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.

  • Resource
  • Published:

Large-scale tethered function assays identify factors that regulate mRNA stability and translation

Abstract

The molecular functions of the majority of RNA-binding proteins (RBPs) remain unclear, highlighting a major bottleneck to a full understanding of gene expression regulation. Here, we develop a plasmid resource of 690 human RBPs that we subject to luciferase-based 3ʹ-untranslated-region tethered function assays to pinpoint RBPs that regulate RNA stability or translation. Enhanced UV-cross-linking and immunoprecipitation of these RBPs identifies thousands of endogenous mRNA targets that respond to changes in RBP level, recapitulating effects observed in tethered function assays. Among these RBPs, the ubiquitin-associated protein 2-like (UBAP2L) protein interacts with RNA via its RGG domain and cross-links to mRNA and rRNA. Fusion of UBAP2L to RNA-targeting CRISPR–Cas9 demonstrates programmable translational enhancement. Polysome profiling indicates that UBAP2L promotes translation of target mRNAs, particularly global regulators of translation. Our tethering survey allows rapid assignment of the molecular activity of proteins, such as UBAP2L, to specific steps of mRNA metabolism.

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: A large-scale tethered function screen identifies RBPs regulating stability and translation.
Fig. 2: eCLIP identifies endogenous RNA targets of candidate stabilizers and destabilizers.
Fig. 3: Integration of eCLIP and RNA-seq data defines regulatory classes of RBPs and transcripts.
Fig. 4: UBAP2L is associated with translating ribosomes and promotes translation.
Fig. 5: UBAP2L binds directly to the ribosome.

Similar content being viewed by others

Data availability

Sequencing data are available at NCBI GEO (accession number GSE117294). Source data are provided with this paper.

Code availability

All code described in the Methods is publicly available and can be found at https://github.com/YeoLab/.

References

  1. Lukong, K. E., Chang, K. W., Khandjian, E. W. & Richard, S. RNA-binding proteins in human genetic disease. Trends Genet. 24, 416–425 (2008).

    Article  CAS  PubMed  Google Scholar 

  2. Baltz, A. G. et al. The mRNA-bound proteome and its global occupancy profile on protein-coding transcripts. Mol. Cell 46, 674–690 (2012).

    Article  CAS  PubMed  Google Scholar 

  3. Castello, A. et al. Insights into RNA biology from an atlas of mammalian mRNA-binding proteins. Cell 149, 1393–1406 (2012).

    Article  CAS  PubMed  Google Scholar 

  4. Gerstberger, S., Hafner, M., Ascano, M. & Tuschl, T. Evolutionary conservation and expression of human RNA-binding proteins and their role in human genetic disease. Adv. Exp. Med. Biol. 825, 1–55 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gerstberger, S., Hafner, M. & Tuschl, T. A census of human RNA-binding proteins. Nat. Rev. Genet. 15, 829–845 (2014).

    Article  CAS  PubMed  Google Scholar 

  6. Trendel, J. et al. The human RNA-binding proteome and its dynamics during translational arrest. Cell 176, 391–403 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. Queiroz, R. M. L. et al. Comprehensive identification of RNA–protein interactions in any organism using orthogonal organic phase separation (OOPS). Nat. Biotechnol. 37, 169–178 (2019); erratum 37, 692 (2019).

  8. Urdaneta, E. C. et al. Purification of cross-linked RNA–protein complexes by phenol-toluol extraction. Nat. Commun. 10, 990 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Graindorge, A. et al. In-cell identification and measurement of RNA–protein interactions. Nat. Commun. 10, 5317 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Beckmann, B. M. et al. The RNA-binding proteomes from yeast to man harbour conserved enigmRBPs. Nat. Commun. 6, 10127 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Van Nostrand, E. L. et al. Robust transcriptome-wide discovery of RNA-binding protein binding sites with enhanced CLIP (eCLIP). Nat. Methods 13, 508–514 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Wheeler, E. C., Van Nostrand, E. L. & Yeo, G. W. Advances and challenges in the detection of transcriptome-wide protein–RNA interactions. Wiley Interdiscip. Rev. RNA 9, e1436 (2018).

