Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Comparative Study
. 2012 May 31;31(22):2794-804.
doi: 10.1038/onc.2011.449. Epub 2011 Oct 10.

Small nucleolar RNA 42 acts as an oncogene in lung tumorigenesis

Affiliations
Comparative Study

Small nucleolar RNA 42 acts as an oncogene in lung tumorigenesis

Y-P Mei et al. Oncogene. .

Abstract

Non-small cell lung cancer (NSCLC) is the leading cause of cancer death, reflecting the need for better understanding the oncogenesis, and developing new diagnostic and therapeutic targets for the malignancy. Emerging evidence suggests that small nucleolar RNAs (snoRNAs) have malfunctioning roles in tumorigenesis. Our recent study demonstrated that small nucleolar RNA 42 (SNORA42) was overexpressed in lung tumors. Here, we investigate the role of SNORA42 in tumorigenesis of NSCLC. We simultaneously assess genomic dosages and expression levels of SNORA42 and its host gene, KIAA0907, in 10 NSCLC cell lines and a human bronchial epithelial cell line. We then determine in vitro functional significance of SNORA42 in lung cancer cell lines through gain- and loss-of-function analyses. We also inoculate cancer cells with SNORA42-siRNA into mice through either tail vein or subcutaneous injection. We finally evaluate expression level of SNORA42 on frozen surgically resected lung tumor tissues of 64 patients with stage I NSCLC by using quantitative reverse transcriptase PCR assay. Genomic amplification and associated high expression of SNORA42 rather than KIAA0907 are frequently observed in lung cancer cells, suggesting that SNORA42 overexpression is activated by its genomic amplification. SNORA42 knockdown in NSCLC cells inhibits in vitro and in vivo tumorigenicity, whereas enforced SNORA42 expression in bronchial epitheliums increases cell growth and colony formation. Such pleiotropy of SNORA42 suppression could be achieved at least partially through increased apoptosis of NSCLC cells in a p53-dependent manner. SNORA42 expression in lung tumor tissue specimens is inversely correlated with survival of NSCLC patients. Therefore, SNORA42 activation could have an oncogenic role in lung tumorigenesis and provide potential diagnostic and therapeutic targets for the malignancy.

PubMed Disclaimer

Conflict of interest statement

Conflict of interest

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
SNORA42 rather than its host gene (KIAA0907) is frequently amplified and overexpressed in lung cancer cells. (a) Schematic diagram of SNORA42 and its location in KIAA0907. (b) Genomic dosage (open bar) and RNA expression level (solid bar) of SNORA42 in 10 NSCLC cell lines and a control cell line, BEAS-2B. The open bars in (b) and (c) represent genomic dosages and the solid ones represent RNA levels. (c) Genomic dosage (open bar) and RNA expression level (solid bar) of KIAA0907 in all cell lines. (d) Southern blot analyses of SNORA42 and KIAA0907 in the cell lines. (e) N blot analyses of SNORA42 and KIAA0907 in the cell lines. (f) KIAA0907 protein expression was tested by western blotting in the cell lines. Each data point represented mean value from all the three independent experiments.
Figure 2
Figure 2
SNORA42 knockdown inhibits cell growth and proliferation of NSCLC cells. (a) SNORA42 was efficiently and specifically reduced by SNORA-siRNA in cancer cells. The figure only shows expression levels of SNORA42 in H460 and H1944 cancer cells 48h after treatment (*P < 0.001). (b) Suppression of cell proliferation by SNORA42 knockdown. (c) Effect of SNORA42 knockdown on cell viability 72h after different treatments in cancer cells. The figure only shows expression levels of SNORA42 in H460 and H1944 cancer cells after treatment (*P < 0.001). (d) KIAA0907 expression was efficiently and specifically knocked down by KIAA0907-siRNA in H1944. (e) KIAA0907 knockdown had no effect on cell viability in H1944. The experiments were repeated independently for three times.
Figure 3
Figure 3
SNORA42 knockdown inhibits in vitro tumorigenicity. (a) Soft-agar colony formation for H460 and H1944 cells transfected with mock control, scrambled siRNA and SNORA42-siRNA, respectively. (b) Colonies were counted after a 3-week culture period. Each data point represented mean value from all three independent experiments (*P < 0.001).
Figure 4
Figure 4
Ectopic expression of SNORA42 enhances cell proliferation and growth ability of BEAS-2B, which was used as a control cell line. (a) SNORA42 expression level in BEAS-2B cells, which was transfected with pCMV control or pCMV SNORA42 (*P < 0.001). (b) Effect of SNORA42 overexpression on cell proliferation of BEAS-2B cells 48 h after treatments (*P < 0.001). (c) SNORA42 overexpression enhances cell proliferation. (d) Soft-agar colony formation for BEAS-2B cells treated with either pCMV SNORA42 or pCMV control, respectively, after a 4 week culture (*P < 0.01). Each experiment was performed independently for at least three times.
Figure 5
Figure 5
SNORA42 knockdown inhibits in vivo tumorigenicity of cancer cells. (a) Effect of SNORA42 suppression on ectopic tumor formation of H1944 cells. After 4 weeks, H1944 cells with scrambled siRNA yielded tumors (red arrow) in 5 of 10 mice. (b) Effect of SNORA42 knockdown on orthotopic tumor formation of H460-luc2 cells. An orthotopic lung tumor in the mouse (left) injected with H460-luc2 cells transfected with scrambled siRNA at week 3. (c) Cross-section of the lungs of orthotopic mice model at week 7 after injection with cancer cells with scrambled siRNA displaying multiple malignant nodules (red arrows in left), whereas there is no nodule on the lungs from mouse injected with cancer cells transfected with SNORA42-siRNA (right).
Figure 6
Figure 6
Suppression of SNORA42 induces apoptosis in H460, H1944 and H292 cancer cells. (a) Annexin V/PI (propidium iodide) staining was performed on H460 cells with different treatments and analyzed by flow cytometry. Percentages of intact cells (Annexin VPI), early apoptotic cells (Annexin V+ PI) and late apoptotic or necrotic cells (Annexin V+ PI+) are shown in the lower panel. The Annexin V/PI staining assay was undertaken on all cell lines, the figure only shows the results from H460 cell line. (b) The sub-G1 peak of cell cycle was induced by SNORA42 knockdown. (c) Cleavages of caspase-3 and poly ADP-ribose polymerase were induced by SNORA42 knockdown. The figure only shows the results from H460 or H1944 cells. (d) The cell lines (H226, A549, H1299, H358, H1792, SK-MES-1 and H522) that are either p53-null cells or cells with mutated p53 do not exhibit apoptosis after SNORA42 knockdown. The figure only shows the results from H1299 or H1792 cells.
Figure 7
Figure 7
SNORA42 expression level inversely correlates with that of p53. (a) p53 protein was downregulated in H1944 cells with enforced SNORA42 expression. (b) p53 protein was upregulated in H1944 cancer cells with SNORA42 knockdown. (c) A tumor developed from H460 cancer cells with SNORA42-siRNA displays low SNORA42 expression by in situ hybridization and positive p53 expression by immunohistochemistry. SNORA42 probe produces one to two staining spots in most of the nucleus (green arrows). Antibody to p53 protein shows presence of staining in nucleus. (d) A xenograft tumor produced from H460 cancer cells with scrambled siRNA shows high SNORA42 expression and negative p53 expression. SNORA42 probe produces more than two staining spots in most of the nucleus (red arrows). The antibody shows complete absence of p53 staining.
Figure 8
Figure 8
SNORA42 expression in human lung tissues and its clinical significance. (a) Higher expression level of SNORA42 in NSCLC specimens compared with noncancerous lung tissues (*P < 0.001). (b) Probability of cancer-specific survival by levels of SNORA42 expression in stage I NSCLC.

Similar articles

Cited by

References

    1. Albertson DG, Ylstra B, Segraves R, Collins C, Dairkee SH, Kowbel D, et al. Quantitative mapping of amplicon structure by array CGH identifies CYP24 as a candidate oncogene. Nat Genet. 2000;25:144–146. - PubMed
    1. Baek SJ, Kim KS, Nixon JB, Wilson LC, Eling TE. Cyclooxygenase inhibitors regulate the expression of a TGF-â superfamily member that has proapoptotic and antitumorigenic activities. Mol Pharmacol. 2001;59:901–908. - PubMed
    1. Chang LS, Lin SY, Lieu AS, Wu TL. Differential expression of human 5S snoRNA genes. Biochem Biophys Res Commun. 2002;299:196–200. - PubMed
    1. Dong XY, Guo P, Boyd J, Sun X, Li Q, Zhou W, et al. Implication of snoRNA U50 in human breast cancer. J Genet Genomics. 2009;8:447–454. - PMC - PubMed
    1. Dong XY, Rodriguez C, Guo P, Sun X, Talbot JT, Zhou W, et al. SnoRNA U50 is a candidate tumor-suppressor gene at 6q14.3 with a mutation associated with clinically significant prostate cancer. Hum Mol Genet. 2008;17:1031–1042. - PMC - PubMed

Publication types

MeSH terms