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:

The ITIM-containing receptor LAIR1 is essential for acute myeloid leukaemia development

Abstract

Conventional strategies are not particularly successful in the treatment of leukaemia, and identification of signalling pathways crucial to the activity of leukaemia stem cells will provide targets for the development of new therapies. Here we report that certain receptors containing the immunoreceptor tyrosine-based inhibition motif (ITIM) are crucial for the development of acute myeloid leukaemia (AML). Inhibition of expression of the ITIM-containing receptor LAIR1 does not affect normal haematopoiesis but abolishes leukaemia development. LAIR1 induces activation of SHP-1, which acts as a phosphatase-independent signalling adaptor to recruit CAMK1 for activation of downstream CREB in AML cells. The LAIR1–SHP-1–CAMK1–CREB pathway sustains the survival and self-renewal of AML stem cells. Intervention in the signalling initiated by ITIM-containing receptors such as LAIR1 may result in successful treatment of AML.

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: Lair1, a representative ITIM receptor, is essential for the growth of human leukaemia cell lines.
Figure 2: Knockdown of lair1 blocks xenograft of human leukaemia cell lines.
Figure 3: LAIR1 enhances development of MLL-AF9 mouse AML during serial transplantation.
Figure 4: LAIR1 deficiency exhausts tumour-initiating cells by apoptosis.
Figure 5: SHP-1 rescues the lair1-null AML phenotype.
Figure 6: SHP-1 functions as an adaptor to recruit CAMK1 in the LAIR1 signalling.
Figure 7: CREB is a transcription factor for LAIR1 signalling in AML cells.
Figure 8: LAIR1 signalling is essential for human leukaemia development.

Similar content being viewed by others

References

  1. Maynadie, M. et al. Twenty-five years of epidemiological recording on myeloid malignancies: data from the specialized registry of hematologic malignancies of Cote d’Or (Burgundy, France). Haematologica 96, 55–61 (2011).

    PubMed  Google Scholar 

  2. Krause, D. S. & Van Etten, R. A. Right on target: eradicating leukemic stem cells. Trends Mol. Med. 13, 470–481 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Lapidot, T. et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367, 645–648 (1994).

    CAS  PubMed  Google Scholar 

  4. Lane, S. W., Scadden, D. T. & Gilliland, D. G. The leukemic stem cell niche: current concepts and therapeutic opportunities. Blood 114, 1150–1157 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Wei, J. et al. Microenvironment determines lineage fate in a human model of MLL-AF9 leukemia. Cancer Cell 13, 483–495 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Ishikawa, F. et al. Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat. Biotechnol. 25, 1315–1321 (2007).

    CAS  PubMed  Google Scholar 

  7. Imbesi, S. et al. Oxidative stress in oncohematologic diseases: an update. Expert Rev. Hematol. 6, 317–325 (2013).

    CAS  PubMed  Google Scholar 

  8. Fathi, A. & Levis, M. FLT3 inhibitors: a story of the old and the new. Curr. Opin. Hematol. 18, 71–76 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Vainchenker, W. & Constantinescu, S. N. JAK/STAT signaling in hematological malignancies. Oncogene 32, 2601–2613 (2013).

    CAS  PubMed  Google Scholar 

  10. Broxmeyer, H. E. Chemokines in hematopoiesis. Curr. Opin. Hematol. 15, 49–58 (2008).

    CAS  PubMed  Google Scholar 

  11. Jin, L., Hope, K. J., Zhai, Q., Smadja-Joffe, F. & Dick, J. E. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat. Med. 12, 1167–1174 (2006).

    PubMed  Google Scholar 

  12. Krause, D. S., Lazarides, K., von Andrian, U. H. & Van Etten, R. A. Requirement for CD44 in homing and engraftment of BCR-ABL-expressing leukemic stem cells. Nat. Med. 12, 1175–1180 (2006).

