New Therapeutic Strategy for Sensitisation of CLL Cells with Inactivation of the DNA Damage Response by Targeting the Deubiquitylating Enzyme USP7-Dependent Pathways,

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3865-3865
Author(s):  
Angelo Agathanggelou ◽  
Anastasia Zlatanou ◽  
Ahmed Gulshanara ◽  
Grant Stewart ◽  
Pamela R Kearns ◽  
...  

Abstract Abstract 3865 Chronic Lymphocytic Leukaemia (CLL) is characterised by marked clinical heterogeneity and tumours with deletion or mutation of the TP53 and ATM genes on chromosomes 17p and 11q are associated with poor outcome. ATM is a protein kinase that, following induction of DNA double strand breaks (DSBs), phosphorylates a number of downstream targets and synchronises a network of cellular responses including p53 activation that leads to induction of pro-apoptotic genes and activation of apoptosis. Consequently, loss of integrity of ATM/p53 pathway results in apoptotic resistance, retention of cells with genomic damage, and tumour progression. Cellular p53 levels are regulated through a p53-Mdm2 regulatory loop whereby Mdm2, a ubiquitin ligase, facilitates p53 polyubiquitination and targeting of p53 for proteasome degradation. It was recently shown that stabilisation of p53 can be achieved by manipulation of this regulatory loop through use of small molecule inhibitors of this p53 degradation pathway, termed nutlins, which prevent p53 ubiquitination. More recently, a class of deubiquitinating enzymes (DUBs) highlighted an additional level of p53-Mdm2 regulation. In particular, a specific DUB, (USP7/HAUSP), has a high affinity for Mdm2 and functions by antagonizing Mdm2 ubiquitination. Unlike nutlins, USP7 also has been implicated in the regulation of cell cycle, mitosis and DNA damage response and we reasoned that USP7 inhibition may sensitise CLL tumours with ATM and TP53 defects. Our analysis of 25 primary CLL tumours with different ATM and TP53 status indicated that USP7 was robustly expressed in all CLL tumour cells tested. Through collaboration with Hybrigenics we obtained a specific USP7 inhibitor, HBX19818. To determine if cell killing could be induced in ATM or TP53 deficient tumours by inhibiting the USP7-Mdm2 pathway, we analysed the induction of cell death over a range of HBX19818 concentrations using both isogenic CLL cell lines with and without ATM and/or p53, as well as 18 primary CLL tumours. We observed a significant cytotoxic effect of HBX19818 in isogenic CLL lines, at concentrations between 1–10μM, irrespective of their ATM and TP53 status. Strikingly, the majority of primary CLL tumours were sensitive to HBX19818 concentrations between 8μM and 16μM to which non-tumour PBMCs were resistant. Western blotting was used to monitor the induction of p53, apoptosis (associated with caspase 7 and PARP1 cleavage), and also whether DNA damage was induced (as measured by H2AX phosphorylation) in response to HBX19818. Our analysis revealed that pharmacological inhibition of USP7 led to p53 upregulation in the p53 proficient CLL cells associated with a robust induction of p21 indicating that the stabilised p53 was active. This response was absent in Mec-1 cell line with non-functional p53. Interestingly, caspase 7 and PARP1 were only cleaved in the ATM wild type CLL cell lines suggesting that activated p53 was capable of inducing cell death. In contrast, the ATM and p53 deficient CLL cell lines did not exhibit any markers suggesting an apoptotic mode of cell death. Rather, these cell lines displayed elevated levels of phospho-H2AX, suggesting the induction of DNA damage, possibly caused by an underlying DNA repair defect. Consistent with our hypothesis, both p53 proficient and p53 non-functional CLL cell lines failed to induce the recruitment of the HR protein Rad51 to sites of IR-induced DNA double strand breaks. Taken together, our data implies that in addition to p53 activation, pharmacological inhibition of USP7 can exert a cytotoxic effect by further mechanisms, possibly by modulating DNA double strand break repair. This is consistent with previous reports suggesting that USP7 regulates monoubiquitination of transcription factor FOXO4 and is involved in the regulation of DNA repair and mitotic progression via its interactions with Claspin and Chfr respectively. We suggest that pharmacological inhibition of USP7 represents a promising target for the treatment of tumours with defective ATM and p53 signalling. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2582-2582
Author(s):  
Monica Pallis ◽  
Martin Grundy ◽  
Claire Seedhouse ◽  
Heather Pimblett ◽  
Nigel Russell

