scholarly journals The cell cycle restricts activation-induced cytidine deaminase activity to early G1

2016 ◽  
Vol 214 (1) ◽  
pp. 49-58 ◽  
Author(s):  
Qiao Wang ◽  
Kyong-Rim Kieffer-Kwon ◽  
Thiago Y. Oliveira ◽  
Christian T. Mayer ◽  
Kaihui Yao ◽  
...  

Activation-induced cytidine deaminase (AID) converts cytosine into uracil to initiate somatic hypermutation (SHM) and class switch recombination (CSR) of antibody genes. In addition, this enzyme produces DNA lesions at off-target sites that lead to mutations and chromosome translocations. However, AID is mostly cytoplasmic, and how and exactly when it accesses nuclear DNA remains enigmatic. Here, we show that AID is transiently in spatial contact with genomic DNA from the time the nuclear membrane breaks down in prometaphase until early G1, when it is actively exported into the cytoplasm. Consistent with this observation, the immunoglobulin (Igh) gene deamination as measured by uracil accumulation occurs primarily in early G1 after chromosomes decondense. Altering the timing of cell cycle–regulated AID nuclear residence increases DNA damage at off-target sites. Thus, the cell cycle–controlled breakdown and reassembly of the nuclear membrane and the restoration of transcription after mitosis constitute an essential time window for AID-induced deamination, and provide a novel DNA damage mechanism restricted to early G1.

Cancers ◽  
2021 ◽  
Vol 13 (3) ◽  
pp. 479
Author(s):  
Pavel Vodicka ◽  
Ladislav Andera ◽  
Alena Opattova ◽  
Ludmila Vodickova

The disruption of genomic integrity due to the accumulation of various kinds of DNA damage, deficient DNA repair capacity, and telomere shortening constitute the hallmarks of malignant diseases. DNA damage response (DDR) is a signaling network to process DNA damage with importance for both cancer development and chemotherapy outcome. DDR represents the complex events that detect DNA lesions and activate signaling networks (cell cycle checkpoint induction, DNA repair, and induction of cell death). TP53, the guardian of the genome, governs the cell response, resulting in cell cycle arrest, DNA damage repair, apoptosis, and senescence. The mutational status of TP53 has an impact on DDR, and somatic mutations in this gene represent one of the critical events in human carcinogenesis. Telomere dysfunction in cells that lack p53-mediated surveillance of genomic integrity along with the involvement of DNA repair in telomeric DNA regions leads to genomic instability. While the role of individual players (DDR, telomere homeostasis, and TP53) in human cancers has attracted attention for some time, there is insufficient understanding of the interactions between these pathways. Since solid cancer is a complex and multifactorial disease with considerable inter- and intra-tumor heterogeneity, we mainly dedicated this review to the interactions of DNA repair, telomere homeostasis, and TP53 mutational status, in relation to (a) cancer risk, (b) cancer progression, and (c) cancer therapy.


2020 ◽  
Vol 21 (11) ◽  
pp. 4127
Author(s):  
Xu Han ◽  
James Kapaldo ◽  
Yueying Liu ◽  
M. Sharon Stack ◽  
Elahe Alizadeh ◽  
...  

The effective clinical application of atmospheric pressure plasma jet (APPJ) treatments requires a well-founded methodology that can describe the interactions between the plasma jet and a treated sample and the temporal and spatial changes that result from the treatment. In this study, we developed a large-scale image analysis method to identify the cell-cycle stage and quantify damage to nuclear DNA in single cells. The method was then tested and used to examine spatio-temporal distributions of nuclear DNA damage in two cell lines from the same anatomic location, namely the oral cavity, after treatment with a nitrogen APPJ. One cell line was malignant, and the other, nonmalignant. The results showed that DNA damage in cancer cells was maximized at the plasma jet treatment region, where the APPJ directly contacted the sample, and declined radially outward. As incubation continued, DNA damage in cancer cells decreased slightly over the first 4 h before rapidly decreasing by approximately 60% at 8 h post-treatment. In nonmalignant cells, no damage was observed within 1 h after treatment, but damage was detected 2 h after treatment. Notably, the damage was 5-fold less than that detected in irradiated cancer cells. Moreover, examining damage with respect to the cell cycle showed that S phase cells were more susceptible to DNA damage than either G1 or G2 phase cells. The proposed methodology for large-scale image analysis is not limited to APPJ post-treatment applications and can be utilized to evaluate biological samples affected by any type of radiation, and, more so, the cell-cycle classification can be used on any cell type with any nuclear DNA staining.


