequence contexts contained 85-90% AT and 5-10% TG. Thus, the replication outcome was largely independent of the identity of the 5 nucleotide adjacent to an Sp-Me-PTE. Furthermore, replication across these lesions was not dependent on the activities of DNA polymerases II, IV, or V; Ada, a protein involved in adaptive response and repair of Sp-Me-PTE in E. coli, however, was essential for the generation of the mutagenic products. Additionally, the Rp diastereomer of Me-PTEs at XT sites and both diastereomers of Me-PTEs at TX sites exhibited error-free replication bypass. Moreover, Sp-nBu-PTEs at XT sites did not strongly impede DNA replication, and other nBu-PTEs displayed moderate blockage effects, with none of them being mutagenic. Taken together, these findings provide in-depth understanding of how alkyl-PTE lesions are recognized by the DNA replication machinery in prokaryotic cells and reveal that Ada contributes to mutagenesis of Sp-Me-PTEs in E. coli.
- Published
- 2020
15. The roles of polymerases ν and θ in replicative bypass of O6- and N2-alkyl-2-deoxyguanosine lesions in human cells.
- Author
-
Du, Hua, Wang, Pengcheng, Wu, Jun, He, Xiaomei, and Wang, Yinsheng
- Subjects
DNA alkylation ,DNA damage ,DNA polymerase ,DNA replication ,MS ,mutagenesis ,mutagenesis mechanism ,polymerase θ ,polymerase ν ,Alkylation ,Chromatography ,High Pressure Liquid ,DNA Repair ,DNA-Directed DNA Polymerase ,Deoxyguanosine ,HEK293 Cells ,Humans ,Mutagenesis ,Tandem Mass Spectrometry - Abstract
Exogenous and endogenous chemicals can react with DNA to produce DNA lesions that may block DNA replication. Not much is known about the roles of polymerase (Pol) ν and Pol θ in translesion synthesis (TLS) in cells. Here we examined the functions of these two polymerases in bypassing major-groove O6-alkyl-2-deoxyguanosine (O6-alkyl-dG) and minor-groove N2-alkyl-dG lesions in human cells, where the alkyl groups are ethyl, n-butyl (nBu), and, for O6-alkyl-dG, pyridyloxobutyl. We found that Pol ν and Pol θ promote TLS across major-groove O6-alkyl-dG lesions. O6-alkyl-dG lesions mainly induced G→A mutations that were modulated by the two TLS polymerases and the structures of the alkyl groups. Simultaneous ablation of Pol ν and Pol θ resulted in diminished mutation frequencies for all three O6-alkyl-dG lesions. Depletion of Pol ν alone reduced mutations only for O6-nBu-dG, and sole loss of Pol θ attenuated the mutation rates for O6-nBu-dG and O6-pyridyloxobutyl-dG. Replication across the two N2-alkyl-dG lesions was error-free, and Pol ν and Pol θ were dispensable for their replicative bypass. Together, our results provide critical knowledge about the involvement of Pol ν and Pol θ in bypassing alkylated guanine lesions in human cells.
- Published
- 2020
16. Allele-Specific Replication Inhibition of Mitochondrial DNA by MITO-PIP Conjugated with Alkylation Reagent
- Author
-
Hidaka, Takuya and Hidaka, Takuya
- Published
- 2022
- Full Text
- View/download PDF
17. Size- and Stereochemistry-Dependent Transcriptional Bypass of DNA Alkyl Phosphotriester Adducts in Mammalian Cells
- Author
-
Ying Tan, Jiabin Wu, Garrit Clabaugh, Lin Li, Hua Du, and Yinsheng Wang
- Subjects
DNA alkylation ,transcriptional mutagenesis ,DNA damage ,Biochemistry ,QD415-436 - Abstract
Environmental, endogenous and therapeutic alkylating agents can react with internucleotide phosphate groups in DNA to yield alkyl phosphotriester (PTE) adducts. Alkyl-PTEs are induced at relatively high frequencies and are persistent in mammalian tissues; however, their biological consequences in mammalian cells have not been examined. Herein, we assessed how alkyl-PTEs with different alkyl group sizes and stereochemical configurations (SP and RP diastereomers of Me and nPr) affect the efficiency and fidelity of transcription in mammalian cells. We found that, while the RP diastereomer of Me- and nPr-PTEs constituted moderate and strong blockages to transcription, respectively, the SP diastereomer of the two lesions did not appreciably perturb transcription efficiency. In addition, none of the four alkyl-PTEs induced mutant transcripts. Furthermore, polymerase η assumed an important role in promoting transcription across the SP-Me-PTE, but not any of other three lesions. Loss of other translesion synthesis (TLS) polymerases tested, including Pol κ, Pol ι, Pol ξ and REV1, did not alter the transcription bypass efficiency or mutation frequency for any of the alkyl-PTE lesions. Together, our study provided important new knowledge about the impact of alkyl-PTE lesions on transcription and expanded the substrate pool of Pol η in transcriptional bypass.
- Published
- 2022
- Full Text
- View/download PDF
18. Assessing the effect of N-oxidation on the mutagenicity of 1-pyrazolines using the Ames assay.
- Author
-
Inami, Keiko, Miura, Motofumi, Yoshida, Masafumi, and Mochizuki, Masataka
- Subjects
AMES test ,DNA alkylation ,ESCHERICHIA coli ,ELECTRON density ,SALMONELLA typhimurium - Abstract
N-Nitrosamines are well known as environmental carcinogens. We have reported that N-nitroso-N-methylbutylamine was oxidized by Fe
2+ -Cu2+ -H2 O2 to 5-methyl-5-nitro-1-pyrazoline, a direct-acting N-oxide. 1-Pyrazolines have not been reported to exhibit genotoxicity. In this study, we investigated the effect of N-oxidation on the mutagenicity of 1-pyrazolines using the Ames assay. The mutagenicity of 5-alkyl-5-nitro-1-pyrazoline 1-oxide (1a; methyl, 1b; ethyl), the N-oxide isomer (3-alkyl-3-nitro-1-pyrazoline 1-oxide; 2a; methyl, 2b; ethyl), and the corresponding nonoxides (3-alkyl-3-nitro-1-pyrazoline; 3a; methyl, 3b; ethyl) was assayed in Salmonella typhimurium TA1535 and Escherichia coli WP2uvrA. The ratios of mutagenic potency in S. typhimurium TA1535 versus E. coli WP2uvrA were compared with those of N-alkylnitrosoureas. To predict the reaction site on the pyrazolines with nucleophiles, the electron density of the pyrazolines was obtained by theoretical calculations. The pyrazolines were mutagenic in S. typhimurium TA1535 and E. coli WP2uvrA. The ratio of S. typhimurium TA1535 to E. coli WP2uvrA 1a (87:13) or 1b (90:10) was similar to that of N-ethyl-N-nitrosourea (70:30). In contrast, the mutagenic ratio of 2a (22:78) or 2b (52:48) was similar to that of N-propyl-N-nitrosourea (48:52) or N-butyl-N-nitrosourea (14:86). The ratio of 3a (53:47) or 3b (54:46) was similar to that of N-propyl-N-nitrosourea or N-butyl-N-nitrosourea. The pyrazolines exhibit genotoxicity, and the mutagenic potency of the 1-pyrazolines is influenced by N-oxidation. We estimated that the mutagenicity of 1a or 1b was caused by DNA ethylation, and the isomers or the nonoxides were mutagenic via formation of alkylated DNA, which contains an alkyl chain longer than the propyl. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