Abstract Clofarabine is a purine nucleoside analogue which has been incorporated into several therapeutic trial protocols for the treatment of leukaemias including acute myeloid leukaemia (AML). The aims of the study are to ascertain mechanisms of clofarabine action in AML using cell lines and presentation samples from patients. We measured apoptosis by TdT assay, cytochrome C release and flow cytometric assays of the mitochondrial membrane potential probe DiOC6. To study the effects of clofarabine on DNA synthesis and DNA double strand breaks, we used bromodeoxyuridine (BrdU) and H2AX assays respectively. Equitoxic doses were established that caused approximately 20% cell death in the AML cell lines HL-60 (0.3 μM), KG1 (1 μM) and MV4.11 (1 μM) after 6 hours of continuous exposure. At these doses, clofarabine induced apoptotic DNA nicks, as measured by the TdT assay, and Cytochrome C release in all three cell lines. However, clofarabine-induced mitochondrial hyperpolarisation and depolarisation were found to be cell-type specific, occurring in HL-60 but not MV4.11 or KG1 cells, i.e. mitochondrial membrane depolarisation is not an essential part of the mechanism of clofarabine-induced apoptosis. Following clofarabine treatment, DNA synthesis was reduced by 91% within 1 hour in KG1 cells and by 80% in HL-60, but by only 9% in MV4.11. Out of the three cell lines, MV4.11 cells, which have a FLT3 internal tandem duplication (ITD), were the only ones to completely repair DNA double strand breaks following clofarabine removal. In 12 samples from patients with AML, the proportion of cells which incorporated BrdU in a 45 minute assay - a measure of the rate of cell cycling - differed considerably between samples, from 0.8% to 23%, median 13%. Cell death induced by clofarabine was correlated with the cycling rate of untreated cells (P=0.016). All 12 samples showed inhibition of DNA synthesis within 60 minutes of clofarabine treatment (range 20%–92% inhibition, median 54%). Mitochondrial membrane hyperpolarisation and depolarisation were not observed in patient cells. However, DNA double strand breaks were induced by clofarabine in patient cells. Paradoxically, i.e. in contrast to the results seen with the MV4.11 cell line, toxicity was greatest in samples with a FLT3 mutation (P=0.007). In conclusion, the FLT3 mutant MV4.11 cell line effectively repairs clofarabine-induced double strand breaks. Cell death in patient cells cultured with clofarabine is correlated with the presence of a FLT3 mutation. As we have previously established that AML samples with FLT3 mutations have upregulated DNA repair activity, this paradox might be explained by cycles of attempted DNA repair frustrated by renewed clofarabine incoporation into DNA, thus increasing the toxicity of the drug.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 173-173
Author(s):  
Francesca Cottini ◽  
Teru Hideshima ◽  
Rikio Suzuki ◽  
Paul G. Richardson ◽  
Kenneth C. Anderson ◽  
...  