2006 ◽  
Vol 26 (1) ◽  
pp. 39-49 ◽  
Author(s):  
Guochun Jiang ◽  
Aziz Sancar

ABSTRACT We developed a chromatin immunoprecipitation method for analyzing the binding of repair and checkpoint proteins to DNA base lesions in any region of the human genome. Using this method, we investigated the recruitment of DNA damage checkpoint proteins RPA, Rad9, and ATR to base damage induced by UV and acetoxyacetylaminofluorene in transcribed and nontranscribed regions in wild-type and excision repair-deficient human cells in G1 and S phases of the cell cycle. We find that all 3 damage sensors tested assemble at the site or in the vicinity of damage in the absence of DNA replication or repair and that transcription enhances recruitment of checkpoint proteins to the damage site. Furthermore, we find that UV irradiation of human cells defective in excision repair leads to phosphorylation of Chk1 kinase in both G1 and S phase of the cell cycle, suggesting that primary DNA lesions as well as stalled transcription complexes may act as signals to initiate the DNA damage checkpoint response.


2007 ◽  
Vol 27 (19) ◽  
pp. 6852-6862 ◽  
Author(s):  
Aimin Peng ◽  
Andrea L. Lewellyn ◽  
James L. Maller

ABSTRACT In Xenopus laevis embryos, the midblastula transition (MBT) at the 12th cell division marks initiation of critical developmental events, including zygotic transcription and the abrupt inclusion of gap phases into the cell cycle. Interestingly, although an ionizing radiation-induced checkpoint response is absent in pre-MBT embryos, introduction of a threshold amount of undamaged plasmid or sperm DNA allows a DNA damage checkpoint response to be activated. We show here that undamaged threshold DNA directly participates in checkpoint signaling, as judged by several dynamic changes, including H2AX phosphorylation, ATM phosphorylation and loading onto chromatin, and Chk1/Chk2 phosphorylation and release from nuclear DNA. These responses on physically separate threshold DNA require γ-H2AX and are triggered by an ATM-dependent soluble signal initiated by damaged DNA. The signal persists in egg extracts even after damaged DNA is removed from the system, indicating that the absence of damaged DNA is not sufficient to end the checkpoint response. The results identify a novel mechanism by which undamaged DNA enhances checkpoint signaling and provide an example of how the transition to cell cycle checkpoint activation during development is accomplished by maternally programmed increases in the DNA-to-cytoplasm ratio.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3444-3444
Author(s):  
Bay T Ho ◽  
Shyh-Jen Shih ◽  
Sheetal P Singh ◽  
Manuel O Diaz ◽  
Dawei Lin ◽  
...  

Abstract Abstract 3444 The MLL gene, encoding a histone methyl transferase, fuses with multiple different partner genes and is a common finding in patients with AML and ALL. This multiplicity of partners is in stark contrast to other fusion oncogenes such as BCR-ABL in CML or PML-RARA in APL, where fusions between the same genes predominate and which has aided the design of targeted therapy. In order to understand the fusion process in more detail, a hot spot for both MLL cleavage and gene fusion adjacent to exon 12 was examined in a series of breast cancer and lymphoma patients, using inverse PCR (IPCR). All patients received chemotherapy containing drugs targeting Topoisomerase II and samples of blood were examined both before and after therapy. Of the fifty patients enrolled in the study a subset was also examined by IPCR, parallel sequencing and custom bioinformatic analysis. The advantage of this approach is that all possible rearrangements may be examined within a single sample. In addition, the technique also records all sequences at the same location that are uninvolved in a translocation. The blood of three out of four patients examined in this way contained no evidence of MLL rearrangements. However in one patient, with a diagnosis of diffuse large B cell lymphoma, a total of thirteen MLL rearrangements were identified that were present prior to therapy. The majority of the fusions were detected for up to 6 months after the end of chemotherapy indicating they were likely of clonal origin. Of the thirteen fusions, five were predicted to provide functional fusion proteins and these included MLL-MLLT3 (AF9), the most common MLL fusion in myeloid leukemia. The remaining fusions predicted to generate functional proteins involved USP46, FER1L5, CCNJL and NKD1. None of these have been previously identified in clinical specimens of MLL linked disease. However, NKD1 is a negative regulator of the WNT pathway that has been linked to the maintenance of the stem cell phenotype in AML. In order to understand the fusion process in more detail, each fusion breakpoint was examined. All thirteen MLL fusions contained microhomology at the breakpoint, ranging from 1 to 6 bp, indicating NHEJ as the likely pathway generating the fusions. Though the presence of microhomology masked the precise breakpoint, using the 3' edge of microhomology as a reference, eight of the thirteen rearrangements were clustered within a 5 bp tract at the base of a putative stem-loop structure, as we have reported before. Such a restricted distribution of breakpoints found within a clonal population suggests a common mechanism is involved in either cleavage, and/or fusion, at this location. In order to address the mechanism driving these rearrangements, a screen of all sequenced material was undertaken. It was observed that within the residual sequenced material, a selective increase in C>T transitions were noted for two cytosines located within the 5bp breakpoint hotspot. The level of C>T transition was 50–100 fold higher than anywhere else within the sequence generated by IPCR. In addition, the region of C>T transition contained the WRC (A/T, A/G, C) motif characteristic of Activation-Induced Cytidine Deaminase (AID) attack. AID deaminates cytosine leading to a uracil which may be repaired by uracil-DNA glycosylase and the base excision repair (BER) pathway. AID has been implicated in both DNA breaks, via the BER pathway, and a linked increase in translocations. If the BER pathway fails to execute appropriately, a C>T transition may occur as the aberrantly located uracil undergoes replication at the next division. The presence of extensive C>T transitions therefore is indicative of both AID function and defective repair pathway(s). These factors may explain both the extensive number of rearrangements observed in this patient and the legacy C>T transitions from multiple AID attacks. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 113 (suppl_1) ◽  
Author(s):  
Ramaswamy Kannappan ◽  
Giorgia Palano ◽  
Polina Goichberg ◽  
Fumihiro Sanada ◽  
Sergio Signore ◽  
...  

