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.

  • Letter
  • Published:

Recurrent mTORC1-activating RRAGC mutations in follicular lymphoma

Subjects

A Corrigendum to this article was published on 27 May 2016

This article has been updated

Abstract

Follicular lymphoma is an incurable B cell malignancy1 characterized by the t(14;18) translocation and mutations affecting the epigenome2,3. Although frequent gene mutations in key signaling pathways, including JAK-STAT, NOTCH and NF-κB, have also been defined2,3,4,5,6,7, the spectrum of these mutations typically overlaps with that in the closely related diffuse large B cell lymphoma (DLBCL)6,7,8,9,10,11,12,13. Using a combination of discovery exome and extended targeted sequencing, we identified recurrent somatic mutations in RRAGC uniquely enriched in patients with follicular lymphoma (17%). More than half of the mutations preferentially co-occurred with mutations in ATP6V1B2 and ATP6AP1, which encode components of the vacuolar H+-ATP ATPase (V-ATPase) known to be necessary for amino acid−induced activation of mTORC1. The RagC variants increased raptor binding while rendering mTORC1 signaling resistant to amino acid deprivation. The activating nature of the RRAGC mutations, their existence in the dominant clone and their stability during disease progression support their potential as an excellent candidate for therapeutic targeting.

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: Identification of frequent RRAGC mutations in follicular lymphoma.
Figure 2: Frequent and co-occurring mutations in ATP6V1B2 and ATP6AP1.
Figure 3: Effects of RagC variants on mTORC1 signaling.
Figure 4: RagC mutants have altered nucleotide binding affinity.

Similar content being viewed by others

Change history

  • 12 January 2016

    In the version of this article initially published online, several funding sources were omitted from the Acknowledgments section. The error has been corrected for the print, PDF and HTML versions of this article.

References

  1. Swenson, W.T. et al. Improved survival of follicular lymphoma patients in the United States. J. Clin. Oncol. 23, 5019–5026 (2005).

    PubMed  Google Scholar 

  2. Okosun, J. et al. Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat. Genet. 46, 176–181 (2014).

    CAS  PubMed  Google Scholar 

  3. Pasqualucci, L. et al. Genetics of follicular lymphoma transformation. Cell Reports 6, 130–140 (2014).

    CAS  PubMed  Google Scholar 

  4. Karube, K. et al. Recurrent mutations of NOTCH genes in follicular lymphoma identify a distinctive subset of tumours. J. Pathol. 234, 423–430 (2014).

    CAS  PubMed  Google Scholar 

  5. Yildiz, M. et al. Activating STAT6 mutations in follicular lymphoma. Blood 125, 668–679 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Morin, R.D. et al. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nat. Genet. 42, 181–185 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Pasqualucci, L. et al. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature 471, 189–195 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Compagno, M. et al. Mutations of multiple genes cause deregulation of NF-κB in diffuse large B-cell lymphoma. Nature 459, 717–721 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Lenz, G. et al. Oncogenic CARD11 mutations in human diffuse large B cell lymphoma. Science 319, 1676–1679 (2008).

    CAS  PubMed  Google Scholar 

  10. Lohr, J.G. et al. Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing. Proc. Natl. Acad. Sci. USA 109, 3879–3884 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Ngo, V.N. et al. Oncogenically active MYD88 mutations in human lymphoma. Nature 470, 115–119 (2011).

    CAS  PubMed  Google Scholar 

  12. Morin, R.D. et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature 476, 298–303 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Pasqualucci, L. et al. Analysis of the coding genome of diffuse large B-cell lymphoma. Nat. Genet. 43, 830–837 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Bödör, C. et al. EZH2 mutations are frequent and represent an early event in follicular lymphoma. Blood 122, 3165–3168 (2013).

    PubMed  PubMed Central  Google Scholar 

  15. Green, M.R. et al. Hierarchy in somatic mutations arising during genomic evolution and progression of follicular lymphoma. Blood 121, 1604–1611 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Beà, S. et al. Landscape of somatic mutations and clonal evolution in mantle cell lymphoma. Proc. Natl. Acad. Sci. USA 110, 18250–18255 (2013).

    PubMed  PubMed Central  Google Scholar 

  17. Landau, D.A. et al. Evolution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell 152, 714–726 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Lohr, J.G. et al. Widespread genetic heterogeneity in multiple myeloma: implications for targeted therapy. Cancer Cell 25, 91–101 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Morin, R.D. et al. Mutational and structural analysis of diffuse large B-cell lymphoma using whole-genome sequencing. Blood 122, 1256–1265 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Schmitz, R. et al. Burkitt lymphoma pathogenesis and therapeutic targets from structural and functional genomics. Nature 490, 116–120 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    PubMed  PubMed Central  Google Scholar 

  22. Nakashima, N., Noguchi, E. & Nishimoto, T. Saccharomyces cerevisiae putative G protein, Gtr1p, which forms complexes with itself and a novel protein designated as Gtr2p, negatively regulates the Ran/Gsp1p G protein cycle through Gtr2p. Genetics 152, 853–867 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Sekiguchi, T., Hirose, E., Nakashima, N., Ii, M. & Nishimoto, T. Novel G proteins, Rag C and Rag D, interact with GTP-binding proteins, Rag A and Rag B. J. Biol. Chem. 276, 7246–7257 (2001).

    CAS  PubMed  Google Scholar 

  24. Kim, E., Goraksha-Hicks, P., Li, L., Neufeld, T.P. & Guan, K.L. Regulation of TORC1 by Rag GTPases in nutrient response. Nat. Cell Biol. 10, 935–945 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Sancak, Y. et al. The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science 320, 1496–1501 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Bar-Peled, L. et al. A tumor suppressor complex with GAP activity for the Rag GTPases that signal amino acid sufficiency to mTORC1. Science 340, 1100–1106 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Bar-Peled, L., Schweitzer, L.D., Zoncu, R. & Sabatini, D.M. Ragulator is a GEF for the Rag GTPases that signal amino acid levels to mTORC1. Cell 150, 1196–1208 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Sancak, Y. et al. Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids. Cell 141, 290–303 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Zoncu, R. et al. mTORC1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H+-ATPase. Science 334, 678–683 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Rebsamen, M. et al. SLC38A9 is a component of the lysosomal amino acid sensing machinery that controls mTORC1. Nature 519, 477–481 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang, S. et al. Metabolism. Lysosomal amino acid transporter SLC38A9 signals arginine sufficiency to mTORC1. Science 347, 188–194 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Forgac, M. Vacuolar ATPases: rotary proton pumps in physiology and pathophysiology. Nat. Rev. Mol. Cell Biol. 8, 917–929 (2007).

    CAS  PubMed  Google Scholar 

  33. Jansen, E.J. & Martens, G.J. Novel insights into V-ATPase functioning: distinct roles for its accessory subunits ATP6AP1/Ac45 and ATP6AP2/(pro) renin receptor. Curr. Protein Pept. Sci. 13, 124–133 (2012).

    CAS  PubMed  Google Scholar 

  34. Iadevaia, V., Huo, Y., Zhang, Z., Foster, L.J. & Proud, C.G. Roles of the mammalian target of rapamycin, mTOR, in controlling ribosome biogenesis and protein synthesis. Biochem. Soc. Trans. 40, 168–172 (2012).

    CAS  PubMed  Google Scholar 

  35. Ben-Sahra, I., Howell, J.J., Asara, J.M. & Manning, B.D. Stimulation of de novo pyrimidine synthesis by growth signaling through mTOR and S6K1. Science 339, 1323–1328 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Tsun, Z.Y. et al. The folliculin tumor suppressor is a GAP for the RagC/D GTPases that signal amino acid levels to mTORC1. Mol. Cell 52, 495–505 (2013).

    CAS  PubMed  Google Scholar 

  37. Feig, L.A. & Cooper, G.M. Relationship among guanine nucleotide exchange, GTP hydrolysis, and transforming potential of mutated ras proteins. Mol. Cell. Biol. 8, 2472–2478 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Feig, L.A. Tools of the trade: use of dominant-inhibitory mutants of Ras-family GTPases. Nat. Cell Biol. 1, E25–E27 (1999).

    CAS  PubMed  Google Scholar 

  39. John, J. et al. Kinetic and structural analysis of the Mg2+-binding site of the guanine nucleotide–binding protein p21H-ras. J. Biol. Chem. 268, 923–929 (1993).

    CAS  PubMed  Google Scholar 

  40. Hara, K. et al. Amino acid sufficiency and mTOR regulate p70 S6 kinase and eIF-4E BP1 through a common effector mechanism. J. Biol. Chem. 273, 14484–14494 (1998).

    CAS  PubMed  Google Scholar 

  41. Nicklin, P. et al. Bidirectional transport of amino acids regulates mTOR and autophagy. Cell 136, 521–534 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Hoffenberg, S. et al. Functional and structural interactions of the Rab5 D136N mutant with xanthine nucleotides. Biochem. Biophys. Res. Commun. 215, 241–249 (1995).

    CAS  PubMed  Google Scholar 

  43. Schmidt, G. et al. Biochemical and biological consequences of changing the specificity of p21ras from guanosine to xanthosine nucleotides. Oncogene 12, 87–96 (1996).

    CAS  PubMed  Google Scholar 

  44. Farnsworth, C.L. & Feig, L.A. Dominant inhibitory mutations in the Mg2+-binding site of RasH prevent its activation by GTP. Mol. Cell. Biol. 11, 4822–4829 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Lai, C.C., Boguski, M., Broek, D. & Powers, S. Influence of guanine nucleotides on complex formation between Ras and CDC25 proteins. Mol. Cell. Biol. 13, 1345–1352 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Proud, C.S. Guanine nucleotides, protein phosphorylation and the control of translation. Trends Biochem. Sci. 11, 73–77 (1986).

    CAS  Google Scholar 

  47. Krengel, U. et al. Three-dimensional structures of H-ras p21 mutants: molecular basis for their inability to function as signal switch molecules. Cell 62, 539–548 (1990).

    CAS  PubMed  Google Scholar 

  48. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. DePristo, M.A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 43, 491–498 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Koboldt, D.C. et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 22, 568–576 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Dayem Ullah, A.Z., Lemoine, N.R. & Chelala, C. SNPnexus: a web server for functional annotation of novel and publicly known genetic variants (2012 update). Nucleic Acids Res. 40, W65–W70 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Saitou, N. & Nei, M. The neighbor-joining method: a new method for reconstructing phylogenetic trees. Mol. Biol. Evol. 4, 406–425 (1987).

    CAS  PubMed  Google Scholar 

  53. Van Loo, P. et al. Allele-specific copy number analysis of tumors. Proc. Natl. Acad. Sci. USA 107, 16910–16915 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Langmead, B. & Salzberg, S.L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  56. Law, C.W., Chen, Y., Shi, W. & Smyth, G.K. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014).

    PubMed  PubMed Central  Google Scholar 

  57. Ritchie, M.E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    PubMed  PubMed Central  Google Scholar 

  58. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Boussif, O. et al. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc. Natl. Acad. Sci. USA 92, 7297–7301 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are indebted to the patients for donating tumor specimens as part of this study. We thank G. Clark at the Francis Crick Institute for automated DNA sequencing and the Queen Mary University of London Genome Centre for Illumina MiSeq sequencing. We acknowledge the support of Barts, Cambridge, Leeds and Southampton's Experimental Cancer Medicine and Cancer Research UK Centers. This work was supported by grants from the Kay Kendall Leukaemia Fund and Cancer Research UK (awarded to J.F.), grants from the US National Institutes of Health (NIH; R01 CA103866 and AI47389) and the US Department of Defense (W81XWH-07-0448) to D.M.S., and fellowship support from the US NIH to R.L.W. (T32 GM007753 and F30 CA189333). D.M.S. is an investigator of the Howard Hughes Medical Institute. J.O. is a recipient of the Kay Kendall Leukaemia Fund Junior Clinical Research Fellowship (KKL 557).

Author information

Authors and Affiliations

Authors

Contributions

J.O. and J.F. conceived the study. J.O., D.M.S. and J.F. directed the study. C.M., G.P., P.J., A.D., J.C.S., M.-Q.D., S.B., A.J., T.A.L., R.A., S.M. and J.G.G. provided patient samples and clinical data. M.C., A.J. and M.-Q.D. conducted pathological review of specimens. J.M. collated clinical information. S.I. prepared and processed samples. H.Q. provided cell line DNA. J.O., R.L.W., S.A., L.W., B.M.C., L.E.-I., A.F.A.S., A.C., A.E., C.B. and R.Z. performed experiments. J.W., J.A.G.-A., S.H.B. and C.C. performed the bioinformatic analysis. J.R. and R.S. coordinated and verified the ICGC data set. J.O., R.L.W., J.W., D.M.S. and J.F. analyzed and interpreted the data. J.O., R.L.W., D.M.S. and J.F. wrote the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Jessica Okosun or David M Sabatini.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Clinical timeline for the discovery WES cases.

This illustrates the timeline of the disease events during the clinical course of each patient’s disease, further indicating the available samples that were sequenced in this study.

Supplementary Figure 2 Phylogenetic reconstruction demonstrating the clonal evolution history of each of the five WES cases.

In each case, a phylogenetic tree was constructed using the somatic nonsynonymous variants detected in the WES analyses. All trees are rooted at the germline (GL) sequence, with the trunk of the tree representing variants shared by all the tumor biopsies, depicting a common ancestral origin. Internal branches indicate variants that are shared by more than one subsequent progressed or relapse tumor, and the terminal branches illustrate variants that are unique or phase specific to that biopsy alone. Early initiating genes are shown on the trunk of the tree. Novel genes identified in this study (RRAGC, ATP6V1B2 and ATP6AP1) are also illustrated. For RRAGC mutations, the superscript numbers in cases B2, B3 and B4 indicate the different RRAGC mutations identified in those individual biopsies.

Supplementary Figure 3 Copy number of RRAGC as compared to other gene loci.

The top panel shows the log R values for each of the gene loci indicated from all 24 samples from the five WES cases, and the bottom panel shows the log R values from our previously published SNP6.0 data set comprising 29 different follicular lymphoma samples and corresponding paired transformed follicular lymphoma. The gene locus for TNFRSF14, 1p36.32, was chosen as a reference locus as it is commonly subject to frequent copy number deletions in follicular lymphoma. The horizontal dashed line indicates the log R value of −0.2, with values below this measure indicative of deletions and those above 0.2 indicative of gains, as reported previously3.

Supplementary Figure 4 Model of components of the amino acid–induced mTORC1 pathway.

At low amino acid levels (left), the Rag heterodimer (RagB-RagC) is in a nucleotide-bound configuration incompatible for the recruitment and activation of mTORC1. In the presence of sufficient amino acids (right), a supercomplex comprising the v-ATPase, Ragulator, SLC38A9 and the Rag GTPase heterodimer translocates to the lysosomal surface. This changes the Rag heterodimer into its active form with RagB being GTP bound and RagC being GDP bound, resulting in the recruitment and activation of mTORC1.

Supplementary Figure 5 Differential gene expression between RRAGC-mutated and wild-type follicular lymphoma cases.

(a) Heat map from the unsupervised hierarchical clustering of genes that are differentially expressed in RRAGC-mutated (red bar; n = 5) and wild-type (blue bar; n = 8) tumors. This consisted of 75 upregulated and 182 downregulated genes, selected on the basis of a double threshold of raw P value < 0.01 and absolute fold change >2. (b) Mean gene expression values for RRAGC, MTOR and the other Rag GTPases. No difference in expression was noted between RRAGC-mutated and wild-type tumors. Expression values were measured using voom log2-cpm (read count per million reads).

Supplementary Figure 6 Representative GSEA plots.

GSEA of gene expression data derived from RNA-seq of five RRAGC-mutated versus eight RRAGC–wild type cases. This showed significant enrichment for gene sets in several processes involved in translation and cell cycle regulation, which were upregulated in the RRAGC-mutated tumors as compared to wild-type tumors. Hits displayed below the graph show where the members of the gene set appear in the ranked list of genes. FDR q values and further gene sets are fully listed in Supplementary Table 10.

Supplementary Figure 7 Recurrent follicular lymphoma RagC mutants activate the mTORC1 pathway.

(a) Follicular lymphoma RagC mutants (RagCS75F, RagCS75N, RagCT90N and RagCW115R) dramatically increase mTORC1 binding (mTOR and raptor), whereas RRAGC mutations identified in solid cancers (p.M121V, p.Y165C, p.D202G, p.L217R and p.R396Q) did not coimmunoprecipitate mTORC1 as strongly. Anti-FLAG immunoprecipitates were collected and analyzed as in Figure 3a. (b) All four RagC mutants coimmunoprecipitate more raptor than wild-type RagC in Raji cells. Anti-FLAG immunoprecipitates from Raji cells stably expressing the indicated proteins were collected and analyzed as in Figure 3a. (c) Three RagC mutants (RagCS75N, RagCS75F and RagCW115R) coimmunoprecipitate more raptor than wild-type RagC when overexpressed in OCI-Ly7 cells. Anti-FLAG immunoprecipitates from OCI-Ly7 cells stably expressing the indicated proteins were collected and analyzed as in Figure 3a. (d) Four RagC mutants increase raptor binding over wild-type RagC in OCI-Ly8 cells. Anti-FLAG immunoprecipitates from Ly8 cells stably expressing the indicated proteins were collected and analyzed as in Figure 3a. (e) Stable overexpression of RagCS75N, RagCS75F, RagCT90N and RagCW115R renders the cells partially insensitive to full amino acid deprivation. HEK293T cells that stably expressed the indicated proteins were starved of amino acids for 50 min and restimulated with amino acids for 10 min. The cell lysates were analyzed as in Figure 3b. (f) Quantification of the amount of phosphorylated S6K1 in Figure 3c, under leucine starvation or starvation followed by restimulation in HEK293T cells stably expressing the indicated proteins. (g) Quantification of the amount of phosphorylated S6K1 in Figure 3d, under arginine starvation or starvation followed by restimulation in HEK293T cells stably expressing the indicated proteins. (h) Quantification of the amount of phosphorylated S6K1 in Figure 3e, under leucine starvation or starvation followed by restimulation in Karpas-422 cells stably expressing the indicated proteins.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Tables 1 and 2 (PDF 1711 kb)

Supplementary Tables 3: Summary of somatic variants identified in the 24 tumor samples (5 cases) (XLS 24 kb)

41588_2016_BFng3473_MOESM32_ESM.xls

Supplementary Tables 4: List of somatic variants identified from whole exome sequencing 5 cases (24 tumor biopsies) (XLS 337 kb)

Supplementary Table 5: List of RRAGC and mTORC1-related gene variants (XLS 52 kb)

Supplementary Table 6: RRAGC mutations identified in solid tumors, from published datasets (XLS 24 kb)

Supplementary Table 7: Targeted resequencing coverage (XLS 25 kb)

Supplementary Table 8: List of ATP6V1B2 and ATP6AP1 variants (XLS 41 kb)

Supplementary Table 9: Details of FL cases for RNA-seq and GSEA analyses (XLS 27 kb)

41588_2016_BFng3473_MOESM38_ESM.xls

Supplementary Table 10: Top significantly enriched gene sets for RRAGC mutants compared to wild-type comparison using the GSEA tool MSigDB collections (XLS 76 kb)

Supplementary Table 11: Details of samples used in this study (XLS 48 kb)

Supplementary Table 12: Clinical characteristics of the sequencing and validation cohort (XLS 24 kb)

Supplementary Table 13: Primers sequences for tagged-amplicon sequencing (XLS 23 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Okosun, J., Wolfson, R., Wang, J. et al. Recurrent mTORC1-activating RRAGC mutations in follicular lymphoma. Nat Genet 48, 183–188 (2016). https://doi.org/10.1038/ng.3473

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3473

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research