19. Archaeal DNA alkylation repair conducted by DNA glycosylase and methyltransferase.
- Author
-
Yin, Youcheng and Zhang, Likui
- Subjects
- *
DNA alkylation , *DNA repair , *METHYLTRANSFERASES , *DNA demethylation , *BACTERIAL proteins , *METHYLGUANINE , *DIOXYGENASES - Abstract
Alkylated bases in DNA created in the presence of endogenous and exogenous alkylating agents are either cytotoxic or mutagenic, or both to a cell. Currently, cells have evolved several strategies for repairing alkylated base. One strategy is a base excision repair process triggered by a specific DNA glycosylase that is used for the repair of the cytotoxic 3-methyladenine. Additionally, the cytotoxic and mutagenic O6-methylguanine (O6-meG) is corrected by O6-methylguanine methyltransferase (MGMT) via directly transferring the methyl group in the lesion to a specific cysteine in this protein. Furthermore, oxidative DNA demethylation catalyzed by DNA dioxygenase is utilized for repairing the cytotoxic 3-methylcytosine (3-meC) and 1-methyladenine (1-meA) in a direct reversal manner. As the third domain of life, Archaea possess 3-methyladenine DNA glycosylase II (AlkA) and MGMT, but no DNA dioxygenase homologue responsible for oxidative demethylation. Herein, we summarize recent progress in structural and biochemical properties of archaeal AlkA and MGMT to gain a better understanding of archaeal DNA alkylation repair, focusing on similarities and differences between the proteins from different archaeal species and between these archaeal proteins and their bacterial and eukaryotic relatives. To our knowledge, it is the first review on archaeal DNA alkylation repair conducted by DNA glycosylase and methyltransferase. Key points: • Archaeal MGMT plays an essential role in the repair of O6-meG • Archaeal AlkA can repair 3-meC and 1-meA [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
20. Deficiency in mammalian STN1 promotes colon cancer development via inhibiting DNA repair.
- Author
-
Dinh Duc Nguyen, Kim, Eugene, Nhat Thong Le, Xianzhong Ding, Jaiswal, Rishi Kumar, Kostlan, Raymond Joseph, Thi Ngoc Thanh Nguyen, Shiva, Olga, Minh Thong Le, and Weihang Chai
- Subjects
- *
DNA mismatch repair , *DNA repair , *COLON cancer , *CARCINOGENESIS , *DNA alkylation , *DNA demethylation - Abstract
The article presents a study which identified STN1 knockout mouse deficiency as a risk factor for colorectal cancer (CRC) and implicated the previously unknown STN1-base excision repair (BER) axis in protecting colon tissues from oxidative damage. Topics discussed include analysis of CTC1 and STN1 genetic alterations, association between mutational signatures and STN1-deficient CRC tumors, and generation of STN1 conditional knockout (cko) mice and genotyping.
- Published
- 2023
- Full Text
- View/download PDF
21. DNA Alkylation Damage by Nitrosamines and Relevant DNA Repair Pathways.
- Author
-
Fahrer, Jörg and Christmann, Markus
- Subjects
- *
DNA alkylation , *DNA adducts , *NITROSOAMINES , *DNA repair , *DNA damage , *DNA synthesis , *ALKYLATING agents - Abstract
Nitrosamines occur widespread in food, drinking water, cosmetics, as well as tobacco smoke and can arise endogenously. More recently, nitrosamines have been detected as impurities in various drugs. This is of particular concern as nitrosamines are alkylating agents that are genotoxic and carcinogenic. We first summarize the current knowledge on the different sources and chemical nature of alkylating agents with a focus on relevant nitrosamines. Subsequently, we present the major DNA alkylation adducts induced by nitrosamines upon their metabolic activation by CYP450 monooxygenases. We then describe the DNA repair pathways engaged by the various DNA alkylation adducts, which include base excision repair, direct damage reversal by MGMT and ALKBH, as well as nucleotide excision repair. Their roles in the protection against the genotoxic and carcinogenic effects of nitrosamines are highlighted. Finally, we address DNA translesion synthesis as a DNA damage tolerance mechanism relevant to DNA alkylation adducts. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
22. Insight Into Factors Governing Formation, Synthesis and Stereochemical Configuration of DNA Adducts Formed by Mitomycins.
- Author
-
Paz, Manuel M. and Champeil, Elise
- Subjects
- *
DNA adducts , *MITOMYCINS , *DNA alkylation , *ALKYLATING agents , *MITOMYCIN C , *ANTINEOPLASTIC agents - Abstract
Mitomycin C, (MC), an antitumor drug used in the clinics, is a DNA alkylating agent. Inert in its native form, MC is reduced to reactive mitosenes in cellulo which undergo nucleophilic attack by DNA bases to form monoadducts as well as interstrand crosslinks (ICLs). These properties constitute the molecular basis for the cytotoxic effects of the drug. The mechanism of DNA alkylation by mitomycins has been studied for the past 30 years and, until recently, the consensus was that drugs of the mitomycins family mainly target CpG sequences in DNA. However, that paradigm was recently challenged. Here, we relate the latest research on both MC and dicarbamoylmitomycin C (DMC), a synthetic derivative of MC which has been used to investigate the regioselectivity of mitomycins DNA alkylation as well as the relationship between mitomycins reductive activation pathways and DNA adducts stereochemical configuration. We also review the different synthetic routes to access mitomycins nucleoside adducts and oligonucleotides containing MC/DMC DNA adducts located at a single position. Finally, we briefly describe the DNA structural modifications induced by MC and DMC adducts and how site specifically modified oligonucleotides have been used to elucidate the role each adduct plays in the drugs cytotoxicity. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
23. NEO212, a Perillyl Alcohol-Temozolomide Conjugate, Triggers Macrophage Differentiation of Acute Myeloid Leukemia Cells and Blocks Their Tumorigenicity.
- Author
-
Chen, Thomas C., Minea, Radu O., Swenson, Steve, Yang, Zhuoyue, Thein, Thu Zan, and Schönthal, Axel H.
- Subjects
- *
THERAPEUTIC use of antineoplastic agents , *CELL differentiation , *DACARBAZINE , *METHYLTRANSFERASES , *MACROPHAGES , *ANTINEOPLASTIC agents , *DRUG resistance , *TEMOZOLOMIDE , *DESCRIPTIVE statistics , *CELL lines , *CYTARABINE , *DATA analysis software , *CELL death , *PHARMACODYNAMICS - Abstract
Simple Summary: Acute myeloid leukemia (AML) is a cancer of the blood and bone marrow, where cancer cells are unable to complete the maturation process towards white blood cells, but instead continue to proliferate. We are developing a novel chemotherapeutic drug, NEO212, that has shown anticancer activity in different preclinical cancer models, including AML. In the current study, we demonstrate that NEO212 forces AML cells to resume the differentiation process and progress towards the macrophage phenotype, which is accompanied by a loss of proliferation. NEO212 is able to achieve this growth-inhibitory effect even in AML cells that are resistant to other chemotherapeutic drugs in clinical use, such as cytarabine and temozolomide. In mouse models with AML, we found that treatment with NEO212 was well tolerated and resulted in an apparent cure of these animals. We propose that NEO212 should be developed further and evaluated in clinical trials with AML patients. Many patients with acute myeloid leukemia (AML) are still dying from this disease. In the past, the alkylating agent temozolomide (TMZ) has been investigated for AML and found to be partially effective; however, the presence of O6-methylguanine DNA methyltransferase (MGMT; a DNA repair enzyme) in tumor cells confers profound treatment resistance against TMZ. We are developing a novel anticancer compound, called NEO212, where TMZ was covalently conjugated to perillyl alcohol (a naturally occurring monoterpene). NEO212 has revealed robust therapeutic activity in a variety of preclinical cancer models, including AML. In the current study, we investigated its impact on a panel of human AML cell lines and found that it exerted cytotoxic potency even against MGMT-positive cells that were highly resistant to TMZ. Furthermore, NEO212 strongly stimulated the expression of a large number of macrophage-associated marker genes, including CD11b/ITGAM. This latter effect could not be mimicked when cells were treated with TMZ or an equimolar mix of individual agents, TMZ plus perillyl alcohol. The superior cytotoxic impact of NEO212 appeared to involve down-regulation of MGMT protein levels. In a mouse model implanted with TMZ-resistant, MGMT-positive AML cells, two 5-day cycles of 25 mg/kg NEO212 achieved an apparent cure, as mice survived >300 days without any signs of disease. In parallel toxicity studies with rats, a 5-day cycle of 200 mg/kg NEO212 was well tolerated by these animals, whereas animals that were given 200 mg/kg TMZ all died due to severe leukopenia. Together, our results show that NEO212 exerts pleiotropic effects on AML cells that include differentiation, proliferation arrest, and eventual cell death. In vivo, NEO212 was well tolerated even at dosages that far exceed the therapeutic need, indicating a large therapeutic window. These results present NEO212 as an agent that should be considered for development as a therapeutic agent for AML. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
24. Insight on the Interaction between the Camptothecin Derivative and DNA Oligomer Mimicking the Target of Topo I Inhibitors.
