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:

Genome organization around nuclear speckles drives mRNA splicing efficiency

Abstract

The nucleus is highly organized, such that factors involved in the transcription and processing of distinct classes of RNA are confined within specific nuclear bodies1,2. One example is the nuclear speckle, which is defined by high concentrations of protein and noncoding RNA regulators of pre-mRNA splicing3. What functional role, if any, speckles might play in the process of mRNA splicing is unclear4,5. Here we show that genes localized near nuclear speckles display higher spliceosome concentrations, increased spliceosome binding to their pre-mRNAs and higher co-transcriptional splicing levels than genes that are located farther from nuclear speckles. Gene organization around nuclear speckles is dynamic between cell types, and changes in speckle proximity lead to differences in splicing efficiency. Finally, directed recruitment of a pre-mRNA to nuclear speckles is sufficient to increase mRNA splicing levels. Together, our results integrate the long-standing observations of nuclear speckles with the biochemistry of mRNA splicing and demonstrate a crucial role for dynamic three-dimensional spatial organization of genomic DNA in driving spliceosome concentrations and controlling the efficiency of mRNA splicing.

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: snRNAs preferentially bind pre-mRNAs of genes that are close to speckles.
Fig. 2: Co-transcriptional splicing efficiency varies based on the proximity to nuclear speckles.
Fig. 3: Expression of a gene from a genomic locus in proximity to nuclear speckles leads to increased mRNA splicing.
Fig. 4: Differential gene positioning around speckles leads to different splicing efficiencies across cell types.
Fig. 5: Pre-mRNA organization around nuclear speckles drives splicing efficiency.
Fig. 6: Integrated model of how gene organization around nuclear speckles affects splicing.

Similar content being viewed by others

Data availability

Sequencing datasets have been deposited into the GEO with accession identifier GSE247833.

Code availability

Additional scripts and data are available at GitHub (https://github.com/GuttmanLab/speckle).

References

  1. Bhat, P., Honson, D. & Guttman, M. Nuclear compartmentalization as a mechanism of quantitative control of gene expression. Nat. Rev. Mol. Cell Biol. 22, 653–670 (2021).

    Article  CAS  PubMed  Google Scholar 

  2. Dundr, M. & Misteli, T. Biogenesis of nuclear bodies. Cold Spring Harb. Perspect. Biol. https://doi.org/10.1101/cshperspect.a000711 (2010).

  3. Spector, D. L. & Lamond, A. I. Nuclear speckles. Cold Spring Harb. Perspect. Biol. 3, a000646 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Mattaj, I. W. Splicing in space. Nature https://doi.org/10.1038/372727a0 (1994).

  5. Lewis, J. D. & Tollervey, D. Like attracts like: getting RNA processing together in the nucleus. Science 288, 1385–1389 (2000).

    Article  ADS  CAS  PubMed  Google Scholar 

  6. Shachar, S. & Misteli, T. Causes and consequences of nuclear gene positioning. J. Cell Sci. 130, 1501–1508 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Belmont, A. S. Nuclear compartments: an incomplete primer to nuclear compartments, bodies, and genome organization relative to nuclear architecture. Cold Spring Harb. Perspect. Biol. 14, a041268 (2022).

    Article  CAS  PubMed  Google Scholar 

  8. Putnam, A., Thomas, L. & Seydoux, G. RNA granules: functional compartments or incidental condensates? Genes Dev. 37, 354–376 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Herzel, L., Ottoz, D. S. M., Alpert, T. & Neugebauer, K. M. Splicing and transcription touch base: co-transcriptional spliceosome assembly and function. Nat. Rev. Mol. Cell Biol. https://doi.org/10.1038/nrm.2017.63 (2017).

  10. Maquat, L. E. Nonsense-mediated mRNA decay: splicing, translation and mRNP dynamics. Nat. Rev. Mol. Cell Biol. 5, 89–99 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  12. Huang, S. & Spector, D. L. U1 and U2 small nuclear RNAs are present in nuclear speckles. Proc. Natl Acad. Sci. USA 89, 305–308 (1992).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Fu, X. D. & Maniatis, T. Factor required for mammalian spliceosome assembly is localized to discrete regions in the nucleus. Nature 343, 437–441 (1990).

    Article  ADS  CAS  PubMed  Google Scholar 

  14. Spector, D. L., Schrier, W. H. & Busch, H. Immunoelectron microscopic localization of snRNPs. Biol. Cell 49, 1–10 (1983).

    Article  CAS  PubMed  Google Scholar 

  15. Lerner, E. A., Lerner, M. R., Janeway, C. A. J. & Steitz, J. A. Monoclonal antibodies to nucleic acid-containing cellular constituents: probes for molecular biology and autoimmune disease. Proc. Natl Acad. Sci. USA 78, 2737–2741 (1981).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Hall, L. L., Smith, K. P., Byron, M. & Lawrence, J. B. Molecular anatomy of a speckle. Anat. Rec. A Discov. Mol. Cell. Evol. Biol. https://doi.org/10.1002/ar.a.20336 (2006).

  17. Cmarko, D. et al. Ultrastructural analysis of transcription and splicing in the cell nucleus after bromo-UTP microinjection. Mol. Biol. Cell 10, 211–223 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Fakan, S., Leser, G. & Martin, T. E. Ultrastructural distribution of nuclear ribonucleoproteins as visualized by immunocytochemistry on thin sections. J. Cell Biol. 98, 358–363 (1984).

    Article  CAS  PubMed  Google Scholar 

  19. Zhang, G., Taneja, K. L., Singer, R. H. & Green, M. R. Localization of pre-mRNA splicing in mammalian nuclei. Nature 372, 809–812 (1994).

    Article  ADS  CAS  PubMed  Google Scholar 

  20. Huang, S., Deerinck, T. J., Ellisman, M. H. & Spector, D. L. In vivo analysis of the stability and transport of nuclear poly(A)+ RNA. J. Cell Biol. 126, 877–899 (1994).

    Article  CAS  PubMed  Google Scholar 

  21. Misteli, T., Cáceres, J. F. & Spector, D. L. The dynamics of a pre-mRNA splicing factor in living cells. Nature 387, 523–527 (1997).

    Article  ADS  CAS  PubMed  Google Scholar 

  22. Jiménez-García, L. F. & Spector, D. L. In vivo evidence that transcription and splicing are coordinated by a recruiting mechanism. Cell 73, 47–59 (1993).

    Article  PubMed  Google Scholar 

  23. Sacco-Bubulya, P. & Spector, D. L. Disassembly of interchromatin granule clusters alters the coordination of transcription and pre-mRNA splicing. J. Cell Biol. 156, 425–436 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Misteli, T. & Spector, D. L. Serine/threonine phosphatase 1 modulates the subnuclear distribution of pre-mRNA splicing factors. Mol. Biol. Cell 7, 1559–1572 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Huang, S. & Spector, D. L. Intron-dependent recruitment of pre-mRNA splicing factors to sites of transcription. J. Cell Biol. 133, 719–732 (1996).

    Article  CAS  PubMed  Google Scholar 

  26. Misteli, T. & Spector, D. L. RNA polymerase II targets pre-mRNA splicing factors to transcription sites in vivo. Mol. Cell 3, 697–705 (1999).

    Article  CAS  PubMed  Google Scholar 

  27. Xie, S. Q., Martin, S., Guillot, P. V., Bentley, D. L. & Pombo, A. Splicing speckles are not reservoirs of RNA polymerase II, but contain an inactive form, phosphorylated on serine2 residues of the C-terminal domain. Mol. Biol. Cell 17, 1723–1733 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Misteli, T. et al. Serine phosphorylation of SR proteins is required for their recruitment to sites of transcription in vivo. J. Cell Biol. 143, 297–307 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Tammer, L. et al. Gene architecture directs splicing outcome in separate nuclear spatial regions. Mol. Cell 82, 1021–1034.e8 (2022).

    Article  CAS  PubMed  Google Scholar 

  30. Barutcu, A. R. et al. Systematic mapping of nuclear domain-associated transcripts reveals speckles and lamina as hubs of functionally distinct retained introns. Mol. Cell 82, 1035–1052.e9 (2022).

    Article  CAS  PubMed  Google Scholar 

  31. Wang, K. et al. Intronless mRNAs transit through nuclear speckles to gain export competence. J. Cell Biol. 217, 3912–3929 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kim, J., Venkata, N. C., Hernandez Gonzalez, G. A., Khanna, N. & Belmont, A. S. Gene expression amplification by nuclear speckle association. J. Cell Biol. 219, e201904046 (2020).

    PubMed  Google Scholar 

  33. Zhong, X.-Y., Wang, P., Han, J., Rosenfeld, M. G. & Fu, X.-D. SR proteins in vertical integration of gene expression from transcription to RNA processing to translation. Mol. Cell 35, 1–10 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lawrence, J. B. & Clemson, C. M. Gene associations: true romance or chance meeting in a nuclear neighborhood? J. Cell Biol. 182, 1035–1038 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Perraud, M., Gioud, M. & Monier, J. C. [Intranuclear structures of monkey kidney cells recognised by immunofluorescence and immuno-electron microscopy using anti-ribonucleoprotein antibodies (author’s transl)]. Ann. Immunol. 130C, 635–647 (1979).

    CAS  Google Scholar 

  36. Chen, Y. et al. Mapping 3D genome organization relative to nuclear compartments using TSA-seq as a cytological ruler. J. Cell Biol. 217, 4025–4048 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Quinodoz, S. A. et al. Higher-order inter-chromosomal hubs shape 3D genome organization in the nucleus. Cell 174, 744–757.e24 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Takei, Y. et al. Integrated spatial genomics reveals global architecture of single nuclei. Nature 590, 344–350 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Quinodoz, S. A. et al. RNA promotes the formation of spatial compartments in the nucleus. Cell 184, 5775–5790.e30 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wilkinson, M. E., Charenton, C. & Nagai, K. RNA splicing by the spliceosome. Annu. Rev. Biochem. 89, 359–388 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Will, C. L. & Lührmann, R. Spliceosome structure and function. Cold Spring Harb. Perspect. Biol. 3, a003707 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Calvet, J. P. & Pederson, T. Base-pairing interactions between small nuclear RNAs and nuclear RNA precursors as revealed by psoralen cross-linking in vivo. Cell 26, 363–370 (1981).

    Article  CAS  PubMed  Google Scholar 

  43. Engreitz, J. M. et al. RNA–RNA interactions enable specific targeting of noncoding RNAs to nascent pre-mRNAs and chromatin sites. Cell 159, 188–199 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wuarin, J. & Schibler, U. Physical isolation of nascent RNA chains transcribed by RNA polymerase II: evidence for cotranscriptional splicing. Mol. Cell. Biol. 14, 7219–7225 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Goronzy, I. N. et al. Simultaneous mapping of 3D structure and nascent RNAs argues against nuclear compartments that preclude transcription. Cell Rep. 41, 111730 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Takei, Y. et al. Single-cell nuclear architecture across cell types in the mouse brain. Science 374, 586–594 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  47. Zhang, L. et al. TSA-seq reveals a largely conserved genome organization relative to nuclear speckles with small position changes tightly correlated with gene expression changes. Genome Res. 31, 251–264 (2020).

    Article  CAS  PubMed  Google Scholar 

  48. Pichon, X., Robert, M.-C., Bertrand, E., Singer, R. H. & Tutucci, E. New generations of MS2 variants and MCP fusions to detect single mRNAs in living eukaryotic cells. Methods Mol. Biol. 2166, 121–144 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Tripathi, V. et al. SRSF1 regulates the assembly of pre-mRNA processing factors in nuclear speckles. Mol. Biol. Cell 23, 3694–3706 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ilık, İ. A. et al. SON and SRRM2 are essential for nuclear speckle formation. eLife 9, e60579 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  52. Olins, A. L., Rhodes, G., Welch, D. B. M., Zwerger, M. & Olins, D. E. Lamin B receptor: multi-tasking at the nuclear envelope. Nucleus 1, 53–70 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Reed, R. & Maniatis, T. A role for exon sequences and splice-site proximity in splice-site selection. Cell 46, 681–690 (1986).

    Article  CAS  PubMed  Google Scholar 

  54. Szymczyna, B. R. et al. Structure and function of the PWI motif: a novel nucleic acid-binding domain that facilitates pre-mRNA processing. Genes Dev. 17, 461–475 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Quinodoz, S. A. & Guttman, M. Essential roles for RNA in shaping nuclear organization. Cold Spring Harb. Perspect. Biol. 14, a039719 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. McCracken, S. et al. The C-terminal domain of RNA polymerase II couples mRNA processing to transcription. Nature 385, 357–360 (1997).

    Article  ADS  CAS  PubMed  Google Scholar 

  57. Ding, F. & Elowitz, M. B. Constitutive splicing and economies of scale in gene expression. Nat. Struct. Mol. Biol. 26, 424–432 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Yuryev, A. et al. The C-terminal domain of the largest subunit of RNA polymerase II interacts with a novel set of serine/arginine-rich proteins. Proc. Natl Acad. Sci. USA 93, 6975–6980 (1996).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  59. Lazarev, D. & Manley, J. L. Concurrent splicing and transcription are not sufficient to enhance splicing efficiency. RNA 13, 1546–1557 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zimber, A., Nguyen, Q.-D. & Gespach, C. Nuclear bodies and compartments: functional roles and cellular signalling in health and disease. Cell Signal. 16, 1085–1104 (2004).

    Article  CAS  PubMed  Google Scholar 

  61. Quinodoz, S. A. et al. SPRITE: a genome-wide method for mapping higher-order 3D interactions in the nucleus using combinatorial split-and-pool barcoding. Nat. Protoc. 17, 36–75 (2022).

    Article  CAS  PubMed  Google Scholar 

  62. Banerjee, A. K. et al. SARS-CoV-2 disrupts splicing, translation, and protein trafficking to suppress host defenses. Cell 183, 1325–1339.e21 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Melsted, P. et al. Modular, efficient and constant-memory single-cell RNA-seq preprocessing. Nat. Biotechnol. 39, 813–818 (2021).

    Article  CAS  PubMed  Google Scholar 

  64. Engreitz, J. M. et al. Local regulation of gene expression by lncRNA promoters, transcription and splicing. Nature 539, 452–455 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  65. Huang, S. et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science 360, 176–182 (2018).

    Article  ADS  Google Scholar 

  66. Huang, S. et al. Mapping and modeling the genomic basis of differential RNA isoform expression at single-cell resolution with LR-Split-seq. Genome Biol. 22, 286 (2021).

    Article  Google Scholar 

  67. Majumdar, D. S. et al. Programmed delayed splicing: a mechanism for timed inflammatory gene expression. Preprint at bioRxiv https://doi.org/10.1101/443796 (2018).

  68. Mayr, C. & Bartel, D. P. Widespread shortening of 3′ UTRs by alternative cleavage and polyadenylation activates oncogenes in cancer cells. Cell 138, 673–684 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Manna, P. T., Davis, L. J. & Robinson, M. S. Fast and cloning-free CRISPR/Cas9-mediated genomic editing in mammalian cells. Traffic 20, 974–982 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Raj, A., van den Bogaard, P., Rifkin, S. A., van Oudenaarden, A. & Tyagi, S. Imaging individual mRNA molecules using multiple singly labeled probes. Nat. Methods 5, 877–879 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Stringer, C., Wang, T., Michaelos, M. & Pachitariu, M. Cellpose: a generalist algorithm for cellular segmentation. Nat. Methods 18, 100–106 (2021).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. Honson, E. Detmar and D. Perez for experimental help; B. Riviere, L. Pachter and N. Ollikainen for computational help; M. Flynn for the bidirectional reporter plasmid; L. Cai, M. Elowitz and A. Raj for reagents; F. Ding, H. Yin, J. Jachowicz, L. Frankiw and Y. Luo for discussions; B. Yeh for discussions about splicing efficiency calculations; I. Antoshechkin for sequencing; G. Spigola for microscopy advice; A. Lin for sequencing advice; B. Yeh, D. Honson and K. Leslie for critical comments on the manuscript; R. Maehr and K. Mohan Parsi for H1 ES cell lines; B. Wold and B. Williams for myocyte cell lines; and S. Hiley for editing. Illustrations in Figs. 1a,c–d,e,g2a,b, 3a,b, 4a, 5a–c and 6 and Extended Data Figs. 4b and 9 were created by I.-M. Strazhnik, Caltech. Imaging was performed at the Biological Imaging Facility with the support of the Caltech Beckman Institute and the Arnold and Mabel Beckman Foundation. This work was funded by NIH T32 GM 7616-40, NIH NRSA CA247447, the UCLA-Caltech Medical Scientist Training Program, a Chen Graduate Innovator Grant, and the Josephine De Karman Fellowship Trust (P.B.); and a HHMI Gilliam Fellowship, NSF GRFP Fellowship, and the HHMI Hanna H. Gray Fellows Program (S.A.Q.). This work was funded by the NIH 4DN program (U01 DK127420), NIH Directors’ Transformative Research Award (R01 DA053178), the NYSCF, CZI Ben Barres Early Career Acceleration Award, and funds from Caltech.

Author information

Authors and Affiliations

Authors

Contributions

P.B. and M.G. conceived the study, analysed data, interpreted results and wrote the manuscript. P.B., A.C. and M.G. designed experiments. B.E. performed RNA FISH and image analyses, including nuclear segmentation and spot detection. P.B., A.C., O.E. and W.H. generated plasmids for the MCP–MS2 experiments and performed co-transfection experiments for FACS. S.A.Q., E.S. and S.-T.G. generated mouse MM14 myocyte SPRITE data. M.R.B. generated H1 human ES cell SPRITE data. M.S. processed imaging data. P.B. and A.B. performed SC35 immunofluorescence combined with fluorescence microscopy for mCherry constructs. P.B. and A.W.C. performed 5EU RNA-seq. P.B and J.L.M.F. performed DNA FISH combined with SRRM1 immunofluorescence. Y.T analysed seqFISH+ data. I.N.G. generated computational assignment of speckle hubs for human SPRITE data. D.K.S. processed myocyte split-seq data using kallisto, and T.C. developed analytical methods for comparison of splicing efficiency between cell types. P.B. and M.G. supervised the work and M.G. acquired funding.

Corresponding author

Correspondence to Mitchell Guttman.

Ethics declarations

Competing interests

S.A.Q. and M.G. are inventors on a patent covering the SPRITE method.

Peer review

Peer review information

Nature thanks Giacomo Cavalli and the other, anonymous, reviewer(s) 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 Correlation between speckle proximity scores between SPRITE datasets and TSA-seq for SON.

A. Chromosome wide view of speckle proximity score at 1 Mb-resolution for three replicates of SPRITE datasets in mouse ES cells. Two collected in Quinodoz et al Cell 2021 and a third dataset collected for this manuscript. Speckle hub regions highlighted on chromosomes in red. Gene density track on bottom. Correlation of SPRITE experiments between: B. RD SPRITE Cell 2021 (Replicate 1) and RD SPRITE Cell 2021 (Replicate 2) (spearman r = 0.94, p < 0.0001, P value is two-tailed). C. RD SPRITE Cell 2021 (Replicate 1) and Bhat et al 2024 (spearman r = 0.90, p < 0.0001, P value is two-tailed). D. RD SPRITE Cell 2021 (Replicate 2) and Bhat et al 2024 (spearman r = 0.87, p < 0.0001, P value is two-tailed). E. Correlation of SPRITE and TSA-seq for speckle protein, SON, in H1 hESCs (spearman r = 0.75, p < 0.0001, P value is two-tailed). F. Chromosome wide view of speckle proximity score (top track) and TSA-seq (middle track, values > 0 shown) at 100-kb resolution for H1 hESCs. Gene density shown on bottom.

Extended Data Fig. 2 snRNA density for differently expressed genomic regions and different nascent transcription density.

A. To ensure that splicing factor difference were not due to expression differences between speckle close and speckle far genes, we divided genes up based on expression ranges: high expression (RPKM = 7.5-20), medium expression (RPKM = 2.5-7.5), low expression (RPKM = 1-2.5). The distribution of expression within these ranges were the same for speckle close and speckle far genes. The number of 100-kb regions analyzed are 8 regions each for high expression speckle close and far, 70 regions for medium expression speckle close and 28 for medium expression speckle far, and 194 for low expression speckle close and 62 for low expression speckle far. In the box plot, the center line represents the median, boxes show the interquartile range, whiskers show the range of values. B. U1 snRNA density is plotted for high (top), medium (middle), and low expression genes (bottom). C. U2 snRNA density is plotted for high (top), medium (middle), and low expression genes (bottom). D. U4 snRNA density is plotted for high (top), medium (middle), and low expression genes (bottom). E. U6 snRNA density is plotted for high (top), medium (middle), and low expression genes (bottom). (F-I): To ensure that splicing factor difference were not due to density of nascent transcription differences between speckle close and speckle far genes, we divided genes up based on transcription density ranges based on the number of nascent RNA reads from 5EU spanning each 100-kb bin. The number of 100-kb regions analyzed are 693 top 20% speckle close and 25 top 20% speckle far, 282 of 60–80% speckle close and 68 of 60–80% speckle far, 101 of 40–60% speckle close and 228 of 40–60% speckle far, 29 of 20–40% speckle close and 428 of 20–40% speckle far, and 7 of bottom 20% speckle close and 362 of bottom 20% speckle far. F. U1 snRNA density is plotted for top 20%, 60–80%, 40–60%, 20–40%, and bottom 20% of nascent transcription density. G. U2 snRNA density is plotted for top 20%, 60–80%, 40–60%, 20–40%, and bottom 20% of nascent transcription density. H. U4 snRNA density is plotted for top 20%, 60–80%, 40–60%, 20–40%, and bottom 20% of nascent transcription density. I. U6 snRNA density is plotted for top 20%, 60–80%, 40–60%, 20–40%, and bottom 20% of nascent transcription density.

Extended Data Fig. 3 snRNA density for junction matched genomic regions, genomic regions harboring genes of different lengths, and U1 AMT RAP-RNA enrichment for junction matched genomic regions.

A. An identical number of regions with an identical number of junctions (179 regions each for speckle close and speckle far regions)were randomly sampled to compare regions with equivalent junction density (See Methods). B. The expression levels were matched to compare the regions in A with similar mean expression per 100-kb bin. C. SPRITE speckle proximity score of filtered speckle close and speckle far regions analyzed in panel A. D. U1 snRNA density is plotted for junction and expression-controlled regions. E. U2 snRNA density is plotted for junction and expression-controlled regions. F. U4 snRNA density is plotted for junction and expression-controlled regions. G. U6 snRNA density is plotted for junction and expression-controlled regions. H. To ensure that splicing factor difference were not due to gene length differences between speckle close and speckle far genes, we divided genes up based on gene length ranges: longest genes (60th to 80th percentile), medium length range genes (40th to 60th percentile), shortest genes (bottom 20%). The distribution of length within these ranges were the same for speckle close and speckle far genes. For the regions with the longest genes, 53 speckle close and 84 speckle far 100-kb regions analyzes. For the regions with the medium length genes, 73 speckle close and 63 speckle far 100-kb regions analyzed. For the regions with the shortest genes, 178 speckle close and 102 speckle far 100-kb regions analyzed. In the box plot, the center line represents the median, boxes show the interquartile range, whiskers show the range of values. I. U1 snRNA density is plotted for longest (top), medium (middle), and shortest length genes (bottom). J. U2 snRNA density is plotted for longest (top), medium (middle), and shortest length genes (bottom). K. U4 snRNA density is plotted for longest (top), medium (middle), and shortest length genes (bottom). L. U6 snRNA density is plotted for longest (top), medium (middle), and shorted length genes (bottom). M. Density plot showing speckle proximity score (100-kb) for genomic regions enriched for U1 binding. N. U1 RAP RNA enrichment per junction (y-axis) versus number of exons per 100-kb genomic bin for speckle close and speckle far regions. Dotted lines are mean U1 enrichment values and error is SEM. Number of regions per point: n = 97, 91, 28, and 12 for speckle far regions exon number = 10, 20, 30 and 40, respectively; n = 18, 68, 70, and 47 for speckle close regions exon number = 10, 20, 30 and 40, respectively.

Extended Data Fig. 4 Higher splicing efficiency in speckle close regions across measurements, cell-types, and when comparing to genes of similar expression, length, and junction density to speckle far regions.

A. i. SPRITE speckle proximity score at 100-kb resolution (x axis) in mESCs and per cent spliced (from chromatin RNA-seq). 50 bins across all contact frequencies were taken and bins with speckle proximity scores between 0 and 200 are shown. Data are presented as mean values and bars represent 95% confidence interval. ii. SPRITE speckle proximity score at 100-kb resolution (x axis) in mESCs and per cent spliced (from SPRITE). 50 bins across all contact frequencies were taken and bins with speckle proximity scores between 0 and 200 are shown. Data are presented as mean values and bars represent 95% confidence interval. B. Schematic of 5EU labeling and nascent RNA sequencing pipeline. C. SPRITE speckle proximity score at 100-kb resolution (x axis) in mESCs and per cent spliced (from 5EU RNA-seq). 50 bins across all contact frequencies were taken and bins with speckle proximity scores between 0 and 200 are shown. Bars represent 95% confidence interval. Spearman r correlation = 0.95, p < 0.0001, P value is two-tailed. D. Correlation of splicing efficiency between previously published chromatin RNA-seq and newly generated 5EU RNA-seq (this paper; Spearman r correlation = 0.79, p < 0.0001), P value is two-tailed. (E-S) Splicing efficiency for speckle close and speckle far regions normalized for with genes that are: E. The top expressed genes (within 80–100% of expressed genes). 96 speckle close and 15 speckle far genes analzyed. F. Within 60–80% of expressed genes. 95 speckle close and 53 speckle far genes analyzed. G. Within 40–60% of expressed genes. 74 speckle close and 62 speckle far genes analyzed. H. Within 20–40% of expressed genes. 78 speckle close and 90 speckle far genes analyzed. I. The least expressed genes (0–20% of expressed genes). 51 speckle close and 173 speckle far genes analyzed. J. The longest genes (80–100% of genes lengths). 30 speckle close and 143 speckle far genes analyzed. K. 60–80% of gene lengths. 57 speckle close and 86 speckle far genes analyzed. L. 40–60% of gene lengths. 59 speckle close and 72 speckle far genes analyzed. M. 20–40% of gene lengths. 101 speckle close and 56 speckle far genes analyzed. N. The shortest genes (0–20% of gene lengths). 147 speckle close and 36 speckle far genes analyzed. O. 2 exons (single intron) per 100-kb region. 15 speckle close and 13 speckle far genes analyzed. P. 3–5 exons per 100-kb region. 50 speckle close and 119 speckle far genes analyzed. Q. 6–10 exons per 100-kb region. 51 speckle close and 202 speckle far genes analyzed. R. 11–15 exons per 100-kb region. 74 speckle close and 153 speckle far genes analyzed. S. 16–20 exons per 100-kb region. 95 speckle close and 78 speckle far genes analyzed. T. SPRITE speckle proximity score at 100-kb resolution (x axis) in H1-hESCs and per cent spliced within genomic bins from SPRITE (y axis) across 50 bins. Spearman r correlation = 0.70, p < 0.0001, P value is two-tailed. Median normalized speckle proximity scores are reported under each raw speckle hub contact value. Median value for H1 hESC = 7.0. U. SPRITE speckle proximity score at 100-kb resolution (x axis) in myocytes and per cent spliced (from nuclear RNA-seq) across 50 bins. Pearson r correlation = 0.64, p < 0.0001, P value is two-tailed. RD SPRITE data was not collected in myocytes for technical reasons. Median normalized speckle proximity scores are reported under each raw speckle hub contact value. Median value for mouse myocytes = 209. The range of speckle proximity scores vary between H1 hESC (1–20) and mouse myocytes (~75–400) due to the myocyte SPRITE data being sequenced more deeply.

Extended Data Fig. 5 Integrated reporter maintains endogenous speckle distances.

A. Representative images and zoom-ins of SRRM1 immunofluorescence combined with DNA FISH for the integrated reporter mini-gene. SRRM1 in magenta, reporter DNA in yellow and DAPI. Scale bar is 10 µm. n = 85 cells from 2 biological replicates. B. ECDF plots showing distance of DNA FISH spots of integrated location to the nearest nuclear speckle in the integrated cell lines (left) or distances computed from DNA seqFISH (right). C. Violin plots showing distance of DNA FISH spots of integrated location to the nearest nuclear speckle in the integrated cell lines (left) or distances computed from DNA seqFISH (right). Same data used as in 3C. Difference in means between speckle close and speckle far regions calculated for integrated loci and endogenous loci are represented above the distributions. D. 2D FACS plots showing GFP splicing levels as a function of BFP transcription levels between speckle close and speckle far integrated cell lines.

Extended Data Fig. 6 SPRITE analysis of myocyte cells and comparison to mES cells.

A. Distribution of SPRITE cluster sizes for myocyte SPRITE. The percentage of reads was calculated for different SPRITE cluster sizes (1, 2–10, 11–100, 101–1000, and over 1001 reads) and reported as the percentage of total reads. Cluster size is defined as the number of reads with the same barcode. B. Alignment statistics. C. A summary of ligation efficiency statistics to confirm tags have successfully ligated to each DNA molecule. D. Mouse myocyte interchromosomal contacts on chromosomes 4, 8, 11. E. Speckle hubs in mouse myocytes highlighted in red on chromosome track. Genome wide distribution of SPRITE speckle proximity scores (100-kb resolution). Gene density track on bottom. F. Distribution of SPRITE speckle proximity scores (100-kb resolution) for normalized mES and myocyte cell SPRITE. G. Distribution of number of genomic regions categorized as speckle hubs in myocyte, ES cells, both, or neither. H. SPRITE speckle proximity score at 100-kb resolution for a 20-Mb region on chromosome 7 in mouse myocytes. Pol II-S2P ChIP-seq density at 1-kb resolution. I. Ser2-P Pol II density (x axis) and normalized speckle proximity score (100-kb resolution) for myocytes. Spearman correlation = 0.69; p < 0.0001, P value is two-tailed. Similar to previous observations in other cellular contexts, we observed that DNA regions located close to speckles correspond to genomic regions containing high-density of RNA Pol II in differentiated myocytes. J. ES cell speckle proximity score (light green) and skeletal muscle speckle proximity score (dark green) for genomic locus near MyoD1 (expressed in myocyte). ∆Pol II refers to difference in Ser2P-Pol II ChIP seq signal between mES cells and myocytes at 100-kb resolution, red is high in myocyte and blue high in ES.

Extended Data Fig. 7 pre-mRNA organization around nuclear speckles drives splicing efficiency.

(A-D) Whole cell imaging of each protein with SC35 immunofluorescence and overlay with nucleus outlined in white for: A. SRRM1. B. SRSF1. C. SRSF3. D. LBR. Scale bars are 10 µm. Experiment was performed three times. (E-H) GFP fluorescence (splicing levels) (y axis) versus BFP fluorescence intensity for constructs with MCP or without MCP for: E. SRRM1. F. SRSF1. G. SRSF3. H. LBR. I. Difference in GFP splicing levels between SRRM1 MCP and no MCP with a four-parameter nonlinear regression. J. Difference in GFP splicing levels between SRSF1 MCP and no MCP with a four-parameter nonlinear regression. K. Four parameter logistic nonlinear fits for SRRM1, SRSF1, SRSF3, and LBR. L. Whole cell imaging of ∆NS SRRM1 with SC35 immunofluorescence overlay. Scale bar is 10 µm. Experiment was performed three times. M. GFP fluorescence (splicing levels) (y axis) versus BFP fluorescence intensity for constructs with MCP or without MCP for ∆NS SRRM1.

Extended Data Fig. 8 Differential versus leveled intron architectures also display speckle dependent splicing efficiency.

A. Schematic of CORO1B (leveled) intron and mapped %GC content across intron and exon boundary. B. Schematic of FRG1 (differential) intron and mapped %GC content across intron and exon boundary. C. GFP levels (y axis) versus fluorescence intensity (levels) of BFP (x axis) (bottom) for three replicates of SRRM1+/− MCP co-transfected with CORO1B splicing reporter. D. GFP levels (y axis) versus fluorescence intensity (levels) of BFP (x axis) (bottom) for three replicates of LBR+/− MCP co-transfected with CORO1B splicing reporter. E. GFP levels (y axis) versus fluorescence intensity (levels) of BFP (x axis) (bottom) for three replicates of SRRM1+/− MCP co-transfected with FRG1 splicing reporter. F. GFP levels (y axis) versus fluorescence intensity (levels) of BFP (x axis) (bottom) for three replicates of LBR+/− MCP co-transfected with FRG1 splicing reporter.

Extended Data Fig. 9 Integrated model for how spliceosome activity and proximity to nuclear speckles impact kinetics of splicing.

There are two components impacting the kinetics of splicing – spliceosome concentration and spliceosome activity. (i) Proximity to nuclear speckles impacts the concentration of spliceosomes at a given pre-mRNA, such that genes that are close to speckles will have higher spliceosome concentration than genes that are far from speckles. (ii) In contrast, splice site strength is defined by the activity of the spliceosome at the splice site53. In this way, spliceosomes engaged at ‘strong’ splice sites would have higher activity while ‘weak’ splice sites would have lower activity. These two components would be expected to have different effects on the kinetics of splicing. Specifically, modulating activity (splice site strength) would be expected to impact the maximum output of the reaction. Conversely, modulating concentration (speckle proximity) would be expected to impact the efficiency of each reaction.

Supplementary information

Supplementary Figures

Reporting Summary

Supplementary Table 1

Speckle distances computed from seqFISH or TSA–seq and correlation with SPRITE.

Supplementary Table 2

Speckle distances computed from SPRITE and associated speckle hubs in mouse and human cells.

Supplementary Table 3

snRNA density, gene length and expression metrics for each genomic bin.

Supplementary Table 4

Splicing efficiency measured by chromatin RNA-seq, 5EU-seq and RD-SPRITE.

Supplementary Table 5

Differences in PolII and SPRITE speckle contact between different cell types.

Supplementary Table 6

DNA FISH probe sequences.

Supplementary Video 1

Co-localization of RNA FISH spots (cyan) and SRRM1 (red) speckles in 3D. MS2-containing reporter RNA preferentially co-localizes with nuclear speckles when co-expressed with SRRM1–MCP using RNA FISH coupled with fluorescence microscopy of mCherry (for SRRM1).

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

Bhat, P., Chow, A., Emert, B. et al. Genome organization around nuclear speckles drives mRNA splicing efficiency. Nature (2024). https://doi.org/10.1038/s41586-024-07429-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41586-024-07429-6

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