    CAS  PubMed  Google Scholar 

  13. Miller, P. G. et al. In vivo RNAi screening identifies a leukemia-specific dependence on integrin beta 3 signaling. Cancer Cell 24, 45–58 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Chao, M. P. et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell 142, 699–713 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Jan, M. et al. Prospective separation of normal and leukemic stem cells based on differential expression of TIM3, a human acute myeloid leukemia stem cell marker. Proc. Natl Acad. Sci. USA 108, 5009–5014 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Horton, S. J. & Huntly, B. J. Recent advances in acute myeloid leukemia stem cell biology. Haematologica 97, 966–974 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Weng, A. P. et al. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science 306, 269–271 (2004).

    CAS  PubMed  Google Scholar 

  18. Zhao, C. et al. Loss of beta-Catenin impairs the renewal of normal and CML stem cells in vivo. Cancer Cell 12, 528–541 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Wang, Y. et al. The Wnt/beta-catenin pathway is required for the development of leukemia stem cells in AML. Science 327, 1650–1653 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhao, C. et al. Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature 458, 776–779 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Krause, D. S. et al. Differential regulation of myeloid leukemias by the bone marrow microenvironment. Nat. Med. 19, 1513–1517 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Heidel, F. H., Mar, B. G. & Armstrong, S. A. Self-renewal related signaling in myeloid leukemia stem cells. Int. J. Hematol. 94, 109–117 (2011).

    PubMed  PubMed Central  Google Scholar 

  23. Konopleva, M. Y. & Jordan, C. T. Leukemia stem cells and microenvironment: biology and therapeutic targeting. J. Clin. Oncol. 29, 591–599 (2011).

    PubMed  PubMed Central  Google Scholar 

  24. Sands, W. A., Copland, M. & Wheadon, H. Targeting self-renewal pathways in myeloid malignancies. Cell Commun. Signal 11, 33 (2013)10.1186/1478-811X-11-33

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zheng, J. et al. Inhibitory receptors bind Angptls and support blood stem cells and leukemia development. Nature 485, 656–660 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Takai, T., Nakamura, A. & Endo, S. Role of PIR-B in autoimmune glomerulonephritis. J. Biomed. Biotechnol. 2011, 275302 (2011).

    PubMed  Google Scholar 

  27. Daeron, M., Jaeger, S., Du Pasquier, L. & Vivier, E. Immunoreceptor tyrosine-based inhibition motifs: a quest in the past and future. Immunol. Rev. 224, 11–43 (2008).

    CAS  PubMed  Google Scholar 

  28. Kim, T. et al. Human LilrB2 is a beta-amyloid receptor and its murine homolog PirB regulates synaptic plasticity in an Alzheimer’s model. Science 341, 1399–1404 (2013).

    CAS  PubMed  Google Scholar 

  29. Tang, X. et al. Leukocyte-associated Ig-like receptor-1-deficient mice have an altered immune cell phenotype. J. Immunol. 188, 548–558 (2012).

    CAS  PubMed  Google Scholar 

  30. Olde Nordkamp, M. J. et al. Leukocyte-associated Ig-like receptor-1 is a novel inhibitory receptor for surfactant protein D. J. Leukoc. Biol. 96, 105–111 (2014).

    PubMed  Google Scholar 

  31. Meyaard, L. LAIR and collagens in immune regulation. Immunol. Lett. 128, 26–28 (2010).

    CAS  PubMed  Google Scholar 

  32. Maasho, K. et al. The inhibitory leukocyte-associated Ig-like receptor-1 (LAIR-1) is expressed at high levels by human naive T cells and inhibits TCR mediated activation. Mol. Immunol. 42, 1521–1530 (2005).

    CAS  PubMed  Google Scholar 

  33. Krivtsov, A. V. et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature 442, 818–822 (2006).

    CAS  PubMed  Google Scholar 

  34. Somervaille, T. C. & Cleary, M. L. Identification and characterization of leukemia stem cells in murine MLL-AF9 acute myeloid leukemia. Cancer Cell 10, 257–268 (2006).

    CAS  PubMed  Google Scholar 

  35. Yan, M. et al. A previously unidentified alternatively spliced isoform of t(8;21) transcript promotes leukemogenesis. Nat. Med. 12, 945–949 (2006).

