scholarly journals Ym155 Induces Oxidative Stress-Mediated DNA Damage and Cell Cycle Arrest, and Causes Programmed Cell Death in Anaplastic Thyroid Cancer Cells

2021 ◽  
Vol 22 (4) ◽  
pp. 1961
Author(s):  
Qinqin Xu ◽  
Ryan P. Mackay ◽  
Adam Y. Xiao ◽  
John A. Copland ◽  
Paul M. Weinberger

Anaplastic thyroid cancer (ATC) is one of the most lethal malignancies with a median survival time of about 4 months. Currently, there is no effective treatment, and the development of new therapies is an important and urgent issue for ATC patients. YM155 is a small molecule that was identified as the top candidate in a high-throughput screen of small molecule inhibitors performed against a panel of ATC cell lines by the National Cancer Institute. However, there were no follow-up studies investigating YM155 in ATC. Here, we determined the effects of YM155 on ATC and human primary benign thyroid cell (PBTC) survival with alamarBlue assay. Our data show that YM155 inhibited proliferation of ATC cell lines while sparing normal thyroid cells, suggesting a high therapeutic window. YM155-induced DNA damage was detected by measuring phosphorylation of γ-H2AX as a marker for DNA double-strand breaks. The formamidopyrimidine-DNA glycosylase (FPG)-modified alkaline comet assay in conjunction with reactive oxygen species (ROS) assay and glutathione (GSH)/glutathione (GSSG) assay suggests that YM155-mediated oxidative stress contributes to DNA damage. In addition, we provide evidence that YM155 causes cell cycle arrest in S phase and in the G2/M transition and causes apoptosis, as seen with flow cytometry. In this study, we show for the first time the multiple effects of YM155 in ATC cells, furthering a potential therapeutic approach for ATC.

2018 ◽  
Vol 25 (5) ◽  
pp. 585-594.e7 ◽  
Author(s):  
Kenichi Shimada ◽  
Eduard Reznik ◽  
Michael E. Stokes ◽  
Lakshmi Krishnamoorthy ◽  
Pieter H. Bos ◽  
...  

2020 ◽  
Author(s):  
Xi Su ◽  
Jiaxin Liu ◽  
Haihong Zhang ◽  
Qingqing Gu ◽  
Xinrui Zhou ◽  
...  

Abstract Background Anaplastic thyroid cancer (ATC) is a kind of rare thyroid cancer with very poor prognosis. It is one of the deadliest cancers in human due to the aggressive behavior and resistance to treatment. Doxorubicin has been approved in ATC treatment as a single agent, but monotherapy still shows no improvement of the total survival in advanced ATC. Lenvatinib was investigated with encouraging results in treating the patients with radioiodine-refractory differentiated thyroid cancer (DTC). However, antitumor efficacy of combination therapy with lenvatinib and doxorubicin remains largely unclear. Methods The antitumor efficacy of combination therapy with lenvatinib and doxorubicin on ATC cell proliferation and was assessed by the MTT assay and colony formation. Flow cytometry were employed to assess ATC cells’ apoptosis and cell cycle arrest in response to combination therapy. Xenograft models were used to test its in vivo antitumor activity. Result Lenvatinib monotherapy was less effective than doxorubicin in treating ATC cell lines and xenografts model. The combination therapy of lenvatinib and doxorubicin significantly inhibited ATC cell proliferation and tumor growth in nude mice, and induced cell apoptosis and cell cycle arrest in compared to lenvatinib or doxorubicin monotherapy. Conclusion Lenvatinib promotes the antitumor effects of doxorubicin in ATC cell and xenografts model. Lenvatinib/doxorubicin combination may be a potential candidate therapeutic approach for ATC.


2018 ◽  
Author(s):  
Kenichi Shimada ◽  
Eduard Reznik ◽  
Michael E. Stokes ◽  
Lakshmi Krishnamoorthy ◽  
Pieter H. Bos ◽  
...  

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1366-1366 ◽  
Author(s):  
Watkins M Krystal ◽  
Russell Walker ◽  
Nathan Fishkin ◽  
Charlene Audette ◽  
Yelena Kovtun ◽  
...  

Abstract IMGN779 is a CD33-targeting ADC consisting of a humanized anti-CD33 antibody, Z4681A, conjugated to DGN462, a novel DNA-alkylating agent, through a cleavable disulfide linker, sulfo-SPDB. CD33 is broadly expressed on leukemic blasts of patients with AML, making it a promising target for AML therapy. DGN462 is a member of the novel IGN class of DNA-acting cytotoxic agents, that consists of an indolino-benzodiazepine dimer containing a mono-imine moiety. Potent killing of AML tumor cells by DGN462 and IMGN779 has previously been demonstrated in vitro (K. Whiteman et. al, ASH 2014 #2321). Here we describe studies elucidating the mechanism of action of this novel payload in the AML cell lines: MV4-11, HL60, and EOL-1. The ability of DGN462 to alkylate DNA was demonstrated in an AML cell line by isolation and analysis of the genomic DNA from cells. DGN462 was found to co-purify with DNA, demonstrating that it covalently reacts with cellular DNA. A comet assay was performed confirming that DGN462 does not cross-link DNA. H2AX is known to be phosphorylated on serine 139 in response to DNA damage. Using flow cytometry, we assessed the phosphorylation of H2AX in AML cell lines following exposure to DGN462 and IMGN779. Increases in phosphorylated H2AX levels were detected as early as 4 hours following exposure to DGN462-SMe and 12 hours following exposure to IMGN779. Additional flow cytometry analysis showed that exposure to DGN462-SMe and IMGN779 leads to S-phase accumulation, G2/M arrest, followed by induction of apoptotic markers (cleaved PARP and cleaved Caspase-3) at later time points. Ex vivo studies using AML patient samples also showed elevation of phosphorylated H2AX and an increase in apoptosis in myeloid blasts following exposure to DGN462-SMe and IMGN779. As a demonstration of proof-of-mechanism in AML disease models, our results suggest that cell killing by IMGN779 is mediated by DNA damage, as a consequence of DNA alkylation. The DNA damage response is accompanied by cell cycle arrest, which leads to apoptosis. This relationship will be further explored in additional AML preclinical models to support the use of phosphorylated H2AX as a pharmacodynamic biomarker for IMGN779 activity in future clinical studies. Disclosures Krystal: ImmunoGen, Inc.: Employment, Equity Ownership. Walker:ImmunoGen, Inc.: Employment, Equity Ownership. Fishkin:ImmunoGen, Inc.: Employment, Equity Ownership. Audette:ImmunoGen, Inc.: Employment, Equity Ownership. Kovtun:ImmunoGen, Inc.: Employment, Equity Ownership. Romanelli:ImmunoGen, Inc.: Employment, Equity Ownership.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1655-1655
Author(s):  
Simone Boehrer ◽  
Lionel Ades ◽  
Nicolas Tajeddine ◽  
Lorenzo Galluzzi ◽  
Stephane de Botton ◽  
...  

Abstract Background: The hypomethylating agents azacytidine (AZA) and decitabine (DEC) have shown clinical efficacy in patients (pts) with MDS. There is in vitro evidence that both agents, in addition to their hypomethylating effect, also function by inducing apoptosis, cell cycle arrest and/or the activation of a DNA damage response (DDR). However, the exact contributions of those mechanisms of action and their functional interdependence remain to be defined. Methods: A panel of MDS (P39, MDS-1)- and AML (HL-60, KG-1)-derived cell lines were incubated with increasing dosages of AZA (1–2μM) and DEC (1–2μM) and the drugs capacity to induce apoptosis (DiOC6(3)/PI), cell cycle arrest (PI) and/or a DDR (immunoflourescence staining of P-ATM, P-Chk-1, P-Chk-2, γ-H2AX) were assessed in absence and presence of the ATM-inhibitor KU-55933 and the Chk-1 inhibitor UCN-01. Results: We show that both drugs induced dose-dependent apoptosis in myeloid cell lines: whereas AZA increased apoptosis in KG-1 and HL-60 by about 10% (48h, 2μM) the respective incubation with DEC augmented apoptosis by about 20% (HL-60) to 30% (KG-1). P39 cells were resistant to AZA and increased apoptosis by 15% after 48h of 2μM DEC, and MDS-1 cells were resistant to both drugs. In addition, both drugs induced a G2/M-arrest in P39 (+15% after 48h with 2μM of AZA or DEC) and HL-60 (+20% after 48h with 2μM of AZA or DEC) cells, but not in KG-1 and MDS-1 cells. Noteworthy, both drugs induced a DDR in the apoptosis-sensitive KG-1 cells (but not P39 cells) as evidenced by the appearance of nuclear P-ATM and γ-H2AX foci. Surprisingly, this activation of P-ATM did not induce the nuclear translocation of P-Chk-1-Ser317 or P-Chk-2-Ser68. To more clearly define the importance of the DDR in AZA- and DEC-induced apoptosis and G2/M-arrest, experiments were recapitulated in the presence of the ATM-inhibitor KU-55933 and the Chk-1 inhibitor UCN-01. Inhibition of ATM abrogated the apoptosis-inducing activity of AZA and DEC in KG-1 cells (without influencing cell cycle progression), whereas inhibition of Chk-1 remained without effect. In contrast, in P39 and HL-60 cells, inhibition of ATM neither affected cell cycle progression, nor sensitivity towards the drugs. Nevertheless, inhibition of Chk-1 by UCN-01 completely abrogated the G2/M-arresting effect of AZA (and diminished that of DEC) in P39 and HL-60 cells. Conclusions: We provide novel evidence for the cell-type dependent capacity of the hypomethylating agents 5-azacytidine and decitabine to induce apoptosis, cell-cycle arrest and DDR in cell lines representing different subtypes of MDS and AML. Moreover, we show the crucial role of ATM and Chk-1 activation – as part of the DDR – in mediating AZA and DEC apoptosis-inducing and cell cycle-arresting effects, respectively, providing evidence that hypomethylating agents confer their beneficial effects by employing different pathways of the DDR.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1833-1833
Author(s):  
Ken Maes ◽  
Miguel Lemaire ◽  
Jordan Gauthier ◽  
Hendrik De Raeve ◽  
Eline Menu ◽  
...  

Abstract Abstract 1833 Multiple myeloma (MM) is still an incurable plasma cell malignancy, thus highlighting the need for alternative treatment options. Currently, strategies for therapy are being developed targeting epigenetic modification using epigenetic modulating agents like histone deacetylase inhibitors (HDACi) and DNA methyltransferase inhibitors (DNMTi). 5-aza-2'-deoxycitidine or decitabine (DAC) is a DNMTi and is FDA approved for treatment of myelodysplastic syndrome and has beneficial clinical effects against leukemia. The anti-tumor effects are ascribed to two non-mutual exclusive modes of action. Relative low doses are thought to lead to passive CpG demethylation resulting in re-expression of genes silence by DNA methylation and apoptosis, while relative high doses are cytotoxic by inducing a DNA damage response together with cell cycle arrest and apoptosis. In multiple myeloma (MM), preclinical data regarding the effects of DAC is, however, limited. Therefore, we investigated the cytotoxic effects of DAC in MM both in vitro and in vivo. In addition, we evaluated the combination of DAC with the pan-HDAC inhibitor JNJ-26481585. First, we assessed the effects of DAC on cell cycle progression and apoptosis on a panel of MM cell lines. We used one murine (5T33MMvt) and 5 human (OPM-2, RPMI 8226, LP-1, KMS-11 and NCI-H929) MM cell lines. In general, DAC could affect cell cycle progression by inducing either a G0/G1-phase arrest or a G2/M-phase arrest. The 5T33MMvt and LP-1 cells were arrested in the G2/M-phase, while OPM-2 and NCI-H929 cells underwent a G0/G1-phase arrest. Subsequently, apoptosis occurred in all cell lines. Interestingly, the 5T33MMvt cells were relatively sensitive, as nM doses of DAC were sufficient to induce massive apoptosis in a relative short incubation time (2 days). The human cell lines were less sensitive since higher doses (μM range) and longer incubation time (3–5 days) were necessary to induce apoptosis, with the OPM-2 cells being the least sensitive. To determine the potential mechanisms more in detail, we focused on the 5T33MMvt and OPM-2 cells. In both cell lines, DAC-mediated apoptosis was associated with caspase activation and PARP cleavage, Bim upregulation and posttranslational changes in Mcl-1 expression. The G2/M-phase arrest in the 5T33MMvt cells was accompanied by phosphorylation of CDK-1 and an increase in cyclinB1 expression. In both cell lines, p27 protein expression was increased, what may contribute to the cell cycle arrest. Furthermore, in the 5T33MMvt cells, a DNA damage response was activated as evidenced by a clear induction of ATM and H2AX phosphorylation. This was not the case for the OPM-2 cells, in which we observed no ATM activation and only a modest H2AX phosphorylation upon DAC treatment. In addition, the tumor suppressor p53 was phosphorylated on ser15 upon DAC treatment in both cell lines, indicating a potential role of p53. However, a p53 inhibitor, pifithrin-α, could not abrogate DAC-induced apoptosis indicating that p53 transactivation is not essential in this process. Next, we used the syngeneic 5T33 murine MM model (5T33MM) to investigate the in vivo effects of DAC. 5T33MM mice were daily treated with 0.1, 0.2 and 0.5 mg/kg DAC. We observed a significant decrease in serum M-protein, bone marrow plasmacytosis and spleno- and hepatomegaly compared to vehicle treated mice. These effects led to a significant increase in survival probability of DAC treated mice (p≤0.001). Lastly, we evaluated the possibility of combining DAC with a pan-HDAC inhibitor JNJ-26481585 (JNJ-585). DAC and JNJ-585 synergistically induced cell death in RPMI-8226, OPM-2 and 5T33MMvt cells. We further demonstrated the combinatory effects of DAC and JNJ-585 in the 5T33MM murine model. Here, we observed enhanced effects of DAC and JNJ-585 on serum M-protein, BM tumor load and survival (p≤0.001) compared to either agent alone. In conclusion, DAC shows potent anti-MM effects both in vitro and in vivo. Mechanistically, we observed induction of a DNA damage response and/or cell cycle arrest. Apoptosis was caspase-mediated but independent of the transactivation of p53. DAC was also efficient in the murine 5T33MM model in which DAC treatment led to a survival benefit. In addition, DAC showed useful in a combination with the HDAC inhibitor JNJ-585. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 42 (1) ◽  
Author(s):  
Ako Matsui ◽  
Kazunari Hashiguchi ◽  
Masao Suzuki ◽  
Qiu-Mei Zhang-Akiyama

Abstract Background DNA damage is generated by various intrinsic and extrinsic sources such as reactive oxygen species (ROS) and environmental mutagens, and causes genomic alterations. DNA damage response (DDR) is activated to induce cell cycle arrest and DNA repair. Oxidation resistance 1 (OXR1) is a protein that defends cells against oxidative stress. We previously reported that OXR1 protein functions in the regulation of G2-phase cell cycle arrest in cells irradiated with gamma-rays, suggesting that OXR1 directly responds to DNA damage. Purpose To clarify the functions of OXR1 against ROS-independent DNA damage, HeLa and OXR1-depleted HeLa cells were treated with heavy-ion beams and the ROS-independent DNA-damaging agent methyl methanesulfonate (MMS). Results First, OXR1-depleted cells exhibited higher sensitivity to MMS and heavy-ion beams than control cells. Next, OXR1 depletion increased micronucleus formation and shortened the duration of G2-phase arrest after treatment with MMS or heavy-ion beams. These results suggest that OXR1 functions in the maintenance of cell survival and genome stability in response to DNA damage. Furthermore, the OXR1 protein level was increased by MMS and heavy-ion beams in HeLa cells. Conclusions Together with our previous study, the present study suggests that OXR1 plays an important role in the response to DNA damage, not only when DNA damage is generated by ROS.


Genes ◽  
2018 ◽  
Vol 9 (10) ◽  
pp. 513 ◽  
Author(s):  
Andrea Maria Guarino ◽  
Annaelena Troiano ◽  
Elio Pizzo ◽  
Andrea Bosso ◽  
Maria Vivo ◽  
...  

The prototype cold-shock Y-box binding protein 1 (YB-1) is a multifunctional protein that regulates a variety of fundamental biological processes including cell proliferation and migration, DNA damage, matrix protein synthesis and chemotaxis. The plethora of functions assigned to YB-1 is strictly dependent on its subcellular localization. In resting cells, YB-1 localizes to cytoplasm where it is a component of messenger ribonucleoprotein particles. Under stress conditions, YB-1 contributes to the formation of stress granules (SGs), cytoplasmic foci where untranslated messenger RNAs (mRNAs) are sorted or processed for reinitiation, degradation, or packaging into ribonucleoprotein particles (mRNPs). Following DNA damage, YB-1 translocates to the nucleus and participates in DNA repair thereby enhancing cell survival. Recent data show that YB-1 can also be secreted and YB-1-derived polypeptides are found in plasma of patients with sepsis and malignancies. Here we show that in response to oxidative insults, YB-1 assembly in SGs is associated with an enhancement of YB-1 protein secretion. An enriched fraction of extracellular YB-1 (exYB-1) significantly inhibited proliferation of receiving cells and such inhibition was associated to a G2/M cell cycle arrest, induction of p21WAF and reduction of Np63 protein level. All together, these data show that acute oxidative stress causes sustained release of YB-1 as a paracrine/autocrine signal that stimulate cell cycle arrest.


Sign in / Sign up

Export Citation Format

Share Document