Knockout of lncRNA UCA1 inhibits drug resistance to gefitinib via targeting STAT3 signaling in NSCLC

2019 ◽  
Vol 110 (3) ◽  
Author(s):  
Biyuan Zhang ◽  
Haiji Wang ◽  
Qi Wang ◽  
Jinpeng Xu ◽  
Peng Jiang ◽  
...  
2020 ◽  
Vol 11 (12) ◽  
Author(s):  
Lakshana Sreenivasan ◽  
Hui Wang ◽  
Shyong Quin Yap ◽  
Pascal Leclair ◽  
Anthony Tam ◽  
...  

AbstractMedulloblastoma (MB) is a high-grade pediatric brain malignancy that originates from neuronal precursors located in the posterior cranial fossa. In this study, we evaluated the role of STAT3 and IL-6 in a tumor microenvironment mediated drug resistance in human MBs. We established that the Group 3 MB cell line, Med8A, is chemosensitive (hence Med8A-S), and this is correlated with a basal low phosphorylated state of STAT3, while treatment with IL-6 induced robust increases in pY705-STAT3. Via incremental selection with vincristine, we derived the stably chemoresistant variant, Med8A-R, that exhibited multi-drug resistance, enhanced IL-6 induced pY705-STAT3 levels, and increased IL6R expression. Consequently, abrogation of STAT3 or IL6R expression in Med8A-R led to restored chemosensitivity to vincristine, highlighting a prominent role for canonical IL-6/STAT3 signaling in acquired drug resistance. Furthermore, Med8A-S subjected to conditioning exposure with IL-6, termed Med8A-IL6+ cells, exhibited enhanced vincristine resistance, increased expression of pY705-STAT3 and IL6R, and increased secretion of IL-6. When cocultured with Med8A-IL6+ cells, Med8A-S cells exhibited increased pY705-STAT3 and increased IL-6 secretion, suggesting a cytokine feedback loop responsible for amplifying STAT3 activity. Similar IL-6 induced phenomena were also observed in the Group 3 MB cell lines, D283 and D341, including increased pY705-STAT3, drug resistance, IL-6 secretion and IL6R expression. Our study unveiled autocrine IL-6 as a promoter of STAT3 signaling in development of drug resistance, and suggests therapeutic benefits for targeting the IL-6/STAT3 signaling axis in Group 3 MBs.


Oncotarget ◽  
2010 ◽  
Vol 1 (1) ◽  
pp. 22-33 ◽  
Author(s):  
Lei Shi ◽  
Siqing Wang ◽  
Maurizio Zangari ◽  
Hongwei Xu ◽  
Thai M. Cao ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2895-2895
Author(s):  
Mark B Meads ◽  
Liang Nong ◽  
Linda Mathews ◽  
William S. Dalton ◽  
Kenneth H. Shain

Abstract Abstract 2895 Multiple myeloma is the second most common hematologic malignancy and remains incurable due to the eventual development of drug resistance. Therapy resistance has been linked to both acquired genetic changes as well as the dynamic influences of soluble and physical factors present in the microenvironment. We and others have shown that adhesion of tumor cells to the extracellular matrix component fibronectin (FN) via integrins leads to cell cycle arrest and protection from chemotherapy or Cell Adhesion-Mediated Drug Resistance (CAM-DR). The pleiotropic cytokine IL-6 has also been shown to mediate drug resistance in both solid and hematopoietic tumors [1]. To date, the influence of these two effectors on drug resistance has been studied separately; however, within the context of the bone marrow myeloma cells are influenced by both soluble and physical effectors of the microenvironment simultaneously. Using a reductionist model of the multivariant microenvironment, we recently demonstrated that unique collaborative signaling between FN-adhered cells and IL-6 leads to increased proliferation of protected tumor cells and a more malignant phenotype[2]. Our results demonstrate the FN-adhesion mediated cell-cycle arrest of myeloma cells was reversed following stimulation of adhered cells with IL-6. This phenotype was associated with a novel amplification of IL-6-induced STAT3 activation in adhered cells [2]. To further characterized the molecular events mediating this unique event we focused on the focal adhesion kinase, Proline-rich tYrosine Kinase 2 (Pyk2/FAK2). Pyk2 is a downstream intermediate of integrin signaling and has been demonstrated to amplify EGFR and cSrc-induced STAT3 activation. As such, we hypothesized that Pyk2 may be an important modulator of the enhanced STAT3 activation following multivariant signaling between beta1 integrins and gp130[3]. Our results link this amplification to Pyk2 in myeloma and murine pro-B cell lines. Western blot analysis demonstrated that Pyk2 autophophorylation on tyrosine 402 is induced following myeloma cell adhesion to FN correlating with the amplification of IL-6-induced STAT3, JAK1, and gp130 phosphorylation in both myeloma and pro B cell line models. Targeting Pyk2 with RNA interference attenuated the adhesion-associated amplification of STAT3 signaling as well as JAK1 and gp130 phosphorylation, but did not influence the limited STAT3 activation in cells grown in suspension. Further we demonstrated that Pyk2 kinase activity was similarly required for the activation of JAK1/STAT3 signaling under collaborative conditions, but not by IL-6 alone. Consistent with previous results ERK1/2 and Akt signaling were not affected by Pyk2 siRNA or kinase activity. Importantly, Pyk2 siRNA did not inhibit myeloma cell adhesion (n=4, p-value >0.05). Our previous results demonstrated that that the enhanced STAT3 signaling involved a FN-adhesion specific binding of unphosphorylated STAT3 with gp130 (independent of IL-6 stimulation). To determine if Pyk2 was similarly recruited to gp130 under co-stimulatory conditions we have initially used confocal microscopy. Confocal imaging with antisera to Pyk2 and gp130 demonstrated colocalization of the two effectors upon adhesion of myeloma cell to FN, but not in those grown in suspension. These data demonstrate that within the context of multivariant stimulation (IL-6 and FN-adhesion) a unique Pyk2-mediated JAK1/STAT3 signaling cascade is associated with unique biologic sequelae. We are currently exploring the biological sequelae of Pyk2-mediated collaborative signaling between integrins and IL-6. Key endpoints currently under investigation include proliferation and drug-response. These findings are significant because they suggests strategies targeting Pyk2 could be used to block cooperative signaling between integrins and IL-6, and inhibit the proliferation and/or therapy resistance conferred to tumor cells by the multivariant bone marrow niche. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 5101-5101
Author(s):  
Danielle C Croucher ◽  
Victor H Jimenez-Zepeda ◽  
Zhi Hua Li ◽  
Ellen Wei ◽  
Brent DG Page ◽  
...  

Abstract Abstract 5101 STAT3 is a cytoplasmic transcription factor, transiently activated in response to external stimuli such as growth factors and cytokines. As a transcription factor, STAT3 induces the expression of genes known to be involved in tumorigenesis, implicating STAT3 dysregulation in a number of hallmark oncogenic processes including tumor cell survival, proliferation, angiogenesis, metastasis, and drug resistance. Aberrant STAT3 signaling is prevalent in hematologic malignancies including Waldenstrom Macroglobulinemia (WM), a rare form of B cell non-Hodgkin lymphoma that is characterized by hyper-monoclonal IgM secretion in the peripheral blood. Despite the development of novel therapies and combinatorial treatment regimes, WM remains uniformly fatal, and as the limits of current chemotherapies have been reached, new approaches to treatment are urgently needed to improve patient outcome. Analysis of the gene and protein expression profiles of WM patients suggests that activation of STAT3 signaling plays a critical role in WM, providing rationale for the therapeutic use of STAT3 inhibitors. We have demonstrated pre-clinical efficacy of a novel, highly specific and potent small molecule STAT3/5 inhibitor, BP-1-102, in both hyper-IgM secreting B cell lymphoma cell lines (Mec-1 and RL) as well as two WM cell lines (MWCL-1 and BCWM-1). BP-1-102 directly targets STAT proteins by specifically blocking the SH2 domain that is a required for the phosphorylation, dimerization and nuclear localization of STAT3, ultimately resulting in inhibition of STAT3 transcriptional activation of target genes. We have shown that BP-1-102 directly interacts with STAT3's SH2 domain and is one of the most effective disruptors of STAT3 activity described to date. Treatment of cell lines with low μM doses of BP-1-102 induced dose-dependent decreases in constitutive and IL10-induced STAT3 phosphorylation (pSTAT3) as well as pSTAT3 nuclear localization. We further evaluated the potency of BP-1-102 against STAT1 and 5 compared to STAT3 using phosphor-flow cytometry to measure STAT phosphorylation status. BP-1-102 effectively inhibited GM-CSF induced STAT5 phosphorylation in AML2 cells at low dose concentrations (< 12.5uM) but only weakly inhibited IFNγ induced STAT1 phosphorylation in U937 leukemic cells at similar dose concentrations confirming the selectivity of BP-1-102 for STAT 3 and 5. Using a STAT3 dependent luciferase reporter construct, we confirmed repression of STAT3 transcriptional activity which correlated with a dose-dependent decrease in expression of STAT3 target genes (Mcl-1, Bcl-XL, Survivin and c-Myc). Inhibition of pSTAT3 resulted in decreased cell viability as assessed by MTT assay after 72 hours of in vitro exposure, with IC50 values ranging from 6uM to 10uM. In addition, treatment of cells with BP-1-102 resulted in caspase-dependent apoptosis which correlated with the activation of caspase-3 and PARP cleavage. Interestingly, co-culture of Mec-1 and RL with bone marrow stroma cells reduced the cytotoxicity of BP-1-102 suggesting stroma-conferred resistance, while MWCL-1 were equally sensitive to the cytotoxic effects of BP-1-102 regardless of either culture condition. Preliminary investigation suggests that the efflux system, used by cells to extrude toxic substances and linked to drug resistance in cancer, may be responsible for conferring stroma-mediate resistance to BP-1-102 in Mec-1 and RL cells. Finally, xenograft experiments to determine in vivo efficacy and safety are planned and will be presented. Collectively, these findings demonstrate a critical role for STAT3 signaling in WM pathology and provide the rationale for further development of STAT3 inhibitors for the treatment of WM. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 10 (10) ◽  
Author(s):  
Ruyi Xu ◽  
Yi Li ◽  
Haimeng Yan ◽  
Enfan Zhang ◽  
Xi Huang ◽  
...  

Abstract We previously showed that the chemokine CCL2 can recruit macrophages (Mφs) to the bone marrow (BM) in multiple myeloma (MM) and that myeloma-associated Mφs are important in drug resistance. Here, we explore the role of increased CCL2 expression in the BM microenvironment of MM and elucidate the underlying mechanism. Our results show that CCL2 expression is associated with the treatment status of MM patients. Mφs interact with MM cells and further upregulate their expression of CCL2. These increased level of CCL2 polarizes Mφs toward the M2-like phenotype and promotes Mφs to protect MM cells from drug-induced apoptosis. Mechanistically, CCL2 upregulated the expression of the immunosuppressive molecular MCP-1-induced protein (MCPIP1) in Mφs. MCPIP1 mediates Mφs’ polarization and protection via dual catalytic activities. Additionally, we found that CCL2 induces MCPIP1 expression via the JAK2-STAT3 signaling pathway. Taken together, our results indicate that increased CCL2 expression in MM patients’ BM polarizes Mφs toward the M2-like phenotype and promotes the protective effect of Mφs through MCPIP1, providing novel insight into the mechanism of Mφs-mediated drug resistance in MM.


2017 ◽  
Vol 37 (6) ◽  
Author(s):  
Hye-Sook Seo ◽  
Jin Mo Ku ◽  
Hee-Jae Lee ◽  
Jong-Kyu Woo ◽  
Chunhoo Cheon ◽  
...  

Overcoming drug resistance is an important task for investigators and clinician to achieve successful chemotherapy in cancer patients. Drug resistance is caused by various factors, including the overexpression of P-glycoprotein (P-gp, MDR1). The development of new, useful compounds that overcome drug resistance is urgent. SH003 is extracted from the mixture of three different herbs, and its anticancer effect has been revealed in different cancer cell types. In the present study, we investigated whether SH003 is able to reverse drug resistance using paclitaxel-resistant breast cancer cells (MCF-7/PAC). In our experiments, SH003 significantly decreased cell growth and colony formation in MCF-7/PAC cells and parental MCF-7 cells. This growth inhibition was related to the accumulation of cells in the sub-G0/G1 apoptotic population and an increase in the number of apoptotic cells. SH003 reduced the mRNA expression of multidrug resistance 1 (MDR1) and multidrug resistance-associated proteins (MRPs) in MCF-7/PAC cells. SH003 also down-regulated the expression of P-gp. SH003 reversed drug efflux from MCF-7/PAC cells, resulting in rhodamine123 (Rho123) accumulation. Inhibition of drug resistance by SH003 is related to the suppression of the signal transducer and activator of transcription 3 (STAT3) signaling pathway. SH003 decreased STAT3 activation (p-STAT3) and its nuclear translocation and inhibited the secretion of VEGF and MMP-2, which are STAT3 target genes. An STAT3 inhibitor, JAK inhibitor I and an HIF-1α inhibitor decreased cell growth in MCF-7 and MCF-7/PAC cells. Taken together, these results demonstrate that SH003 can overcome drug resistance, and SH003 might be helpful for chemotherapy in cancer patients.


2021 ◽  
Vol 1 (4) ◽  
pp. 100111
Author(s):  
Jin Mo Ku ◽  
Se Hyang Hong ◽  
Hyo In Kim ◽  
Min Jeong Kim ◽  
Su-Jeong Ku ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4021-4021
Author(s):  
Kenneth Shain ◽  
Mark Meads ◽  
Liang Nong ◽  
Jennifer Gemmer ◽  
Linda Mathews ◽  
...  

Abstract Abstract 4021 Multiple myeloma remains an incurable bone marrow resident malignancy, in part, due to the eventual development of drug resistance linked to both acquired genetic changes and the dynamic influences of the microenvironment. We and others have shown that adhesion of tumor cells to the extracellular matrix component fibronectin (FN) via integrins leads to cell cycle arrest and protection from chemotherapy or Cell Adhesion-Mediated Drug Resistance (CAM-DR). The pleiotropic cytokine IL-6 has also been shown to mediate drug resistance in both solid and hematopoietic tumors [1]. The influence of these two effectors on drug resistance has primarily been studied separately; however, within the context of the bone marrow myeloma cells are influenced by both soluble and physical effectors simultaneously. Using a reductionist model of the multivariant microenvironment, we demonstrated that unique collaborative signaling between FN-adhered cells and IL-6 leads to increased proliferation of protected tumor cells and a more malignant phenotype [2]. Our results demonstrate the FN-adhesion mediated cell-cycle arrest of myeloma cells was reversed following stimulation of adhered cells with IL-6 and drug resistance was maintained. This phenotype was associated with a novel amplification of IL-6-induced STAT3 activation in adhered cells [2]. We have since validated the FN-adhesion-dependent amplification of IL-6-induced STAT3 phosphorylation in myeloma patient specimens by flow cytometry. To further characterize post-translational events induced under multivariant conditions, we performed immune-affinity phosphotyrosine proteomic screening in RMPI8226 myeloma cells maintained in suspension or adhered to FN with or without IL-6 stimulation. Screening identified 338 differentially tyrosine phosphorylated peptides. Among these were proteins involved in signal transduction, cytoskeleton assembly, survival, and metabolic pathways. Proline-rich tYrosine Kinase 2 (Pyk2/FAK2) was highly phosphorylated at Y597 in FN-adhered RPMI8226 myeloma cells. Eighty-three phosphorylation events were up-regulated by adhesion, including paxillin at pY118 and p130CAS at pY249 indicating focal adhesion formation. Pyk2 is a downstream intermediate of integrin signaling and has been demonstrated to amplify EGFR and cSrc-induced STAT3 activation. As such, we hypothesized that Pyk2 may be an important modulator of the enhanced STAT3 activation following multivariant signaling between beta1 integrins and gp130 [3]. In addition to enhanced phosphorylation of Pyk2 at Y579 identified by proteomic screening, western blot analysis demonstrated Pyk2 autophosphorylation of Y402 following myeloma cell adhesion to FN correlating with the amplification of IL-6-induced STAT3 and JAK1. Targeting Pyk2 with RNA interference attenuated the adhesion-associated amplification of STAT3 and JAK1 phosphorylation, but did not influence the limited STAT3 activation in cells grown in suspension. Pyk2 siRNA did not inhibit myeloma cell adhesion (n=4, p-value >0.05). Our previous results demonstrated that that the enhanced STAT3 signaling involved a FN-adhesion specific binding of unphosphorylated STAT3 with gp130 (independent of IL-6 stimulation). To determine if Pyk2 was similarly recruited to gp130 under co-stimulatory conditions we have used proximal ligation assay (PLA). Confocal imaging of the PLA reaction with antisera to Pyk2 and gp130 demonstrated colocalization of the two effectors upon adhesion of patient myeloma cells to FN, but not in those grown in suspension. We have also demonstrated that STAT3 activation is markedly enhanced in myeloma cell lines adhered to patient bone marrow stroma (BMS), but not cells grown in transwell coculture (no myeloma cell-BMS contact). Critically, the protective advantage afforded myeloma cells in coculture was attenuated only by dual knockdown of STAT3 and Pyk2 using novel antisense oligonucleotides (ISIS Pharmaceuticals, n=4, p-value 0.027). These data demonstrate a novel Pyk2-mediated JAK1/STAT3 signaling cascade within the context of multivariant stimulation. Moreover, these data suggest that strategies targeting of both lateral and vertical signaling (Pyk2 & STAT3) may be required to overcome therapy resistance conferred to tumor cells by the multivariant bone marrow niche. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document