RNA disruption is associated with response to multiple classes of chemotherapy drugs in tumor cell lines

Rashmi Narendrula, Kyle Mispel-Beyer, Baoqing Guo, Amadeo M Parissenti, Laura B Pritzker, Ken Pritzker, Twinkle Masilamani, Xiaohui Wang, Carita Lannér, Rashmi Narendrula, Kyle Mispel-Beyer, Baoqing Guo, Amadeo M Parissenti, Laura B Pritzker, Ken Pritzker, Twinkle Masilamani, Xiaohui Wang, Carita Lannér

Abstract

Background: Cellular stressors and apoptosis-inducing agents have been shown to induce ribosomal RNA (rRNA) degradation in eukaryotic cells. Recently, RNA degradation in vivo was observed in patients with locally advanced breast cancer, where mid-treatment tumor RNA degradation was associated with complete tumor destruction and enhanced patient survival. However, it is not clear how widespread chemotherapy induced "RNA disruption" is, the extent to which it is associated with drug response or what the underlying mechanisms are.

Methods: Ovarian (A2780, CaOV3) and breast (MDA-MB-231, MCF-7, BT474, SKBR3) cancer cell lines were treated with several cytotoxic chemotherapy drugs and total RNA was isolated. RNA was also prepared from docetaxel resistant A2780DXL and carboplatin resistant A2780CBN cells following drug exposure. Disruption of RNA was analyzed by capillary electrophoresis. Northern blotting was performed using probes complementary to the 28S and 18S rRNA to determine the origins of degradation bands. Apoptosis activation was assessed by flow cytometric monitoring of annexin-V and propidium iodide (PI) binding to cells and by measuring caspase-3 activation. The link between apoptosis and RNA degradation (disruption) was investigated using a caspase-3 inhibitor.

Results: All chemotherapy drugs tested were capable of inducing similar RNA disruption patterns. Docetaxel treatment of the resistant A2780DXL cells and carboplatin treatment of the A2780CBN cells did not result in RNA disruption. Northern blotting indicated that two RNA disruption bands were derived from the 3'-end of the 28S rRNA. Annexin-V and PI staining of docetaxel treated cells, along with assessment of caspase-3 activation, showed concurrent initiation of apoptosis and RNA disruption, while inhibition of caspase-3 activity significantly reduced RNA disruption.

Conclusions: Supporting the in vivo evidence, our results demonstrate that RNA disruption is induced by multiple chemotherapy agents in cell lines from different tissues and is associated with drug response. Although present, the link between apoptosis and RNA disruption is not completely understood. Evaluation of RNA disruption is thus proposed as a novel and effective biomarker to assess response to chemotherapy drugs in vitro and in vivo.