Doxorubicin (DOXO) causes dilated cardiomyopathy and heart failure. We have documented previously that DOXO-mediated cardiotoxicity is dictated by functional alterations of cardiac stem cells (CSCs). DOXO-induced myopathy was coupled with a reduction in CSC number due to increased death, inhibition of proliferation, and senescence. We raised the possibility that survival and growth of CSCs following DOXO treatment may be enhanced by modulating the intracellular level of p53, which plays a critical role in the determination of stem cell fate. For this purpose, transgenic mice carrying an additional p53 allele (Sp53) were studied. With respect to wild-type mice (WT), CSCs isolated from Sp53 mice (Sp53-CSCs) showed increased apoptosis with accumulation of the pro-apoptotic p53 targets BAX, PUMA and Pidd. Conversely, the expression of p21Cip1, a cell cycle inhibitor and inducer of cell senescence, was lower in Sp53-CSCs than WT cells. Upon DOXO treatment, Sp53-CSCs exhibited accelerated onset of apoptosis. However, viable Sp53-CSCs showed enhanced formation of DNA damage response foci, indicative of a very efficient DNA repair mechanism. Following removal of DOXO, Sp53-CSCs re-entered the cell cycle and divided, while WT cells continued to die by apoptosis or became senescent. The response of WT-CSCs to DOXO involved the pro-apoptotic Bcl2 family member Noxa and the senescence-associated protein p16INK4a. In contrast, exposure of Sp53-CSCs to DOXO provoked pulses of p53 expression, which favored sustained upregulation of Mdm2. Mdm2 antagonized the inhibitory effect of p53 on cell growth and prevented apoptosis. Ultimately, Sp53-CSCs showed accumulation of PCNA, which is required for DNA repair and synthesis. Importantly, IGF-1 release was higher in Sp53-CSCs, promoting their replication through an autocrine-paracrine mechanism. Collectively, our data demonstrate that changes in the pattern of p53 expression have beneficial effects on CSCs by amplifying the DNA repair response, facilitating the clearance of cells with non-repairable DNA damage, and enabling the proliferation of cells in which DNA lesions are effectively removed. Thus, targeting p53 expression in CSCs may protect the heart from the toxic effects of chemotherapy.


2004 ◽  
Vol 24 (23) ◽  
pp. 10126-10144 ◽  
Author(s):  
Michela Clerici ◽  
Veronica Baldo ◽  
Davide Mantiero ◽  
Francisca Lottersberger ◽  
Giovanna Lucchini ◽  
...  

ABSTRACT In Saccharomyces cerevisiae, Mec1/ATR plays a primary role in sensing and transducing checkpoint signals in response to different types of DNA lesions, while the role of the Tel1/ATM kinase in DNA damage checkpoints is not as well defined. We found that UV irradiation in G1 in the absence of Mec1 activates a Tel1/MRX-dependent checkpoint, which specifically inhibits the metaphase-to-anaphase transition. Activation of this checkpoint leads to phosphorylation of the downstream checkpoint kinases Rad53 and Chk1, which are required for Tel1-dependent cell cycle arrest, and their adaptor Rad9. The spindle assembly checkpoint protein Mad2 also partially contributes to the G2/M arrest of UV-irradiated mec1Δ cells independently of Rad53 phosphorylation and activation. The inability of UV-irradiated mec1Δ cells to undergo anaphase can be relieved by eliminating the anaphase inhibitor Pds1, whose phosphorylation and stabilization in these cells depend on Tel1, suggesting that Pds1 persistence may be responsible for the inability to undergo anaphase. Moreover, while UV irradiation can trigger Mec1-dependent Rad53 phosphorylation and activation in G1- and G2-arrested cells, Tel1-dependent checkpoint activation requires entry into S phase independently of the cell cycle phase at which cells are UV irradiated, and it is decreased when single-stranded DNA signaling is affected by the rfa1-t11 allele. This indicates that UV-damaged DNA molecules need to undergo structural changes in order to activate the Tel1-dependent checkpoint. Active Clb-cyclin-dependent kinase 1 (CDK1) complexes also participate in triggering this checkpoint and are required to maintain both Mec1- and Tel1-dependent Rad53 phosphorylation, suggesting that they may provide critical phosphorylation events in the DNA damage checkpoint cascade.


Sign in / Sign up

Export Citation Format

Share Document