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:

Neuropathy of haematopoietic stem cell niche is essential for myeloproliferative neoplasms

Abstract

Myeloproliferative neoplasms (MPNs) are diseases caused by mutations in the haematopoietic stem cell (HSC) compartment. Most MPN patients have a common acquired mutation of Janus kinase 2 (JAK2) gene in HSCs1,2,3,4 that renders this kinase constitutively active, leading to uncontrolled cell expansion. The bone marrow microenvironment might contribute to the clinical outcomes of this common event. We previously showed that bone marrow nestin+ mesenchymal stem cells (MSCs) innervated by sympathetic nerve fibres regulate normal HSCs5,6. Here we demonstrate that abrogation of this regulatory circuit is essential for MPN pathogenesis. Sympathetic nerve fibres, supporting Schwann cells and nestin+ MSCs are consistently reduced in the bone marrow of MPN patients and mice expressing the human JAK2(V617F) mutation in HSCs. Unexpectedly, MSC reduction is not due to differentiation but is caused by bone marrow neural damage and Schwann cell death triggered by interleukin-1β produced by mutant HSCs. In turn, in vivo depletion of nestin+ cells or their production of CXCL12 expanded mutant HSC number and accelerated MPN progression. In contrast, administration of neuroprotective or sympathomimetic drugs prevented mutant HSC expansion. Treatment with β3-adrenergic agonists that restored the sympathetic regulation of nestin+ MSCs5,6 prevented the loss of these cells and blocked MPN progression by indirectly reducing the number of leukaemic stem cells. Our results demonstrate that mutant-HSC-driven niche damage critically contributes to disease manifestation in MPN and identify niche-forming MSCs and their neural regulation as promising therapeutic targets.

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: Apoptosis of BM nestin+ HSC niche cells contributes to MPN progression.
Figure 2: BM Schwann cell death triggered by HSPC-derived interleukin-1β precedes nestin+ MSC apoptosis in MPN.
Figure 3: Treatment with β3-adrenergic agonist or neuroprotective drug blocks MPN progression.
Figure 4: Compensation of BM neuropathy rescues MSCs and prevents mutant HSC expansion in MPN.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

The gene expression data have been deposited in the Gene Expression Omnibus (GEO) databank (http://www.ncbi.nlm.nih.gov/geo) under the accession number GSE55802.

References

  1. James, C. et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 434, 1144–1148 (2005)

    Article  CAS  ADS  Google Scholar 

  2. Kralovics, R. et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N. Engl. J. Med. 352, 1779–1790 (2005)

    Article  CAS  Google Scholar 

  3. Levine, R. L. et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 7, 387–397 (2005)

    Article  CAS  Google Scholar 

  4. Baxter, E. J. et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 365, 1054–1061 (2005)

    Article  CAS  Google Scholar 

  5. Méndez-Ferrer, S. et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 466, 829–834 (2010)

    Article  ADS  Google Scholar 

  6. Méndez-Ferrer, S., Lucas, D., Battista, M. & Frenette, P. S. Haematopoietic stem cell release is regulated by circadian oscillations. Nature 452, 442–447 (2008)

    Article  ADS  Google Scholar 

  7. Raaijmakers, M. H. et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature 464, 852–857 (2010)

    Article  CAS  ADS  Google Scholar 

  8. Walkley, C. R. et al. A microenvironment-induced myeloproliferative syndrome caused by retinoic acid receptor gamma deficiency. Cell 129, 1097–1110 (2007)

    Article  CAS  Google Scholar 

  9. Walkley, C. R., Shea, J. M., Sims, N. A., Purton, L. E. & Orkin, S. H. Rb regulates interactions between hematopoietic stem cells and their bone marrow microenvironment. Cell 129, 1081–1095 (2007)

    Article  CAS  Google Scholar 

  10. Colmone, A. et al. Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells. Science 322, 1861–1865 (2008)

    Article  CAS  ADS  Google Scholar 

  11. Iriuchishima, H. et al. Neovascular niche for human myeloma cells in immunodeficient mouse bone. PLoS ONE 7, e30557 (2012)

