Abstract 688: Acetylation of poly-ADP-ribose polymerase and Ku70 by histone deacetylase inhibitors promote abnormal binding to DNA double strand breaks and decrease repair efficiency in leukemia cells

Author(s):  
Carine Robert ◽  
Ivana Gojo ◽  
Feyruz Rassool
2020 ◽  
Vol 62 (1) ◽  
pp. 25-33
Author(s):  
Yuki Sakamoto ◽  
Tetsuya Kokuta ◽  
Ai Teshigahara ◽  
Kenta Iijima ◽  
Hiroyuki Kitao ◽  
...  

Abstract The choice of repair pathways of DNA double-strand breaks (DSBs) is dependent upon the cell cycle phases. While homologous recombination repair (HRR) is active between the S and G2 phases, its involvement in mitotic DSB repair has not been examined in detail. In the present study, we developed a new reporter assay system to detect homology-directed repair (HDR), a major pathway used for HRR, in combination with an inducible DSB-generation system. As expected, the maximal HDR activity was observed in the late S phase, along with minimal activity in the G1 phase and at the G1/S boundary. Surprisingly, significant HDR activity was observed in M phase, and the repair efficiency was similar to that observed in late S phase. HDR was also confirmed in metaphase cells collected with continuous colcemid exposure. ChIP assays revealed the recruitment of RAD51 to the vicinity of DSBs in M phase. In addition, the ChIP assay for gamma-H2AX and phosphorylated DNA-PKcs indicated that a part of M-phase cells with DSBs could proceed into the next G1 phase. These results provide evidence showing that a portion of mitotic cell DSBs are undoubtedly repaired through action of the HDR repair pathway.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2662-2662
Author(s):  
Laura Desbourdes ◽  
Nicole Ishac ◽  
Thomas Charbonnier ◽  
Aurore Iltis ◽  
Elfi Ducrocq ◽  
...  

Abstract Introduction The contribution of the stromal cell compartment to leukemogenesis remains poorly understood. Several studies have described abnormalities involving this compartment in acute myeloid leukemias (AML) and myelodysplastic syndromes (MDS) including proliferative defect of mesenchymal stem/stromal cells (MSCs) as reported by our group in AML patients (Domenech et al., Haematologica 2012, EHA meeting, abstr. 37). Recently, evidence of an involvement of MSCs in the leukemic process has been provided in murine models (Walkley et al., Cell 2007; Raaijmakers et al. Nature 2010). In the present study, we investigated potential modifications of human AML blast cell biology induced in vitro by MSCs from healthy individuals and AML patients. Methods Bone marrow MSCs from 6 AML patients (3 M0, 3 M1 [FAB classification]) were compared to those from 6 normal individuals. All the MSCs were analyzed at the end of the second passage of culture. Capacity of MSCs to influence leukemic cell behavior was assessed by co-culture with immature leukemia cells from KG1a line and with heterologous or autologous primary blast cells from the AML patients. Apoptotic cell frequencies, cell cycle phase distributions, and presence of DNA double-strand breaks of leukemia cells with or without MSCs were performed by flow cytometry. Results In co-cultures, no difference in leukemia cell adhesion were found on AML and normal MSCs. The presence of MSCs reduced apoptosis of primary blast cells (-30%, p=0.002) although no effect was observed on campthotecin-induced apoptosis of KG1a cells, as compared to MSC-free culture conditions. Cell cycle analysis revealed that G0/G1 ratios of KG1a cells were strongly increased by the presence of MSCs considering the whole cells (+128%, p=0.005) as well as the cell fraction adherent to MSCs (+138%, p=0.006). Comparable results were obtained with primary blast cells considering all the cells (+51%, p=0.117), especially within the adherent cell fraction (+229%, p=0.004). In addition, direct contact with MSCs was associated with a slight decrease in the proportion of blast cells with DNA double-strand breaks compared to MSC-free cultures (-10%, p= 0.034). No significant differences were found for all these results when co-cultures with AML and normal MSCs were compared. Likewise, no differences existed between co-cultures with M0 and M1 AML MSCs or between co-cultures of autologous and heterologous primary blast cells with AML MSCs. Conclusions This study shows that MSCs influence leukemic cell behavior, irrespective of their healthy or leukemic origin. In particular, they protect blast cells from apoptosis and induce their quiescence, (mainly by direct contact) which could contribute to decreased yields of DNA double-strand breaks, a source of genetic instability. Further experiments are in progress to evaluate potential changes in the capacity of AML MSCs to support normal hematopoiesis. Disclosures: Gyan: FRESENIUS KABI: Consultancy, Research Funding. Domenech:Celgene Corporation: Research Funding.


