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:

Juvenile myelomonocytic leukemia displays mutations in components of the RAS pathway and the PRC2 network

Abstract

Juvenile myelomonocytic leukemia (JMML) is a rare and severe myelodysplastic and myeloproliferative neoplasm of early childhood initiated by germline or somatic RAS-activating mutations1,2,3. Genetic profiling and whole-exome sequencing of a large JMML cohort (118 and 30 cases, respectively) uncovered additional genetic abnormalities in 56 cases (47%). Somatic events were rare (0.38 events/Mb/case) and restricted to sporadic (49/78; 63%) or neurofibromatosis type 1 (NF1)-associated (8/8; 100%) JMML cases. Multiple concomitant genetic hits targeting the RAS pathway were identified in 13 of 78 cases (17%), disproving the concept of mutually exclusive RAS pathway mutations and defining new pathways activated in JMML involving phosphoinositide 3-kinase (PI3K) and the mTORC2 complex through RAC2 mutation. Furthermore, this study highlights PRC2 loss (26/78; 33% of sporadic JMML cases) that switches the methylation/acetylation status of lysine 27 of histone H3 in JMML cases with altered RAS and PRC2 pathways. Finally, the association between JMML outcome and mutational profile suggests a dose-dependent effect for RAS pathway activation, distinguishing very aggressive JMML rapidly progressing to acute myeloid leukemia.

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: Combinations of multiple hits targeting the RAS pathway and PRC2 network.
Figure 2: Alteration profiles in individual JMML cases.
Figure 3: Clonal evolution of JMML.
Figure 4: The p.Asp63Val substitution in RAC2 results in a gain-of-function effect associated with an increase in effector binding and a massive decrease in GAP function, leading to AKT activation via two distinct pathways.
Figure 5: Reduced PRC2 dosage and histone H3 modifications.
Figure 6: Overall survival of patients with sporadic JMML according to the presence and type of additional somatic mutations.

Similar content being viewed by others

Accession codes

Primary accessions

ArrayExpress

Referenced accessions

NCBI Reference Sequence

Protein Data Bank

References

  1. Chang, T.Y., Dvorak, C.C. & Loh, M.L. Bedside to bench in juvenile myelomonocytic leukemia: insights into leukemogenesis from a rare pediatric leukemia. Blood 124, 2487–2497 (2014).

    Article  CAS  PubMed  Google Scholar 

  2. Niemeyer, C.M. et al. Chronic myelomonocytic leukemia in childhood: a retrospective analysis of 110 cases. European Working Group on Myelodysplastic Syndromes in Childhood (EWOG-MDS). Blood 89, 3534–3543 (1997).

    CAS  PubMed  Google Scholar 

  3. Locatelli, F. & Niemeyer, C.M. How I treat juvenile myelomonocytic leukemia. Blood 125, 1083–1090 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Niemeyer, C.M. RAS diseases in children. Haematologica 99, 1653–1662 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Shlien, A. et al. Combined hereditary and somatic mutations of replication error repair genes result in rapid onset of ultra-hypermutated cancers. Nat. Genet. 47, 257–262 (2015).

    Article  CAS  PubMed  Google Scholar 

  6. Sakaguchi, H. et al. Exome sequencing identifies secondary mutations of SETBP1 and JAK3 in juvenile myelomonocytic leukemia. Nat. Genet. 45, 937–941 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Strullu, M. et al. Juvenile myelomonocytic leukaemia and Noonan syndrome. J. Med. Genet. 51, 689–697 (2014).

    Article  CAS  PubMed  Google Scholar 

  8. Niemeyer, C.M. et al. Germline CBL mutations cause developmental abnormalities and predispose to juvenile myelomonocytic leukemia. Nat. Genet. 42, 794–800 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Pérez, B. et al. Germline mutations of the CBL gene define a new genetic syndrome with predisposition to juvenile myelomonocytic leukaemia. J. Med. Genet. 47, 686–691 (2010).

    Article  PubMed  CAS  Google Scholar 

  10. Makishima, H. et al. Somatic SETBP1 mutations in myeloid malignancies. Nat. Genet. 45, 942–946 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Brown, K.M. et al. Phosphodiesterase-8A binds to and regulates Raf-1 kinase. Proc. Natl. Acad. Sci. USA 110, E1533–E1542 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Flex, E. et al. Activating mutations in RRAS underlie a phenotype within the RASopathy spectrum and contribute to leukaemogenesis. Hum. Mol. Genet. 23, 4315–4327 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Shang, X. et al. R-Ras and Rac GTPase cross-talk regulates hematopoietic progenitor cell migration, homing, and mobilization. J. Biol. Chem. 286, 24068–24078 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kawazu, M. et al. Transforming mutations of RAC guanosine triphosphatases in human cancers. Proc. Natl. Acad. Sci. USA 110, 3029–3034 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Emanuel, P.D. Hallway gossip between Ras and PI3K pathways. Blood 123, 2751–2753 (2014).