    CAS  PubMed  Google Scholar 

  36. Sugihara, E. et al. Ink4a and Arf are crucial factors in the determination of the cell of origin and the therapeutic sensitivity of Myc-induced mouse lymphoid tumor. Oncogene 31, 2849–2861 (2012).

    CAS  PubMed  Google Scholar 

  37. Tapley, P. et al. Increased G-CSF responsiveness of bone marrow cells from hematopoietic cell phosphatase deficient viable motheaten mice. Exp. Hematol. 25, 122–131 (1997).

    CAS  PubMed  Google Scholar 

  38. Paulson, R. F., Vesely, S., Siminovitch, K. A. & Bernstein, A. Signalling by the W/Kit receptor tyrosine kinase is negatively regulated in vivo by the protein tyrosine phosphatase Shp1. Nat. Genet. 13, 309–315 (1996).

    CAS  PubMed  Google Scholar 

  39. Lorenz, U. et al. Genetic analysis reveals cell type-specific regulation of receptor tyrosine kinase c-Kit by the protein tyrosine phosphatase SHP1. J. Exp. Med. 184, 1111–1126 (1996).

    CAS  PubMed  Google Scholar 

  40. Timms, J. F. et al. Identification of major binding proteins and substrates for the SH2-containing protein tyrosine phosphatase SHP-1 in macrophages. Mol. Cell. Biol. 18, 3838–3850 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Minoo, P., Zadeh, M. M., Rottapel, R., Lebrun, J. J. & Ali, S. A novel SHP-1/Grb2-dependent mechanism of negative regulation of cytokine-receptor signaling: contribution of SHP-1 C-terminal tyrosines in cytokine signaling. Blood 103, 1398–1407 (2004).

    CAS  PubMed  Google Scholar 

  42. Xu, M., Zhao, R. & Zhao, Z. J. Identification and characterization of leukocyte-associated Ig-like receptor-1 as a major anchor protein of tyrosine phosphatase SHP-1 in hematopoietic cells. J. Biol. Chem. 275, 17440–17446 (2000).

    CAS  PubMed  Google Scholar 

  43. Shankar, D. B. et al. The role of CREB as a proto-oncogene in hematopoiesis and in acute myeloid leukemia. Cancer Cell 7, 351–362 (2005).

    CAS  PubMed  Google Scholar 

  44. Liu, F., Thompson, M. A., Wagner, S., Greenberg, M. E. & Green, M. R. Activating transcription factor-1 can mediate Ca(2 +)- and cAMP-inducible transcriptional activation. J. Biol. Chem. 268, 6714–6720 (1993).

    CAS  PubMed  Google Scholar 

  45. Tyson, D. R., Swarthout, J. T., Jefcoat, S. C. & Partridge, N. C. PTH induction of transcriptional activity of the cAMP response element-binding protein requires the serine 129 site and glycogen synthase kinase-3 activity, but not casein kinase II sites. Endocrinology 143, 674–682 (2002).

    CAS  PubMed  Google Scholar 

  46. Xiao, X., Li, B. X. & Xie, F. Pharmaceutical compositions comprising naphthamides as antitumor agents, WO 2013067379 (2012)

  47. Li, B. X. & Xiao, X. Discovery of a small-molecule inhibitor of the KIX-KID interaction. Chembiochem 10, 2721–2724 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Eklund, E. A., Goldenberg, I., Lu, Y., Andrejic, J. & Kakar, R. SHP1 protein-tyrosine phosphatase regulates HoxA10 DNA binding and transcriptional repression activity in undifferentiated myeloid cells. J. Biol. Chem. 277, 36878–36888 (2002).

    CAS  PubMed  Google Scholar 

  49. Tibaldi, E. et al. Lyn-mediated SHP-1 recruitment to CD5 contributes to resistance to apoptosis of B-cell chronic lymphocytic leukemia cells. Leukemia 25, 1768–1781 (2011).

