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 transcriptional terminator XRN2 and the RNA-binding protein Sam68 link alternative polyadenylation to cell cycle progression in prostate cancer

Abstract

Alternative polyadenylation (APA) yields transcripts differing in their 3′-end, and its regulation is altered in cancer, including prostate cancer. Here we have uncovered a mechanism of APA regulation impinging on the interaction between the exonuclease XRN2 and the RNA-binding protein Sam68, whose increased expression in prostate cancer is promoted by the transcription factor MYC. Genome-wide transcriptome profiling revealed a widespread impact of the Sam68/XRN2 complex on APA. XRN2 promotes recruitment of Sam68 to its target transcripts, where it competes with the cleavage and polyadenylation specificity factor for binding to strong polyadenylation signals at distal ends of genes, thus promoting usage of suboptimal proximal polyadenylation signals. This mechanism leads to 3′ untranslated region shortening and translation of transcripts encoding proteins involved in G1/S progression and proliferation. Thus, our findings indicate that the APA program driven by Sam68/XRN2 promotes cell cycle progression and may represent an actionable target for therapeutic intervention.

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

Fig. 1: XRN2 physically interacts with Sam68.
Fig. 2: XRN2 and Sam68 expression are positively correlated in PC.
Fig. 3: MYC positively controls XRN2 expression in PC.
Fig. 4: Genome-wide regulation of APA by XRN2 and Sam68 in PC cells.
Fig. 5: Sam68 and XRN2 globally modulate pA selection in the 3′UTR of target transcripts.
Fig. 6: Sam68 and XRN2 repress strong, canonical target pAs.
Fig. 7: XRN2 and Sam68 promotes cell cycle progression through APA modulation.

Similar content being viewed by others

Data availability

The sequencing data generated in this study are deposited in the Gene Expression Omnibus at GSE198872. Public sequencing data used in this study are deposited under GSE37401 (https://doi.org/10.1016/j.celrep.2012.05.003)54, GSE85164 (https://doi.org/10.1038/nature21715)45, GSE46691(https://doi.org/10.1371/journal.pone.0066855) 41, GSE29079 (https://doi.org/10.1186/1471-2407-11-507)43 and GSE21034 (https://doi.org/10.1016/j.ccr.2010.05.026)42. Prostate adenocarcinoma (TGCA, PanCancer Atlas, 494 samples) gene expression data (mRNA) and clinical data (Progression-Free Survival) were downloaded from cBioPortal (https://www.cbioportal.org/study/summary?id=prad_tcga_pan_can_atlas_2018). Source data are provided with this paper.

Code availability

Code used to analyze the 3′READS-seq is available at https://github.com/DinghaiZ/3-prime-READS-plus.

References

  1. Proudfoot, N. J. Transcriptional termination in mammals: stopping the RNA polymerase II juggernaut. Science 352, aad9926 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Tian, B. & Manley, J. L. Alternative polyadenylation of mRNA precursors. Nat. Rev. Mol. Cell Biol. 18, 18–30 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Tian, B. & Graber, J. H. Signals for pre-mRNA cleavage and polyadenylation. Wiley Interdiscip. Rev. RNA 3, 385–396 (2012).

    Article  CAS  PubMed  Google Scholar 

  4. Elkon, R., Ugalde, A. P. & Agami, R. Alternative cleavage and polyadenylation: extent, regulation and function. Nat. Rev. Genet. 14, 496–506 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Zhu, Y. et al. Molecular mechanisms for CFIm-mediated regulation of mRNA alternative polyadenylation. Mol. Cell 69, 62–74 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Derti, A. et al. A quantitative atlas of polyadenylation in five mammals. Genome Res. 22, 1173–1183 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hwang, H. W. et al. PAPERCLIP identifies microRNA targets and a role of CstF64/64tau in promoting non-canonical poly(A) site usage. Cell Rep. 15, 423–435 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Venkataraman, K., Brown, K. M. & Gilmartin, G. M. Analysis of a noncanonical poly(A) site reveals a tripartite mechanism for vertebrate poly(A) site recognition. Genes Dev. 19, 1315–1327 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Schwich, O. D. et al. SRSF3 and SRSF7 modulate 3′UTR length through suppression or activation of proximal polyadenylation sites and regulation of CFIm levels. Genome Biol. 22, 82 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chatrikhi, R. et al. RNA binding protein CELF2 regulates signal-induced alternative polyadenylation by competing with enhancers of the polyadenylation machinery. Cell Rep. 28, 2795–2806 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Naro, C. et al. Functional interaction between U1snRNP and Sam68 insures proper 3′ end pre-mRNA processing during germ cell differentiation. Cell Rep. 26, 2929–2941 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Kaida, D. et al. U1 snRNP protects pre-mRNAs from premature cleavage and polyadenylation. Nature 468, 664–668 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Jenal, M. et al. The poly(A)-binding protein nuclear 1 suppresses alternative cleavage and polyadenylation sites. Cell 149, 538–553 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Nazim, M. et al. Competitive regulation of alternative splicing and alternative polyadenylation by hnRNP H and CstF64 determines acetylcholinesterase isoforms. Nucleic Acids Res. 45, 1455–1468 (2017).

    CAS  PubMed  Google Scholar 

  15. Kyburz, A., Friedlein, A., Langen, H. & Keller, W. Direct interactions between subunits of CPSF and the U2 snRNP contribute to the coupling of pre-mRNA 3′ end processing and splicing. Mol. Cell 23, 195–205 (2006).

    Article  CAS  PubMed  Google Scholar 

  16. Millevoi, S. et al. An interaction between U2AF 65 and CF I(m) links the splicing and 3′ end processing machineries. EMBO J. 25, 4854–4864 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gruber, A. J. & Zavolan, M. Alternative cleavage and polyadenylation in health and disease. Nat. Rev. Genet. 20, 599–614 (2019).

    Article  CAS  PubMed  Google Scholar 

  18. Sandberg, R., Neilson, J. R., Sarma, A., Sharp, P. A. & Burge, C. B. Proliferating cells express mRNAs with shortened 3′ untranslated regions and fewer microRNA target sites. Science 320, 1643–1647 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Singh, P. et al. Global changes in processing of mRNA 3′ untranslated regions characterize clinically distinct cancer subtypes. Cancer Res. 69, 9422–9430 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Xia, Z. et al. Dynamic analyses of alternative polyadenylation from RNA-seq reveal a 3′-UTR landscape across seven tumour types. Nat. Commun. 5, 5274 (2014).

    Article  CAS  PubMed  Google Scholar 

  21. Gruber, A. J. et al. Discovery of physiological and cancer-related regulators of 3′ UTR processing with KAPAC. Genome Biol. 19, 44 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Lee, S. H. et al. Widespread intronic polyadenylation inactivates tumour suppressor genes in leukaemia. Nature 561, 127–131 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Masamha, C. P. et al. CFIm25 links alternative polyadenylation to glioblastoma tumour suppression. Nature 510, 412–416 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Elkon, R. et al. E2F mediates enhanced alternative polyadenylation in proliferation. Genome Biol. 13, R59 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Van Etten, J. L. et al. Targeting a single alternative polyadenylation site coordinately blocks expression of androgen receptor mRNA splice variants in prostate cancer. Cancer Res. 77, 5228–5235 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Attard, G. et al. Prostate cancer. Lancet 387, 70–82 (2016).