    Article  CAS  PubMed  Google Scholar 

  16. Goodwin, C.B. et al. PI3K p110δ uniquely promotes gain-of-function Shp2-induced GM-CSF hypersensitivity in a model of JMML. Blood 123, 2838–2842 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Worzfeld, T. et al. Genetic dissection of plexin signaling in vivo. Proc. Natl. Acad. Sci. USA 111, 2194–2199 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Innocenti, M. et al. Phosphoinositide 3-kinase activates Rac by entering in a complex with Eps8, Abi1, and Sos-1. J. Cell Biol. 160, 17–23 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Offenhäuser, N. et al. The eps8 family of proteins links growth factor stimulation to actin reorganization generating functional redundancy in the Ras/Rac pathway. Mol. Biol. Cell 15, 91–98 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Cutts, B.A. et al. Nf1 deficiency cooperates with oncogenic K-RAS to induce acute myeloid leukemia in mice. Blood 114, 3629–3632 (2009).

    Article  CAS  PubMed  Google Scholar 

  21. Xu, J. et al. Dominant role of oncogene dosage and absence of tumor suppressor activity in Nras-driven hematopoietic transformation. Cancer Discov. 3, 993–1001 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Wang, J. et al. Endogenous oncogenic Nras mutation initiates hematopoietic malignancies in a dose- and cell type–dependent manner. Blood 118, 368–379 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Khan, S.N. et al. Multiple mechanisms deregulate EZH2 and histone H3 lysine 27 epigenetic changes in myeloid malignancies. Leukemia 27, 1301–1309 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Son, J., Shen, S.S., Margueron, R. & Reinberg, D. Nucleosome-binding activities within JARID2 and EZH1 regulate the function of PRC2 on chromatin. Genes Dev. 27, 2663–2677 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kinkel, S.A. et al. Jarid2 regulates hematopoietic stem cell function by acting with Polycomb repressive complex 2. Blood 125, 1890–1900 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ciferri, C. et al. Molecular architecture of human Polycomb repressive complex 2. eLife 1, e00005 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Puda, A. et al. Frequent deletions of JARID2 in leukemic transformation of chronic myeloid malignancies. Am. J. Hematol. 87, 245–250 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Zhang, Y. et al. Corepressor protein CDYL functions as a molecular bridge between Polycomb repressor complex 2 and repressive chromatin mark trimethylated histone lysine 27. J. Biol. Chem. 286, 42414–42425 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Abdel-Wahab, O. et al. ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression. Cancer Cell 22, 180–193 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Abdel-Wahab, O. & Levine, R.L. Mutations in epigenetic modifiers in the pathogenesis and therapy of acute myeloid leukemia. Blood 121, 3563–3572 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kim, E. et al. SRSF2 mutations contribute to myelodysplasia by mutant-specific effects on exon recognition. Cancer Cell 27, 617–630 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Huether, R. et al. The landscape of somatic mutations in epigenetic regulators across 1,000 paediatric cancer genomes. Nat. Commun. 5, 3630 (2014).

    Article  PubMed  CAS  Google Scholar 

  33. Yang, F. PRC2 dysfunction through multiple mechanisms in myeloid malignancies. Epigenomics 5, 481 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Pasini, D. et al. Characterization of an antagonistic switch between histone H3 lysine 27 methylation and acetylation in the transcriptional regulation of Polycomb group target genes. Nucleic Acids Res. 38, 4958–4969 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Leiserson, M.D. et al. Pan-cancer network analysis identifies combinations of rare somatic mutations across pathways and protein complexes. Nat. Genet. 47, 106–114 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Lee, W. et al. PRC2 is recurrently inactivated through EED or SUZ12 loss in malignant peripheral nerve sheath tumors. Nat. Genet. 46, 1227–1232 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zhang, M. et al. Somatic mutations of SUZ12 in malignant peripheral nerve sheath tumors. Nat. Genet. 46, 1170–1172 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. De Raedt, T. et al. PRC2 loss amplifies Ras-driven transcription and confers sensitivity to BRD4-based therapies. Nature 514, 247–251 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Jaiswal, S. et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 371, 2488–2498 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Genovese, G. et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 371, 2477–2487 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Jacobs, K.B. et al. Detectable clonal mosaicism and its relationship to aging and cancer. Nat. Genet. 44, 651–658 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Laurie, C.C. et al. Detectable clonal mosaicism from birth to old age and its relationship to cancer. Nat. Genet. 44, 642–650 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Locatelli, F. et al. Hematopoietic stem cell transplantation (HSCT) in children with juvenile myelomonocytic leukemia (JMML): results of the EWOG-MDS/EBMT trial. Blood 105, 410–419 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Takagi, M. et al. Autoimmunity and persistent RAS-mutated clones long after the spontaneous regression of JMML. Leukemia 27, 1926–1928 (2013).

