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:

Ubiquitination of hnRNPA1 by TRAF6 links chronic innate immune signaling with myelodysplasia

A Corrigendum to this article was published on 22 March 2017

This article has been updated

Abstract

Toll-like receptor (TLR) activation contributes to premalignant hematologic conditions, such as myelodysplastic syndromes (MDS). TRAF6, a TLR effector with ubiquitin (Ub) ligase activity, is overexpressed in MDS hematopoietic stem/progenitor cells (HSPCs). We found that TRAF6 overexpression in mouse HSPC results in impaired hematopoiesis and bone marrow failure. Using a global Ub screen, we identified hnRNPA1, an RNA-binding protein and auxiliary splicing factor, as a substrate of TRAF6. TRAF6 ubiquitination of hnRNPA1 regulated alternative splicing of Arhgap1, which resulted in activation of the GTP-binding Rho family protein Cdc42 and accounted for hematopoietic defects in TRAF6-expressing HSPCs. These results implicate Ub signaling in coordinating RNA processing by TLR pathways during an immune response and in premalignant hematologic diseases, such as MDS.

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: Overexpression of TRAF6 results in HSPC defects.
Figure 2: TRAF6 regulates ubiquitination of RNA-binding proteins and RNA splicing.
Figure 3: TRAF6 ubiquitinates hnRNPA1.
Figure 4: hnRNPA1 contributes to the hematopoietic phenotype in Vav-TRAF6 mice.
Figure 5: TRAF6 regulates RNA splicing of Arhgap1.
Figure 6: Exclusion of exon 2 results in reduced Arhgap1 protein and Cdc42 activation.
Figure 7: Cdc42 contributes to HSPC defects in Vav-TRAF6 mice.
Figure 8: Cdc42 activation is associated with primary human MDS.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Change history

  • 23 January 2017

    In the version of this article initially published online, the 16th author's surname was spelled incorrectly as 'Salamonis'. The correct spelling is 'Salomonis'. The error has been corrected in the PDF and HTML versions of this article.

References

  1. Esplin, B.L. et al. Chronic exposure to a TLR ligand injures hematopoietic stem cells. J. Immunol. 186, 5367–5375 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Wong, J.J. et al. Orchestrated intron retention regulates normal granulocyte differentiation. Cell 154, 583–595 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Shalek, A.K. et al. Single-cell transcriptomics reveals bimodality in expression and splicing in immune cells. Nature 498, 236–240 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Nagai, Y. et al. Toll-like receptors on hematopoietic progenitor cells stimulate innate immune system replenishment. Immunity 24, 801–812 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Starczynowski, D.T. et al. Identification of miR-145 and miR-146a as mediators of the 5q- syndrome phenotype. Nat. Med. 16, 49–58 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Kristinsson, S.Y. et al. Chronic immune stimulation might act as a trigger for the development of acute myeloid leukemia or myelodysplastic syndromes. J. Clin. Oncol. 29, 2897–2903 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Wei, Y. et al. Toll-like receptor alterations in myelodysplastic syndrome. Leukemia 27, 1832–1840 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Caputi, M., Mayeda, A., Krainer, A.R. & Zahler, A.M. hnRNP A/B proteins are required for inhibition of HIV-1 pre-mRNA splicing. EMBO J. 18, 4060–4067 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Nimer, S.D. Myelodysplastic syndromes. Blood 111, 4841–4851 (2008).

    Article  CAS  PubMed  Google Scholar 

  10. Wei, Y. et al. Toll-like receptor alterations in myelodysplastic syndrome. Leukemia 27, 1832–1840 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Barreyro, L. et al. Overexpression of IL-1 receptor accessory protein in stem and progenitor cells and outcome correlation in AML and MDS. Blood 120, 1290–1298 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Dimicoli, S. et al. Overexpression of the toll-like receptor (TLR) signaling adaptor MYD88, but lack of genetic mutation, in myelodysplastic syndromes. PLoS One 8, e71120 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Chen, X. et al. Induction of myelodysplasia by myeloid-derived suppressor cells. J. Clin. Invest. 123, 4595–4611 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Varney, M.E. et al. Loss of Tifab, a del(5q) MDS gene, alters hematopoiesis through derepression of Toll-like receptor-TRAF6 signaling. J. Exp. Med. 212, 1967–1985 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ogilvy, S. et al. Promoter elements of vav drive transgene expression in vivo throughout the hematopoietic compartment. Blood 94, 1855–1863 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. Lin, Y.W., Slape, C., Zhang, Z. & Aplan, P.D. NUP98-HOXD13 transgenic mice develop a highly penetrant, severe myelodysplastic syndrome that progresses to acute leukemia. Blood 106, 287–295 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ivanova, N.B. et al. A stem cell molecular signature. Science 298, 601–604 (2002).

