Open Access

Effect of membrane‑bound complement regulatory proteins on tumor cell sensitivity to complement‑dependent cytolysis triggered by heterologous expression of the α‑gal xenoantigen

  • Authors:
    • Yu Wang
    • Juan Liao
    • Ya‑Jun Yang
    • Zhu Wang
    • Feng Qin
    • Sheng‑Ming Zhu
    • Hong Zheng
    • Yan‑Ping Wang
  • View Affiliations

  • Published online on: April 12, 2018     https://doi.org/10.3892/ol.2018.8478
  • Pages: 9061-9068
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Engineering malignant cells to express a heterologous α‑gal antigen can induce heterograft hyperacute rejection, resulting in complement‑dependent cytolysis (CDC) of tumor cells, which has been considered as a novel strategy for antitumor therapy. A549 cells engineered to express Galα1‑3Galβ1‑4GlcNAc‑R (α‑gal) epitope exhibited strong resistance to CDC treated by normal human serum (NHS) in a previous study. We hypothesized that the expression of membrane‑bound complement regulatory proteins (mCRPs) decay accelerating factor (CD55) and protectin (CD59) influenced the efficacy of the α‑gal/NHS‑mediated antitumor effect to tumor cells in vitro. The present study confirmed that A549 cells expressed high levels of CD55 and CD59, whereas Lovo cells expressed relatively low levels of these proteins. A549 and Lovo cells transfected with plasmids containing or lacking the α‑gal epitope were evaluated for their susceptibility to CDC by NHS and detected using a trypan blue exclusion assay. α‑gal‑expressing Lovo (Lovo‑GT) cells were almost completely killed by α‑gal‑mediated CDC following incubation with 50% NHS, whereas no cytolysis was observed in α‑gal expressing A549 (A549‑GT) cells. Abrogating CD55 and CD59 function from A549‑GT cells by various concentrations of phosphatidylinositol‑specific phospholipase C (PI‑PLC) or blocking antibodies increased the susceptibility of cells to CDC, and the survival rate decreased significantly comparing to the controls (P<0.05). The findings of the present study indicated that using the α‑gal/NHS system to eliminate tumor cells via inducing the complement cascade reaction might represent a feasible approach for the treatment of cancer. However, high levels of mCRP expression may limit the efficacy of this approach. Therefore, an improved efficacy of cancer cell killing may be achieved by combining strategies of heterologous α‑gal expression and mCRP downregulation.
View Figures
View References

Related Articles

Journal Cover

June-2018
Volume 15 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Y, Liao J, Yang YJ, Wang Z, Qin F, Zhu SM, Zheng H and Wang YP: Effect of membrane‑bound complement regulatory proteins on tumor cell sensitivity to complement‑dependent cytolysis triggered by heterologous expression of the α‑gal xenoantigen. Oncol Lett 15: 9061-9068, 2018
APA
Wang, Y., Liao, J., Yang, Y., Wang, Z., Qin, F., Zhu, S. ... Wang, Y. (2018). Effect of membrane‑bound complement regulatory proteins on tumor cell sensitivity to complement‑dependent cytolysis triggered by heterologous expression of the α‑gal xenoantigen. Oncology Letters, 15, 9061-9068. https://doi.org/10.3892/ol.2018.8478
MLA
Wang, Y., Liao, J., Yang, Y., Wang, Z., Qin, F., Zhu, S., Zheng, H., Wang, Y."Effect of membrane‑bound complement regulatory proteins on tumor cell sensitivity to complement‑dependent cytolysis triggered by heterologous expression of the α‑gal xenoantigen". Oncology Letters 15.6 (2018): 9061-9068.
Chicago
Wang, Y., Liao, J., Yang, Y., Wang, Z., Qin, F., Zhu, S., Zheng, H., Wang, Y."Effect of membrane‑bound complement regulatory proteins on tumor cell sensitivity to complement‑dependent cytolysis triggered by heterologous expression of the α‑gal xenoantigen". Oncology Letters 15, no. 6 (2018): 9061-9068. https://doi.org/10.3892/ol.2018.8478