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:

SHP-1 inhibition targets leukaemia stem cells to restore immunosurveillance and enhance chemosensitivity by metabolic reprogramming

Abstract

Leukaemia stem cells (LSCs) in acute myeloid leukaemia present a considerable treatment challenge due to their resistance to chemotherapy and immunosurveillance. The connection between these properties in LSCs remains poorly understood. Here we demonstrate that inhibition of tyrosine phosphatase SHP-1 in LSCs increases their glycolysis and oxidative phosphorylation, enhancing their sensitivity to chemotherapy and vulnerability to immunosurveillance. Mechanistically, SHP-1 inhibition leads to the upregulation of phosphofructokinase platelet (PFKP) through the AKT–β-catenin pathway. The increase in PFKP elevates energy metabolic activities and, as a consequence, enhances the sensitivity of LSCs to chemotherapeutic agents. Moreover, the upregulation of PFKP promotes MYC degradation and, consequently, reduces the immune evasion abilities of LSCs. Overall, our study demonstrates that targeting SHP-1 disrupts the metabolic balance in LSCs, thereby increasing their vulnerability to chemotherapy and immunosurveillance. This approach offers a promising strategy to overcome LSC resistance in acute myeloid leukaemia.

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

Fig. 1: SHP-1 inhibition enhances chemotherapy efficacy in MLL-AF9-induced AML mice.
Fig. 2: SHP-1 inhibition enhances chemotherapy efficacy in mouse and human AML.
Fig. 3: Ptpn6 deletion suppresses crLSCs in AML.
Fig. 4: Ptpn6 deletion upregulates PFKP in crLSCs through the AKT–β-catenin axis.
Fig. 5: Ptpn6 deletion elevates energy metabolism and chemosensitivity in a PFKP-dependent manner.
Fig. 6: Ptpn6 deletion restores innate immunosurveillance in crLSCs.
Fig. 7: Ptpn6 deletion promotes innate immunosurveillance in crLSCs through the PFKP–MYC axis.
Fig. 8: The metabolic regulation of crLSCs and cycling LSCs through the SHP-1–PFKP axis.

Similar content being viewed by others

Data availability

scRNA-seq supporting the findings of this study have been deposited at the GEO under accession code GSE244234. Clinical data were obtained from the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) and The Cancer Genome Atlas (TCGA) database of the AML project. The paired human AML scRNA-seq data were obtained from Zenodo (https://zenodo.org/,4905250). The paired human AML RNA-seq data were obtained from GEO studies GSE199451 and GSE83533. The original western blot images have been provided and are publicly available. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

Code availability

All codes and packages used in this study are either publicly available or are available at GitHub (https://github.com/fruitman1984/SHP1-2023).

References

  1. Ferrara, F. & Schiffer, C. A. Acute myeloid leukaemia in adults. Lancet 381, 484–495 (2013).

    Article  PubMed  Google Scholar 

  2. Kreso, A. & Dick, J. E. Evolution of the cancer stem cell model. Cell Stem Cell 14, 275–291 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Shlush, L. I. et al. Tracing the origins of relapse in acute myeloid leukaemia to stem cells. Nature 547, 104–108 (2017).

    Article  ADS  CAS  PubMed  Google Scholar 

  4. 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).

    Article  CAS  PubMed  Google Scholar 

  5. Vacchelli, E. et al. Chemotherapy-induced antitumor immunity requires formyl peptide receptor 1. Science 350, 972–978 (2015).

    Article  CAS  PubMed  Google Scholar 

  6. Zitvogel, L., Galluzzi, L., Smyth, M. J. & Kroemer, G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity 39, 74–88 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Paczulla, A. M. et al. Absence of NKG2D ligands defines leukaemia stem cells and mediates their immune evasion. Nature 572, 254–259 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Miao, Y. et al. Adaptive immune resistance emerges from tumor-initiating stem cells. Cell 177, 1172–1186 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lagadinou, E. D. et al. BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell Stem Cell 12, 329–341 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Ye, H. et al. Leukemic stem cells evade chemotherapy by metabolic adaptation to an adipose tissue niche. Cell Stem Cell 19, 23–37 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Jones, C. L. et al. Inhibition of amino acid metabolism selectively targets human leukemia stem cells. Cancer Cell 35, 333–335 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Brand, A. et al. LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 24, 657–671 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Cairns, R. A., Harris, I. S. & Mak, T. W. Regulation of cancer cell metabolism. Nat. Rev. Cancer 11, 85–95 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Boyd, A. L. et al. Identification of chemotherapy-induced leukemic-regenerating cells reveals a transient vulnerability of human AML recurrence. Cancer Cell 34, 483–498 (2018).