    Article  CAS  PubMed  Google Scholar 

  18. Clark, T.A., Sugnet, C.W. & Ares, M. Jr. Genomewide analysis of mRNA processing in yeast using splicing-specific microarrays. Science 296, 907–910 (2002).

    Article  CAS  PubMed  Google Scholar 

  19. Purdom, E. et al. FIRMA: a method for detection of alternative splicing from exon array data. Bioinformatics 24, 1707–1714 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ray, D. et al. A compendium of RNA-binding motifs for decoding gene regulation. Nature 499, 172–177 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Iervolino, A. et al. hnRNP A1 nucleocytoplasmic shuttling activity is required for normal myelopoiesis and BCR/ABL leukemogenesis. Mol. Cell. Biol. 22, 2255–2266 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Fan, Y. et al. Lysine 63-linked polyubiquitination of TAK1 at lysine 158 is required for tumor necrosis factor alpha- and interleukin-1beta-induced IKK/NF-kappaB and JNK/AP-1 activation. J. Biol. Chem. 285, 5347–5360 (2010).

    Article  CAS  PubMed  Google Scholar 

  23. Zhang, W. et al. Crystal structures and RNA-binding properties of the RNA recognition motifs of heterogeneous nuclear ribonucleoprotein L: insights into its roles in alternative splicing regulation. J. Biol. Chem. 288, 22636–22649 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Han, K., Yeo, G., An, P., Burge, C.B. & Grabowski, P.J. A combinatorial code for splicing silencing: UAGG and GGGG motifs. PLoS Biol. 3, e158 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zearfoss, N.R., Clingman, C.C., Farley, B.M., McCoig, L.M. & Ryder, S.P. Quaking regulates Hnrnpa1 expression through its 3′ UTR in oligodendrocyte precursor cells. PLoS Genet. 7, e1001269 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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 

  27. Tavanez, J.P., Madl, T., Kooshapur, H., Sattler, M. & Valcárcel, J. hnRNP A1 proofreads 3′ splice site recognition by U2AF. Mol. Cell 45, 314–329 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Florian, M.C. et al. Cdc42 activity regulates hematopoietic stem cell aging and rejuvenation. Cell Stem Cell 10, 520–530 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wang, L., Yang, L., Filippi, M.D., Williams, D.A. & Zheng, Y. Genetic deletion of Cdc42GAP reveals a role of Cdc42 in erythropoiesis and hematopoietic stem/progenitor cell survival, adhesion, and engraftment. Blood 107, 98–105 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Yang, L. et al. Rho GTPase Cdc42 coordinates hematopoietic stem cell quiescence and niche interaction in the bone marrow. Proc. Natl. Acad. Sci. USA 104, 5091–5096 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Stoilov, P., Lin, C.H., Damoiseaux, R., Nikolic, J. & Black, D.L. A high-throughput screening strategy identifies cardiotonic steroids as alternative splicing modulators. Proc. Natl. Acad. Sci. USA 105, 11218–11223 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Florian, M.C. et al. A canonical to non-canonical Wnt signalling switch in haematopoietic stem-cell ageing. Nature 503, 392–396 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Dolatshad, H. et al. Disruption of SF3B1 results in deregulated expression and splicing of key genes and pathways in myelodysplastic syndrome hematopoietic stem and progenitor cells. Leukemia 29, 1092–1103 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Iglesias, M.J. et al. Combined chromatin and expression analysis reveals specific regulatory mechanisms within cytokine genes in the macrophage early immune response. PLoS One 7, e32306 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 368, 2059–2074 (2013).

  36. Wells, C.D. et al. A Rich1/Amot complex regulates the Cdc42 GTPase and apical-polarity proteins in epithelial cells. Cell 125, 535–548 (2006).