- Author
-
Bocian, Wojciech, Naumczuk, Beata, Urbanowicz, Magdalena, Sitkowski, Jerzy, Bednarek, Elżbieta, Wiktorska, Katarzyna, Pogorzelska, Anna, Wielgus, Ewelina, and Kozerski, Lech
- Subjects
- *
CAMPTOTHECIN , *DNA topoisomerase I , *DEOXYRIBOZYMES , *DNA , *MOLECULAR dynamics , *DNA alkylation , *LIGAND binding (Biochemistry) - Abstract
The understanding of the mechanism of Topo I inhibition by organic ligands is a crucial source of information that has led to the design of more effective and safe pharmaceuticals in oncological chemotherapy. The vast number of inhibitors that have been studied in this respect over the last decades have enabled the creation of a concept of an 'interfacial inhibitor', thereby describing the machinery of Topo I inhibition. The central module of action of this machinery is the interface of a Topo I/DNA/inhibitor ternary complex. Most of the 'interfacial inhibitors' are primarily kinetic inhibitors that form molecular complexes with an "on–off" rate timing; therefore, all of the contacts between the inhibitor and both the enzyme and the DNA are essential to keep the complex stable and reduce the "off rate". To test this hypothesis, we designed the compound using a C-9-(N-(2′-hydroxyethyl)amino)methyl substituent in an SN38 core, with a view that a flexible substituent may bind inside the nick of a model of the DNA and stabilize the complex, leading to a reduction in the "off rate" of a ligand in a potential ternary complex in vivo. Using docking analysis and molecular dynamics, free energy calculations on the level of the MM-PBSA and MM-GBSA model, here we presented the in silico-calculated structure of a ternary complex involving the studied compound 1. This confirmed our suggestion that compound 1 is situated in a groove of the nicked DNA model in a few conformations. The number of hydrogen bonds between the components of a ternary complex was established, which strengthens the complex and supports our view. The docking analysis and free energy calculations for the receptor structures which were obtained in the MD simulations of the ternary complex 1/DNA/Topo I show that the binding constant is stronger than it was for similar complexes with TPT, CPT, and SN38, which are commonly considered as strong Topo I inhibitors. The binary complex structure 1/DNA was calculated and compared with the experimental results of a complex that was in a solution. The analysis of the cross-peaks in NOESY spectra allowed us to assign the dipolar interactions between the given protons in the calculated structures. A DOSY experiment in the solution confirmed the strong binding of a ligand in a binary complex, having a Ka of 746 mM−1, which was compared with a Ka of 3.78 mM−1 for TPT. The MALDI-ToF MS showed the presence of the biohybrid, thus evidencing the occurrence of DNA alkylation by compound 1. Because of it having a strong molecular complex, alkylation is the most efficient way to reduce the "on–off" timing as it acts as a tool that causes the cog to brake in a working gear, and this is this activity we want to highlight in our contribution. Finally, the Topo I inhibition test showed a lower IC50 of the studied compound than it did for CPT and SN38. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
25. Development of Cyclic and Hairpin Pyrrole-Imidazole Polyamides for Specific Recognition of Disease-Associated DNA Sequences
- Author
-
Hirose, Yuki and Hirose, Yuki
- Published
- 2024
26. University of California Researcher Adds New Findings in the Area of Glioblastomas [Dnar-14. Modulating Populational Variance of Methyl-guanine Methyl Transferase (Mgmt) Expression Through Microrna Degradation: A Novel Mechanism of Temozolomide...].
- Subjects
DNA alkylation ,COENZYMES ,ALKYLATING agents ,ATAXIA telangiectasia ,TRANSFERASES ,METHYLGUANINE - Abstract
Researchers at the University of California have discovered a novel mechanism of temozolomide resistance in glioblastomas involving the degradation of microRNA miR-181d. This degradation leads to an increase in the mean level of MGMT expression and a widening of the variance in MGMT expression within cell populations. The study suggests that modulating both the mean and variance of MGMT expression can impact temozolomide sensitivity, highlighting the potential implications for microRNA biology in general. [Extracted from the article]
- Published
- 2024
27. New Glioblastomas Research Reported from University of California (Path-30. Clinical Outcomes And Predictive Biomarkers For Idh-wildtype Glioblastomas Developing Hypermutation Following Temozolomide Treatment).
- Subjects
DNA ligases ,ALKYLATING agents ,DNA alkylation ,TEMOZOLOMIDE ,DISEASE risk factors - Abstract
Research from the University of California discusses the development of hypermutation in IDH-wildtype glioblastomas following temozolomide treatment. The study found that a small percentage of patients developed hypermutation at recurrence, leading to longer overall survival. Specific methylation patterns in the MGMT and KCNQ1DN genes were identified as potential biomarkers for predicting favorable outcomes in patients with glioblastomas. This research provides insights into potential predictive biomarkers and treatment strategies for patients with glioblastomas. [Extracted from the article]
- Published
- 2024
28. Reports from University of California Riverside Describe Recent Advances in Drug Research (n2-alkyl-dg Lesions Elicit R-loop Accumulation In the Genome).
- Subjects
DNA alkylation ,NUCLEIC acids ,CANCER chemotherapy ,ALKYLATING agents ,COENZYMES ,DNA adducts - Abstract
A recent study from the University of California Riverside explores the impact of minor-groove N-2-alkyl-dG lesions on genome integrity by eliciting R-loop accumulation in chromatin and DNA. The research suggests that these lesions may disrupt transcription elongation and compromise genome integrity, potentially leading to genome instability. The findings propose a potential therapeutic strategy involving the combination of R-loop helicase inhibitors with DNA alkylating drugs. This study sheds light on the intricate relationship between DNA damage, R-loop formation, and genome stability, offering insights for future drug research and development. [Extracted from the article]
- Published
- 2024
29. Studies from University Hospital Clinic Valladolid Reveal New Findings on Cancer (Dual-Action Therapeutics: DNA Alkylation and Antimicrobial Peptides for Cancer Therapy).
- Subjects
DNA alkylation ,ANTIMICROBIAL peptides ,DRUG therapy ,CANCER treatment ,ALKYLATING agents - Abstract
A recent report from the University Hospital Clinic Valladolid in Spain explores the potential of dual-action therapeutics for cancer treatment. The researchers discuss the combination of DNA alkylating agents and antimicrobial peptides as a promising approach to fighting cancer. DNA alkylation involves attaching alkyl groups to DNA, causing damage and cell death, while antimicrobial peptides disrupt cancer cell membranes and modulate immune responses. The study examines the mechanisms of action and current research findings of these therapeutic modalities, highlighting their potential to improve the efficacy and specificity of cancer treatments. [Extracted from the article]
- Published
- 2024
30. Mechanism of DNA alkylation-induced transcriptional stalling, lesion bypass, and mutagenesis
- Author
-
Xu, Liang, Wang, Wei, Wu, Jiabin, Shin, Ji Hyun, Wang, Pengcheng, Unarta, Ilona Christy, Chong, Jenny, Wang, Yinsheng, and Wang, Dong
- Subjects
Genetics ,Aetiology ,2.1 Biological and endogenous factors ,Alkylation ,DNA ,DNA Repair ,DNA Replication ,Humans ,Mutagenesis ,RNA Polymerase II ,Saccharomyces cerevisiae ,Transcription ,Genetic ,transcription ,DNA alkylation ,transcriptional lesion bypass ,transcriptional mutagenesis ,RNA polymerase II - Abstract
Alkylated DNA lesions, induced by both exogenous chemical agents and endogenous metabolites, interfere with the efficiency and accuracy of DNA replication and transcription. However, the molecular mechanisms of DNA alkylation-induced transcriptional stalling and mutagenesis remain unknown. In this study, we systematically investigated how RNA polymerase II (pol II) recognizes and bypasses regioisomeric O2-, N3-, and O4-ethylthymidine (O2-, N3-, and O4-EtdT) lesions. We observed distinct pol II stalling profiles for the three regioisomeric EtdT lesions. Intriguingly, pol II stalling at O2-EtdT and N3-EtdT sites is exacerbated by TFIIS-stimulated proofreading activity. Assessment for the impact of the EtdT lesions on individual fidelity checkpoints provided further mechanistic insights, where the transcriptional lesion bypass routes for the three EtdT lesions are controlled by distinct fidelity checkpoints. The error-free transcriptional lesion bypass route is strongly favored for the minor-groove O2-EtdT lesion. In contrast, a dominant error-prone route stemming from GMP misincorporation was observed for the major-groove O4-EtdT lesion. For the N3-EtdT lesion that disrupts base pairing, multiple transcriptional lesion bypass routes were found. Importantly, the results from the present in vitro transcriptional studies are well correlated with in vivo transcriptional mutagenesis analysis. Finally, we identified a minor-groove-sensing motif from pol II (termed Pro-Gate loop). The Pro-Gate loop faces toward the minor groove of RNA:DNA hybrid and is involved in modulating the translocation of minor-groove alkylated DNA template after nucleotide incorporation opposite the lesion. Taken together, this work provides important mechanistic insights into transcriptional stalling, lesion bypass, and mutagenesis of alkylated DNA lesions.
- Published
- 2017
31. Bloom syndrome DNA helicase mitigates mismatch repair-dependent apoptosis.
- Author
-
Uechi, Yuka, Fujikane, Ryosuke, Morita, Sho, Tamaoki, Sachio, and Hidaka, Masumi
- Subjects
- *
DNA repair , *DOUBLE-strand DNA breaks , *DNA alkylation , *APOPTOSIS , *DNA helicases , *ALKYLATING agents - Abstract
Generation of O6-methylguanine (O6-meG) by DNA-alkylating agents such as N-methyl N-nitrosourea (MNU) activates the multiprotein mismatch repair (MMR) complex and the checkpoint response involving ATR/CHK1 and ATM/CHK2 kinases, which may in turn trigger cell cycle arrest and apoptosis. The Bloom syndrome DNA helicase BLM interacts with the MMR complex, suggesting functional relevance to repair and checkpoint responses. We observed a strong interaction of BLM with MMR proteins in HeLa cells upon treatment with MNU as evidenced by co-immunoprecipitation as well as colocalization in the nucleus as revealed by dual immunofluorescence staining. Knockout of BLM sensitized HeLa MR cells to MNU-induced cell cycle disruption and enhanced expression of the apoptosis markers cleaved caspase-9 and PARP1. MNU-treated BLM-deficient cells also exhibited a greater number of 53BP1 foci and greater phosphorylation levels of H2AX at S139 and RPA32 at S8, indicating the accumulation of DNA double-strand breaks. These findings suggest that BLM prevents double-strand DNA breaks during the MMR-dependent DNA damage response and mitigates O6-meG-induced apoptosis. • BLM associates with mismatch repair complex upon alkylated damage. • Deficiency in BLM sensitizes cells to alkylating agent. • BLM suppresses accumulation of double strand breaks caused by DNA alkylation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Inhibition of human DNA alkylation damage repair enzyme ALKBH2 by HIV protease inhibitor ritonavir.
- Author
-
Shaji, Unnikrishnan P., Tuti, Nikhil, Alim, S.K., Mohan, Monisha, Das, Susmita, Meur, Gargi, Swamy, Musti J., and Anindya, Roy
- Subjects
- *
DNA ligases , *HIV protease inhibitors , *DNA alkylation , *ALKYLATING agents , *ANTI-HIV agents , *DNA adducts - Abstract
The human DNA repair enzyme AlkB homologue-2 (ALKBH2) repairs methyl adducts from genomic DNA and is overexpressed in several cancers. However, there are no known inhibitors available for this crucial DNA repair enzyme. The aim of this study was to examine whether the first-generation HIV protease inhibitors having strong anti-cancer activity can be repurposed as inhibitors of ALKBH2. We selected four such inhibitors and performed in vitro binding analysis against ALKBH2 based on alterations of its intrinsic tryptophan fluorescence and differential scanning fluorimetry. The effect of these HIV protease inhibitors on the DNA repair activity of ALKBH2 was also evaluated. Interestingly, we observed that one of the inhibitors, ritonavir, could inhibit ALKBH2-mediated DNA repair significantly via competitive inhibition and sensitized cancer cells to alkylating agent methylmethane sulfonate (MMS). This work may provide new insights into the possibilities of utilizing HIV protease inhibitor ritonavir as a DNA repair antagonist. • Four HIV drugs with anti-cancer properties were evaluated as ALKBH2 antagonist. • Intrinsic Trp fluorescence quenching showed that ritonavir binds to ALKBH2. • Thermal unfolding showed ritonavir binding requires ALKBH2 R110 residue. • Enzyme kinetics results suggested that ritonavir competitively inhibits ALKBH2. • Ritonavir exposure reduced cancer cell survival following MMS treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. Michael addition-activated alkylation of G-quadruplex DNA with methylamine-protected vinyl-quinazolinone derivatives.
- Author
-
Chen, Yutong, Onizuka, Kazumitsu, and Nagatsugi, Fumi
- Subjects
- *
DNA alkylation , *ACTIVATION (Chemistry) , *CHEMICAL systems , *ALKYLATION , *QUADRUPLEX nucleic acids - Abstract
[Display omitted] The role of G-quadruplex (G4) in cellular processes can be investigated by the covalent modification of G4-DNA using alkylating reagents. Controllable alkylating reagents activated by external stimuli can react elegantly and selectively. Herein, we report a chemical activation system that can significantly boost the reaction rate of methylamine-protected vinyl-quinazolinone (VQ) derivative for the alkylation of G4‐DNA. The two screened activators can transform low-reactive VQ-NHR' to highly reactive intermediates following the Michael addition mechanism. This approach expands the toolbox of activable G4 alkylating reagents. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. A DNA repair-independent role for alkyladenine DNA glycosylase in alkylation-induced unfolded protein response.
