ijms-logo

Journal Browser

Journal Browser

DNA Damage, DNA Repair, and Cancer 2.0

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Oncology".

Deadline for manuscript submissions: closed (31 August 2023) | Viewed by 13987

Special Issue Editor


E-Mail Website
Guest Editor
Department of Diagnostic Pathology, Kindai University Hospital, Osaka 589-8511, Japan
Interests: transcription-coupled DNA damage; lymphomagenesis; mRNA export; breast carcinogenesis; antibody engineering
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

DNA lesions produced by a variety of genotoxic stresses are precisely repaired to avoid the genomic instability which is associated with a high predisposition to cancer development. The occurrence of unrepaired DNA lesions is censored by DNA-damage response (DDR) composed of complex cascades followed by the induction of cell-cycle arrest or apoptosis. As most tumors show impaired DDR, many studies have focused on the understanding of detailed molecular mechanisms in DDR-pathways and/or cancer-related DNA repair defects to reveal the cause of cancers. In addition, this information has provided us new therapeutic targets. Recently, when the germ-line mutations exist in breast cancers, PARP inhibitors offer a new therapeutic option based on the concept of synthetic lethality. Moreover, it is known that various inhibitors targeting molecules associated with DDR are in clinical trials. Comprehensive analyses of canonical and non-canonical DNA repair pathways and the interactions of each molecule defective in various cancers are promising to search for potential therapeutic targets. Therefore, authors are invited to submit original research and review articles that address the physiological, pathophysiological, pharmacological, and epidemiological importance of DDR and/or DNA repair pathways in cancer development.

Topics include, but are not limited to:

  • Involvement of canonical or non-canonical DNA repair pathways in cancer development;
  • Identification of the novel therapeutic targets in DNA repair pathway;
  • Animal models mimicking the development of various human cancers;
  • Pathological significance of aberrantly expressed DNA repair-related molecules in human clinical samples.

Dr. Kazuhiko Kuwahara
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • DNA-damage response
  • DNA repair molecules
  • genome instability
  • cellular senescence
  • animal models
  • DNA damage
  • DNA repair and cancer

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

2 pages, 161 KiB  
Editorial
DNA Damage, DNA Repair, and Cancer: Second Edition
by Kazuhiko Kuwahara
Int. J. Mol. Sci. 2023, 24(23), 16835; https://doi.org/10.3390/ijms242316835 - 28 Nov 2023
Viewed by 700
Abstract
Following our first Special Issue, we are pleased to present this Special Issue in the International Journal of Molecular Sciences, titled ‘DNA Damage, DNA Repair, and Cancer: Second Edition’ [...] Full article
(This article belongs to the Special Issue DNA Damage, DNA Repair, and Cancer 2.0)

Research

Jump to: Editorial, Review

23 pages, 4393 KiB  
Article
AZD-7648, a DNA-PK Inhibitor, Induces DNA Damage, Apoptosis, and Cell Cycle Arrest in Chronic and Acute Myeloid Leukemia Cells
by Beatriz Santos Lapa, Maria Inês Costa, Diana Figueiredo, Joana Jorge, Raquel Alves, Ana Raquel Monteiro, Beatriz Serambeque, Mafalda Laranjo, Maria Filomena Botelho, Isabel Marques Carreira, Ana Bela Sarmento-Ribeiro and Ana Cristina Gonçalves
Int. J. Mol. Sci. 2023, 24(20), 15331; https://doi.org/10.3390/ijms242015331 - 18 Oct 2023
Cited by 2 | Viewed by 1725
Abstract
The non-homologous end joining pathway is vital for repairing DNA double-strand breaks (DSB), with DNA-dependent protein kinase (DNA-PK) playing a critical role. Altered DNA damage response (DDR) in chronic (CML) and acute myeloid leukemia (AML) offers potential therapeutic opportunities. We studied the therapeutic [...] Read more.
The non-homologous end joining pathway is vital for repairing DNA double-strand breaks (DSB), with DNA-dependent protein kinase (DNA-PK) playing a critical role. Altered DNA damage response (DDR) in chronic (CML) and acute myeloid leukemia (AML) offers potential therapeutic opportunities. We studied the therapeutic potential of AZD-7648 (DNA-PK inhibitor) in CML and AML cell lines. This study used two CML (K-562 and LAMA-84) and five AML (HEL, HL-60, KG-1, NB-4, and THP-1) cell lines. DDR gene mutations were obtained from the COSMIC database. The copy number and methylation profile were evaluated using MS-MLPA and DDR genes, and telomere length using qPCR. p53 protein expression was assessed using Western Blot, chromosomal damage through cytokinesis-block micronucleus assay, and γH2AX levels and DSB repair kinetics using flow cytometry. Cell density and viability were analyzed using trypan blue assay after treatment with AZD-7648 in concentrations ranging from 10 to 200 µM. Cell death, cell cycle distribution, and cell proliferation rate were assessed using flow cytometry. The cells displayed different DNA baseline damage, DDR gene expressions, mutations, genetic/epigenetic changes, and p53 expression. Only HEL cells displayed inefficient DSB repair. The LAMA-84, HEL, and KG-1 cells were the most sensitive to AZD-7648, whereas HL-60 and K-562 showed a lower effect on density and viability. Besides the reduction in cell proliferation, AZD-7648 induced apoptosis, cell cycle arrest, and DNA damage. In conclusion, these results suggest that AZD-7648 holds promise as a potential therapy for myeloid leukemias, however, with variations in drug sensitivity among tested cell lines, thus supporting further investigation to identify the specific factors influencing sensitivity to this DNA-PK inhibitor. Full article
(This article belongs to the Special Issue DNA Damage, DNA Repair, and Cancer 2.0)
Show Figures

Figure 1

20 pages, 4479 KiB  
Article
Chemical Inhibition of RPA by HAMNO Alters Cell Cycle Dynamics by Impeding DNA Replication and G2-to-M Transition but Has Little Effect on the Radiation-Induced DNA Damage Response
by Rositsa Dueva, Lisa Marie Krieger, Fanghua Li, Daxian Luo, Huaping Xiao, Martin Stuschke, Eric Metzen and George Iliakis
Int. J. Mol. Sci. 2023, 24(19), 14941; https://doi.org/10.3390/ijms241914941 - 6 Oct 2023
Cited by 2 | Viewed by 1171
Abstract
Replication protein A (RPA) is the major single-stranded DNA (ssDNA) binding protein that is essential for DNA replication and processing of DNA double-strand breaks (DSBs) by homology-directed repair pathways. Recently, small molecule inhibitors have been developed targeting the RPA70 subunit and preventing RPA [...] Read more.
Replication protein A (RPA) is the major single-stranded DNA (ssDNA) binding protein that is essential for DNA replication and processing of DNA double-strand breaks (DSBs) by homology-directed repair pathways. Recently, small molecule inhibitors have been developed targeting the RPA70 subunit and preventing RPA interactions with ssDNA and various DNA repair proteins. The rationale of this development is the potential utility of such compounds as cancer therapeutics, owing to their ability to inhibit DNA replication that sustains tumor growth. Among these compounds, (1Z)-1-[(2-hydroxyanilino) methylidene] naphthalen-2-one (HAMNO) has been more extensively studied and its efficacy against tumor growth was shown to arise from the associated DNA replication stress. Here, we study the effects of HAMNO on cells exposed to ionizing radiation (IR), focusing on the effects on the DNA damage response and the processing of DSBs and explore its potential as a radiosensitizer. We show that HAMNO by itself slows down the progression of cells through the cell cycle by dramatically decreasing DNA synthesis. Notably, HAMNO also attenuates the progression of G2-phase cells into mitosis by a mechanism that remains to be elucidated. Furthermore, HAMNO increases the fraction of chromatin-bound RPA in S-phase but not in G2-phase cells and suppresses DSB repair by homologous recombination. Despite these marked effects on the cell cycle and the DNA damage response, radiosensitization could neither be detected in exponentially growing cultures, nor in cultures enriched in G2-phase cells. Our results complement existing data on RPA inhibitors, specifically HAMNO, and suggest that their antitumor activity by replication stress induction may not extend to radiosensitization. However, it may render cells more vulnerable to other forms of DNA damaging agents through synthetically lethal interactions, which requires further investigation. Full article
(This article belongs to the Special Issue DNA Damage, DNA Repair, and Cancer 2.0)
Show Figures

Figure 1

21 pages, 4558 KiB  
Article
Transcriptomic Analysis of CRISPR/Cas9-Mediated PARP1-Knockout Cells under the Influence of Topotecan and TDP1 Inhibitor
by Nadezhda S. Dyrkheeva, Anastasia A. Malakhova, Aleksandra L. Zakharenko, Larisa S. Okorokova, Dmitriy N. Shtokalo, Sophia V. Pavlova, Sergey P. Medvedev, Suren M. Zakian, Anna A. Nushtaeva, Alexey E. Tupikin, Marsel R. Kabilov, Svetlana N. Khodyreva, Olga A. Luzina, Nariman F. Salakhutdinov and Olga I. Lavrik
Int. J. Mol. Sci. 2023, 24(6), 5148; https://doi.org/10.3390/ijms24065148 - 7 Mar 2023
Cited by 3 | Viewed by 2236
Abstract
Topoisomerase 1 (TOP1) is an enzyme that regulates DNA topology and is essential for replication, recombination, and other processes. The normal TOP1 catalytic cycle involves the formation of a short-lived covalent complex with the 3′ end of DNA (TOP1 cleavage complex, TOP1cc), which [...] Read more.
Topoisomerase 1 (TOP1) is an enzyme that regulates DNA topology and is essential for replication, recombination, and other processes. The normal TOP1 catalytic cycle involves the formation of a short-lived covalent complex with the 3′ end of DNA (TOP1 cleavage complex, TOP1cc), which can be stabilized, resulting in cell death. This fact substantiates the effectiveness of anticancer drugs—TOP1 poisons, such as topotecan, that block the relegation of DNA and fix TOP1cc. Tyrosyl-DNA phosphodiesterase 1 (TDP1) is able to eliminate TOP1cc. Thus, TDP1 interferes with the action of topotecan. Poly(ADP-ribose) polymerase 1 (PARP1) is a key regulator of many processes in the cell, such as maintaining the integrity of the genome, regulation of the cell cycle, cell death, and others. PARP1 also controls the repair of TOP1cc. We performed a transcriptomic analysis of wild type and PARP1 knockout HEK293A cells treated with topotecan and TDP1 inhibitor OL9-119 alone and in combination. The largest number of differentially expressed genes (DEGs, about 4000 both up- and down-regulated genes) was found in knockout cells. Topotecan and OL9-119 treatment elicited significantly fewer DEGs in WT cells and negligible DEGs in PARP1-KO cells. A significant part of the changes caused by PARP1-KO affected the synthesis and processing of proteins. Differences under the action of treatment with TOP1 or TDP1 inhibitors alone were found in the signaling pathways for the development of cancer, DNA repair, and the proteasome. The drug combination resulted in DEGs in the ribosome, proteasome, spliceosome, and oxidative phosphorylation pathways. Full article
(This article belongs to the Special Issue DNA Damage, DNA Repair, and Cancer 2.0)
Show Figures

Figure 1

18 pages, 3460 KiB  
Article
VRK1 Kinase Activity Modulating Histone H4K16 Acetylation Inhibited by SIRT2 and VRK-IN-1
by Eva Monte-Serrano and Pedro A. Lazo
Int. J. Mol. Sci. 2023, 24(5), 4912; https://doi.org/10.3390/ijms24054912 - 3 Mar 2023
Cited by 6 | Viewed by 2528
Abstract
The accessibility of DNA to different cellular functions requires a dynamic regulation of chromatin organization that is mediated by different epigenetic modifications, which regulate chromatin accessibility and degree of compaction. These epigenetic modifications, particularly the acetylation of histone H4 in lysine 14 (H4K16ac), [...] Read more.
The accessibility of DNA to different cellular functions requires a dynamic regulation of chromatin organization that is mediated by different epigenetic modifications, which regulate chromatin accessibility and degree of compaction. These epigenetic modifications, particularly the acetylation of histone H4 in lysine 14 (H4K16ac), determine the degree of chromatin accessibility to different nuclear functions, as well as to DNA damage drugs. H4K16ac is regulated by the balance between two alternative histone modifications, acetylation and deacetylation, which are mediated by acetylases and deacetylases. Tip60/KAT5 acetylates, and SIRT2 deacetylates histone H4K16. However, the balance between these two epigenetic enzymes is unknown. VRK1 regulates the level of H4K16 acetylation by activating Tip60. We have shown that the VRK1 and SIRT2 are able to form a stable protein complex. For this work, we used in vitro interaction, pull-down and in vitro kinase assays. In cells, their interaction and colocalization were detected by immunoprecipitation and immunofluorescence. The kinase activity of VRK1 is inhibited by a direct interaction of its N-terminal kinase domain with SIRT2 in vitro. This interaction causes a loss of H4K16ac similarly to the effect of a novel VRK1 inhibitor (VRK-IN-1) or VRK1 depletion. The use of specific SIRT2 inhibitors in lung adenocarcinoma cells induces H4K16ac, contrary to the novel VRK-IN-1 inhibitor, which prevents H4K16ac and a correct DNA damage response. Therefore, the inhibition of SIRT2 can cooperate with VRK1 in the accessibility of drugs to chromatin in response to DNA damage caused by doxorubicin. Full article
(This article belongs to the Special Issue DNA Damage, DNA Repair, and Cancer 2.0)
Show Figures

Figure 1

12 pages, 3813 KiB  
Article
The Combination of Panobinostat and Melphalan for the Treatment of Patients with Multiple Myeloma
by Maria Gkotzamanidou, Evangelos Terpos, Meletios A. Dimopoulos and Vassilis L. Souliotis
Int. J. Mol. Sci. 2022, 23(24), 15671; https://doi.org/10.3390/ijms232415671 - 10 Dec 2022
Cited by 3 | Viewed by 1665
Abstract
Histone deacetylase inhibitors show synergy with several genotoxic drugs. Herein, we investigated the biological impact of the combined treatment of panobinostat and melphalan in multiple myeloma (MM). DNA damage response (DDR) parameters and the expression of DDR-associated genes were analyzed in bone marrow [...] Read more.
Histone deacetylase inhibitors show synergy with several genotoxic drugs. Herein, we investigated the biological impact of the combined treatment of panobinostat and melphalan in multiple myeloma (MM). DNA damage response (DDR) parameters and the expression of DDR-associated genes were analyzed in bone marrow plasma cells (BMPCs) and peripheral blood mononuclear cells (PBMCs) from 26 newly diagnosed MM patients. PBMCs from 25 healthy controls (HC) were examined in parallel. Compared with the ex vivo melphalan-only treatment, combined treatment with panobinostat and melphalan significantly reduced the efficiency of nucleotide excision repair (NER) and double-strand-break repair (DSB/R), enhanced the accumulation of DNA lesions (monoadducts and DSBs), and increased the apoptosis rate only in patients’ BMPCs (all p < 0.001); marginal changes were observed in PBMCs from the same patients or HC. Accordingly, panobinostat pre-treatment decreased the expression levels of critical NER (DDB2, XPC) and DSB/R (MRE11A, PRKDC/DNAPKc, RAD50, XRCC6/Ku70) genes only in patients’ BMPCs; no significant changes were observed in PBMCs from patients or HC. Together, our findings demonstrate that panobinostat significantly increased the melphalan sensitivity of malignant BMPCs without increasing the melphalan sensitivity of PBMCs from the same patients, thus paving the way for combination therapies in MM with improved anti-myeloma efficacy and lower side effects. Full article
(This article belongs to the Special Issue DNA Damage, DNA Repair, and Cancer 2.0)
Show Figures

Graphical abstract

Review

Jump to: Editorial, Research

14 pages, 1600 KiB  
Review
The Initial Hepatitis B Virus-Hepatocyte Genomic Integrations and Their Role in Hepatocellular Oncogenesis
by Tomasz I. Michalak
Int. J. Mol. Sci. 2023, 24(19), 14849; https://doi.org/10.3390/ijms241914849 - 3 Oct 2023
Cited by 2 | Viewed by 1364
Abstract
Hepatitis B virus (HBV) remains a dominant cause of hepatocellular carcinoma (HCC). Recently, it was shown that HBV and woodchuck hepatitis virus (WHV) integrate into the hepatocyte genome minutes after invasion. Retrotransposons and transposable sequences were frequent sites of the initial insertions, suggesting [...] Read more.
Hepatitis B virus (HBV) remains a dominant cause of hepatocellular carcinoma (HCC). Recently, it was shown that HBV and woodchuck hepatitis virus (WHV) integrate into the hepatocyte genome minutes after invasion. Retrotransposons and transposable sequences were frequent sites of the initial insertions, suggesting a mechanism for spontaneous HBV DNA dispersal throughout the hepatocyte genome. Several somatic genes were also identified as early insertional targets in infected hepatocytes and woodchuck livers. Head-to-tail joints (HTJs) dominated amongst fusions, indicating their creation by non-homologous end-joining (NHEJ). Their formation coincided with the robust oxidative damage of hepatocyte DNA. This was associated with the activation of poly(ADP-ribose) polymerase 1 (PARP1)-mediated dsDNA repair, as reflected by the augmented transcription of PARP1 and XRCC1; the PARP1 binding partner OGG1, a responder to oxidative DNA damage; and increased activity of NAD+, a marker of PARP1 activation, and HO1, an indicator of cell oxidative stress. The engagement of the PARP1-mediated NHEJ repair pathway explains the HTJ format of the initial merges. The findings show that HBV and WHV are immediate inducers of oxidative DNA damage and hijack dsDNA repair to integrate into the hepatocyte genome, and through this mechanism, they may initiate pro-oncogenic processes. Tracking initial integrations may uncover early markers of HCC and help to explain HBV-associated oncogenesis. Full article
(This article belongs to the Special Issue DNA Damage, DNA Repair, and Cancer 2.0)
Show Figures

Figure 1

19 pages, 1294 KiB  
Review
Poly(ADP-ribose) Polyremase-1 (PARP-1) Inhibition: A Promising Therapeutic Strategy for ETS-Expressing Tumours
by Arnaud J. Legrand, Souhaila Choul-li, Vincent Villeret and Marc Aumercier
Int. J. Mol. Sci. 2023, 24(17), 13454; https://doi.org/10.3390/ijms241713454 - 30 Aug 2023
Cited by 1 | Viewed by 1409
Abstract
ETS transcription factors are a highly conserved family of proteins involved in the progression of many cancers, such as breast and prostate carcinomas, Ewing’s sarcoma, and leukaemias. This significant involvement can be explained by their roles at all stages of carcinogenesis progression. Generally, [...] Read more.
ETS transcription factors are a highly conserved family of proteins involved in the progression of many cancers, such as breast and prostate carcinomas, Ewing’s sarcoma, and leukaemias. This significant involvement can be explained by their roles at all stages of carcinogenesis progression. Generally, their expression in tumours is associated with a poor prognosis and an aggressive phenotype. Until now, no efficient therapeutic strategy had emerged to specifically target ETS-expressing tumours. Nevertheless, there is evidence that pharmacological inhibition of poly(ADP-ribose) polymerase-1 (PARP-1), a key DNA repair enzyme, specifically sensitises ETS-expressing cancer cells to DNA damage and limits tumour progression by leading some of the cancer cells to death. These effects result from a strong interplay between ETS transcription factors and the PARP-1 enzyme. This review summarises the existing knowledge of this molecular interaction and discusses the promising therapeutic applications. Full article
(This article belongs to the Special Issue DNA Damage, DNA Repair, and Cancer 2.0)
Show Figures

Figure 1

Back to TopTop