    Article  CAS  PubMed  Google Scholar 

  37. Jean-Philippe, J., Paz, S. & Caputi, M. hnRNP A1: the Swiss army knife of gene expression. Int. J. Mol. Sci. 14, 18999–19024 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Elias, H.K. et al. Stem cell origin of myelodysplastic syndromes. Oncogene 33, 5139–5150 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Baldridge, M.T., King, K.Y., Boles, N.C., Weksberg, D.C. & Goodell, M.A. Quiescent haematopoietic stem cells are activated by IFN-gamma in response to chronic infection. Nature 465, 793–797 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Essers, M.A. et al. IFNalpha activates dormant haematopoietic stem cells in vivo. Nature 458, 904–908 (2009).

    Article  CAS  PubMed  Google Scholar 

  41. Wang, L., Yang, L., Debidda, M., Witte, D. & Zheng, Y. Cdc42 GTPase-activating protein deficiency promotes genomic instability and premature aging-like phenotypes. Proc. Natl. Acad. Sci. USA 104, 1248–1253 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chambers, S.M. et al. Aging hematopoietic stem cells decline in function and exhibit epigenetic dysregulation. PLoS Biol. 5, e201 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Han, D. et al. Dendritic cell expression of the signaling molecule TRAF6 is critical for gut microbiota-dependent immune tolerance. Immunity 38, 1211–1222 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Emig, D. et al. AltAnalyze and DomainGraph: analyzing and visualizing exon expression data. Nucleic Acids Res. 38, W755–W762 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. 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 

  46. McLeay, R.C. & Bailey, T.L. Motif Enrichment Analysis: a unified framework and an evaluation on ChIP data. BMC Bioinformatics 11, 165 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wu, J. et al. SpliceTrap: a method to quantify alternative splicing under single cellular conditions. Bioinformatics 27, 3010–3016 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Wijeratne, A.B., Manning, J.R., Schultz, Jel.J. & Greis, K.D. Quantitative phosphoproteomics using acetone-based peptide labeling: method evaluation and application to a cardiac ischemia/reperfusion model. J. Proteome Res. 12, 4268–4279 (2013).

    Article  CAS  PubMed  Google Scholar 

  49. Cyphert, T.J., Suchanek, A.L., Griffith, B.N. & Salati, L.M. Starvation actively inhibits splicing of glucose-6-phosphate dehydrogenase mRNA via a bifunctional ESE/ESS element bound by hnRNP K. Biochim. Biophys. Acta 1829, 905–915 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Rhyasen, G.W. et al. Targeting IRAK1 as a therapeutic approach for myelodysplastic syndrome. Cancer Cell 24, 90–104 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Starczynowski, D.T. et al. TRAF6 is an amplified oncogene bridging the RAS and NF-κB pathways in human lung cancer. J. Clin. Invest. 121, 4095–4105 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Fang, J. et al. Cytotoxic effects of bortezomib in myelodysplastic syndrome/acute myeloid leukemia depend on autophagy-mediated lysosomal degradation of TRAF6 and repression of PSMA1. Blood 120, 858–867 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wang, L., Yang, L., Burns, K., Kuan, C.Y. & Zheng, Y. Cdc42GAP regulates c-Jun N-terminal kinase (JNK)-mediated apoptosis and cell number during mammalian perinatal growth. Proc. Natl. Acad. Sci. USA 102, 13484–13489 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Starczynowski laboratory for discussions. We thank J. Bailey and V. Summey for assistance with transplantations (Comprehensive Mouse and Cancer Core). We also thank L. Salati (West Virginia University) for advice on the RIP assays, and H. Singh (CCHMC) for helpful discussion. MDSL cells were kindly provided by K. Tohyama (Kawasaki Medical University). Traf6-floxed mice were kindly provided by Y. Choi (University of Pennsylvania). This work was supported by Cincinnati Children's Hospital Research Foundation, American Society of Hematology (ASH), National Institute of Health (RO1HL111103, RO1DK102759, RO1HL114582), Gabrielle's Angel Foundation for Cancer Research, and Edward P. Evans Foundation grants to D.T.S. D.T.S. is a Leukemia Lymphoma Society Scholar. Cores are supported through the NIDDK Centers of Excellence in Experimental Hematology (P30DK090971).

Author information

Authors and Affiliations

Authors

Contributions

J.F., L.C.B., M.-D.F., K.D.G., H.G., Y.Z. and D.T.S. designed and performed experiments. X.L., S.C., S.A. and R.K. provided technical assistance. K.C. provided bioinformatics support. X.C., N.S. and M.T.W. performed the RNA splicing and exon binding analysis. P.S. provided reagents and advice on RNA splicing. D.W. provided histopathology advice. K.G. performed the mass spectrometry analysis. G.G.-M. and J.P.M. provided patient samples. J.F. and D.T.S. wrote the manuscript.

Corresponding author

Correspondence to Daniel T Starczynowski.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Generation of a hematopoietic-specific TRAF6 overexpression mouse to mimic expression of TRAF6 in human MDS.

(a) A schematic of the Vav-TRAF6 construct used for pronuclear injection to generate Vav-TRAF6 transgenic mice. The 5’ and 3’ Vav regulatory elements, simian virus 40 (SV40) intron and polyadenlyation signal are indicated. A FLAG-tagged human TRAF6 cDNA was cloned downstream of a Vav promoter. (b) Immunoblotting for TRAF6 in spleen and liver cells from WT (FVB/NJ) and Vav-TRAF6 mice. (c) Immunoblotting for TRAF6 in Lin- BM cells from WT and 3 independent genetic Vav-TRAF6 founder mice on the FVB/NJ or C57/Bl6 background. (d) Summary of spleen weight from WT (FVB/NJ) and moribund Vav-TRAF6 mice (left). Representative spleens from WT (FVB/NJ) and Vav-TRAF6 mice (right). (e) Summary of CD11b/Gr1 (myeloid), Ter119 (erythroid), B220 (B lymphocytes), and CD3 (T lymphocytes) immunophenotypic proportions within the spleen of WT (FVB/NJ; n = 4) and moribund Vav-TRAF6 mice (n = 4).

Supplementary Figure 2 TRAF6 overexpression results in HSPC defects following competitive BM transplantations.

(a) Experimental overview of competitive BMT. BM MNC or LT-HSC (CD45.2+) from WT (C57Bl/6) or Vav-TRAF6 mice were mixed with competitor BM cells (CD45.1+) at 1:1 ratio, and transplanted into lethally-irradiated recipient mice (BoyJ) as the primary (10) cBMT. BM cells were recovered from recipients at 5 months post 10 cBMT, and transplanted into lethally-irradiated recipient mice (BoyJ) for the secondary (20) cBMT. (b) Peripheral blood chimerism of donor-derived (CD45.2+) MNC was determined after 10 and/or 20 cBMT using MNC or LT-HSC. (n > 5 per group). (c) BM chimerism of myeloid (CD11b+) and lymphoid (CD3+ and B220+) cells of donor-derived peripheral blood (CD45.2+) was examined after the 20 BMT (n > 5 per group). (d) BM chimerism of HSPC was determined after the 10 cBMT. (n > 6 per group). (e) Proportion of myeloid (CD11b+) and lymphoid (CD3+ and B220+) cells in the competitor cell-derived compartment (CD45.1+) after competitive transplantation with either BM mononuclear cells (MNC, left) or LT-HSC (right). (n > 7 mice per genotype). *, P < 0.05.

Supplementary Figure 3 TRAF6 overexpression alters myeloid differentiation, innate immune, and hematopoietic stem/progenitor gene profiles.

(a) Differentially expressed genes in WT (FVB/NJ; n = 4) and Vav-TRAF6 (n = 3) LSK at 6 months of age. (b) Gene set enrichment analysis (GSEA) performed on LSK RNA isolated from WT and Vav-TRAF6 mice.

Supplementary Figure 4 hnRNPA1 is an ubiquitin substrate of TRAF6.