Figures

Fig. 1
Fig. 1
Dose and time-dependent ribosomal RNA disruption in response to taxanes. A2780 and CaOV3 cells were exposed to increasing concentrations of either docetaxel (DXL) or paclitaxel (TAX) for times ranging from 24 to 72 h. Total RNA was isolated from cells following drug exposure and RNA quality was analyzed by capillary electrophoresis. Electropherograms showing RNA mobility were converted to gel images using the Bioanalyzer software. a Gel image of RNA from A2780 cells treated with paclitaxel. b RDI analysis of RNA isolated from paclitaxel treated A2780 cells. c Gel image of RNA from A2780 cells treated with docetaxel. d RDI analysis of RNA isolated from docetaxel treated A2780 cells. e Gel image of RNA isolated from CaOV3 cells treated with docetaxel. f RDI analysis of RNA isolated from CaOV3 cells treated with docetaxel
Fig. 2
Fig. 2
Dose dependence of carboplatin-induced RNA disruption in A2780 and CaOV3 cells. A2780 and CaOV3 cells were treated with increasing concentrations of carboplatin (CBN) for 72 h, the length of time required to detect the RNA disruption response to carboplatin. Total RNA was isolated from cells following drug exposure and RNA quality was analyzed by capillary electrophoresis. a Gel image of RNA from A2780 cells treated with carboplatin. b RDI analysis of RNA isolated from carboplatin treated A2780 cells. c Gel image of RNA from CaOV3 cells treated with carboplatin. d RDI analysis of RNA isolated from carboplatin treated CaOV3 cells
Fig. 3
Fig. 3
Multiple chemotherapy agents induce RNA disruption in breast and ovarian cancer cell lines. Multiple breast and ovarian cancer cell lines were exposed to various chemotherapy agents and total RNA was isolated from the cells following drug exposure. a Gel image of total RNA isolated from A2780 cells treated with multiple chemotherapy agents. All treatments were for 72 h, except carboplatin, which was treated for 120 h. b Gel image of RNA isolated from MDA-MB-231, a breast cancer cell line, following treatment with multiple chemotherapy agents. All treatments were for 72 h, except for the control (0 μM) and cisplatin which were treated for 96 h. c Gel image of RNA isolated from various breast (MCF-7, MB231, SKBR3, BT474) and ovarian cancer (A2780, CaOV3) cells following 72 h docetaxel treatment, except for MDA-MB-231-0 μM, from which RNA was isolated after 96 h. Abbreviations: TAX-paclitaxel, DXL-docetaxel, CBN-carboplatin, CIS-cisplatin, ETOP-etoposide, VIN-vincristine, IRN-irinotecan, DOX-doxorubicin
Fig. 4
Fig. 4
Northern blot analysis of RNA isolated from A2780 cells treated with docetaxel. A2780 cells were treated with 0.2 μM docetaxel for 48 h and total RNA was isolated. RNA was resolved by agarose gel electrophoresis and transferred to PVDF membranes for hybridization with the 32P-end-labeled oligonucleotide probe, 28S-5. a The panel on the left shows the agarose gel and the panel on the right shows the Northern blot of the gel. RNA bands are indicated by arrows with the size of the band in nucleotides alongside. b A schematic diagram of the 28S rRNA sequence showing conserved and variable regions, based on the structure of the 28S rRNA as defined by Gorski et al. (1987) [53] and Wakeman et al. [28]. Location of the probes in the 28S rRNA sequence is shown above the diagram using arrows and the location of the cleavage sites and resulting bands are shown below the diagram
Fig. 5
Fig. 5
Lack of RNA disruption response in drug resistant cells. A2780 and A2780DXL (resistant to docetaxel) cells were treated with 0, 0.005 and 0.2 μM docetaxel (DXL) for 48 and 72 h. RNA isolated from the cells was analyzed by capillary gel electrophoresis. A2780 and A2780CBN (resistant to carboplatin) cells were treated with 0 and 10 μM carboplatin (CBN) for 72 h. To test for cross-resistance, A2780DXL cells were treated with 0 and 5 μM carboplatin while A2780CBN cells were treated with 0 and 0.2 μM docetaxel. RNA isolated from the cells was analyzed by capillary gel electrophoresis. a RDI analysis of RNA isolated from A2780 and A2780DXL cells treated with docetaxel. b RDI analysis of RNA isolated from A2780 and A2780CBN cells treated with carboplatin. c RDI analysis of A2780DXL cells treated with 0 and 5 μM carboplatin. d RDI analysis of RNA isolated from A2780CBN cells treated with 0 and 0.2 μM docetaxel
Fig. 6
Fig. 6
Early and late markers of apoptosis in A2780 cells treated with 0.2 μM docetaxel.A2780 cells were treated with 0.2 μM docetaxel (DXL) for 8, 24, 48 and 72 h. a Cells were stained with annexin V-FITC and propidium iodide and analyzed by flow cytometry. Scatter plots of stained cells at each time point are shown. b A2780 cells were stained with propidium iodide only and analyzed by flow cytometry. Histograms of stained cells are shown for each time point. c DNA laddering in A2780 cells treated with 0 and 0.2 μM docetaxel for times ranging from 2 to 72 h. Jurkat cells were treated with 5 and 30 μM etoposide as a positive control. DNA was isolated from cells at each time point and analyzed by agarose gel electrophoresis. A representative ethidium bromide stained gel is shown
Fig. 7
Fig. 7
Recovery and proliferation of A2780 cells following docetaxel treatment. In order to assess whether the A2780 cells treated with docetaxel (DXL) that show RNA degradation are able to recover and proliferate, cells were treated with 0, 0.005 or 0.2 μM docetaxel for 24, 48 and 72 h. Following the treatment end point, cells were collected and replated in fresh drug-free medium, and their proliferation was assessed following recovery for 24, 48, 72 and 96 h of replating. a Recovery of cells after 24 h treatment. b Recovery after 48 h drug treatment. c Recovery following 72 h drug treatment
Fig. 8
Fig. 8
Caspase-3 activation is associated with RNA disruption after docetaxel treatment in A2780 cells. A2780 cells were treated with 0.2 μM docetaxel (DXL) for 24, 48 or 72 h. a Caspase-3 activity, assayed using a DEVD substrate, in lysates from A2780 cells. b Effect of a caspase-3 inhibitor on caspase-3 activation in lysates from docetaxel treated A2780 cells. c Gel image of RNA disruption in A2780 cells treated with docetaxel, in the presence (+) or absence (−) of caspase-3 inhibitor (Q-DEVD-Oph). d RDI analysis of RNA from A2780 cells treated with docetaxel for 72 h, with and without the caspase-3 inhibitor

