Abstract 1811: ERβ sensitizes NSCLC cells to chemotherapeutic agents by regulating DNA damage response

Author(s):  
Igor Bado ◽  
Fotis Nikolos ◽  
Jan-Åke Gustafsson ◽  
Christoforos Thomas
2021 ◽  
Vol 12 (12) ◽  
Author(s):  
Yu Mao ◽  
Yu Sun ◽  
Zhixuan Wu ◽  
Jingzhi Zheng ◽  
Jianing Zhang ◽  
...  

AbstractAberrant and exclusive expression of chromatin regulators in retinoblastoma (RB) in contrast to terminally differentiated normal retina presents a unique opportunity of selective targeting for RB. However, precise roles of these chromatin regulators in RB development and their potential as therapeutic targets have not been defined thoroughly. Here, we report that targeting of disruptor of telomeric silencing 1-like (DOT1L), a histone H3K79 methyltransferase, sensitizes RB cells to chemotherapeutic drugs by impairing the DNA damage response and thereby potentiating apoptosis while it is largely inefficacious as a single-agent therapy. Moreover, we identified high mobility group AT-hook 2 (HMGA2) as a novel DOT1L target gene in RB cells and found that its aberrant expression is dependent on DOT1L. As HMGA2 depletion reduced CHK1 phosphorylation during DNA damage response and augmented the drug sensitivity in RB cells, our results suggested that DOT1L targeting has a dual role in chemosensitization of RB cells by directly interfering with the immediate involvement of DOT1L in early DNA damage response upon genotoxic insults and also by downregulating the expression of HMGA2 as a rather late effect of DOT1L inhibition. Furthermore, we provide the first preclinical evidence demonstrating that combined therapy with a DOT1L inhibitor significantly improves the therapeutic efficacy of etoposide in murine orthotopic xenografts of RB by rendering the response to etoposide more potent and stable. Taken together, these results support the therapeutic benefits of DOT1L targeting in combination with other chemotherapeutic agents in RB, with mechanistic insights into how DOT1L targeting can improve the current chemotherapy in an RB cell-selective manner.


2021 ◽  
Vol 12 (12) ◽  
Author(s):  
Dawid Mehlich ◽  
Michał Łomiak ◽  
Aleksandra Sobiborowicz ◽  
Alicja Mazan ◽  
Dagmara Dymerska ◽  
...  

AbstractChemoresistance constitutes a major challenge in the treatment of triple-negative breast cancer (TNBC). Mixed-Lineage Kinase 4 (MLK4) is frequently amplified or overexpressed in TNBC where it facilitates the aggressive growth and migratory potential of breast cancer cells. However, the functional role of MLK4 in resistance to chemotherapy has not been investigated so far. Here, we demonstrate that MLK4 promotes TNBC chemoresistance by regulating the pro-survival response to DNA-damaging therapies. We observed that MLK4 knock-down or inhibition sensitized TNBC cell lines to chemotherapeutic agents in vitro. Similarly, MLK4-deficient cells displayed enhanced sensitivity towards doxorubicin treatment in vivo. MLK4 silencing induced persistent DNA damage accumulation and apoptosis in TNBC cells upon treatment with chemotherapeutics. Using phosphoproteomic profiling and reporter assays, we demonstrated that loss of MLK4 reduced phosphorylation of key DNA damage response factors, including ATM and CHK2, and compromised DNA repair via non-homologous end-joining pathway. Moreover, our mRNA-seq analysis revealed that MLK4 is required for DNA damage-induced expression of several NF-кB-associated cytokines, which facilitate TNBC cells survival. Lastly, we found that high MLK4 expression is associated with worse overall survival of TNBC patients receiving anthracycline-based neoadjuvant chemotherapy. Collectively, these results identify a novel function of MLK4 in the regulation of DNA damage response signaling and indicate that inhibition of this kinase could be an effective strategy to overcome TNBC chemoresistance.


2018 ◽  
Vol 2018 ◽  
pp. 1-21 ◽  
Author(s):  
Pierpaola Davalli ◽  
Gaetano Marverti ◽  
Angela Lauriola ◽  
Domenico D’Arca

Cancer is a death cause in economically developed countries that results growing also in developing countries. Improved outcome through targeted interventions faces the scarce selectivity of the therapies and the development of resistance to them that compromise the therapeutic effects. Genomic instability is a typical cancer hallmark due to DNA damage by genetic mutations, reactive oxygen and nitrogen species, ionizing radiation, and chemotherapeutic agents. DNA lesions can induce and/or support various diseases, including cancer. The DNA damage response (DDR) is a crucial signaling-transduction network that promotes cell cycle arrest or cell death to repair DNA lesions. DDR dysregulation favors tumor growth as downregulated or defective DDR generates genomic instability, while upregulated DDR may confer treatment resistance. Redox homeostasis deeply and capillary affects DDR as ROS activate/inhibit proteins and enzymes integral to DDR both in healthy and cancer cells, although by different routes. DDR regulation through modulating ROS homeostasis is under investigation as anticancer opportunity, also in combination with other treatments since ROS affect DDR differently in the patients during cancer development and treatment. Here, we highlight ROS-sensitive proteins whose regulation in oxidatively induced DDR might allow for selective strategies against cancer that are better tailored to the patients.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3668-3668
Author(s):  
Min Wu ◽  
Li Li ◽  
Max Hamaker ◽  
Donald Small ◽  
Amy S. Duffield

Abstract Acute myeloid leukemia (AML) is an aggressive hematologic neoplasm characterized by clonal expansion of myeloid blasts. The FMS-like tyrosine kinase-3 (FLT3) receptor gene is the most commonly mutated gene in AML, and patients who harbor a FLT3/ITD mutation have a relatively poor prognosis. Utilizing a co-immunoprecipitation/mass spectrometry analysis, we found that FLT3/ITD interacts with Dedicator of Cytokinesis 2 (DOCK2), which is a guanine nucleotide exchange factor for Rac GTPases. Expression of DOCK2 is limited to hematopoietic cells, and it is expressed in leukemic blasts of patients with FLT3/ITD AML. Knockdown (KD) of DOCK2 results in reduced Rac1 activity and leads to decreased survival of leukemic cells with elevated FLT3 activity, both alone and in combination with cytarabine (Ara-C) treatment. Moreover, DOCK2 KD in transplanted FLT3/ITD leukemic cells prolongs disease progression in a mouse xenograft model. We further investigated the mechanisms by which DOCK2 affects cell proliferation and response to chemotherapeutic agents in FLT3/ITD leukemic cells. DOCK2 KD via shRNA in a leukemia cell line that expresses FLT3/ITD (MV4;11) resulted in significantly elevated sensitivity towards Ara-C treatment, as indicated by a markedly increased apoptosis rate and reduced cell proliferation rate. Moreover, the survival of immunodeficient NSG mice transplanted with DOCK2 KD MV4;11 cells was prolonged after treatment with a relatively low dose of Ara-C, in contrast to no survival benefit observed in mice transplanted with control cells. To investigate the mechanism of the increased sensitivity to Ara-C, we used a BrdU incorporation assay to investigate cell proliferation +/- drug treatment. Both control and DOCK2 KD cells showed arrested DNA synthesis and proliferation in response to Ara-C via the activation of DNA damage response (DDR), as evidenced by activation of the DDR effectors Chk1 and Wee1. The control cells continued to synthesize DNA and proliferate, albeit at a reduced rate, following a brief partial arrest; However, DNA synthesis was completely abrogated in DOCK2 KD MV4;11 cells within two hours of Ara-C treatment, and the arrest of DNA replication and cell proliferation persisted until apoptosis was initiated. As the resumption of DNA synthesis and cell proliferation requires efficient repair of damaged DNA, the persistent cell cycle arrest in DOCK2 KD cells suggests a deficiency in DNA repair. To investigate the roles that DOCK2 plays in DNA repair, we quantified DNA damage via γH2AX levels. Western blot analysis revealed a progressive elevation of γH2AX levels in DOCK2 KD MV4;11 cells after Ara-C treatment, while the levels remained stable in control cells. Furthermore, DOCK2 deficiency in MV4;11 cells also led to reduced activity of Wee1 and Chk1, as well as decreased levels of Rad51, indicating multifaceted regulatory effects of DOCK2 on DNA repair pathways. Another suggestion as to the function of the DOCK2/Rac1 pathway comes from the differential response of DOCK2 KD cells treated with Ara-C and 5-fluorouracil (5-FU). While DOCK2 KD MV4;11 cells exhibited decreased proliferation and elevated apoptosis when treated with Ara-C, they demonstrated a significantly higher proliferation rate and lower apoptosis rate than control cells when treated with 5-FU. In contrast to Ara-C, treatment of DOCK2 KD MV4;11 cells with 5-FU resulted in stable γH2AX levels while γH2AX levels increased in control cells. These effects appear to be dependent on elevated FLT3 activity, as REH cells that express wild-type FLT3 did not show any observable difference in response to Ara-C or 5-FU. Interestingly, this differential response to Ara-C and 5-FU is characteristic of mismatch repair (MMR)-deficient colorectal cancer cells, which have decreased activity of key MMR components, such as MSH2 and MLH1. We found that levels of both MSH2 and MLH1 were significantly reduced in DOCK2 KD MV4;11 cells, suggesting that the MMR may also play a role in the response of FLT3/ITD leukemic cells to Ara-C. Our observations suggest that FLT3/ITD-mediated differences in DDR and MMR may play a role in the relatively poor response of FLT3/ITD AML to standard AML chemotherapeutic regimens. Thus, the addition of DDR inhibitors may be useful in the treatment of FLT3/ITD AML, and pharmacologic inhibition of the Rac signaling pathways via DOCK2 provides a novel and promising therapeutic target for FLT3/ITD AML. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 19 (1) ◽  
pp. 595-611
Author(s):  
Yuan Yang ◽  
◽  
Lingshan Zhou ◽  
Xi Gou ◽  
Guozhi Wu ◽  
...  

<abstract> <sec><title>Objective</title><p>Gastric cancer (GC) is the fifth most common malignancy and the fourth leading cause of cancer-related mortality worldwide. The identification of valuable predictive signatures to improve the prognosis of patients with GC is becoming a realistic prospect. DNA damage response-related long noncoding ribonucleic acids (drlncRNAs) play an important role in the development of cancers. However, their prognostic and therapeutic values remain sparse in gastric cancer (GC).</p> </sec> <sec><title>Methods</title><p>We obtained the transcriptome data and clinical information from The Cancer Genome Atlas Stomach Adenocarcinoma (TCGA-STAD) cohort. Co-expression network analyses were performed to discover functional modules using the igaph package. Subsequently, lncRNA pairs were identified by bioinformation analysis, and prognostic pairs were determined by univariate analysis, respectively. In addition, we utilized least absolute shrinkage and selection operator (LASSO) cox regression analysis to construct the risk model based on lncRNA pairs. Then, we distinguished between the high- or low- risk groups from patients with GC based on the optimal model. Finally, we reevaluated the association between risk score and overall survival, tumor immune microenvironment, specific tumor-infiltrating immune cells related biomarkers, and the sensitivity of chemotherapeutic agents.</p> </sec> <sec><title>Results</title><p>32 drlncRNA pairs were obtained, and a 17-drlncRNA pairs signature was constructed to predict the overall survival of patients with GC. The ROC was 0.797, 0.812 and 0.821 at 1, 2, 3 years, respectively. After reclassifying these patients into different risk-groups, we could differentiate between them based on negative overall survival outcome, specialized tumor immune infiltration status, higher expressed immune cell related biomarkers, and a lower chemotherapeutics sensitivity. Compared with previous models, our model showed better performance with a higher ROC value.</p> </sec> <sec><title>Conclusion</title><p>The prognostic and therapeutic signature established by novel lncRNA pairs could provide promising prediction value, and guide individual treatment strategies in the future.</p> </sec> </abstract>


Diabetes ◽  
2020 ◽  
Vol 69 (Supplement 1) ◽  
pp. 2118-P
Author(s):  
CHAY TENG YEO ◽  
BRYNDON OLESON ◽  
JOHN A. CORBETT ◽  
JAMIE K. SCHNUCK

Sign in / Sign up

Export Citation Format

Share Document