(a) A schematic representation of the comparative Ub (di-Glycine) proteomic assay to identify novel TRAF6 ubiquitinated substrates in TF1 cells with (+) and without (-) TRAF6 (left panel), and in vitro ubiquitin reconstitution assay on individual protein substrates (right panel). (b) TF1 cells expressing a doxycyclin (DOX)-inducible shRNA targeting TRAF6 were used for comparative Ub (di-Glycine) capture proteomics. The table contains the number of identified peptides at each step of the process (upper). The Venn diagram represents the final number of unique peptides in each experimental group (lower). (c) Immunoprecipitated (IP) hnRNPA1 was immunoblotted (IB) for Ub in TF1 cells expressing a DOX-inducible TRAF6 shRNA. (+ DOX, TRAF6 knockdown). (d) HEK293 cells transfected with HA-Ub, MYC-hnRNPA1, and/or FLAG-TRAF6 were IB for IP MYC-hnRNPA1 (anti HA-Ub).

Supplementary Figure 5 TRAF6 affects RNA isoform expression and splicing in HSPC.

(a) FIRMA Index values and RT-PCR analysis (using primers flanking the skipped exon) of the indicated cassette exons in Vav-TRAF6 and WT (FVB/NJ) LSK (left) and LPS-treated WT Lin- BM cells (right). Values under the plot represent the short isoform as a percentage of the short and long isoforms. (b-d) Endogenous Cep164 exon 7 usage as measured by qRT-PCR using primers to the exon 6-8 junction (as depicted in the schematic on the right) and normalized to total Cep164 in WT Lin- BM cells stimulated with 10 ng/mL of LPS (b), LSK and Lin- BM cells from WT (FVB/NJ) and Vav-TRAF6 mice (c), and TRAF6-deficient Lin- BM cells (C57Bl/6 Traf6WT/WT;Mx1Cre and Traf6fl/fl;Mx1Cre mice treated with PolyIC) stimulated with 10 ng/mL of LPS (d). (n > 3 per experimental group). *, P < 0.05.

Supplementary Figure 6 Loss of Arhgap1 contributes to myeloid dysplasia.

(a) Wright-Giemsa-stained peripheral (PB) blood and bone marrow (BM) from WT (C57Bl/6) and Arhgap1-deficient (Arhgap1+/-) mice. Red arrows indicate dysplastic myeloid cells with Pseudo-Pelger Huet anomaly (upper) and neutrophils with hypersegmentation (lower). (b) Percent BM neutrophil dysplasia was determined in age-matched WT (n = 3) and Arhgap1+/- (Gap1+/-) (n = 6) mice. At least 100 cells were examined for each mouse. *, P < 0.05.

Supplementary Figure 7 Role of hnRNPA1 in TRAF6-mediated exon skipping of ARHGAP17 in human MDS.

(a) Schematic representation of the human ARHGAP17 gene structure with the approximate position of exons and introns. Overview of ARHGAP17 exon usage is shown for normal expression (black lines) and in THP1 cells stimulated with LPS (P = 0.0087) or MDSL cells (red lines). Below is the predicted protein products from ARHGAP17-DEx18 with the approximate positions of the BAR, Gap, and SH3 domains. (b) Knockdown of human hnRNPA1 by 2 independent shRNAs (shhnRNPA1-1 [TRCN0000006583] and shhnRNPA1-2 [TRCN0000006586]) was measured by qRT-PCR in HEK293 and MDSL (left) and by immunblotting in HEK293 (right). (c) RT-PCR analysis using primers flanking endogenous human ARHGAP17 exon 18 (as depicted in the schematic on the right) in HEK293 or MDSL cells following knockdown of hnRNPA1 (shA1). (d) Immunoblotting of ARHGAP17 and hnRNPA1 in MDSL cells transduced with independent shRNAs targeting hnRNPA1. (e) Overview of TRAF6 regulation of RNA splicing function during transient and chronic TLR signaling: TRAF6 ubiquitinates hnRNPA1 at the RNA recognition domain. Ubiquitinated hnRNPA1 binds directly and/or indirectly to select exons and results in increased exon exclusion and isoform expression. One of the signaling pathways affected by differential splicing is the small G-protein GTPase family, including Cdc42. Altered splicing of Arhgap1 (a negative regulator of Cdc42) results in active Cdc42, which in part contributes to the HSPC defects and myeloid dysplasia associated with BM failure.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Tables 1–6 (PDF 1978 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fang, J., Bolanos, L., Choi, K. et al. Ubiquitination of hnRNPA1 by TRAF6 links chronic innate immune signaling with myelodysplasia. Nat Immunol 18, 236–245 (2017). https://doi.org/10.1038/ni.3654

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3654

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