    Article  CAS  PubMed  Google Scholar 

  15. Duy, C. et al. Chemotherapy induces senescence-like resilient cells capable of initiating AML recurrence. Cancer Discov. 11, 1542–1561 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zeng, A. G. X. et al. A cellular hierarchy framework for understanding heterogeneity and predicting drug response in acute myeloid leukemia. Nat. Med. 28, 1212–1223 (2022).

    Article  CAS  PubMed  Google Scholar 

  17. Neel, B. G., Gu, H. & Pao, L. The ‘Shp’ing news: SH2 domain-containing tyrosine phosphatases in cell signaling. Trends Biochem. Sci. 28, 284–293 (2003).

    Article  CAS  PubMed  Google Scholar 

  18. Ostman, A., Hellberg, C. & Böhmer, F. D. Protein-tyrosine phosphatases and cancer. Nat. Rev. Cancer 6, 307–320 (2006).

    Article  PubMed  Google Scholar 

  19. Jiang, L. et al. SHP-1 regulates hematopoietic stem cell quiescence by coordinating TGF-β signaling. J. Exp. Med. 215, 1337–1347 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Xiao, W., Ando, T., Wang, H. Y., Kawakami, Y. & Kawakami, T. Lyn- and PLC-β3-dependent regulation of SHP-1 phosphorylation controls Stat5 activity and myelomonocytic leukemia-like disease. Blood 116, 6003–6013 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Reich, D. et al. SHP1 regulates a STAT6-ITGB3 axis in FLT3ITD-positive AML cells. Leukemia 34, 1444–1449 (2020).

    Article  PubMed  Google Scholar 

  22. Kang, X. et al. The ITIM-containing receptor LAIR1 is essential for acute myeloid leukaemia development. Nat. Cell Biol. 17, 665–677 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Corral, J. et al. An Mll-AF9 fusion gene made by homologous recombination causes acute leukemia in chimeric mice: a method to create fusion oncogenes. Cell 85, 853–861 (1996).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  25. 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).

    Article  CAS  PubMed  Google Scholar 

  26. Kundu, S. et al. Novel SHP-1 inhibitors tyrosine phosphatase inhibitor-1 and analogs with preclinical anti-tumor activities as tolerated oral agents. J. Immunol. 184, 6529–6536 (2010).

    Article  CAS  PubMed  Google Scholar 

  27. Mohr, S. et al. Hoxa9 and Meis1 cooperatively induce addiction to Syk signaling by suppressing miR-146a in acute myeloid leukemia. Cancer Cell 31, 549–562 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Boyer, M. W. et al. The role of B7 costimulation by murine acute myeloid leukemia in the generation and function of a CD8+ T-cell line with potent in vivo graft-versus-leukemia properties. Blood 89, 3477–3485 (1997).

    Article  CAS  PubMed  Google Scholar 

  29. 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).

    Article  CAS  PubMed  Google Scholar 

  30. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Chen, R. et al. Regulation of Akt/PKB activation by tyrosine phosphorylation. J. Biol. Chem. 276, 31858–31862 (2001).

    Article  CAS  PubMed  Google Scholar 

  32. Luan, B. et al. Deficiency of a β-arrestin-2 signal complex contributes to insulin resistance. Nature 457, 1146–1149 (2009).

    Article  ADS  CAS  PubMed  Google Scholar 

  33. Zheng, Y., Peng, M., Wang, Z., Asara, J. M. & Tyner, A. L. Protein tyrosine kinase 6 directly phosphorylates AKT and promotes AKT activation in response to epidermal growth factor. Mol. Cell. Biol. 30, 4280–4292 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. He, X. C. et al. PTEN-deficient intestinal stem cells initiate intestinal polyposis. Nat. Genet. 39, 189–198 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Perry, J. M. et al. Overcoming Wnt-β-catenin dependent anticancer therapy resistance in leukaemia stem cells. Nat. Cell Biol. 22, 689–700 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Casey, S. C., Baylot, V. & Felsher, D. W. The MYC oncogene is a global regulator of the immune response. Blood 131, 2007–2015 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Dhanasekaran, R. et al. The MYC oncogene—the grand orchestrator of cancer growth and immune evasion. Nat. Rev. Clin. Oncol. 19, 23–36 (2022).

    Article  CAS  PubMed  Google Scholar 

  38. Devaiah, B. N. et al. MYC protein stability is negatively regulated by BRD4. Proc. Natl Acad. Sci. USA 117, 13457–13467 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Gregory, M. A., Qi, Y. & Hann, S. R. Phosphorylation by glycogen synthase kinase-3 controls c-myc proteolysis and subnuclear localization. J. Biol. Chem. 278, 51606–51612 (2003).

    Article  CAS  PubMed  Google Scholar 

  40. Vander Heiden, M. G. & DeBerardinis, R. J. Understanding the intersections between metabolism and cancer biology. Cell 168, 657–669 (2017).

    Article  PubMed Central  Google Scholar 

  41. Li, X. et al. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat. Rev. Clin. Oncol. 16, 425–441 (2019).

    Article  CAS  PubMed  Google Scholar 

  42. Lechman, E. R. et al. miR-126 regulates distinct self-renewal outcomes in normal and malignant hematopoietic stem cells. Cancer Cell 29, 214–228 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Naka, K. et al. TGF-β-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia. Nature 463, 676–680 (2010).

    Article  ADS  CAS  PubMed  Google Scholar 

  44. Sykes, S. M. et al. AKT/FOXO signaling enforces reversible differentiation blockade in myeloid leukemias. Cell 146, 697–708 (2011).

    Article  CAS  PubMed  Google Scholar 

  45. Jaiswal, S. et al. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell 138, 271–285 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Chen, J. et al. SLAMF7 is critical for phagocytosis of haematopoietic tumour cells via Mac-1 integrin. Nature 544, 493–497 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  47. Feng, M. et al. Phagocytosis checkpoints as new targets for cancer immunotherapy. Nat. Rev. Cancer 19, 568–586 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lee, K. M. et al. MYC and MCL1 cooperatively promote chemotherapy-resistant breast cancer stem cells via regulation of mitochondrial oxidative phosphorylation. Cell Metab. 26, 633–647 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Freeman, A. J. et al. Natural killer cells suppress T cell-associated tumor immune evasion. Cell Rep. 28, 2784–2794 (2019).

    Article  CAS  PubMed  Google Scholar 

  50. Ferrari de Andrade, L. et al. Inhibition of MICA and MICB shedding elicits NK-cell-mediated immunity against tumors resistant to cytotoxic T cells. Cancer Immunol. Res. 8, 769–780 (2020).

    Article  PubMed  Google Scholar 

  51. Liu, L. et al. An RFC4/Notch1 signaling feedback loop promotes NSCLC metastasis and stemness. Nat. Commun. 12, 2693 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  52. Li, S. et al. Distinct evolution and dynamics of epigenetic and genetic heterogeneity in acute myeloid leukemia. Nat. Med. 22, 792–799 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Cocciardi, S. et al. Clonal evolution patterns in acute myeloid leukemia with NPM1 mutation. Nat. Commun. 10, 2031 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  54. Petti, A. A. et al. Genetic and transcriptional contributions to relapse in normal karyotype acute myeloid leukemia. Blood Cancer Discov. 3, 32–49 (2022).