    Article  CAS  PubMed  Google Scholar 

  45. Matsuda, K. et al. Spontaneous improvement of hematologic abnormalities in patients having juvenile myelomonocytic leukemia with specific RAS mutations. Blood 109, 5477–5480 (2007).

    Article  CAS  PubMed  Google Scholar 

  46. Matsuda, K. et al. Acquisition of loss of the wild-type NRAS locus with aggressive disease progression in a patient with juvenile myelomonocytic leukemia and a heterozygous NRAS mutation. Haematologica 92, 1576–1578 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Kato, M. et al. Aggressive transformation of juvenile myelomonocytic leukemia associated with duplication of oncogenic KRAS due to acquired uniparental disomy. J. Pediatr. 162, 1285–1288 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Kalra, R., Paderanga, D.C., Olson, K. & Shannon, K.M. Genetic analysis is consistent with the hypothesis that NF1 limits myeloid cell growth through p21ras. Blood 84, 3435–3439 (1994).

    CAS  PubMed  Google Scholar 

  49. Chan, R.J., Cooper, T., Kratz, C.P., Weiss, B. & Loh, M.L. Juvenile myelomonocytic leukemia: a report from the 2nd International JMML Symposium. Leuk. Res. 33, 355–362 (2009).

    Article  PubMed  Google Scholar 

  50. Pérez, B. et al. Genetic typing of CBL, ASXL1, RUNX1, TET2 and JAK2 in juvenile myelomonocytic leukaemia reveals a genetic profile distinct from chronic myelomonocytic leukaemia. Br. J. Haematol. 151, 460–468 (2010).

    Article  CAS  PubMed  Google Scholar 

  51. Ahmadian, M.R., Stege, P., Scheffzek, K. & Wittinghofer, A. Confirmation of the arginine-finger hypothesis for the GAP-stimulated GTP-hydrolysis reaction of Ras. Nat. Struct. Biol. 4, 686–689 (1997).

    Article  CAS  PubMed  Google Scholar 

  52. Hemsath, L. & Ahmadian, M.R. Fluorescence approaches for monitoring interactions of Rho GTPases with nucleotides, regulators, and effectors. Methods 37, 173–182 (2005).

    Article  CAS  PubMed  Google Scholar 

  53. Eberth, A. & Ahmadian, M.R. In vitro GEF and GAP assays. Curr. Protoc. Cell Biol. Chapter 14, Unit 14.9 (2009).

  54. Jaiswal, M., Dubey, B.N., Koessmeier, K.T., Gremer, L. & Ahmadian, M.R. Biochemical assays to characterize Rho GTPases. Methods Mol. Biol. 827, 37–58 (2012).

    Article  CAS  PubMed  Google Scholar 

  55. Eberth, A. et al. A BAR domain–mediated autoinhibitory mechanism for RhoGAPs of the GRAF family. Biochem. J. 417, 371–377 (2009).

    Article  CAS  PubMed  Google Scholar 

  56. Jaiswal, M., Dubey, B.N., Koessmeier, K.T., Gremer, L. & Ahmadian, M.R. Biochemical assays to characterize Rho GTPases. Methods Mol. Biol. 827, 37–58 (2012).

    Article  CAS  PubMed  Google Scholar 

  57. Gremer, L. et al. Germline KRAS mutations cause aberrant biochemical and physical properties leading to developmental disorders. Hum. Mutat. 32, 33–43 (2011).

    Article  CAS  PubMed  Google Scholar 

  58. Nakhaei-Rad, S. et al. The function of embryonic stem cell–expressed RAS (E-RAS), a unique RAS family member, correlates with its additional motifs and its structural properties. J. Biol. Chem. 290, 15892–15903 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Worthylake, D.K., Rossman, K.L. & Sondek, J. Crystal structure of Rac1 in complex with the guanine nucleotide exchange region of Tiam1. Nature 408, 682–688 (2000).

    Article  CAS  PubMed  Google Scholar 

  60. Morreale, A. et al. Structure of Cdc42 bound to the GTPase binding domain of PAK. Nat. Struct. Mol. Biol. 7, 384–388 (2000).

    Article  CAS  Google Scholar 

  61. Nassar, N., Hoffman, G.R., Manor, D., Clardy, J.C. & Cerione, R.A. Structures of Cdc42 bound to the active and catalytically compromised forms of Cdc42GAP. Nat. Struct. Biol. 5, 1047–1052 (1998).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank N. Roodur, M. Keita (Unité de Génétique Moléculaire, Hospital Robert Debré) and L. Coste-Sarguet (INSERM UMR 1131) for expert technical assistance. We thank E. Martin, J.-P. Sarava and M. Letexier (Integragen) for advice and assistance in whole-exome sequencing analysis. SNP array analyses were performed by the Plateforme de Génomique Constitutionnelle–Nord (PfGC-Nord) (J. Soulier, S. Quentin and S. Drunat). We thank the technical team, and more particularly P. Aubin, from the Service de Biologie Cellulaire, Hôpital Saint Louis, for excellent technical help in the management of myeloid progenitor assays. We also thank S. Rasika for careful editing of the manuscript.

This work was supported by the Ligue contre le Cancer (LCC)–Ile-de-France, by the Société Française de Lutte contre les Cancers et Leucémies de l'Enfant (SFCE), the Association pour la Recherche et pour les Etudes dans les Maladies Infantiles Graves (AREMIG, Nancy) and the German Research Foundation (Deutsche Forschungsgemeinschaft, DFG) through the Collaborative Research Center 974 (SFB 974) Communication and Systems Relevance during Liver Injury and Regeneration and the International Graduate School of Protein Science and Technology (iGRASP).

Author information

Authors and Affiliations

Authors

Contributions

A. Caye collected subject samples, participated in the study design, performed laboratory assays, analyzed data and wrote the manuscript. M.S. and J.L. performed analyses and collected clinical data. F.G. performed epigenetic studies, and B.C. and E.V. performed colony assays. S.G. performed biostatistical analyses. O.F. and E.L. performed the cytological review. K.N., S.N.-R., R.D., D.H. and M.R.A. performed the functional and biochemical RAC2 studies. J.V. collected clinical data. S.P. performed laboratory assays. D.V. performed NF1 diagnosis. J.-H.D., A.B., C. Paillard, C. Picard, C.G., A.P., Y.R., F.M., B.N., Y.B., M.P., D.A., N.S. and A. Contet contributed subject samples and clinical data. M.S., A.B., C.C., B.C. and M.R.A. reviewed the manuscript. H.C. collected subject samples, designed and coordinated the study, analyzed data and wrote the manuscript.

Corresponding author

Correspondence to Hélène Cavé.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Spectrum of somatically acquired mutations identified by combining WES and genome-wide DNA array analysis in the discovery cohort of 30 JMML cases.

A total of 85 somatically acquired alterations were found, including 64 nonsynonymous point mutations or small insertion-deletions (indels) identified in the coding regions of these tumors by WES and 21 somatic cytogenetic alterations evidenced by SNP/CGH array, WES and/or metaphase cytogenetics.

Supplementary Figure 2 Graphical representation of the type of data obtained by sample in a cohort of 118 patients with JMML.

Both the discovery cohort (top; n = 30) and validation cohort (bottom; n = 88) are represented.

Supplementary Figure 3 Distribution of RAS-related mutations in a cohort of 118 patients with JMML as detected by routine workup.

Distribution of RAS-related mutations in 118 consecutively diagnosed JMML cases as detected by routine workup and detailed spectrum of KRAS (n = 18) and NRAS (n = 22) mutations.

Supplementary Figure 4 Histogram showing the type and number of additional somatic mutations per patient with JMML, according to genetic subgroup.

Supplementary Figure 5 Proportion of mutations predicted to be deleterious versus non-pathogenic substitutions.

The pathogenicity of somatic nonsynonymous exonic missense variants with respect to gene function was predicted using the SIFT, PolyPhen-2 and MutationTaster algorithms (Supplementary Table 6). A total of 91% of all missense mutations were predicted to result in functionally relevant alterations by at least two of the three methods used for functional prediction. This percentage was similar when considering only initiating mutations, known to be deleterious in all cases (92%), as well as secondary mutations (89%).

Supplementary Figure 6 Sequence electrophoregram showing the presence of three concomitant mutations targeting the RAS pathway at diagnosis of JMML_89.

Mutated nucleotides are indicated by a red arrow. The subclonal pattern of NF1 mutation is consistent with late acquisition. NF1 haploinsufficiency was due to a recurrent c.2033delG mutation of a G homopolymer within the NF1 coding region (exon 18). The frequency of this mutation appeared strikingly higher among somatic variants (5/6 cases with a secondary NF1 mutation) than among germline variants (Leiden Open Variation Database, LOVD).

Supplementary Figure 7 Hyperactive RAC2 Asp63Val contributes to AKT activation via both the PI3K-PDK1 and mTORC2 cascades.

Pull-down experiments (a,b) and immunoblot (IB) analysis were conducted using total cell lysates (cg) derived from transfected COS-7 cells with FLAG-tagged RAC2 and RAS variants. The GTPase-binding domain (GBD) of the RAC effector PAK1 was used as a GST fusion protein for the pulldown experiment. All experiments were performed three times. (a) Pulldown analysis showed that RAC2 Asp63Val largely exists in an active, GTP-bound state as compared to wild-type RAC2 (RAC2 WT), but activation is not as strong as for constitutively active RAC2 Gly12Val. Total RAC2 and RAS proteins were detected using antibody to FLAG and pan-RAS antibody to show the total amounts of the transfected FLAG-tagged RAC2 and RAS variants. (b) The RAC2 protein bands in a were densitometrically quantified (depicted as numbers and bars) as the amount of the GTP-bound RAC2 protein relative to wild-type RAC2. Coexpression of constitutively active NRAS Gly12Val, HRAS Gly12Val and KRAS Gly12Val did not change the level of GTP-bound RAC2 Asp63Val. (c) Total cell lysates were analyzed for the phosphorylation levels of AKT (pAKT 308 and pAKT 473), MEK1/2 (pMEK1/2) and ERK1/2 (pERK1/2). Total amounts of these kinases were used as loading controls. AKT is phosphorylated at Thr308 by the PI3K-PDK1 pathway, whereas the mTORC2 complex phosphorylates AKT at Ser473. (dg) The protein bands in c were densitometrically quantified (depicted as numbers and bars), clearly showing that the presence of RAC2 Asp63Val resulted in strong AKT phosphorylation and slight MEK phosphorylation but no ERK phosphorylation. Interestingly, a comparison of RAC2 Asp63Val and RAC2 Gly12Val showed that the relative amount of phosphorylated protein was proportional to the amount of the GTP-bound protein.

Supplementary Figure 8 Sequence electrophoregram showing progressive LOH of the KRAS locus with an allelic imbalance in favor of the oncogenic allele in JMML_24.

Wild-type (WT) and mutated nucleotides are indicated by black and red arrows, respectively.

Supplementary Figure 9 Overall survival in sporadic JMML according to initiating RAS-activating lesion.

Kaplan-Meier representation of the overall survival (%) in 96 patients with JMML evaluable for follow-up. Patients with Noonan syndrome were excluded from the analysis.

Supplementary Figure 10 Mean coverage of whole-exome sequencing in 30 paired JMML and germline DNA samples.

Supplementary Figure 11 Performances of PCR-based targeted deep sequencing in 75 JMML samples.

(a) The mean coverage of the coding regions is plotted for each gene by 25× descending order. (b) The mean depth of sequencing is plotted for each gene on a logarithmic scale, by descending order.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11, Supplementary Tables 1, 4 and 5 and Supplementary Note. (PDF 2807 kb)

Supplementary Table 2

List of variants identified by whole-exome sequencing. (XLSX 44 kb)

Supplementary Table 3

List of copy number abnormalities and copy-neutral loss of heterozygosity evidenced by genome-wide DNA arrayanalysis and whole-exome sequencing. (XLSX 22 kb)

Supplementary Table 6

List of all sequence mutations detected in the cohort of 118 JMML cases. (XLSX 55 kb)

Supplementary Table 7

Genetic analysis of myeloid colonies obtained by in vitroculture on methyl-cellulose. (XLSX 30 kb)

Supplementary Table 8

List of PCR primers used for targeted deep sequencing and Sanger targeted sequencing. (XLSX 116 kb)

Supplementary Table 9

Tabular data for survival curves. (XLSX 21 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Caye, A., Strullu, M., Guidez, F. et al. Juvenile myelomonocytic leukemia displays mutations in components of the RAS pathway and the PRC2 network. Nat Genet 47, 1334–1340 (2015). https://doi.org/10.1038/ng.3420

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3420

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer