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:

piRNA-independent function of PIWIL1 as a co-activator for anaphase promoting complex/cyclosome to drive pancreatic cancer metastasis

An Author Correction to this article was published on 15 April 2020

This article has been updated

Abstract

Piwi proteins are normally restricted in germ cells to suppress transposons through associations with Piwi-interacting RNAs (piRNAs), but they are also frequently activated in many types of human cancers. A great puzzle is the lack of significant induction of corresponding piRNAs in cancer cells, as we document here in human pancreatic ductal adenocarcinomas (PDACs), which implies that such germline-specific proteins are somehow hijacked to promote tumorigenesis through a different mode of action. Here, we show that in the absence of piRNAs, human PIWIL1 in PDAC functions as an oncoprotein by activating the anaphase promoting complex/cyclosome (APC/C) E3 complex, which then targets a critical cell adhesion-related protein, Pinin, to enhance PDAC metastasis. This is in contrast to piRNA-dependent PIWIL1 ubiquitination and removal by APC/C during late spermiogenesis. These findings unveil a piRNA-dependent mechanism to switch PIWIL1 from a substrate in spermatids to a co-activator of APC/C in human cancer cells.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: PIWIL1 is largely present in the apo state, without piRNA binding, in human cancer cells.
Fig. 2: PIWIL1 acts as a novel co-activator of the APC/C E3 complex.
Fig. 3: PIWIL1 leads to the constitutive activation of APC/C in PDAC cells.
Fig. 4: The APC/C co-activator function of PIWIL1 is required for its role in PDAC metastasis but not tumour growth.
Fig. 5: Pinin is a substrate of APC/CPIWIL1 in PDAC cells.
Fig. 6: Pinin inhibits cell migration and metastasis but not cell proliferation and tumour growth in PDAC cells.
Fig. 7: Ectopic Pinin overrides the stimulatory effect of PIWIL1 on PDAC metastasis, but not tumour growth.
Fig. 8: Comparison of PIWIL1 and Pinin expression in human PDAC specimens.

Similar content being viewed by others

Data availability

The RNA sequencing data supporting the findings of this study have been deposited in the GEO repository under accession code GSE141340. The MS data have been deposited in ProteomeXchange with the primary accession code PXD017452. The human cancer data were derived from the TCGA Research Network at http://cancergenome.nih.gov/. The dataset derived from this resource that supports the findings of this study can be downloaded from BROAD (http://firebrowse.org/viewGene.html?gene=piwil1). All other data supporting the findings of this study are available from the corresponding authors upon reasonable request.

Change history

References

  1. Juliano, C., Wang, J. & Lin, H. Uniting germline and stem cells: the function of Piwi proteins and the piRNA pathway in diverse organisms. Annu. Rev. Genet. 45, 447–469 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Cox, D. N. et al. A novel class of evolutionarily conserved genes defined by piwi are essential for stem cell self-renewal. Genes Dev. 12, 3715–3727 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Harris, A. N. & Macdonald, P. M. aubergine encodes a Drosophila polar granule component required for pole cell formation and related to eIF2C. Development 128, 2823–2832 (2001).

    Article  CAS  PubMed  Google Scholar 

  4. Deng, W. & Lin, H. miwi, a murine homolog of piwi, encodes a cytoplasmic protein essential for spermatogenesis. Dev. Cell 2, 819–830 (2002).

    Article  CAS  PubMed  Google Scholar 

  5. Kuramochi-Miyagawa, S. et al. Mili, a mammalian member of piwi family gene, is essential for spermatogenesis. Development 131, 839–849 (2004).

    Article  CAS  PubMed  Google Scholar 

  6. Carmell, M. A. et al. MIWI2 is essential for spermatogenesis and repression of transposons in the mouse male germline. Dev. Cell 12, 503–514 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Batista, P. J. et al. PRG-1 and 21U-RNAs interact to form the piRNA complex required for fertility in C. elegans. Mol. Cell 31, 67–78 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Houwing, S., Berezikov, E. & Ketting, R. F. Zili is required for germ cell differentiation and meiosis in zebrafish. EMBO J. 27, 2702–2711 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Aravin, A. et al. A novel class of small RNAs bind to MILI protein in mouse testes. Nature 442, 203–207 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Girard, A., Sachidanandam, R., Hannon, G. J. & Carmell, M. A. A germline-specific class of small RNAs binds mammalian Piwi proteins. Nature 442, 199–202 (2006).

    Article  PubMed  Google Scholar 

  11. Grivna, S. T., Beyret, E., Wang, Z. & Lin, H. A novel class of small RNAs in mouse spermatogenic cells. Genes Dev. 20, 1709–1714 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lau, N. C. et al. Characterization of the piRNA complex from rat testes. Science 313, 363–367 (2006).

    Article  CAS  PubMed  Google Scholar 

  13. Siomi, M. C., Sato, K., Pezic, D. & Aravin, A. A. PIWI-interacting small RNAs: the vanguard of genome defence. Nat. Rev. Mol. Cell Biol. 12, 246–258 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Iwasaki, Y. W., Siomi, M. C. & Siomi, H. PIWI-interacting RNA: its biogenesis and functions. Annu. Rev. Biochem. 84, 405–433 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Czech, B. & Hannon, G. J. One loop to rule them all: the ping-pong cycle and piRNA-guided silencing. Trends Biochem. Sci. 41, 324–337 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ozata, D. M., Gainetdinov, I., Zoch, A., O’Carroll, D. & Zamore, P. D. PIWI-interacting RNAs: small RNAs with big functions. Nat. Rev. Genet. 20, 89–108 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Gou, L. T. et al. Pachytene piRNAs instruct massive mRNA elimination during late spermiogenesis. Cell Res. 24, 680–700 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Watanabe, T. & Lin, H. Posttranscriptional regulation of gene expression by Piwi proteins and piRNAs. Mol. Cell 56, 18–27 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhang, P. et al. MIWI and piRNA-mediated cleavage of messenger RNAs in mouse testes. Cell Res. 25, 193–207 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Goh, W. S. et al. piRNA-directed cleavage of meiotic transcripts regulates spermatogenesis. Genes Dev. 29, 1032–1044 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Barckmann, B. et al. Aubergine iCLIP reveals piRNA-dependent decay of mRNAs involved in germ cell development in the early embryo. Cell Rep. 12, 1205–1216 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Vourekas, A., Alexiou, P., Vrettos, N., Maragkakis, M. & Mourelatos, Z. Sequence-dependent but not sequence-specific piRNA adhesion traps mRNAs to the germ plasm. Nature 531, 390–394 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Watanabe, T., Cheng, E. C., Zhong, M. & Lin, H. Retrotransposons and pseudogenes regulate mRNAs and lncRNAs via the piRNA pathway in the germline. Genome Res. 25, 368–380 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Shen, E. Z. et al. Identification of piRNA binding sites reveals the Argonaute regulatory landscape of the C. elegans germline. Cell 172, 937–951 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhang, D. et al. The piRNA targeting rules and the resistance to piRNA silencing in endogenous genes. Science 359, 587–592 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Rojas-Rios, P. & Simonelig, M. piRNAs and PIWI proteins: regulators of gene expression in development and stem cells. Development 145, dev161786 (2018).

    Article  PubMed  CAS  Google Scholar 

  27. Dai, P. et al. A translation-activating function of MIWI/piRNA during mouse spermiogenesis. Cell 179, 1566–1581 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Dai, P., Wang, X. & Liu, M. F. A dual role of the PIWI/piRNA machinery in regulating mRNAs during mouse spermiogenesis. Sci. China Life Sci. https://doi.org/10.1007/s11427-020-1632-5 (2020).

  29. Sasaki, T., Shiohama, A., Minoshima, S. & Shimizu, N. Identification of eight members of the Argonaute family in the human genome. Genomics 82, 323–330 (2003).

    Article  CAS  PubMed  Google Scholar 

  30. Suzuki, R., Honda, S. & Kirino, Y. PIWI expression and function in cancer. Front. Genet. 3, 204 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Litwin, M., Szczepanska-Buda, A., Piotrowska, A., Dziegiel, P. & Witkiewicz, W. The meaning of PIWI proteins in cancer development. Oncol. Lett. 13, 3354–3362 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Simpson, A. J., Caballero, O. L., Jungbluth, A., Chen, Y. T. & Old, L. J. Cancer/testis antigens, gametogenesis and cancer. Nat. Rev. Cancer 5, 615–625 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. Siddiqi, S. & Matushansky, I. Piwis and piwi-interacting RNAs in the epigenetics of cancer. J. Cell. Biochem. 113, 373–380 (2012).

    Article  CAS  PubMed  Google Scholar 

  34. Ng, K. W. et al. Piwi-interacting RNAs in cancer: emerging functions and clinical utility. Mol. Cancer 15, 5 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Zhao, S. et al. piRNA-triggered MIWI ubiquitination and removal by APC/C in late spermatogenesis. Dev. Cell 24, 13–25 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Peters, J. M. The anaphase promoting complex/cyclosome: a machine designed to destroy. Nat. Rev. Mol. Cell Biol. 7, 644–656 (2006).

    Article  CAS  PubMed  Google Scholar 

  37. Pines, J. Cubism and the cell cycle: the many faces of the APC/C. Nat. Rev. Mol. Cell Biol. 12, 427–438 (2011).

    Article  CAS  PubMed  Google Scholar 

  38. Chang, L. & Barford, D. Insights into the anaphase-promoting complex: a molecular machine that regulates mitosis. Curr. Opin. Struct. Biol. 29, 1–9 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Zhang, J., Wan, L., Dai, X., Sun, Y. & Wei, W. Functional characterization of anaphase promoting complex/cyclosome (APC/C) E3 ubiquitin ligases in tumorigenesis. Biochim. Biophys. Acta 1845, 277–293 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Ouyang, P. & Sugrue, S. P. Identification of an epithelial protein related to the desmosome and intermediate filament network. J. Cell Biol. 118, 1477–1488 (1992).

    Article  CAS  PubMed  Google Scholar 

  41. Ouyang, P. & Sugrue, S. P. Characterization of pinin, a novel protein associated with the desmosome-intermediate filament complex. J. Cell Biol. 135, 1027–1042 (1996).

    Article  CAS  PubMed  Google Scholar 

  42. Yu, B. et al. Methylation as a crucial step in plant microRNA biogenesis. Science 307, 932–935 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gou, L. T. et al. Ubiquitination-deficient mutations in human Piwi cause male infertility by impairing histone-to-protamine exchange during spermiogenesis. Cell 169, 1090–1104 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hasuwa, H., Ishino, K. & Siomi, H. Human PIWI (HIWI) is an azoospermia factor. Sci. China Life Sci. 61, 348–350 (2018).

    Article  PubMed  Google Scholar 

  45. Passmore, L. A. et al. Doc1 mediates the activity of the anaphase-promoting complex by contributing to substrate recognition. EMBO J. 22, 786–796 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Carroll, C. W., Enquist-Newman, M. & Morgan, D. O. The APC subunit Doc1 promotes recognition of the substrate destruction box. Curr. Biol. 15, 11–18 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Glotzer, M., Murray, A. W. & Kirschner, M. W. Cyclin is degraded by the ubiquitin pathway. Nature 349, 132–138 (1991).

    Article  CAS  PubMed  Google Scholar 

  48. Schwab, M., Neutzner, M., Möcker, D. & Seufert, W. Yeast Hct1 recognizes the mitotic cyclin Clb2 and other substrates of the ubiquitin ligase APC. EMBO J. 20, 5165–5175 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Pfleger, C. M. & Kirschner, M. W. The KEN box: an APC recognition signal distinct from the D box targeted by Cdh1. Genes Dev. 14, 655–665 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Dongre, A. & Weinberg, R. A. New insights into the mechanisms of epithelial–mesenchymal transition and implications for cancer. Nat. Rev. Mol. Cell Biol. 20, 69–84 (2019).

    Article  CAS  PubMed  Google Scholar 

  51. Shi, Y., Ouyang, P. & Sugrue, S. P. Characterization of the gene encoding pinin/DRS/memA and evidence for its potential tumor suppressor function. Oncogene 19, 289–297 (2000).

    Article  CAS  PubMed  Google Scholar 

  52. Shi, Y., Tabesh, M. & Sugrue, S. P. Role of cell adhesion-associated protein, pinin (DRS/memA), in corneal epithelial migration. Investig. Ophthalmol. Vis. Sci. 41, 1337–1345 (2000).

    CAS  Google Scholar 

  53. Joo, J. H. et al. Reduction of Pnn by RNAi induces loss of cell–cell adhesion between human corneal epithelial cells. Mol. Vis. 11, 133–142 (2005).

    CAS  PubMed  Google Scholar 

  54. Joo, J. H. et al. Pinin modulates expression of an intestinal homeobox gene, Cdx2, and plays an essential role for small intestinal morphogenesis. Dev. Biol. 345, 191–203 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Genzor, P., Cordts, S. C., Bokil, N. V. & Haase, A. D. Aberrant expression of select piRNA-pathway genes does not reactivate piRNA silencing in cancer cells. Proc. Natl Acad. Sci. USA 116, 11111–11112 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Manchado, E. et al. Targeting mitotic exit leads to tumor regression in vivo: modulation by Cdk1, Mastl, and the PP2A/B55alpha, delta phosphatase. Cancer Cell 18, 641–654 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. Wan, L. et al. The APC/C E3 ligase complex activator FZR1 restricts BRAF oncogenic function. Cancer Discov. 7, 424–441 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Yuan, P. et al. KRAS/NF-kappaB/YY1/miR-489 signaling axis controls pancreatic cancer metastasis. Cancer Res. 77, 100–111 (2017).

