Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

The MBNL3 splicing factor promotes hepatocellular carcinoma by increasing PXN expression through the alternative splicing of lncRNA-PXN-AS1

Abstract

Understanding the roles of splicing factors and splicing events during tumorigenesis would open new avenues for targeted therapies. Here we identify an oncofetal splicing factor, MBNL3, which promotes tumorigenesis and indicates poor prognosis of hepatocellular carcinoma patients. MBNL3 knockdown almost completely abolishes hepatocellular carcinoma tumorigenesis. Transcriptomic analysis revealed that MBNL3 induces lncRNA-PXN-AS1 exon 4 inclusion. The transcript lacking exon 4 binds to coding sequences of PXN mRNA, causes dissociation of translation elongation factors from PXN mRNA, and thereby inhibits PXN mRNA translation. In contrast, the transcript containing exon 4 preferentially binds to the 3′ untranslated region of PXN mRNA, protects PXN mRNA from microRNA-24–AGO2 complex-induced degradation, and thereby increases PXN expression. Through inducing exon 4 inclusion, MBNL3 upregulates PXN, which mediates the pro-tumorigenic roles of MBNL3. Collectively, these data demonstrate detailed mechanistic links between an oncofetal splicing factor, a splicing event and tumorigenesis, and establish splicing factors and splicing events as potential therapeutic targets.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: MBNL3 expression is increased in HCC, and NANOG, OCT4 and SOX2 upregulate MBNL3 expression.
Figure 2: MBNL3 drives HCC tumorigenesis.
Figure 3: MBNL3 is required for tumorigenesis of HCC cells.
Figure 4: MBNL3 induces transcriptomic change and alternative splicing of lncRNA-PXN-AS1.
Figure 5: PXN-AS1-L and PXN-AS1-S have different effects on PXN mRNA stability.
Figure 6: PXN-AS1-L and PXN-AS1-S have different effects on PXN mRNA translation.
Figure 7: PXN-AS1-L and PXN-AS1-S have opposing effects on tumorigenesis of HCC cells.
Figure 8: The correlation between MBNL3 expression, lncRNA-PXN-AS1 splicing, and PXN expression in HCC tissues.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

References

  1. Torre, L. A. et al. Global cancer statistics, 2012. CA Cancer J. Clin. 65, 87–108 (2015).

    Article  PubMed  Google Scholar 

  2. Lee, J. S. et al. A novel prognostic subtype of human hepatocellular carcinoma derived from hepatic progenitor cells. Nat. Med. 12, 410–416 (2006).

    Article  CAS  PubMed  Google Scholar 

  3. Becker, D. et al. Genetic signatures shared in embryonic liver development and liver cancer define prognostically relevant subgroups in HCC. Mol. Cancer 11, 55 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Yong, K. J. et al. Oncofetal gene SALL4 in aggressive hepatocellular carcinoma. N. Engl. J. Med. 368, 2266–2276 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bechara, E. G., Sebestyen, E., Bernardis, I., Eyras, E. & Valcarcel, J. RBM5, 6, and 10 differentially regulate NUMB alternative splicing to control cancer cell proliferation. Mol. Cell 52, 720–733 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Zhang, J. & Manley, J. L. Misregulation of pre-mRNA alternative splicing in cancer. Cancer Discov. 3, 1228–1237 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Oltean, S. & Bates, D. O. Hallmarks of alternative splicing in cancer. Oncogene 33, 5311–5318 (2014).

    Article  CAS  PubMed  Google Scholar 

  8. Kornblihtt, A. R. et al. Alternative splicing: a pivotal step between eukaryotic transcription and translation. Nat. Rev. Mol. Cell Biol. 14, 153–165 (2013).

    CAS  PubMed  Google Scholar 

  9. Lee, Y. & Rio, D. C. Mechanisms and regulation of alternative pre-mRNA splicing. Annu. Rev. Biochem. 84, 291–323 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang, Y. et al. The splicing factor RBM4 controls apoptosis, proliferation, and migration to suppress tumor progression. Cancer Cell 26, 374–389 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhou, X. et al. BCLAF1 and its splicing regulator SRSF10 regulate the tumorigenic potential of colon cancer cells. Nat. Commun. 5, 4581 (2014).

    Article  CAS  PubMed  Google Scholar 

  12. Salton, M. et al. Inhibition of vemurafenib-resistant melanoma by interference with pre-mRNA splicing. Nat. Commun. 6, 7103 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Lu, Z. X. et al. Transcriptome-wide landscape of pre-mRNA alternative splicing associated with metastatic colonization. Mol. Cancer Res. 13, 305–318 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Holoch, D. & Moazed, D. RNA-mediated epigenetic regulation of gene expression. Nat. Rev. Genet. 16, 71–84 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Fatica, A. & Bozzoni, I. Long non-coding RNAs: new players in cell differentiation and development. Nat. Rev. Genet. 15, 7–21 (2014).

    Article  CAS  PubMed  Google Scholar 

  16. Yuan, J. H. et al. A long noncoding RNA activated by TGF-β promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell 25, 666–681 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Wang, Y. et al. The long noncoding RNA lncTCF7 promotes self-renewal of human liver cancer stem cells through activation of Wnt signaling. Cell Stem Cell 16, 413–425 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. McHugh, C. A. et al. The Xist lncRNA interacts directly with SHARP to silence transcription through HDAC3. Nature 521, 232–236 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wang, F. et al. Oncofetal long noncoding RNA PVT1 promotes proliferation and stem cell-like property of hepatocellular carcinoma cells by stabilizing NOP2. Hepatology 60, 1278–1290 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Li, T. et al. Multi-stage analysis of gene expression and transcription regulation in C57/B6 mouse liver development. Genomics 93, 235–242 (2009).

    Article  CAS  PubMed  Google Scholar 

  21. Lee, J. S. et al. Transcriptional ontogeny of the developing liver. BMC Genomics 13, 33 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Roessler, S. et al. A unique metastasis gene signature enables prediction of tumor relapse in early-stage hepatocellular carcinoma patients. Cancer Res. 70, 10202–10212 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Chen, X. et al. Gene expression patterns in human liver cancers. Mol. Biol. Cell 13, 1929–1939 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Tung, E. K. et al. Clinicopathological and prognostic significance of serum and tissue Dickkopf-1 levels in human hepatocellular carcinoma. Liver Int. 31, 1494–1504 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Zhu, P. et al. ZIC2-dependent OCT4 activation drives self-renewal of human liver cancer stem cells. J. Clin. Invest. 125, 3795–3808 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Lee, T. K. et al. CD24+ liver tumor-initiating cells drive self-renewal and tumor initiation through STAT3-mediated NANOG regulation. Cell Stem Cell 9, 50–63 (2011).

    Article  CAS  PubMed  Google Scholar 

  27. Shan, J. et al. Nanog regulates self-renewal of cancer stem cells through the insulin-like growth factor pathway in human hepatocellular carcinoma. Hepatology 56, 1004–1014 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Han, H. et al. PBX3 is targeted by multiple miRNAs and is essential for liver tumour-initiating cells. Nat. Commun. 6, 8271 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Hoshida, Y. et al. Prognostic gene expression signature for patients with hepatitis C-related early-stage cirrhosis. Gastroenterology 144, 1024–1030 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Sznajder, L. J. et al. Mechanistic determinants of MBNL activity. Nucleic Acids Res. 44, 10326–10342 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Orengo, J. P., Bundman, D. & Cooper, T. A. A bichromatic fluorescent reporter for cell-based screens of alternative splicing. Nucleic Acids Res. 34, e148 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Poulos, M. G. et al. Progressive impairment of muscle regeneration in muscleblind-like 3 isoform knockout mice. Hum. Mol. Genet. 22, 3547–3558 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Sen, A. et al. Paxillin mediates extranuclear and intranuclear signaling in prostate cancer proliferation. J. Clin. Invest. 122, 2469–2481 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hu, C. T., Cheng, C. C., Wu, J. R., Pan, S. M. & Wu, W. S. PKCɛ-mediated c-Met endosomal processing directs fluctuant c-Met-JNK-paxillin signaling for tumor progression of HepG2. Cell Signal 27, 1544–1555 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Marasek, P. et al. Paxillin-dependent regulation of IGF2 and H19 gene cluster expression. J. Cell Sci. 128, 3106–3116 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chen, D. L. et al. Overexpression of paxillin induced by miR-137 suppression promotes tumor progression and metastasis in colorectal cancer. Carcinogenesis 34, 803–811 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Kato, H. et al. Length-dependent recognition of double-stranded ribonucleic acids by retinoic acid-inducible gene-I and melanoma differentiation-associated gene 5. J. Exp. Med. 205, 1601–1610 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gantier, M. P. & Williams, B. R. The response of mammalian cells to double-stranded RNA. Cytokine Growth Factor Rev. 18, 363–371 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Wu, D. W., Chen, C. Y., Chu, C. L. & Lee, H. Paxillin confers resistance to tyrosine kinase inhibitors in EGFR-mutant lung cancers via modulating BIM and Mcl-1 protein stability. Oncogene 35, 621–630 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Jia, H. L. et al. Gene expression profiling reveals potential biomarkers of human hepatocellular carcinoma. Clin. Cancer Res. 13, 1133–1139 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. Kalsotra, A. et al. A postnatal switch of CELF and MBNL proteins reprograms alternative splicing in the developing heart. Proc. Natl Acad. Sci. USA 105, 20333–20338 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Pascual, M., Vicente, M., Monferrer, L. & Artero, R. The Muscleblind family of proteins: an emerging class of regulators of developmentally programmed alternative splicing. Differentiation 74, 65–80 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Wang, E. T. Transcriptome-wide regulation of pre-mRNA splicing and mRNA localization by muscleblind proteins. Cell 150, 710–724 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Han, H. et al. MBNL proteins repress ES-cell-specific alternative splicing and reprogramming. Nature 498, 241–245 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kanadia, R. N. et al. Developmental expression of mouse muscleblind genes Mbnl1, Mbnl2 and Mbnl3. Gene Expr. Patterns 3, 459–462 (2003).

    Article  CAS  PubMed  Google Scholar 

  46. Zhao, W. et al. 1B50-1, a mAb raised against recurrent tumor cells, targets liver tumor-initiating cells by binding to the calcium channel α2delta1 subunit. Cancer Cell 23, 541–556 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Faghihi, M. A. et al. Expression of a noncoding RNA is elevated in Alzheimer’s disease and drives rapid feed-forward regulation of β-secretase. Nat. Med. 14, 723–730 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Yoon, J. H. et al. LincRNA-p21 suppresses target mRNA translation. Mol. Cell 47, 648–655 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Carrieri, C. et al. Long non-coding antisense RNA controls Uchl1 translation through an embedded SINEB2 repeat. Nature 491, 454–457 (2012).

    CAS  PubMed  Google Scholar 

  50. Wu, D. W. et al. Paxillin promotes tumor progression and predicts survival and relapse in oral cavity squamous cell carcinoma by microRNA-218 targeting. Carcinogenesis 35, 1823–1829 (2014).

    Article  CAS  PubMed  Google Scholar 

  51. Zhao, C. J., Du, S. K., Dang, X. B. & Gong, M. Expression of paxillin is correlated with clinical prognosis in colorectal cancer patients. Med. Sci. Monit. 21, 1989–1995 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Alisi, A. et al. Focal adhesion kinase (FAK) mediates the induction of pro-oncogenic and fibrogenic phenotypes in hepatitis C virus (HCV)-infected cells. PLoS ONE 7, e44147 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wang, R. Y. et al. MUC15 inhibits dimerization of EGFR and PI3K-AKT signaling and is associated with aggressive hepatocellular carcinomas in patients. Gastroenterology 145, 1436–1448.e12 (2013).

    Article  CAS  PubMed  Google Scholar 

  54. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Sharma, E., Sterne-Weiler, T., O’Hanlon, D. & Blencowe, B. J. Global mapping of human RNA–RNA interactions. Mol. Cell 62, 618–626 (2016).

    Article  CAS  PubMed  Google Scholar 

  57. El-Naggar, A. M. et al. Translational activation of HIF1α by YB-1 promotes sarcoma metastasis. Cancer Cell 27, 682–697 (2015).

    Article  CAS  PubMed  Google Scholar 

  58. Faller, W. J. et al. mTORC1-mediated translational elongation limits intestinal tumour initiation and growth. Nature 517, 497–500 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank T. A. Cooper from Baylor College of Medicine, USA, for the gift of the RG6 minigene. This work was supported by grants from the National Key Basic Research Program (973 project) (2015CB554004) from the Ministry of Science and Technology of China, the National Natural Science Foundation of China (Grant No. 81572792, 81330037, 81372240, and 81171937), the Shanghai Sailing Program (15YF1400100), and the Natural Science Foundation of Shanghai (15XD1504500 and 14JC1407800).