    Article  CAS  ADS  Google Scholar 

  12. Pikman, Y. et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 3, e270 (2006)

    Article  Google Scholar 

  13. Pardanani, A. D. et al. MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients. Blood 108, 3472–3476 (2006)

    Article  CAS  Google Scholar 

  14. Klampfl, T. et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N. Engl. J. Med. 369, 2379–2390 (2013)

    Article  CAS  Google Scholar 

  15. Nangalia, J. et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N. Engl. J. Med. 369, 2391–2405 (2013)

    Article  CAS  Google Scholar 

  16. Shih, A. H., Abdel-Wahab, O., Patel, J. P. & Levine, R. L. The role of mutations in epigenetic regulators in myeloid malignancies. Nature Rev. Cancer 12, 599–612 (2012)

    Article  CAS  Google Scholar 

  17. Vannucchi, A. M. et al. Mutations and prognosis in primary myelofibrosis. Leukemia 27, 1861–1869 (2013)

    Article  CAS  Google Scholar 

  18. Isern, J. et al. Self-renewing human bone marrow mesenspheres promote hematopoietic stem cell expansion. Cell Rep. 3, 1714–1724 (2013)

    Article  CAS  Google Scholar 

  19. Tiedt, R. et al. Ratio of mutant JAK2-V617F to wild-type Jak2 determines the MPD phenotypes in transgenic mice. Blood 111, 3931–3940 (2008)

    Article  CAS  Google Scholar 

  20. Kubovcakova, L. et al. Differential effects of hydroxyurea and INC424 on mutant allele burden and myeloproliferative phenotype in a JAK2-V617F polycythemia vera mouse model. Blood 121, 1188–1199 (2013)

    Article  CAS  Google Scholar 

  21. Schepers, K. et al. Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche. Cell Stem Cell 13, 285–299 (2013)

    Article  CAS  Google Scholar 

  22. Yamazaki, S. et al. Nonmyelinating Schwann cells maintain hematopoietic stem cell hibernation in the bone marrow niche. Cell 147, 1146–1158 (2011)

    Article  CAS  Google Scholar 

  23. Nagasawa, T. et al. Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature 382, 635–638 (1996)

    Article  CAS  ADS  Google Scholar 

  24. Tzeng, Y. S. et al. Loss of Cxcl12/Sdf-1 in adult mice decreases the quiescent state of hematopoietic stem/progenitor cells and alters the pattern of hematopoietic regeneration after myelosuppression. Blood 117, 429–439 (2011)

    Article  CAS  Google Scholar 

  25. Rameshwar, P., Denny, T. N., Stein, D. & Gascon, P. Monocyte adhesion in patients with bone marrow fibrosis is required for the production of fibrogenic cytokines. Potential role for interleukin-1 and TGF-beta. J. Immunol. 153, 2819–2830 (1994)

    CAS  PubMed  Google Scholar 

  26. Lucas, D. et al. Chemotherapy-induced bone marrow nerve injury impairs hematopoietic regeneration. Nature Med. 19, 695–703 (2013)

    Article  CAS  ADS  Google Scholar 

  27. Mignone, J. L., Kukekov, V., Chiang, A. S., Steindler, D. & Enikolopov, G. Neural stem and progenitor cells in nestin-GFP transgenic mice. J. Comp. Neurol. 469, 311–324 (2004)

    Article  CAS  Google Scholar 

  28. Balordi, F. & Fishell, G. Mosaic removal of hedgehog signaling in the adult SVZ reveals that the residual wild-type stem cells have a limited capacity for self-renewal. J. Neurosci. 27, 14248–14259 (2007)

    Article  CAS  Google Scholar 

  29. Sousa, V. H., Miyoshi, G., Hjerling-Leffler, J., Karayannis, T. & Fishell, G. Characterization of Nkx6-2-derived neocortical interneuron lineages. Cereb. Cortex 19 (Suppl. 1). i1–i10 (2009)

    Article  Google Scholar 

  30. Brockschnieder, D., Pechmann, Y., Sonnenberg-Riethmacher, E. & Riethmacher, D. An improved mouse line for Cre-induced cell ablation due to diphtheria toxin A, expressed from the Rosa26 locus. Genesis 44, 322–327 (2006)