    Article  CAS  PubMed  Google Scholar 

  55. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Sato, N., Tamada, Y., Yu, G. & Okuno, Y. CBNplot: Bayesian network plots for enrichment analysis. Bioinformatics 38, 2959–2960 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Pinho, S. et al. VCAM1 confers innate immune tolerance on haematopoietic and leukaemic stem cells. Nat. Cell Biol. 24, 290–298 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Guzzi, N. et al. Pseudouridine-modified tRNA fragments repress aberrant protein synthesis and predict leukaemic progression in myelodysplastic syndrome. Nat. Cell Biol. 24, 299–306 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We acknowledge support from the National Key Research and Development Program of China (2022YFA1103300 and 2022YFA1104100 to Meng Zhao), the National Natural Science Foundation of China (82325002 to Meng Zhao, 92268205 to L.J., 32370845 and 82300181 to X.X., and 82300209 to Y.Y.), the Guangdong Innovative and Entrepreneurial Research Team Program (2019ZT08Y485 to L.J.), the Outstanding Youths Development Scheme of Nanfang Hospital, Southern Medical University (2020J001 to S.R.) and the Sanming Project of Medicine in Shenzhen (SZSM201911004 to Meng Zhao). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the paper.

Author information

Authors and Affiliations

Authors

Contributions

X.X., Y.Y., W.Z., W.M. and C.H. designed and performed most of the experiments, analysed the data and generated the figures. Minyi Zhao and Qifa Liu contributed to the clinical data. G.Q. and J.X. contributed to the bioinformatic analysis. X.W. and Qiong Liu contributed to the plasmid construction and pull-down assays. F.T. contributed to ChIP–seq analysis. J.M.P., S.R. and X.K. contributed to the scientific discussion and paper preparation. L.J. and Meng Zhao supervised the study and wrote the paper.

Corresponding authors

Correspondence to Shuan Rao, Xunlei Kang, Meng Zhao or Linjia Jiang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Weiyi Peng and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 SHP-1 inhibition enhances the chemotherapy efficacy in AML.

a, Quantification of western blots of SHP-1 and pSHP-1 levels in MLL-AF9 AML mice with or without chemotherapy as illustrated (n = 3 mice per group) (Related to Fig. 1b). b, Quantification of SHP-1 expression in AML patients from GEO dataset. c,d, The proportion (c) and quantification (d) of SHP-1 in human LSPCs. Data were adapted from Petti et al. PMID: 35019859 (4905250, Zenodo). e, Western blots of SHP-1 and pSHP-1 in human CD34+CD38 LSPCs from paired AML patients. β-actin was a loading control. Eight individual AML patients were presented. f, Cell viability of Shp-1Δ/Δ and Shp-1f/f AML cells at 48 hours after indicated treatments (n = 3 mice per group). g-i, The cell number (g), apoptosis rate (h) of HSCs, and the colony-forming ability (i) in the bone marrow of indicated mice (n = 3 mice per group). j, Western blots of indicated proteins in Shp-1Δ/Δ AML cells from MLL-AF9-induced AML mice with or without chemotherapy as illustrated. β-actin was a loading control. 1#, 2#, and 3# indicated three individual mice. k, Cell growth of Shp-1Δ/Δ MLL-AF9 AML cells with or without SHP-2 knock-down and chemotherapy as illustrated (n = 3 individual experiments). l, SHP-2 expression in the TARGET dataset (n = 94 in remission group, n = 38 in relapse group, n = 155 in death with disease group). m, Schematic experiment strategy for MLL-AF9-induced AML mice. Chemotherapy (DOX, doxorubicin; Ara-C, cytarabine). n-o, The percentage of GFP+ leukaemic cells in peripheral blood (n), and Kaplan-Meier survival curves (o) of AML mice with indicated treatment (n = 5 mice per group). Error bars represent the means ± s.d. The P values in o were determined using Kaplan–Meier survival analysis. The P values in b, d were determined by paired t test, The P values in f were determined by unpaired two-tailed Student’s t test.The P values in a, g, h, i k, and n were determined by a one-way ANOVA followed by Dunnett’s test.

Source data

Extended Data Fig. 2 The effects of SHP-1 in LSCs and HSCs.

a, Schematic strategy to quantify the functional LSCs in Shp-1Δ/Δ and Shp-1f/f AML mice without chemotherapy. b,c, Kaplan-Meier survival curves of AML mice that received the indicated numbers of Shp-1Δ/Δ or Shp-1f/f AML cells (b), and plot of competitive repopulating units (CRUs) calculated using ELDA (c) (n = 5 mice per group). d, Cell growth of Shp-1Δ/Δ and Shp-1f/f MLL-AF9 AML cells. e, Representative images (left) and quantification (right) of the colony-forming cells of Shp-1Δ/Δ and Shp-1f/f AML cells (n = 3 individual experiments). f, Cell viability of AML cell lines after SHP-1 knock-down at 48 hours after indicated treatments. g-l, The cell number (g), apoptosis rate (h) of HSCs, and the colony-forming ability of normal cells (i) in the bone marrow; and the frequency of T cells (j), B cells (k), and myeloid cells (l) in peripheral blood of AML mice with indicated treatments (n = 3 mice per group in g, h, i and n = 5 mice per group in j, k, l). m, Western blots (left) and quantification (right) of SHP-1 and pSHP-1 levels in HSCs from normal mice and LSCs from MLL-AF9 AML mice with or without chemotherapy (n = 4 mice per group). Western blots from 2 individual mice were presented. Error bars represent the means ± s.d. The P values of c were determined using the ELDA platform. The P values d and e were determined by unpaired two-tailed Student’s t test. The P values in f, g, h, i, j, k, l and m were determined by a one-way ANOVA followed by Dunnett’s test.

Source data

Extended Data Fig. 3 The effects of SHP-1 in mouse and human AML.

a, Western blot of SHP-1 in C1498 AML cells. β-actin was a loading control. Knock-down (KD). b,c, Cell viability of C1498 AML cells at 48 hours after indicated treatments. DOX, doxorubicin; Ara-C, cytarabine. d, Schematic depicting the strategy to analyse the function of SHP-1 in Hoxa9-Meis1-induced AML mice with or without chemotherapy (doxorubicin and cytarabine). e,f, The percentage of GFP+ leukaemic cells in peripheral blood (e) and Kaplan-Meier survival curves (f) of AML mice with indicated treatments (n = 5 mice per group). g, Western blot of SHP-1 in primary AML cells with or without SHP-1 knock-down. β-actin was a loading control. 10 individual AML patients were presented. h, Representative FACS plot of human CD45+CD33+ and CD34+CD38 cells in the bone marrow of B-NDG recipients engrafted with human AML cells at 8w after transplantation. Error bars represent the means ± s.d. The P values in f were determined by Kaplan–Meier survival analysis. The P values in b, c were determined by unpaired two-tailed Student’s t test. The P values in e were determined by a one-way ANOVA followed by Dunnett’s test.

Source data

Extended Data Fig. 4 Single-cell atlas of mouse and human AML cells.

a, Schematic strategy for scRNA-seq. b, MLL-AF9 expression in 16 cell clusters, including leukaemic cell clusters crLSCs (clusters 1, 5, and 6), cycling LSCs (clusters 0, 2, and 3), Blast leukaemic cells (clusters 4, 7, 8, and 11), and normal hematopoietic cells (clusters 9, 10, 12, 13, 14, and 15). c, Representative FACS plots (left) and the absolute number (right) of Shp-1f/f bone marrow cells from AML mice for sc-RNAseq. d, The frequency of each cell clusters. e, Pearson’s correlation (colour bar) of the average gene expression profiles for cell clusters. f, UMAP of bone marrow cells from Shp-1f/f and Shp-1Δ/Δ AML mice (n = 5 mice per group). g, Pathways enriched by upregulated genes in crLSCs compared to cycling LSCs of MLL-AF9-induced AML mice. h, Pseudotime curve analysis of MLL-AF9 AML cells. i, Hoxa9-Meis1 expression in cell clusters. j, UMAP of bone marrow cells from Hoxa9-Meis1 AML mice (n = 5 mice per group). k, The frequency of cell clusters in Hoxa9-Meis1-induced AML mice. l, Cluster of differentially expressed genes (rows) in the cell clusters (columns) in Hoxa9-Meis1-induced AML cells. m, GSEA signature enrichment plots for comparison between cycling LSCs and crLSCs in Hoxa9-Meis1-induced AML mice. The P value was derived from Fisher’s exact test. n, The expression profile of ex vivo drug sensitivity genes in crLSCs and cycling LSCs in Hoxa9-Meis1-induced AML. o, Cluster of differentially expressed genes (rows) in the cell clusters (columns) in human LSPCs and cycling-LSPCs. p, GSEA signature enrichment plots for comparison between human LSPCs and cycling-LSPCs in indicated pathways. q, The expression profile of ex vivo drug sensitivity genes of human LSPCs and cycling-LSPCs. r-t, Quantification of SHP-1 expression in crLSCs and cycling LSCs from MLL-AF9 (r), Hoxa9-Meis1(s) and human (t) AML cells. Error bars represent the means ± s.d. The P values in c, r, s and t were determined by unpaired two-tailed Student’s t test.

Source data

Extended Data Fig. 5 Functional characterization of crLSCs in mouse AML.

a-c, The reactive oxygen species (ROS) level (a), oxygen consumption rate (OCR) (b), and extracellular acidification rate (ECAR) (c) in crLSCs, cycling LSCs, and Blast leukaemic cells (n = 5 mice). d, Phagocytic index by macrophages of crLSCs, cycling LSCs, and Blast leukaemic cells phagocytosed by macrophages. (n = 4 mice per group). e, Quantification of the killing efficiency of NK cells to crLSCs, cycling LSCs, and Blast leukaemic cells (n = 3 replicates from 6 mice per group). f, Representative FACS plot of crLSCs, cycling LSCs, and Blast leukaemic cells in the bone marrow of Hoxa9-Meis1-induced AML mice. g, The heatmap of stemness and differentiation genes expression levels in crLSCs, cycling LSCs, and Blast cells of Hoxa9-Meis1-induced AML mice. h, Cell viability of crLSCs and cycling LSCs from Hoxa9-Meis1-induced AML mice at 48 hours after Ara-C or DOX treatments (n = 3- mice per group). i, Schematic treatment strategy, Kaplan-Meier survival curves, and plot of competitive repopulating units (CRUs) calculated using ELDA for crLSCs and cycling LSCs from Hoxa9-Meis1-induced AML mice with or without chemotherapy (n = 5 mice per group). j-l, The absolute number (j), apoptosis rate (k), and cell cycle (l) of crLSCs, cycling LSCs, and Blast leukaemic cells in the bone marrow of Hoxa9-Meis1-induced AML mice with or without chemotherapy (n = 3-4 mice per group). m, Pathways enriched by upregulated genes in cycling LSCs compared to crLSCs in Hoxa9-Meis1 AML model. Error bars represent the means ± s.d. The P values in a, d, e, j, k and l were determined by a one-way ANOVA followed by Dunnett’s test. The P values in h were determined by unpaired two-tailed Student’s t test. The P values of i were determined using the ELDA platform.

Source data

Extended Data Fig. 6 The effects of SHP-1 and SHP-2 in AML cells.

a, GO term analysis of downregulated genes in Shp-1Δ/Δ crLSCs compared to Shp-1f/f crLSCs from MLL-AF9-induced AML mice. b, GSEA signature enrichment plots for comparison between Shp-1f/f crLSCs and Shp-1Δ/Δ crLSCs in indicated pathways in MLL-AF9 AML model. The P value was derived from Fisher’s exact test. c, The cell cycle analysis of cycling LSCs and Blast leukaemic cells in the bone marrow of Shp-1f/f or Shp-1Δ/ΔMLL-AF9-induced AML mice. d,e, The absolute number of cycling LSCs (d) and Blast leukaemic cells (e) in Shp-1f/f or Shp-1Δ/Δ AML cells from bone marrow of MLL-AF9-induced AML mice with or without chemotherapy (n = 3-5 mice per group). f-j, The relative glucose uptake (f), intracellular ATP concentration (ATP) (g), intracellular pyruvate concentration (PA) (h), extracellular lactate level (Lactate) (i) and intracellular LDH activity (LDH) (j) in MLL-AF9 AML cells with SHP-1 and/or SHP-2 knock-down with or without chemotherapy (n = 3 mice per group). k,l, Phagocytic index by macrophages of MLL-AF9 AML cells with indicated treatments (n = 5 mice per group). m, Quantification of the killing efficiency of NK cells to MLL-AF9 AML cells with indicated treatments (n = 3 mice per group). Error bars represent the means ± s.d. The P values in c, d, e, f, g, h, i, j, k, l and m were determined by a one-way ANOVA followed by Dunnett’s test.

Source data

Extended Data Fig. 7 SHP-1 regulates the AKT-β-catenin-PFKP axis.

a, The Bayesian network plot showing the relationship between pathways enriched in Shp-1Δ/Δ crLSCs compared to Shp-1f/f crLSCs from MLL-AF9-induced AML mice. b, IC50 values of MLL-AF9 AML cells with AKT inhibitor (MK2206) and chemotherapy as indicated (n = 5 replicates). DOX, doxorubicin; Ara-C, cytarabine. c, Western blots of PFKM and PFKL in Shp-1f/f and Shp-1Δ/Δ crLSCs. β-actin was a loading control. 1#, 2#, and 3# indicated three individual mice. d, Western blots of PFKP in AML cells from MLL-AF9 AML mice with or without TPI treatment. 1#, 2# and 3# indicated three individual mice. e, Schematic of experimental strategy, western blots, and quantification of AKT, β-catenin-pSer552, and PFKP proteins in MLL-AF9 AML cells with indicated treatment. 1#, 2# and 3# indicated three individual mice. β-actin was a loading control. f, Western blot of exogenously overexpressed AKT and its mutant (T308A, S473A, 2 A) proteins in 293 T cells. β-actin was a loading control. g, Westen blots of AKT and SHP-1 in GST pulldown assay. GST-SHP-1 and His-AKT were purified from bacteria. h, Western blots of indicated proteins in in vitro de-phosphorylation assay with AKT and SHP-1. HA-SHP-1 and Flag-AKT were purified from 293 T cells. i, Western blots of indicated protein with AKT, AKT mutants, and SHP-1 overexpression in 293 T cells. j, β-catenin-pSer552 bound to Pfkp genes in the promoter region. k, Relative luciferase activity of Pfkp reporter with or without β-catenin overexpression in 293 T cells. Error bars represent the means ± s.d., and the P values in b, e and k were determined by a one-way ANOVA followed by Dunnett’s test.

Source data

Extended Data Fig. 8 SHP-1-PFKP axis regulates energy metabolism and chemosensitivity of human AML cells.

a, Western blots of indicated proteins in primary AML cells from relapsed patients with SHP-1 knock-down. β-actin was a loading control. KD1 and KD2 indicated two individual SHP-1 knock-down shRNA sequences. b, Western blot of PFKP in THP-1 cells with PFKP overexpression. β-actin was a loading control. c-h, Intracellular F1,6BP concentration (c), relative glucose uptake (d), intracellular ATP concentration (ATP) (e), intracellular pyruvate concentration (PA) (f), extracellular lactate level (g), and intracellular LDH activity (LDH) (h) in THP-1 cells after PFKP overexpression. i, The cell death rate in AML cell lines at 48 hours after indicated treatments (n = 4 independent experiments). Error bars represent the means ± s.d., and the P values in c, d, e, f, g, h were determined by unpaired two-tailed Student’s t-test.

Source data

Extended Data Fig. 9 Shp-1 deletion restores innate immunosurveillance in crLSCs in a PFKP-dependent manner.

a,b, Representative FACS plots (left) and quantification (right) of CD47 (a) and CD24 (b) expression in Shp-1Δ/Δ and Shp-1f/f crLSCs with or without PFKP knock-down (n = 4-5 mice per group). c, Representative photomicrographs of phagocytosis by macrophages (left), and quantification (right) of Shp-1Δ/Δ and Shp-1f/f crLSCs (n = 6 mice per group). Scale bars, 50 μm. d, The cell viability of sorted Shp-1Δ/Δ and Shp-1f/f crLSCs with or without PFKP knock-down (n = 3 mice per group). e, Representative photomicrographs (left) and quantification (right) of phagocytic index by macrophages of Shp-1Δ/Δ and Shp-1f/f crLSCs with or without PFKP knock-down (n = 6 mice per group). Scale bars, 50 μm. f,g, The phagocytic index by macrophages (f) and the killing efficiency of NK cells (g) to THP-1 cells after SHP-1 knock-down. KD1, and KD2 indicated two individual SHP-1 knock-down shRNA (n = 3-5 independent experiments). h,i, The phagocytic index by macrophages (h) and the killing efficiency of NK cells (i) to THP-1 cells with or without PFKP overexpression (n = 3-5 independent experiments). j, The heatmap of relative mRNA expression of immune-checkpoint genes in primary AML cells. 4#, 5# idicated two individual AML patients (n = 3 replicates for each patient). k,l, The percentage of human CD45+ leukaemic cells in peripheral blood (k), and survival curves (l) of B-NDG mice with PBMC infusion and chemotherapy treatment as indicated (n = 5 mice per group). Error bars represent the means±s.d. The P values in l were determined by Kaplan–Meier survival analysis. The P values in c, h, i were determined by unpaired two-tailed Student’s t test. The P values in a, b, d, e, f, g, k were determined by a one-way ANOVA followed by Dunnett’s test.

Source data

Extended Data Fig. 10 SHP-1-PFKP axis regulates c-MYC protein and CD47 expression in AML cells.

a, Western blots of c-MYC in crLSCs, cycling LSCs, and Blast cells from MLL-AF9 model. β-actin was a loading control. 1# and 2# indicated two individual mice. b, The quantification of indicated protein levels in Shp-1Δ/Δ and Shp-1f/f MLL-AF9 AML cells with or without PFKP knock-down (Related to Fig. 7c). β-actin was a loading control. (n = 3 independent experiments). c, The quantification of PFKP, c-MYC-pT58, c-MYC-pS62, and c-MYC protein levels in THP-1 cells with PFKP knock-down or PFKP overexpression (Related to Fig. 7d, n = 4 independent experiments). d, Western blot and quantification of c-MYC in the cytoplasm and nucleus of THP-1 cells with PFKP knock-down or PFKP overexpression. e, The relative mRNA expression of c-MYC in THP-1 cells with or without PFKP knock-down and PFKP overexpression. f, Western blots of His-PFKP, HA-GSK3β, and Flag-c-MYC in His-pulldown assay. g, Western blots of GST-c-MYC and HA-PFKP in GST-pulldown assay. h, Western blots of His-GSK3β and HA-PFKP in His-pulldown assay. i, Western blot and quantification of c-MYC in the cytoplasm and nucleus of Shp-1Δ/Δ and Shp-1f/f crLSCs. j, ChIP-qPCR of CD47 promoter region with anti-c-MYC antibody or IgG control in MLL-AF9 AML cells. k, Western blots of c-MYC and SHP-1 (left) and quantification of CD47 cells (right) in Shp-1Δ/Δ and Shp-1f/f crLSCs with or without c-MYC overexpression (n = 3 mice). l, Western blots of PFKP and c-MYC (left) and quantification of CD47 cells (right) in crLSCs with or without PFKP or c-MYC overexpression (n = 3 mice). Error bars represent the means ± s.d. The P values in b, c, d, e, i, k, and l were determined by a one-way ANOVA followed by Dunnett’s test. The P values in j were determined by unpaired two-tailed Student’s t-test.

Source data

Supplementary information

Supplementary Fig. 1

Gating strategy for Figs. 1–7 and Extended Data Figs. 1–3 and 5–9.

Reporting Summary

Supplementary Table

Clinical sample information.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, X., Yu, Y., Zhang, W. et al. SHP-1 inhibition targets leukaemia stem cells to restore immunosurveillance and enhance chemosensitivity by metabolic reprogramming. Nat Cell Biol 26, 464–477 (2024). https://doi.org/10.1038/s41556-024-01349-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-024-01349-3

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