Autophagy Promotes the Repair of Radiation-Induced DNA Damage in Bone Marrow Hematopoietic Cells via Enhanced STAT3 Signaling

2017 ◽  
Vol 187 (3) ◽  
pp. 382 ◽  
Author(s):  
Fei Xu ◽  
Xin Li ◽  
Lili Yan ◽  
Na Yuan ◽  
Yixuan Fang ◽  
...  
2002 ◽  
Vol 76 (15) ◽  
pp. 7790-7798 ◽  
Author(s):  
Masanobu Kitagawa ◽  
Shuichi Yamaguchi ◽  
Maki Hasegawa ◽  
Kaoru Tanaka ◽  
Toshihiko Sado ◽  
...  

ABSTRACT Exposure of hematopoietic progenitors to gamma irradiation induces p53-dependent apoptosis. However, host responses to DNA damage are not uniform and can be modified by various factors. Here, we report that a split low-dose total-body irradiation (TBI) (1.5 Gy twice) to the host causes prominent apoptosis in bone marrow cells of Friend leukemia virus (FLV)-infected C3H mice but not in those of FLV-infected DBA mice. In C3H mice, the apoptosis occurs rapidly and progressively in erythroid cells, leading to lethal host anemia, although treatment with FLV alone or TBI alone induced minimal apoptosis in bone marrow cells. A marked accumulation of P53 protein was demonstrated in bone marrow cells from FLV-infected C3H mice 12 h after treatment with TBI. Although a similar accumulation of P53 was also observed in bone marrow cells from FLV-infected DBA mice treated with TBI, the amount appeared to be parallel to that of mice treated with TBI alone and was much lower than that of FLV- plus TBI-treated C3H mice. To determine the association of p53 with the prominent enhancement of apoptosis in FLV- plus TBI-treated C3H mice, p53 knockout mice of the C3H background (C3H p53−/− ) were infected with FLV and treated with TBI. As expected, p53 knockout mice exhibited a very low frequency of apoptosis in the bone marrow after treatment with FLV plus TBI. Further, C3H p53−/− → C3H p53+/+ bone marrow chimeric mice treated with FLV plus TBI survived even longer than the chimeras treated with FLV alone. These findings indicate that infection with FLV strongly enhances radiation-induced apoptotic cell death of hematopoietic cells in host animals and that the apoptosis occurs through a p53-associated signaling pathway, although the response was not uniform in different host strains.


Blood ◽  
2000 ◽  
Vol 95 (8) ◽  
pp. 2645-2650 ◽  
Author(s):  
Lisa A. Porter ◽  
Gurmit Singh ◽  
Jonathan M. Lee

Abstract γ-Radiation is a potent inducer of apoptosis. There are multiple pathways regulating DNA damage-induced apoptosis, and we set out to identify novel mechanisms regulating γ-radiation–induced apoptosis in hematopoietic cells. In this report, we present data implicating the cyclin B1 protein as a regulator of apoptotic fate following DNA damage. Cyclin B1 is the regulatory subunit of the cdc2 serine/threonine kinase, and accumulation of cyclin B1 in late G2 phase of the cell cycle is a prerequisite for mitotic initiation in mammalian cells. We find that abundance of the cyclin B1 protein rapidly increases in several mouse and human hematopoietic cells (Ramos, DP16, HL60, thymocytes) undergoing γ-radiation–induced apoptosis. Cyclin B1 accumulation occurs in all phases of the cell cycle. Antisense inhibition of cyclin B1 accumulation decreases apoptosis, and ectopic cyclin B1 expression is sufficient to induce apoptosis. These observations are consistent with the idea that cyclin B1 is both necessary and sufficient for γ-radiation-induced apoptosis.


Blood ◽  
2021 ◽  
Author(s):  
Tao Cheng ◽  
Linping Hu ◽  
Xiuxiu Yin ◽  
Yawen Zhang ◽  
Aiming Pang ◽  
...  