    CAS  PubMed  Google Scholar 

  50. Shultz, L. D. et al. Mutations at the murine motheaten locus are within the hematopoietic cell protein-tyrosine phosphatase (Hcph) gene. Cell 73, 1445–1454 (1993).

    CAS  PubMed  Google Scholar 

  51. Picciotto, M. R., Zoli, M., Bertuzzi, G. & Nairn, A. C. Immunochemical localization of calcium/calmodulin-dependent protein kinase I. Synapse 20, 75–84 (1995).

    CAS  PubMed  Google Scholar 

  52. Stedman, D. R., Uboha, N. V., Stedman, T. T., Nairn, A. C. & Picciotto, M. R. Cytoplasmic localization of calcium/calmodulin-dependent protein kinase I-alpha depends on a nuclear export signal in its regulatory domain. FEBS Lett. 566, 275–280 (2004).

    CAS  PubMed  Google Scholar 

  53. Zhang, C. C., Kaba, M., Iizuka, S., Huynh, H. & Lodish, H. F. Angiopoietin-like 5 and IGFBP2 stimulate ex vivo expansion of human cord blood hematopoietic stem cells as assayed by NOD/SCID transplantation. Blood 111, 3415–3423 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Zheng, J. et al. Ex vivo expanded hematopoietic stem cells overcome the MHC barrier in allogeneic transplantation. Cell Stem Cell 9, 119–130 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Huynh, H. et al. IGF binding protein 2 supports the survival and cycling of hematopoietic stem cells. Blood 118, 3236–3243 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Zheng, J., Huynh, H., Umikawa, M., Silvany, R. & Zhang, C. C. Angiopoietin-like protein 3 supports the activity of hematopoietic stem cells in the bone marrow niche. Blood 117, 470–479 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank H. Saya from Keio University School of Medicine for the pMX-IG N-Myc vector. We appreciate the support of staff of the tissue bank at the Department of Hematopathology, the University of Texas MD Anderson Cancer Center. Support to C.C.Z. was from NIH grant 1R01CA172268, Leukemia & Lymphoma Society Awards 1024-14 and TRP-6024-14, CPRIT RP140402, March of Dimes Foundation grant 1-FY14-201, Robert A. Welch Foundation grant I-1834, and When Everyone Survives Foundation. J.W.T. is supported by grants from the V Foundation for Cancer Research, the William Lawrence and Blanche Hughes Fund, and the National Cancer Institute (4 R00CA151457-03), and the Leukemia & Lymphoma Society. X.X. is supported by Susan G. Komen Foundation and RO1GM087305. J.E.C. is supported by the intramural program of the National Institute of Allergy and Infectious Diseases. M.J.Y. is supported in part by NIH/NCI R01 CA164346, Ladies Leukemia League, Developmental Research Awards in Leukemia SPORE CA100632, and Center for Inflammation and Cancer, IRG, Center for Genetics and Genomics, Sister Institution Network Fund and Physician Scientist Award of the University of Texas MD Anderson Cancer Center.

Author information

Authors and Affiliations

Authors

Contributions

X.K. performed most of the experiments, analysed data and contributed to writing the paper; Z.L. performed the TCGA and clustering analyses; C.C. performed the CAMK experiments; M.D. performed the LILRB knockdown experiments. Y.F., F.X. and X.X. performed the CREB inhibitor experiments and provided advice; B.D., X.H., R.H.C. and M.J.Y. collected the primary AML samples and provided advice; J.W.T. helped with experiments using leukaemia cell lines and contributed to paper writing; J.E.C. provided LAIR1-deficient mice and contributed to paper writing; C.C.Z. conceived, coordinated and supervised the project, designed experiments, analysed data and wrote the paper.

Corresponding author

Correspondence to Cheng Cheng Zhang.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 4 Lair1, a representative ITIM receptor, is essential for the growth of human leukemia cell lines.

(a) Expression of certain human ITIM receptor mRNAs negatively correlates with the overall survival of AML patients. A total of 58 ITIM receptors were selected based on the criteria that (1) they are plasma membrane receptors and (2) they use ITIM as the main signalling motifs. Data were obtained from the TCGA AML database, and analysed without normalization (condition 1) or normalized to GADPH expression (condition 2), Affymetrix housekeeping gene expression (condition 3), or total mRNA (condition 4). More information about data analysis can be found in Methods. (b) Effects of inhibition of expression of indicated ITIM receptors using shRNAs as determined by real-time RT-PCR. Data are from a single experiment, representative of 3 independent experiments. (c) In silico analysis of the correlation between human lair1 mRNA expression and the overall survival of AML patients younger than 65 years old. Data were obtained from the TCGA AML database (n = 52 patient samples for each groups, p = 0.0271, log-rank test). (d) An in silico analysis of human lair1 mRNA expression in 43 human AML samples as described previously1. (e) SP-D is not highly expressed in the bone marrow environment compared with mRNA expression in lung epithelial cells, as determined by real-time RT-PCR. Data are from a single experiment, representative of 3 independent experiments. (f) Schematic summary of the LAIR1 chimeric receptor signalling reporter cells system. (g) In the LAIR1 chimeric receptor signalling reporter cells system, flow cytometry analysis demonstrated that, while the immobilized collagen 1 (1 μg ml−1) or anti-LAIR1 antibody induced LAIR1 activation (as shown by increased GFP induction), the immobilized or soluble SP-D (1 μg ml−1) was unable to do so (the upper panels are for control cells, and the lower panels are for the hLAIR1 reporter cells).

Supplementary Figure 5 Depletion of lair1 suppresses growth of human leukemia cell lines in vitro and in vivo.

(a) Representative images (from at least 3 similar images) showing the marked reduction of MV4-11 cell growth on treatment with shRNAs targeting lair1 at 4 days after viral infection. Scale bar is 50 μM. (b) No effects on 562 cell growth were observed on treatment with shRNAs targeting lair1. Data from one experiment with n = 3 technical replicate samples are shown. The experiment was repeated 3 times with similar results. (c) No significant cell cycle change was detected between control and lair1-deficient MV4-11 cells at 3 and 6 days after infection with virus encoding a control shRNA or virus encoding shRNA 226, respectively. Data from one experiment with n = 3 technical replicate samples are shown. The experiment was repeated 3 times with similar results. (d) GFP+ LAIR1high cells in the LAIR1 shRNA 226 knockdown samples isolated from the transplanted NSG mice contained much less LAIR1 shRNA sequences than the original GFP+ LAIR1low counterparts before transplantation. Real-time PCR was performed to quantitate the shRNA sequences from these populations by using the shRNA 226 specific primer (GCTAGTCCATCTGAGTCAG-forward) together with the vector primer (AAGCGAGCTTATCGATACCG-reverse). Data are from a single experiment, representative of 3 independent experiments. (e) Flow cytometry analysis showing the decreased engraftment of lair1-deficient MV4-11 cells in BM of individual recipient mice (scrambled control versus lair1-shRNA treated samples).

Supplementary Figure 6 Flow cytometry analysis showing the decreased engraftment of lair1-deficient 697 cells in BM of individual recipient mice (scrambled control versus lair1-shRNA treated samples).

Supplementary Figure 7 LAIR1 deficient mice maintain normal hematopoiesis.

(a) WT and lair1-null neonatal liver Lin cells (1 × 105 cells) were transplanted together with 1 × 105 CD45.1 competitor cells into lethally irradiated (10 Gy) CD45.1 mice (n = 5 mice). Peripheral blood engraftments are shown at 6 and 20 weeks after transplantation. (b) Comparison of mutilineage contribution between WT and lair1-null cells at 20 weeks after transplantation (n = 5 mice). (c) WT and lair1-null BM cells (1 × 106 cells) from primary engraftments were transplanted together with 2 × 105 CD45.1 competitor cells into lethally irradiated (10 Gy) CD45.1 mice (n = 5 mice). Peripheral blood engraftments are shown at 4 and 16 weeks after transplantation. (d) Comparison of mutilineage contributions of WT and lair1-null cells at 16 weeks after secondary transplantation (n = 5 mice). (e) WT and lair1-null HSCs home similarly to the recipient BM. BM cells from WT or lair1-null mice (n = 5 mice) were labelled with carboxyfluorescein succinimidyl ester (CFSE), and 1 × 107 cells were transplanted into lethally irradiated recipients. After 12 h, the total percentage of CFSE+ cells in the BM, spleen, and liver and LT-HSCs (CFSE+LinSca-1+Kit+Flk2CD34 cells) in BM were determined by flow cytometry.

Supplementary Figure 8 LAIR1 enhances leukemia development in several mouse leukemia models during serial transplantation.

(a) No significant difference in AML development were observed on primary transplantation with MLL-AF9-infected WT or lair1-null hematopoietic progenitors in primary transplantation; survival curves are shown (n = 10 mice; p = 0.1121, log-rank test). The experiment was repeated three times with similar results. (b) Summary of percentages of YFP+ AML cells, YFP+Mac-1+Gr-1+, and YFP+Mac-1+Kit+ cells in BM of primary recipient mice transplanted with the WT or lair1-null MLL-AF9 AML cells (n = 10 mice), the experiment was repeated three times with similar results. (c) The clonal relationship between the transplanted cells was studied by southern blotting on genomic DNA isolated from 4 pairs of WT and lair1 null MLL-AF9 induced leukemia samples. The result demonstrates that the leukemias were oligoclonal. The southern blotting was performed using a probe for YFP. (d) Southern blotting analysis of the oliogocolnal nature of MLL-AF9 integration in genomes of 3 primarily and 2 secondarily transplanted mice. The secondarily transplanted AML mice contained multiple MLL-AF9 clones from different primary samples. The bands indicated by the red number coordinates between primary and secondary samples. (e) Survival curves of mice receiving N-Myc infected WT or lair1-null hematopoietic progenitors in primary transplantation (n = 10 mice,p = 0.4204, log-rank test). (f) Survival curves of mice receiving 3,000 pooled GFP+ BM cells that were collected from primary recipients transplanted with WT or lair1-null N-Myc B-ALL cells (n = 5 mice, p = 0.0031, log-rank test). (gi) Leukemia development vanishes on secondary transplantation of lair1-null MLL-AF9 cells. Summary of percentages of YFP+ AML cells and YFP+Mac-1+Kit+, YFP+Mac-1+Gr-1+, YFP+B220+, and YFP+CD3+ cells in (f) BM, (g) PB, and (h) spleen of secondary recipient mice transplanted with the WT or lair1-null MLL-AF9 AML cells at day 28 post-transplant (mean ± s.e.m., Student’s t-test; n = 5 mice; (g) Mac1+/Gr1+ **p = 0.0015; B220+ **p = 0.0019; CD3+ ***p < 0.0001; (h) Mac1+/Gr1+ **p = 0.0031; B220+ **p = 0.0022; CD3+ **p = 0.0017; (i) Mac1+/Gr1+ **p = 0.0002; B220+**p = 0.0035; CD3+ ***p < 0.0001).

Supplementary Figure 9 SHP-1-CAMK1 rescues the lair1-null AML phenotype.

(a) Retrovirally-expressed SHP-1 increased CFU numbers of lair1-null AML cells in secondary plating. Data from one experiment with n = 3 technical replicate samples are shown. The experiment was repeated 3 times with similar results. (b) The expression of endogenous shp-1 was inhibited by Cre virus infection, as determined by Q-PCR at 48 h after infection. Data are from a single experiment, representative of 3 independent experiments. (c,d) SHP-1 inhibitors have little effect on (c) CFU activity of MLL-AF9 AML cells or (d) cell growth capacity of human AML leukemia cell line (MV4-11). Data from one experiment with n = 3 technical replicate samples are shown. The experiment was repeated 3 times with similar results. (e) Retrovirally-expressed SHP-1 WT, C453S, and 4YF(278, 303, 538, 566), but not SHP-1 PTPc, increased CFU numbers of lair1-null AML cells in secondary plating. Data from one experiment with n = 3 technical replicate samples are shown. The experiment was repeated 3 times with similar results. (f) Retrovirally-expressed CAMK1 increased CFU numbers of lair1-null AML cells in secondary plating. Data from one experiment with n = 3 technical replicate samples are shown. The experiment was repeated 3 times with similar results. (g) IP-western assay of MLL-AF9 BM cells of wide-type mouse by precipitating SHP-1, followed by detection of CAMK1 or LAIR1. (h) Additional 4 times of western-blot analyses as in Fig. 4a, showing SHP-1 protein levels in both primarily and secondarily transplanted WT and LAIR1 null leukemic mice.

Supplementary Figure 10 Transcription factor CREB is necessary for LAIR1-mediated signalling in AML cells.

(a) Retrovirally-expressed WT CREB, but not CREB S129A, S133A, or the double mutant S129/S133A, increased CFU numbers of lair1-null AML cells. Data from one experiment with n = 3 technical replicate samples are shown. The experiment was repeated 3 times with similar results. (b,c) Treatment of (b) 697 or (c) U937 cells with CREB inhibitor XX15 inhibited growth. YFP+ WT AML cells (20,000) were sorted by flow cytometry, plated in 1.55-cm wells, and treated with the indicated concentration of XX15. Cell numbers were determined on days 1, 2, and 3 from triplicate wells. Data from one experiment with n = 3 technical replicate samples are shown. The experiment was repeated 3 times with similar results. (d) Retrovirally-expressed CREB S219/133A decreased the total SHP-1 levels in the WT AML mouse bone marrow samples as shown by western blotting.

Supplementary Figure 11 LAIR1–SHP-1–CAMK1 axis supports human AML development.

(a) LAIR1high primary AML cells have greater colony-forming ability difference in both first and second plating (samples B1 and B2), whereas no significant difference in colony-forming ability was detected between LAIR1high and LAIR1low cord blood mononuclear cells (sample A). Data from one experiment with n = 3 technical replicate samples are shown. The experiment was repeated 3 times with similar results. (b) Endogenous shp-1 expression was inhibited using shRNAs (201, 698, 786) in MV4-11 cells as determined by Q-PCR at 48 h after infection. Data are from a single experiment, representative of 3 independent experiments. (c) No clear association between shp-2 expression and AML patient survival was observed. Data were obtained from the TCGA AML database (n = 82 patient samples for high or n = 83 patient samples for low, p = 0.9451, log-rank test). (d) Endogenous camk1 expression was inhibited using an shRNA in MV4-11 cells as determined by Q-PCR at 48 h after infection. Data are from a single experiment, representative of 3 independent experiments. (e) LAIR1 expression is independent of the selected human AML stem cell phenotypic markers. The expression of LAIR1 and phenotypic markers (CD34/CD38/CD90) were analysed by flow cytometry in four AML clinical samples. (f) LAIR1 knockdown decreased colony-forming ability in all seven tested primary human AML cells as determined by CFU assays. Data from one experiment with n = 3 technical replicate samples are shown. The experiment was repeated 3 times with similar results. (g) The survival curves of mice receiving control or LAIR1-knockdown primary human patient AML cells (sample# 6). n = 9 mice; p < 0.0001, log-rank test. (h) Schematic summary of the novel signalling pathway mediated by the ITIM receptor LAIR1 in leukemia cells.

Supplementary Figure 12 Original uncropped images of western blots.

Supplementary Table 1 In silico analysis of the correlation between human ITIM domain-containing receptor mRNA expression and the overall survival of AML patients.
Supplementary Table 2 shRNA targeting sequences for the indicated ITIM receptors.
Supplementary Table 3 Clinical sample information.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1019 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kang, X., Lu, Z., Cui, C. et al. The ITIM-containing receptor LAIR1 is essential for acute myeloid leukaemia development. Nat Cell Biol 17, 665–677 (2015). https://doi.org/10.1038/ncb3158

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3158

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