Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2020

Open Access 01-12-2020 | Kidney Cancer | Research

RETRACTED ARTICLE: HnRNP A1 - mediated alternative splicing of CCDC50 contributes to cancer progression of clear cell renal cell carcinoma via ZNF395

Authors: Guoliang Sun, Hui Zhou, Ke Chen, Jin Zeng, Yangjun Zhang, Libin Yan, Weimin Yao, Junhui Hu, Tao Wang, Jinchun Xing, Kefeng Xiao, Lily Wu, Zhangqun Ye, Hua Xu

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2020

Login to get access

Abstract

Background

Aberrant alternative splicing events play critical roles in carcinogenesis and progression of many cancers, while sparse studies regarding to alternative splicing are available for clear cell renal cell carcinoma (ccRCC). We identified that alternative splicing of coiled-coil domain containing 50 (CCDC50) was dysregulated in ccRCC, whereas the clinical significance of this splicing event and its splicing regulation mechanisms were still elusive.

Methods

Bioinformatic algorithm was utilized to identify significant exon skipping events in ccRCC via exon sequencing data from The Cancer Genome Atlas. Semi-quantitative real-time polymerase chain reaction and western blot were used to validate the aberrant expression of different transcripts in renal cancer tissues, cell lines and corresponding noncancerous controls. Short hairpin RNA targeting CCDC50 and overexpressing plasmids for each transcript were introduced into ccRCC cell lines, followed by a series of in vitro and in vivo functional experiments. Moreover, a panel of splicing factors were identified and their roles on splicing regulation of CCDC50 precursor mRNA (pre-mRNA) were studied. Furthermore, RNAseq data were analyzed to elucidate downstream molecules of CCDC50. Two-way analysis of variance and unpaired Student t test were used in statistical analysis.

Results

Pre-mRNA of CCDC50 generated two transcripts, full-length transcript (CCDC50-FL) and truncated transcript (CCDC50-S) with exon 6 skipped. CCDC50-S was overexpressed in ccRCC tissues and cell lines compared to noncancerous counterparts, but CCDC50-FL was only detected in noncancerous tissues and normal renal epithelial cells. Higher percent spliced-in index was associated with better survival in ccRCC patients. In vitro and in vivo functional experiments indicated that CCDC50-S transcript promoted the proliferation, migration, invasion and tumorigenesis of ccRCC, while CCDC50-FL exerted opposite tumor suppressive functions. Besides, we identified that heterogeneous nuclear ribonucleoprotein A1 (HnRNP A1) could promote the skipping of exon 6, which resulted in higher portion of CCDC50-S and oncogenic transformation. Moreover, zinc finger protein 395 (ZNF395) was identified as a downstream protein of CCDC50-S, and the interaction initiated oncogenic pathways which were involved in ccRCC progression.