Total body irradiation (TBI) is commonly used in host conditioning regimens for human hematopoietic stem cell (HSC) transplantation to treat various hematological disorders. Exposure to TBI not only induces acute myelosuppression and immunosuppression but also impairs the various components of the HSC niche in recipients. Our previous study demonstrated that radiation-induced bystander effects (RIBE) of irradiated recipients decreased the long-term repopulating ability of transplanted mouse HSCs. However, RIBE on human HSCs have not been studied. Here, we report that RIBE on transplanted human hematopoietic cells impaired the long-term hematopoietic reconstitution of human HSCs as well as the colony-forming ability of human hematopoietic progenitor cells (HPCs). Our further studies found that the RIBE-affected human hematopoietic cells showed enhanced DNA damage responses, cell cycle arrest and p53-dependent apoptosis, mainly due to oxidative stress. Moreover, multiple antioxidants could mitigate these bystander effects, though at different efficacies both in vitro and in vivo. Taken together, these findings suggest that RIBE impairs human HSCs by oxidative DNA damage. This study provides definitive evidence for RIBE in transplanted human HSCs and further justifies the necessity for conducting clinical trials to assess the ability of multiple antioxidants to improve the efficacy of HSC transplantation for patients with hematological or non-hematological disorders.


2016 ◽  
Vol 113 (52) ◽  
pp. 15024-15029 ◽  
Author(s):  
Michael I. Carr ◽  
Justine E. Roderick ◽  
Hong Zhang ◽  
Bruce A. Woda ◽  
Michelle A. Kelliher ◽  
...  

The p53 tumor suppressor acts as a guardian of the genome by preventing the propagation of DNA damage-induced breaks and mutations to subsequent generations of cells. We have previously shown that phosphorylation of the Mdm2 oncoprotein at Ser394 by the ATM kinase is required for robust p53 stabilization and activation in cells treated with ionizing radiation, and that loss of Mdm2 Ser394 phosphorylation leads to spontaneous tumorigenesis and radioresistance in Mdm2S394A mice. Previous in vitro data indicate that the c-Abl kinase phosphorylates Mdm2 at the neighboring residue (Tyr393) in response to DNA damage to regulate p53-dependent apoptosis. In this present study, we have generated an Mdm2 mutant mouse (Mdm2Y393F) to determine whether c-Abl phosphorylation of Mdm2 regulates the p53-mediated DNA damage response or p53 tumor suppression in vivo. The Mdm2Y393F mice develop accelerated spontaneous and oncogene-induced tumors, yet display no defects in p53 stabilization and activity following acute genotoxic stress. Although apoptosis is unaltered in these mice, they recover more rapidly from radiation-induced bone marrow ablation and are more resistant to whole-body radiation-induced lethality. These data reveal an in vivo role for c-Abl phosphorylation of Mdm2 in regulation of p53 tumor suppression and bone marrow failure. However, c-Abl phosphorylation of Mdm2 Tyr393 appears to play a lesser role in governing Mdm2-p53 signaling than ATM phosphorylation of Mdm2 Ser394. Furthermore, the effects of these phosphorylation events on p53 regulation are not additive, as Mdm2Y393F/S394A mice and Mdm2S394A mice display similar phenotypes.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5103-5103
Author(s):  
Debajyoti Majumdar ◽  
Eric Pietras ◽  
Jason Stumhofer ◽  
Snehalata A Pawar

Abstract Background: Bone marrow (BM) failure is a common side-effect of toxicity to the hematopoietic tissue both in the clinical setting as well as in case of total body irradiation (TBI) exposure in the event of nuclear terrorism or disasters. Particularly IR-induced myelosuppression is considered a significant risk factor for infections and increased risk for long-term hematopoietic dysfunction and myelodysplasia. Although a lot is known about the key regulatory proteins of steady-state hematopoiesis, not much is known about the regulatory factors involved in stress-induced hematopoiesis. Therefore delineating the mechanisms underlying the effects of IR stress-induced hematopoiesis is critical for the development of novel interventions with the potential to prevent or alleviate IR-induced BM injury/failure. The transcription factor CCAAT/enhancer-binding protein delta (Cebpd; C/EBPδ) plays an important role in the regulation of inflammatory and stress responses, and in the innate and adaptive immune responses. We have previously reported that Cebpd-knockout (KO) mice display increased neutropenia, thrombocytopenia and myelotoxicity in response to IR exposure, which correlated with increased apoptosis of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). In the present study, we further investigated the underlying mechanisms of IR-induced bone marrow failure in the absence of C/EBPδ. Methods: Cebpd+/+and Cebpd-/- mice (C57BL/6J background) aged 3 months were exposed to 6 Gy TBI. Bone marrow mononuclear cells (BM-MNCs) were isolated from femurs and tibiae harvested at early time-points (1h, 4h and 24h) as well as 2 weeks post-irradiation. The presence of reactive oxygen species (ROS) was measured using MitoSOX and the extent of DNA damage was measured using an antibody specific to g-H2AX at the above time points. BM-MNCs were labelled with fluorophore-tagged antibodies and analyzed by flow cytometry to measure the absolute numbers of long term-HSCs, multipotent progenitors, common lymphoid progenitors and myeloid progenitors at 2 weeks post-6 Gy. The changes in cell cycle distribution in response to IR exposure will also be analyzed by flow cytometry using a DNA-binding dye in conjunction with Ki67. Results: Preliminary results reveal: (a) reduced numbers of HSCs, HPCs, common myeloid progenitors, myeloid-erythroid progenitors and granulocyte-monocyte progenitors and (b) increased accumulation of ROS and the DNA damage marker, γ-H2AX in HSCs and HPCs in Cebpd-/- mice compared to Cebpd+/+ mice at 2 weeks post-irradiation. These results suggest that C/EBPδ may play a protective role in radiation-induced bone marrow injury. Studies are underway to further examine changes in cell cycle, ROS and DNA damage in the various BM cell lineages post-TBI. (Funding support by NIGMS P20GM109005 & Department of Defense W81XWH-15-1-0489 is gratefully acknowledged) Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1839-1839
Author(s):  
Hang Su ◽  
Suthakar Ganapathy ◽  
Zhi-Min Yuan ◽  
Chul S. Ha

Abstract Introduction Though radiolabeled anti-CD20 antibody has expanding roles in the management of B-cell lymphoma, its main drawback has been radiation-induced damage to the bone marrow leading to acute grade 3 and 4 hematological toxicity and potential contribution to the development of myelodysplastic syndrome and secondary leukemia. Arsenic trioxide is currently used to treat acute promyelocytic leukemia and known as a cytotoxic agent. However, we have recently demonstrated that arsenic trioxide can act as a cytoprotective agent at much lower dose. This is due to its ability to temporarily and reversibly suppress p53 activation caused by DNA-damaging treatments such as chemotherapy or radiotherapy. It has been also demonstrated that this protective effect is selective to normal tissues, as it requires functional p53. We have developed a preclinical model to assess the efficacy of low dose arsenic trioxide (LDA) as a cytoprotective agent against bone marrow toxicity induced by radioimmunotherapy using Y-90 ibritumomab tiuxetan as a model. Methods To test the hypothesis that LDA protects bone marrow against Y-90 ibritumomab tiuxetan induced damage, sex-matched BAL/c mice (4-6 weeks of age) were randomized into four groups: control, LAD only, Y-90 ibritumomab tiuxetan only, LAD pretreatment followed by Y-90 ibritumomab tiuxetan. LDA pretreatment was carried out by feeding mice with water containing 1 mg/L arsenic trioxide for three days. Y-90 ibritumomab tiuxetan was then injected into mice at the dose of 200uCi via tail vein. Tissue samples were collected at different time points (3 hours to 5 weeks) after treatment. Bone marrow damage was analyzed histologically with H&E staining, and DNA damage was assessed with pH2AX staining. To test the hypothesis that LDA does not protect malignant cells, a mouse xenograft model was generated using a CD20 expressing lymphoma cell line, Karpas 422. Treatments were initiated 1 week after implantation when tumors became palpable. Tumor volumes were measured with a caliper periodically. Tumor volume was calculated using the equation: volume = length × width × depth × 0.5236 mm3. Two independent experiments were done and the tumor volumes are expressed as means ± SE. Results Y-90 ibritumomab tiuxetan treatments were associated with severe damages to bone marrow cells, and such damages were significantly reduced by LDA pretreatment (Fig 1). Consistent with this observation, much more DNA damage was accumulated in mice treated with Y-90 ibritumomab tiuxetan from as early as 3 hours to a week after treatment, as compared to mice pretreated with LDA (Fig 2). Remarkably, while DNA damage was eliminated in LDA-pretreated mice by 2wk to 5wk after treatment, damage was still observed in mice without LAD pretreatment. In tumor xenograft models, the tumor volume of the control group continued to increase with time. LDA pretreatment did not have any detectable effect on the growth of the implanted tumors. As expected, treatment with a single dose of Y-90 ibritumomab tiuxetan resulted in marked tumor growth suppression. LDA pretreatment showed little effect on radiation-induced tumor growth suppression (Fig 3). Conclusion Our results demonstrate that a brief pretreatment with LDA is associated with a marked protection of bone marrow without compromising the ability of irradiation to kill lymphoma cells. A clinical trial is being developed based on our findings. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 266-266
Author(s):  
Yuki Kagoya ◽  
Shunya Arai ◽  
Akihide Yoshimi ◽  
Takako Tsuruta-Kishino ◽  
Keisuke Kataoka ◽  
...  