References

    1. Houseley J, Tollervey D. The many pathways of RNA degradation. Cell. 2009;136:763–76. doi: 10.1016/j.cell.2009.01.019.
    1. Lebreton A, Tomecki R, Dziembowski A, Seraphin B. Endonucleolytic RNA cleavage by a eukaryotic exosome. Nature. 2008;456:993–6. doi: 10.1038/nature07480.
    1. LaRiviere FJ, Cole SE, Ferullo DJ, Moore MJ. A late-acting quality control process for mature eukaryotic rRNAs. Mol Cell. 2006;24:619–26. doi: 10.1016/j.molcel.2006.10.008.
    1. Fujii K, Kitabatake M, Sakata T, Ohno M. 40S subunit dissociation and proteasome-dependent RNA degradation in nonfunctional 25S rRNA decay. EMBO J. 2012;31:2579–89. doi: 10.1038/emboj.2012.85.
    1. Vasudevan S, Steitz JA. AU-rich-element-mediated upregulation of translation by FXR1 and Argonaute 2. Cell. 2007;128:1105–18. doi: 10.1016/j.cell.2007.01.038.
    1. Franks TM, Lykke-Andersen J. The control of mRNA decapping and P-body formation. Mol Cell. 2008;32:605–15. doi: 10.1016/j.molcel.2008.11.001.
    1. Kadaba S, Wang X, Anderson JT. Nuclear RNA surveillance in Saccharomyces cerevisiae: Trf4p-dependent polyadenylation of nascent hypomethylated tRNA and an aberrant form of 5S rRNA. RNA. 2006;12:508–21. doi: 10.1261/rna.2305406.
    1. Cole SE, LaRiviere FJ, Merrikh CN, Moore MJ. A convergence of rRNA and mRNA quality control pathways revealed by mechanistic analysis of nonfunctional rRNA decay. Mol Cell. 2009;34:440–50. doi: 10.1016/j.molcel.2009.04.017.
    1. Kraft C, Deplazes A, Sohrmann M, Peter M. Mature ribosomes are selectively degraded upon starvation by an autophagy pathway requiring the Ubp3p/Bre5p ubiquitin protease. Nat Cell Biol. 2008;10:602–10. doi: 10.1038/ncb1723.
    1. Guchelaar HJ, Vermes A, Vermes I, Haanen C. Apoptosis: molecular mechanisms and implications for cancer chemotherapy. Pharm World Sci. 1997;19:119–25. doi: 10.1023/A:1008654316572.
    1. Lafarga M, Lerga A, Andres MA, Polanco JI, Calle E, Berciano MT. Apoptosis induced by methylazoxymethanol in developing rat cerebellum: organization of the cell nucleus and its relationship to DNA and rRNA degradation. Cell Tissue Res. 1997;289:25–38. doi: 10.1007/s004410050849.
    1. Houge G, Robaye B, Eikhom TS, Golstein J, Mellgren G, Gjertsen BT, et al. Fine mapping of 28S rRNA sites specifically cleaved in cells undergoing apoptosis. Mol Cell Biol. 1995;15:2051–62. doi: 10.1128/MCB.15.4.2051.
    1. Houge G, Doskeland SO, Boe R, Lanotte M. Selective cleavage of 28S rRNA variable regions V3 and V13 in myeloid leukemia cell apoptosis. FEBS Lett. 1993;315:16–20. doi: 10.1016/0014-5793(93)81123-H.
    1. Gjertsen BT, Cressey LI, Ruchaud S, Houge G, Lanotte M, Doskeland SO. Multiple apoptotic death types triggered through activation of separate pathways by cAMP and inhibitors of protein phosphatases in one (IPC leukemia) cell line. J Cell Sci. 1994;107(Pt 12):3363–77.
    1. Hoat TX, Nakayashiki H, Tosa Y, Mayama S. Specific cleavage of ribosomal RNA and mRNA during victorin-induced apoptotic cell death in oat. Plant J. 2006;46:922–33. doi: 10.1111/j.1365-313X.2006.02752.x.
    1. Mroczek S, Kufel J. Apoptotic signals induce specific degradation of ribosomal RNA in yeast. Nucleic Acids Res. 2008;36:2874–88. doi: 10.1093/nar/gkm1100.
    1. King KL, Jewell CM, Bortner CD, Cidlowski JA. 28S ribosome degradation in lymphoid cell apoptosis: evidence for caspase and Bcl-2-dependent and -independent pathways. Cell Death Differ. 2000;7:994–1001. doi: 10.1038/sj.cdd.4400731.
    1. Parissenti AM, Chapman JA, Kahn HJ, Guo B, Han L, O'Brien P, et al. Association of low tumor RNA integrity with response to chemotherapy in breast cancer patients. Breast Cancer Res Treat. 2010;119:347–56. doi: 10.1007/s10549-009-0531-x.
    1. Schroeder A, Mueller O, Stocker S, Salowsky R, Leiber M, Gassmann M, et al. The RIN: an RNA integrity number for assigning integrity values to RNA measurements. BMC Mol Biol. 2006;7:3. doi: 10.1186/1471-2199-7-3.
    1. Parissenti AM, Guo B, Pritzker LB, Pritzker KP, Wang X, Zhu M, et al. Tumor RNA disruption predicts survival benefit from breast cancer chemotherapy. Breast Cancer Res Treat. 2015;153:135–44. doi: 10.1007/s10549-015-3498-9.
    1. Jordan P, Carmo-Fonseca M. Cisplatin inhibits synthesis of ribosomal RNA in vivo. Nucleic Acids Res. 1998;26:2831–6. doi: 10.1093/nar/26.12.2831.
    1. Burger K, Muhl B, Harasim T, Rohrmoser M, Malamoussi A, Orban M, et al. Chemotherapeutic drugs inhibit ribosome biogenesis at various levels. J Biol Chem. 2010;285:12416–25. doi: 10.1074/jbc.M109.074211.
    1. Armstrong SR, Narendrula R, Guo B, Parissenti AM, McCallum KL, Cull S, et al. Distinct genetic alterations occur in ovarian tumor cells selected for combined resistance to carboplatin and docetaxel. J Ovarian Res. 2012;5:40. doi: 10.1186/1757-2215-5-40.
    1. He K, Zhou HR, Pestka JJ. Targets and intracellular signaling mechanisms for deoxynivalenol-induced ribosomal RNA cleavage. Toxicol Sci. 2012;127:382–90. doi: 10.1093/toxsci/kfs134.
    1. Nadano D, Sato TA. Caspase-3-dependent and -independent degradation of 28 S ribosomal RNA may be involved in the inhibition of protein synthesis during apoptosis initiated by death receptor engagement. J Biol Chem. 2000;275:13967–73. doi: 10.1074/jbc.275.18.13967.
    1. Brown T, Mackey K, et al. Analysis of RNA by Northern and Slot Blot Hybridization. In: Frederick M, Ausubel RB, Kingston RE, DAvid D, Morre JGS, Smith JA, Struhl K, et al., editors. Current Protocols in Molecular Biology. Volume 1. United States of America: John Wiley and Sons, Inc; 1997. pp. 4.9.1–4.9.16.
    1. Guo B, Hembruff SL, Villeneuve DJ, Kirwan AF, Parissenti AM. Potent killing of paclitaxel- and doxorubicin-resistant breast cancer cells by calphostin C accompanied by cytoplasmic vacuolization. Breast Cancer Res Treat. 2003;82:125–41. doi: 10.1023/B:BREA.0000003969.21267.81.
    1. Wakeman JA, Maden BE. 28 S ribosomal RNA in vertebrates. Locations of large-scale features revealed by electron microscopy in relation to other features of the sequences. Biochem J. 1989;258:49–56. doi: 10.1042/bj2580049.
    1. Li G, Xiang Y, Sabapathy K, Silverman RH. An apoptotic signaling pathway in the interferon antiviral response mediated by RNase L and c-Jun NH2-terminal kinase. J Biol Chem. 2004;279:1123–31. doi: 10.1074/jbc.M305893200.
    1. He K, Zhou HR, Pestka JJ. Mechanisms for ribotoxin-induced ribosomal RNA cleavage. Toxicol Appl Pharmacol. 2012;265:10–8. doi: 10.1016/j.taap.2012.09.017.
    1. Maestre AM, Garzon A, Rodriguez D. Equine torovirus (BEV) induces caspase-mediated apoptosis in infected cells. PLoS ONE. 2011;6:e20972. doi: 10.1371/journal.pone.0020972.
    1. Naito T, Yokogawa T, Kim HS, Futagami M, Wataya Y, Matsuda A, et al. Anticancer mechanisms of 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl) cytosine (ECyd, TAS-106). Nucleic Acids Res Suppl. 2002;241–2.
    1. Olmo N, Turnay J. Gonzalez de Buitrago G, Lopez de Silanes I, Gavilanes JG, Lizarbe MA. Cytotoxic mechanism of the ribotoxin alpha-sarcin. Induction of cell death via apoptosis. Eur J Biochem. 2001;268:2113–23. doi: 10.1046/j.1432-1327.2001.02086.x.
    1. Wataya Y, Takenaka K, Yokogawa T, Matsuda A, Sasaki T, Fukushima M. Cytotoxic mechanism of 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)cytosine (ECyd) Nucleic Acids Symp Ser. 1999;42:133–4. doi: 10.1093/nass/42.1.133.
    1. Samali A, Gilje B, Doskeland SO, Cotter TG, Houge G. The ability to cleave 28S ribosomal RNA during apoptosis is a cell-type dependent trait unrelated to DNA fragmentation. Cell Death Differ. 1997;4:289–93. doi: 10.1038/sj.cdd.4400246.
    1. Banerjee S, An S, Zhou A, Silverman RH, Makino S. RNase L-independent specific 28S rRNA cleavage in murine coronavirus-infected cells. J Virol. 2000;74:8793–802. doi: 10.1128/JVI.74.19.8793-8802.2000.
    1. Hashimoto T, Kikkawa U, Kamada S. Contribution of caspase(s) to the cell cycle regulation at mitotic phase. PLoS ONE. 2011;6:e18449. doi: 10.1371/journal.pone.0018449.
    1. Hashimoto T, Yamauchi L, Hunter T, Kikkawa U, Kamada S. Possible involvement of caspase-7 in cell cycle progression at mitosis. Genes Cells. 2008;13:609–21. doi: 10.1111/j.1365-2443.2008.01192.x.
    1. Yamaguchi K, Uzzo R, Dulin N, Finke JH, Kolenko V. Renal carcinoma cells undergo apoptosis without oligonucleosomal DNA fragmentation. Biochem Biophys Res Commun. 2004;318:710–3. doi: 10.1016/j.bbrc.2004.04.086.
    1. Sakahira H, Enari M, Ohsawa Y, Uchiyama Y, Nagata S. Apoptotic nuclear morphological change without DNA fragmentation. Curr Biol. 1999;9:543–6. doi: 10.1016/S0960-9822(99)80240-1.
    1. Yuste VJ, Bayascas JR, Llecha N, Sanchez-Lopez I, Boix J, Comella JX. The absence of oligonucleosomal DNA fragmentation during apoptosis of IMR-5 neuroblastoma cells: disappearance of the caspase-activated DNase. J Biol Chem. 2001;276:22323–31. doi: 10.1074/jbc.M100072200.
    1. Ossareh-Nazari B, Nino CA, Bengtson MH, Lee JW, Joazeiro CA, Dargemont C. Ubiquitylation by the Ltn1 E3 ligase protects 60S ribosomes from starvation-induced selective autophagy. J Cell Biol. 2014;204:909–17. doi: 10.1083/jcb.201308139.
    1. Fujii K, Kitabatake M, Sakata T, Miyata A, Ohno M. A role for ubiquitin in the clearance of nonfunctional rRNAs. Genes Dev. 2009;23:963–74. doi: 10.1101/gad.1775609.
    1. Sprowl JA, Reed K, Armstrong SR, Lanner C, Guo B, Kalatskaya I, et al. Alterations in tumor necrosis factor signaling pathways are associated with cytotoxicity and resistance to taxanes: a study in isogenic resistant tumor cells. Breast Cancer Res. 2012;14:R2. doi: 10.1186/bcr3083.
    1. Wajant H, Pfizenmaier K, Scheurich P. Tumor necrosis factor signaling. Cell Death Differ. 2003;10:45–65. doi: 10.1038/sj.cdd.4401189.
    1. MacEwan DJ. TNF ligands and receptors--a matter of life and death. Br J Pharmacol. 2002;135:855–75. doi: 10.1038/sj.bjp.0704549.
    1. Kalyanaraman B, Joseph J, Kalivendi S, Wang S, Konorev E, Kotamraju S. Doxorubicin-induced apoptosis: Implications in cardiotoxicity. In Oxygen/Nitrogen Radicals: Cell Injury and Disease. Edited by Vallyathan V, Castranova, Vince, Xianglin Shi. US: Springer; 2002: 119–24: Developments in Molecular and Cellular Biochemistry© 2002.
    1. Cao DX, Qiao B, Ge ZQ, Yuan YJ. Comparison of burst of reactive oxygen species and activation of caspase-3 in apoptosis of K562 and HL-60 cells induced by docetaxel. Cancer Lett. 2004;214:103–13. doi: 10.1016/j.canlet.2004.03.047.
    1. Li D, Ueta E, Kimura T, Yamamoto T, Osaki T. Reactive oxygen species (ROS) control the expression of Bcl-2 family proteins by regulating their phosphorylation and ubiquitination. Cancer Sci. 2004;95:644–50. doi: 10.1111/j.1349-7006.2004.tb03323.x.
    1. Moungjaroen J, Nimmannit U, Callery PS, Wang L, Azad N, Lipipun V, et al. Reactive oxygen species mediate caspase activation and apoptosis induced by lipoic acid in human lung epithelial cancer cells through Bcl-2 down-regulation. J Pharmacol Exp Ther. 2006;319:1062–9. doi: 10.1124/jpet.106.110965.
    1. Simon HU, Haj-Yehia A, Levi-Schaffer F. Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis. 2000;5:415–8. doi: 10.1023/A:1009616228304.
    1. Liudkovska V, Seweryn M, Czarnocka-Cieciura A, Kufel J. Nucleases involved in rRNA degradation in yeast during apoptosis and autophagy, Abstract 432. In: RNA Society 2014 meeting proceedings. Quebec City, Quebec, Ontario, Canada: The RNA Society; 2014. p. 634–7166. Abstract 432.
    1. Gorski JL, Gonzalez IL, Schmickel RD. The secondary structure of human 28S rRNA: the structure and evolution of a mosaic rRNA gene. J Mol Evol. 1987; 24:236–51.

Source: PubMed

3
購読する