Conclusions

Aberrant alternative splicing of CCDC50 is regulated by HnRNP A1 in ccRCC. This splicing event contributes to cancer progression through the downstream pathway involving ZNF395.
Appendix
Available only for authorised users
Literature
1.
go back to reference Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67:7–30.CrossRef Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67:7–30.CrossRef
2.
go back to reference Riazalhosseini Y, Lathrop M. Precision medicine from the renal cancer genome. Nat Commun. 2016;12:655–66. Riazalhosseini Y, Lathrop M. Precision medicine from the renal cancer genome. Nat Commun. 2016;12:655–66.
3.
go back to reference Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, Heng DY, Larkin J, Ficarra V. Renal cell carcinoma. Nat Rev Dis Primers. 2017;3:17009.CrossRef Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, Heng DY, Larkin J, Ficarra V. Renal cell carcinoma. Nat Rev Dis Primers. 2017;3:17009.CrossRef
4.
go back to reference Wang ET, Sandberg R, Luo S, Khrebtukova I, Zhang L, Mayr C, Kingsmore SF, Schroth GP, Burge CB. Alternative isoform regulation in human tissue transcriptomes. Nature. 2008;456:470–6.CrossRef Wang ET, Sandberg R, Luo S, Khrebtukova I, Zhang L, Mayr C, Kingsmore SF, Schroth GP, Burge CB. Alternative isoform regulation in human tissue transcriptomes. Nature. 2008;456:470–6.CrossRef
5.
go back to reference Pan Q, Shai O, Lee LJ, Frey BJ, Blencowe BJ. Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat Genet. 2008;40:1413–5.CrossRef Pan Q, Shai O, Lee LJ, Frey BJ, Blencowe BJ. Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat Genet. 2008;40:1413–5.CrossRef
6.
go back to reference Ghigna C, Valacca C, Biamonti G. Alternative splicing and tumor progression. Curr Genomics. 2008;9:556–70.CrossRef Ghigna C, Valacca C, Biamonti G. Alternative splicing and tumor progression. Curr Genomics. 2008;9:556–70.CrossRef
7.
go back to reference David CJ, Manley JL. Alternative pre-mRNA splicing regulation in cancer: pathways and programs unhinged. Genes Dev. 2010;24:2343–64.CrossRef David CJ, Manley JL. Alternative pre-mRNA splicing regulation in cancer: pathways and programs unhinged. Genes Dev. 2010;24:2343–64.CrossRef
8.
go back to reference Valletti A, Gigante M, Palumbo O, Carella M, Divella C, Sbisa E, Tullo A, Picardi E, D'Erchia AM, Battaglia M, et al. Genome-wide analysis of differentially expressed genes and splicing isoforms in clear cell renal cell carcinoma. PLoS One. 2013;8:e78452.CrossRef Valletti A, Gigante M, Palumbo O, Carella M, Divella C, Sbisa E, Tullo A, Picardi E, D'Erchia AM, Battaglia M, et al. Genome-wide analysis of differentially expressed genes and splicing isoforms in clear cell renal cell carcinoma. PLoS One. 2013;8:e78452.CrossRef
9.
go back to reference Chen K, Xiao H, Zeng J, Yu G, Zhou H, Huang C, Yao W, Xiao W, Hu J, Guan W, et al. Alternative splicing of EZH2 pre-mRNA by SF3B3 contributes to the tumorigenic potential of renal Cancer. Clin Cancer Res. 2017;23:3428–41.CrossRef Chen K, Xiao H, Zeng J, Yu G, Zhou H, Huang C, Yao W, Xiao W, Hu J, Guan W, et al. Alternative splicing of EZH2 pre-mRNA by SF3B3 contributes to the tumorigenic potential of renal Cancer. Clin Cancer Res. 2017;23:3428–41.CrossRef
10.
go back to reference Modamio-Hoybjor S, Moreno-Pelayo MA, Mencia A, del Castillo I, Chardenoux S, Armenta D, Lathrop M, Petit C, Moreno F. A novel locus for autosomal dominant nonsyndromic hearing loss (DFNA44) maps to chromosome 3q28-29. Hum Genet. 2003;112:24–8.CrossRef Modamio-Hoybjor S, Moreno-Pelayo MA, Mencia A, del Castillo I, Chardenoux S, Armenta D, Lathrop M, Petit C, Moreno F. A novel locus for autosomal dominant nonsyndromic hearing loss (DFNA44) maps to chromosome 3q28-29. Hum Genet. 2003;112:24–8.CrossRef
11.
go back to reference Modamio-Hoybjor S, Mencia A, Goodyear R, del Castillo I, Richardson G, Moreno F, Moreno-Pelayo MA. A mutation in CCDC50, a gene encoding an effector of epidermal growth factor-mediated cell signaling, causes progressive hearing loss. Am J Hum Genet. 2007;80:1076–89.CrossRef Modamio-Hoybjor S, Mencia A, Goodyear R, del Castillo I, Richardson G, Moreno F, Moreno-Pelayo MA. A mutation in CCDC50, a gene encoding an effector of epidermal growth factor-mediated cell signaling, causes progressive hearing loss. Am J Hum Genet. 2007;80:1076–89.CrossRef
12.
go back to reference Vazza G, Picelli S, Bozzato A, Mostacciuolo ML. Identification and characterization of C3orf6, a new conserved human gene mapping to chromosome 3q28. Gene. 2003;314:113–20.CrossRef Vazza G, Picelli S, Bozzato A, Mostacciuolo ML. Identification and characterization of C3orf6, a new conserved human gene mapping to chromosome 3q28. Gene. 2003;314:113–20.CrossRef
13.
go back to reference Tashiro K, Konishi H, Sano E, Nabeshi H, Yamauchi E, Taniguchi H. Suppression of the ligand-mediated down-regulation of epidermal growth factor receptor by Ymer, a novel tyrosine-phosphorylated and ubiquitinated protein. J Biol Chem. 2006;281:24612–22.CrossRef Tashiro K, Konishi H, Sano E, Nabeshi H, Yamauchi E, Taniguchi H. Suppression of the ligand-mediated down-regulation of epidermal growth factor receptor by Ymer, a novel tyrosine-phosphorylated and ubiquitinated protein. J Biol Chem. 2006;281:24612–22.CrossRef
14.
go back to reference Kameda H, Watanabe M, Bohgaki M, Tsukiyama T, Hatakeyama S. Inhibition of NF-κB signaling via tyrosine phosphorylation of Ymer. Biochem Biophys Res Commun. 2009;378:744–9.CrossRef Kameda H, Watanabe M, Bohgaki M, Tsukiyama T, Hatakeyama S. Inhibition of NF-κB signaling via tyrosine phosphorylation of Ymer. Biochem Biophys Res Commun. 2009;378:744–9.CrossRef
15.
go back to reference Tsukiyama T, Matsuda-Tsukiyama M, Bohgaki M, Terai S, Tanaka S, Hatakeyama S. Ymer acts as a multifunctional regulator in nuclear factor-kappaB and Fas signaling pathways. Mol Med. 2012;18:587–97.CrossRef Tsukiyama T, Matsuda-Tsukiyama M, Bohgaki M, Terai S, Tanaka S, Hatakeyama S. Ymer acts as a multifunctional regulator in nuclear factor-kappaB and Fas signaling pathways. Mol Med. 2012;18:587–97.CrossRef
16.
go back to reference Farfsing A, Engel F, Seiffert M, Hartmann E, Ott G, Rosenwald A, Stilgenbauer S, Dohner H, Boutros M, Lichter P, Pscherer A. Gene knockdown studies revealed CCDC50 as a candidate gene in mantle cell lymphoma and chronic lymphocytic leukemia. Leukemia. 2009;23:2018–26.CrossRef Farfsing A, Engel F, Seiffert M, Hartmann E, Ott G, Rosenwald A, Stilgenbauer S, Dohner H, Boutros M, Lichter P, Pscherer A. Gene knockdown studies revealed CCDC50 as a candidate gene in mantle cell lymphoma and chronic lymphocytic leukemia. Leukemia. 2009;23:2018–26.CrossRef
17.
go back to reference Wang H, Zhang CZ, Lu SX, Zhang MF, Liu LL, Luo RZ, Yang X, Wang CH, Chen SL, He YF, et al. A coiled-coil domain containing 50 splice variant is modulated by serine/arginine-rich splicing factor 3 and promotes hepatocellular carcinoma in mice by the Ras signaling pathway. Hepatology. 2019;69:179–95.CrossRef Wang H, Zhang CZ, Lu SX, Zhang MF, Liu LL, Luo RZ, Yang X, Wang CH, Chen SL, He YF, et al. A coiled-coil domain containing 50 splice variant is modulated by serine/arginine-rich splicing factor 3 and promotes hepatocellular carcinoma in mice by the Ras signaling pathway. Hepatology. 2019;69:179–95.CrossRef
18.
go back to reference Shilo A, Siegfried Z, Karni R. The role of splicing factors in deregulation of alternative splicing during oncogenesis and tumor progression. Mol Cell Oncol. 2015;2:e970955.CrossRef Shilo A, Siegfried Z, Karni R. The role of splicing factors in deregulation of alternative splicing during oncogenesis and tumor progression. Mol Cell Oncol. 2015;2:e970955.CrossRef
19.
go back to reference Yu G, Yao W, Wang J, Ma X, Xiao W, Li H, Xia D, Yang Y, Deng K, Xiao H, et al. LncRNAs expression signatures of renal clear cell carcinoma revealed by microarray. PLoS One. 2012;7:e42377.CrossRef Yu G, Yao W, Wang J, Ma X, Xiao W, Li H, Xia D, Yang Y, Deng K, Xiao H, et al. LncRNAs expression signatures of renal clear cell carcinoma revealed by microarray. PLoS One. 2012;7:e42377.CrossRef
20.
go back to reference Bohgaki M, Tsukiyama T, Nakajima A, Maruyama S, Watanabe M, Koik T, Hatakeyama S. Involvement of Ymer in suppression of NF-kappaB activation by regulated interaction with lysine-63-linked polyubiquitin chain. Biochim Biophys Acta. 2008;1783:826–37.CrossRef Bohgaki M, Tsukiyama T, Nakajima A, Maruyama S, Watanabe M, Koik T, Hatakeyama S. Involvement of Ymer in suppression of NF-kappaB activation by regulated interaction with lysine-63-linked polyubiquitin chain. Biochim Biophys Acta. 2008;1783:826–37.CrossRef
21.
go back to reference Pollazzon M, Grosso S, Papa FT, Katzaki E, Marozza A, Mencarelli MA, Uliana V, Balestri P, Mari F, Renieri A. A 9.3 Mb microdeletion of 3q27.3q29 associated with psychomotor and growth delay, tricuspid valve dysplasia and bifid thumb. Eur J Med Genet. 2009;52:131–3.CrossRef Pollazzon M, Grosso S, Papa FT, Katzaki E, Marozza A, Mencarelli MA, Uliana V, Balestri P, Mari F, Renieri A. A 9.3 Mb microdeletion of 3q27.3q29 associated with psychomotor and growth delay, tricuspid valve dysplasia and bifid thumb. Eur J Med Genet. 2009;52:131–3.CrossRef
22.
go back to reference Chuang WY, Chang H, Chang GJ, Wang TH, Chang YS, Wang TH, Yeh CJ, Ueng SH, Chien HP, Chang CY, et al. Pleomorphic mantle cell lymphoma morphologically mimicking diffuse large B cell lymphoma: common cyclin D1 negativity and a simple immunohistochemical algorithm to avoid the diagnostic pitfall. Histopathology. 2017;70:986–99.CrossRef Chuang WY, Chang H, Chang GJ, Wang TH, Chang YS, Wang TH, Yeh CJ, Ueng SH, Chien HP, Chang CY, et al. Pleomorphic mantle cell lymphoma morphologically mimicking diffuse large B cell lymphoma: common cyclin D1 negativity and a simple immunohistochemical algorithm to avoid the diagnostic pitfall. Histopathology. 2017;70:986–99.CrossRef
23.
go back to reference Zhang J, Manley JL. Misregulation of pre-mRNA alternative splicing in cancer. Cancer Discov. 2013;3:1228–37.CrossRef Zhang J, Manley JL. Misregulation of pre-mRNA alternative splicing in cancer. Cancer Discov. 2013;3:1228–37.CrossRef
24.
