ijms-logo

Journal Browser

Journal Browser

DNA Replication Stress and Chromosomal Instability

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Pathology, Diagnostics, and Therapeutics".

Deadline for manuscript submissions: closed (31 March 2021) | Viewed by 33214

Special Issue Editors


E-Mail Website
Guest Editor
Institute of Human Genetics (IGH), Univ Montpellier, CNRS UMR 9002 Montpellier, France
Interests: DNA replication stress; DNA signaling and repair; ATR/Chk1 pathway; TopBP1 condensate
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Institute of Human Genetics (IGH), Univ Montpellier, CNRS UMR 9002 Montpellier, France
Interests: DNA replication, telomeres, nuclear structure, DNA damage response
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Faithful duplication of the genetic information during DNA synthesis is essential for the cell to maintain its genome integrity. DNA lesions generated by exogenous or endogenous insults perturb DNA synthesis and lead to the stalling of replication forks. Interference with DNA replication causes DNA replication stress, which is a well-established hallmark of cancer and a source of genomic instability.  

Eukaryotic cells have evolved a sophisticated DNA damage response (DDR) mechanism to tackle the threats caused by lesions during DNA synthesis in order to avoid replication forks collapsing. Due to their high level of replicative stress, cancer cells rely more on DDR to survive than normal cells. Therefore, targeting the DDR pathway is extensively studied to improve cancer therapies. Thus, there is a need to increase our knowledge on how cells deal with replicative stress.

In the recent years, new sources of DNA replicative stress have been uncovered (DNA-protein crosslinks, mechanical stress, etc.) as well as crosstalks with other pathways (splicing, aldehydes metabolism, etc). This Special Issue aims to explore in depth how these diverse threats are detected and processed by the cells and to study their impact on genome integrity and cancer.

Dr. Jihane Basbous
Dr. Cyril Ribeyre
Guest Editors

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 
  • DNA replication stress 
  • DNA repair 
  • Checkpoint activation 
  • Genomic instability 
  • Tumorigenesis 
  • Chemotherapies

Published Papers (8 papers)

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

Research

Jump to: Review

21 pages, 1763 KiB  
Article
Proteins Marking the Sequence of Genotoxic Signaling from Irradiated Mesenchymal Stromal Cells to CD34+ Cells
by Vanessa Kohl, Oliver Drews, Victor Costina, Miriam Bierbaum, Ahmed Jawhar, Henning Roehl, Christel Weiss, Susanne Brendel, Helga Kleiner, Johanna Flach, Birgit Spiess, Wolfgang Seifarth, Daniel Nowak, Wolf-Karsten Hofmann, Alice Fabarius and Henning D. Popp
Int. J. Mol. Sci. 2021, 22(11), 5844; https://doi.org/10.3390/ijms22115844 - 29 May 2021
Cited by 2 | Viewed by 2770
Abstract
Non-targeted effects (NTE) of ionizing radiation may initiate myeloid neoplasms (MN). Here, protein mediators (I) in irradiated human mesenchymal stromal cells (MSC) as the NTE source, (II) in MSC conditioned supernatant and (III) in human bone marrow CD34+ cells undergoing genotoxic NTE were [...] Read more.
Non-targeted effects (NTE) of ionizing radiation may initiate myeloid neoplasms (MN). Here, protein mediators (I) in irradiated human mesenchymal stromal cells (MSC) as the NTE source, (II) in MSC conditioned supernatant and (III) in human bone marrow CD34+ cells undergoing genotoxic NTE were investigated. Healthy sublethal irradiated MSC showed significantly increased levels of reactive oxygen species. These cells responded by increasing intracellular abundance of proteins involved in proteasomal degradation, protein translation, cytoskeleton dynamics, nucleocytoplasmic shuttling, and those with antioxidant activity. Among the increased proteins were THY1 and GNA11/14, which are signaling proteins with hitherto unknown functions in the radiation response and NTE. In the corresponding MSC conditioned medium, the three chaperones GRP78, CALR, and PDIA3 were increased. Together with GPI, these were the only four altered proteins, which were associated with the observed genotoxic NTE. Healthy CD34+ cells cultured in MSC conditioned medium suffered from more than a six-fold increase in γH2AX focal staining, indicative for DNA double-strand breaks, as well as numerical and structural chromosomal aberrations within three days. At this stage, five proteins were altered, among them IQGAP1, HMGB1, and PA2G4, which are involved in malign development. In summary, our data provide novel insights into three sequential steps of genotoxic signaling from irradiated MSC to CD34+ cells, implicating that induced NTE might initiate the development of MN. Full article
(This article belongs to the Special Issue DNA Replication Stress and Chromosomal Instability)
Show Figures

Figure 1

21 pages, 3367 KiB  
Article
Low Replicative Stress Triggers Cell-Type Specific Inheritable Advanced Replication Timing
by Lilas Courtot, Elodie Bournique, Chrystelle Maric, Laure Guitton-Sert, Miguel Madrid-Mencía, Vera Pancaldi, Jean-Charles Cadoret, Jean-Sébastien Hoffmann and Valérie Bergoglio
Int. J. Mol. Sci. 2021, 22(9), 4959; https://doi.org/10.3390/ijms22094959 - 07 May 2021
Cited by 3 | Viewed by 3631
Abstract
DNA replication timing (RT), reflecting the temporal order of origin activation, is known as a robust and conserved cell-type specific process. Upon low replication stress, the slowing of replication forks induces well-documented RT delays associated to genetic instability, but it can also generate [...] Read more.
DNA replication timing (RT), reflecting the temporal order of origin activation, is known as a robust and conserved cell-type specific process. Upon low replication stress, the slowing of replication forks induces well-documented RT delays associated to genetic instability, but it can also generate RT advances that are still uncharacterized. In order to characterize these advanced initiation events, we monitored the whole genome RT from six independent human cell lines treated with low doses of aphidicolin. We report that RT advances are cell-type-specific and involve large heterochromatin domains. Importantly, we found that some major late to early RT advances can be inherited by the unstressed next-cellular generation, which is a unique process that correlates with enhanced chromatin accessibility, as well as modified replication origin landscape and gene expression in daughter cells. Collectively, this work highlights how low replication stress may impact cellular identity by RT advances events at a subset of chromosomal domains. Full article
(This article belongs to the Special Issue DNA Replication Stress and Chromosomal Instability)
Show Figures

Graphical abstract

20 pages, 5528 KiB  
Article
PARP Inhibition Increases the Reliance on ATR/CHK1 Checkpoint Signaling Leading to Synthetic Lethality—An Alternative Treatment Strategy for Epithelial Ovarian Cancer Cells Independent from HR Effectiveness
by Patrycja Gralewska, Arkadiusz Gajek, Agnieszka Marczak, Michał Mikuła, Jerzy Ostrowski, Agnieszka Śliwińska and Aneta Rogalska
Int. J. Mol. Sci. 2020, 21(24), 9715; https://doi.org/10.3390/ijms21249715 - 19 Dec 2020
Cited by 19 | Viewed by 5133
Abstract
Poly (ADP-ribose) polymerase inhibitor (PARPi, olaparib) impairs the repair of DNA single-strand breaks (SSBs), resulting in double-strand breaks (DSBs) that cannot be repaired efficiently in homologous recombination repair (HRR)-deficient cancers such as BRCA1/2-mutant cancers, leading to synthetic lethality. Despite the efficacy of olaparib [...] Read more.
Poly (ADP-ribose) polymerase inhibitor (PARPi, olaparib) impairs the repair of DNA single-strand breaks (SSBs), resulting in double-strand breaks (DSBs) that cannot be repaired efficiently in homologous recombination repair (HRR)-deficient cancers such as BRCA1/2-mutant cancers, leading to synthetic lethality. Despite the efficacy of olaparib in the treatment of BRCA1/2 deficient tumors, PARPi resistance is common. We hypothesized that the combination of olaparib with anticancer agents that disrupt HRR by targeting ataxia telangiectasia and Rad3-related protein (ATR) or checkpoint kinase 1 (CHK1) may be an effective strategy to reverse ovarian cancer resistance to olaparib. Here, we evaluated the effect of olaparib, the ATR inhibitor AZD6738, and the CHK1 inhibitor MK8776 alone and in combination on cell survival, colony formation, replication stress response (RSR) protein expression, DNA damage, and apoptotic changes in BRCA2 mutated (PEO-1) and HRR-proficient BRCA wild-type (SKOV-3 and OV-90) cells. Combined treatment caused the accumulation of DNA DSBs. PARP expression was associated with sensitivity to olaparib or inhibitors of RSR. Synergistic effects were weaker when olaparib was combined with CHK1i and occurred regardless of the BRCA2 status of tumor cells. Because PARPi increases the reliance on ATR/CHK1 for genome stability, the combination of PARPi with ATR inhibition suppressed ovarian cancer cell growth independently of the efficacy of HRR. The present results were obtained at sub-lethal doses, suggesting the potential of these inhibitors as monotherapy as well as in combination with olaparib. Full article
(This article belongs to the Special Issue DNA Replication Stress and Chromosomal Instability)
Show Figures

Graphical abstract

Review

Jump to: Research

21 pages, 19417 KiB  
Review
DNA Damage Response in the Adaptive Arm of the Immune System: Implications for Autoimmunity
by Theodora Manolakou, Panayotis Verginis and Dimitrios T. Boumpas
Int. J. Mol. Sci. 2021, 22(11), 5842; https://doi.org/10.3390/ijms22115842 - 29 May 2021
Cited by 5 | Viewed by 3891
Abstract
In complex environments, cells have developed molecular responses to confront threats against the genome and achieve the maintenance of genomic stability assuring the transfer of undamaged DNA to their progeny. DNA damage response (DDR) mechanisms may be activated upon genotoxic or environmental agents, [...] Read more.
In complex environments, cells have developed molecular responses to confront threats against the genome and achieve the maintenance of genomic stability assuring the transfer of undamaged DNA to their progeny. DNA damage response (DDR) mechanisms may be activated upon genotoxic or environmental agents, such as cytotoxic drugs or ultraviolet (UV) light, and during physiological processes requiring DNA transactions, to restore DNA alterations that may cause cellular malfunction and affect viability. In addition to the DDR, multicellular organisms have evolved specialized immune cells to respond and defend against infections. Both adaptive and innate immune cells are subjected to DDR processes, either as a prerequisite to the immune response, or as a result of random endogenous and exogenous insults. Aberrant DDR activities have been extensively studied in the immune cells of the innate arm, but not in adaptive immune cells. Here, we discuss how the aberrant DDR may lead to autoimmunity, with emphasis on the adaptive immune cells and the potential of therapeutic targeting. Full article
(This article belongs to the Special Issue DNA Replication Stress and Chromosomal Instability)
Show Figures

Figure 1

36 pages, 2107 KiB  
Review
SUMO-Targeted Ubiquitin Ligases and Their Functions in Maintaining Genome Stability
by Ya-Chu Chang, Marissa K. Oram and Anja-Katrin Bielinsky
Int. J. Mol. Sci. 2021, 22(10), 5391; https://doi.org/10.3390/ijms22105391 - 20 May 2021
Cited by 23 | Viewed by 5806
Abstract
Small ubiquitin-like modifier (SUMO)-targeted E3 ubiquitin ligases (STUbLs) are specialized enzymes that recognize SUMOylated proteins and attach ubiquitin to them. They therefore connect the cellular SUMOylation and ubiquitination circuits. STUbLs participate in diverse molecular processes that span cell cycle regulated events, including DNA [...] Read more.
Small ubiquitin-like modifier (SUMO)-targeted E3 ubiquitin ligases (STUbLs) are specialized enzymes that recognize SUMOylated proteins and attach ubiquitin to them. They therefore connect the cellular SUMOylation and ubiquitination circuits. STUbLs participate in diverse molecular processes that span cell cycle regulated events, including DNA repair, replication, mitosis, and transcription. They operate during unperturbed conditions and in response to challenges, such as genotoxic stress. These E3 ubiquitin ligases modify their target substrates by catalyzing ubiquitin chains that form different linkages, resulting in proteolytic or non-proteolytic outcomes. Often, STUbLs function in compartmentalized environments, such as the nuclear envelope or kinetochore, and actively aid in nuclear relocalization of damaged DNA and stalled replication forks to promote DNA repair or fork restart. Furthermore, STUbLs reside in the same vicinity as SUMO proteases and deubiquitinases (DUBs), providing spatiotemporal control of their targets. In this review, we focus on the molecular mechanisms by which STUbLs help to maintain genome stability across different species. Full article
(This article belongs to the Special Issue DNA Replication Stress and Chromosomal Instability)
Show Figures

Figure 1

12 pages, 715 KiB  
Review
Regulation of DNA Replication Licensing and Re-Replication by Cdt1
by Hui Zhang
Int. J. Mol. Sci. 2021, 22(10), 5195; https://doi.org/10.3390/ijms22105195 - 14 May 2021
Cited by 13 | Viewed by 3471
Abstract
In eukaryotic cells, DNA replication licensing is precisely regulated to ensure that the initiation of genomic DNA replication in S phase occurs once and only once for each mitotic cell division. A key regulatory mechanism by which DNA re-replication is suppressed is the [...] Read more.
In eukaryotic cells, DNA replication licensing is precisely regulated to ensure that the initiation of genomic DNA replication in S phase occurs once and only once for each mitotic cell division. A key regulatory mechanism by which DNA re-replication is suppressed is the S phase-dependent proteolysis of Cdt1, an essential replication protein for licensing DNA replication origins by loading the Mcm2-7 replication helicase for DNA duplication in S phase. Cdt1 degradation is mediated by CRL4Cdt2 ubiquitin E3 ligase, which further requires Cdt1 binding to proliferating cell nuclear antigen (PCNA) through a PIP box domain in Cdt1 during DNA synthesis. Recent studies found that Cdt2, the specific subunit of CRL4Cdt2 ubiquitin E3 ligase that targets Cdt1 for degradation, also contains an evolutionarily conserved PIP box-like domain that mediates the interaction with PCNA. These findings suggest that the initiation and elongation of DNA replication or DNA damage-induced repair synthesis provide a novel mechanism by which Cdt1 and CRL4Cdt2 are both recruited onto the trimeric PCNA clamp encircling the replicating DNA strands to promote the interaction between Cdt1 and CRL4Cdt2. The proximity of PCNA-bound Cdt1 to CRL4Cdt2 facilitates the destruction of Cdt1 in response to DNA damage or after DNA replication initiation to prevent DNA re-replication in the cell cycle. CRL4Cdt2 ubiquitin E3 ligase may also regulate the degradation of other PIP box-containing proteins, such as CDK inhibitor p21 and histone methylase Set8, to regulate DNA replication licensing, cell cycle progression, DNA repair, and genome stability by directly interacting with PCNA during DNA replication and repair synthesis. Full article
(This article belongs to the Special Issue DNA Replication Stress and Chromosomal Instability)
Show Figures

Figure 1

26 pages, 1541 KiB  
Review
Replication Stress, Genomic Instability, and Replication Timing: A Complex Relationship
by Lina-Marie Briu, Chrystelle Maric and Jean-Charles Cadoret
Int. J. Mol. Sci. 2021, 22(9), 4764; https://doi.org/10.3390/ijms22094764 - 30 Apr 2021
Cited by 12 | Viewed by 3694
Abstract
The replication-timing program constitutes a key element of the organization and coordination of numerous nuclear processes in eukaryotes. This program is established at a crucial moment in the cell cycle and occurs simultaneously with the organization of the genome, thus indicating the vital [...] Read more.
The replication-timing program constitutes a key element of the organization and coordination of numerous nuclear processes in eukaryotes. This program is established at a crucial moment in the cell cycle and occurs simultaneously with the organization of the genome, thus indicating the vital significance of this process. With recent technological achievements of high-throughput approaches, a very strong link has been confirmed between replication timing, transcriptional activity, the epigenetic and mutational landscape, and the 3D organization of the genome. There is also a clear relationship between replication stress, replication timing, and genomic instability, but the extent to which they are mutually linked to each other is unclear. Recent evidence has shown that replication timing is affected in cancer cells, although the cause and consequence of this effect remain unknown. However, in-depth studies remain to be performed to characterize the molecular mechanisms of replication-timing regulation and clearly identify different cis- and trans-acting factors. The results of these studies will potentially facilitate the discovery of new therapeutic pathways, particularly for personalized medicine, or new biomarkers. This review focuses on the complex relationship between replication timing, replication stress, and genomic instability. Full article
(This article belongs to the Special Issue DNA Replication Stress and Chromosomal Instability)
Show Figures

Figure 1

24 pages, 1695 KiB  
Review
DNA2 in Chromosome Stability and Cell Survival—Is It All about Replication Forks?
by Jessica J. R. Hudson and Ulrich Rass
Int. J. Mol. Sci. 2021, 22(8), 3984; https://doi.org/10.3390/ijms22083984 - 13 Apr 2021
Cited by 7 | Viewed by 3579
Abstract
The conserved nuclease-helicase DNA2 has been linked to mitochondrial myopathy, Seckel syndrome, and cancer. Across species, the protein is indispensable for cell proliferation. On the molecular level, DNA2 has been implicated in DNA double-strand break (DSB) repair, checkpoint activation, Okazaki fragment processing (OFP), [...] Read more.
The conserved nuclease-helicase DNA2 has been linked to mitochondrial myopathy, Seckel syndrome, and cancer. Across species, the protein is indispensable for cell proliferation. On the molecular level, DNA2 has been implicated in DNA double-strand break (DSB) repair, checkpoint activation, Okazaki fragment processing (OFP), and telomere homeostasis. More recently, a critical contribution of DNA2 to the replication stress response and recovery of stalled DNA replication forks (RFs) has emerged. Here, we review the available functional and phenotypic data and propose that the major cellular defects associated with DNA2 dysfunction, and the links that exist with human disease, can be rationalized through the fundamental importance of DNA2-dependent RF recovery to genome duplication. Being a crucial player at stalled RFs, DNA2 is a promising target for anti-cancer therapy aimed at eliminating cancer cells by replication-stress overload. Full article
(This article belongs to the Special Issue DNA Replication Stress and Chromosomal Instability)
Show Figures

Figure 1

Back to TopTop