Abstract 1001: Expression of cytochrome C oxidase 4 (COX4) in thyroid cancer cells

2016 
Background: Targeting cell metabolism has emerged as a therapeutic strategy for the treatment of cancer. Metabolically active thyroid cancers are resistant to treatment with radioactive iodine. Aberrant expression of genes controlling glycolysis was demonstrated in thyroid cancer cells, but little is known regarding the role of mitochondrial proteins in thyroid carcinogenesis. Cytochrome c oxidase 4 (COX4) plays pivotal roles in oxidative phosphorylation and the cellular response to oxidative stress. COX4 may thus represent a promising therapeutic target. Objectives: We examined expression of COX4 in human thyroid tumors and performed functional studies using thyroid cancer cell lines. Material and Methods Expression of COX4 was examined by immunostaining in 25 follicular adenomas (FAs), 22 follicular cancers (FTCs), 90 papillary cancers (PTCs) and in 48 samples from normal thyroid tissue. FTC-derived (FTC133) and PTC-derived (BCPAP) cell lines were used to create COX4-deficient cells by lentiviral transfection. The efficiency of COX4 inhibition was examined. Mitochondrial membrane potential was examined by JC-1 staining. DNA-damage signaling was examined after cell exposure to γ radiation (6-18 Gy). We also determined the effects of 2-deoxyglucose (2DG) on viability of thyroid cancer cells with compromised COX4. Caspase-3 and PARP cleavage assays were performed to measure apoptosis. Results Positive immunostaining with anti-COX4 was detected in 6/48 (12.5%) normal tissue, 5/20 (25%) FAs, 7/22 (31%) FTCs, and 51/90 (56%) PTCs. The intensity of COX4 immunostaining was significantly higher in thyroid cancers than in either normal thyroid (p = 0.0001) or benign FAs (0.001). COX4 expression was more frequently detected in PTCs than in FTCs (p = 0.03). The mRNA level of COX4 was higher in BCPAP and FTC133 cells compared to normal thyroid. In both cell lines, silencing of COX4 altered intra-cellular distribution of JC-1 staining. In control cells, JC-1 staining was perinuclear, but in COX4-deficient cells it became diffusely cytoplasmic. COX4 silencing affected cell growth and response to γradiation in a cell type specific manner. In BCPAP cells, downregulation of COX4 was associated with inhibition of cell growth, block in G1 phase and inhibition of Cyclin D1. In BCPAP cells, COX4 silencing activated DNA-damage signaling and increased sensitivity to γ radiation. Inhibitor of glycolysis (2DG) was more efficient against COX4-deficient than COX4-expressing BCPAP cells. In FTC133 cells, silencing of COX4 increased the rate of growth and induced expression of Cyclin D1. COX4 silencing did not increase FTC133 cell sensitivity to γradiation nor to treatment with 2DG. Conclusion COX4 is implicated in regulation of thyroid cancer cell growth and response to DNA damaging or metabolic treatments. These data suggest that evaluation of COX4 in thyroid cancer could serve as a biomarker of response to treatment with metabolic agents. Citation Format: Vasyl V. Vasko, Athanasios Bikas, Aneeta Patel, John Costello, Rok Tkavc, Kenneth D. Burman, Kirk Jensen. Expression of cytochrome C oxidase 4 (COX4) in thyroid cancer cells. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1001.
    • Correction
    • Source
    • Cite
    • Save
    • Machine Reading By IdeaReader
    0
    References
    0
    Citations
    NaN
    KQI
    []