  13. Licatalosi, D. D. et al. HITS-CLIP yields genome-wide insights into brain alternative RNA processing. Nature 456, 464–469 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hafner, M. et al. Transcriptome-wide identification of RNA-binding protein and microRNA target sites by PAR-CLIP. Cell 141, 129–141 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Konig, J. et al. iCLIP reveals the function of hnRNP particles in splicing at individual nucleotide resolution. Nat. Struct. Mol. Biol. 17, 909–915 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Kapeli, K. et al. Distinct and shared functions of ALS-associated proteins TDP-43, FUS and TAF15 revealed by multisystem analyses. Nat. Commun. 7, 12143 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Martinez, F. J. et al. Protein–RNA networks regulated by normal and ALS-associated mutant HNRNPA2B1 in the nervous system. Neuron 92, 780–795 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Van Nostrand, E. L., Huelga, S. C. & Yeo, G. W. Experimental and computational considerations in the study of RNA-binding protein–RNA interactions. Adv. Exp. Med. Biol. 907, 1–28 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Van Nostrand, E. L. et al. A large-scale binding and functional map of human RNA binding proteins. Nature https://doi.org/10.1038/s41586-020-2077-3 (2020).

  20. Ule, J. et al. CLIP identifies Nova-regulated RNA networks in the brain. Science 302, 1212–1215 (2003).

    Article  CAS  PubMed  Google Scholar 

  21. Ule, J. et al. An RNA map predicting Nova-dependent splicing regulation. Nature 444, 580–586 (2006).

    Article  CAS  PubMed  Google Scholar 

  22. Coller, J. M., Gray, N. K. & Wickens, M. P. mRNA stabilization by poly(A) binding protein is independent of poly(A) and requires translation. Genes Dev. 12, 3226–3235 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Coller, J. & Wickens, M. Tethered function assays: an adaptable approach to study RNA regulatory proteins. Methods Enzymol. 429, 299–321 (2007).

    Article  CAS  PubMed  Google Scholar 

  24. Bos, T. J., Nussbacher, J. K., Aigner, S. & Yeo, G. W. Tethered function assays as tools to elucidate the molecular roles of RNA-binding proteins. Adv. Exp. Med. Biol. 907, 61–88 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Batra, R. et al. Elimination of toxic microsatellite repeat expansion RNA by RNA-targeting Cas9. Cell 170, 899–912 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nelles, D. A. et al. Programmable RNA tracking in live cells with CRISPR/Cas9. Cell 165, 488–496 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. O’Connell, M. R. et al. Programmable RNA recognition and cleavage by CRISPR/Cas9. Nature 516, 263–266 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Punta, M. et al. The Pfam protein families database. Nucleic Acids Res. 40, D290–D301 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Attwood, T. K. et al. PRINTS and its automatic supplement, prePRINTS. Nucleic Acids Res. 31, 400–402 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lykke-Andersen, J. & Wagner, E. Recruitment and activation of mRNA decay enzymes by two ARE-mediated decay activation domains in the proteins TTP and BRF-1. Genes Dev. 19, 351–361 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Benjamini, Y., Krieger, A. M. & Yekutieli, D. Adaptive linear step-up procedures that control the false discovery rate. Biometrika 93, 491–507 (2006).

    Article  Google Scholar 

  32. Nishimura, T. et al. The eIF4E-binding protein 4E-T is a component of the mRNA decay machinery that bridges the 5ʹ and 3ʹ termini of target mRNAs. Cell Rep. 11, 1425–1436 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Wang, X. et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature 505, 117–120 (2014).