Abstract Myeloproliferative neoplasms (MPN) are clonal myeloid disorders associated with a high prevalence of JAK2V617F mutation. One of the most dismal complications in MPN is their transformation to acute myeloid leukemia (AML) after accumulation of additive genetic mutations. Curiously, the transformed AML cells frequently lack the JAK2 mutation, indicating that these leukemia cells are derived from JAK2V617F-negative clones which coexisted with JAK2V617F-positive ones during MPN phase. In this study, we investigated the pathogenesis underlying these phenomena using murine MPN model induced by transplantation of JAK2V617F-IRES-GFP-transduced bone marrow cells. First, we evaluated accumulation of DNA damage by immunofluorescence staining of γH2AX in GFP-positive and GFP-negative fractions. Surprisingly, we observed increased γH2AX foci formation in both GFP-positive and negative hematopoietic stem/progenitor cells. Moreover, when normal hematopoietic cells were cultured in conditioned media (CM) by JAK2V617F-positive cells, the cultured cells showed significantly elevated intracellular reactive oxygen species (ROS) levels and accumulation of γH2AX foci formation. These findings indicate that JAK2V617F-positive clones confer genetic instability and DNA damage accumulation to both themselves and neighboring normal cells in a paracrine manner. To clarify the mechanism of JAK2V617F-induced paracrine DNA damage, we analyzed gene expression profiles of JAK2V617F-positive hematopoietic stem/progenitor cells compared with normal counterparts in murine and human MPN. Since we were interested in elucidating the cause for paracrine effects evoked by JAK2V617F-positive clones, we focused on 8 genes encoding soluble factors included in the list of the genes that showed significantly elevated expression in JAK2V617-positive cells. Through shRNA-mediated knockdown of the individual genes in JAK2V617F-positive cells, we found that repression of lipocalin-2 (LCN2), one of the pro-inflammatory adipokines, strikingly alleviated the paracrine DNA damage response mediated by JAK2V617F-positive cells. Consistent with the result, exposure of hematopoietic cells to LCN2 resulted in elevated intracellular ROS levels and increased γ-H2AX foci formation. Collectively, these data demonstrate that LCN2 secreted from JAK2V617F-positive clones should be associated with the induction of oxidative DNA damage into neighboring cells in a paracrine fashion. Next, we explored how DNA damage is evoked by LCN2 exposure. Interestingly, we found that hematopoietic cells treated with LCN2 had significantly higher levels of intracellular iron, and the effect was further augmented with addition of FeSO4 in the medium. Furthermore, the intracellular ROS levels elevated by LCN2 were reverted to normal with co-treatment of iron chelating agent deferoxamine, suggesting that LCN2-triggered paracrine oxidative stress is mediated by intracellular iron overload. Finally, we studied the response of normal clones to increased oxidative stress. Importantly, we found that hematopoietic cells treated with JAK2V617F CM or LCN2 had increased phosphorylation of p53 and elevated expression of the pro-apoptotic genes regulated by p53 pathway, such as Bax, Noxa, and Puma. They also showed decreased cell proliferation and increased apoptosis rate. In contrast, when bone marrow cells derived from p53-deficient mice were treated with LCN2, they showed no significant change in apoptosis rate, suggesting that p53 pathway activation is required for the LCN2-treated cells to exclude themselves when severely damaged, and prevent the emergence of clones with various genetic aberrations. These results are consistent with clinical findings that p53 mutation, which might accelerate the DNA damage accumulation, is frequently seen in leukemic clones transformed from MPN. In summary, we demonstrate that JAK2V617F-positive cells evoke DNA damage to adjacent normal clones through LCN2-mediated iron overload, which is associated with increased risk for leukemogenesis from both JAK2V617F-positive and negative cells within MPN bone marrow. Disrupting these cascades presents a promising therapeutic approach for preventing the leukemic transformation from MPN. Disclosures: Kurokawa: Celgene: Consultancy, Research Funding; Bristol-Myers Squibb: Research Funding; Novartis: Consultancy, Research Funding.


PLoS ONE ◽  
2019 ◽  
Vol 14 (4) ◽  
pp. e0205626 ◽  
Author(s):  
Desirée Bartolini ◽  
Yanzhong Wang ◽  
Jie Zhang ◽  
Daniela Giustarini ◽  
Ranieri Rossi ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document