go back to reference Bates DO, Morris JC, Oltean S, Donaldson LF. Pharmacology of modulators of alternative splicing. Pharmacol Rev. 2017;69:63–79.CrossRef Bates DO, Morris JC, Oltean S, Donaldson LF. Pharmacology of modulators of alternative splicing. Pharmacol Rev. 2017;69:63–79.CrossRef
25.
go back to reference Camacho-Vanegas O, Weighardt F, Ghigna C, Amaldi F, Riva S, Biamonti G. Growth-dependent and growth-independent translation of messengers for heterogeneous nuclear ribonucleoproteins. Nucleic Acids Res. 1997;25:3950–4.CrossRef Camacho-Vanegas O, Weighardt F, Ghigna C, Amaldi F, Riva S, Biamonti G. Growth-dependent and growth-independent translation of messengers for heterogeneous nuclear ribonucleoproteins. Nucleic Acids Res. 1997;25:3950–4.CrossRef
26.
go back to reference Pollard VW, Michael WM, Nakielny S, Siomi MC, Wang F, Dreyfuss G. A novel receptor-mediated nuclear protein import pathway. Cell. 1996;86:985–94.CrossRef Pollard VW, Michael WM, Nakielny S, Siomi MC, Wang F, Dreyfuss G. A novel receptor-mediated nuclear protein import pathway. Cell. 1996;86:985–94.CrossRef
27.
go back to reference Roy R, Huang Y, Seckl MJ, Pardo OE. Emerging roles of hnRNPA1 in modulating malignant transformation. Wiley Interdiscip Rev RNA. 2017;8:e1431. Roy R, Huang Y, Seckl MJ, Pardo OE. Emerging roles of hnRNPA1 in modulating malignant transformation. Wiley Interdiscip Rev RNA. 2017;8:e1431.
28.
go back to reference Fujiya M, Konishi H, Mohamed Kamel MK, Ueno N, Inaba Y, Moriichi K, Tanabe H, Ikuta K, Ohtake T, Kohgo Y. microRNA-18a induces apoptosis in colon cancer cells via the autophagolysosomal degradation of oncogenic heterogeneous nuclear ribonucleoprotein A1. Oncogene. 2014;33:4847–56.CrossRef Fujiya M, Konishi H, Mohamed Kamel MK, Ueno N, Inaba Y, Moriichi K, Tanabe H, Ikuta K, Ohtake T, Kohgo Y. microRNA-18a induces apoptosis in colon cancer cells via the autophagolysosomal degradation of oncogenic heterogeneous nuclear ribonucleoprotein A1. Oncogene. 2014;33:4847–56.CrossRef
29.
go back to reference Chettouh H, Fartoux L, Aoudjehane L, Wendum D, Claperon A, Chretien Y, Rey C, Scatton O, Soubrane O, Conti F, et al. Mitogenic insulin receptor-a is overexpressed in human hepatocellular carcinoma due to EGFR-mediated dysregulation of RNA splicing factors. Cancer Res. 2013;73:3974–86.CrossRef Chettouh H, Fartoux L, Aoudjehane L, Wendum D, Claperon A, Chretien Y, Rey C, Scatton O, Soubrane O, Conti F, et al. Mitogenic insulin receptor-a is overexpressed in human hepatocellular carcinoma due to EGFR-mediated dysregulation of RNA splicing factors. Cancer Res. 2013;73:3974–86.CrossRef
30.
go back to reference Akerman M, Fregoso OI, Das S, Ruse C, Jensen MA, Pappin DJ, Zhang MQ, Krainer AR. Differential connectivity of splicing activators and repressors to the human spliceosome. Genome Biol. 2015;16:119.CrossRef Akerman M, Fregoso OI, Das S, Ruse C, Jensen MA, Pappin DJ, Zhang MQ, Krainer AR. Differential connectivity of splicing activators and repressors to the human spliceosome. Genome Biol. 2015;16:119.CrossRef
31.
go back to reference David CJ, Chen M, Assanah M, Canoll P, Manley JL. HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature. 2010;463:364–8.CrossRef David CJ, Chen M, Assanah M, Canoll P, Manley JL. HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature. 2010;463:364–8.CrossRef
32.