Abstract Background: Multiple myeloma (MM) cells show a variable combination of chromosomal translocations, copy-number variations, somatic mutations and clonal heterogeneity, which makes every patient unique. We have recently shown that MM cells have signs of ongoing DNA-damage, which activates an ATM/ABL1-dependent DNA damage response (DDR) without overt apoptosis (Cottini et al., Nat Med, 2014). Here we further characterize the mechanisms of DNA damage and replicative stress in MM, which provide the basis for a novel synthetic lethality treatment approach. Results: MM cell lines with active DNA damage have enrichment in pathways of DNA replication and cell cycle. These same MM cell lines also present 53BP1, RPA and RAD51 foci with activated ATR and CHK1. Of note, 53BP1, RPA and RAD51 foci are markers of replicative stress, associated with DNA hyper-replication and stalled replication forks. Importantly, replicative stress markers are also present in primary MM cells. We also demonstrated a gene expression signature specific for increased chromosomal instability and DNA damage in a cohort of MM patients versus normal plasma cells. Remarkably, a subset (20 percent) of patients with myeloma overexpress genes belonging to the instability signature; this group also shows an unfavorable prognosis due to a more aggressive disease. These findings suggest that some patients present a similar phenotype to the cell lines, characterized by extensive replicative stress and activation of hyper-replicative pathways. We therefore hypothesized that MM cells might be sensitive to replicative stress overload, which occurs when cells fail to endure the presence of an excess of unrepaired DNA. To evaluate this hypothesis, we used shRNAs to silence ATR, a protein involved in the control of stalled replication origins, in two myeloma cell lines, one with normal TP53 (H929) and another with mutant TP53 (OPM-2). Inhibition of ATR caused a reduction in cell growth and induction of apoptosis, both more evident in MM cell lines with mutant TP53. A similar phenotype was observed when MM cell lines were incubated with VE-821, a specific ATR inhibitor. The strongest response occurred in TP53 mutant cell lines, which are representative of a model of aggressive MM, consistent with the concept of replicative stress overload. Indeed, p53 is normally phosphorylated and active in MM, while TP53 loss in the context of hyper-replication may prevent activation of salvage checkpoint, thereby favoring cell death in the absence of ATR. ATR inhibition also induces an increase in DNA double strand breaks, as evidenced by the higher number of γ-H2A.X foci. Reactive oxygen species (ROS) can also mediate DNA damage, and treatment with an antioxidant reagent N-Acetylcysteine (NAC), which helps scavenging ROS by replenishing glutathione stores, was indeed capable of reducing DNA double strand breaks and replicative stress markers. Since cancer cells are particularly sensitive to oxidative stress, we then evaluated the anti-MM activity of piperlongumine (PL), a drug that induces ROS accumulation. MM cell lines were sensitive to PL treatment, while PBMCs were minimally affected. As expected, the apoptotic effects of PL were abrogated upon co-incubation with NAC, indicating the specific activity of PL on ROS. We next exploited the possibility of combining replicative and oxidative stress in myeloma cells, hoping to overcome the threshold of tolerance to unrepaired DNA. H929 and OPM-2 cells were transfected with ATR shRNAs or treated with VE-821 and incubated with DMSO or 1-2.5 μM PL; synergic effects by the combination treatment were evident in both myeloma cell lines and also in patient MM cells. Conclusion: Replicative stress is present in a group of MM patients, who have aggressive disease, myeloma cell hyperproliferation and poor prognosis. Strategies aimed to shift the balance towards high DNA damage by ROS production and reduced DNA repair can decrease MM growth and may benefit patients with otherwise unfavorable outcomes. Disclosures No relevant conflicts of interest to declare.


1998 ◽  
Vol 18 (6) ◽  
pp. 646-657 ◽  
Author(s):  
Jun Chen ◽  
Koichi Uchimura ◽  
R. Anne Stetler ◽  
Raymond L. Zhu ◽  
Masaki Nakayama ◽  
...  

Using in situ hybridization, Northern blot analysis, Western blot analysis, and immunocytochemistry, mRNA and protein expression of the novel DNA damage-inducible gene GADD45 was examined in the rat brain at 0.5, 2, 4, 8, 16, 24, 48, and 72 hours after 15 minutes of transient global ischemia. Transient ischemia produced by the four-vessel occlusion method resulted in DNA double-strand breaks and delayed neuronal cell death in vulnerable neurons of the hippocampal CA1 sector, the hilus, dorsal caudate-putamen, and thalamus, as shown by in situ DNA nick end-labeling and histologic staining. GADD45 mRNA was transiently increased in less-vulnerable regions such as the parietal cortex (up to 8 hours after ischemia) and dentate granule cells (up to 24 hours after ischemia) but was persistently increased in vulnerable neurons such as CA1 pyramidal neurons (up to 48 hours). GADD45 immunoreactivity was increased in both vulnerable and less-vulnerable regions at earlier reperfusion periods (4 to 16 hours), but thereafter immunoreactivity was decreased below control levels in most vulnerable regions before delayed cell death and DNA double-strand breaks. At 72 hours after transient ischemia, a moderate increase in GADD45 immunoreactivity was still detectable in some CA3 neurons and in a few surviving neurons in the CA1 region. Double staining performed at 16 to 72 hours after ischemia revealed that GADD45 immunoreactivity was persistently increased in neurons that did not develop DNA damage. Because GADD45 protein may participate in the DNA excision repair process and because it has been shown that this protein is also overexpressed in neurons that survive focal ischemia and kainate-induced epileptic seizures, the results reported here support the hypothesis that GADD45 could have a protective role in neuronal injury.


Author(s):  
B. Zhai ◽  
A. Steino ◽  
J. Bacha ◽  
D. Brown ◽  
M. Daugaard

Dianhydrogalactitol (VAL-083) is a unique bi-functional alkylating agent causing N7-guanine-methylation and inter-strand DNA crosslinks. VAL-083 readily crosses the blood-brain barrier, accumulates in brain tumor tissue and has shown activity in prior NCI-sponsored clinical trials against various cancers, including glioblastoma (GBM) and medulloblastoma. VAL-083 is also active against GBM cancer stem cells and acts as a radiosensitizer independent of O6-methylguanine-DNA methyltransferase activity (in contrast to e.g. temozolomide and BCNU). Here we report new insights into VAL-083 mechanism of action by showing that VAL-083 induces irreversible cell-cycle arrest and cell death caused by replication-dependent DNA damage. In lung (H2122, H1792, H23, A549) and prostate (PC3, LNCaP) cancer cell lines VAL-083 treatment caused irreversible S/G2 cell-cycle arrest and cell death (IC50 range 3.06-25.7 µM). VAL-083 pulse-treatment led to persistent phosphorylation of DNA double-strand breaks (DSB) sensors ATM, single-strand DNA-binding Replication Protein A (RPA32), and histone variant H2A.X, suggesting persistent DNA lesions. After 10 months in culture with increasing VAL-083 concentrations, H1792 and LNCaP cells survive at concentrations up to 9.4 µM and 7.4 µM, respectively, suggesting that efficient resistance mechanisms are not easily acquired by the cancer cells. Taken together with previous results showing that VAL-083 circumvents cisplatin-resistance and is less dependent on p53 activity than cisplatin, these results suggest a molecular mechanism for VAL-083 that differs from both TMZ, BCNU and cisplatin. They further suggest that irreparable DNA damage induced by VAL-083 is impervious to common strategies employed by cancer cells to escape effects of alkylating drugs used in GBM treatment.


2021 ◽  
Vol 7 (25) ◽  
pp. eabg0993
Author(s):  
Naoki Takahashi ◽  
Soichi Inagaki ◽  
Kohei Nishimura ◽  
Hitoshi Sakakibara ◽  
Ioanna Antoniadi ◽  
...  

Plants have a high ability to cope with changing environments and grow continuously throughout life. However, the mechanisms by which plants strike a balance between stress response and organ growth remain elusive. Here, we found that DNA double-strand breaks enhance the accumulation of cytokinin hormones through the DNA damage signaling pathway in the Arabidopsis root tip. Our data showed that activation of cytokinin signaling suppresses the expression of some of the PIN-FORMED genes that encode efflux carriers of another hormone, auxin, thereby decreasing the auxin signals in the root tip and causing cell cycle arrest at G2 phase and stem cell death. Elevated cytokinin signaling also promotes an early transition from cell division to endoreplication in the basal part of the root apex. We propose that plant hormones spatially coordinate differential DNA damage responses, thereby maintaining genome integrity and minimizing cell death to ensure continuous root growth.


2021 ◽  
Author(s):  
David Yves Zander ◽  
Sandy S Burkart ◽  
Sandra Wuest ◽  
Vladimir Goncalves Magalhaes ◽  
Marco Binder

Properly responding to DNA damage is vital for eukaryotic cells, including the induction of DNA repair, growth arrest and, as a last resort to prevent neoplastic transformation, cell death. Besides being crucial for ensuring homeostasis, the same pathways and mechanisms are at the basis of chemoradiotherapy in cancer treatment, which involves therapeutic induction of DNA damage by chemical or physical (radiological) measures. Apart from typical DNA damage response mediators, the relevance of cell-intrinsic antiviral signaling pathways in response to DNA breaks has recently emerged. Originally known for combatting viruses via expression of antiviral factors including interferons (IFNs) and establishing of an antiviral state, retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) were found to be critical for adequate induction of cell death upon the introduction of DNA double-strand breaks. We here show that presence of IRF3 is crucial in this process, most likely through direct activation of pro-apoptotic factors rather than transcriptional induction of canonical downstream components, such as IFNs. Investigating genes reported to be involved in both DNA damage response and antiviral signaling, we demonstrate that IRF1 is an obligatory factor for DNA damage-induced cell death. Interestingly, its regulation does not require activation of RLR signaling, but rather sensing of DNA double strand breaks by ATM and ATR. Hence, even though independently regulated, both RLR signaling and IRF1 are essential for proper induction/execution of intrinsic apoptosis. Our results not only support more broadly developing IRF1 as a biomarker predictive for the effectiveness of chemoradiotherapy, but also suggest investigating a combined pharmacological stimulation of RLR and IRF1 signaling as a potential adjuvant regimen in tumor therapy.


2010 ◽  
Vol 49 (S 01) ◽  
pp. S64-S68
Author(s):  
E. Dikomey

SummaryIonising irradiation acts primarily via induction of DNA damage, among which doublestrand breaks are the most important lesions. These lesions may lead to lethal chromosome aberrations, which are the main reason for cell inactivation. Double-strand breaks can be repaired by several different mechanisms. The regulation of these mechanisms appears be fairly different for normal and tumour cells. Among different cell lines capacity of doublestrand break repair varies by only few percents and is known to be determined mostly by genetic factors. Knowledge about doublestrand break repair mechanisms and their regulation is important for the optimal application of ionising irradiation in medicine.


2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Deepti Sharma ◽  
Louis De Falco ◽  
Sivaraman Padavattan ◽  
Chang Rao ◽  
Susana Geifman-Shochat ◽  
...  

AbstractThe poly(ADP-ribose) polymerase, PARP1, plays a key role in maintaining genomic integrity by detecting DNA damage and mediating repair. γH2A.X is the primary histone marker for DNA double-strand breaks and PARP1 localizes to H2A.X-enriched chromatin damage sites, but the basis for this association is not clear. We characterize the kinetics of PARP1 binding to a variety of nucleosomes harbouring DNA double-strand breaks, which reveal that PARP1 associates faster with (γ)H2A.X- versus H2A-nucleosomes, resulting in a higher affinity for the former, which is maximal for γH2A.X-nucleosome that is also the activator eliciting the greatest poly-ADP-ribosylation catalytic efficiency. The enhanced activities with γH2A.X-nucleosome coincide with increased accessibility of the DNA termini resulting from the H2A.X-Ser139 phosphorylation. Indeed, H2A- and (γ)H2A.X-nucleosomes have distinct stability characteristics, which are rationalized by mutational analysis and (γ)H2A.X-nucleosome core crystal structures. This suggests that the γH2A.X epigenetic marker directly facilitates DNA repair by stabilizing PARP1 association and promoting catalysis.


Author(s):  
Sang-Min Jang ◽  
Christophe E. Redon ◽  
Haiqing Fu ◽  
Fred E. Indig ◽  
Mirit I. Aladjem

Abstract Background The p97/valosin-containing protein (VCP) complex is a crucial factor for the segregation of ubiquitinated proteins in the DNA damage response and repair pathway. Objective We investigated whether blocking the p97/VCP function can inhibit the proliferation of RepID-deficient cancer cells using immunofluorescence, clonogenic survival assay, fluorescence-activated cell sorting, and immunoblotting. Result p97/VCP was recruited to chromatin and colocalized with DNA double-strand breaks in RepID-deficient cancer cells that undergo spontaneous DNA damage. Inhibition of p97/VCP induced death of RepID-depleted cancer cells. This study highlights the potential of targeting p97/VCP complex as an anticancer therapeutic approach. Conclusion Our results show that RepID is required to prevent excessive DNA damage at the endogenous levels. Localization of p97/VCP to DSB sites was induced based on spontaneous DNA damage in RepID-depleted cancer cells. Anticancer drugs targeting p97/VCP may be highly potent in RepID-deficient cells. Therefore, we suggest that p97/VCP inhibitors synergize with RepID depletion to kill cancer cells.


Sign in / Sign up

Export Citation Format

Share Document