    Article  PubMed  CAS  Google Scholar 

  34. Garneau, N. L., Wilusz, J. & Wilusz, C. J. The highways and byways of mRNA decay. Nat. Rev. Mol. Cell Biol. 8, 113–126 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Hu, W., Yuan, B. & Lodish, H. F. Cpeb4-mediated translational regulatory circuitry controls terminal erythroid differentiation. Dev. Cell 30, 660–672 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Vicens, Q., Kieft, J. S. & Rissland, O. S. Revisiting the closed-loop model and the nature of mRNA 5ʹ–3ʹ communication. Mol. Cell 72, 805–812 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Rissland, O. S. The organization and regulation of mRNA–protein complexes. Wiley Interdiscip. Rev. RNA 8, e1369 (2017).

  38. Bicknell, A. A. & Ricci, E. P. When mRNA translation meets decay. Biochem. Soc. Trans. 45, 339–351 (2017).

    Article  CAS  PubMed  Google Scholar 

  39. Radhakrishnan, A. & Green, R. Connections underlying translation and mRNA stability. J. Mol. Biol. 428, 3558–3564 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Roy, B. & Jacobson, A. The intimate relationships of mRNA decay and translation. Trends Genet. 29, 691–699 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Fu, X. F. et al. DAZ family proteins, key players for germ cell development. Int. J. Biol. Sci. 11, 1226–1235 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Rosario, R., Childs, A. J. & Anderson, R. A. RNA-binding proteins in human oogenesis: balancing differentiation and self-renewal in the female fetal germline. Stem Cell Res. 21, 193–201 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Julaton, V. T. & Reijo Pera, R. A. NANOS3 function in human germ cell development. Hum. Mol. Genet. 20, 2238–2250 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Cirillo, L. et al. UBAP2L forms distinct cores that act in nucleating stress granules upstream of G3BP1. Curr. Biol. 30, 698–707 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Markmiller, S. et al. Context-dependent and disease-specific diversity in protein interactions within stress granules. Cell 172, 590–604 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Youn, J. Y. et al. High-density proximity mapping reveals the subcellular organization of mRNA-associated granules and bodies. Mol. Cell 69, 517–532.e11 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Deragon, J. M. & Bousquet-Antonelli, C. The role of LARP1 in translation and beyond. Wiley Interdiscip. Rev. RNA 6, 399–417 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Gray, G. A. & Gray, N. K. A tail of translational regulation. Elife 6, e29104 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Andreev, D. E. et al. Translation control of mRNAs encoding mammalian translation initiation factors. Gene 651, 174–182 (2018).

    Article  CAS  PubMed  Google Scholar 

  50. Hortsch, M., Griffiths, G. & Meyer, D. I. Restriction of docking protein to the rough endoplasmic reticulum: immunocytochemical localization in rat liver. Eur. J. Cell Biol. 38, 271–279 (1985).

    CAS  PubMed  Google Scholar 

  51. Gehman, L. T. et al. The splicing regulator Rbfox1 (A2BP1) controls neuronal excitation in the mammalian brain. Nat. Genet. 43, 706–711 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Duncan, P. I., Stojdl, D. F., Marius, R. M., Scheit, K. H. & Bell, J. C. The Clk2 and Clk3 dual-specificity protein kinases regulate the intranuclear distribution of SR proteins and influence pre-mRNA splicing. Exp. Cell Res. 241, 300–308 (1998).

    Article  CAS  PubMed  Google Scholar 

  53. Scotti, M. M. & Swanson, M. S. RNA mis-splicing in disease. Nat. Rev. Genet. 17, 19–32 (2016).

    Article  CAS  PubMed  Google Scholar 

  54. Chen, C. Y., Xu, N., Zhu, W. & Shyu, A. B. Functional dissection of hnRNP D suggests that nuclear import is required before hnRNP D can modulate mRNA turnover in the cytoplasm. RNA 10, 669–680 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Luna, R., Rondon, A. G. & Aguilera, A. New clues to understand the role of THO and other functionally related factors in mRNP biogenesis. Biochim. Biophys. Acta 1819, 514–520 (2012).

    Article  CAS  PubMed  Google Scholar 

  56. Sundararaman, B. et al. Resources for the comprehensive discovery of functional RNA elements. Mol. Cell 61, 903–913 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Miyasaka, T. et al. Interaction of antiproliferative protein Tob with the CCR4–NOT deadenylase complex. Cancer Sci. 99, 755–761 (2008).

