Abstract

BackgroundAcquired resistance towards apoptosis is a hallmark of cancer. Elimination of cells bearing activated oncogenes or stimulation of tumor suppressor mediators may provide a selection pressure to overcome resistance. KC-53 is a novel biyouyanagin analogue known to elicit strong anti-inflammatory and anti-viral activity. The current study was designed to evaluate the anticancer efficacy and molecular mechanisms of KC-53 against human cancer cells.MethodsUsing the MTT assay we examined initially how KC-53 affects the proliferation rates of thirteen representative human cancer cell lines in comparison to normal peripheral blood mononuclear cells (PBMCs) and immortalized cell lines. To decipher the key molecular events underlying its mode of action we selected the human promyelocytic leukemia HL-60 and the acute lymphocytic leukemia CCRF/CEM cell lines that were found to be the most sensitive to the antiproliferative effects of KC-53.ResultsKC-53 promoted rapidly and irreversibly apoptosis in both leukemia cell lines at relatively low concentrations. Apoptosis was characterized by an increase in membrane-associated TNFR1, activation of Caspase-8 and proteolytic inactivation of the death domain kinase RIP1 indicating that KC-53 induced mainly the extrinsic/death receptor apoptotic pathway. Regardless, induction of the intrinsic/mitochondrial pathway was also achieved by Caspase-8 processing of Bid, activation of Caspase-9 and increased translocation of AIF to the nucleus. FADD protein knockdown restored HL-60 and CCRF/CEM cell viability and completely blocked KC-53-induced apoptosis. Furthermore, KC-53 administration dramatically inhibited TNFα-induced serine phosphorylation on TRAF2 and on IκBα hindering therefore p65/NF-κΒ translocation to nucleus. Reduced transcriptional expression of pro-inflammatory and pro-survival p65 target genes, confirmed that the agent functionally inhibited the transcriptional activity of p65.ConclusionsOur findings demonstrate, for the first time, the selective anticancer properties of KC-53 towards leukemic cell lines and provide a detailed understanding of the molecular events underlying its dual anti-proliferative and pro-apoptotic properties. These results provide new insights into the development of innovative and targeted therapies for the treatment of some forms of leukemia.Electronic supplementary materialThe online version of this article (doi:10.1186/s12885-016-2310-5) contains supplementary material, which is available to authorized users.

Highlights

  • Acquired resistance towards apoptosis is a hallmark of cancer

  • We have determined the effects of KC-53 on the viability of human breast (MCF-7, MDA-MB-231TXSA), lung (A-549), prostate (PC-3), colon (LoVo), endometrial (Ishikawa), osteosarcoma (KHOS), gastric (STSA), leukemia (Jurkat, HL-60, CRF/CEM) and lymphoma (Raji, Daudi) tumorigenic cells

  • HL-60 viability increased from 43 to 60 % in the presence of z.vad.fmk while no alterations were observed in the viability of CCRF/CEM. These findings indicate that inhibition of cell proliferation by KC-53 might be mediated by both caspase -dependent (CD) and -independent (CID) programmed cell death in a cell-context-specific manner

Read more

Summary

Introduction

Acquired resistance towards apoptosis is a hallmark of cancer. Elimination of cells bearing activated oncogenes or stimulation of tumor suppressor mediators may provide a selection pressure to overcome resistance. Alterations affecting key molecules of these pathways such as Bcl-2, p53 and the nuclear factor κΒ (NF-κB) lead to accumulation of malignant cells Among the latter, NF-κB promotes the transcription of genes encoding proteins involved in the suppression of cell death by both the intrinsic and the extrinsic pathway [3, 4]. TRAF2 phosphorylation, occurring on Ser 11, promotes receptorinteracting protein 1 (RIP1) ubiquitination, facilitating the recruitment and activation of the downstream IκB kinase complex (IKK). This leads to NF-κΒ activation [5, 6] and concurrently preventing RIP1 from interacting with Fas-associated death domain (FADD) protein and procaspase-8 [7, 8]. Active Caspase-8 cleaves and inactivates RIP1 initiating the extrinsic pathway of apoptosis [9, 10]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call