An Argonaute phosphorylation cycle promotes microRNA-mediated silencing (original) (raw)

References

  1. Mendell, J. T. & Olson, E. N. MicroRNAs in stress signaling and human disease. Cell 148, 1172–1187 (2012)
    Article CAS PubMed PubMed Central Google Scholar
  2. Vidigal, J. A. & Ventura, A. The biological functions of miRNAs: lessons from in vivo studies. Trends Cell Biol . 25, 137–147 (2015)
    Article CAS PubMed Google Scholar
  3. Bartel, D. P. MicroRNAs: target recognition and regulatory functions. Cell 136, 215–233 (2009)
    Article CAS PubMed PubMed Central Google Scholar
  4. Jonas, S. & Izaurralde, E. Towards a molecular understanding of microRNA-mediated gene silencing. Nature Rev. Genet . 16, 421–433 (2015)
    Article CAS PubMed Google Scholar
  5. Grimson, A. et al. MicroRNA targeting specificity in mammals: determinants beyond seed pairing. Mol. Cell 27, 91–105 (2007)
    Article CAS PubMed PubMed Central Google Scholar
  6. Bosson, A. D., Zamudio, J. R. & Sharp, P. A. Endogenous miRNA and target concentrations determine susceptibility to potential ceRNA competition. Mol. Cell 56, 347–359 (2014)
    Article CAS PubMed PubMed Central Google Scholar
  7. Denzler, R., Agarwal, V., Stefano, J., Bartel, D. P. & Stoffel, M. Assessing the ceRNA hypothesis with quantitative measurements of miRNA and target abundance. Mol. Cell 54, 766–776 (2014)
    Article CAS PubMed PubMed Central Google Scholar
  8. Jo, M. H. et al. Human Argonaute 2 has diverse reaction pathways on target RNAs. Mol. Cell 59, 117–124 (2015)
    Article CAS PubMed Google Scholar
  9. Salomon, W. E., Jolly, S. M., Moore, M. J., Zamore, P. D. & Serebrov, V. Single-molecule imaging reveals that Argonaute reshapes the binding properties of its nucleic acid guides. Cell 162, 84–95 (2015)
    Article CAS PubMed PubMed Central Google Scholar
  10. Parry, D. H., Xu, J. & Ruvkun, G. A whole-genome RNAi screen for C. elegans miRNA pathway genes. Curr. Biol . 17, 2013–2022 (2007)
    Article CAS PubMed PubMed Central Google Scholar
  11. Zhou, R. et al. Comparative analysis of argonaute-dependent small RNA pathways in Drosophila. Mol. Cell 32, 592–599 (2008)
    Article CAS PubMed PubMed Central Google Scholar
  12. Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87 (2014)
    Article CAS PubMed ADS Google Scholar
  13. Wang, T., Wei, J. J., Sabatini, D. M. & Lander, E. S. Genetic screens in human cells using the CRISPR-Cas9 system. Science 343, 80–84 (2014)
    Article CAS PubMed ADS Google Scholar
  14. Jallepalli, P. V. et al. Securin is required for chromosomal stability in human cells. Cell 105, 445–457 (2001)
    Article CAS PubMed Google Scholar
  15. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nature Methods 11, 783–784 (2014)
    Article CAS PubMed PubMed Central Google Scholar
  16. Luo, B. et al. Highly parallel identification of essential genes in cancer cells. Proc. Natl Acad. Sci. USA 105, 20380–20385 (2008)
    Article CAS PubMed PubMed Central ADS Google Scholar
  17. Stefansson, B., Ohama, T., Daugherty, A. E. & Brautigan, D. L. Protein phosphatase 6 regulatory subunits composed of ankyrin repeat domains. Biochemistry 47, 1442–1451 (2008)
    Article CAS PubMed Google Scholar
  18. He, T. C. et al. Identification of c-MYC as a target of the APC pathway. Science 281, 1509–1512 (1998)
    Article CAS PubMed ADS Google Scholar
  19. Zuber, J. et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 478, 524–528 (2011)
    Article CAS PubMed PubMed Central ADS Google Scholar
  20. O’Donnell, K. A., Wentzel, E. A., Zeller, K. I., Dang, C. V. & Mendell, J. T. c-Myc-regulated microRNAs modulate E2F1 expression. Nature 435, 839–843 (2005)
    Article PubMed ADS CAS Google Scholar
  21. Ji, M. et al. The miR-17-92 microRNA cluster is regulated by multiple mechanisms in B-cell malignancies. Am. J. Pathol . 179, 1645–1656 (2011)
    Article CAS PubMed PubMed Central Google Scholar
  22. Lee, Y. S. & Dutta, A. The tumor suppressor microRNA let-7 represses the HMGA2 oncogene. Genes Dev . 21, 1025–1030 (2007)
    Article CAS PubMed PubMed Central Google Scholar
  23. Mayr, C., Hemann, M. T. & Bartel, D. P. Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation. Science 315, 1576–1579 (2007)
    Article CAS PubMed PubMed Central ADS Google Scholar
  24. Tokumaru, S., Suzuki, M., Yamada, H., Nagino, M. & Takahashi, T. let-7 regulates Dicer expression and constitutes a negative feedback loop. Carcinogenesis 29, 2073–2077 (2008)
    Article CAS PubMed Google Scholar
  25. Kinoshita, E., Kinoshita-Kikuta, E., Takiyama, K. & Koike, T. Phosphate-binding tag, a new tool to visualize phosphorylated proteins. Mol. Cell. Proteomics 5, 749–757 (2006)
    Article CAS PubMed Google Scholar
  26. Zeng, Y., Sankala, H., Zhang, X. & Graves, P. R. Phosphorylation of Argonaute 2 at serine-387 facilitates its localization to processing bodies. Biochem. J . 413, 429–436 (2008)
    Article CAS PubMed Google Scholar
  27. Schirle, N. T. & MacRae, I. J. The crystal structure of human Argonaute2. Science 336, 1037–1040 (2012)
    Article CAS PubMed PubMed Central ADS Google Scholar
  28. Elkayam, E. et al. The structure of human argonaute-2 in complex with miR-20a. Cell 150, 100–110 (2012)
    Article CAS PubMed PubMed Central Google Scholar
  29. Liu, Q. et al. miR-16 family induces cell cycle arrest by regulating multiple cell cycle genes. Nucleic Acids Res . 36, 5391–5404 (2008)
    Article CAS PubMed PubMed Central Google Scholar
  30. Flores-Jasso, C. F., Salomon, W. E. & Zamore, P. D. Rapid and specific purification of Argonaute-small RNA complexes from crude cell lysates. RNA 19, 271–279 (2013)
    Article CAS PubMed PubMed Central Google Scholar
  31. Cummins, J. M. et al. The colorectal microRNAome. Proc. Natl Acad. Sci. USA 103, 3687–3692 (2006)
    Article CAS PubMed PubMed Central ADS Google Scholar
  32. Pillai, R. S., Artus, C. G. & Filipowicz, W. Tethering of human Ago proteins to mRNA mimics the miRNA-mediated repression of protein synthesis. RNA 10, 1518–1525 (2004)
    Article CAS PubMed PubMed Central Google Scholar
  33. Knippschild, U. et al. The casein kinase 1 family: participation in multiple cellular processes in eukaryotes. Cell. Signal . 17, 675–689 (2005)
    Article CAS PubMed Google Scholar
  34. Wang, C. C., Tao, M., Wei, T. & Low, P. S. Identification of the major casein kinase I phosphorylation sites on erythrocyte band 3. Blood 89, 3019–3024 (1997)
    Article CAS PubMed Google Scholar
  35. Schirle, N. T., Sheu-Gruttadauria, J. & MacRae, I. J. Structural basis for microRNA targeting. Science 346, 608–613 (2014)
    Article CAS PubMed PubMed Central ADS Google Scholar
  36. Hansen, T. B. et al. Natural RNA circles function as efficient microRNA sponges. Nature 495, 384–388 (2013)
    Article CAS PubMed ADS Google Scholar
  37. Van Nostrand, E. L. et al. Robust transcriptome-wide discovery of RNA-binding protein binding sites with enhanced CLIP (eCLIP). Nature Methods 13, 508–514 (2016)
    Article CAS PubMed PubMed Central Google Scholar
  38. Tani, H. et al. Genome-wide determination of RNA stability reveals hundreds of short-lived noncoding transcripts in mammals. Genome Res . 22, 947–956 (2012)
    Article CAS PubMed PubMed Central Google Scholar
  39. Kluiver, J. et al. Rapid generation of microRNA sponges for microRNA inhibition. PLoS ONE 7, e29275 (2012)
    Article CAS PubMed PubMed Central ADS Google Scholar
  40. Largaespada, D. A. & Collier, L. S. Transposon-mediated mutagenesis in somatic cells: identification of transposon-genomic DNA junctions. Methods Mol. Biol . 435, 95–108 (2008)
    Article CAS PubMed PubMed Central Google Scholar
  41. Wessel, D. & Flügge, U. I. A method for the quantitative recovery of protein in dilute solution in the presence of detergents and lipids. Anal. Biochem . 138, 141–143 (1984)
    Article CAS PubMed Google Scholar
  42. Rüdel, S., Flatley, A., Weinmann, L., Kremmer, E. & Meister, G. A multifunctional human Argonaute2-specific monoclonal antibody. RNA 14, 1244–1253 (2008)
    Article PubMed PubMed Central CAS Google Scholar
  43. Patel, R. K. & Jain, M. NGS QC Toolkit: a toolkit for quality control of next generation sequencing data. PLoS ONE 7, e30619 (2012)
    Article CAS PubMed PubMed Central ADS Google Scholar
  44. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol . 14, R36 (2013)
    Article PubMed PubMed Central CAS Google Scholar
  45. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014)
    Article CAS PubMed Google Scholar
  46. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010)
    Article CAS PubMed Google Scholar
  47. Marcel, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.journal 17, 10–12 (2011)
    Google Scholar
  48. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010)
    Article CAS PubMed PubMed Central Google Scholar
  49. Kim, Y. K., Kim, B. & Kim, V. N. Re-evaluation of the roles of DROSHA, Export in 5, and DICER in microRNA biogenesis. Proc. Natl Acad. Sci. USA 113, E1881–E1889 (2016)
    Article CAS PubMed PubMed Central Google Scholar
  50. Jiang, H. & Wong, W. H. SeqMap: mapping massive amount of oligonucleotides to the genome. Bioinformatics 24, 2395–2396 (2008)
    Article CAS PubMed PubMed Central Google Scholar