go back to reference Chen M, Zhang J, Manley JL. Turning on a fuel switch of cancer: hnRNP proteins regulate alternative splicing of pyruvate kinase mRNA. Cancer Res. 2010;70:8977–80.CrossRef Chen M, Zhang J, Manley JL. Turning on a fuel switch of cancer: hnRNP proteins regulate alternative splicing of pyruvate kinase mRNA. Cancer Res. 2010;70:8977–80.CrossRef
33.
go back to reference Sichtig N, Korfer N, Steger G. Papillomavirus binding factor binds to SAP30 and represses transcription via recruitment of the HDAC1 co-repressor complex. Arch Biochem Biophys. 2007;467:67–75.CrossRef Sichtig N, Korfer N, Steger G. Papillomavirus binding factor binds to SAP30 and represses transcription via recruitment of the HDAC1 co-repressor complex. Arch Biochem Biophys. 2007;467:67–75.CrossRef
34.
go back to reference Schroeder L, Herwartz C, Jordanovski D, Steger G. ZNF395 is an activator of a subset of IFN-stimulated genes. Mediat Inflamm. 2017;2017:1248201.CrossRef Schroeder L, Herwartz C, Jordanovski D, Steger G. ZNF395 is an activator of a subset of IFN-stimulated genes. Mediat Inflamm. 2017;2017:1248201.CrossRef
35.
go back to reference Herwartz C, Castillo-Juarez P, Schroder L, Barron BL, Steger G. The transcription factor ZNF395 is required for the maximal hypoxic induction of Proinflammatory cytokines in U87-MG cells. Mediat Inflamm. 2015;2015:804264.CrossRef Herwartz C, Castillo-Juarez P, Schroder L, Barron BL, Steger G. The transcription factor ZNF395 is required for the maximal hypoxic induction of Proinflammatory cytokines in U87-MG cells. Mediat Inflamm. 2015;2015:804264.CrossRef
36.
go back to reference Yao X, Tan J, Lim KJ, Koh J, Ooi WF, Li Z, Huang D, Xing M, Chan YS, Qu JZ, et al. VHL deficiency drives enhancer activation of oncogenes in clear cell renal cell carcinoma. Cancer Discov. 2017;7:1284–305.CrossRef Yao X, Tan J, Lim KJ, Koh J, Ooi WF, Li Z, Huang D, Xing M, Chan YS, Qu JZ, et al. VHL deficiency drives enhancer activation of oncogenes in clear cell renal cell carcinoma. Cancer Discov. 2017;7:1284–305.CrossRef
37.
go back to reference Srivastava S, Mohibi S, Mirza S, Band H, Band V. Epidermal growth factor receptor activation promotes ADA3 acetylation through the AKT-p300 pathway. Cell Cycle. 2017;16:1515–25.CrossRef Srivastava S, Mohibi S, Mirza S, Band H, Band V. Epidermal growth factor receptor activation promotes ADA3 acetylation through the AKT-p300 pathway. Cell Cycle. 2017;16:1515–25.CrossRef
38.
go back to reference Franovic A, Gunaratnam L, Smith K, Robert I, Patten D, Lee S. Translational up-regulation of the EGFR by tumor hypoxia provides a nonmutational explanation for its overexpression in human cancer. Proc Natl Acad Sci U S A. 2007;104:13092–7.CrossRef Franovic A, Gunaratnam L, Smith K, Robert I, Patten D, Lee S. Translational up-regulation of the EGFR by tumor hypoxia provides a nonmutational explanation for its overexpression in human cancer. Proc Natl Acad Sci U S A. 2007;104:13092–7.CrossRef
Metadata
Title
RETRACTED ARTICLE: HnRNP A1 - mediated alternative splicing of CCDC50 contributes to cancer progression of clear cell renal cell carcinoma via ZNF395
Authors
Guoliang Sun
Hui Zhou
Ke Chen
Jin Zeng
Yangjun Zhang
Libin Yan
Weimin Yao
Junhui Hu
Tao Wang
Jinchun Xing
Kefeng Xiao
Lily Wu
Zhangqun Ye
Hua Xu
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2020
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-020-01606-x

Other articles of this Issue 1/2020

Journal of Experimental & Clinical Cancer Research 1/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine