Abstract

Complement protein C1q is the first recognition subcomponent of the complement classical pathway that plays a vital role in the clearance of immune complexes, pathogens, and apoptotic cells. C1q also has a homeostatic role involving immune and non-immune cells; these functions not necessarily involve complement activation. Recently, C1q has been shown to be expressed locally in the microenvironment of a range of human malignant tumors, where it can promote cancer cell adhesion, migration, and proliferation, without involving complement activation. C1q has been shown to be present in the ascitic fluid formed during ovarian cancers. In this study, we have examined the effects of human C1q and its globular domain on an ovarian cancer cell line, SKOV3. We show that C1q and the recombinant globular head modules induce apoptosis in SKOV3 cells in a time-dependent manner. C1q expression was not detectable in the SKOV3 cells. Exogenous treatment with C1q and globular head modules at the concentration of 10 µg/ml induced apoptosis in approximately 55% cells, as revealed by immunofluorescence microscopy and FACS. The qPCR and caspase analysis suggested that C1q and globular head modules activated tumor necrosis factor (TNF)-α and upregulated Fas. The genes of mammalian target of rapamycin (mTOR), RICTOR, and RAPTOR survival pathways, which are often overexpressed in majority of the cancers, were significantly downregulated within few hours of the treatment of SKOV3 cells with C1q and globular head modules. In conclusion, C1q, via its globular domain, induced apoptosis in an ovarian cancer cell line SKOV3 via TNF-α induced apoptosis pathway involving upregulation of Bax and Fas. This study highlights a potentially protective role of C1q in certain cancers.

Highlights

  • C1q is the first subcomponent of the C1 complex that recognizes the IgG- or IgM-containing immune complexes and initiates the complement classical pathway

  • The malignant ovarian tumor cells isolated from ascitic fluid (AF) have been shown to have C1q and C2 deposited on the surface, which were rendered susceptible to complement-mediated killing by AF in the presence of anti-CD59-neutralizing antibody [9]

  • The presence of mammalian target of rapamycin (mTOR) was detected abundantly in the cytoplasm of the untreated cells, compared to SKOV3 cells treated with C1q and globular heads at 15 h time point via immunofluorescence microscopy (Figure 8B)

Read more

Summary

Introduction

C1q is the first subcomponent of the C1 complex that recognizes the IgG- or IgM-containing immune complexes and initiates the complement classical pathway. Complement activation appears to be dampened due to the expression of membrane regulators such as CD46, CD55, and CD59 on the ovarian cancer cells, rendering complement system as an inefficient immune surveillance mechanism. The presence of C1q has been shown in the stroma and vascular endothelium of a number of human malignant tumors, including lung adenocarcinoma, melanoma, colon adenocarcinoma, breast adenocarcinoma, and pancreatic carcinoma [6]. This has raised a tumor growth-fostering role for locally synthesized C1q via promotion of adhesion, migration, and proliferation

Methods
Results
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