    Article  PubMed  Google Scholar 

  27. Phillips, J. W. et al. Pathway-guided analysis identifies Myc-dependent alternative pre-mRNA splicing in aggressive prostate cancers. Proc. Natl Acad. Sci. USA 117, 5269–5279 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhang, D. et al. Intron retention is a hallmark and spliceosome represents a therapeutic vulnerability in aggressive prostate cancer. Nat. Commun. 11, 2089 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Li, L. et al. 3′UTR shortening identifies high-risk cancers with targeted dysregulation of the ceRNA network. Sci. Rep. 4, 5406 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Burd, C. J. et al. Cyclin D1b variant influences prostate cancer growth through aberrant androgen receptor regulation. Proc. Natl Acad. Sci. USA 103, 2190–2195 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. David, C. J., Chen, M., Assanah, M., Canoll, P. & Manley, J. L. HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature 463, 364–368 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Caggiano, C., Pieraccioli, M., Panzeri, V., Sette, C. & Bielli, P. c-MYC empowers transcription and productive splicing of the oncogenic splicing factor Sam68 in cancer. Nucleic Acids Res. 47, 6160–6171 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Paronetto, M. P. et al. Alternative splicing of the cyclin D1 proto-oncogene is regulated by the RNA-binding protein Sam68. Cancer Res. 70, 229–239 (2010).

    Article  CAS  PubMed  Google Scholar 

  34. Stockley, J. et al. The RNA-binding protein Sam68 regulates expression and transcription function of the androgen receptor splice variant AR-V7. Sci. Rep. 5, 13426 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. La Rosa, P. et al. Sam68 promotes self-renewal and glycolytic metabolism in mouse neural progenitor cells by modulating Aldh1a3 pre-mRNA 3′-end processing. eLife 5, e20750 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Iijima, Y. et al. SAM68-specific splicing is required for proper selection of alternative 3′ UTR isoforms in the nervous system. iScience 22, 318–335 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Bielli, P., Busa, R., Paronetto, M. P. & Sette, C. The RNA-binding protein Sam68 is a multifunctional player in human cancer. Endocr. Relat. Cancer 18, R91–R102 (2011).

    Article  CAS  PubMed  Google Scholar 

  38. Bielli, P. et al. The transcription factor FBI-1 inhibits SAM68-mediated BCL-X alternative splicing and apoptosis. EMBO Rep. 15, 419–427 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Busa, R. et al. The RNA-binding protein Sam68 contributes to proliferation and survival of human prostate cancer cells. Oncogene 26, 4372–4382 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Rajan, P. et al. The RNA-binding and adaptor protein Sam68 modulates signal-dependent splicing and transcriptional activity of the androgen receptor. J. Pathol. 215, 67–77 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Jenkins, R. B., Qian, J., Lieber, M. M. & Bostwick, D. G. Detection of c-myc oncogene amplification and chromosomal anomalies in metastatic prostatic carcinoma by fluorescence in situ hybridization. Cancer Res. 57, 524–531 (1997).

    CAS  PubMed  Google Scholar 

  42. Taylor, B. S. et al. Integrative genomic profiling of human prostate cancer. Cancer Cell 18, 11–22 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Brase, J. C. et al. TMPRSS2-ERG -specific transcriptional modulation is associated with prostate cancer biomarkers and TGF-β signaling. BMC Cancer 11, 507 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sathianathen, N. J., Konety, B. R., Crook, J., Saad, F. & Lawrentschuk, N. Landmarks in prostate cancer. Nat. Rev. Urol. 15, 627–642 (2018).

    Article  PubMed  Google Scholar 

  45. Aktas, T. et al. DHX9 suppresses RNA processing defects originating from the Alu invasion of the human genome. Nature 544, 115–119 (2017).

    Article  CAS  PubMed  Google Scholar 

  46. Eaton, J. D. et al. Xrn2 accelerates termination by RNA polymerase II, which is underpinned by CPSF73 activity. Genes Dev. 32, 127–139 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Nimura, K. et al. Regulation of alternative polyadenylation by Nkx2-5 and Xrn2 during mouse heart development. eLife 5, e16030 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Hoque, M. et al. Analysis of alternative cleavage and polyadenylation by 3′ region extraction and deep sequencing. Nat. Methods 10, 133–139 (2013).

    Article  CAS  PubMed  Google Scholar 

  49. Herzel, L., Ottoz, D. S. M., Alpert, T. & Neugebauer, K. M. Splicing and transcription touch base: co-transcriptional spliceosome assembly and function. Nat. Rev. Mol. Cell Biol. 18, 637–650 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Pandya-Jones, A. & Black, D. L. Co-transcriptional splicing of constitutive and alternative exons. RNA 15, 1896–1908 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Tang, P. et al. Alternative polyadenylation by sequential activation of distal and proximal PolyA sites. Nat. Struct. Mol. Biol. 29, 21–31 (2022).

    Article  CAS  PubMed  Google Scholar 

  52. Fong, N. et al. Effects of transcription elongation rate and Xrn2 exonuclease activity on RNA polymerase II termination suggest widespread kinetic competition. Mol. Cell 60, 256–267 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Shi, Y. Alternative polyadenylation: new insights from global analyses. RNA 18, 2105–2117 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Martin, G., Gruber, A. R., Keller, W. & Zavolan, M. Genome-wide analysis of pre-mRNA 3′ end processing reveals a decisive role of human cleavage factor I in the regulation of 3′ UTR length. Cell Rep. 1, 753–763 (2012).

    Article  CAS  PubMed  Google Scholar 

  55. Galarneau, A. & Richard, S. The STAR RNA binding proteins GLD-1, QKI, SAM68 and SLM-2 bind bipartite RNA motifs. BMC Mol. Biol. 10, 47 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Feracci, M. et al. Structural basis of RNA recognition and dimerization by the STAR proteins T-STAR and Sam68. Nat. Commun. 7, 10355 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Baxley, R. M. & Bielinsky, A. K. Mcm10: a dynamic scaffold at eukaryotic replication forks. Genes (Basel) 8, 73 (2017).

    Article  PubMed Central  Google Scholar 

  58. Da-Silva, L. F. & Duncker, B. P. ORC function in late G1: maintaining the license for DNA replication. Cell Cycle 6, 128–130 (2007).

    Article  CAS  PubMed  Google Scholar 

  59. Paronetto, M. P. et al. Sam68 regulates translation of target mRNAs in male germ cells, necessary for mouse spermatogenesis. J. Cell Biol. 185, 235–249 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Labbe, D. P. & Brown, M. Transcriptional regulation in prostate cancer. Cold Spring Harb. Perspect. Med. 8, a030437 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Brumbaugh, J. et al. Nudt21 controls cell fate by connecting alternative polyadenylation to chromatin signaling. Cell 172, 106–120 (2018).

    Article  CAS  PubMed  Google Scholar 

  62. Li, W. et al. Alternative cleavage and polyadenylation in spermatogenesis connects chromatin regulation with post-transcriptional control. BMC Biol. 14, 6 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Yao, C. et al. Transcriptome-wide analyses of CstF64-RNA interactions in global regulation of mRNA alternative polyadenylation. Proc. Natl Acad. Sci. USA 109, 18773–18778 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Oh, J. M. et al. U1 snRNP regulates cancer cell migration and invasion in vitro. Nat. Commun. 11, 1 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Morris, A. R. et al. Alternative cleavage and polyadenylation during colorectal cancer development. Clin. Cancer Res. 18, 5256–5266 (2012).

    Article  CAS  PubMed  Google Scholar 

  66. Garcia-Gutierrez, L., Delgado, M. D. & Leon, J. MYC oncogene contributions to release of cell cycle brakes. Genes (Basel) 10, 244 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  67. Bennett, C. F., Krainer, A. R. & Cleveland, D. W. Antisense oligonucleotide therapies for neurodegenerative diseases. Annu. Rev. Neurosci. 42, 385–406 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all members of Sette’s laboratory for fruitful discussions throughout this study. The research reported in this paper was supported by Ministero della Salute ‘Ricerca Finalizzata 2011’ (GR-2011-02348423 to P.B.) and ‘Ricerca Finalizzata 2016’ (RF-2016-02363460 to C.S.), by the Associazione Italiana Ricerca sul Cancro (AIRC IG23416 to C.S.), Ministero della Salute – Ricerca Corrente 2021 and 2022 to IRCCS Fondazione Policlinico Gemelli, and by the National Institutes of Health (GM084089 and GM129069 to B.T.). We acknowledge financial support from the Università Cattolica del Sacro Cuore (UCSC) for the execution and publication of this study.

Author information

Authors and Affiliations

Authors

Contributions

C.S. and P.B. conceived the study. M.P., P.B. and C.S. wrote the manuscript. M.P., C.C., P.B. and C.S. designed the experiments. M.P., C.C., L.M. and P.B. performed the experiments. B.T., M.P., G.B. and C.Z. performed the 3′READS data and bioinformatic analyses. R.L. and S.D.S. performed PC immunohistochemistry analysis.

Corresponding authors

Correspondence to Claudio Sette or Pamela Bielli.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks David Elliott and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editors: Beth Moorefield, Tiago Faial and Carolina Perdigoto, in collaboration with the Nature Structural & Molecular Biology team. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 XRN2 physically interacts with Sam68 (Related to Fig. 1).

a, Nucleotide sequence alignment between XRN2 (CCDS 13144.1, GRCh38.p13) and Clone 177 (Cln177) retrieved from the two-hybrid screen. b, Nucleotide and aminoacid sequence of the region of interaction of XRN2 with Sam68 identified by the two-hybrid screen.

Extended Data Fig. 2 XRN2 physically interacts with Sam68 (Related to Fig. 1).

a,b, Western blot (WB) analysis and Coomassie blue staining of the GST pull-down assay (n = 2) performed using LNCaP nuclear extracts (N.E.) in presence of GST-Sam68 full-length (a) and deletion mutants (b). GST was used as negative control (a,b). A scheme of GST-Sam68 fusion proteins is also shown (b).

Source data

Extended Data Fig. 3 XRN2 and Sam68 expression are positively correlated in PC (Related to Fig. 2).

a-d, Pearson’s correlation between XRN2 and Sam68 expression (a,c) and XRN2 expression in Sam68low (blue circles) and Sam68high (red squares) patient groups (b,d) retrieved from Sawyers (GSE21034) (a,b) and Sueltman (GSE29079) (c,d) datasets. Pearson’s correlation coefficient (r) (two-sided) and the p-values (P) are reported (95% confidence interval) (a,c). In b and d statistical significance was calculated by Mann-Whitney test (two-sided) and the p-values are reported (95% confidence interval). e, Scatter-plot analysis showing the positive correlation (R2 = 0.887) between the expression of XRN2 and Sam68 proteins in PC specimens.

Extended Data Fig. 4 XRN2 and MYC expression are correlated in PC (Related to Fig. 3).

a-d, Pearson’s correlation between XRN2 and MYC expression (a,c) and distribution of XRN2 expression in MYClow (blue circles) and MYChigh (red squares) groups (b,d) retrieved from Sawyers (GSE21034) (a,b) and Sueltman (GSE29079) (c,d) datasets. Pearson’s correlation coefficient (r) (two-sided) and the p-values (P) of the correlation (95% confidence interval) were reported in a and c panels. In b and d statistical significance was calculated by Mann-Whitney test (two-sided) and the p-values are reported (95% confidence interval). e, UCSC Genome Browser snapshot of RNAPII, H3K27Ac and H3K4Me3 ChIP-seq profiles surrounding the TSS of the XRN2 gene. RNAPII (POLR2A), MYC and MAX binding regions are indicated (dark box). f, Schematic representation of the putative XRN2 promoter cloned upstream of the luciferase-based report pGL3-basic plasmid. The putative MYC binding site (E-box) is indicated in bold. g, Bar graph (left panel) represents luciferase activity of XRN2 promoter compared to an intergenic region (intergenic), used as negative control. The luciferase assay was performed in 293 T cells transfected, or not (empty vector, EV), with MYC-pCDNA3 vector (MYC). h,i, qPCR (h) and Western blot (i) analyses of MYC, XRN2 and Sam68 expression in LNCaP and 22Rv1 PC cells lines transfected with Control (si-scr#2) and MYC (si-MYC#2) siRNAs. The expression was reported as fold enrichment (ΔΔCq) of Histone 3. g-i, Data represent mean ± SD of three biological replicates. Statistical significance was calculated by unpaired Student’s t-test (two-sided). In g, the p-values are: intergenic P = 0.686, XRN2 P = 9.6 × 10−6. In h: MYC/LNCaP P = 2 × 10−4, MYC/22Rv1 P = 5.1 × 10−3, XRN2/LNCaP P = 1.5 × 10−3, XRN2/22Rv1 P = 2.7 × 10−3, Sam68/LNCaP P = 8.4 × 10−8, Sam68/22Rv1 P = 3.1 × 10−3). In the representation of panels, statistical value is reported as ** P < 0.01; *** P < 0.001; n.s. not significant.

Source data

Extended Data Fig. 5 Genome-wide regulation of APA by XRN2 and Sam68 in PC cells (Related to Fig. 4).

a, Representative Western-blot analysis of LNCaP cells transfected twice with control (si-Scr), Sam68 (si-Sam68) and XRN2 (si-XRN2) siRNAs. β-actin was used as loading control (n = 3). b, Principal Component Analysis showing variance of 3′READS data from two biological replicates. The red circles, green triangles and blue squares represent pA selection data in control, Sam68 and XRN2 silenced cells, respectively. The proportion of variance (%) for both the first and second principal components is reported. c, 3′READS sample distance analysis. The heatmap show the Euclidean distances between samples. Dendrogram of clustering results are also shown. d, Venn diagram showing the overlap between common regulated genes undergoing to expression (GE) or APA changes in absence of Sam68 (si-Sam68) and XRN2 (si-XRN2) (ns: not significant, modified Fisher’s test). e, Representative 3′RACE PCR analysis (n = 2) of four genes (RCC2, SCAMP2, LAMC1, CD164) undergoing UTR lengthening in absence of Sam68 and XRN2. Downregulated and up-regulated pAs are indicated in orange and purple, respectively.

Source data

Extended Data Fig. 6 Genome-wide regulation of APA by XRN2 and Sam68 in PC cells (Related to Fig. 4).

a,b, Representative Western blot analysis of LNCaP cells transiently (a) or stably (b) depleted for Sam68 and XRN2 (n = 3). β-actin was used as loading control. ce, Bar graphs showing qRT-PCR analyses of pA usage evaluated in 24 representative genes undergoing APA regulation in LNCaP cells treated as in a and b. Fold change of d-pA relative to p-pA was calculated by the ΔCq method. In e, unvalidated genes are shown. Data represent mean ± SD of three biological replicates (ce). In ce, statistical significance was calculated by unpaired Student’s t-test, two-sided (exact p-values reported in source data). In the representation of panels, statistical value is reported as * P < 0.05; ** P < 0.01; *** P < 0.001. UCSC genome browser tracks showing APA regulation for each event analyzed is also shown on the right side of each graph. Purple and orange boxes indicate up- and down-regulated events, respectively.

Source data

Extended Data Fig. 7 Sam68 and XRN2 globally modulate pA selection in the 3′UTR of target transcripts (Related to Fig. 5).

a, Representative Western blot and densitometric (bar graphs) analyses of nuclear matrix subcellular fraction isolated in control (sh-scr), Sam68 (sh-Sam68) and XRN2 (sh-XRN2) stably depleted LNCaP cells. Lamin β-1 was used as loading control. b, Bar graphs showing qPCR analysis of pA usage evaluated in three genes undergoing 3′UTR-APA regulation in cells knocked down for XRN2 targeting 3′UTR (sh-XRN2-3′UTR) and transfected with empty vector (EV), XRN2 wild-type (WT) and catalytically-death mutant (D235A). LNCaP cells stably depleted with sh targeting CDS (sh-XRN2) were used as control. Fold change of distal (d-pA) relative to the proximal pA (p-pA) in the 3′UTR was calculated by the ΔCq method. c, CLIP assays performed in LNCaP cells stably depleted for XRN2 (sh-XRN2) using Sam68 antibody or IgGs, as negative control. RNA associated with Sam68 was quantified by qPCR using primers located upstream of regulated and non-regulated pAs and represented as percentage (%) of input. d, Bar graph showing the qPCR analysis of 4sU-labeled RNA isolated from LNCaP cells stably transduced with control (sh-scr) and XRN2 (sh-XRN2) shRNAs. Labeled RNA is represented as percentage (%) of total RNA used for the assay (input). e, CLIP assays performed in LNCaP cells transfected as in b using Sam68 antibody or IgGs, as negative control. RNA associated with Sam68 was reported as in c. ae, Data represent mean ± SD of three biological replicates. Statistical significance was calculated by unpaired Student’s t-test (two-sided). In panels a,c and b,e the exact p-value is reported in figure and source data, respectively. When not indicated (b,e), p-values are reported as *P < 0.05; ** P < 0.01; *** P < 0.001; n.s.: not significant.

Source data

Extended Data Fig. 8 Sam68 and XRN2 represses strong, distal PAS (Related to Fig. 6).

a, Changes of APA isoform abundance (ΔAbn) of genes presenting at least one regulated pA in LNCaP cells depleted for Sam68 (si-Sam68) or XRN2 (si-XRN2). Mean values (Mean) and number of events (n) are reported. Statistical significance was calculated by unpaired Student’s t-test (two-sided). The p-value is reported. In boxplot, band and box indicate the median and the 25-75th percentile, respectively. Whiskers indicate ±1.5x interquartile range. b, Frequency distribution of the U (upper panel) and C (lower panel) nucleotide in up- (purple line), down- (orange line) and un-regulated (black line) region between −100/+100nt from CS (0). ce, Metagene analyses of CSTF64 (c), CPSF30 (d), and Sam68 (e) CLIP-binding profile with respect to CS (0) in upregulated (purple), downregulated (orange) and non-regulated (black) PASs. f, Scheme of wild-type (FLNB WT) and mutant (FLNB mut) nucleotide sequence surrounding FLNB distal PAS (highlighted in bold). The putative Sam68 binding sites (underline) and mutated bases (red) are indicated. g, RT-PCR (agarose gel) and qPCR (bar graph) analyses of pA usage of wild-type (WT) and mutant (Mut) FLNB minigene evaluated in LNCaP cells transfected, or not, with Sam68-GFP plasmid. Representative Western blot of protein expression is also shown. h, Western blot analysis of RNA-pulldown assay performed using biotin-labeled FLNB WT or Mut RNA. Streptavidin beads were used as control (−) (n = 1). i,j, CLIP assays performed in sh-Sam68 and sh-XRN2 LNCaP cells using CPSF30 antibody. IgG was used as negative control. FLNB and SCARB2 RNA associated with CPSF30 (i) or CSTF64 (j) factors was quantified by qPCR and represented as percentage (%) of input. In g,i,j, statistical significance was calculated by unpaired Student’s t-test, two-sided (n = 3). In g, WT(Sam68-GFP/EV) P = 1.4 × 10−3, Mut(Sam68-GFP/EV) P = 0.777, WT Sam68-GFP/Mut Sam68-GFP P = 9.0 × 10−3; in i, FLNB: CPSF30(sh-Sam68/sh-scr) P = 5.0 × 10−4, CPSF30(sh-XRN2/sh-scr) P = 9.0 × 10−4, SCARB2: CPSF30(sh-Sam68/sh-scr) P = 1.0 × 10−4, CPSF30(sh-XRN2/sh-scr) P = 9.0 × 10−4; in j, FLNB downreg: CSTF64(sh-Sam68/sh-scr) P = 0.1999, CSTF64(sh-XRN2/sh-scr) P = 0.2830; FLNB upreg: CSTF64(sh-Sam68/sh-scr) P = 3.1 × 10−3, CSTF64(sh-XRN2/sh-scr) P = 0.043; SCARB2 downreg: CSTF64(sh-Sam68/sh-scr) P = 0.4242, CSTF64(sh-XRN2/sh-scr) P = 0.4723; SCARB2 upreg: CSTF64(sh-Sam68/sh-scr) P = 1.0 × 10−4, CSTF64(sh-XRN2/sh-scr) P = 0.0468). In the representation of panels, statistical value is reported as *P < 0.05; ** P < 0.01; *** P < 0.001; n.s. not significant.

Source data

Extended Data Fig. 9 XRN2 and SAM68 promotes cell cycle progression through APA modulation (Related to Fig. 7).

a,b, Cell cycle (a) and sub-G1 (b) distribution assessed by PI staining in asynchronous LNCaP cells stably depleted for Sam68 and XRN2. c, Cell cycle distribution assessed by PI staining in asynchronous LNCaP cells stably depleted for MCM10 and ORC2. ac, Data represent mean ± SD of three biological replicates. Statistical significance was calculated by unpaired Student’s t-test, two-sided. In a, the p-values are: G1: sh-Sam68/sh-scr P = 2.2 × 10−3, sh-XRN2/sh-scr P = 8.6 × 10−3; S: sh-Sam68/sh-scr P = 2.0 × 10−4, sh-XRN2/sh-scr P = 1.2 × 10−3; G2-M: sh-Sam68/sh-scr P = 0.037, sh-XRN2/sh-scr P = 0.036; in b, sh-Sam68/sh-scr P = 0.2264, sh-XRN2/sh-scr P = 0.6388; in C, G1: si-MCM10/si-scr P = 2.6 × 10−6, si-ORC2/si-scr P = 3.6 × 10−3; S: si-MCM10/si-scr P = 3.0 × 10−4, si-ORC2/si-scr P = 0.1679; G2-M: si-MCM10/si-scr P = 2 × 10−4, si-ORC2/si-scr P = 0.0917). In the representation of panels, statistical value is reported as * P < 0.05; ** P < 0.01; *** P < 0.001.

Supplementary information

Supplementary Information

Supplementary methods, Tables 1–3 and Fig. 1.

Reporting Summary

Peer Review File

Source data

Source Data Fig. 1

Unprocessed western blots and agarose gel related to Fig. 1.

Source Data Fig. 3

Unprocessed western blots and agarose gels related to Fig. 3.

Source Data Fig. 4

Unprocessed western blots related to Fig. 4.

Source Data Fig. 5

Unprocessed western blots related to Fig. 5.

Source Data Fig. 6

Unprocessed western blots and agarose gels related to Fig. 6.

Source Data Fig. 6

Statistical source data related to Fig. 6.

Source Data Fig. 7

Unprocessed western blots and agarose gels related to Fig. 7.

Source Data Fig. 7

Statistical source data related to Fig. 7.

Source Data Extended Data Fig. 2

Unprocessed western blots and Coomassie gel related to Extended Data 2.

Source Data Extended Data Fig. 4

Unprocessed western blots related to Extended Data 4.

Source Data Extended Data Fig. 5

Unprocessed western blots and agarose gels related to Fig. 5.

Source Data Extended Data Fig. 6

Unprocessed western blots related to Extended Data 6.

Source Data Extended Data Fig. 6

Statistical source data related to Extended Data 6.

Source Data Extended Data Fig. 7

Unprocessed western blots related to Extended Data 7.

Source Data Extended Data Fig. 7

Statistical source data related to Extended Data 7.

Source Data Extended Data Fig. 8

Unprocessed western blots and agarose gels related to Extended Data 8.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pieraccioli, M., Caggiano, C., Mignini, L. et al. The transcriptional terminator XRN2 and the RNA-binding protein Sam68 link alternative polyadenylation to cell cycle progression in prostate cancer. Nat Struct Mol Biol 29, 1101–1112 (2022). https://doi.org/10.1038/s41594-022-00853-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-022-00853-0

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