    Article  CAS  PubMed  Google Scholar 

  59. Yang, Q. et al. Single-cell CAS-seq reveals a class of short PIWI-interacting RNAs in human oocytes. Nat Commun. 10, 3389 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Jiang, S. et al. MicroRNA-155 functions as an OncomiR in breast cancer by targeting the suppressor of cytokine signaling 1 gene. Cancer Res. 70, 3119–3127 (2010).

    Article  CAS  PubMed  Google Scholar 

  61. Jiang, S. et al. A novel miR-155/miR-143 cascade controls glycolysis by regulating hexokinase 2 in breast cancer cells. EMBO J. 31, 1985–1998 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Zhang, L. F. et al. Suppression of miR-199a maturation by HuR is crucial for hypoxia-induced glycolytic switch in hepatocellular carcinoma. EMBO J. 34, 2671–2685 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Hudson, S. V. et al. Targeted noninvasive imaging of EGFR-expressing orthotopic pancreatic cancer using multispectral optoacoustic tomography. Cancer Res. 74, 6271–6279 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Kraft, C., Gmachl, M. & Peters, J. M. Methods to measure ubiquitin-dependent proteolysis mediated by the anaphase-promoting complex. Methods 38, 39–51 (2006).

    Article  CAS  PubMed  Google Scholar 

  65. Williamson, A., Jin, L. & Rape, M. Preparation of synchronized human cell extracts to study ubiquitination and degradation. Methods Mol. Biol. 545, 301–312 (2009).

    Article  CAS  PubMed  Google Scholar 

  66. Zhong, B. et al. Negative regulation of IL-17-mediated signaling and inflammation by the ubiquitin-specific protease USP25. Nat. Immunol. 13, 1110–1117 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Wisniewski, J. R., Zougman, A., Nagaraj, N. & Mann, M. Universal sample preparation method for proteome analysis. Nat. Methods 6, 359–362 (2009).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank X.-D. Fu from the University of California, San Diego, Y. Zheng from UT Southwestern and members of the M.-F.L. Lab for helpful comments. We thank L. Zhan from the Institute of Nutrition and Health, SIBS, CAS, Y. Yu from Tianjin Medical University, and Q. Shi from the University of Science and Technology of China for experimental assistance. This work was supported by grants from the National Key R&D Program of China (2017YFA0504400), the National Natural Science Foundation of China (91940305, 31830109, 31821004, 31961133022 and 91640201), the Chinese Academy of Sciences (“Strategic Priority Research Program” grant XDB19010203) and the Shanghai Municipal Science and Technology Major Project (17JC1420100, 19JC1410200 and 2017SHZDZX01).

Author information

Authors and Affiliations

Authors

Contributions

M.-F.L. and S.Z. planned the project. F.L., P.Y., M.R., Y.L., L.W., T.L., W.L., J.L., D.L., Y.L., Y.-F.R., S.Z. and M.-F.L. designed the experiments. F.L., P.Y., M.R., W.T., C.-H.J., Y.-P.H., F.Z., T.W., Q.Y. and S.Z. conducted the experiments. F.L., P.Y., S.Z. and M.-F.L. analysed the data. F.L., P.Y., S.Z., D.L. and M.-F.L. wrote the paper. All authors discussed the results and commented on the manuscript. M.-F.L. and S.Z. supervised the overall work.

Corresponding authors

Correspondence to Shuang Zhao or Mo-Fang Liu.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Piwil1 (Hiwi) is aberrantly expressed in various types of human tumors and cancer cell lines.

a, Comparison of Piwil1 expression between primary tumor (red column) and paired adjacent normal tissues (blue column) in various types of human cancers from TCGA database, with the rectangle indicating the range between the third and the first quartile values and the segment inside the rectangle indicating the median values, the whiskers indicating the maximum (top) and minimum (bottom) values, and the unfilled circles as outlier values indicating variations in each box-whisker plot dataset. BLCA, Bladder urothelial carcinoma; BRCA, Breast invasive carcinoma; CESC, Cervical squamous cell carcinoma and endocervical adenocarcinoma; CHOL, Cholangiocarcinoma; COAD, Colon adenocarcinoma; COADREAD, Colorectal adenocarcinoma; ESCA, Esophageal carcinoma; GBM, Glioblastoma multiforme; LGG, Lower grade glioma; HNSC, Head and neck squamous cell carcinoma; KICH, Kidney chromophobe; KICH, Pan-kidney cohort; KIPAN, KICH+KIRC+KIRP; KIRC, Kidney renal clear cell carcinoma; KIRP, Kidney renal papillary cell carcinoma; LIHC, Liver hepatocellular carcinoma; LUAD, Lung adenocarcinoma; LUSC, Lung squamous cell carcinoma; PAAD, Pancreatic adenocarcinoma; PCPG, Pheochromocytoma and paraganglioma; PRAD, Prostate adenocarcinoma; READ, Rectum adenocarcinoma; STAD, Stomach adenocarcinoma; STES, Stomach and esophageal carcinoma; THCA, Thyroid carcinoma; THYM, Thymoma; UCEC, Uterine corpus endometrial carcinoma. The data were downloaded from BROAD (http://firebrowse.org/viewGene.html?gene=piwil1). b, c, qRT-PCR (b) and western blot analyses (c) of Piwil1 expression in various types of human cancer cell lines (left) and PDAC cell lines (right), with 293 T (lane 1) and Flag-PIWIL1-transfected 293 T cells (lane 2) serving as negative and positive controls, respectively. Results shown in b are mean ± SD of three separate experiments. Statistical analysis was performed using two-tailed Student t test. Results shown in c are representative of three independent experiments. Unprocessed blots and statistical source data are provided in Source Data Extended Data Fig. 1 and Statistical Source Data Extended Data Fig. 1, respectively.

Source data

Extended Data Fig. 2 Piwil1 knockdown inhibits the proliferation and migration of PDAC cells.

a-h, Piwil1 knockdown in BxPC-3 cells reduced cell proliferation (a, n=4 independent experiments), anchorage-independent growth (b, n=3 independent experiments), xenograft tumor growth in nude mice (c, n=3 animals), cell migration (d, n=3 independent experiments), invasion (e, n=3 independent experiments), and metastatic colonization in NOD/SCID mice through either orthotopic (f, n=3 animals), spleen (g, n=3 animals) or tail vein injection (h, n=3 animals). il, Piwil1 knockdown in AsPC-1 cells reduced cell proliferation (i, n=4 independent experiments), anchorage-independent growth (j, n=3 independent experiments), cell migration (k, n=3 independent experiments) and invasion (l, n=3 independent experiments). BxPC-3 and AsPC-1 cells were respectively transfected with Piwil1 siRNA (a, b, d, e and i–l) or infected with shPiwil1 pseudovirus (c and f–h), and the assays were performed at 24 h post-transfection. a and i, Cell proliferation by MTT assay. b and j, Soft agar colony formation assay, with the relative number of soft agar foci per field. c, Xenograft assay in nude mice, with the time course of xenograft tumour growth. d and k, Wound healing assay, with the quantitative results of wound closure. e and l, Transwell invasion assay, with the invasive cell numbers per field at 24 h after the cells plated. f, Orthotopic xenograft assay in NOD/SCID mice. Left, the average volumes of xenograft tumors in the pancreata; right, quantification of liver metastases area. g and h, Liver or lung metastatic colonization assays in NOD/SCID mice through spleen (g) or tail vein injection (h), respectively, with the bioluminescence quantification of metastasis tumors. The mean ± SD was plotted in all graphs. Statistical analysis was performed using two-tailed Student t test. Statistical source data are provided in Statistical Source Data Extended Data Fig. 2.

Source data

Extended Data Fig. 3 Ectopic expression of Piwil1 promotes the proliferation and migration of PDAC cells.

Ectopic expression of Piwil1 in PANC-1 cells potently promoted cell proliferation (a, n=4 independent experiments), anchorage-independent growth (b, n=3 independent experiments), xenograft tumor growth in nude mice (c, n=3 animals), cell migration (d, n=3 independent experiments), invasion (e, n=3 independent experiments), and metastatic colonization in NOD/SCID mice through either orthotopic (f, n=3 animals), spleen (g, n=3 animals) or tail vein injection (h, n=3 animals). PANC-1 cells were respectively transfected with Flag-PIWIL1 or control vector p3×Flag, and the assays were performed at 24 h post-transfection. The procedures are similar to that described in Extended Data Fig. 2. The mean ± SD was plotted in all graphs. Statistical analysis was performed using two-tailed Student t test. Statistical source data are provided in Statistical Source Data Extended Data Fig. 3.

Source data

Extended Data Fig. 4 Cyclin B1 is not a direct substrate for APC/CPIWIL1 E3 complex in PDAC cells.

a, PIWIL1 was persistently associated with APC/C complex in AsPC-1 cells during the cell cycle. Both HeLa (lanes 1–4) and AsPC-1 cells (lanes 5–8) were synchronized in G1/S phase, then released and harvested at indicated time points for anti-APC3 co-IP assay of the association of Cdc20, Cdh1, or PIWIL1 with APC/C (top), and cell cycles were examined by FACS (bottom). The phosphorylated APC3 was indicated by “*”. b, c, Piwil1 knockdown (c) or overexpression (d) barely affected Cyclin B1 ubiquitination in PDAC cells. BxPC-3 and AsPC-1 cells were co-transfected with pEF-HA-Ub and scramble siRNA (lanes 1, 4, 7 and 10), Cdc20 siRNA (lanes 2, 3, 8 and 9) or Piwil1 siRNAs (lanes 5, 6, 11 and 12), respectively. PANC-1, Capan-1 and MIA PaCa-2 cells were co-transfected with pEF-HA-Ub and p3xFlag control vector (lanes 1, 3, 5, 7, 9 and 11) or p3xFlag-PIWIL1 (lanes 2, 4, 6, 8, 10 and 12), respectively. Top, anti-Cyclin B1 IP pellets were immunoblotted by anti-Ub for Cyclin B1-Ub conjugates (upper) or anti-Cyclin B1 to normalize the loading amounts (lower). Bottom, cell lysates were immunoblotted for PIWIL1, Cdc20, and Cyclin B1, with β-actin serving as a loading control. d, PIWIL1 depletion did not alter the expression pattern of Cyclins and progression of cell cycle in BxPC-3 cells. 24 h post-transfection with Piwil1 siRNA (lanes 7–12) or control scramble siRNA (lanes 1–6), BxPC-3 cells were synchronized in G1/S phase, then released and harvested at indicated time points for immunoblotting of indicated proteins in cell lysates (left), or FACS analyses of cell cycles (right). e, Gating strategy for analysis of synchronized cells. Results shown in ad are representative of three independent experiments. Unprocessed blots are provided in Source Data Extended Data Fig. 4.

Source data

Extended Data Fig. 5 The APC/C co-activator function is required for PIWIL1 action on cell migration but not proliferation in PDAC cells.

ad, C-box mutations in PIWIL1 did not alter its effect on cell proliferation (a, n=4 independent experiments), anchorage-independent growth (b, n=3 independent experiments), but markedly impaired its stimulatory effect on cell migration (c, n = 3 independent experiments) and invasion (d, n=3 independent experiments) in MIA PaCa-2 cells. The cells were respectively transfected with control p3×Flag or Flag-tagged wildtype PIWIL1, PIWIL1-C-boxmut, PIWIL1-N or PIWIL1-N-C-boxmut, and the assays were performed at 24 h post-transfection. eh, The co-activator-deficient C-box mutant MIWI effectively resorted cell proliferation (e, n=5 independent experiments) and anchorage-independent growth (f, n= 3 independent experiments), but failed to rescue cell migration (g, n=3 independent experiments) and invasion (h, n=3 independent experiments) in PIWIL1-depleted BxPC-3 cells. The cells were respectively co-transfected with Piwil1 siRNAs and control vector p3×Flag or Flag-tagged MIWI or MIWI-C-boxmut, and the assays were performed at 24 h post-transfection. The procedures are similar to that described in Extended Data Fig. 2. The mean ± SD was plotted in all graphs. Statistical analysis was performed using two-tailed Student t test. Statistical source data are provided in Statistical Source Data Extended Data Fig. 5.

Source data

Extended Data Fig. 6 Downregulation of IDH3B, ACTL6A or ARPC3 by PIWIL1 is not involved in its oncogenic function in PDAC cells.

a–d, Ectopic expression of IDH3B, ACTL6A or ARPC3 barely affected cell proliferation (a, n=4 independent experiments), anchorage-independent growth (b, n=3 independent experiments), cell migration (c, n=3 independent experiments) and invasion (d, n=3 independent experiments) in BxPC-3 cells. The cells were respectively transfected with Myc-tagged IDH3B, ACTL6A, ARPC3 or control vector pCMV-Myc, and the assays were performed at 24 h post-transfection. e–h, Co-transfection of Myc-tagged IDH3B, ACTL6A or ARPC3 vectors barely affected the stimulatory effect of PIWIL1 on cell proliferation (e, n=4 independent experiments), anchorage-independent growth (f, n=3: independent experiments), cell migration (c, n=3 independent experiments) and invasion (d, n=3 independent experiments) in PANC-1 cells. The cells were respectively co-transfected with Flag-PIWIL1 and Myc-tagged IDH3B, ACTL6A, or ARPC3, with pCMV-Myc and Myc-Pinin respectively serving as negative and positive controls, and the assays were performed at 24 h post-transfection. The procedures are similar to that described in Extended Data Fig. 2. The mean ± SD was plotted in all graphs. Statistical analysis was performed using two-tailed Student t test. Statistical source data are provided in Statistical Source Data Extended Data Fig. 6.

Source data

Extended Data Fig. 7 Pinin is specifically ubiquitinated by APC/CPIWIL1.

a, Cdh1 or Cdc20 knockdown marginally altered Pinin ubiquitination in BxPC-3 cells. Top, anti-Pinin IP pellets were immunoblotted with anti-Ub or anti-Pinin; bottom, cell lysates were immunoblotted for indicated proteins, with β-actin serving as a loading control. The cells were transfected with indicated siRNAs (lanes 1–4), with Apc10 siRNAs (lane 5) serving as positive control. b, The PIWIL1-N peptide, but not its C-box mutant, promoted Pinin ubiquitination by APC/C in vitro. Left, in vitro Pinin ubiquitination using PIWIL1-N peptides (lanes 6 and 7), with omission of one component or more (lanes 1–5) as negative controls; right, Coomassie blue staining of recombinant PIWIL1-N peptides. PIWIL1-N peptides and Pinin were bacterially expressed and purified. c, The D-box (red) and KEN-box (green) are conserved in vertebrate Pinin. d–g, Double mutations of D-Box and KEN-box resulted in Pinin resistant to ubiquitination by APC/CPIWIL1. (d), Schematic representation of the construction of D-Box or KEN-box mutant Pinin. (e), Pinin ubiquitination in PIWIL1 co-transfected-PANC-1 cells. Myc-Pinin or its mutants was respectively co-transfected with Flag-PIWIL1 and HA-Ub. Top, anti-Myc IP pellets were immunoblotted with anti-Ub or anti-Myc; bottom, cell lysates were immunoblotted for Flag-PIWIL1 or Myc-Pinin, with β-actin serving as a loading control. (f), Ubiquitination of Pinin or Pinin-DK mutant by APC/C in vitro. (g), The stability of Pinin proteins in PIWIL1-expressing PANC-1 cells. Myc-tagged Pinin (lanes 1–6) or Pinin-DK mutant (lanes 7–12) was co-transfected with Flag-PIWIL1. 48 h post-transfection, cells were treated with 50 μg/ml cycloheximide and harvested for immunoblotting of PIWIL1 or Pinin at indicated time points, with β-actin serving as a loading control. Results shown in a, b and eg are representative of three independent experiments. Unprocessed blots are provided in Source Data Extended Data Fig. 7.

Source data

Extended Data Fig. 8 Pinin inhibits cell migration and metastasis but not cell proliferation and tumor growth in PDAC cells.

Pinin knockdown in PANC-1 cells barely altered cell proliferation (a, n=3 independent experiments), anchorage-independent growth (b, n=3 independent experiments), and xenograft tumor growth in nude mice (c, n=3 animals), but promoted cell migration (d, n=3 independent experiments), invasion (e, n=3 independent experiments), and metastatic colonization in NOD/SCID mice through either orthotopic (f, n=3 animals), spleen (g, n=3 animals) or tail vein injection (h, n=3 animals). The cells were respectively transfected with Pinin siRNA (Pinin-siR1 or Pinin-siR2) or scrambled siRNA (scr siR) (a, b, d, and e), or shPinin or control pSilencer (c and fh), and the assays were performed at 24 h post-transfection. The procedures are similar to that described in Extended Data Fig. 2. The mean ± SD was plotted in all graphs. Statistical analysis was performed using two-tailed Student t test. Statistical source data are provided in Statistical Source Data Extended Data Fig. 8.

Source data

Extended Data Fig. 9 Pinin knockdown rescues the inhibitory effect of PIWIL1 depletion on PDAC metastasis but not tumor growth.

a–d, Co-transfection of Myc-Pinin in MIA PaCa-2 cells barely affected the stimulatory effect of PIWIL1 on cell proliferation (a, n=4 independent experiments) and anchorage-independent growth (b, n=3 independent experiments), but effectively attenuated the stimulatory effect of PIWIL1 on cell migration (c, n=3 independent experiments) and invasion (d, n=3 independent experiments). The cells were co-transfected with indicated vectors, and the assays were performed at 24 h post-transfection. eh, Pinin knockdown in BxPC-1 cells barely affected the inhibitory effect of PIWIL1 depletion on cell proliferation (e, n=6 independent experiments) and anchorage-independent growth (f, n=3 independent experiments), but effectively attenuated the inhibitory effect of PIWIL1 depletion on cell migration (g, n=3 independent experiments) and invasion (h, n=3 independent experiments) in cultured cells and metastatic colonization in the liver of NOD/SCID mice through either orthotopic or spleen (i, j, n=3 animals). The cells were co-transfected with indicated siRNAs (eh) or shRNAs (i and j), and the assays were performed at 24 h post-transfection. The procedures are similar to that described in Extended Data Fig. 2. k, Dispase-based dissociation assay showed the functional requirement of APC/CPIWIL1-Pinin axis in controlling intercellular cohesion in BxPC-3 cells. The cells were transfected with indicated siRNAs, and the assay was performed at 48 h post-transfection. Left, representative images; right, the average fragment number from 6 independent experiments, reporting as Tukey box-plots (middle lines, median values; lower and upper sides of the rectangles, the 1st and 3rd percentile, whiskers, confidence interval). The mean ± SD was plotted in all graphs. Statistical analysis was performed using two-tailed Student t test. Statistical source data are provided in Statistical Source Data Extended Data Fig. 9.

Source data

Extended Data Fig. 10 Clinical relevance of the APCPIWIL1-Pinin axis in PDAC patients.

a, Comparison of PIWIL1 staining intensity between early and late stages of acinar-to-ductal metaplasia (ADM). Representative images of Hematoxylin and eosin (H&E), α-Amylase, CK19 or PIWIL1 immunohistochemical staining (brown) of pancreas sections of chronic pancreatitis specimens without ADM (top), or with ADM at early (middle) or late stages (bottom), respectively. Magnification for pancreas sections, 40×; scale bars, 100 µm. b, Kaplan–Meier curves for overall survival analysis of the 30 patients recruited to our study with PIWIL1 (top) or Pinin expression (bottom), respectively. c, PIWIL1 and Pinin immunohistochemical (IHC) staining of serial sections from human primary PDAC tumor (middle panels), paired adjacent normal tissue (top panels) and liver metastatic nodule (bottom panels) from the 7 patients that showed liver metastasis after surgery. Scale bar, 100 μm. Bottom panels in each row showing an enlargement of black framed region on the top. d, Comparison of PIWIL1 (left) and Pinin IHC staining intensity (right) between primary PDAC tumor and paired liver metastatic nodule sections from the 7 patients. The positively stained cells were analyzed and the average values of staining intensity were calculated using Image-Pro Plus software, and then normalized with those in paired adjacent normal tissues. Results shown in ad are representative of three independent experiments. Statistical analysis was performed using two-tailed Student t test. Statistical source data are provided in Statistical Source Data Extended Data Fig. 10.

Source data

Supplementary information

Reporting Summary

Supplementary Tables

Table 1: The list of proteins upregulated in PIWIL1 siRNA-transfected BxPC-3 cells relative to Scr siR control. Table 2: Relevance of PIWIL1 and Pinin expression with clinicopathological features in PDAC. Table 3: The sequences of chemically synthesized DNA and RNA oligonucleotides. Table 4: The list of antibodies and reagents.

Source data

Source Data Fig. 1

Statistical source data

Source Data Fig. 1

Unprocessed gels

Source Data Fig. 2

Unprocessed western blots

Source Data Fig. 3

Unprocessed western blots

Source Data Fig. 4

Statistical source data

Source Data Fig. 4

Unprocessed western blots

Source Data Fig. 5

Statistical source data

Source Data Fig. 5

Unprocessed western blots

Source Data Fig. 6

Statistical source data

Source Data Fig. 6

Unprocessed western blots

Source Data Fig. 7

Statistical source data

Source Data Fig. 7

Unprocessed western blots

Source Data Fig. 8

Statistical source data

Source Data Extended Data Fig. 1

Statistical source data

Source Data Extended Data Fig. 1

Unprocessed western blots

Source Data Extended Data Fig. 2

Statistical source data

Source Data Extended Data Fig. 3

Statistical source data

Source Data Extended Data Fig. 4

Unprocessed western blots

Source Data Extended Data Fig. 5

Statistical source data

Source Data Extended Data Fig. 6

Statistical source data

Source Data Extended Data Fig. 7

Unprocessed western blots

Source Data Extended Data Fig. 8

Statistical source data

Source Data Extended Data Fig. 9

Statistical source data

Source Data Extended Data Fig. 10

Statistical source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, F., Yuan, P., Rao, M. et al. piRNA-independent function of PIWIL1 as a co-activator for anaphase promoting complex/cyclosome to drive pancreatic cancer metastasis. Nat Cell Biol 22, 425–438 (2020). https://doi.org/10.1038/s41556-020-0486-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-020-0486-z

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