Foxo3-Mediated Redox Regulation Is Required for DNA Damage Response in Hematopoietic Stem Cells.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2295-2295
Author(s):  
Carolina L. Bigarella ◽  
Pauline Rimmele ◽  
Brigitte Izac ◽  
Valentina d'Escamard ◽  
Saghi Ghaffari

Abstract Abstract 2295 Stringent regulation of redox status is critical to the control of hematopoietic stem cell (HSC) quiescence and to the maintenance of HSC pool. However mechanisms by which oxidative stress controls HSC quiescence versus cycling remain unknown. Foxo3 transcription factor is required for the regulation of HSC quiescence and for the maintenance of hematopoietic and leukemic stem cell pool. Redox regulation is key to the Foxo3 control of HSC pool. ROS accumulation in Foxo3 null HSC mediates in vivo activation of p53, and increased p21 expression leading to an arrest in the G2/M phase of cell cycle associated with loss of quiescence. We hypothesized that ROS may regulate HSC quiescence versus cycling via control of DNA damage repair program. To address this question, we examined whether Foxo3 is involved in DNA damage response of HSC. We first evaluated by immunostaining phosphorylation of histone H2AX variant (γH2AX), a hallmark sensor of DNA strand break, in LSK (Lin−Sca-1+c-Kit+) cells freshly isolated from Foxo3−/− bone marrow. We found the number of cells with nuclear γH2AX foci significantly increased in Foxo3−/− LSK cells (n=100; >5 foci/nuclei) in comparison with wild type (WT)-LSK. We subsequently confirmed and quantified these data by flow cytometry analysis of γH2AX. Together these analyses showed that loss of Foxo3 leads to increased γH2AX levels in LSK cells at the steady state. We next evaluated the presence of DNA breaks, by submitting Foxo3−/− versus WT LSK FACS-sorted cells to single-cell gel electrophoresis (Comet Assay). These investigations confirmed that LSK cells from Foxo3−/− mice accumulate DNA breaks at the steady state, as the percentage of comet shape cells (4 fold) and comet length (3 fold) were all increased in Foxo3 mutant LSK. We then asked whether the increased ROS accumulation had any direct role in damaging DNA in Foxo3−/− LSK. Using a fluorescent probe specific for the most common oxidative DNA damage lesion, the 8-hydroxyguanine base (8-OxoG), we further showed that Foxo3−/− LSK cells exhibit oxidative DNA damage. To further investigate the potential function of ROS in the control of HSC DNA damage response, we treated Foxo3−/− and WT mice for 14 days with the ROS scavenger N-acetyl-cysteine (NAC; 100 mg/Kg/day) in vivo. NAC treatment reduced by four fold γH2AX in Foxo3−/− LSK cells to levels similar to that in WT-LSK cells. Similarly, comet assay analysis of FACS-sorted LSK cells from NAC-treated WT and Foxo3−/− mice showed a two fold reduction of DNA breaks. These results suggest that increase in ROS damage DNA and triggers DNA damage response in Foxo3−/− LSK cells at the steady state. Additionally, expression of a number of genes involved in DNA damage repair including Xrcc5 (Ku80) and Xrcc6 (Ku70) was highly downregulated in both long-term-HSC (LT-HSC, LSK-CD150+CD48−) and LSK populations as evidenced by Q-RT-PCR on the Fluidigm™ microfluidics array technology. Together these results strongly suggest that Foxo3-mediated redox regulation is required for protection of DNA from accumulating damage at the steady state in HSC. We further investigated whether ROS-mediated activation of p53 in Foxo3 null HSCs limits the extent of accumulation of DNA damage in HSC. To address this question we crossed p53+/−Foxo3+/− double heterozygous animals to generate p53-Foxo3 double knockout mice. Loss of p53 in Foxo3−/− mice led to significant rise in lymphocyte counts and decrease in neutrophil counts in comparison with Foxo3−/−, indicating a potential shift in lineage determination from HSC. To our surprise, loss of one allele of p53 in Foxo3-null mice significantly reduced gH2AX staining and DNA breaks, as analyzed respectively by flow cytometry and comet assay of sorted LSK cells. While the rescue of DNA damage in Foxo3−/− HSCs as result of loss of p53 was unexpected it is not clear whether it is related to the impact on the fate of HSC. The clarification of these questions in future studies will be important for understanding mechanisms that control the emergence of leukemic stem cells. Together these studies suggest that Foxo3 guards DNA from damage in HSC at the steady state. In addition they indicate an important function for ROS modulation in the in vivo regulation of DNA damage response in HSC. Altogether understanding mechanisms that control ROS modulation of DNA damage response are likely to advance our understanding of the regulation of normal hematopoietic and leukemic stem cell quiescence. Disclosures: No relevant conflicts of interest to declare.