- Author
-
Milano, Larissa, Charlier, Clara F., Andreguetti, Rafaela, Cox, Thomas, Healing, Eleanor, Thom(e, Marcos P., Elliott, Ruan M., Samson, Leona D., Masson, Jean-Yves, Lenz, Guido, Henriques, João Antonio P., Nohturff, Axel, and Meira, Lisiane B.
- Subjects
- *
UNFOLDED protein response , *DNA alkylation , *ALKYLATING agents , *COMMERCIAL products , *DNA , *DNA repair - Abstract
Alkylating agents damage DNA and proteins and are widely used in cancer chemotherapy. While cellular responses to alkylationinduced DNA damage have been explored, knowledge of how alkylation affects global cellular stress responses is sparse. Here, we examined the effects of the alkylating agent methylmethane sulfonate (MMS) on gene expression in mouse liver, using mice deficient in alkyladenine DNA glycosylase (Aag), the enzyme that initiates the repair of alkylated DNA bases. MMS induced a robust transcriptional response in wild-type liver that included markers of the endoplasmic reticulum (ER) stress/unfolded protein response (UPR) known to be controlled by XBP1, a key UPR effector. Importantly, this response is significantly reduced in the Aag knockout. To investigate how AAG affects alkylation-induced UPR, the expression of UPR markers after MMS treatment was interrogated in human glioblastoma cells expressing different AAG levels. Alkylation induced the UPR in cells expressing AAG; conversely, AAG knockdown compromised UPR induction and led to a defect in XBP1 activation. To verify the requirements for the DNA repair activity of AAG in this response, AAG knockdown cells were complemented with wild-type Aag or with an Aag variant producing a glycosylase-deficient AAG protein. As expected, the glycosylasedefective Aag does not fully protect AAG knockdown cells against MMS-induced cytotoxicity. Remarkably, however, alkylationinduced XBP1 activation is fully complemented by the catalytically inactive AAG enzyme. This work establishes that, besides its enzymatic activity, AAG has noncanonical functions in alkylationinduced UPR that contribute to cellular responses to alkylation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
35. The ASCC2 CUE domain in the ALKBH3-ASCC DNA repair complex recognizes adjacent ubiquitins in K63-linked polyubiquitin.
- Author
-
Lombardi, Patrick M., Haile, Sara, Rusanov, Timur, Rodell, Rebecca, Anoh, Rita, Baer, Julia G., Burke, Kate A., Gray, Lauren N., Hacker, Abigail R., Kebreau, Kayla R., Ngandu, Christine K., Orland, Hannah A., Osei-Asante, Emmanuella, Schmelyun, Dhane P., Shorb, Devin E., Syed, Shaheer H., Veilleux, Julianna M., Majumdar, Ananya, Mosammaparast, Nima, and Wolberger, Cynthia
- Subjects
- *
DNA repair , *DNA alkylation , *N-terminal residues , *UBIQUITIN , *POISONS - Abstract
Alkylation of DNA and RNA is a potentially toxic lesion that can result in mutations and even cell death. In response to alkylation damage, K63-linked polyubiquitin chains are assembled that localize the Alpha-ketoglutarate-dependent dioxygenase alkB homolog 3-Activating Signal Cointegrator 1 Complex Subunit (ASCC) repair complex to damage sites in the nucleus. The protein ASCC2, a subunit of the ASCC complex, selectively binds K63-linked polyubiquitin chains via its coupling of ubiquitin conjugation to ER degradation (CUE) domain. The basis for polyubiquitin-binding specificity was unclear, because CUE domains in other proteins typically bind a single ubiquitin and do not discriminate among different polyubiquitin linkage types. We report here that the ASCC2 CUE domain selectively binds K63-linked diubiquitin by con-tacting both the distal and proximal ubiquitin. The ASCC2 CUE domain binds the distal ubiquitin in a manner similar to that reported for other CUE domains bound to a single ubiquitin, whereas the contacts with the proximal ubiquitin are unique to ASCC2. Residues in the N-terminal portion of the ASCC2 α1 helix contribute to the binding interaction with the proximal ubiquitin of K63-linked diubiquitin. Mutation of residues within the N-terminal portion of the ASCC2 a1 helix decreases ASCC2 recruitment in response to DNA alkylation, supporting the functional significance of these interactions during the alkylation damage response. Our study reveals the versatility of CUE domains in ubiquitin recognition. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
36. Hoogsteen Base Pairings: A New Paradigm for DNA Replication, DNA Recognition, and DNA Repair.
- Author
-
Datta, Neelabh
- Subjects
- *
BASE pairs , *DNA repair , *DNA structure , *DNA alkylation , *BANKING industry , *DNA sequencing , *DNA - Abstract
In contrast to Watson-Crick (WC) base pairing, Hoogsteen (HG) base pairing involves flipping a purine base 180° between its anti and syn conformation. Recent studies have shown that HG pairs coexist in dynamical equilibrium, and several biological functions depend on them. This significance has stirred computational research on this base-pairing transition. However, a methodical reproduction of sequence variations has continued to be out of reach. It is becoming increasingly clear that Hoogsteen base pairs play a crucial role in DNA replication, recognition, damage repair, and incorrect sequence repair. The Protein Data Bank contains a variety of Hoogsteen base pairing modes that include the preference for A-T versus G-C bps, TA versus GG pairs, and a preference for 5'-purines at terminal ends. RNA A-form duplexes are strongly disfavoured by Hoogsteen base pairs, in stark contrast to B-form DNA. Therefore, N1-methyl adenosine and N1-methyl guanosine, which is found in DNA as alkylation impairment and in RNA as posttranscriptional adjustments, have great differences in effects. They create G-C+ and A-U Hoogsteen base pairs in duplex DNA that preserve the structural integrity of the double helix but obstruct base pairing altogether and induce local duplex melting in RNA, providing a mechanism for potently disrupting RNA structure through posttranscriptional modifications. In duplex DNA, they maintain the structural integrity of the double helix by creating G-C+ and A-U Hoogsteen base pairs, but block base pairing altogether and cause local duplex melting in RNA, thus providing a potent means for disrupting RNA structure post transcriptionally. As a result of the markedly different inclinations for BDNA and A-RNA to form Hoogsteen base pairs, they may be able to balance the opposing demands of maintaining genome stability and dynamically modulating the epitranscriptome. This review examines the occurrence of Hoogsteen base pairs in DNA and RNA duplexes. Keywords: Hoogsteen bp, DNA structure, DNA dynamics, DNA sequencing, Hoogsteen bp dynamicsIn contrast to Watson-Crick (WC) base pairing, Hoogsteen (HG) base pairing involves flipping a purine base 180° between its anti and syn conformation. Recent studies have shown that HG pairs coexist in dynamical equilibrium, and several biological functions depend on them. This significance has stirred computational research on this base-pairing transition. However, a methodical reproduction of sequence variations has continued to be out of reach. It is becoming increasingly clear that Hoogsteen base pairs play a crucial role in DNA replication, recognition, damage repair, and incorrect sequence repair. The Protein Data Bank contains a variety of Hoogsteen base pairing modes that include the preference for A-T versus G-C bps, TA versus GG pairs, and a preference for 5'-purines at terminal ends. RNA A-form duplexes are strongly disfavoured by Hoogsteen base pairs, in stark contrast to B-form DNA. Therefore, N1-methyl adenosine and N1-methyl guanosine, which is found in DNA as alkylation impairment and in RNA as posttranscriptional adjustments, have great differences in effects. They create G-C+ and A-U Hoogsteen base pairs in duplex DNA that preserve the structural integrity of the double helix but obstruct base pairing altogether and induce local duplex melting in RNA, providing a mechanism for potently disrupting RNA structure through posttranscriptional modifications. In duplex DNA, they maintain the structural integrity of the double helix by creating G-C+ and A-U Hoogsteen base pairs, but block base pairing altogether and cause local duplex melting in RNA, thus providing a potent means for disrupting RNA structure post transcriptionally. As a result of the markedly different inclinations for BDNA and A-RNA to form Hoogsteen base pairs, they may be able to balance the opposing demands of maintaining genome stability and dynamically modulating the epitranscriptome. This review examines the occurrence of Hoogsteen base pairs in DNA and RNA duplexes. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
37. DNA Damaging Agents in Chemical Biology and Cancer
- Author
-
Basilius Sauter and Dennis Gillingham
- Subjects
dna alkylation ,cnacer ,chemotherapy ,covalent drugs ,nucleic acids ,Chemistry ,QD1-999 - Abstract
Despite their toxicity, DNA alkylating drugs remain a cornerstone of anticancer therapy. The classical thinking was that rapidly dividing tumour cells left more of its DNA in an exposed single-stranded state, making these rapidly dividing cells more susceptible to alkylating drugs. As our understanding of DNA repair pathways has matured it is becoming clear that compromised DNA repair – a hallmark of cancer – plays a role as well in defining the therapeutic window of these toxic drugs. Hence, although new alkylating motifs are unlikely to progress through the clinic, the legacy of these medicines is that we now understand the therapeutic potential of targeting DNA damage repair pathways. Here we look at the history of alkylating agents as anticancer drugs, while also summarizing the different mechanistic approaches to covalent DNA modification. We also provide several case studies on how insights into compromised DNA repair pathways are paving the way for potent and less toxic targeted medicines against the DNA damage response.