Author information

Authors and Affiliations

Authors

Contributions

S.-h.S., J.-h.Y. and F.W. designed the experiments. J.-h.Y., X.-n.L., T.-t.W. and W.P. performed the experiments. J.-h.Y., F.W., X.-n.L., T.-t.W., W.P. and Q.-f.T. analysed the data. W.-p.Z. and Q.-f.T. provided clinical specimens. J.-h.Y. wrote the manuscript. S.-h.S. supervised the project and helped with the writing of the manuscript. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Fang Wang or Shu-han Sun.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 MBNL3 expression is increased in HCC, and NANOG, OCT4, and SOX2 upregulate MBNL3 expression.

(a) Mbnl3 expression levels during mouse liver development from GSE13149 data. (b) Mbnl3 expression levels during mouse liver development from GSE21224 data. (c) MBNL3 expression levels in human HCC tissues (n = 268 independent samples) and noncancerous hepatic tissues (n = 243 independent samples) from GSE25097 data. P < 0.0001 by Mann–Whitney test. (d) MBNL3 expression levels in human HCC tissues (n = 247 independent samples) and noncancerous hepatic tissues (n = 239 independent samples) from GSE14520 data. P < 0.0001 by Mann–Whitney test. For c and d, the horizontal lines in the box plots represent the medians, the boxes represent the interquartile range, and the whiskers represent the minimum and maximum values. (e) The representative images of MBNL3 IHC staining in HCC tissues and noncancerous hepatic tissues. Scale bar, 50 μm. (f) Western blot analysis of MBNL3 expression in human immortalized, nontransformed liver cell line (QSG-7701), HCC cell lines (SMMC-7721, HCCLM3, MHCC97H, Huh7 and Hep3B), and hepatoblastoma cell line (HepG2). (g) Western blot analysis of NANOG, OCT4, and SOX2 expression in SMMC-7721 cells overexpressing NANOG, OCT4, and SOX2. (h) Western blot analysis of NANOG, OCT4, and SOX2 expression in NANOG, OCT4, and SOX2 depleted SMMC-7721 cells. (i) Western blot analysis of NANOG, OCT4, and SOX2 expression in human immortalized, nontransformed liver cell line (QSG-7701), HCC cell lines (SMMC-7721, HCCLM3, and Huh7), and hepatoblastoma cell line (HepG2). (j) Schematic outlining the predicted binding sites for NANOG, OCT4, and SOX2 in the MBNL3 promoter. (k) ChIP assays in SMMC-7721 cells using anti-NANOG, anti-OCT4, anti-SOX2, or nonspecific IgG. Enrichment was determined relative to input controls. Site N was used as negative control with no binding sites for NANOG, OCT4, and SOX2. Data are mean ± s.d. of n = 3 independent experiments. P < 0.05, P < 0.01, P < 0.001 by Student’s t-test. Source data are available in Supplementary Table 8. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 2 MBNL3 drives HCC tumorigenesis and is required for HCC tumorigenesis.

(ab) Tumors generated by subcutaneous injection with MBNL3 stably overexpressed SMMC-7721 cells into nude mice were stained for Ki67 (a) or TUNEL (b). n = 5 independent samples. Scale bars, 50 μm. (c,d) Tumors generated by subcutaneous injection with MBNL3 stably overexpressed QSG-7701 cells into nude mice were stained for Ki67 (c) or TUNEL (d). n = 4 independent samples. Scale bars, 50 μm. For ad, data are mean ± s.d. P < 0.05, P < 0.01 by Mann–Whitney test. (e) Representative hematoxylin and eosin-stained images of liver tissues isolated from the mice at 28 day after intrasplenic injection with indicated SMMC-7721 cells. Scale bars, 500 μm. (f) The mRNA and protein levels of MBNL3 in MBNL3 stably silenced Huh7 cells. (g,h) Cell proliferations were measured using CCK-8 assays (g) or EdU immunofluorescence staining (h) in MBNL3 stably silenced Huh7 cells. Scale bars, 100 μm. (i) Colony formation assays of MBNL3 stably silenced Huh7 cells. Scale bars, 5 mm. (j) Cell apoptosis was detected by TUNEL staining in MBNL3 stably silenced Huh7 cells. Scale bars, 100 μm. For fj, data are mean ± s.d. of n = 3 independent experiments. P < 0.05, P < 0.01, P < 0.001 by Student’s t-test. (k) Western blot analysis of apoptosis markers in MBNL3 stably silenced Huh7 cells. (l) Effects of MBNL3 knockdown in SMMC-7721 cells on subcutaneous tumor growth. Image of mice 28 days after injection is shown. (m) Effects of another independent shRNA knockdown of MBNL3 in SMMC-7721 cells on subcutaneous tumor growth. Image of mice 35 days after injection is shown. (n) Effects of MBNL3 knockdown in Huh7 cells on subcutaneous tumor growth. Images of mice and tumors 28 days after injection are shown. Scale bars, 1 cm. (o) Representative hematoxylin and eosin-stained images of liver tissues isolated from the mice 28 days after intrasplenic injection with indicated SMMC-7721 cells. Scale bars, 500 μm. Source data are available in Supplementary Table 8. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 3 Depletion of MBNL3 inhibits HCC cells growth in vitro and in vivo.

(ad) The protein levels of MBNL3 in MBNL3 stably depleted MHCC97H (a), HCCLM3 (b), HepG2 (c), or Hep3B (d) cells. (eh) Cell proliferations were measured using CCK-8 assays in MBNL3 stably depleted MHCC97H (e), HCCLM3 (f), HepG2 (g), or Hep3B (h) cells. (il) Cell proliferations were assessed using EdU immunofluorescence staining in MBNL3 stably depleted MHCC97H (i), HCCLM3 (j), HepG2 (k), or Hep3B (l) cells. Scale bars, 100 μm. (m-p) Cell apoptosis was detected by TUNEL staining in MBNL3 stably depleted MHCC97H (m), HCCLM3 (n), HepG2 (o), or Hep3B (p) cells. Scale bars, 100 μm. For e-p, data are mean ± s.d. of n = 3 independent experiments. P < 0.05, P < 0.01, P < 0.001 by Student’s t-test. (qx) Effects of MBNL3 depletion on MHCC97H (qu), HCCLM3 (rv), HepG2 (sw), or Hep3B (tx) cells subcutaneous tumor growth. Tumor volumes were measured every 7 days (qt). The mice were killed at 28 day after injection, and the tumors were excised and weighed (ux). Data are mean ± s.d., n = 8 mice in each group for MHCC97H and HCCLM3 cells, n = 7 mice in each group for HepG2 and Hep3B cells, P < 0.05, P < 0.01, P < 0.001 by Mann–Whitney test.

Supplementary Figure 4 MBNL3 induced alternative splicing of lncRNA-PXN-AS1.

(a) GSEA of gene signatures reported to be associated with poor or good survival in MBNL3 shRNA-2 stably transduced SMMC-7721 cells versus control cells. NES, normalized enrichment score. (b) Inclusion of exon 4 was examined in MBNL1 or MBNL2 overexpressed SMMC-7721 cells by RT-PCR. (c) LncRNA-PXN-AS1 isoforms expression levels was examined in SMMC-7721 cells overexpressing MBNL1 or MBNL2 by real-time qPCR. (d) A schematic showing PXN-AS1 minigene containing exon 4, 250bp of its upstream intron 3, and 302bp of its downstream intron 4. (e) The insert sequence of pre-lncRNA-PXN-AS1. The underlined sequences are exon 4. The previous reported putative MBNL3 binding motif YGCY were highlighted in red. (fg) RT-PCR analysis of PXN-AS1 minigene coexpressed with MBNL3 expressing plasmid (f) or MBNL3 shRNA (g). (h) Binding of MBNL3 with pre-lncRNA-PXN-AS1 was detected by CLIP assays in SMMC-7721 cells exogenously expressing PXN-AS1 minigene with primer pairs complementary to MBNL3 intron 3, exon 4, intron 4, or S100A4 by real-time qPCR. (i) RT-PCR analysis of the effects of MBNL3 overexpression on wild type PXN-AS1 minigene (PXN-AS1-wt), PXN-AS1 minigene with mutation of the predicated MBNL3 binding sites in intron 3, exon 4, or intron 4. (j) Inclusion of exon 4 was examined in human fetal livers and adult livers by RT-PCR. (k) Inclusion of exon 4 was examined in human nontransformed liver cell line and HCC cell lines by RT-PCR. For b,c, and fk, data are mean ± s.d. of n = 3 independent experiments. P < 0.05, P < 0.01 by Student’s t-test. Source data are available in Supplementary Table 8. Uncropped images of gels are shown in Supplementary Fig. 9.

Supplementary Figure 5 Characterization of lncRNA-PXN-AS1 and the different effects of lncRNA-PXN-AS1 isoforms on PXN.

(a) Left; representative images of PCR products from the 5′-RACE or 3′-RACE procedure. The major PCR product is marked by an arrow. Right; sequencing of PCR products revealed the boundary of lncRNA-PXN-AS1 sequences. (b) The nucleotide sequence of full-length PXN-AS1-L and PXN-AS1-S. (c) The levels of PXN-AS1-L and PXN-AS1-S in purified polyadenylated RNAs. β-actin and U6 serve as poly (A) positive and negative control, respectively. (d) The levels of PXN-AS1-L and PXN-AS1-S in purified cytoplasmic or nuclear RNAs. β-actin and U6 serve as cytoplasmic and nuclear control, respectively. (e) Confocal RNA FISH images showed cytoplasmic localization of lncRNA-PXN-AS1. Scale bars, 10 μm. The probe detects both PXN-AS1-L and PXN-AS1-S. (f) PXN protein levels in PXN-AS1-L or PXN-AS1-S overexpressed QSG-7701 cells. (g) AMT crosslinking followed by proximity ligation of interacting RNA and sequencing confirmed the interactions between PXN-AS1-L exon 4 and PXN 3′UTR. (h) miR-24 expression in QSG-7701, SMMC-7721, HCCLM3, Huh7, and HepG2 cells. For c,d,h, data are mean ± s.d. of n = 3 independent experiments. P < 0.05, P < 0.01, P < 0.001 by Student’s t-test. (i) SMMC-7721 cell lysates were incubated with PXN full-length mRNA and biotin-labeled PXN-AS1-L, or PXN-AS1-L and biotin-labeled PXN full-length mRNA; after pull-down, bound proteins were extracted and assessed by western blot, and bound mRNAs were extracted and assessed by real-time qPCR. (j) Polysome profiles from SMMC-7721 cells incubated with harringtonine for 0 s (up) and 180 s (low) before harvest. The absorbance profile in 254 nm is outlined. (k,l) SMMC-7721 cells transiently expressing indicated transcripts of lncRNA-PXN-AS1 were incubated with harringtonine for 0 s and 180 s. Then polysomes in cytoplasmic extracts from these cells were fractionated through sucrose gradients, and the relative distributions of PXN (k) or β-actin (l) mRNAs in gradient fractions were measured by real-time qPCR, and polysomes fractions are shown as percentage of total RNA in the gradient. Source data are available in Supplementary Table 8. Uncropped images of gels and blots are shown in Supplementary Fig. 9.

Supplementary Figure 6 PXN modulates MCL1 and BIM expression, and promotes HCC tumorigenesis.

(a) PXN protein levels in SMMC-7721 cells stably overexpressing PXN. (b) Western blot analysis of MCL1 and BIM protein levels in SMMC-7721 cells stably overexpressing PXN. (cf) Western blot analysis of MCL1 and BIM protein levels in SMMC-7721 (c) or QSG-7701(d) cells stably overexpressing MBNL3, SMMC-7721 (e) or Huh7 (f) cells stably silencing MBNL3. (g) Cell proliferations were measured using CCK-8 assays in SMMC-7721 cells stably overexpressing PXN. (h) Cell proliferations were assessed using EdU immunofluorescence staining in SMMC-7721 cells stably overexpressing PXN. Scale bars, 100 μm. (i) Colony formation assays of SMMC-7721 cells stably overexpressing PXN. Scale bars, 5 mm. (j) Cell apoptosis was detected by TUNEL staining in SMMC-7721 cells stably overexpressing PXN. Scale bars, 100 μm. For g,j, data are mean ± s.d. of n = 3 independent experiments P < 0.05, P < 0.01 by Student’s t-test. (k) Western blot analysis of apoptosis markers in SMMC-7721 cells stably overexpressing PXN. (l,n) Effects of PXN overexpression in SMMC-7721 cells on subcutaneous tumor growth. Tumor volumes were measured every 7 days (l). The mice were killed 21 days after injection, and the tumors were excised and weighed (m,n). Scale bars, 1 cm. Data are mean ± s.d. of n = 10 mice in each group, P < 0.01 by Mann–Whitney test. Source data are available in Supplementary Table 8. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 7 The biologic effects of MBNL3 on HCC cells tumorigenesis are partially medicated by AS of lncRNA-PXN-AS1.

(a) SMMC-7721 cells with stable coexpression of shMBNL3 and PXN-AS1-L were generated. PXN-AS1-L expression levels in indicated cells were confirmed by real-time qPCR. (b) PXN protein levels in indicated SMMC-7721 cells. (c,d) Cell proliferations were measured using CCK-8 assays (c) or EdU immunofluorescence staining (d) in indicated cells. Scale bars, 100 μm. (e) Cell apoptosis was detected by TUNEL staining in indicated cells. Scale bars, 100 μm. (f) Western blot analysis of apoptosis markers in indicated cells. (g,h) PXN-AS1-L partially rescues the effects of MBNL3 depletion on subcutaneous tumor growth. Tumor volumes were measured every 7 days (g). The mice were killed 28 days after injection, and the tumors were excised and weighed (h). Scale bars, 1 cm. (i) SMMC-7721 cells with stable coexpression of MBNL3 and PXN-AS1-S were generated. PXN-AS1-S expression levels in indicated cells were confirmed by real-time qPCR analysis. (j) PXN protein levels in indicated SMMC-7721 cells. (k,l) Cell proliferations were measured using CCK-8 assays (k) or EdU immunofluorescence staining (l) in indicated cells. Scale bars, 100 μm. (m) Cell apoptosis was detected by TUNEL staining in indicated cells. Scale bars, 100 μm. (n) Western blot analysis of apoptosis markers in indicated cells. (o,p) PXN-AS1-S partially rescues the effects of MBNL3 overexpression on subcutaneous tumor growth. Tumor volumes were measured every 7 days (o). The mice were killed 28 days after injection, and the tumors were excised and weighed (p). Scale bars, 1 cm. For a,ce,i and k-m data are mean ± s.d. of n = 3 independent experiments P < 0.05, P < 0.01, P < 0.001 by Student’s t-test. For g,h,o,p, data are mean ± s.d. of n = 8 mice in each group for g,h, and n = 7 mice in each group for o,p P < 0.05, P < 0.01,P < 0.001 by Mann–Whitney test. Source data are available in Supplementary Table 8. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 8 Depletion of PXN abolishes the biological effects of MBNL3 overexpression.

(a) SMMC-7721 cells with stable coexpression of MBNL3/shPXN or controls were generated. The protein levels of PXN in indicated cells were confirmed by western blot. (b) Cell proliferations were measured using CCK-8 assays in indicated SMMC-7721 cells. (c) Cell proliferations were assessed using EdU immunofluorescence staining in indicated SMMC-7721 cells. Scale bars, 100 μm. (d) Colony formation assays of indicated SMMC-7721 cells. Scale bars, 5 mm. (e) Cell apoptosis was detected by TUNEL staining in indicated SMMC-7721 cells. Scale bars, 100 μm. For be, data are mean ± s.d. of n = 3 independent experiments P < 0.05, P < 0.01, P < 0.001 by Student’s t-test. (f) Western blot analysis of apoptosis markers in indicated SMMC-7721 cells. (gi) Depletion of PXN abolishes the effects of MBNL3 overexpression on subcutaneous tumor growth. Tumor volumes were measured every 7 days (g). The mice were killed at 21 day after injection, and the tumors were excised and weighed (hi). Scale bars, 1 cm. Data are mean ± s.d. of n = 8 mice in each group P < 0.001 by Mann–Whitney test. Source data are available in Supplementary Table 8. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary information

Supplementary Information

Supplementary Information (PDF 5797 kb)

Supplementary Table 1

Supplementary Information (XLSX 10 kb)

Supplementary Table 2

Supplementary Information (XLSX 9 kb)

Supplementary Table 3

Supplementary Information (XLSX 10 kb)

Supplementary Table 4

Supplementary Information (XLSX 16 kb)

Supplementary Table 5

Supplementary Information (XLSX 56 kb)

Supplementary Table 6

Supplementary Information (XLSX 10 kb)

Supplementary Table 7

Supplementary Information (XLSX 12 kb)

Supplementary Table 8

Supplementary Information (XLSX 100 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yuan, Jh., Liu, Xn., Wang, Tt. et al. The MBNL3 splicing factor promotes hepatocellular carcinoma by increasing PXN expression through the alternative splicing of lncRNA-PXN-AS1. Nat Cell Biol 19, 820–832 (2017). https://doi.org/10.1038/ncb3538

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3538

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer