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:

Nucleolar URB1 ensures 3′ ETS rRNA removal to prevent exosome surveillance

Subjects

Abstract

The nucleolus is the most prominent membraneless condensate in the nucleus. It comprises hundreds of proteins with distinct roles in the rapid transcription of ribosomal RNA (rRNA) and efficient processing within units comprising a fibrillar centre and a dense fibrillar component and ribosome assembly in a granular component1. The precise localization of most nucleolar proteins and whether their specific localization contributes to the radial flux of pre-rRNA processing have remained unknown owing to insufficient resolution in imaging studies2,3,4,5. Therefore, how these nucleolar proteins are functionally coordinated with stepwise pre-rRNA processing requires further investigation. Here we screened 200 candidate nucleolar proteins using high-resolution live-cell microscopy and identified 12 proteins that are enriched towards the periphery of the dense fibrillar component (PDFC). Among these proteins, unhealthy ribosome biogenesis 1 (URB1) is a static, nucleolar protein that ensures 3′ end pre-rRNA anchoring and folding for U8 small nucleolar RNA recognition and the subsequent removal of the 3′ external transcribed spacer (ETS) at the dense fibrillar component–PDFC boundary. URB1 depletion leads to a disrupted PDFC, uncontrolled pre-rRNA movement, altered pre-rRNA conformation and retention of the 3′ ETS. These aberrant 3′ ETS-attached pre-rRNA intermediates activate exosome-dependent nucleolar surveillance, resulting in decreased 28S rRNA production, head malformations in zebrafish and delayed embryonic development in mice. This study provides insight into functional sub-nucleolar organization and identifies a physiologically essential step in rRNA maturation that requires the static protein URB1 in the phase-separated nucleolus.

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: Image-based screening identifies 12 nucleolar proteins that are enriched in the PDFC.
Fig. 2: URB1 is a conserved PDFC protein and is required for PDFC integrity.
Fig. 3: URB1 is required during early development.
Fig. 4: URB1 depletion disrupts 3′ ETS removal by altering U8 snoRNA binding to pre-32S rRNA.
Fig. 5: URB1 depletion impairs the distribution of 3′ ETS-containing pre-rRNA molecules and nucleolar morphology.
Fig. 6: URB1 depletion activates exosome-dependent pre-rRNA surveillance, impairing embryonic development.

Similar content being viewed by others

Data availability

All data supporting the findings of this study are available in the manuscript, Source Data, and at https://data.mendeley.com/datasets/8fmmx5vpkt/draft?a=c1a207f4-eda5-48be-99a9-92863496c868. SHAPE-MaP data with NAI and DMS, and iCLIP–seq datasets of Flag–URB1 are available at the Gene Expression Omnibus under accessions GSE194413 and GSE196625Source data are provided with this paper.

Code availability

Custom code used in SHAPE-MaP and iCLIP–seq analysis and the ImageJ script used in image analysis in this study are available from https://github.com/YangLab/Nucleolar-URB1-ensures-3-ETS-rRNA-removal-to-prevent-exosome--surveillance.

References

  1. Lafontaine, D. L. J., Riback, J. A., Bascetin, R. & Brangwynne, C. P. The nucleolus as a multiphase liquid condensate. Nat. Rev. Mol. Cell Bio. 22, 165–182 (2021).

    Article  CAS  Google Scholar 

  2. Thul, P. J. et al. A subcellular map of the human proteome. Science 356, eaal3321 (2017).

    Article  PubMed  Google Scholar 

  3. Huh, W. K. et al. Global analysis of protein localization in budding yeast. Nature 425, 686–691 (2003).

    Article  ADS  CAS  PubMed  Google Scholar 

  4. Yao, R. W. et al. Nascent pre-rRNA sorting via phase separation drives the assembly of dense fibrillar components in the human nucleolus. Mol. Cell 76, 767–783.e711 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Wu, M. et al. lncRNA SLERT controls phase separation of FC/DFCs to facilitate Pol I transcription. Science 373, 547–555 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  6. Boisvert, F. M., van Koningsbruggen, S., Navascues, J. & Lamond, A. I. The multifunctional nucleolus. Nat. Rev. Mol. Cell Biol. 8, 574–585 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Andersen, J. S. et al. Nucleolar proteome dynamics. Nature 433, 77–83 (2005).

    Article  ADS  CAS  PubMed  Google Scholar 

  8. Andersen, J. S. et al. Directed proteomic analysis of the human nucleolus. Curr. Biol. 12, 1–11 (2002).

    Article  PubMed  Google Scholar 

  9. Frottin, F. et al. The nucleolus functions as a phase-separated protein quality control compartment. Science 365, 342–347 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

  10. Scherl, A. et al. Functional proteomic analysis of human nucleolus. Mol. Biol. Cell 13, 4100–4109 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hu, R. et al. ZNF668 functions as a tumor suppressor by regulating p53 stability and function in breast cancer. Cancer Res. 71, 6524–6534 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Guo, D. C. et al. Loss-of-function mutations in YY1AP1 lead to grange syndrome and a fibromuscular dysplasia-like vascular disease. Am. J. Hum. Genet. 100, 21–30 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Stenstrom, L. et al. Mapping the nucleolar proteome reveals a spatiotemporal organization related to intrinsic protein disorder. Mol. Syst. Biol. 16, e9469 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Cuylen, S. et al. Ki-67 acts as a biological surfactant to disperse mitotic chromosomes. Nature 535, 308–312 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Meszaros, B., Erdos, G. & Dosztanyi, Z. IUPred2A: context-dependent prediction of protein disorder as a function of redox state and protein binding. Nucleic Acids Res. 46, W329–W337 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Feric, M. et al. Coexisting liquid phases underlie nucleolar subcompartments. Cell 165, 1686–1697 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Grob, A., Colleran, C. & McStay, B. Construction of synthetic nucleoli in human cells reveals how a major functional nuclear domain is formed and propagated through cell division. Genes Dev. 28, 220–230 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Drygin, D. et al. Anticancer activity of CX-3543: a direct inhibitor of rRNA biogenesis. Cancer Res. 69, 7653–7661 (2009).

    Article  CAS  PubMed  Google Scholar 

  19. Granick, D. Nucleolar necklaces in chick embryo fibroblast cells. I. Formation of necklaces by dichlororibobenzimidazole and other adenosine analogues that decrease RNA synthesis and degrade preribosomes. J. Cell Biol. 65, 398–417 (1975).

    Article  CAS  PubMed  Google Scholar 

  20. Mnaimneh, S. et al. Exploration of essential gene functions via titratable promoter alleles. Cell 118, 31–44 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. Dez, C. et al. Npa1p, a component of very early pre-60S ribosomal particles, associates with a subset of small nucleolar RNPs required for peptidyl transferase center modification. Mol. Cell. Biol. 24, 6324–6337 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Narla, A. & Ebert, B. L. Ribosomopathies: human disorders of ribosome dysfunction. Blood 115, 3196–3205 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Tafforeau, L. et al. The complexity of human ribosome biogenesis revealed by systematic nucleolar screening of Pre-rRNA processing factors. Mol. Cell 51, 539–551 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Zubradt, M. et al. DMS-MaPseq for genome-wide or targeted RNA structure probing in vivo. Nat. Methods 14, 75–82 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Peculis, B. A. & Steitz, J. A. Sequence and structural elements critical for U8 snRNP function in Xenopus oocytes are evolutionarily conserved. Genes Dev. 8, 2241–2255 (1994).

    Article  CAS  PubMed  Google Scholar 

  26. Badrock, A. P. et al. Analysis of U8 snoRNA variants in zebrafish reveals how bi-allelic variants cause leukoencephalopathy with calcifications and cysts. Am. J. Hum. Genet. 106, 694–706 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Leone, S., Bar, D., Slabber, C. F., Dalcher, D. & Santoro, R. The RNA helicase DHX9 establishes nucleolar heterochromatin, and this activity is required for embryonic stem cell differentiation. EMBO Rep. 18, 1248–1262 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Calo, E. et al. RNA helicase DDX21 coordinates transcription and ribosomal RNA processing. Nature 518, 249–253 (2015).

    Article  ADS  CAS  PubMed  Google Scholar 

  29. Bond, A. T., Mangus, D. A., He, F. & Jacobson, A. Absence of Dbp2p alters both nonsense-mediated mRNA decay and rRNA processing. Mol. Cell. Biol. 21, 7366–7379 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Martin, R., Straub, A. U., Doebele, C. & Bohnsack, M. T. DExD/H-box RNA helicases in ribosome biogenesis. RNA Biol. 10, 4–18 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Thomson, E., Ferreira-Cerca, S. & Hurt, E. Eukaryotic ribosome biogenesis at a glance. J. Cell Sci. 126, 4815–4821 (2013).

    Article  CAS  PubMed  Google Scholar 

  32. Farley-Barnes, K. I. et al. Diverse regulators of human ribosome biogenesis discovered by changes in nucleolar number. Cell Rep. 22, 1923–1934 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Schmid, M. & Jensen, T. H. The nuclear RNA exosome and its cofactors. Adv. Exp. Med. Biol. 1203, 113–132 (2019).

    Article  CAS  PubMed  Google Scholar 

  34. Wasmuth, E. V., Januszyk, K. & Lima, C. D. Structure of an Rrp6–RNA exosome complex bound to poly(A) RNA. Nature 511, 435–439 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  35. Dez, C., Houseley, J. & Tollervey, D. Surveillance of nuclear-restricted pre-ribosomes within a subnucleolar region of Saccharomyces cerevisiae. EMBO J. 25, 2662–2662 (2006).

    Article  CAS  PubMed Central  Google Scholar 

  36. Lafontaine, D. L. J. A. ‘Garbage can’ for ribosomes: how eukaryotes degrade their ribosomes. Trends Biochem. Sci 35, 267–277 (2010).

    Article  CAS  PubMed  Google Scholar 

  37. Lu, Z. et al. RNA duplex map in living cells reveals higher-order transcriptome structure. Cell 165, 1267–1279 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Dunn, K. W., Kamocka, M. M. & McDonald, J. H. A practical guide to evaluating colocalization in biological microscopy. Am. J. Physiol. Cell Physiol. 300, C723–C742 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Liu, C. X. et al. Structure and degradation of circular RNAs regulate PKR activation in innate immunity. Cell 177, 865–880.e821 (2019).

    Article  CAS  PubMed  Google Scholar 

  40. Walker, M. B. & Kimmel, C. B. A two-color acid-free cartilage and bone stain for zebrafish larvae. Biotech. Histochem. 82, 23–28 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. Shen, B. et al. Efficient genome modification by CRISPR–Cas9 nickase with minimal off-target effects. Nat. Methods 11, 399–402 (2014).

    Article  CAS  PubMed  Google Scholar 

  42. Thisse, C. & Thisse, B. High-resolution in situ hybridization to whole-mount zebrafish embryos. Nat. Protoc. 3, 59–69 (2008).

    Article  CAS  PubMed  Google Scholar 

  43. Oka, Y. & Sato, T. N. Whole-mount single molecule FISH method for zebrafish embryo. Sci. Rep. 5, 8571 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  44. Cordero, P., Kladwang, W., VanLang, C. C. & Das, R. Quantitative dimethyl sulfate mapping for automated RNA secondary structure inference. Biochemistry 51, 7037–7039 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Deigan, K. E., Li, T. W., Mathews, D. H. & Weeks, K. M. Accurate SHAPE-directed RNA structure determination. Proc. Natl Acad. Sci. USA 106, 97–102 (2009).

    Article  ADS  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Chen laboratory for discussions; J. Zhang, B.-Y. Zou and J.-G. Li for supporting our experiments; and the core facilities of Zhejiang University Medical Center and Liangzhu Laboratory, and Optofem Technology for technical support with STED. This study was supported by the CAS Project for Young Scientists in Basic Research (YSBR-009), the National Key R&D Program of China (2021YFA1100203), the Shanghai Municipal Commission for Science and Technology (20JC1410300), the National Natural Science Foundation of China (NSFC) (31830108, 31821004, and 31725009), the Center for Excellence in Molecular Cell Science (CEMCS) (2020DF03), the HHMI International Program (55008728) and the Xplorer Prize to L.-L.C.

Author information

Authors and Affiliations

Authors

Contributions

L.-L.C. supervised and conceived the project. L.-L.C., L.S. and G.X. designed experiments. Y.L. performed CLEM, supervised by L. Yu. L.Z. generated Urb1−/− mice, supervised by J.L. S.-X.G. performed SILAC, supervised by C.C.L.W. P.-H.Z. performed computational analyses, supervised by L. Yang. L.S., G.X., Y.H., R.-W.Y., P.-F.L., Y.-H.P., X.G., S.L., L.-Z.Y., S.-M.C., Z.-H.Y. and Y.W. performed all other experiments and analyses, supervised by L.-L.C. L.S., G.X. and L.-L.C. drafted the manuscript. L.-L.C. edited the manuscript.

Corresponding author

Correspondence to Ling-Ling Chen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Jean-Denis Beaudoin, Prasanth Kannanganattu and Denis Lafontaine for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 The schematic of pre-rRNA processing and experiment procedure of image-based nucleolar protein identification.

a. Coordination of the sub-nucleolar compartment with its function in ribosome biogenesis. b. The schematic of pre-rRNA processing in human cells. c. The workflow of live-cell imaging-based mapping of nucleolar proteins. d. Composition of nucleolar protein candidates in this paper2,7. e. Workflow of acquiring, processing and analyzing image data by Fiji/ImageJ. f. Cluster distribution of candidate nucleolar proteins. SCORENPM1 and SCOREDKC1 (see Methods for details) are shown to distinguish different nucleolar sub-regions. g. Distribution analysis procedure of candidate nucleolar proteins. h. The list of 140 proteins in different nucleolar sub-regions. i. Composition of 140 nucleolar enriched proteins from different reports2,7,10. j. Proportion of different nucleolar sub-regions (FC, DFC, PDFC, NR, GC and PNC) of the 140 nucleolar enriched proteins. k. Biological process GO-enrichment of nucleolar proteins enriched in FC, DFC, PDFC and GC. P-values are calculated by two-sided Mann-Whitney tests. l. Biological process enrichments in FC, DFC, PDFC, NR, GC and PNC. The circle areas are scaled by the number of proteins in each category.

Source data

Extended Data Fig. 2 Gallery of nucleolar protein candidates.

Representative images of all screened 200 candidate proteins by exogenous expression of EGFP tagged candidate protein in the dual-color reference knock-in cells shown in Extended Data Fig. 1c. Data was collected from n = 4 cells. Scale bar 2 μm. a. Representative images of nucleolar proteins enriched in FC. b. Representative images of nucleolar proteins enriched in PNC. c. Representative images of nucleolar proteins enriched in DFC. d. Representative images of nucleolar proteins enriched in PDFC. e. Representative images of nucleolar proteins enriched only in GC. f. Representative images of nucleolar proteins enriched in NR. g. Representative images of nucleolar proteins enriched in both GC and the nucleus. h. Representative images of nucleolar proteins enriched in both GC and the cytoplasm. i. Representative images of nucleolar proteins enriched in GC, the nucleus and the cytoplasm. j. Representative images of nucleolar proteins formed aggregations in GC. k. Representative images of protein candidates that obtained multiple distribution patterns. l. Representative images of protein candidates that did not show obvious signals in nucleoli.

Extended Data Fig. 3 Validation of nucleolar sub-regions.

a. Representative wide-field images of each individual PDFC protein in fixed HeLa cells with DKC1/NPM1 as markers of DFC/GC, respectively. Data was collected from n = 4 cells. Scale bar 2 μm. b. CLEM images of EGFP-KI URB1. Left, the merged image of EGFP-URB1 acquired by light microscopy and correlative nucleolus acquired by TEM. Right, the zoomed-in image from the white square. c. Validation of PDFC in H9 cells. Representative SIM image of PDFC (URB1) and DFC (FBL) by direct anti-URB1 and anti-FBL IF in H9 cells. d. Validation of PDFC in different cell types. Representative wide-field images of PDFC marker proteins URB1 and URB2 in K562, U2OS and HEK293 cells by exogenous expression of EGFP-tagged individual candidate proteins, mRuby3-DKC1and mTagBFP2-NPM1. Data was collected from n = 4 cells. Scale bar 2 μm. e. Diameters of different nucleolar compartments. RPA194, DKC1, URB1, and NPM1 were used as FC, DFC, PDFC and GC marker proteins, respectively, n = 60. f. Representative wide-field images of classical and non-classical nucleolar sub-regions. Left panel, classical nucleolar sub-regions are represented by exogenous expression of UBTF (FC marker), NOP56 (DFC marker) and DDX18 (GC marker) in DKC1/NPM1-KI dual-color cells as described in Extended Data Fig. 1c. Non-classical nucleolar sub-regions are marked by exogenous expression of URB1 (PDFC marker), PTBP1 (PNC marker) and MKI67 (NR marker). Data was collected from n = 4 cells. Scale bar 2 μm. g. Validation of GC condensates in different cell types. Representative wide-field images of GC condensates in K562, U2OS and HEK293 cells by exogenous expression of EGFP-RBM4, mRuby3-DKC1and mTagBFP2-NPM1. Data was collected from n = 4 cells. Scale bar 2 μm.

Source data

Extended Data Fig. 4 Characterization of nucleolar PDFC proteins.

a. The disorder tendency of 12 PDFC proteins calculated by IUPred2A. b. The rank of PDFC proteins about the disorder sequence content. URB1 and URB2 obtain the lowest disorder sequence content among all PDFC proteins. c. Two different perspectives of the URB1 and URB2 protein structures predicted by AlphaFold2. The predicted RNA binding domains are shown in magenta. d. Representative images of PDFC (URB1) and DFC (FBL) in HeLa cells treated with FBS starvation (24 h and 48 h), or UV radiation (60J). Data was collected from n = 4 cells. Scale bar 2 μm. e. URB2 surrounds DFC under different treatments. Representative images of DKC1 (DFC), URB2 (PDFC), and NPM1 (GC) proteins under DMSO (control), ActD, cx-5461 and DRB treatments in live HeLa cells. Of note, mRuby3-DKC1/mTagBFP2-NPM1 knock-in HeLa cells were introduced with EGFP-URB2 expression, followed by different treatments (ActD: 10 ng/mL, 3 h; DRB: 50 μM, 1.5 h; CX-5461:2 μM, 1 h). f. PDFC proteins surrounds DFC under Pol I inhibition. Representative images of DFC, PDFC (DDX47, NCL and DHX9) and GC proteins under ActD treatment in live HeLa cells. Data was collected from n = 4 cells. Scale bar 2 μm. g. RT-qPCR validation of the shRNA knockdown efficiency in URB1 KD and NCL KD cells. Data are presented as mean values +/− SEM. P-values are calculated by two-tailed Student’s t-test. n = 3 independent experiments.

Source data

Extended Data Fig. 5 Generation of URB1 depleted zebrafish and mouse models.

a. The conservation analysis of URB1 between human, mouse and zebrafish. URB1 has two conserved domains in N- and C- terminus. The conserved amino acids are shown in red according to conservative degrees. b. urb1 is barely expressed in zebrafish embryos at 8-cell and 4dpf stages, as shown by WISH with either the sense (control, upper panels) or the antisense urb1 (bottom panels) probe. Scale bar 100 μm. c. The schematic of the MO targeting site and KD efficiency detecting primers in zebrafish urb1 gene locus. d. RT-PCR validation of the MO efficiency targeting urb1. The agarose gel shows the MO KD efficiency detected by PCR using detecting primers in un-injected and urb1-MO zebrafish, n = 15. The red asterisk indicated the splicing-blocked mRNA of urb1 after MO injection. e. Statistics of small head deformity with different dosages of urb1 MO in zebrafish. f. Introducing urb1 mRNA rescues the urb1 MO phenotypes in zebrafish. Representative images of zebrafish cartilages stained with alcian blue after injection of urb1 MO alone, or urb1 MO and in vitro transcribed zebrafish urb1 mRNA. Scale bar 100 μm. g. Statistics of the head size in (f). n = 45 biologically independent animals. Data are presented as mean values +/− SEM. h. The schematic of the URB1-sgRNA targeting site in zebrafish urb1 gene locus. i. The sanger sequencing result of urb1+/− zebrafish confirmed the urb1+/− zebrafish has a 17-nucleotides deletion at exon 24, which resulted in a frameshift in the following urb1−/− embryos. j. URB1 depletion results in zebrafish cranial cartilage hypoplasia. Lateral view of alcian blue stained control and urb1−/− larvae at 3dpf and 4dpf. n = 6 animals. Scale bar 100 μm. k. URB1 depletion results in zebrafish cranial cartilage malformations. Statistics of width, length and head area (illustrated on left) in control, urb1 MO-injected and urb1−/− larvae at 4dpf. n = 62 biologically independent animals. Error bars, mean ± SEM. P-values are calculated by two-tailed Student’s t-test. l. Dorsal and lateral view of alcian blue stained control, urb1+/− and urb1−/− larvae. Ratio of observed phenotypes is labeled lower right of the panel. Scale bar 100 μm. m. Statistics of the head length, head width, length/width and head size in (j). urb1−/− larvae have a retardation in various aspects detected in head development compared to control and urb1+/− larvae. n = 102 independent animals. Data are presented as mean values +/− SEM. P-values are calculated by two-tailed Student’s t-test. n. Sanger sequencing confirmed that the Urb1+/− mouse received a 2-nucleotides insertion at exon 1, which resulted in a frameshift in Urb1−/− embryos. o. Statistics of blastocysts (E4.5) isolated from Urb1+/− intercrosses. p. URB1 depletion impaired the colony formation ability of H9 cells. Data are presented as mean values +/− SEM. P-values are calculated by two-tailed Student’s t-test.

Source data

Extended Data Fig. 6 URB1 depletion impairs 32S pre-rRNA biogenesis.

a. WB validation of URB1 depletion efficiency in URB1 KD cell lines. b. URB1 depletion affects pre-rRNA biogenesis. Left, schematic of human pre-rRNA processing and positions of probes used in Northern Blots (NB). Right, NB results of pre-rRNA intermediates in scramble shRNA-treated and URB1-KD HeLa cells, with probes recognizing 5′ ETS and ITS1, respectively. c. Quantification of pre-rRNA intermediates detected by different NB probes shown in (b). Each indicated rRNA intermediate is normalized to scramble shRNA-treated cells and shown as the relative intensities. n = 4 independent experiments. Data are presented as mean values +/− SEM. d. The aberrant 32S pre-rRNA intermediates contain the 5.8S rRNA region. Left, schematic representation of human 5.8S probe related rRNA intermediates in NB. Right, NB analyses of 5.8S probe related rRNA intermediates in scramble and URB1 KD HeLa cells. e. Aberrant 3′ 32S pre-rRNA takes up a majority of total 32S pre-rRNA after URB1 depletion. Left, schematic of human 28S-3′ ETS related rRNA intermediates in NB. Right, NB of 28S-3′ ETS probe related rRNA intermediates in scramble shRNA-treated and URB1 KD HeLa cells. f. URB1 depletion disrupts 32S pre-rRNA biogenesis in 293FT cells. NB analyses of ITS2 and 3′ ETS probe related rRNA intermediates in scramble shRNA-treated and URB1 KD 293FT cells. Red pentacle, aberrant intermediates. g. NB analyses of ITS2 probe related rRNA intermediates show no obvious reduction in 32S pre-rRNA and no accumulation of aberrant intermediates after URB2 KD compared to URB1 KD. Red pentacle, aberrant intermediates. h. NB analyses of rRNA intermediates in scramble shRNA-treated, DDX21 KD and DDX50 KD HeLa cells, respectively with 5′ ETS, ITS1 and ITS2 probe shows no obvious or subtle reduction in 32S pre-rRNA compared to URB1 KD.

Source data

Extended Data Fig. 7 The depletion of URB1 leads to an increase in the SHAPE value of binding region and its upstream revealed by SHAPE-MaP.

a. Schematic of primers used in SHAPE-MaP experiments. b. Predicated secondary structural modes of 28S-3′ ETS region in scramble shRNA-treated and URB1 KD HeLa cells based on NAI labeling. Red region, URB1 binding region. c. The DMS scores of 28S-3′ ETS region in scramble shRNA-treated and URB1 KD HeLa cells labeled by DMS. d. Predicated secondary structure modes in (c). Red region, URB1 binding region. e. The SHAPE scores of 28S-3′ ETS region and U8-snoRNA binding region in scramble shRNA-treated and URB1 KD HeLa cells labeled by NAI.

Extended Data Fig. 8 Examined PDFC proteins prefer to bind the 28S and 3′ end regions of pre-rRNAs.

a. Schematic of pulse-chase EU labelling experiment. b. Several examined PDFC proteins preferentially bind to the 3′ end of 28S pre-rRNA analyzed by RNA Immunoprecipitation (RIP) in HeLa cells. Left, the schematic of RIP experiments. Right, the heatmap of PDFC proteins binding to the 47S pre-rRNA. Data was collected from three independent experiments; heatmap values were generated by averaging IP/Input of each examined PDFC protein, and normalized to the highest binding capability, respectively.

Source data

Extended Data Fig. 9 URB1 depletion alters the distribution of both 3′ ETS-attached pre-rRNAs and U8 snoRNAs.

a. 3′ ETS-attached pre-rRNAs localize at the periphery of DFC in H9 cells. Top, representative single-z-section SIM images of 3′ ETS-attached pre-rRNAs (red) and DFC (FBL, green). Bottom, representative averaged images. b. U8 snoRNAs localize at the periphery of DFC. Top, representative single-z-section SIM images of U8 snoRNAs (magenta) and DFC (FBL, green) in HeLa cells. Bottom, representative single-z-section SIM images of U8 snoRNA and DFC (FBL) in H9 cells. c. URB1 KD alters the distribution of U8 snoRNAs. U8 snoRNAs (magenta) detected by smFISH were aberrantly dispersed to GC after URB1 knockdown in HeLa cells. Top, representative single-z-section SIM images of U8 snoRNAs and DFC in scramble and URB1 KD cells. Bottom, averaged images shown above, n = 20. d. Statistics of the altered distributions of U8 snoRNAs in (c). The floating bar shows the range of values, and the center line represents the median. P-values are calculated by two-sided Mann-Whitney tests. e. URB1 depletion led to reduced ribosome production in 293FT cells and HeLa cells, as shown by ribosome fractionations. The position of URB1 KD is noted by the green line and the scramble is noted by the black line. f. URB1 KD leads to reduced GC volume. Representative single z-section (upper panels) and 3D-reconstructed (bottom panels) SIM images of DKC1 (magenta) and NPM1 (blue) in scramble shRNA-treated and URB1-KD HeLa cells. White arrows show that some DFCs are extruded outside of GC. g. Statistics of the GC volume, the mean distance between DFCs and DFC numbers per cell in (f), n = 15. The floating bar shows the range of values. Data are presented as mean values +/− SEM. P-values are calculated by two-tailed Student’s t-test. (NS, not significant). h. URB1 KD led to decreased mobility of NPM1 in HeLa cells. FRAP analysis of NPM1 in scramble shRNA-treated and URB1 KD HeLa cells, n = 10. i. URB1 KD led to retarded cell proliferation in HeLa cells, revealed by MTT cell proliferation assays, n = 3. Data are presented as mean values +/− SEM. j. 3′ ETS-attached pre-rRNAs and U8 snoRNAs localize at the periphery of merged FC/DFC in zebrafish embryo. U8-snoRNAs (top, red) and 3′ ETS-attached pre-rRNAs (bottom, red) detected by smFISH surround z-fbl (blue) in zebrafish 8 hpf embryos, n = 3. Data are presented as mean values +/− SEM. k. urb1−/− larvae exhibit a less-defined midbrain-hindbrain boundaryand disorganized trunk vasculature (arrowheads) compared to siblings at 24 hpf. Scale bar 100 μm. l. mUrb1 KD impairs the radial flux processing of pre-rRNAs. 3′ ETS-attached pre-rRNAs (red) detected by 3′ ETS smFISH probes were diffused to the outside of DFC (FBL detected by IF) after mUrb1 knockdown in mouse NIH-3T3 cells, n = 20. m. mUrb1 depletion impairs the radial flux processing of pre-rRNAs in blastocyst (E4.0). Representative images of 3′ ETS-attached pre-rRNAs in control and mUrb1-KO (knockout) blastocyst by smFISH showed a diffused pattern of pre-rRNAs into GC region after mUrb1 depletion, n = 3. Scale bar 10 μm.

Source data

Extended Data Fig. 10 Loss of URB1 activates the exosome surveillance in the nucleolus.

a. The workflow of stable isotope labeling using amino acids in cell culture (SILAC) followed by mass spectrometry (MS) in scramble shRNA-treated and URB1 KD Hela cells. b. Volcano plot of the changes in protein expression upon URB1 KD. 4,688 proteins were overlapped in independent replicates. P-values are calculated by two-sided Mann-Whitney tests. c. The number of up-regulated and down-regulated proteins in URB1 KD HeLa cells. 274 proteins are up-regulated >2-fold and 170 proteins are downregulated >2-fold. d. GO/KEGG analyses of expression changed proteins upon URB1 KD in HeLa cells. Briefly, the expression levels of ribosome components (especially LSU) are down regulated and EIF4E (RNA transport factor) is up regulated. e. WB validation of the related changes in protein expression upon URB1 KD. EXOSC8 is up regulated, RPL10 and RPL23 are down regulated. f. Exosome components accumulate with 3′ ETS-attached pre-rRNAs in nucleoli upon URB1 KD. Representative images of EXOSC8, EXOSC9 and MTR4 with 3′ ETS-attached pre-rRNAs in scramble shRNA-treated and URB1 KD HeLa cells are shown by smFISH and IF. g. NB analyses of rRNA intermediates in scramble shRNA-treated, URB1, EXOSC1/3/4, EXOSC7/9/10, DIS3 and DIS3/MTR4 KD HeLa cells, respectively with ITS2 or 3′ ETS probes. Left, depletion of different exosome components led to a reduction or no change in 32S pre-rRNA expression, and no obvious aberrant intermediates were observed. Right, depletion of different exosome components did not affect 3′ ETS processing. h. Most exosome components KD rescues the abnormal exosome-dependent 32S pre-rRNA degradation. Left, the schematic of URB1 and exosome components that affect the 32S pre-rRNA degradation. Right, NB detection of the ITS2-containing pre-rRNA intermediates in scramble shRNA-treated and URB1 KD cells that were further depleted exosome components. i. RT-qPCR validation of the shRNA knockdown efficiency in cells KD of URB1 and exosome components. P-values are calculated by two-tailed Student’s t-test. j. URB1 depletion generated 3′ ETS-attached pre-rRNAs carry short poly(A) tails. NB detection using the 3′ ETS probes after oligo-dT pull-down in scramble shRNA-treated and URB1 KD HeLa cells. k. 3′ RACE analysis of pre-rRNA intermediates in scramble shRNA-treated and URB1 KD HeLa cells shows short poly(A) tails in 3′ ETS-attached pre-rRNAs.

Source data

Supplementary information

Supplementary Figure 1

The unprocessed Immunoblots associated with the data presented in main and extended data figures.

Reporting Summary

Supplementary Table 1

Oligonucleotide sequences used in this study, comprising: sheet1—primer sequences used for plasmid construction, qPCR analysis, SHAPE-MaP analysis and 3′ RACE analysis; sheet 2—sgRNA sequences used for genome editing; sheet 3—morpholino and shRNA sequences used for knockdown; and sheet 4—smFISH and northern blot probes used in this study.

Supplementary Table 2

Source data

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shan, L., Xu, G., Yao, RW. et al. Nucleolar URB1 ensures 3′ ETS rRNA removal to prevent exosome surveillance. Nature 615, 526–534 (2023). https://doi.org/10.1038/s41586-023-05767-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-05767-5

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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