K562 Cell Sensitization to 5-Fluorouracil- or Interferon-Alpha-Induced Apoptosis Via Cordycepin (3′-Deoxyadenosine): Fine Control of Cell Apoptosis Via Poly(A) Polymerase Upregulation

2004 ◽  
Vol 19 (1) ◽  
pp. 58-66
Author(s):  
G.C. Lallas ◽  
N. Courtis ◽  
M. Havredaki

K562 cells represent a classical model for the study of drug resistance. Induction of apoptosis is accompanied by concomitant distinct modulations of poly(A) polymerase (PAP) and other proteins involved in mRNA maturation. Recent data suggest the involvement of mRNA stability in the induction of specific apoptosis pathways. In this study we used a specific polyadenylation inhibitor, cordycepin (3-deoxyadenosine), to investigate the involvement of polyadenylation in K562 cell apoptosis and drug resistance. The combination of cordycepin with either 5-fluorouracil or interferon-alpha sensitized chemoresistant K562 cells to apoptosis. This sensitization was followed by distinct PAP modulations before and after the appearance of characteristic apoptosis pointers (DNA laddering, DAPI staining, mitochondrial transmembrane potential). PAP modulations appeared essential for K562 sensitization. mRNA polyadenylation therefore seemed to be involved not only in apoptosis but also in drug resistance. Polyadenylation inhibition by cordycepin under certain conditions sensitized chemoresistant K562 cells to apoptosis and thus polyadenylation could prove to be a fine target for overcoming drug resistance.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4414-4414
Author(s):  
Xiao-hong Zhang ◽  
Li-da Su ◽  
Xiao-ying Zhao ◽  
Qing-hua Lv

Abstract Summery: Anti-apoptosis is one of drug resistance mechanisms in leukemic cells. It was found in our early study that As2O3 can induce apoptosis of K562 cells, and this effect involve the degradation of IκB-αand consequently the activation of NF-κB. The relationship between drug resistance of leukemic cells and the expression of both IκB-αand NF-κB associated with apoptosis induced by arsenic trioxide(As2O3) was studied in K562 and K562/ADR cells. Methods: Apoptosis was induced in K562 and K562/ADR cells cultured with As2O3 in different concentrations. Western blot was used to analyze the expression of NF-κB in nuclear and IκB-α in cytoplasm of these cells. Apoptosis and degradation of IκB-αprotein were also observed by flow cytometry. Results: The suppressive effect of As2O3 on proliferation of K562/ADR was lower than that in K562 cell, IC50 values were 19.07μmol/L and 5.26μmol/L, respectively. After exposure to As2O3, the ratio of apoptosis cells increased with the concentration of As2O3 in K562 cells, from(13.25±1.83)% to (50.56±8.62)% with variation of As2O3 from 1μmol/L to 4μmol/L(P<0.05). The ratio of apoptosis cells in K562/ADR cultured with 4μmol/L As2O3 was significantly lower than that in K562 cells, (8.00±1.47)% vs. (50.56±8.62)%, (P<0.05). The level of IκB-α in K562 cytoplasm was down-regulated from (88.07±0.99)% to (49.21±0.95)%, (P<0.01) after As2O3 stimulation, while NF-κB in nuclear was up-regulated, that was not found in K562/ADR cells. Conclusion: As2O3 could induce apoptosis of K562 cells, associated with the degradation of IκB-αand the activation of NF-κB. There were resistance to As2O3 induced apoptosis and an abnormal regulation of NF-κB expression by As2O3 in K562/ADR cells.


Author(s):  
Weilan Lan ◽  
Jinyan Zhao ◽  
Wujin Chen ◽  
Haixia Shang ◽  
Jun Peng ◽  
...  

Background: Anlotinib is a multi-target tyrosine kinase inhibitor that has been reported to have activity against colorectal cancer. However, the mechanisms of how anlotinib mediates drug-resistance of colorectal cancer have not been fully described. Particularly the potential mechanisms regarding to the inhibition of proliferation and induction of apoptosis remain unknown. Objective: In this study, we intended to study the effect and related-mechanism of the proliferation, migration, invasion and induced apoptosis of anlotinib overcoming multidrug resistant colorectal cancer cells through in vitro experiments. Methods: Cell viability was determined by MTT assays and the resistant index was calculated. Colony formation and PI/RNase Staining were used for testing the proliferation of resistant cells. DAPI staining and Annexin V-FITC/PI staining were used to detect cell apoptosis. Migration and invasion were examined by transwell. Protein expression and activation of PI3K/AKT pathway were detected by western blot. LY294002 was used to verify whether anlotinib overcomes the drug-resistance of CRC cells by inactivating the PI3K/AKT pathway. Results: The results showed that the HCT-8/5-FU cells were resistant to multiple chemotherapy drugs (5-FU, ADM and DDP). Anlotinib significantly inhibited the cell viability, proliferation, migration, invasion and induced the cell apoptosis. Moreover, anlotinib downregulated the expression of survivin, cyclin D1, CDK4, caspase-3, Bcl-2, MMP-2, MMP-9, vimentin and N-cadherin, but up-regulated cleaved-caspase-3, Bax and E-cadherin and blocked the activity of the PI3K/AKT in HCT-8/5-FU cells. We found anlotinib and LY294002 overcame the drug resistance of HCT-8/5-FU cells by reducing the expression of PI3K/p-AKT. Conclusions: Anlotinib inhibited the proliferation, migration, invasion and induced apoptosis of HCT-8/5-FU cells, and the mechanisms may be that anlotinib conquered multidrug resistance of colorectal cancer cells via inactivating of PI3K/AKT pathway.


2020 ◽  
Vol 11 (11) ◽  
Author(s):  
Jie-hui Tan ◽  
Rong-chang Cao ◽  
Lei Zhou ◽  
Zhi-tao Zhou ◽  
Huo-ji Chen ◽  
...  

Abstract Treatment of acute pancreatitis (AP) and chronic pancreatitis (CP) remains problematic due to a lack of knowledge about disease-specific regulatory targets and mechanisms. The purpose of this study was to screen proteins related to endoplasmic reticulum (ER) stress and apoptosis pathways that may play a role in pancreatitis. Human pancreatic tissues including AP, CP, and healthy volunteers were collected during surgery. Humanized PRSS1 (protease serine 1) transgenic (PRSS1Tg) mice were constructed and treated with caerulein to mimic the development of human AP and CP. Potential regulatory proteins in pancreatitis were identified by proteomic screen using pancreatic tissues of PRSS1Tg AP mice. Adenoviral shRNA-mediated knockdown of identified proteins, followed by functional assays was performed to validate their roles. Functional analyses included transmission electron microscopy for ultrastructural analysis; qRT-PCR, western blotting, co-immunoprecipitation, immunohistochemistry, and immunofluorescence for assessment of gene or protein expression, and TUNEL assays for assessment of acinar cell apoptosis. Humanized PRSS1Tg mice could mimic the development of human pancreatic inflammatory diseases. EMC6 and APAF1 were identified as potential regulatory molecules in AP and CP models by proteomic analysis. Both EMC6 and APAF1 regulated apoptosis and inflammatory injury in pancreatic inflammatory diseases. Moreover, APAF1 was regulated by EMC6, induced apoptosis to injure acinar cells and promoted inflammation. In the progression of pancreatitis, EMC6 was activated and then upregulated APAF1 to induce acinar cell apoptosis and inflammatory injury. These findings suggest that EMC6 may be a new therapeutic target for the treatment of pancreatic inflammatory diseases.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5048-5048
Author(s):  
Jin-Hwang Liu ◽  
Chin-Cheng Liu ◽  
Chueh-Chuan Yen ◽  
Jyh-Pyng Gau ◽  
Cheng-Hwai Tzeng

Abstract Imatinib induces apoptosis in chronic myeloid leukemic (CML) cells even with mutant p53. Ras provokes promyelocytic leukemia protein (pml), which promotes apoptosis and senescence in untransformed cells. In CML cells, ras is usually activated; however, the function of pml seems inadequate to induce apoptosis without imatinib. In both BCR-ABL+ p53mutant K562 and Meg-01 CML cells but not in BCR-ABL− HL60 cells, we found imatinib upregulated phosphorylation of p38 mitogen-activated protein kinase (MAPK), checkpoint kinase 2 (chk2) and transactivation-competent (TA) p73, formation of PML-nuclear body (NB), co-localization of TAp73/PML-NB, and expression of bax. Co-immpunoprecipitation of TAp73 and pml was also consistent with the TAp73/PML-NB co-localization. The induced co-localization also occurred to primary CML cells from three out of six patients, including the two with p53mutant. In K562 cells, both inhibiting p38MAPK with SB203580 and silencing pml or TAp73 with short interfering RNAs (siRNAs) abolished the imatinib-induced apoptosis whereas interferon alpha-2a induced additively with imatinib the apoptosis. Notably, interferon alpha-2a increased additively while SB203580 hindered the imatinib-induced phosphorylation of TAp73 and the TAp73/PML-NB co-localization. Together, imatinib induced in some CML cells, especially with mutant p53, a p53-independent proapoptotic mechanism via a p38MAPK/chk2-pml/TAp73 signaling.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2578-2578
Author(s):  
Zhanxiang Wang ◽  
Seiji Fukuda ◽  
Louis M. Pelus

Abstract The Bcr-abl oncogene is found in cells of >95% of patients with CML and encodes a cytoplasmic protein with constitutive tyrosine kinase activity. Bcr-abl induces hematopoietic cell transformation and protects cells from apoptosis induced by numerous stimuli. Bcr-abl activates Myc, Ras, raf, Pl3K, and c-jun kinases that are critical for transforming activity, however, the signaling pathways between Bcr-abl and the apoptosis machinery are just beginning to be determined. It is known that Bcr-abl can exert an antiapoptotic effect by blocking mitochondrial release of cytochrome C. Although increased knowledge of Bcr-abl pathways resulted in the design of selective tyrosine kinase inhibitors such as STI571 (Gleevec), the development of drug resistance limits efficacy. Survivin is a member of the highly conserved inhibitor-of-apoptosis (IAP) family of endogenous caspase inhibitors. Similar to other IAPs, Survivin blocks apoptosis by inhibiting caspases 3, 7 and 9. However in contrast to other IAPs, Survivin is not expressed in most adult tissues but aberrantly overexpressed in all cancers and hematopoietic malignancies. Targeting of Survivin by antisense (AS) or dominant-negative (DN) strategies in transformed cell models induces apoptosis. Survivin is expressed in Bcr-abl+ CML cells in blast crisis, but not in cells from patients with Bcr-ablneg CML. Furthermore, high expression of Survivin is found in Adriamycin resistant Bcr-abl+ K562 cells. These findings led us to investigate whether Survivin is involved in the antiapoptotic effects of Bcr-abl and if Survivin disruption can facilitate apoptosis in Bcr-abl+ cells. Transient transfection of human Mo7e and mouse BaF3 hematopoietic cells with the Bcr-abl oncongene results in significantly elevated expression of Survivin mRNA and protein. The mRNA of 2 other Survivin splice variants, Survivin-2B and Survivin-ΔEx3 were also upregulated. In transfected Mo7e cells, Survivin promoter activity was upregulated 2–4 fold compared to parental Mo7e cells, determined using a luciferase reporter construct. In addition to Survivin, the IAP family member ILP was also upregulated by Bcr-abl. Disruption of Survivin expression/function in K562 cells that contain endogenous Bcr-abl and in Bcr-abl transfected Mo7e cells by ectopic expression of AS or DN T34A or C84A mutant Survivin constructs, significantly promoted apoptosis induced by the Bcr-abl tyrosine kinase inhibitor, Gleevec by ≥2-fold, in a time and dose dependent manner. Enhanced apoptosis induced by AS or DN Survivin was accompanied by caspase dependent cleavage of Bcr-abl oncoprotein (>50% decrease in Bcr-abl protein) that was blocked by the caspase inhibitor Z-VAD-fmk; disruption of mitochondria membrane potential (>35% increase in TMRMnegexpression); and enhanced cytochrome C release, quantified by westerns. Similarly, disruption of Survivin mRNA in K562 cells by a 20-mer antisense oligonucleotide resulted in >40% increase in mitochondrial disruption. In contrast, forced expression of wild-type Survivin in K562 cells protected cells from Gleevec-induced apopotosis. In summary, our results demonstrate that the Bcr-abl oncogene regulates Survivin transcription and production, which represents a new signaling pathway downstream of Bcr-abl that may be helpful in understanding the pathophysiology of CML. Targeted Survivin disruption may sensitize Bcr-abl+ CML cells to Gleevec-induced apoptosis and have therapeutic potential, particularly in the development of drug resistance.


2019 ◽  
Vol 20 (9) ◽  
pp. 2271 ◽  
Author(s):  
Shu-Huey Chen ◽  
Jyh-Ming Chow ◽  
Yao-Yu Hsieh ◽  
Chun-Yu Lin ◽  
Kai-Wen Hsu ◽  
...  

Since imatinib (Glivec or Gleevec) has been used to target the BCR-ABL fusion protein, chronic myeloid leukemia (CML) has become a manageable chronic disease with long-term survival. However, 15%–20% of CML patients ultimately develop resistance to imatinib and then progress to an accelerated phase and eventually to a blast crisis, limiting treatment options and resulting in a poor survival rate. Thus, we investigated whether histone deacetylase inhibitors (HDACis) could be used as a potential anticancer therapy for imatinib-resistant CML (IR-CML) patients. By applying a noninvasive apoptosis detection sensor (NIADS), we found that panobinostat significantly enhanced cell apoptosis in K562 cells. A further investigation showed that panobinostat induced apoptosis in both K562 and imatinib-resistant K562 (IR-K562) cells mainly via H3 and H4 histone acetylation, whereas panobinostat targeted cancer stem cells (CSCs) in IR-K562 cells. Using CRISPR/Cas9 genomic editing, we found that HDAC1 and HDAC2 knockout cells significantly induced cell apoptosis, indicating that the regulation of HDAC1 and HDAC2 is extremely important in maintaining K562 cell survival. All information in this study indicates that regulating HDAC activity provides therapeutic benefits against CML and IR-CML in the clinic.


Author(s):  
Zhiming Wang ◽  
Jin He ◽  
Duc-hiep Bach ◽  
Yung-hsing Huang ◽  
Zongwei Li ◽  
...  

Abstract Background Therapeutic resistance occurs in most patients with multiple myeloma (MM). One of the key mechanisms for MM drug resistance comes from the interaction between MM cells and adipocytes that inhibits drug-induced apoptosis in MM cells; MM cells reprogram adipocytes to morph into different characterizations, including exosomes, which are important for tumor-stroma cellular communication. However, the mechanism by which exosomes mediate the cellular machinery of the vicious cycle between MM cells and adipocytes remains unclear. Methods Adipocytes were either isolated from bone marrow aspirates of healthy donors or MM patients or derived from mesenchymal stem cells. Co-culturing normal adipocytes with MM cells was used to generate MM-associated adipocytes. Exosomes were collected from the culture medium of adipocytes. Annexin V-binding and TUNEL assays were performed to assess MM cell apoptosis. Methyltransferase activity assay and dot blotting were used to access the m6A methylation activity of methyltransferase like 7A (METTL7A). RIP, MeRIP-seq, and RNA–protein pull down for assessing the interaction between long non-cording RNAs (LncRNAs) and RNA binding proteins were performed. Adipocyte-specific enhancer of zeste homolog 2 (EZH2) knockout mice and MM-xenografted mice were used for evaluating MM therapeutic response in vivo. Results Exosomes collected from MM patient adipocytes protect MM cells from chemotherapy-induced apoptosis. Two LncRNAs in particular, LOC606724 and SNHG1, are significantly upregulated in MM cells after exposure to adipocyte exosomes. The raised LncRNA levels in MM cells are positively correlated to worse outcomes in patients, indicating their clinical relevancy in MM. The functional roles of adipocyte exosomal LOC606724 or SNHG1 in inhibition of MM cell apoptosis are determined by knockdown in adipocytes or overexpression in MM cells. We discovered the interactions between LncRNAs and RNA binding proteins and identified methyltransferase like 7A (METTL7A) as an RNA methyltransferase. MM cells promote LncRNA package into adipocyte exosomes through METTL7A-mediated LncRNA m6A methylation. Exposure of adipocytes to MM cells enhances METTL7A activity in m6A methylation through EZH2-mediated protein methylation. Conclusion This study elucidates an unexplored mechanism of how adipocyte-rich microenvironment exacerbates MM therapeutic resistance and indicates a potential strategy to improve therapeutic efficacy by blocking this vicious exosome-mediated cycle.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 5053-5053
Author(s):  
Shuangfeng Xie ◽  
Songmei Yin ◽  
Dan Li ◽  
Danian Nie ◽  
Yiqing Li ◽  
...  

Abstract Abstract 5053 Introduction Failure for chemotherapy in leukemia patients is often due to innate or acquired multi-drug resistance of leukemia cells. The X-linked inhibitor of apoptosis protein (XIAP) is a potent inhibitor of apoptosis and is also involved in drug resistance. The relationship between XIAP and drug resistance is not fully clarified. Here, we manipulated the expression of XIAP in K562 human chronic myelogenous leukemia cells and explore the changes in cell apoptosis and sensitivity to cytarabin. Methods Small interfering RNA (siRNA) targeting XIAP was created, K562 cells were either transfected with XIAP siRNA or control siRNA by Nucleofector transfection. The siRNAs were labelled with Cy3. Real-time RT-PCR and Western blot were used to determine the expression of XIAP in K562 cells. Flow cytometry was used to detect apoptotic cells and 50% inhibiting concentration (IC50) of cytarabin was also determined. Results The transfection rate was 87% in general. The transfection was also confirmed by fluorescence microscopy. After 48h, the expression levels of XIAP mRNA and protein were much lower in XIAP siRNA-transfected cells than in control siRNA-transfected cells (mRNA, 0.37±0.10 vs. 1.41±0.13, P<0.05 ; protein, 0.39±0.03 vs. 0.99±0.08, P<0.05). There was no significant difference in the fractions of apoptotic cells between these two groups at 48h after transfection (37.04%±1.77% vs. 47.43%±1.13%, P>0.05). Cytarabin was added in different concentrations right after Nucleotransfection. After 48h, the IC50 of Cytarabin in the XIAP siRNA-transfected cells was 2.28-fold lower than that in the control siRNA-transfected cells (82.60±21.56μg/ml vs. 188.67±44.48μg/ml, P<0.05). The percentage of apoptotic cells was much higher in the XIAP knockdown cells than in the control cells (47.43%±1.13% vs. 37.04%±1.77% P<0.05). Conclusions XIAP siRNA can specifically down-regulate the expressions of XIAP mRNA and protein, promote the drug-sensitivity to cytarabin and increase the cell apoptosis in K562 cells. This might be a novel choice for leukemia treatment. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 27 (13) ◽  
pp. 2118-2132 ◽  
Author(s):  
Aysegul Hanikoglu ◽  
Hakan Ozben ◽  
Ferhat Hanikoglu ◽  
Tomris Ozben

: Elevated Reactive Oxygen Species (ROS) generated by the conventional cancer therapies and the endogenous production of ROS have been observed in various types of cancers. In contrast to the harmful effects of oxidative stress in different pathologies other than cancer, ROS can speed anti-tumorigenic signaling and cause apoptosis of tumor cells via oxidative stress as demonstrated in several studies. The primary actions of antioxidants in cells are to provide a redox balance between reduction-oxidation reactions. Antioxidants in tumor cells can scavenge excess ROS, causing resistance to ROS induced apoptosis. Various chemotherapeutic drugs, in their clinical use, have evoked drug resistance and serious side effects. Consequently, drugs having single-targets are not able to provide an effective cancer therapy. Recently, developed hybrid anticancer drugs promise great therapeutic advantages due to their capacity to overcome the limitations encountered with conventional chemotherapeutic agents. Hybrid compounds have advantages in comparison to the single cancer drugs which have usually low solubility, adverse side effects, and drug resistance. This review addresses two important treatments strategies in cancer therapy: oxidative stress induced apoptosis and hybrid anticancer drugs.


Sign in / Sign up

Export Citation Format

Share Document