- Published
- 2020
- Full Text
- View/download PDF
38. Quantitation of DNA by nuclease P1 digestion and UPLC-MS/MS to assess binding efficiency of pyrrolobenzodiazepine
- Author
-
Yong Ma, Buyun Chen, and Donglu Zhang
- Subjects
Nuclease P1 ,UPLC-MS/MS ,DNA quantitation ,DNA alkylation ,Pyrrolobenzodiazepine (PBD-Dimer) ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Accurate DNA quantitation is a prerequisite in many biomedical and pharmaceutical studies. Here we established a new DNA quantitation method by nuclease P1 digestion and UPLC-MS/MS analysis. DNA fragments can be efficiently hydrolyzed to single deoxyribonucleotides by nuclease P1 in a short time. The decent stabilities of all the four deoxyribonucleotides were confirmed under different conditions. Deoxyadenosine monophosphate (dAMP) was selected as the surrogate for DNA quantitation because dAMP showed the highest sensitivity among the four deoxyribonucleotides in the UPLC-MS/MS analysis. The linear range in DNA quantitation by this method is 1.2–5000 ng/mL. In the validation, the inter-day and intra-day accuracies were within 90%–110%, and the inter-day and intra-day precision were acceptable (RSD
- Published
- 2020
- Full Text
- View/download PDF
39. Understanding the Alkylation Mechanism of 3‐Chloropiperidines – NMR Kinetic Studies and Isolation of Bicyclic Aziridinium Ions.
- Author
-
Helbing, Tim, Georg, Mats, Stöhr, Fabian, Carraro, Caterina, Becker, Jonathan, Gatto, Barbara, and Göttlich, Richard
- Subjects
- *
ALKYLATION , *IONS , *DNA alkylation , *SILVER salts , *SINGLE crystals - Abstract
The present study describes the kinetic analysis of the 3‐chloropiperidine alkylation mechanism. These nitrogen mustard‐based compounds are expected to react via a highly electrophilic bicyclic aziridinium ion, which is readily attacked by nucleophiles. Halide abstraction using silver salts with weakly coordinating anions lead to the isolation of these proposed intermediates, whereas their structure was confirmed by single crystal XRD. Kinetic studies of the aziridinium ions also revealed notable reactivity differences of the C5 gem‐methylated compounds and their unmethylated counterparts. The observed reactivity trends were also reflected by NMR studies in aqueous solution and DNA alkylation experiments of the related 3‐chloropiperidines. Therefore, the underlying Thorpe‐Ingold effect might be considered as another option to adjust the alkylation activity of these compounds. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
40. New Cancer Research from Swiss Federal Institute of Technology Zurich (ETH) Outlined (Acylfulvenes covalently interact with thioredoxin as an additional cancer target).
- Subjects
PROTEIN drugs ,DNA alkylation ,THIOREDOXIN ,REPORTERS & reporting ,CHEMICAL biology - Abstract
A new study from the Swiss Federal Institute of Technology Zurich (ETH) explores the interactions between acylfulvenes (AFs), a class of DNA alkylating drugs, and thioredoxin (Trx), a key redox regulating enzyme. The study found that AFs, particularly hydroxymethylacylfulvene (HMAF), inhibited Trx activity by covalently modifying its active site cysteines. This research suggests that Trx could be a potential target for AFs in cancer treatment. The study provides insights into the molecular interactions that govern the impact of AFs on cancer cells. [Extracted from the article]
- Published
- 2024
41. Data from University of Alabama at Birmingham Update Knowledge in Glioblastomas (Temozolomide and the PARP Inhibitor Niraparib Enhance Expression of Natural Killer Group 2D Ligand ULBP1 and Gamma-Delta T Cell Cytotoxicity in Glioblastoma).
- Subjects
DNA alkylation ,ALKYLATING agents ,DEVELOPMENTAL biology ,GENE expression ,TEMOZOLOMIDE - Abstract
A recent study conducted at the University of Alabama at Birmingham has explored the potential of combining the PARP inhibitor niraparib with the DNA alkylator temozolomide (TMZ) to enhance immune cell recognition in glioblastoma (GBM). The researchers found that the combination therapy increased the expression of natural killer group 2D ligand ULBP1 and gamma-delta T cell cytotoxicity in GBM. These findings suggest that the combination of PARP inhibition, DNA alkylation, and gamma-delta T cell therapy could be a promising treatment approach for GBM. [Extracted from the article]
- Published
- 2024
42. Versatile cell-based assay for measuring DNA alkylation damage and its repair.
- Author
-
Li, Yong, Mao, Peng, Basenko, Evelina Y., Lewis, Zachary, Smerdon, Michael J., and Czaja, Wioletta
- Subjects
- *
DNA alkylation , *DNA damage , *CANCER chemotherapy , *CARCINOGENS , *METABOLITES , *MUTAGENESIS - Abstract
DNA alkylation damage induced by environmental carcinogens, chemotherapy drugs, or endogenous metabolites plays a central role in mutagenesis, carcinogenesis, and cancer therapy. Base excision repair (BER) is a conserved, front line DNA repair pathway that removes alkylation damage from DNA. The capacity of BER to repair DNA alkylation varies markedly between different cell types and tissues, which correlates with cancer risk and cellular responses to alkylation chemotherapy. The ability to measure cellular rates of alkylation damage repair by the BER pathway is critically important for better understanding of the fundamental processes involved in carcinogenesis, and also to advance development of new therapeutic strategies. Methods for assessing the rates of alkylation damage and repair, especially in human cells, are limited, prone to significant variability due to the unstable nature of some of the alkyl adducts, and often rely on indirect measurements of BER activity. Here, we report a highly reproducible and quantitative, cell-based assay, named alk-BER (alkylation Base Excision Repair) for measuring rates of BER following alkylation DNA damage. The alk-BER assay involves specific detection of methyl DNA adducts (7-methyl guanine and 3-methyl adenine) directly in genomic DNA. The assay has been developed and adapted to measure the activity of BER in fungal model systems and human cell lines. Considering the specificity and conserved nature of BER enzymes, the assay can be adapted to virtually any type of cultured cells. Alk-BER offers a cost efficient and reliable method that can effectively complement existing approaches to advance integrative research on mechanisms of alkylation DNA damage and repair. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
43. "Repaired and Activated" DNAzyme Enables the Monitoring of DNA Alkylation Repair in Live Cells.
