SRSF2 mutations drive oncogenesis by activating a global program of aberrant alternative splicing in hematopoietic cells

Yang Liang, Toma Tebaldi, Kai Rejeski, Poorval Joshi, Giovanni Stefani, Ashley Taylor, Yuanbin Song, Radovan Vasic, Jamie Maziarz, Kunthavai Balasubramanian, Anastasia Ardasheva, Alicia Ding, Alessandro Quattrone, Stephanie Halene, Yang Liang, Toma Tebaldi, Kai Rejeski, Poorval Joshi, Giovanni Stefani, Ashley Taylor, Yuanbin Song, Radovan Vasic, Jamie Maziarz, Kunthavai Balasubramanian, Anastasia Ardasheva, Alicia Ding, Alessandro Quattrone, Stephanie Halene

Abstract

Recurrent mutations in the splicing factor SRSF2 are associated with poor clinical outcomes in myelodysplastic syndromes (MDS). Their high frequency suggests these mutations drive oncogenesis, yet the molecular explanation for this process is unclear. SRSF2 mutations could directly affect pre-mRNA splicing of a vital gene product; alternatively, a whole network of gene products could be affected. Here we determine how SRSF2 mutations globally affect RNA binding and splicing in vivo using HITS-CLIP. Remarkably, the majority of differential binding events do not translate into alternative splicing of exons with SRSF2P95H binding sites. Alternative splice alterations appear to be dominated by indirect effects. Importantly, SRSF2P95H targets are enriched in RNA processing and splicing genes, including several members of the hnRNP and SR families of proteins, suggesting a "splicing-cascade" phenotype wherein mutation of a single splicing factor leads to widespread modifications in multiple RNA processing and splicing proteins. We show that splice alteration of HNRNPA2B1, a splicing factor differentially bound and spliced by SRSF2P95H, impairs hematopoietic differentiation in vivo. Our data suggests a model whereby the recurrent mutations in splicing factors set off a cascade of gene regulatory events that together affect hematopoiesis and drive cancer.