Download references

Acknowledgements

We thank D. Bartel, C. Cepko, D. Sabatini, P. Sharp, D. Trono, T. Tuschl, and F. Zhang for plasmids; A. Guzman and R. Bruce in the McDermott Center Next Generation Sequencing Core; A. Mobley and the University of Texas Southwestern Flow Cytometry Core; H. Ball and the University of Texas Southwestern Protein Chemistry Technology Core; S. Johnson for assistance with software implementation; J. Cabrera for assistance with figure preparation; and K. O’Donnell for advice on the manuscript. This work was supported by grants from the Cancer Prevention and Research Institute of Texas (CPRIT) (R1008 and RP160249 to J.T.M., RP101251 to Y.X., RP120718 to Z.J.C., and RR150033 to V.S.T.) and the National Institutes of Health (R01CA120185 and R35CA197311 to J.T.M., R01CA152301 to Y.X., and R00DK099254 to V.S.T.). T.L. is supported by a fellowship from Cancer Research Institute. F.K. is supported by the Leopoldina Fellowship Program (LPDS 2014-12) from the German National Academy of Sciences Leopoldina. J.T.M. and V.S.T. are CPRIT Scholars in Cancer Research. J.T.M. and Z.J.C. are Investigators of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

  1. Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, 75390, Texas, USA
    Ryan J. Golden, Tuo Li, Juliane Braun, Hema Manjunath, Xiang Chen, Tsung-Cheng Chang, Florian Kopp, Andres Ramirez-Martinez, Vincent S. Tagliabracci, Zhijian J. Chen & Joshua T. Mendell
  2. Medical Scientist Training Program, University of Texas Southwestern Medical Center, Dallas, 75390, Texas, USA
    Ryan J. Golden
  3. Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, 75390, Texas, USA
    Beibei Chen & Yang Xie
  4. Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, 75390, Texas, USA
    Beibei Chen & Yang Xie
  5. Department of Microbiology and Immunology, University of California San Francisco, San Francisco, 94143, California, USA
    Jiaxi Wu
  6. Eugene McDermott Center for Human Growth & Development, University of Texas Southwestern Medical Center, Dallas, 75390, Texas, USA
    Vanessa Schmid
  7. Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, 75390, Texas, USA
    Zhijian J. Chen & Joshua T. Mendell
  8. Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, 75390, Texas, USA
    Yang Xie & Joshua T. Mendell
  9. Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, 75390, Texas, USA
    Joshua T. Mendell

Authors

  1. Ryan J. Golden
    You can also search for this author inPubMed Google Scholar
  2. Beibei Chen
    You can also search for this author inPubMed Google Scholar
  3. Tuo Li
    You can also search for this author inPubMed Google Scholar
  4. Juliane Braun
    You can also search for this author inPubMed Google Scholar
  5. Hema Manjunath
    You can also search for this author inPubMed Google Scholar
  6. Xiang Chen
    You can also search for this author inPubMed Google Scholar
  7. Jiaxi Wu
    You can also search for this author inPubMed Google Scholar
  8. Vanessa Schmid
    You can also search for this author inPubMed Google Scholar
  9. Tsung-Cheng Chang
    You can also search for this author inPubMed Google Scholar
  10. Florian Kopp
    You can also search for this author inPubMed Google Scholar
  11. Andres Ramirez-Martinez
    You can also search for this author inPubMed Google Scholar
  12. Vincent S. Tagliabracci
    You can also search for this author inPubMed Google Scholar
  13. Zhijian J. Chen
    You can also search for this author inPubMed Google Scholar
  14. Yang Xie
    You can also search for this author inPubMed Google Scholar
  15. Joshua T. Mendell
    You can also search for this author inPubMed Google Scholar

Contributions

R.J.G. performed most experiments and B.C. performed most bioinformatics analyses. T.L., X.C., J.W., and R.J.G. performed mass spectrometry analyses. J.B. and H.M. generated plasmid constructs, cell lines, and performed CLIP validation experiments. V.S. provided technical assistance for sequencing sgRNA libraries. T.-C.C. performed qPCR analyses. F.K. assisted with CLIP experiments. A.R.-M. generated plasmid constructs. B.C., Y.X., and J.T.M. performed bioinformatics analyses. V.S.T. provided guidance for the in vitro kinase assays. R.J.G., J.T.M., B.C., Y.X., Z.J.C., and T.L. designed most experiments. R.J.G and J.T.M. wrote the manuscript.

Corresponding author

Correspondence toJoshua T. Mendell.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks S. Tavazoie, W. Wei and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Figure 1 Knockout of candidate miRNA regulators in HCT116_EGFP_ cells.

Flow cytometry analysis of EGFP in HCT116_EGFP_ cells after transduction with lentiCRISPR vectors targeting the indicated genes.

Extended Data Figure 2 BRD4, CTNNB1, and POU2F1 positively regulate miR-19 expression.

a, Model depicting how each gene may promote expression of the miR-17-92 cluster. bd, Western blot analysis confirming loss of expression of the indicated gene in HCT116 knockout clones. Asterisk indicates non-specific band. For each protein, all lanes came from the same blot but irrelevant lanes were removed. eg, qRT–PCR assays demonstrating reduced expression of MYC (e), pri-miR-17-92 (f), or mature miR-19a/b (g) in _BRD4_−/−, _CTNNB1_−/−, or _POU2F1_−/− cells. For gel source data, see Supplementary Fig. 1.

Extended Data Figure 4 General impairment of miRNA-mediated silencing in _ANKRD52_–/– cells.

a, b, Flow cytometry analysis of EGFP expression in HCT116 cells stably expressing reporters for miR-16 (a) or miR-200 (b) after transduction with lentiCRISPR vectors targeting ANKRD52 or expressing a non-targeting sgRNA. c, qRT–PCR showing de-repression of established let-7 targets (DICER1 or HMGA2) in _AGO2_–/– or _ANKRD52_–/– cells. *P < 0.05, **P < 0.01, two-tailed Student’s _t_-test comparing _AGO2_–/– or ANKRD52_–/– to parental. (n = 3 biological replicates, each assayed in triplicate.) d, qRT–PCR analysis of DICER1 and HMGA2 in non-transfected (NT) HCT116_EGFP-miR-19 cells or after transfection with miR-19 antisense oligonucleotides (Anti-miR-19) confirms that these transcripts are not regulated by miR-19. Upregulation of the EGFP miR-19 reporter transcript served as a positive control in this experiment. (n = 3 biological replicates, each assayed in triplicate.) e, qRT–PCR was performed for the indicated miRNAs and expression levels were normalized to U6 snRNA (n = 2 biological replicates, each assayed in triplicate).

Extended Data Figure 5 The ANKRD52–PPP6C complex interacts with and dephosphorylates AGO proteins.

a, Co-immunoprecipitation of Flag–HA-AGO2 (FH-AGO2) with V5-ANKRD52 or V5-PPP6C with or without RNase A treatment. b, Phos-tag electrophoresis demonstrating AGO2 hyperphosphorylation in multiple ANKRD52/PPP6C-deficient cell lines. c, Phos-tag western blot analysis of Flag–HA-AGO1 (FH-AGO1) stably expressed in ANKRD52+/+ and _ANKRD52_−/− HCT116 cells. For gel source data, see Supplementary Fig. 1.

Extended Data Figure 6 Identification of multiple definitively phosphorylated residues in the S824–S834 region of AGO2 by mass spectrometry.

a, Full-scan mass spectra zoomed to the region for the AGO2 815–837 peptide. The unphosphorylated and multiply phosphosphorylated precursor ions are shown in red. Peak labels indicate the mass-to-charge ratios and the charge state. The singly charged ion with grey label (top panel) does not correspond to peptides 815–837. Data at two close elution time points are shown for ANKRD52−/− to illustrate the unphosphorylated (0P), singly (1P), doubly (2P), and triply (3P) phosphorylated peptides. b, Quantification of the indicated endogenous AGO2 phosphopeptides relative to unphosphorylated peptide as determined by mass spectrometry. Labels 1P, 2P, or 3P respectively denote singly, doubly, or triply phosphorylated peptides spanning residues 815–837 of AGO2. Superscript indicates peptide charge state. ND, not detected. c, MS/MS spectra demonstrating phosphorylation of endogenous AGO2 at S824 in _ANKRD52_−/− cells. Red bars denote site-determining ions. d, e, MS/MS spectra demonstrating phosphorylation of FH-AGO2 (T830A) at S824 and S828 (d) or phosphorylation of FH-AGO2 (S824A/T830A) at S828 and S831(e) in _ANKRD52_−/− cells.

Extended Data Figure 7 Phosphomimetic mutants of FH-AGO2 do not exhibit reduced miRNA association.

a, Western blots showing expression of the indicated FH-AGO2 mutants. Within each panel (top, middle, bottom), all lanes came from the same blot but irrelevant lanes were removed. b, miRNA association of wild-type or mutant FH-AGO2 assessed as described in Fig. 3a (n = 4 biological replicates, each assayed in triplicate). For gel source data, see Supplementary Fig. 1.

Extended Data Figure 8 Analysis of serine/threonine kinases identified in the CRISPR–Cas9 suppressor screen.

a, b, Flow cytometry demonstrating EGFP expression in HCT116_EGFP-miR19_ (a) or HCT116_EGFP_ cells (b) after transduction with lentiCRISPR vectors targeting the indicated genes. c, Flow cytometry demonstrating EGFP expression in HCT116_EGFP-miR19_ cells treated with the indicated dose of rapamycin. NT, not treated. d, Phos-tag western blot analysis of AGO2 in ANKRD52−/− cells after treatment with rapamycin. For gel source data, see Supplementary Fig. 1.

Extended Data Figure 9 Functional characterization of CSNK1A1 and AGO2 target binding mutants.

a, Western blot analysis confirms loss of CSNK1A1 expression in HCT116 ANKRD52_−/−;CSNK1A1_−/− clonal knockout cells. All lanes came from the same blot but irrelevant lanes were removed. b, miR-19 expression normalized to U6 expression, assessed by qRT–PCR, in cells of the indicated genotypes (n = 4 biological replicates, each assayed in triplicate). c, Co-immunoprecipitation of V5-CSNK1A1 with FH-AGO2, with or without RNase A treatment. d, miRNA association of FH-AGO2 assessed as in Fig. 3e (n = 4 biological replicates, each assayed in triplicate). *P < 0.05 comparing mutant to wild-type AGO2, two-tailed Student’s _t_-test. For gel source data, see Supplementary Fig. 1.

Extended Data Figure 10 Generation and eCLIP analysis of _AGO2_–/– cells reconstituted with AGO2WT or AGO25XA.

a, Western blot showing equivalent expression of FH-AGO2WT and FH-AGO25XA at physiological levels. For gel source data, see Supplementary Fig. 1. b, Distribution of AGO2 binding sites determined by eCLIP. c, Validation of targets identified by eCLIP using FH-AGO2 pull-down assays performed in reconstituted _AGO2_−/− cells. Experiment was performed as in Fig. 3a except anti-Flag antibody was used for immunoprecipitation (n = 3 biological replicates, each assayed in triplicate). *P < 0.05, **P < 0.01, one-tailed Student’s _t_-test comparing FH-AGO25XA with FH-AGO2WT. NS, not significant. d, FH-AGO2WT CLIP coverage (normalized total number of reads in clusters in a given 3′ UTR divided by the FPKM) of genes whose AGO2-mediated repression is or is not rescued by FH-AGO25XA. e, The 8-, 7-, or 6-nucleotide binding sites for active miRNAs in HCT116 were identified within FH-AGO2WT/FH-AGO25XA-common CLIP clusters or FH-AGO25XA-unique CLIP clusters in 3′ UTRs. CDF plots show CLIP coverage for each class of site (normalized number of crosslinking events within ten nucleotides of each site). NS, not significant, assessed by Kolmogorov–Smirnov test. f, CDF plot showing the fold change in CLIP coverage comparing FH-AGO25XA to FH-AGO2WT for transcripts with long half-lives (top quartile) versus those with short half-lives (bottom quartile). g, Summary of the newly defined AGO2 phosphorylation cycle. Target engagement triggers the hierarchical, multi-site phosphorylation of AGO2 by CSNK1A1, which inhibits target binding. The ANKRD52–PPP6C phosphatase complex dephosphorylates these residues, allowing AGO2 to engage new targets. Continual phosphorylation/de-phosphorylation of AGO2 through this cycle is necessary to maintain the global efficiency of miRNA-mediated silencing.

Supplementary information

Supplementary Figure 1

This file contains the gel source data. (PDF 1155 kb)

Supplementary Table 1.

This file contains the simulated sgRNA enrichment in top 0.5% brightest cells. (XLSX 9 kb)

Supplementary Table 2.

This file contains the RIGER analysis of CRISPR-Cas9 screen in HCT116_EGFP-miR19_ cells. (XLSX 2290 kb)

Supplementary Table 3.

This file contains the RIGER analysis of CRISPR-Cas9 screen in HCT116_EGFP_ cells. (XLSX 2291 kb)

Supplementary Table 4.

This file contains the RNA-seq results showing genes with altered expression in AGO2 −/−, ANKRD52 −/−, and CSNK1A1 −/−; ANKRD52 −/− HCT116 cells compared to wild-type HCT116 cells. (XLSX 160 kb)

Supplementary Table 5.

This file contains the RIGER analysis of CRISPR-Cas9 suppressor screen in ANKRD52 −/− HCT116_EGFP-miR19_ cells. (XLSX 2096 kb)

Supplementary Table 6.

This file contains the RNA-seq results showing genes with altered expression in _AGO2_−/− cells and _AGO2_−/− cells reconstituted with FH-AGO2WT or FH-AGO25XA compared to wild-type HCT116 cells. (XLSX 886 kb)

Supplementary Table 7.

This file contains the eCLIP results. (XLSX 271 kb)

Supplementary Table 8.

This file contains the oligonucleotides and peptides used in this study. (XLSX 18 kb)

PowerPoint slides

Rights and permissions

About this article

Cite this article

Golden, R., Chen, B., Li, T. et al. An Argonaute phosphorylation cycle promotes microRNA-mediated silencing.Nature 542, 197–202 (2017). https://doi.org/10.1038/nature21025

Download citation