    Article  CAS  PubMed  Google Scholar 

  58. Lovci, M. T. et al. Rbfox proteins regulate alternative mRNA splicing through evolutionarily conserved RNA bridges. Nat. Struct. Mol. Biol. 20, 1434–1442 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Lee, D. S. M., Ghanem, L. R. & Barash, Y. Integrative analysis reveals RNA G-quadruplexes in UTRs are selectively constrained and enriched for functional associations. Nat. Commun. 11, 527 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Urano, J., Fox, M. S. & Reijo Pera, R. A. Interaction of the conserved meiotic regulators, BOULE (BOL) and PUMILIO-2 (PUM2). Mol. Reprod. Dev. 71, 290–298 (2005).

    Article  CAS  PubMed  Google Scholar 

  61. Moore, F. L. et al. Human Pumilio-2 is expressed in embryonic stem cells and germ cells and interacts with DAZ (Deleted in AZoospermia) and DAZ-like proteins. Proc. Natl Acad. Sci. USA 100, 538–543 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Schmidt, E. K., Clavarino, G., Ceppi, M. & Pierre, P. SUnSET, a nonradioactive method to monitor protein synthesis. Nat. Methods 6, 275–277 (2009).

    Article  CAS  PubMed  Google Scholar 

  63. Liu, B. & Qian, S. B. Characterizing inactive ribosomes in translational profiling. Translation 4, e1138018 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Fujii, K., Susanto, T. T., Saurabh, S. & Barna, M. Decoding the function of expansion segments in ribosomes. Mol. Cell 72, 1013–1020 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Maeda, M. et al. Arginine methylation of ubiquitin-associated protein 2-like is required for the accurate distribution of chromosomes. FASEB J. 30, 312–323 (2016).

    Article  CAS  PubMed  Google Scholar 

  66. Natchiar, S. K., Myasnikov, A. G., Kratzat, H., Hazemann, I. & Klaholz, B. P. Visualization of chemical modifications in the human 80S ribosome structure. Nature 551, 472–477 (2017).

    Article  CAS  PubMed  Google Scholar 

  67. Lutz, C. S., Cooke, C., O’Connor, J. P., Kobayashi, R. & Alwine, J. C. The snRNP-free U1A (SF-A) complex(es): identification of the largest subunit as PSF, the polypyrimidine-tract binding protein-associated splicing factor. RNA 4, 1493–1499 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Boelens, W. C. et al. The human U1 snRNP-specific U1A protein inhibits polyadenylation of its own pre-mRNA. Cell 72, 881–892 (1993).

    Article  CAS  PubMed  Google Scholar 

  69. Fensterl, V. & Sen, G. C. Interferon-induced Ifit proteins: their role in viral pathogenesis. J. Virol. 89, 2462–2468 (2015).

    Article  PubMed  CAS  Google Scholar 

  70. Liang, D., Halpert, M. M., Konduri, V. & Decker, W. K. Stepping out of the cytosol: AIMp1/p43 potentiates the link between innate and adaptive immunity. Int. Rev. Immunol. 34, 367–381 (2015).

    Article  CAS  PubMed  Google Scholar 

  71. Protter, D. S. W. & Parker, R. Principles and properties of stress granules. Trends Cell Biol. 26, 668–679 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Hanson, G., Alhusaini, N., Morris, N., Sweet, T. & Coller, J. Translation elongation and mRNA stability are coupled through the ribosomal A-site. RNA 24, 1377–1389 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Fischer, J. W., Busa, V. F., Shao, Y. & Leung, A. K. L. Structure-mediated RNA decay by UPF1 and G3BP1. Mol. Cell 78, 70–84 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Cox, D. B. T. et al. RNA editing with CRISPR–Cas13. Science 358, 1019–1027 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Brannan, K. W. et al. SONAR discovers RNA-binding proteins from analysis of large-scale protein–protein interactomes. Mol. Cell 64, 282–293 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Castello, A. et al. Comprehensive identification of RNA-binding domains in human cells. Mol. Cell 63, 696–710 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Rual, J. F. et al. Human ORFeome version 1.1: a platform for reverse proteomics. Genome Res. 14, 2128–2135 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. The Gene Ontology Consortium. Expansion of the Gene Ontology knowledgebase and resources. Nucleic Acids Res. 45, D331–D338 (2017).

