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:

Rbfox proteins regulate alternative mRNA splicing through evolutionarily conserved RNA bridges

Abstract

Alternative splicing (AS) enables programmed diversity of gene expression across tissues and development. We show here that binding in distal intronic regions (>500 nucleotides (nt) from any exon) by Rbfox splicing factors important in development is extensive and is an active mode of splicing regulation. Similarly to exon-proximal sites, distal sites contain evolutionarily conserved GCATG sequences and are associated with AS activation and repression upon modulation of Rbfox abundance in human and mouse experimental systems. As a proof of principle, we validated the activity of two specific Rbfox enhancers in KIF21A and ENAH distal introns and showed that a conserved long-range RNA-RNA base-pairing interaction (an RNA bridge) is necessary for Rbfox-mediated exon inclusion in the ENAH gene. Thus we demonstrate a previously unknown RNA-mediated mechanism for AS control by distally bound RNA-binding proteins.

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

Figure 1: Characteristics of Rbfox binding in distal intronic regions.
Figure 2: The Rbfox binding motif TGCATG is the most enriched hexamer in conserved regions in distal intronic space around alternatively spliced exons.
Figure 3: Both proximal and distal Rbfox motifs regulate splicing.
Figure 4: Distal conserved regions containing Rbfox sites control splicing of upstream alternative exons.
Figure 5: An RNA bridge between ENAH E11a and a conserved distal RBFOX site is necessary for exon inclusion.

Similar content being viewed by others

Accession codes

Primary accessions

Sequence Read Archive

References

  1. Black, D.L. Mechanisms of alternative pre-messenger RNA splicing. Annu. Rev. Biochem. 72, 291–336 (2003).

    Article  CAS  PubMed  Google Scholar 

  2. Matlin, A.J., Clark, F. & Smith, C.W. Understanding alternative splicing: towards a cellular code. Nat. Rev. Mol. Cell Biol. 6, 386–398 (2005).

    Article  CAS  PubMed  Google Scholar 

  3. Wang, Z. & Burge, C.B. Splicing regulation: from a parts list of regulatory elements to an integrated splicing code. RNA 14, 802–813 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. 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 

  5. Yeo, G.W. et al. An RNA code for the FOX2 splicing regulator revealed by mapping RNA-protein interactions in stem cells. Nat. Struct. Mol. Biol. 16, 130–137 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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 

  7. Polymenidou, M. et al. Long pre-mRNA depletion and RNA missplicing contribute to neuronal vulnerability from loss of TDP-43. Nat. Neurosci. 14, 459–468 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tollervey, J.R. et al. Characterizing the RNA targets and position-dependent splicing regulation by TDP-43. Nat. Neurosci. 14, 452–458 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lagier-Tourenne, C. et al. Divergent roles of ALS-linked proteins FUS/TLS and TDP-43 intersect in processing long pre-mRNAs. Nat. Neurosci. 15, 1488–1497 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Huelga, S.C. et al. Integrative genome-wide analysis reveals cooperative regulation of alternative splicing by hnRNP proteins. Cell Rep. 1, 167–178 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wilbert, M.L. et al. LIN28 binds messenger RNAs at GGAGA motifs and regulates splicing factor abundance. Mol. Cell 48, 195–206 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zarnack, K. et al. Direct competition between hnRNP C and U2AF65 protects the transcriptome from the exonization of Alu elements. Cell 152, 453–466 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. König, 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 

  14. Hoell, J.I. et al. RNA targets of wild-type and mutant FET family proteins. Nat. Struct. Mol. Biol. 18, 1428–1431 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  16. Yeo, G.W., Van Nostrand, E.L. & Liang, T.Y. Discovery and analysis of evolutionarily conserved intronic splicing regulatory elements. PLoS Genet. 3, e85 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Xue, Y. et al. Genome-wide analysis of PTB-RNA interactions reveals a strategy used by the general splicing repressor to modulate exon inclusion or skipping. Mol. Cell 36, 996–1006 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Zhang, C. et al. Defining the regulatory network of the tissue-specific splicing factors Fox-1 and Fox-2. Genes Dev. 22, 2550–2563 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Barash, Y. et al. Deciphering the splicing code. Nature 465, 53–59 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Guo, N. & Kawamoto, S. An intronic downstream enhancer promotes 3′ splice site usage of a neural cell-specific exon. J. Biol. Chem. 275, 33641–33649 (2000).