- Author
-
Wang, Xiangnan, Yi, Xin, Huang, Zhimei, He, Jianjun, Wu, Zhenkun, Chu, Xia, and Jiang, Jian‐Hui
- Subjects
- *
DNA alkylation , *DNA repair , *DEOXYRIBOZYMES , *DIAGNOSIS , *CANCER treatment - Abstract
Direct measurement of DNA repair is critical for the annotation of their clinical relevance and the discovery of drugs for cancer therapy. Here we reported a "repaired and activated" DNAzyme (RADzyme) by incorporating a single methyl lesion (O6MeG, 3MeC, or 1MeA) at designated positions through systematic screening. We found that the catalytic activity of the RADzyme was remarkably suppressed and could be restored via enzyme‐mediated DNA repair. Benefit from these findings, a fluorogenic RADzyme sensor was developed for the monitoring of MGMT‐mediated repair of O6MeG lesion. Importantly, the sensor allowed the evaluation of MGMT repair activity in different cells and under drugs treatment. Furthermore, another RADzyme sensor was engineered for the monitoring of ALKBH2‐mediated repair of 3MeC lesion. This strategy provides a simple and versatile tool for the study of the basic biology of DNA repair, clinical diagnosis and therapeutic assessment. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
44. ROLE OF DNA REPAIR IN AGING AND MALIGNANCY.
- Author
-
Fatima, Kaynat, Raza, Syed Tasleem, Rizvi, Saliha, and Srivastava, Sanchita
- Subjects
- *
DNA ligases , *DNA alkylation , *DNA damage , *NUCLEAR DNA , *REACTIVE oxygen species , *MITOCHONDRIAL DNA - Abstract
DNA repair enzymes are proteins that detect and repair physical damage to DNA induced by radiation, ultraviolet light, or reactive oxygen species. The repair of DNA damage prevents the loss of genetic information, the creation of double-strand breaks, and the formation of DNA crosslinks. The time-dependent reduction of functional properties is known as aging. Mitochondrial malfunction and the buildup of genetic damage are two common factors of aging. In fact, the poor maintenance of nuclear and mitochondrial DNA is likely a major factor in aging. When the DNA repair machinery isn't operating fine, DNA lesions and mutations can occur, which can lead to cancer development. In fact, the poor maintenance of nuclear and mitochondrial DNA is likely a major factor in aging. When the DNA repair enzymes isn't operating fine, DNA lesions and mutations can occur, which can lead to cancer development. The large number of alterations per cell, which can reach 105, has been identified as a driving mechanism in oncogenesis. These findings show that abnormalities in the DNA repair pathway contribute to the senescence as well as cancer. Nucleotide excision repair (NER), base excision repair (BER), double-strand break repair, mismatch repair (MMR), are all major DNA repair processes in mammalian cells. BER excises mostly oxidative and alkylation DNA damage, NER removes bulky, helix-distorting lesions from DNA (e.g., ultraviolet (UV) photodimers), MMR corrects replication errors. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
45. Specific transport of temozolomide does not override DNA repair-mediated chemoresistance.
- Author
-
Bahrami, Katayun, Kärkkäinen, Jussi, Bibi, Sania, Huttunen, Johanna, Tampio, Janne, Montaser, Ahmed B., Moody, Catherine L., Lehtonen, Marko, Rautio, Jarkko, Wheelhouse, Richard T., and Huttunen, Kristiina M.
- Subjects
- *
DNA alkylation , *DNA mismatch repair , *EXCISION repair , *TEMOZOLOMIDE , *DRUG resistance in cancer cells - Abstract
Temozolomide (TMZ) a DNA alkylating agent, is the standard-of-care for brain tumors, such as glioblastoma multiforme (GBM). Although the physicochemical and pharmacokinetic properties of TMZ, such as chemical stability and the ability to cross the blood-brain barrier (BBB), have been questioned in the past, the acquired chemoresistance has been the main limiting factor of long-term clinical use of TMZ. In the present study, an L -type amino acid transporter 1 (LAT1)-utilizing prodrug of TMZ (TMZ-AA, 6) was prepared and studied for its cellular accumulation and cytotoxic properties in human squamous cell carcinoma, UT-SCC-28 and UT-SCC-42B cells, and TMZ-sensitive human glioma, U-87MG cells that expressed functional LAT1. TMZ-AA 6 accumulated more effectively than TMZ itself into those cancer cells that expressed LAT1 (UT-SCC-42B). However, this did not correlate with decreased viability of treated cells. Indeed, TMZ-AA 6 , similarly to TMZ itself, required adjuvant inhibitor(s) of DNA-repair systems, O 6-methylguanine-DNA methyl transferase (MGMT) and base excision repair (BER), as well as active DNA mismatch repair (MMR), for maximal growth inhibition. The present study shows that improving the delivery of this widely-used methylating agent is not the main barrier to improved chemotherapy, although utilizing a specific transporter overexpressed at the BBB or glioma cells can have targeting advantages. To obtain a more effective anticancer prodrug, the compound design focus should shift to altering the major DNA alkylation site or inhibiting DNA repair systems. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
46. Synthesis of 2‐Chloro‐3‐amino indenone derivatives and their evaluation as inhibitors of DNA dealkylation repair.
- Author
-
Nigam, Richa, Raveendra Babu, Kaki, Ghosh, Topi, Kumari, Bhavini, Das, Prolay, Anindya, Roy, and Ahmed Khan, Faiz
- Subjects
- *
DNA repair , *DNA alkylation , *AROMATIC amines , *METHYL methanesulfonate , *ALKYLATING agents , *IRON compounds , *DNA synthesis - Abstract
DNA alkylation damage, emanating from the exposure to environmental alkylating agents or produced by certain endogenous metabolic processes, affects cell viability and genomic stability. Fe(II)/2‐oxoglutarate‐dependent dioxygenase enzymes, such as Escherichia coli AlkB, are involved in protecting DNA from alkylation damage. Inspired by the natural product indenone derivatives reported to inhibit this class of enzymes, and a set of 2‐chloro‐3‐amino indenone derivatives was synthesized and screened for their inhibitory properties against AlkB. The synthesis of 2‐chloro‐3‐amino indenone derivatives was achieved from 2,3‐dichloro indenones through addition–elimination method using alkyl/aryl amines under catalyst‐free conditions. Using an in vitro reconstituted DNA repair assay, we have identified a 2‐chloro‐3‐amino indenone compound 3o to be an inhibitor of AlkB. We have determined the binding affinity, mode of interaction, and kinetic parameters of inhibition of 3o and tested its ability to sensitize cells to methyl methanesulfonate that mainly produce DNA alkylation damage. This study established the potential of indenone‐derived compounds as inhibitors of Fe(II)/2‐oxoglutarate‐dependent dioxygenase AlkB. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
47. Photoinduced DNA Interstrand Cross‐Linking by 1,1′‐Biphenyl Analogues: Substituents and Leaving Groups Combine to Determine the Efficiency of Cross‐Linker.
- Author
-
Fan, Heli, Sun, Huabing, Zhang, Qi, and Peng, Xiaohua
- Subjects
- *
CARBOCATIONS , *DNA , *NITROPHENYL compounds , *FREE radicals , *DNA alkylation - Abstract
Two series of 1,1′‐biphenyl analogues with various leaving groups (L=OAc, OCH3, OCHCH=CH2, OCH2Ph, SPh, SePh, and Ph3P+) were synthesized. Their reactivity towards DNA and the reaction mechanism were investigated by determining DNA interstrand cross‐link (ICL) efficiency, radical and carbocation formation, and the cross‐linking reaction sites. All compounds induced DNA ICL formation upon 350 nm irradiation via a carbocation that was generated from oxidation of the corresponding free radicals. The ICL efficiency and the reaction rate strongly depended on the combined effect of the leaving group and the substituent. Among all compounds tested, the high ICL efficiency (30–43 %) and fast reaction rate were observed with compounds carrying a nitrophenyl group and acetate (2 a), ether (2 b and 2 c), or triphenylphosphonium salt (2 g) as leaving groups. Most compounds with a 4‐methoxybenzene group showed similar DNA ICL efficiency (≈30 %) with a slow DNA cross‐linking reaction rate. Both cation trapping and free radical trapping adducts were detected in the photo activation process of these compounds, which provided direct evidence for the proposed mechanism. Heat stability study in combination with sequence study suggested that these photo‐generated benzyl cations alkylate DNA at dG, dA, and dC sites. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
48. Generation of reconstituted hemato-lymphoid murine embryos by placental transplantation into embryos lacking HSCs.
- Author
-
Jeon, Hyojung, Asano, Keigo, Wakimoto, Arata, Kulathunga, Kaushalya, Tran, Mai Thi Nhu, Nakamura, Megumi, Yokomizo, Tomomasa, Hamada, Michito, and Takahashi, Satoru
- Subjects
- *
DNA alkylation , *EMBRYOS , *HEMATOPOIETIC stem cells , *LEUCOCYTES , *HEMATOPOIESIS - Abstract
In order to increase the contribution of donor HSC cells, irradiation and DNA alkylating agents have been commonly used as experimental methods to eliminate HSCs for adult mice. But a technique of HSC deletion for mouse embryo for increase contribution of donor cells has not been published. Here, we established for the first time a procedure for placental HSC transplantation into E11.5 Runx1-deficient mice mated with G1-HRD-Runx1 transgenic mice (Runx1-/-::Tg mice) that have no HSCs in the fetal liver. Following the transplantation of fetal liver cells from mice (allogeneic) or rats (xenogeneic), high donor cell chimerism was observed in Runx1-/-::Tg embryos. Furthermore, chimerism analysis and colony assay data showed that donor fetal liver hematopoietic cells contributed to both white blood cells and red blood cells. Moreover, secondary transplantation into adult recipient mice indicated that the HSCs in rescued Runx1-/-::Tg embryos had normal abilities. These results suggest that mice lacking fetal liver HSCs are a powerful tool for hematopoiesis reconstruction during the embryonic stage and can potentially be used in basic research on HSCs or xenograft models. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
49. Evaluation of the DNA Alkylation Properties of a Chlorambucil‐Conjugated Cyclic Pyrrole‐Imidazole Polyamide.
- Author
-
Hirose, Yuki, Hashiya, Kaori, Bando, Toshikazu, and Sugiyama, Hiroshi
- Subjects
- *
DNA alkylation , *POLYAMIDES , *CAPILLARY electrophoresis , *DNA , *HIGH performance liquid chromatography , *ANTINEOPLASTIC agents - Abstract
Hairpin pyrrole‐imidazole polyamides (hPIPs) and their chlorambucil (Chb) conjugates (hPIP‐Chbs) can alkylate DNA in a sequence‐specific manner, and have been studied as anticancer drugs. Here, we conjugated Chb to a cyclic PIP (cPIP), which is known to have a higher binding affinity than the corresponding hPIP, and investigated the DNA alkylation properties of the resulting cPIP‐Chb using the optimized capillary electrophoresis method and conventional HPLC product analysis. cPIP‐Chb conjugate 3 showed higher alkylation activity at its binding sites than did hPIP‐Chb conjugates 1 and 2. Subsequent HPLC analysis revealed that the alkylation site of conjugate 3, which was identified by capillary electrophoresis, was reliable and that conjugate 3 alkylates the N3 position of adenine as do hPIP‐Chbs. Moreover, conjugate 3 showed higher cytotoxicity against LNCaP prostate cancer cells than did conjugate 1 and cytotoxicity comparable to that of conjugate 2. These results suggest that cPIP‐Chbs could be novel DNA alkylating anticancer drugs. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
50. In silico profiling and structural insights of zinc metal ion on O6-methylguanine methyl transferase and its interactions using molecular dynamics approach.
- Author
-
Gharouni, Marzieh, Mosaddeghi, Hamid, Mehrzad, Jamshid, Es-haghi, Ali, and Motavalizadehkakhky, Alireza
- Subjects
- *
MOLECULAR dynamics , *PROLIFERATING cell nuclear antigen , *METAL ions , *AMINO acid sequence , *AMINO acid analysis , *ZINC ions , *DNA alkylation , *AMINO acid residues - Abstract
O6-methylguanine DNA methyl transferase (MGMT) is a metalloenzyme participating in the repair of alkylated DNA. In this research, we performed a comparative study for evaluating the impact of zinc metal ion on the behavior and interactions of MGMT in the both enzymatic forms of apo MGMT and holo MGMT. DNA and proliferating cell nuclear antigen (PCNA), as partners of MGMT, were utilized to evaluate molecular interactions by virtual microscopy of molecular dynamics simulation. The stability and conformational alterations of each forms (apo and holo) MGMT-PCNA, and (apo and holo) MGMT-DNA complexes were calculated by MM/PBSA method. A total of seven systems including apo MGMT, holo MGMT, free PCNA, apo MGMT-PCNA, holo MGMT-PCNA, apo MGMT–DNA, and holo MGMT-DNA complexes were simulated. In this study, we found that holo MGMT was more stable and had better folding and functional properties than that of apo MGMT. Simulation analysis of (apo and holo) MGMT-PCNA complexes displayed that the sequences of the amino acids involved in the interactions were different in the two forms of MGMT. The important amino acids of holo MGMT involved in its interaction with PCNA included E92, K101, A119, G122, N123, P124, and K125, whereas the important amino acids of apo MGMT included R128, R135, S152, N157, Y158, and L162. Virtual microscopy of molecular dynamics simulation showed that the R128 and its surrounding residues were important amino acids involved in the interaction of holo MGMT with DNA that was exactly consistent with X-ray crystallography structure. In the apo form of the protein, the N157 and its surrounding residues were important amino acids involved in the interaction with DNA. The binding free energies of − 387.976, − 396.226, − 622.227, and − 617.333 kcal/mol were obtained for holo MGMT-PCNA, apo MGMT-PCNA, holo MGMT-DNA, and apo MGMT-DNA complexes, respectively. The principle result of this research was that the area of molecular interactions differed between the two states of MGMT. Therefore, in investigations of metalloproteins, the metal ion must be preserved in their structures. Finally, it is recommended to use the holo form of metalloproteins in in vitro and in silico researches. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.