Cell Cycle ◽  
2006 ◽  
Vol 5 (9) ◽  
pp. 994-1000 ◽  
Author(s):  
Margaret Nieborowska-Skorska ◽  
Tomasz Stoklosa ◽  
Mandrita Datta ◽  
Agnieszka Czechowska ◽  
Lori Rink ◽  
...  

2015 ◽  
Vol 112 (50) ◽  
pp. E6907-E6916 ◽  
Author(s):  
Damon Meyer ◽  
Becky Xu Hua Fu ◽  
Wolf-Dietrich Heyer

Maintenance of genome stability is carried out by a suite of DNA repair pathways that ensure the repair of damaged DNA and faithful replication of the genome. Of particular importance are the repair pathways, which respond to DNA double-strand breaks (DSBs), and how the efficiency of repair is influenced by sequence homology. In this study, we developed a genetic assay in diploid Saccharomyces cerevisiae cells to analyze DSBs requiring microhomologies for repair, known as microhomology-mediated end-joining (MMEJ). MMEJ repair efficiency increased concomitant with microhomology length and decreased upon introduction of mismatches. The central proteins in homologous recombination (HR), Rad52 and Rad51, suppressed MMEJ in this system, suggesting a competition between HR and MMEJ for the repair of a DSB. Importantly, we found that DNA polymerase delta (Pol δ) is critical for MMEJ, independent of microhomology length and base-pairing continuity. MMEJ recombinants showed evidence that Pol δ proofreading function is active during MMEJ-mediated DSB repair. Furthermore, mutations in Pol δ and DNA polymerase 4 (Pol λ), the DNA polymerase previously implicated in MMEJ, cause a synergistic decrease in MMEJ repair. Pol λ showed faster kinetics associating with MMEJ substrates following DSB induction than Pol δ. The association of Pol δ depended on RAD1, which encodes the flap endonuclease needed to cleave MMEJ intermediates before DNA synthesis. Moreover, Pol δ recruitment was diminished in cells lacking Pol λ. These data suggest cooperative involvement of both polymerases in MMEJ.


2012 ◽  
Vol 31 (3) ◽  
pp. 298-305 ◽  
Author(s):  
Anna Walczak ◽  
Pawel Rusin ◽  
Lukasz Dziki ◽  
Hanna Zielinska-Blizniewska ◽  
Jurek Olszewski ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1982-1982
Author(s):  
Artur Slupianek ◽  
Michal O. Nowicki ◽  
Tomasz Skorski

Abstract Clinical observations and experimental findings indicated that BCR/ABL stimulates genomic instability leading to mutations and chromosomal abnormalities. The accumulation of genetic errors is believed to be responsible for the transition from a relatively benign CML chronic phase (CML-CP) to the aggressive blast crisis phase (CML-BC) and the resistance to imatinib mesylate. BCR/ABL- positive leukemia cells accumulate an excess of potentially lethal DNA double-strand breaks (DSBs) caused by reactive oxygen species (ROS) or genotoxic treatment. However, BCR/ABL tyrosine kinase facilitates the repair of DSBs and promotes survival. Therefore, the infidelity of DSBs repair processes may contribute to genomic instability in leukemia cells exposed to elevated numbers of spontaneous and/or induced DSBs. To test this hypothesis DSBs repair efficiency and fidelity was examined and compared in parental and BCR/ABL-transformed cells. Nuclear foci detected by γ-H2AX (the form of H2AX histone that is quickly phosphorylated on Serine 139 by ATM, ATR and/or DNA-PKcs kinases on megabase-length fragments near DSB sites) immunofluorescence served as indicators of DSBs. We found that BCR/ABL-positive leukemia cells acquire more DSBs after γ-irradiation in comparison to normal cells. Homologous recombination (HR) and non-homologous end-joining (NHEJ) represent two major mechanisms of DSBs repair in mammalian cells. HR and NHEJ reaction sites in the nuclei can be visualized by double-immunofluorescence detecting co-localization of γ-H2AX foci with RAD51 or Ku70, respectively. NHEJ and HR appear to work in a time-dependent fashion (NHEJ followed by HR) and be more active in BCR/ABL-transformed cells in comparison to normal counterparts. Time-dependent engagements of NHEJ and HR mechanisms in repair of DSBs after γ-irradiation are accompanied by elevated accumulation of Ku70 and RAD51 proteins in cell lysates obtained from BCR/ABL cells. Specific DSBs repair assays confirmed that BCR/ABL leukemia cells in comparison to normal cells displayed enhanced capability of HR and NHEJ. However, analysis of DSBs repair products revealed that the repair mechanisms were less faithful in former cells generating large deletions and point mutations during NHEJ and HR, respectively. In summary, BCR/ABL leukemia cells display facilitated, but unfaithful HR and NHEJ, which may contribute to accumulation of genetic errors in surviving leukemia cells leading to malignant disease progression.


Sign in / Sign up

Export Citation Format

Share Document