  79. Ashburner, M. et al. Gene Ontology: tool for the unification of biology. Nat. Genet. 25, 25–29 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Clement, S. L. & Lykke-Andersen, J. A tethering approach to study proteins that activate mRNA turnover in human cells. Methods Mol. Biol. 419, 121–133 (2008).

    Article  CAS  PubMed  Google Scholar 

  81. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  82. Conway, A. E. et al. Enhanced CLIP uncovers IMP protein–RNA targets in human pluripotent stem cells important for cell adhesion and survival. Cell Rep. 15, 666–679 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Yeo lab, in particular E. Wheeler and F. Krach, for helpful discussions and A. Palazzo for advice on polysome fractionation. E.-C.L. was partly supported by a study-abroad graduate student fellowship from the Taiwanese government. J.C.S. was partially supported by a Natural Sciences and Engineering Research Council of Canada Postgraduate Scholarships–Doctoral (PGS D-532649-2019). Y.H. was supported by the UCSD Frontiers of Innovation Scholars Program. G.A.P. was supported by a graduate fellowship from the National Science Foundation. F.E.T. was supported by a postdoctoral fellowship from the American Cancer Society (129547-PF-16-060-01-RMC). S.M. was supported by a postdoctoral fellowship from the Larry L. Hillblom Foundation (2014-A-027-FEL). This work was supported by grants from the NIH (R01HG004659, U19MH107367, R01NS103172 and U41HG009889) to G.W.Y.

Author information

Authors and Affiliations

Authors

Contributions

G.W.Y. designed the study; S.A., E.-C.L. and G.W.Y. wrote the manuscript. D.E.H. provided RBP ORF plasmids; J.L.N., D.B.S., J.C.S. and D.B.S. collected, built and validated the tagged RBP libraries. J.L.N. designed and validated the tethering reporters. E.-C.L. and J.L.N. performed RT–qPCR and luciferase assays. E.-C.L. performed knockdown and overexpression assays, western blots and polysome profiling experiments. E.-C.L. and A.S. performed eCLIP and immunofluorescence and microscopy. E.-C.L. generated RNA-seq and polysome profiling RNA-seq libraries. S.M. generated the UBAP2L-knockout cell line. F.E.T. and E.-C.L. performed the RCas9 reporter assay. J.L.S. performed the SUnSET assay. E.-C.L., B.A.Y., S.S., Y.H. and G.A.P. performed bioinformatics analyses.

Corresponding author

Correspondence to Gene W. Yeo.

Ethics declarations

Competing interests

G.W.Y. is a cofounder and a member of the board of directors, is on the scientific advisory board and is an equity holder and paid consultant for Locana and Eclipse BioInnovations and a visiting professor at the National University of Singapore. The interests of G.W.Y. have been reviewed and approved by the University of California, San Diego in accordance with its conflict-of-interest policies. The authors declare no other competing financial interests.

Additional information

Peer review information Anke Sparmann was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Source of RBP ORFs, correlation between luciferase levels and RBP sizes, concordance between two luciferase systems, and correlation of reporter RNA and luciferase levels.

a, Western dot blot analysis of transiently expressed MCP-V5-tagged RBP ORFs in HeLa cells using a V5 antibody. Blue circles denote negative controls (no plasmid), red circles denote positive controls (CNOT7-V5-MCP). The order of wells and fold changes over negative controls are listed in Extended Data Table 1. b, Distribution of known classical and nonclassical RNA-binding domains in RBPs represented in our library. c, Distribution of molecular categories for RNA-related functions of RBPs represented in our library. d, Scatter plot of RBP size and luciferase effect. R, Pearson correlation coefficient. e, Luciferase activities from two different reporter constructs. Bar graphs showing log2-fold changes of the activity of Renilla (top) or firefly (bottom) luciferase reporters in presence of the MCP-fusion ORFs over FLAG control. Each vertical line represents a tethered ORF. f, Scatter plot of luciferase activities from the two reporter constructs. Values are expressed as log2-fold changes of the mean luciferase activity in the presence of MCP-fusion ORFs over FLAG controls. R, Pearson correlation coefficient.

Extended Data Fig. 2 IP validation from eCLIP experiments, correlation between eCLIP libraries, and de novo sequence motifs and metagene maps for candidate RBPs.

a,b, In-line western blots of eCLIP IPs of candidate RBPs. Extracts from HEK293T cells (a) or HEK293T cells transfected with the indicated V5-tagged RBP ORFs (b) immunoprecipitated with nonimmune (IgG) control antibodies, and western blot analysis using either RBP-specific (a) or anti-V5 (b) antibodies. The molecular weights (in kilodaltons) of standards are indicated on the right. Arrowheads indicate the calculated molecular weight for each RBP or RBP-V5 fusion protein. c, Heatmap of the Pearson correlation coefficients of fold enrichment of eCLIP peaks for the indicated 14 RBPs analyzed in duplicate. d, De novo sequence motifs in significant eCLIP peaks of the indicated RBP candidates enriched above background, with associated binomial P value. e–h, Metagene maps showing the distribution of eCLIP peak densities at target transcripts. The x axis indicates the relative length of each region. Dark red lines indicate the average number of significantly enriched peaks (≥4-fold enriched and P ≤ 10−2 versus SMInput) of eCLIP peak densities at all transcripts for BOLL, IFIT2, MEX3C, AIMP1 and CNOT7 (e), which show peak enrichment in 3ʹ UTR; DDX6, TOB1, NANOS3 and TOB2 (f), which show peak enrichment in 5ʹ UTR/3ʹ UTR; PARN and CLK3 (g), which show peak enrichment in 5ʹ UTR; and UBAP2L and MTDH (h), which show peak enrichment in CDS. Light shaded areas denote the 95% confidence interval.

Extended Data Fig. 3 Confirmation of RBP knockdown/overexpression, numbers of genes differentially regulated/unaffected by RBP perturbation, and region-level analysis of bound transcripts.

a,b, shRNA-mediated depletion of RBPs in HEK293T cells using 3-5 distinct shRNAs for each RBP, as indicated, compared to nontargeting shRNA control. a, Western blots with GAPDH or tubulin serving as loading controls, as indicated. b, Bar graphs indicating RBP transcript levels determined by RT–qPCR, normalized to levels of 18S rRNA. Data are shown as mean ± SD (n = 3 replicates). Asterisks denote significance at P < 0.05 determined by two-sided Student’s t-test. c, Overexpression of RBPs in HEK293T cells. Bar plots showing transcript levels (RPKM) for each RBP following transfection of RBP expression constructs or FLAG vector control. Data are shown as mean ± s.d. (n = 2 replicates). d–g, Numbers of up- or downregulated (log2-fold change ≥ 1.23 and FDR-corrected P ≤ 0.05) or unchanged genes for transcripts bound (≥4-fold enriched and P ≤ 10−2 versus eCLIP SMInput) or not bound by the indicated RBP for knockdown (d) and overexpression (e) of destabilizing RBPs and for knockdown (f) and overexpression (g) of stabilizing RBPs. h,i, Volcano plots showing the distribution of fold changes in transcript levels, with distribution histograms at the top, upon depletion of the destabilizer PARN (h) and depletion of the stabilizer CLK3 (left) and overexpression of the destabilizer IFIT2 (right) (i). Transcripts with log2(fold change) ≥1.23 and FDR-corrected P ≤ 0.05 are in color, with red and green denoting transcripts with or without at least one significant RBP binding peak (≥4-fold enriched and P ≤ 10−2 versus SMInput in eCLIP), respectively. j, Heatmap showing significance in differential expression of genes significantly differentially expressed (log2(fold change) ≥ 1.23 and FDR-corrected P ≤ 0.05) and significantly bound (≥4-fold enriched and P ≤ 10−2 versus SMInput in eCLIP) versus all unbound genes upon knockdown (KD) or overexpression (OE) of candidate RBPs in each region. Significance was calculated using a two-tailed Mann-Whitney U test. Uncropped images for a are available as source data online, and data for graphs in b, c are available as source data online.

Source data

Extended Data Fig. 4 Translation monitoring in a UBAP2L knockout replicate line, replicate concordance, and validation of polysome analyses in UBAP2L knockouts.

a, Translation monitoring using puromycin incorporation. Anti-puromycin western blot of extracts from puromycin-treated UBAP2L knockout (KO1) and parental (WT) HEK293T cell lines. GAPDH served as loading control. b,c, Polysome profile of UBAP2L after treatment of cells with 0.5 mM puromycin (b) and treatment of lysates with 30 mM EDTA (c). Top, absorbance (at 260 nm) plot of a HEK293T cell lysate fractionated through a 10-50% sucrose gradient. Bottom, western blots of UBAP2L from the corresponding fractions. d, Polysome profiles of HEK293T cells (WT, n = 2) and UBAP2L knockout HEK293T cells (KO, n = 4) fractionated through 10-50% sucrose gradients. Light-colored lines indicate means from each set (WT, light blue; KO, pink), and darkly shaded areas denote s.d. (WT, blue; KO, red). e, Bar graphs showing percentages of transcripts with RPKM ≥ 1 of all transcripts with ≥10 reads per transcript for two UBAP2L knockout lines (KO, 2 replicates each) and control samples (WT, two replicates). f, Scatter plots showing correlation of log2-transformed ratios of input-normalized polysome transcript levels (RPKM) between the two UBAP2L knockout HEK293T lines. R, Pearson correlation coefficient. g, Bar graph showing the percentage of regulated transcripts in UBAP2L targets and nontargets. *P < 0.0001 (χ2 test with Yates’s correction). h, RT–qPCR validation of reduced polysome association for the indicated transcripts. Transcript levels in inputs and polysome fractions were measured for KO and WT samples. KO/WT ratios of input-normalized polysome association of transcripts were then calculated. i, Western blots of DDX54, EIF4G1, EIF3B, and EEF2 in UBAP2L knockout cells (KO1, KO2). GAPDH served as a loading control. j,k, Quantitative flow cytometry reporter assay for mRNA translation using RCas9-fused 4EBP1. j, Plasmid design for the RCas9-4EBP1 experiment. k, Bar graph showing mean YFP levels in RCas9-4EBP1-expressing cells, normalized to RCas9-expressing cells, on each targeting site. Error bars denote s.d. from n > 5,000 RCas9-4EBP1-expressing and n > 5,000 rCas9-expressing cells per site. *P < 0.005; n.s., not significant (P > 0.5); two-tailed Student’s t-test. Uncropped images for ac and i and data for graphs in h and k are available as source data online.

Source data

Extended Data Fig. 5 Repetitive element analysis of UBAP2L eCLIP data.

a, Immunofluorescence images showing UBAP2L (green) in HEK293T cells. DAPI (blue) marks nuclei. Scale bar, 10 μm. b, Pie chart showing fractions of UBAP2L replicate 2 eCLIP reads unambiguously mapping to repeat families in HEK293T cells. c, Line plot of UBAP2L binding sites on rRNAs. Fold enrichment of reads for IP over SMInput is plotted against the nucleotide positions of 18S and 28S rRNAs. Asterisk (*) denotes relative entropy ≥0.01. d–g, Location of UBAP2L binding sites on rRNA. d, ES15L; e, ES7S; f, ES27L; g, ES31L. Nucleotides with significant binding are highlighted in yellow. h, RIP of UBAP2L-RIP and RT–PCR in HEK293T cell lysates. The RIP assay was performed using anti-UBAP2L antibody or rabbit nonimmune IgG. RT–PCR was performed using primer sets within UBAP2L target regions ES7S, ES7L, ES15L, and ES31L. Uncropped images for h are available as source data online.