Conflict of interest statement

DISCLOSURE DECLARATION

The authors declare no conflict of interest

Figures

Figure 1. The SRSF2 P95H mutation alters…
Figure 1. The SRSF2 P95H mutation alters SRSF2 in vivo RNA interactome
(a) Overview of the HITS-CLIP procedure. Top: generation of lentiviral vector constructs expressing C-terminally Flag-tagged SRSF2 WT and P95H in a doxycycline inducible manner. Center: HITS-CLIP key experimental steps. Bottom: computational identification of differentially bound regions, preferentially bound by WT (in red) or by P95H (in cyan) SRSF2. (b) Dose dependent inducible expression of Flag-tagged SRSF2 (WT and P95H). (c) Colony Forming Unit Assay for control CD34+ cells (mCherry), and cells with transient induction of SRSF2 WT or P95H. Left panel: total number of colonies. Right panel: composition of colonies (mean values + SEM). Total colony numbers and colony type percentages were compared between WT and P95H by two-tailed t-test (*P<0.05, **P<0.01, ***P<0.001). (d) Percentage of HITS-CLIP reads aligned to exonic, intronic and intergenic regions. In the left panel, percentages were scaled using total region lengths as a normalizing factor. (e) Percentage of HITS-CLIP crosslinking induced deletions mapping to each RNA nucleotide. (f) Volcano plot displaying fold changes and false discovery rate values for each HITS-CLIP binding site. Significant differentially bound regions are highlighted in red and cyan (preferentially bound by WT or P95H SRSF2 respectively). (g) Venn diagram of genes with at least one SRSF2 differential binding site. (h) Percentage of SRSF2 differential binding sites located within 5′UTR, 3′UTR, and CDS regions of protein coding genes.
Figure 2. The SRSF2 P95H mutation alters…
Figure 2. The SRSF2 P95H mutation alters SRSF2 in vivo RNA motif specificity
(a) Top enriched motifs for WT and P95H SRSF2 binding sites, identified by discriminative analysis of kmer composition. Corresponding p-values are displayed on top of each logo. (b) Relative enrichment of the SSNG (S=C/G, N=C/G/A/U) RNA consensus motifs in RNA regions preferentially bound by WT versus P95H SRSF2. The number of motif occurrences and differential enrichment p-values are displayed for each bar.
Figure 3. Differentially bound SRSF2 targets are…
Figure 3. Differentially bound SRSF2 targets are enriched in RNA binding and splicing genes
(a) Functional annotation enrichment analysis of differentially bound transcripts by WT and P95H SRSF2. The number of genes belonging to each category is displayed. (b) Protein-protein interaction network of SRSF2 RNA interactors associated with splicing. The size of each node is proportional to the number of differential SRSF2 binding sites: WT (in red), P95H (in cyan) or both (in violet).
Figure 4. SRSF2 P95H mutations promote alternative…
Figure 4. SRSF2 P95H mutations promote alternative splicing with inclusion of CCNG rich exons and enrichment in RNA binding and splicing genes
(a) Determination of differential alternative splice events via rMATS analysis in HEL cells engineered to express SRSF2 WT vs P95H. Left panel: five classes of alternative splicing events were considered: cassette exon (CE), alternative 5′ splice site (A5SS), alternative 3′ splice site (A3SS), mutually exclusive exons (MXE) and retained intron (RI). Right panel: the number of significant events with more inclusion in WT or P95H cells is displayed. (b) Relative enrichment of the SSNG (S=C/G, N=C/G/A/T) RNA consensus motifs in cassette exons preferentially spliced in WT vs preferentially spliced in P95H SRSF2 expressing cells. The number of motif occurrences and enrichment p-values are displayed for each bar. (c) Overlap between genes with differential binding and differential splicing in HEL cells expressing either WT or P95H SRSF2. The significance of the overlap is displayed. (d) Functional annotation enrichment analysis of differentially spliced genes in WT vs P95H HEL cells. The number of genes falling into each category is indicated beside each bar.
Figure 5. SRSF2 mutations results in differential…
Figure 5. SRSF2 mutations results in differential binding and splicing of HNRNP proteins
(a–c) Differential binding and splicing in HNRNP proteins is shown for HNRNPA2B1 (a), HNRNPH1 (b), and HNRNPM (c). Left panels: transcript maps showing WT (red) and P95H (cyan) SRSF2 binding profiles. The maps display mean normalized HITS-CLIP signal with nucleotide resolution. Standard errors for each position are shown as ribbons under mean lines. Crosslinking-induced deletions are marked in black. Exon boundaries are represented as vertical dotted lines. Differential interaction sites are highlighted on the transcript. Center panels: RT-PCR capturing differentially bound exons was performed in 3–4 replicates in HEL cells with or without doxycycline induction of SRSF2 WT or P95H expression, and with or without knockdown of endogenous SRSF2, and in human fetal liver CD34+ cells transduced with empty, SRSF2 WT or SRSF2 P95H expressing lentivirus. Right panels: primary patient derived samples - alternative splice events were quantified in normal CD34+ (n=2), WT MDS/AML (n=6), and MUT MDS/AML (n=6) samples. The magnitude of the alternative splice event in % was calculated as ratio of alternative splice event to total expression. Considered bands are marked by an asterisk (*). Predicted band sizes and transcript sizes are indicated to the right of the gel. (d) Direct differential splicing of the HNRNPA2B1 exon 9 by WT vs P95H SRSF2 verified by a minigene splicing assay. RG6-HNRNPA2B1 plasmid was co-transfected with empty vector, SRSF2 WT, SRSF2 P95H/L/R, and control or SRSF2 siRNA. Alternative splicing of exon 9 was determined via semi-quantitative PCR by measuring the ratio of alternative exon exclusion over total (exclusion+inclusion) band intensities (n=3). (e) Colony Forming Unit Assay for control cells (siNTC), cells silenced for HNRNPM or HNRNPH1, cells induced to splice out HNRNPA2B1 exon 9, and cells treated for all three modifications together (siALL). The same number of cells was plated in all experiments. The total number and the composition of colonies is displayed. Total colony numbers and colony type percentages were compared to the control (siNTC). Standard errors (SE) for colony numbers are displayed. In panels (a–e), significance values were determined by one way ANOVA with Sidák Post Hoc Test (*P<0.05, **P<0.01, ***P<0.001).

References

    1. Yoshida K, Sanada M, Shiraishi Y, Nowak D, Nagata Y, Yamamoto R, et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature. 2011;478:64–9.
    1. Meggendorfer M, Roller A, Haferlach T, Eder C, Dicker F, Grossmann V, et al. SRSF2 mutations in 275 cases with chronic myelomonocytic leukemia (CMML) Blood. 2012;120:3080–8.
    1. Papaemmanuil E, Gerstung M, Malcovati L, Tauro S, Gundem G, Van Loo P, et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood. 2013;122:3616–3627.
    1. Lee SC-W, Dvinge H, Kim E, Cho H, Micol J-B, Chung YR, et al. Modulation of splicing catalysis for therapeutic targeting of leukemia with mutations in genes encoding spliceosomal proteins. Nat Med. 2016;22:672–678.
    1. Howard JM, Sanford JR. The RNAissance family: SR proteins as multifaceted regulators of gene expression. Wiley Interdiscip Rev RNA. 2015;6:93–110.
    1. Graveley BR, Maniatis T. Arginine/serine-rich domains of SR proteins can function as activators of pre-mRNA splicing. Mol Cell. 1998;1:765–71.
    1. Wu JY, Maniatis T. Specific interactions between proteins implicated in splice site selection and regulated alternative splicing. Cell. 1993;75:1061–70.
    1. Mayeda A, Screaton GR, Chandler SD, Fu XD, Krainer AR. Substrate specificities of SR proteins in constitutive splicing are determined by their RNA recognition motifs and composite pre-mRNA exonic elements. Mol Cell Biol. 1999;19:1853–63.
    1. Long JC, Caceres JF. The SR protein family of splicing factors: master regulators of gene expression. Biochem J. 2009;417:15–27.
    1. Kim E, Ilagan JO, Liang Y, Daubner GM, Lee SC-W, Ramakrishnan A, et al. SRSF2 Mutations Contribute to Myelodysplasia by Mutant-Specific Effects on Exon Recognition. Cancer Cell. 2015;27:617–30.
    1. Daubner GM, Cléry A, Jayne S, Stevenin J, Allain FH-T. A syn-anti conformational difference allows SRSF2 to recognize guanines and cytosines equally well. EMBO J. 2012;31:162–74.
    1. Zhang J, Lieu YK, Ali AM, Penson A, Reggio KS, Rabadan R, et al. Disease-associated mutation in SRSF2 misregulates splicing by altering RNA-binding affinities. Proc Natl Acad Sci U S A. 2015;112:E4726–34.
    1. Kon A, Yamazaki S, Nannya Y, Kataoka K, Ota Y, Nakagawa MM, et al. Physiological Srsf2 P95H expression causes impaired hematopoietic stem cell functions and aberrant RNA splicing in mice. Blood. 2018;131:621–635.
    1. Lin S, Coutinho-Mansfield G, Wang D, Pandit S, Fu X-D. The splicing factor SC35 has an active role in transcriptional elongation. Nat Struct Mol Biol. 2008;15:819–826.
    1. Müller-McNicoll M, Botti V, de Jesus Domingues AM, Brandl H, Schwich OD, Steiner MC, et al. SR proteins are NXF1 adaptors that link alternative RNA processing to mRNA export. Genes Dev. 2016;30:553–66.
    1. Änkö M-L, Müller-McNicoll M, Brandl H, Curk T, Gorup C, Henry I, et al. The RNA-binding landscapes of two SR proteins reveal unique functions and binding to diverse RNA classes. Genome Biol. 2012;13:R17.
    1. Änkö M-L. Regulation of gene expression programmes by serine–arginine rich splicing factors. Semin Cell Dev Biol. 2014;32:11–21.
    1. Liu HX, Chew SL, Cartegni L, Zhang MQ, Krainer AR. Exonic splicing enhancer motif recognized by human SC35 under splicing conditions. Mol Cell Biol. 2000;20:1063–71.
    1. Chandler SD, Mayeda A, Yeakley JM, Krainer AR, Fu XD. RNA splicing specificity determined by the coordinated action of RNA recognition motifs in SR proteins. Proc Natl Acad Sci U S A. 1997;94:3596–601.
    1. Pandit S, Zhou Y, Shiue L, Coutinho-Mansfield G, Li H, Qiu J, et al. Genome-wide analysis reveals SR protein cooperation and competition in regulated splicing. Mol Cell. 2013;50:223–35.
    1. Zhu J, Mayeda A, Krainer AR. Exon identity established through differential antagonism between exonic splicing silencer-bound hnRNP A1 and enhancer-bound SR proteins. Mol Cell. 2001;8:1351–61.
    1. Bradley T, Cook ME, Blanchette M. SR proteins control a complex network of RNA-processing events. RNA. 2015;21:75–92.
    1. Ule J, Jensen K, Mele A, Darnell RB. CLIP: a method for identifying protein-RNA interaction sites in living cells. Methods. 2005;37:376–86.
    1. Ule J, Jensen KB, Ruggiu M, Mele A, Ule A, Darnell RB. CLIP identifies Nova-regulated RNA networks in the brain. Science. 2003;302:1212–5.
    1. Stefani G, Chen X, Zhao H, Slack FJ. A novel mechanism of LIN-28 regulation of let-7 microRNA expression revealed by in vivo HITS-CLIP in C. elegans. RNA. 2015;21:985–96.
    1. Shen S, Park JW, Lu Z, Lin L, Henry MD, Wu YN, et al. rMATS: Robust and flexible detection of differential alternative splicing from replicate RNA-Seq data. Proc Natl Acad Sci. 2014;111:E5593–E5601.
    1. Sarma NJ, Takeda A, Yaseen NR. Colony forming cell (CFC) assay for human hematopoietic cells. J Vis Exp. 2010 doi: 10.3791/2195.
    1. Itzykson R, Kosmider O, Renneville A, Morabito M, Preudhomme C, Berthon C, et al. Clonal architecture of chronic myelomonocytic leukemias. Blood. 2013;121:2186–2198.
    1. Kishore S, Jaskiewicz L, Burger L, Hausser J, Khorshid M, Zavolan M. A quantitative analysis of CLIP methods for identifying binding sites of RNA-binding proteins. Nat Methods. 2011;8:559–64.
    1. Granneman S, Kudla G, Petfalski E, Tollervey D. Identification of protein binding sites on U3 snoRNA and pre-rRNA by UV cross-linking and high-throughput analysis of cDNAs. Proc Natl Acad Sci U S A. 2009;106:9613–8.
    1. Zhang C, Darnell RB. Mapping in vivo protein-RNA interactions at single-nucleotide resolution from HITS-CLIP data. Nat Biotechnol. 2011;29:607–14.
    1. Sugimoto Y, König J, Hussain S, Zupan B, Curk T, Frye M, et al. Analysis of CLIP and iCLIP methods for nucleotide-resolution studies of protein-RNA interactions. Genome Biol. 2012;13:R67.
    1. Sureau A, Gattoni R, Dooghe Y, Stévenin J, Soret J. SC35 autoregulates its expression by promoting splicing events that destabilize its mRNAs. EMBO J. 2001;20:1785–96.
    1. Dreumont N, Hardy S, Behm-Ansmant I, Kister L, Branlant C, Stévenin J, et al. Antagonistic factors control the unproductive splicing of SC35 terminal intron. Nucleic Acids Res. 2010;38:1353–66.
    1. Keene JD. RNA regulons: coordination of post-transcriptional events. Nat Rev Genet. 2007;8:533–543.
    1. Sanford JR, Wang X, Mort M, Vanduyn N, Cooper DN, Mooney SD, et al. Splicing factor SFRS1 recognizes a functionally diverse landscape of RNA transcripts. Genome Res. 2009;19:381–94.
    1. Ankö M-L, Neugebauer KM. RNA-protein interactions in vivo: global gets specific. Trends Biochem Sci. 2012;37:255–62.
    1. Guil S, Gattoni R, Carrascal M, Abián J, Stévenin J, Bach-Elias M. Roles of hnRNP A1, SR proteins, and p68 helicase in c-H-ras alternative splicing regulation. Mol Cell Biol. 2003;23:2927–41.
    1. Rooke N, Markovtsov V, Cagavi E, Black DL. Roles for SR proteins and hnRNP A1 in the regulation of c-src exon N1. Mol Cell Biol. 2003;23:1874–84.
    1. Expert-Bezançon A, Sureau A, Durosay P, Salesse R, Groeneveld H, Lecaer JP, et al. hnRNP A1 and the SR proteins ASF/SF2 and SC35 have antagonistic functions in splicing of beta-tropomyosin exon 6B. J Biol Chem. 2004;279:38249–59.
    1. Zahler AM, Damgaard CK, Kjems J, Caputi M. SC35 and heterogeneous nuclear ribonucleoprotein A/B proteins bind to a juxtaposed exonic splicing enhancer/exonic splicing silencer element to regulate HIV-1 tat exon 2 splicing. J Biol Chem. 2004;279:10077–84.
    1. Han J, Ding J-H, Byeon CW, Kim JH, Hertel KJ, Jeong S, et al. SR Proteins Induce Alternative Exon Skipping through Their Activities on the Flanking Constitutive Exons. Mol Cell Biol. 2011;31:793–802.
    1. Komeno Y, Huang Y-J, Qiu J, Lin L, Xu Y, Zhou Y, et al. SRSF2 Is Essential for Hematopoiesis, and Its Myelodysplastic Syndrome-Related Mutations Dysregulate Alternative Pre-mRNA Splicing. Mol Cell Biol. 2015;35:3071–82.
    1. Qiu J, Zhou B, Thol F, Zhou Y, Chen L, Shao C, et al. Distinct splicing signatures affect converged pathways in myelodysplastic syndrome patients carrying mutations in different splicing regulators. RNA. 2016;22:1535–49.
    1. Busch A, Hertel KJ. Evolution of SR protein and hnRNP splicing regulatory factors. Wiley Interdiscip Rev RNA. 2012;3:1–12.
    1. Han SP, Friend LR, Carson JH, Korza G, Barbarese E, Maggipinto M, et al. Differential subcellular distributions and trafficking functions of hnRNP A2/B1 spliceoforms. Traffic. 2010;11:886–898.
    1. McGlincy NJ, Tan L-Y, Paul N, Zavolan M, Lilley KS, Smith CWJ. Expression proteomics of UPF1 knockdown in HeLa cells reveals autoregulation of hnRNP A2/B1 mediated by alternative splicing resulting in nonsense-mediated mRNA decay. BMC Genomics. 2010;11:565.
    1. Huelga SC, Vu AQ, Arnold JD, Liang TY, Liu PP, Yan BY, et al. Integrative genome-wide analysis reveals cooperative regulation of alternative splicing by hnRNP proteins. Cell Rep. 2012;1:167–78.
    1. Bergeron D, Pal G, Beaulieu YB, Chabot B, Bachand F. Regulated Intron Retention and Nuclear Pre-mRNA Decay Contribute to PABPN1 Autoregulation. Mol Cell Biol. 2015;35:2503–17.
    1. Martinez FJ, Pratt GA, Van Nostrand EL, Batra R, Huelga SC, Kapeli K, et al. Protein-RNA Networks Regulated by Normal and ALS-Associated Mutant HNRNPA2B1 in the Nervous System. Neuron. 2016;92:780–795.
    1. Obeng EA, Chappell RJ, Seiler M, Chen MC, Campagna DR, Schmidt PJ, et al. Physiologic Expression of Sf3b1(K700E) Causes Impaired Erythropoiesis, Aberrant Splicing, and Sensitivity to Therapeutic Spliceosome Modulation. Cancer Cell. 2016;30:404–417.
    1. Shirai CL, Ley JN, White BS, Kim S, Tibbitts J, Shao J, et al. Mutant U2AF1 Expression Alters Hematopoiesis and Pre-mRNA Splicing In Vivo. Cancer Cell. 2015;27:631–43.
    1. Molliex A, Temirov J, Lee J, Coughlin M, Kanagaraj AP, Kim HJ, et al. Phase separation by low complexity domains promotes stress granule assembly and drives pathological fibrillization. Cell. 2015;163:123–33.
    1. Alarcón CR, Goodarzi H, Lee H, Liu X, Tavazoie S, Tavazoie SF. HNRNPA2B1 Is a Mediator of m(6)A-Dependent Nuclear RNA Processing Events. Cell. 2015;162:1299–308.
    1. Kataoka K, Nagata Y, Kitanaka A, Shiraishi Y, Shimamura T, Yasunaga J, et al. Integrated molecular analysis of adult T cell leukemia/lymphoma. Nat Genet. 2015;47:1304–1315.

Source: PubMed

3
Sottoscrivi