    Article  CAS  PubMed  Google Scholar 

  21. Lapuk, A. et al. Exon-level microarray analyses identify alternative splicing programs in breast cancer. Mol. Cancer Res. 8, 961–974 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Coté, J., Dupuis, S., Jiang, Z. & Wu, J.Y. Caspase-2 pre-mRNA alternative splicing: Identification of an intronic element containing a decoy 3′ acceptor site. Proc. Natl. Acad. Sci. USA 98, 938–943 (2001).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Lim, L.P. & Sharp, P.A. Alternative splicing of the fibronectin EIIIB exon depends on specific TGCATG repeats. Mol. Cell Biol. 18, 3900–3906 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Baraniak, A.P., Lasda, E.L., Wagner, E.J. & Garcia-Blanco, M.A. A stem structure in fibroblast growth factor receptor 2 transcripts mediates cell-type-specific splicing by approximating intronic control elements. Mol. Cell Biol. 23, 9327–9337 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Dirksen, W.P., Mohamed, S.A. & Fisher, S.A. Splicing of a myosin phosphatase targeting subunit 1 alternative exon is regulated by intronic cis-elements and a novel bipartite exonic enhancer/silencer element. J. Biol. Chem. 278, 9722–9732 (2003).

    Article  CAS  PubMed  Google Scholar 

  26. Lenasi, T., Peterlin, B.M. & Dovc, P. Distal regulation of alternative splicing by splicing enhancer in equine beta-casein intron 1. RNA 12, 498–507 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kim, K.K., Kim, Y.C., Adelstein, R.S. & Kawamoto, S. Fox-3 and PSF interact to activate neural cell-specific alternative splicing. Nucleic Acids Res. 39, 3064–3078 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Gehman, L.T. et al. The splicing regulator Rbfox2 is required for both cerebellar development and mature motor function. Genes Dev. 26, 445–460 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. 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 

  30. Yeo, G.W. et al. Alternative splicing events identified in human embryonic stem cells and neural progenitors. PLoS Comput. Biol. 3, e196 (2007).

    Article  PubMed Central  CAS  Google Scholar 

  31. Gallagher, T.L. et al. Rbfox-regulated alternative splicing is critical for zebrafish cardiac and skeletal muscle functions. Dev. Biol. 359, 251–261 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Venables, J.P. et al. RBFOX2 is an important regulator of mesenchymal tissue-specific splicing in both normal and cancer tissues. Mol. Cell Biol. 33, 396–405 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Minovitsky, S., Gee, S.L., Schokrpur, S., Dubchak, I. & Conboy, J.G. The splicing regulatory element, UGCAUG, is phylogenetically and spatially conserved in introns that flank tissue-specific alternative exons. Nucleic Acids Res. 33, 714–724 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Shibata, H., Huynh, D.P. & Pulst, S.M. A novel protein with RNA-binding motifs interacts with ataxin-2. Hum. Mol. Genet. 9, 1303–1313 (2000).

    Article  CAS  PubMed  Google Scholar 

  35. Lim, J. et al. A protein-protein interaction network for human inherited ataxias and disorders of Purkinje cell degeneration. Cell 125, 801–814 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Bhalla, K. et al. The de novo chromosome 16 translocations of two patients with abnormal phenotypes (mental retardation and epilepsy) disrupt the A2BP1 gene. J. Hum. Genet. 49, 308–311 (2004).

    Article  PubMed  Google Scholar 

  37. Martin, C.L. et al. Cytogenetic and molecular characterization of A2BP1/FOX1 as a candidate gene for autism. Am. J. Med. Genet. B. Neuropsychiatr. Genet. 144B, 869–876 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Sebat, J. et al. Strong association of de novo copy number mutations with autism. Science 316, 445–449 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Davis, L.K. et al. Rare inherited A2BP1 deletion in a proband with autism and developmental hemiparesis. Am. J. Med. Genet. A. 158A, 1654–1661 (2012).

    Article  CAS  PubMed  Google Scholar 

  40. Pistoni, M. et al. Rbfox1 downregulation and altered calpain 3 splicing by FRG1 in a mouse model of facioscapulohumeral muscular dystrophy (FSHD). PLoS Genet. 9, e1003186 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sato, D. et al. SHANK1 deletions in males with autism spectrum disorder. Am. J. Hum. Genet. 90, 879–887 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Johansson, J.U. et al. An ancient duplication of exon 5 in the Snap25 gene is required for complex neuronal development/function. PLoS Genet. 4, e1000278 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Xie, J. & McCobb, D.P. Control of alternative splicing of potassium channels by stress hormones. Science 280, 443–446 (1998).

    Article  CAS  PubMed  Google Scholar 

  44. Damianov, A. & Black, D.L. Autoregulation of Fox protein expression to produce dominant negative splicing factors. RNA 16, 405–416 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Yeo, G.W., Nostrand, E.L. & Liang, T.Y. Discovery and analysis of evolutionarily conserved intronic splicing regulatory elements. PLoS Genet. 3, e85 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Maher, B. ENCODE: the human encyclopaedia. Nature 489, 46–48 (2012).

    Article  PubMed  Google Scholar 

  47. Salemi, M. et al. KIF21A mRNA expression in patients with Down syndrome. Neurol. Sci. 34, 569–571 (2013).

    Article  PubMed  Google Scholar 

  48. Heidary, G., Engle, E.C. & Hunter, D.G. Congenital fibrosis of the extraocular muscles. Semin. Ophthalmol. 23, 3–8 (2008).

    Article  PubMed  Google Scholar 

  49. Parra, M.K., Gee, S., Mohandas, N. & Conboy, J.G. Efficient in vivo manipulation of alternative pre-mRNA splicing events using antisense morpholinos in mice. J. Biol. Chem. 286, 6033–6039 (2011).

    Article  CAS  PubMed  Google Scholar 

  50. Warzecha, C.C. et al. An ESRP-regulated splicing programme is abrogated during the epithelial-mesenchymal transition. EMBO J. 29, 3286–3300 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Dittmar, K.A. et al. Genome-wide determination of a broad ESRP-regulated posttranscriptional network by high-throughput sequencing. Mol. Cell Biol. 32, 1468–1482 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Goguel, V. & Rosbash, M. Splice site choice and splicing efficiency are positively influenced by pre-mRNA intramolecular base pairing in yeast. Cell 72, 893–901 (1993).

    Article  CAS  PubMed  Google Scholar 

  53. Plass, M., Codony-Servat, C., Ferreira, P.G., Vilardell, J. & Eyras, E. RNA secondary structure mediates alternative 3′ss selection in Saccharomyces cerevisiae. RNA 18, 1103–1115 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Rogic, S. et al. Correlation between the secondary structure of pre-mRNA introns and the efficiency of splicing in Saccharomyces cerevisiae. BMC Genomics 9, 355 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Raker, V.A., Mironov, A.A., Gelfand, M.S. & Pervouchine, D.D. Modulation of alternative splicing by long-range RNA structures in Drosophila. Nucleic Acids Res. 37, 4533–4544 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kreahling, J.M. & Graveley, B.R. The iStem, a long-range RNA secondary structure element required for efficient exon inclusion in the Drosophila Dscam pre-mRNA. Mol. Cell Biol. 25, 10251–10260 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Pervouchine, D.D. et al. Evidence for widespread association of mammalian splicing and conserved long-range RNA structures. RNA 18, 1–15 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Nasim, F.U., Hutchison, S., Cordeau, M. & Chabot, B. High-affinity hnRNP A1 binding sites and duplex-forming inverted repeats have similar effects on 5′ splice site selection in support of a common looping out and repression mechanism. RNA 8, 1078–1089 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. McManus, C.J. & Graveley, B.R. RNA structure and the mechanisms of alternative splicing. Curr. Opin. Genet. Dev. 21, 373–379 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Warf, M.B., Diegel, J.V., von Hippel, P.H. & Berglund, J.A. The protein factors MBNL1 and U2AF65 bind alternative RNA structures to regulate splicing. Proc. Natl. Acad. Sci. USA 106, 9203–9208 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Wu, T.D. & Nacu, S. Fast and SNP-tolerant detection of complex variants and splicing in short reads. Bioinformatics 26, 873–881 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    PubMed  PubMed Central  Google Scholar 

  64. Zisoulis, D.G. et al. Comprehensive discovery of endogenous Argonaute binding sites in Caenorhabditis elegans. Nat. Struct. Mol. Biol. 17, 173–179 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Dale, R.K., Pedersen, B.S. & Quinlan, A.R. Pybedtools: a flexible Python library for manipulating genomic datasets and annotations. Bioinformatics 27, 3423–3424 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Quinlan, A.R. & Hall, I.M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Smit, A., Hubley, R. & Green, P. RepeatMasker Open-3.0. (1996–2010).

  68. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Underwood, J.G., Boutz, P.L., Dougherty, J.D., Stoilov, P. & Black, D.L. Homologues of the Caenorhabditis elegans Fox-1 protein are neuronal splicing regulators in mammals. Mol. Cell Biol. 25, 10005–10016 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Parkhomchuk, D. et al. Transcriptome analysis by strand-specific sequencing of complementary DNA. Nucleic Acids Res. 37, e123 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Morcos, P.A., Li, Y. & Jiang, S. Vivo-Morpholinos: a non-peptide transporter delivers morpholinos into a wide array of mouse tissues. Biotechniques 45, 613–623 (2008).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank A. Pasquinelli, N. Chi, K. Willert and L. Goldstein and members of the Yeo, Conboy and Goldstein labs for critical reading of the manuscript. M.T.L. is supported as a National Science Foundation GK12 Fellow. This work was supported by grants from the National Institute of Health to G.W.Y. (U54 HG007005, R01 HG004659, R01 GM084317 and R01 NS075449) and to J.G.C. (HL045182 and DK094699) and partially supported by grants to J.W.G. (CA112970 and CA126551). J.G.C. also acknowledges support from DK032094. This work was also supported by the Director, Office of Science, and Office of Biological & Environmental Research of the US Department of Energy under Contract No. DE-AC02-05CH1123. D.L.B. and L.T.G. were supported by US National Institutes of Health grant RO1 GM49662 to D.L.B. D.L.B. is an Investigator of the Howard Hughes Medical Institute. M.T.L. and G.W.Y. are grateful for a gift from P. Yang at Genentech that supported M.T.L. G.W.Y. is supported as an Alfred P. Sloan Research Fellow.

Author information

Authors and Affiliations

Authors

Contributions

M.T.L. and G.A.P. conducted the bioinformatics analyses. D.G., H.M., J.A., S.G., M.P., T.Y.L., T.J.S., S.H. and K.B.M. conducted biological experiments. L.T.G. and D.L.B. generated the Rbfox mutant mice and isolated brain RNA. J.W.G., J.G.C. and G.W.Y. designed the study. M.T.L., D.G., J.G.C. and G.W.Y. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to John G Conboy or Gene W Yeo.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 CLIP-seq to identify Rbfox binding sites, motifs and gene ontology analyses.

(a) Autoradiograph of Rbfox protein-RNA complexes from mouse brain immunoprecipitated with Rbfox-specific antibodies and trimmed with optimized unit (U). concentrations of micrococcal nuclease (MNase). (b) Venn diagram showing the number of genes bound by Rbfox1 and Rbfox2 in mouse brain. (c) List of the top three motifs significantly enriched in the Rbfox1 CLIP compared to appropriate background controls. Similar results were found for Rbfox2 (not shown). (d) Box-plots of hexamer Z-scores, comparing Rbfox1 CLIP clusters to randomly located clusters in each genic region. GCAUG and UGCAUG, the known Rbfox motifs, and a GU-rich 6-mer were enriched in all genic regions except 5′ UTRs. (e) Venn diagrams showing the number of Rbfox1 (red) and Rbfox2 (blue) CLIP-seq clusters that overlap (yellow) each other by ≥ 1nt (“All clusters”). Clusters are then restricted to those within 50 nt of a GCAUG motif in mouse (“GCAUG in mouse”), or where the GCAUG motif is conserved (“Conserved GCAUG”) at increasing branch-length (BL) scores (the higher the score, the more conserved the GCAUG sequence across multiple genomes). Analyses of proximal (“PI”) and distal intronic (“DI”) clusters are displayed on the left and right columns. (f) Bar plots represent the fraction of clusters that overlap relative to the Rbfox1 (red) or Rbfox2 (blue) clusters corresponding to the different restrictions in (e). (g) Dendrogram showing hierarchical clustering with Euclidean distance of statistically significant gene ontology terms (in at least one experimental condition) using negative log10 P values that measure the enrichment of genes within the ontology terms for various experimental conditions.

Supplementary Figure 2 Genome browser views of selected alternatively spliced genes containing distal RBFOX binding sites.

(a-e) Exons and introns are indicated in blue (UCSC genes) or red (Ensembl Genes) with thick lines representing exons and thin lines overlaid with arrows representing introns. The direction of the arrows denotes the direction of transcription. Read density from Rbfox1 and Rbfox2 CLIP-seq (orange and green tracks respectively) on several genes is shown with positive and negative values corresponding to the direction of transcription of aligned reads. Shank1 (a), Snap25 (b), Kcnma1 (c), Rbfox1 (d) and Rbfox2 (e) possess alternatively spliced exons (red arrows). PhastCons evolutionary conservation scores for placental mammals are displayed at the bottom of each panel in dark green. Genomic GCATG sites are indicated above the conservation track in black. Distal and proximal highly conserved regions are designated along the top with purple and green filled rectangles, respectively. Scale bars at the top define the size of region displayed.

Supplementary Figure 3 Discovery and characterization of conserved regions.

Contiguous regions within the human transcriptome were divided into three categories based on their degree of evolutionary conservation as determined by phastCons scores, S. Conserved regions that overlap known repetitive elements and transcription factor binding sites were removed. Pie charts illustrate the distribution of the three categories of lowly (L), moderately (M) and highly (H) conserved regions of approximately similar lengths (box-plots on the right) within different genic regions (5′UTR, exon, proximal intron, distal intron and 3′UTR) in protein-coding genes. Bar charts show the fraction of total nucleotides in each genic region covered by a highly-conserved region. (b) Results from a de novo motif search using HOMER1 for enriched hexamer motifs is shown for distal and proximal highly-conserved regions from (a). Up to 10 motifs are shown with their associated P values indicated to the right.

Supplementary Figure 4 Identification of Rbfox-dependent expression and splicing changes by RNA-seq.

(a, b) Scatter plots of RPKM (reads per kilobase mapped in exons per million reads) values between Rbfox1 and Rbfox2 knockout (KO; y-axis) compared to wildtype (WT; x-axis) sibling pairs showed that 46 and 6 genes are significantly up-regulated (red points), and 11 and 9 genes are down-regulated (green points) upon loss of Rbfox1 and Rbfox2 in mouse brain, respectively (P < 0.001). (c–e) Scatter plots of RPKM values comparing ectopic expression (EE) of RBFOX1, RBFOX2 and RBFOX3 (y-axes) to a plasmid control (x-axis). (f) Scatter plots of percent-spliced-in (ψ) values of cassette exons comparing Rbfox1 and Rbfox2 KO (y-axes) to WT (x-axes); and ectopic expression (EE) of RBFOX1, RBFOX2 and RBFOX3 to plasmid control. Mouse exons that are alternatively spliced (|Δψ| ≥ 5%) upon loss of Rbfox1 (in red), Rbfox2 (in blue) and either (orange) are marked on top two plots. Human exons that are alternatively spliced upon EE of RBFOX1 (in red), RBFOX2 (in blue) and RBFOX3 (in green) are marked on the bottom three plots. A linear fit of the data (black line) and associated R2 value are shown. (g) Venn diagrams show the number of cassette exon splicing events that are in common upon loss of Rbfox1 and Rbfox2 in mouse (top) and upon ectopic expression of RBFOX proteins in human 293T cells (bottom), considering exons with |Δψ| ≥ 5% (any change, left), Δψ ≥ 5% (included, right top) and Δψ ≤ −5% (excluded, right bottom). RBFOX1-, RBFOX2- and RBFOX3-specific events are colored red, blue and green, with orange and dark green representing shared events. (h) A scatter plot shows Δψ values from RT-PCR2,3 (x-axis) and RNA-seq-derived RPKM (y-axis) measurements of exon inclusion for manually validated cassette splicing exon events in mouse brain for Rbfox1 (red) and Rbfox2 (blue). A linear fit to each set of points and associated R2 value are shown. (i) Bar-plots show a comparison between Δψ values using RT-PCR or RNA-seq. RT-PCR values were obtained from refs. 2,3.

Supplementary Figure 5 Distal association of Rbfox sites with Rbfox-dependent alternatively spliced exons.

(a, b) The fractions of alternatively spliced exons compared to unchanged that have GCATG motifs (at increasing levels of conservation) and CLIP-defined Rbfox1 and Rbfox2 sites either upstream or downstream, for both distal (“DI”) and proximal (“PI”) regions, are represented as vertical bars. Rbfox1 and Rbfox2 knockout (KO) mice compared to sibling WT pair are represented in (a). Ectopic expression (EE) of RBFOX1, RBFOX2 and RBFOX3 are represented in (B). The fractions are represented on the log-scale (y-axis). Blue bars represent exons with RNA-seq evidence for inclusion (Δψ > 5%); goldenrod bars represent exons with RNA-seq evidence for exclusion (Δψ < −5%); grey bars represent exons with no change by RNA-seq (−2% < Δψ < 2%). Ties with “*” symbols indicate statistically significant differences when categories of exons are compared (*P < 0.05, **P < 0.001; Fisher's exact test). The maximal change for each exon was used when combining evidence from all experiments, as represented by their unions (U; y-axis) (the final rows for (a) and (b)). (c) The cumulative distributions of Δψ values for exons with >1 GCAUG motif or CLIP-defined Rbfox binding site within either upstream or downstream, for both distal (“DI”) and proximal (“PI”) regions were compared to exons with no flanking conserved GCATG motifs. P values by two-sample Kolgomorov-Smirnov tests are indicated when statistically significant (P < 0.05).

Supplementary Figure 6 Un-cropped gel images.

Un-cropped gel images are shown for RT-PCR and biotin pull-down assays. These include slow-migrating bands, suspected heteroduplexes, which are excluded from main figures.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 (PDF 12812 kb)

Supplementary Table 1

CLIP-seq library statistics (XLS 22 kb)

Supplementary Table 2

Gene ontology results (XLS 4488 kb)

Supplementary Table 3

Gene expression analysis (XLS 17451 kb)

Supplementary Table 4

Splicing analysis (XLS 15717 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lovci, M., Ghanem, D., Marr, H. et al. Rbfox proteins regulate alternative mRNA splicing through evolutionarily conserved RNA bridges. Nat Struct Mol Biol 20, 1434–1442 (2013). https://doi.org/10.1038/nsmb.2699

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2699

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