Source data

Supplementary information

Supplementary Information

Supplementary Note.

Reporting Summary

Supplementary Table 1

For each RBP isoform in the tethered function ORF library, this table lists GenBank gene symbol and accession number(s), calculated molecular weight, prioritized GO term, whether the RBP was identified in Baltz et al. (2012) and/or Castello et al. (2012), prioritized known RNA-binding domain, the source of the ORF construct and expression levels in overexpression assays relative to the negative control.

Supplementary Table 2

For each RBP isoform (ORF) tested, this table lists RLuc–RBP and corresponding RLuc–FLAG luciferase levels of triplicate measurements and means for both; FLuc–RBP and corresponding FLuc–FLAG luciferase levels of triplicate measurements and means for both; mean fold changes (FLuc–RBP/RLuc–FLAG and RLuc–RBP/FLuc–FLAG ratios); P values; and whether the criterion of FDR < 0.01 was fulfilled.

Supplementary Table 3

This table lists the means of the log2-transformed normalized fold changes of luciferase levels of the FLuc reporter in the presence of the indicated tethered RBP and corresponding means of the reporter transcript levels as measured by RT–qPCR.

Supplementary Table 4

This table lists the oligonucleotide primer sequences for RT–qPCR of the luciferase reporter transcripts (RT–qPCR_luciferase assay), validation of shRNA-mediated RBP knockdown for RNA-seq assays (RT–qPCR_shRNA), validation of polysome profiling results (RT–qPCR_polysome) and RT–PCR assays used for RIP analysis on rRNAs (RIP-RT–PCR).

Supplementary Table 5

This table lists the manually curated functional categories and the direction of regulation.

Supplementary Table 6

This table lists the sources of the antibodies used for western blot (WB) and immunofluorescence staining (IF) with dilutions used and for IP in eCLIP (IP).

Supplementary Table 7

This table lists the numbers of raw reads, uniquely mapped reads and usable reads (that is, after PCR duplicate removal) as well as peak numbers (called peaks and significant peaks at thresholds ≥4-fold enrichment and P ≤ 10−2; χ2 test) for the eCLIP libraries generated. Each eCLIP experiment consists of a SMInput (Input) sample and an IP sample. For CNOT7 and AIMP1, the same input library was used for both RBPs due to their similar apparent molecular weight on SDS gel. OE, overexpression.

Supplementary Table 8

This table lists the numbers of raw reads, and numbers and fractions of uniquely mapping reads, for the RNA-seq libraries generated from lentiviral shRNA knockdown and overexpression (OE) samples.

Supplementary Table 9

This table lists the sources and target sequences for the lentiviral shRNA constructs used. TRC, The RNAi Consortium.

Supplementary Table 10

This table lists the numbers of raw reads and the numbers and fractions of uniquely mapping reads for the RNA-seq libraries generated in the polysome profiling studies.

Source data

Source Data Fig. 1

Statistical source data for Fig. 1k,l.

Source Data Fig. 4

Uncropped western blots for Fig. 4a–c,i.

Source Data Fig. 4

Statistical source data for Fig. 4b,i,k,l.

Source Data Fig. 5

Uncropped western blots for Fig. 5b,c.

Source Data Extended Data Fig. 3

Uncropped western blots for Extended Data Fig. 3a.

Source Data Extended Data Fig. 3

Statistical source data for Extended Data Fig. 3b,c.

Source Data Extended Data Fig. 4

Uncropped western blots for Extended Data Fig. 4a–c.

Source Data Extended Data Fig. 4

Statistical source data for Extended Data Fig. 4h,k.

Source Data Extended Data Fig. 5

Uncropped gels for Extended Data Fig. 5h.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Luo, EC., Nathanson, J.L., Tan, F.E. et al. Large-scale tethered function assays identify factors that regulate mRNA stability and translation. Nat Struct Mol Biol 27, 989–1000 (2020). https://doi.org/10.1038/s41594-020-0477-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-020-0477-6

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing