scholarly journals Cannabidiol promotes apoptosis via regulation of XIAP/Smac in gastric cancer

2019 ◽  
Vol 10 (11) ◽  
Author(s):  
Soyeon Jeong ◽  
Min Jee Jo ◽  
Hye Kyeong Yun ◽  
Dae Yeong Kim ◽  
Bo Ram Kim ◽  
...  

Abstract According to recent studies, Cannabidiol (CBD), one of the main components of Cannabis sativa, has anticancer effects in several cancers. However, the exact mechanism of CBD action is not currently understood. Here, CBD promoted cell death in gastric cancer. We suggest that CBD induced apoptosis by suppressing X-linked inhibitor apoptosis (XIAP), a member of the IAP protein family. CBD reduced XIAP protein levels while increasing ubiquitination of XIAP. The expression of Smac, a known inhibitor of XIAP, was found to be elevated during CBD treatment. Moreover, CBD treatment increased the interaction between XIAP and Smac by increasing Smac release from mitochondria to the cytosol. CBD has also been shown to affect mitochondrial dysfunction. Taken together, these results suggest that CBD may have potential as a new therapeutic target in gastric cancer.

2019 ◽  
Vol 10 (12) ◽  
Author(s):  
Larissa Ewald ◽  
Jessica Dittmann ◽  
Meike Vogler ◽  
Simone Fulda

AbstractDespite advances in the treatment of acute myeloid leukemia (AML), prognosis of AML patients is still dismal and better treatment options are required. B-cell Lymphoma 2 (BCL-2) homology domain 3 (BH3)-mimetics are emerging as a novel class of apoptosis-inducing agents that are currently being tested for the treatment of different hematological malignancies including AML. Particularly, the selective BCL-2 inhibitor ABT-199/Venetoclax is demonstrating clinical responses and has recently been approved in combination for the treatment of AML. Compounds targeting the related protein MCL-1 have recently entered clinical trials, highlighting the urgency to compare the different BH3-mimetics and identify the most promising antiapoptotic target in AML. We performed a side-by-side comparison of different highly selective and potent BH3-mimetics targeting BCL-2 (ABT-199), MCL-1 (S63845) or BCL-xL (A1331852) in a panel of AML cell lines and primary patient cells. Gene knockdown using siRNAs was utilized to investigate the functional relevance of BCL-2 proteins. Western blotting and immunoprecipitations were used to explore the influence of BH3-mimetics on interactions between pro- and antiapoptotic BCL-2 proteins. A1331852 induced apoptosis only in selected cases, indicating that BCL-xL is not a very promising therapeutic target in AML. However, S63845 displayed higher potency than ABT-199, with more cell lines and primary cells responding to S63845 than to ABT-199. MCL-1 dependency in AML cells was confirmed by siRNA-mediated knockdown of MCL-1, which was sufficient to induce apoptosis. S63845-induced cell death was accompanied by a displacement of the BH3-only protein BIM as well as BAK, resulting in BAK-dependent apoptosis. In contrast, ABT-199-induced cell death was mediated by BAX rather than BAK, indicating distinct non-redundant molecular functions of BCL-2 and MCL-1 in AML. Our study reveals that MCL-1 may be a more prevalent therapeutic target than BCL-2 in AML and identifies BIM and BAK as important mediators of S63845-induced apoptosis in AML.


2014 ◽  
Vol 2014 ◽  
pp. 1-12 ◽  
Author(s):  
Kuan-Yu Hung ◽  
Shin-Yun Liu ◽  
Te-Cheng Yang ◽  
Tien-Ling Liao ◽  
Shu-Huei Kao

Human peritoneal mesothelial cells (HPMCs) are a critical component of the peritoneal membrane and play a pivotal role in dialysis adequacy. Loss of HPMCs can contribute to complications in peritoneal dialysis. Compelling evidence has shown that high-dialysate glucose is a key factor causing functional changes and cell death in HPMCs. We investigated the mechanism of HPMC apoptosis induced by high-dialysate glucose, particularly the role of mitochondria in the maintenance of HPMCs. HPMCs were incubated at glucose concentrations of 5 mM, 84 mM, 138 mM, and 236 mM. Additionally, N-acetylcysteine (NAC) was used as an antioxidant to clarify the mechanism of high-dialysate-glucose-induced apoptosis. Exposing HPMCs to high-dialysate glucose resulted in substantial apoptosis with cytochrome c release, followed by caspase activation and poly(ADP-ribose) polymerase cleavage. High-dialysate glucose induced excessive reactive oxygen species production and lipid peroxidation as well as oxidative damage to DNA. Mitochondrial fragmentation, multiple mitochondrial DNA deletions, and dissipation of the mitochondrial membrane potential were also observed. The mitochondrial dysfunction and cell death were suppressed using NAC. These results indicated that mitochondrial dysfunction is one of the main causes of high-dialysate-glucose-induced HPMC apoptosis.


Cells ◽  
2020 ◽  
Vol 9 (2) ◽  
pp. 351 ◽  
Author(s):  
Maria Feoktistova ◽  
Roman Makarov ◽  
Sihem Brenji ◽  
Anne T. Schneider ◽  
Guido J. Hooiveld ◽  
...  

The ubiquitin-editing protein A20 (TNFAIP3) is a known key player in the regulation of immune responses in many organs. Genome-wide associated studies (GWASs) have linked A20 with a number of inflammatory and autoimmune disorders, including psoriasis. Here, we identified a previously unrecognized role of A20 as a pro-apoptotic factor in TNF-induced cell death in keratinocytes. This function of A20 is mediated via the NF-κB-dependent alteration of cIAP1/2 expression. The changes in cIAP1/2 protein levels promote NIK stabilization and subsequent activation of noncanonical NF-κB signaling. Upregulation of TRAF1 expression triggered by the noncanonical NF-κB signaling further enhances the NIK stabilization in an autocrine manner. Finally, stabilized NIK promotes the formation of the ripoptosome and the execution of cell death. Thus, our data demonstrate that A20 controls the execution of TNF-induced cell death on multiple levels in keratinocytes. This signaling mechanism might have important implications for the development of new therapeutic strategies for the treatment of A20-associated skin diseases.


2018 ◽  
Vol 115 (12) ◽  
pp. E2762-E2771 ◽  
Author(s):  
Yanfang Wu ◽  
Xia Li ◽  
Junying Jia ◽  
Yanpeng Zhang ◽  
Jing Li ◽  
...  

The accumulation of misfolded proteins in the endoplasmic reticulum (ER) causes ER stress and triggers the unfolded protein response (UPR). Failure to resolve ER stress leads to apoptotic cell death via a yet unclear mechanism. Here, we show that RNF183, a membrane-spanning RING finger protein, localizes to the ER and exhibits classic E3 ligase activities. Sustained ER stress induced by different treatments increases RNF183 protein levels posttranscriptionally in an IRE1α-dependent manner. Activated IRE1 reduces the level of miR-7, which increases the stability of RNF183 transcripts. In addition, overexpression of RNF183 leads to increased apoptosis and its depletion alleviates ER stress-induced apoptosis. Furthermore, RNF183 interacts with Bcl-xL, an antiapoptotic member of the Bcl-2 family, and polyubiquitinates Bcl-xL for degradation. Thus, RNF183 plays an important role in executing programmed cell death upon prolonged ER stress, likely by inducing apoptosis through Bcl-xL.


Molecules ◽  
2018 ◽  
Vol 23 (9) ◽  
pp. 2264 ◽  
Author(s):  
Jeong Yun ◽  
Seon Woo ◽  
Seung Seo ◽  
Kyoung-Jin Min ◽  
Dong Kim ◽  
...  

Niclosamide is used to treat intestinal parasite infections, as being an anthelmintic drug. Recently, several papers suggest the niclosamide inhibits multiple signaling pathways, which are highly activated and mutated in cancer. Here, niclosamide was evaluated for identifying strategies to overcome tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) resistance. Although niclosamide (100–200 nM) alone did not bring about cell death, combinations of niclosamide and TRAIL led to apoptotic cell death in carcinoma cells, but not in normal cells. Niclosamide markedly increased DR5 protein levels, including cell-surface DR5, and decreased c-FLIP protein levels. Down-regulation of DR5 by specific small interfering RNA (siRNA) and ectopic expression of c-FLIP markedly blocked niclosamide plus TRAIL-induced apoptosis. Our findings provide that niclosamide could overcome resistance to TRAIL through up-regulating DR5 on the cell surface and down-regulating c-FLIP in cancer cells. Taken together, niclosamide may be an attractive candidate to overcome TRAIL resistance.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 528-528
Author(s):  
Bing Z. Carter ◽  
Duncan H. Mak ◽  
Wendy D. Schober ◽  
Martin Dietrich ◽  
Clemencia Pinilla ◽  
...  

Abstract Triptolide, a recently identified anticancer agent from a Chinese herb, has been shown to synergistically enhance TRAIL-induced cell death in various solid tumor cell lines. We have found that triptolide potently induces apoptosis in leukemic cell lines and blasts from AML patients at least in part by decreasing XIAP levels. XIAP is known to be a resistance factor in TRAIL-induced cell death. XIAP is highly expressed in AML blasts and primary AML cells generally are insensitive to TRAIL. We therefore hypothesize that triptolide will sensitize AML cells to TRAIL-induced apoptosis. We treated OCI-AML3, U937, and Jurkat cells with sub-optimal concentrations of triptolide, TRAIL, and their combination. At concentrations showing no or minimal effects by each agent alone, their combinations significantly promoted cell death with combination indices (CI) <1.0 for all three leukemia cell lines. To ensure that XIAP contributes to TRAIL resistance, we treated U937 cells overexpressing XIAP (U937XIAP) and the control cells (U937neo) with TRAIL. U937XIAP cells were 6-fold more resistant to TRAIL (IC50=455.8 ng/ml) than U937neo cells (IC50=74.3 ng/ml). We then treated these cells with TRAIL and 1396–11, a small molecule XIAP antagonist that binds to BIR2 of XIAP and promotes caspase-dependent apoptosis. At 3μM, 1396–11 had no significant effect on survival of either U937neo or U939XIAP cells. When combined with TRAIL, 1396–11 augmented TRAIL-induced cell death of both U937neo (IC50=45.0 ng/ml with 1396-11 vs. IC50=74.3 ng/ml without) and U939XIAP cells (IC50=318.9 ng/ml with 1396–11 vs. IC50=455.8 ng/ml without). Furthermore, we observed that triptolide decreased MDM2 and increased p53 protein levels in p53 wild type OCI-AML3 but not in p53 null U937 cells. TRAIL receptor DR5 has been shown to be regulated by p53. To elucidate the role of p53 in TRAIL-induced cell death sensitized by triptolide, we treated OCI-AML3 and U937 cells with triptolide and examined DR5 expression. We found that triptolide induced DR5 protein levels in OCI-AML3, but much less so in U937 cells. Treatment of OCI-AML3 cells with Nutlin3a, a MDM2 inhibitor that binds to MDM2 and stabilizes p53, increased DR5 protein levels and sensitized to TRAIL-induced cell death. Knockdown of p53 with retrovirus expressing p53siRNA in OCI-AML3 cells abolished cellular responses to nutlin3a and significantly decreased the sensitization to TRAIL by nutlin3a. Finally, we treated OCI-AML3 cells with 1396–11, nutlin3a, and TRAIL and our results showed that the triple combination (CI=0.045 at 24 hours, average of ED50, ED75, and ED90) was more effective in inducing cell death than either 1396–11 and TRAIL (CI=0.066) or nutlin3a and TRAIL (CI=0.190) combinations supporting our notion that triptolide sensitizes to TRAIL-induced cell death by modulating both XIAP expression and p53 signaling. Collectively, our studies suggest that inhibition of XIAP and induction of DR5 mediated by p53 activation both independently sensitize leukemic cells to TRAIL-induced apoptosis. Triptolide not only inhibits XIAP, which is overexpressed in AML, but also activates p53 signaling, which is intact in the majority of AMLs. Thus, combinations of triptolide and TRAIL may provide a novel strategy for treating AML patients by overcoming critical mechanisms of apoptosis resistance.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2945-2945
Author(s):  
Yuji Shimura ◽  
Junya Kuroda ◽  
Hisao Nagoshi ◽  
Mio Yamamoto-Sugitani ◽  
Tsutomu Kobayashi ◽  
...  

Abstract Abstract 2945 Background: Despite recent advances of therapeutic strategies, multiple myeloma (MM) still remains mostly incurable due to its drug resistance by both various cell intrinsic molecular abnormalities and the supports of myeloma microenvironment factors. The search for new therapeutic target molecule upon which various MM-promoting signalings converge is essential for the development of more effective therapy for MM. RSK2 is a member of the 90 kDa ribosomal S6 kinases (RSKs) family of serine (Ser) /threonine (Thr) kinases and lies downstream of RAS/ERK-1/2 signaling cascade. In MM, it has been shown that RSK2 mediates signaling initiated by FGFR3 activation for cell survival, but the functional role and the value as the therapeutic target of RSK2 have not been clearly defined. In this study, we precisely studied the association with RSK2 activation and chromosomal status, especially focusing on RSK2Ser227, which is responsible for downstream signaling phosphorylation, and the value of RSK2 as the therapeutic target in MM. Methods: Twelve human myeloma-derived cell lines and primary MM cells were utilized in this study. The use of human samples was approved by the ethical review board of our institute. Cell killing assay was performed using flow cytometry. Growth inhibitory assay was performed with modified MTT assay. Protein levels were examined by Western blotting. BI-D1870, a RSK2 inhibitor, was purchased from Symansis Limited (Auckland, New Zealand). RNA interference for RSK2 was performed by transfection of small interfering RNA (siRNA) into NCI-H929 cells by means of CLB-Transfection Kit (Lonza, Swiss) with protocol 9. Drug combination assays were evaluated with CalcuSyn software (Biosoft, Cambridge, UK). Results: RSK2Ser227 was shown to be phosphorylated in all 12 MM cell lines and 6 of 9 primary MM cells. t(4;14) involving IgH/FGFR3 was detected only in 7 cell lines and high FGFR3 expression was identified only in 6 cell lines. RSK2Ser227 was phosphorylated even in the absence of ERK1/2 or RSK2Tyr529 phosphorylation in several MM cell lines, indicating that their activation was not mandatory to RSK2Ser227 phosphorylation. Our study also did not show any positive relationship between RSK2Ser227 phosphorylation and other so-called “high-risk” cytogenetic abnormalities, such as 13q-, t(11;14), t(8;14), t(14;16), 1q21+, or 17p-. Treatment with BI-D1870 (RSK2 inhibitor) resulted in complete dephosphorylation of RSK2Ser227, but not RSK2Tyr529. The phosphorylation status of ERK1/2, or related signaling kinases, such as AKT, p38 MAPK, or JNK was also not affected. BI-D1870 showed dose-dependent growth inhibition effect in 6 MM-derived cell lines by inducing apoptosis. At molecular level, Mcl-1, p21WAF1/CIP1, cyclin D2 and c-Myc was significantly down-regulated and Bim was up-regulated by BI-D1870 intervention in NCI-H929 cell. BI-D1870 didn't modulate expression level of IRF-4. These change were also observed in other MM cell lines (KMS-34, AMO-1). To exclude the possibility that above BI-D1870 induced changes in protein expression and cell death are due to its off-target effects, we performed gene knockdown experiments of RSK2. Transient gene knockdown of RSK2 by means of RNAi caused downregulation of cyclin D2, p21WAF1/CIP1, c-Myc, while it induced cell death in NCI-H929 cells. In contrast, Bim expression was not altered by RSK2 knockdown. Lenalidomide (LEN) is the most powerful currently available IMiD for MM treatment. While cell death induction by LEN (100μM) was partly prevented by IL-6 (50ng/ml) in NCI-H929 cells, co-administration of BI-D1870 (0.2 μM, at the degree of low concentration not to affect cell viability when used solely) overcame the resistance to LEN induced by IL-6. KMS-11/BTZ were highly resistant to bortezomib (BTZ)-induced cell death, by contrast, it showed the similar sensitivities to BI-D1870-induced cell death when compared with their parental cells. And more, BI-D1870 showed synergistic or additive effects with upcoming new anti-MM agents, such as RAD001, MS-275 and ABT-263 in MM cell lines. Conclusions: RSK2, especially RSK2Ser227, may be a universal therapeutic target for MMs with diverse molecular signatures. Disclosures: Iida: Janssen Pharmaceutical K.K.: Honoraria.


Sign in / Sign up

Export Citation Format

Share Document