2014 ◽  
Vol 211 (9) ◽  
pp. 1759-1777 ◽  
Author(s):  
Cesare Lancini ◽  
Paul C.M. van den Berk ◽  
Joseph H.A. Vissers ◽  
Gaetano Gargiulo ◽  
Ji-Ying Song ◽  
...  

Histone ubiquitination at DNA breaks is required for activation of the DNA damage response (DDR) and DNA repair. How the dynamic removal of this modification by deubiquitinating enzymes (DUBs) impacts genome maintenance in vivo is largely unknown. To address this question, we generated mice deficient for Ub-specific protease 3 (USP3; Usp3Δ/Δ), a histone H2A DUB which negatively regulates ubiquitin-dependent DDR signaling. Notably, USP3 deletion increased the levels of histone ubiquitination in adult tissues, reduced the hematopoietic stem cell (HSC) reserves over time, and shortened animal life span. Mechanistically, our data show that USP3 is important in HSC homeostasis, preserving HSC self-renewal, and repopulation potential in vivo and proliferation in vitro. A defective DDR and unresolved spontaneous DNA damage contribute to cell cycle restriction of Usp3Δ/Δ HSCs. Beyond the hematopoietic system, Usp3Δ/Δ animals spontaneously developed tumors, and primary Usp3Δ/Δ cells failed to preserve chromosomal integrity. These findings broadly support the regulation of chromatin ubiquitination as a key pathway in preserving tissue function through modulation of the response to genotoxic stress.


2019 ◽  
Vol 24 (4) ◽  
pp. 551-565.e8 ◽  
Author(s):  
Giulia Schiroli ◽  
Anastasia Conti ◽  
Samuele Ferrari ◽  
Lucrezia della Volpe ◽  
Aurelien Jacob ◽  
...  

2008 ◽  
Vol 295 (6) ◽  
pp. F1678-F1688 ◽  
Author(s):  
Natalia I. Dmitrieva ◽  
Maurice B. Burg

We previously reported that, both in cell culture and in the renal inner medulla in vivo, elevating NaCl increased the number of DNA breaks, which persisted as long as NaCl remained high but were rapidly repaired when NaCl was lowered. Furthermore, those breaks did not induce the DNA repair protein γH2AX or cause activation of the MRN (Mre11, Rad50, Nbs1) complex. In contrast, others recently reported that high NaCl does induce γH2AX and MRN complex formation and concluded that these activities are associated with repair of the DNA (Sheen MR, Kim SW, Jung JY, Ahn JY, Rhee JG, Kwon HM, Woo SK. Am J Physiol Renal Physiol 291: F1014–F1020, 2006). The purpose of the present studies was to resolve the disparity. The important difference is that HeLa cells, which were the main subject of the later report, are much less tolerant of high NaCl than are the mIMCD3 cells, which were our main subject. mIMCD3 cells survive levels of NaCl that kill HeLa cells by apoptosis. Here we demonstrate that in both cell types raising NaCl to a level that the cells survive (higher for mIMCD3 than HeLa) increases DNA breaks without inducing γH2AX or activating the MRN complex and that the DNA breaks persist as long as NaCl remains elevated, but are rapidly repaired when it is lowered. Importantly, in both cell types, raising NaCl further to cause apoptosis activates these DNA damage response proteins and greatly fragments DNA, associated with cell death. We conclude that γH2AX induction and MRN activation in response to high NaCl are associated with apoptosis, not DNA repair.


Cell Cycle ◽  
2007 ◽  
Vol 6 (19) ◽  
pp. 2371-2376 ◽  
Author(s):  
Derrick J. Rossi ◽  
Jun Seita ◽  
Agnieszka Czechowicz ◽  
Deepta Bhattacharya ◽  
David Bryder ◽  
...  

2016 ◽  
Vol 14 (3) ◽  
pp. 147-154 ◽  
Author(s):  
Tangliang Li ◽  
Zhong-Wei Zhou ◽  
Zhenyu Ju ◽  
Zhao-Qi Wang

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 864-864 ◽  
Author(s):  
Fukun Guo ◽  
Jie Li ◽  
Wei Du ◽  
Shuangmin Zhang ◽  
Wei Liu ◽  
...  

Abstract Abstract 864 The mammalian target of rapamycin (mTOR) integrates signals from nutrients, growth factors, and cellular energy status to control protein synthesis, cell growth, proliferation, survival and metabolism in various cancer cells, but its physiological function in the hematopoiesis process and signaling role in hematopoietic stem cell (HSC) regulation remain unknown. By using the inhibitor rapamycin, mTOR has previously been suggested to regulate megakaryocyte and dendritic cell proliferation and differentiation. Hyperactivation of mTOR by deletion of the negative regulators of mTOR, TSC1/TSC2 or PTEN, causes a loss of quiescence and long-term exhaustion of HSCs. Since conventional gene targeting of mTOR leads to early embryonic lethality, a conditional mTOR knockout mouse model has recently been generated. We have produced mTORflox/flox; Mx-Cre compound mice that allow interferon-induced mTOR deletion in bone marrow (BM) following a transplantation and polyI:C induction protocol. We found that depletion of mTOR drastically affected hematopoiesis: the mTORflox/flox;Mx-Cre BM recipient mice showed a marked reduction in total BM cellularity and in the numbers and frequency of myeloid and lymphoid cells, erythrocytes, and platelets in peripheral blood, bone marrow, and thymus, after induced mTOR deletion, resulting in bone marrow failure and lethality. Interestingly, the numbers of hematopoietic stem and progenitor cells (HSPCs; Lin−Sca-1+c-Kit+) and HSCs (CD150+ CD41−CD48− Lin−Sca-1+c-Kit+) in bone marrow increased transiently by approximately 5- and 8-fold, respectively, while the numbers of early progenitors (CMP, GMP, MEP, CLP) were mildly affected in the mutant mice 7–14 days after polyI:C treatment. While the more mature lineage committed mTOR−/− blood cells showed a cell cycle blockage and significantly increased apoptosis, mTOR−/− HSPCs and HSCs displayed a loss of quiescence and increased proliferation, as assessed by 5-bromodeoxyuridine incorporation assays, and a normal survival index. Transplantation of mTOR−/− BM cells into immunodeficient or syngeneic mice demonstrated that the mTOR−/− HSPCs failed to engraft and repopulate in the recipients. At the molecular level, mRNA microarray, quantitative real-time PCR and immunoblotting analyses of mTOR−/− HSPCs or Lin− cells revealed that the cell cycle inhibitor Rb was downregulated while the positive regulator of cell cycle E2F5 and pro-survival regulators MCL1 and BCL-xL were upregulated. mTOR deficiency abolished the activation of translational regulators S6K and 4E-BP but led to an increased activation of Akt. In addition, mTOR deficiency sensitized Lin− cells to DNA damage induced in vitro or in vivo by melphalan or mitomycin C (MMC), evidenced by a marked increase in γH2AX foci as well as DNA double-strand breaks (comet-tailed value of 30.2 ± 7.6 in mTOR−/− cells treated in vitro with melphalan and 37.6 ± 3.4 in mTOR−/− cells treated in vivo with MMC compared to 7.6 ± 2.1 in melphalan-treated WT cells and 17.3 ± 6.7 in MMC-treated WT cells, respectively). The increased DNA damage response can be attributed to an ∼300-fold reduction of the expression of FANCD2, a key component of the Fanconi DNA damage repair complex. Significantly, the effect of mTOR deficiency on Fanconi gene expression was specific to FANCD2, because the expression of other Fanconi proteins such as FANCA and FANCC was not affected in mTOR−/− Lin− cells. Intriguingly, the mTOR−/− Lin− cells phenocopied the DNA damage response of FANCD2−/− Lin− cells in vitro and in vivo. Similar effects of reduced FANCD2 expression and dampened DNA damage response were observed in human lymphoblasts treated with pp242, a mTOR kinase inhibitor. FANCD2-deficient human Fanconi anemia patient cells recapitulated the pp242-induced DNA damage phenotypes that could be rescued by FANCD2 reconstitution. Taken together, these results demonstrate that mTOR is a critical regulator of HSC quiescence and engraftment through the regulation of cell cycle machinery and is essential in multiple stages of hematopoiesis. Moreover, mTOR is required for maintaining genomic stability of HSPCs through modulation of the Fanconi anemia DNA damage response pathway. Disclosures: No relevant conflicts of interest to declare.


NAR Cancer ◽  
2021 ◽  
Vol 3 (1) ◽  
Author(s):  
Luisa Statello ◽  
Mohamad M Ali ◽  
Silke Reischl ◽  
Sagar Mahale ◽  
Subazini Thankaswamy Kosalai ◽  
...  

Abstract Despite the rapid improvements in unveiling the importance of lncRNAs in all aspects of cancer biology, there is still a void in mechanistic understanding of their role in the DNA damage response. Here we explored the potential role of the oncogenic lncRNA SCAT7 (ELF3-AS1) in the maintenance of genome integrity. We show that SCAT7 is upregulated in response to DNA-damaging drugs like cisplatin and camptothecin, where SCAT7 expression is required to promote cell survival. SCAT7 silencing leads to decreased proliferation of cisplatin-resistant cells in vitro and in vivo through interfering with cell cycle checkpoints and DNA repair molecular pathways. SCAT7 regulates ATR signaling, promoting homologous recombination. Importantly, SCAT7 also takes part in proteasome-mediated topoisomerase I (TOP1) degradation, and its depletion causes an accumulation of TOP1–cc structures responsible for the high levels of intrinsic DNA damage. Thus, our data demonstrate that SCAT7 is an important constituent of the DNA damage response pathway and serves as a potential therapeutic target for hard-to-treat drug resistant cancers.


2020 ◽  
Vol 11 (7) ◽  
Author(s):  
Nan Huang ◽  
Chang Xu ◽  
Liang Deng ◽  
Xue Li ◽  
Zhixuan Bian ◽  
...  

AbstractPhosphoribosylaminoimidazole carboxylase, phosphoribosylaminoimidazole succinocarboxamide synthetase (PAICS), an essential enzyme involved in de novo purine biosynthesis, is connected with formation of various tumors. However, the specific biological roles and related mechanisms of PAICS in gastric cancer (GC) remain unclear. In the present study, we identified for the first time that PAICS was significantly upregulated in GC and high expression of PAICS was correlated with poor prognosis of patients with GC. In addition, knockdown of PAICS significantly induced cell apoptosis, and inhibited GC cell growth both in vitro and in vivo. Mechanistic studies first found that PAICS was engaged in DNA damage response, and knockdown of PAICS in GC cell lines induced DNA damage and impaired DNA damage repair efficiency. Further explorations revealed that PAICS interacted with histone deacetylase HDAC1 and HDAC2, and PAICS deficiency decreased the expression of DAD51 and inhibited its recruitment to DNA damage sites by impairing HDAC1/2 deacetylase activity, eventually preventing DNA damage repair. Consistently, PAICS deficiency enhanced the sensitivity of GC cells to DNA damage agent, cisplatin (CDDP), both in vitro and in vivo. Altogether, our findings demonstrate that PAICS plays an oncogenic role in GC, which act as a novel diagnosis and prognostic biomarker for patients with GC.


Sign in / Sign up

Export Citation Format

Share Document