    Article  CAS  Google Scholar 

  31. Frenette, P. S., Subbarao, S., Mazo, I. B., von Andrian, U. H. & Wagner, D. D. Endothelial selectins and vascular cell adhesion molecule-1 promote hematopoietic progenitor homing to bone marrow. Proc. Natl Acad. Sci. USA 95, 14423–14428 (1998)

    Article  CAS  ADS  Google Scholar 

  32. Stemple, D. L. & Anderson, D. J. Isolation of a stem cell for neurons and glia from the mammalian neural crest. Cell 71, 973–985 (1992)

    Article  CAS  Google Scholar 

  33. Pajtler, K. et al. Production of chick embryo extract for the cultivation of murine neural crest stem cells. J. Vis. Exp. 45, e2380 (2010)

    Google Scholar 

  34. Biernaskie, J. A., McKenzie, I. A., Toma, J. G. & Miller, F. D. Isolation of skin-derived precursors (SKPs) and differentiation and enrichment of their Schwann cell progeny. Nature Protocols 1, 2803–2812 (2007)

    Article  Google Scholar 

  35. Davis, S. & Meltzer, P. S. GEOquery: a bridge between the Gene Expression Omnibus (GEO) and BioConductor. Bioinformatics 23, 1846–1847 (2007)

    Article  Google Scholar 

  36. Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127 (2007)

    Article  Google Scholar 

Download references

Acknowledgements

We thank S. Martín-Salamanca, A. M. Martín, A. B. Ricote, J. M. Ligos, S. Bartlett and the CNIC Comparative Medicine, Genomics and Bioinformatics Units for assistance; members of S.M.-F. and B. Ibáñez groups and M. García-Fernández for data discussion; G. Enikolopov, G. Fishell and D. Riethmacher for providing mice. This work was supported by Fundación CNIC, Spanish Ministry of Economy and Competitiveness (TerCel, Spanish Cell Therapy Network; Plan Nacional SAF-2011-30308) and ConSEPOC-Comunidad de Madrid S2010/BMD-2542 grants to S.M.-F.; Ramón y Cajal Program grants RYC-2009-04703/2011-09726 and Marie Curie grants FP7-PEOPLE-2011-RG-294262/294096 to A.S.-A. and S.M.-F.; Swiss National Science Foundation 310000-120724/1, 32003BB_135712/1 and Swiss Cancer League KLS-02398-02-2009 grants to R.C.S.; A.S.-A. received a Research Fellowship from the European Hematology Association. S.M.-F. is supported in part by an International Early Career Scientist grant of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

L.A. designed and performed most experiments and analyses, prepared figures and wrote the manuscript. A.S.-A. performed in vivo transplantation assays. D.M.-P. performed qPCR, genome-wide expression analyses and together with X.L. provided technical assistance. J.I. performed neonatal cell isolation and culture. X.L. and A.T. performed histological analyses. S.M. provided human samples. Y.-S.T. and D.-M.L. provided Cxcl12-floxed mice. P.L., J.S. and R.C.S. provided samples, designed experiments and human studies. S.M.-F. designed the overall study, supervised the experiments and wrote the manuscript.

Corresponding author

Correspondence to Simón Méndez-Ferrer.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 BM MSC reduction in compound mutant mice and recipients of mutant haematopoietic cells is not due to cell differentiation.

a, b, Blood counts of Nes-gfp;Mx1-cre;JAK2(V617F) and control mice 4-8 weeks after pIpC treatment (a) and Nes-gfp mice 10-12 weeks after transplantation with BM cells from MPN and control mice (b). Note the similar erythrocytosis, neutrophilia and thrombocytosis in compound mutant mice and Nes-gfp mice transplanted with mutant cells. Each dot represents a mouse. c, GFP fluorescence in skull BM of Nes-gfp mice 6–8 weeks after transplantation with MPN and control BM cells (n = 8; scale bar, 100 µm). d, Frequency of CD45CD31Ter119Nes-GFP+ BM cells in MPN (n = 9) and control (n = 7) mice 6–8 weeks after pIpC induction. e, Giemsa-stained fibroblastic colony-forming units (CFU-F) from immunomagnetically-enriched BM CD45Ter119 cells 30 weeks after transplantation with MPN and control BM cells. f, FACS analyses of BM CD45CD31Ter119 cells from MPN and control mice 8 weeks after pIpC treatment (n = 3). The specified MSC markers were used. Mean ± s.e.m. g, BM reticulin staining; MPN mice showed incipient fibrosis (arrow) 6 weeks after pIpC treatment (magnification, ×200). h, i, Lineage-tracing studies of BM nestin+ cells in MPN. h, Experimental design. Nes-creERT2;RCE:loxP mice were transplanted with MPN and control BM cells and fed with tamoxifen diet. Disease development was monitored over 28 weeks. i, Blood counts showing progressive neutrophilia and thrombocytosis in recipients of MPN BM cells (n = 3). j, Femoral haematoxylin and eosin stainings showing abnormal bone formation in recipients of MPN BM cells (magnification, ×100). *P < 0.05, **P < 0.01, ***P < 0.001 (unpaired two-tailed t test).

Extended Data Figure 2 Treatment with the JAK1/2 inhibitor ruxolitinib reduces haematopoietic cell expansion in MPN but it does not rescue BM MSCs.

Nes-gfp mice were transplanted with BM cells from pIpC-induced Mx1-cre;JAK2(V617F) mice and treated with ruxolitinib (n = 4) or vehicle (n = 6) over 8 weeks, starting 2 weeks after transplantation. a, Peripheral blood counts. b, Frequency of immunophenotypically-defined BM CD45CD31Ter119 cells. Mean ± s.e.m. *P < 0.05 (unpaired two-tailed t test).

Extended Data Figure 3 Depletion of nestin+ cells or their Cxcl12 production accelerates MPN.

a, Experimental design for in vivo nestin+ cell depletion. Nes-creERT2;iDTA and control iDTA mice were transplanted with BM cells from pIpC-treated Mx1-cre;JAK2(V617F) and control mice and treated with tamoxifen (n = 3). be, Unlike Schwann cells, MSCs are reduced in Nes-creERT2;iDTA BM. b, c, Frequency of BM CD45CD31Ter119CD90+ cells (b) and fibroblastic colony-forming units (CFU-F, c) in BM CD45Ter119 cells from Nes-creERT2;iDTA and control mice after 4-week tamoxifen treatment. d, Expression of glial fibrillary acidic protein (Gfap) mRNA (normalized to Gapdh) in the BM of mice in a. e, Quantification of Gfap immunostaining of femoral BM from mice in b, c. fk, Early mutant HSC expansion and mobilisation correlates with reduced Cxcl12 expression in BM nestin+ cells. f, g, BM linsca-1+c-kit+ (f) CD34Flt3 long-term (LT) and CD34+Flt3 short-term (ST) HSCs, and CD34Flt3 multipotent progenitors (MPP) and (g) frequency of haematopoietic colony-forming units in culture (CFU-C) from BM nucleated cells, spleen or peripheral blood from Nes-Gfp;Mx1-cre;JAK2(V617F) (n = 6) and control (n = 11) mice 6 weeks after pIpC treatment. h, Appearance (inset; scale bar, 1 cm) and haematoxylin and eosin stainings of spleens (magnification, ×100). ik, Cxcl12 protein (i) and mRNA levels (j, k) in BM extracellular fluid (i), nucleated cells (j) and stromal Nes-GFP+ cells (k) of Mx1-cre;JAK2(V617F) and control mice 6 weeks after pIpC treatment (i, j) or Nes-gfp mice 10 weeks after transplantation with mutant BM (n = 4) or control cells (n = 7) (k). l, Circulating platelets in Nes-creERT2;Cxcl12fl/fl (n = 11) and control Cxcl12fl/fl littermates (n = 14) 18 weeks after transplantation with BM cells from MPN mice and 12 weeks after tamoxifen treatment. Mean ± s.e.m. *P < 0.05, **P < 0.01 (unpaired two-tailed t test).

Extended Data Figure 4 Neuroglial damage in MPN-affected BM stroma.

ad, Expression of neuroglial gene sets in MPN BM Nes-GFP+ cells. ac, Selected transcript expression in fragments per kilobase of exon per million fragments mapped (FPKM) of mesenchymal genes (a), HSC-niche related genes (b) and neural-related genes (c). RNA-Seq in CD45CD31Ter119GFP+ cells isolated from compound MPN and control mice 6 weeks after pIpC treatment (samples pooled from 3 mice). d, Gene set enrichment analyses (GSEA) of RNA-Seq data. e, f, Enrichment plot of coordinated changes in neuroactive ligand-receptor interactions (e) and the transcriptome of neurons, astrocytes and oligodendrocytes (f). g, h, BM Nes-GFP+ cells are different from sympathetic nerve fibres and mature Schwann cells. Immunostaining of tyrosine hydroxylase (TH) to visualize sympathetic nerve fibres (g) and glial fibrillary acidic protein (GFAP) for mature Schwann cells (h) in Nes-gfp BM. Note the close association of Nes-GFP+ (green) cells to distinctive TH+ or GFAP+ (red) cells. i, BM biopsies from (left and middle) control or (right) MPN patients immunostained with (left) secondary Ab as negative control or (middle and right) anti-TH antibodies. Nuclei were counterstained with DAPI (blue). Scale bars, 75 µm (g), 50 µm (h), 100 µm (i).

Extended Data Figure 5 Contribution of IL-1β to early MPN pathogenesis.

a, Increased BM IL-1β levels early in MPN. Multiplex ELISA analysis of pro-inflammatory (IL-1β, TNF-α, IFN-γ, IL-6), regulatory (IL-17) and anti-inflammatory (IL-10) mediators in plasma (control, n = 16; MPN, n = 13) and BM extracellular fluid samples (control, n = 11; MPN, n = 6; only IL-1β and TNF-α were detectable) from control and Mx1-cre;JAK2(V617F) mice 4–8 weeks after pIpC treatment. bf, Nes-gfp mice were transplanted with BM cells from MPN (n = 6) and control (n = 3) mice and analysed 2 weeks later. bd, mRNA expression of IL-1β (b) and its activating enzyme caspase-1 (Casp1, c), and BM frequencies of linsca-1+c-kit+ haematopoietic progenitors and CD11b+Ly-6G(1A8) monocytes (d). e, f, mRNA expression of IL-1 receptor (IL1r) (e) and its antagonist (IL1ra) (f) in BM CD45CD31Ter119Nes-GFP+/− cells. g, Number of circulating platelets in WT mice transplanted with MPN and control BM cells and treated over 18 weeks with IL1ra, starting 2 weeks after transplant. h, Frequency of BM CD45CD31Ter119CD90+ cells in mice in g. i, j, qPCR analyses of IL-1β (i) and Casp1 (j) mRNA expression in haematopoietic progenitors and monocytes isolated from the BM of mice in g. Gapdh was used as housekeeping gene (n = 5). Mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001 (unpaired two-tailed t test).

Extended Data Figure 6 Compensatory treatment with β3-adrenergic agonist BRL37344 blocks MPN progression.

a, Expression of β2- and β3-adrenergic receptors in immunomagnetically-enriched CD45+ and CD45 BM cells from control and pIpC-induced Mx1-cre;JAK2(V617F) mice (n = 3). b-c, Blood counts of WT mice (b) 4 weeks after transplantation with MPN or control BM cells, before (control, n = 11; MPN, n = 13) and (c) 4-12 weeks after chronic BRL37344 (2 mg kg−1) or vehicle treatment (i.p., twice a day; n = 5). CD11b+ monocytes and granulocytes, B220+ B cells and CD3+ T cells were determined by flow cytometry. d, Blood counts of WT mice transplanted with control BM cells and treated with BRL37344 (n = 6) or vehicle (n = 4) as described above. Mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001 (unpaired two-tailed t test).

Extended Data Figure 7 β3-adrenergic agonists or neuroprotective drugs prevent neuroglial gene induction in nestin+ cells and indirectly affect mutant haematopoietic cells.

a, Inhibition of mutant HSC expansion by β3-adrenergic agonists or neuroprotective drugs is HSC-niche dependent. Immunomagnetically-enriched CD45+ haematopoietic cells were obtained from the BM of Nes-gfp;Mx1-cre;JAK2(V617F) mice and control littermates 16 weeks after pIpC treatment (n = 3). BRL37344 (BRL) and 4-methylcatechol (4MC) were added in vitro at the indicated concentrations and the frequency of colony-forming units in culture (CFU-C) was scored after one week in culture. b, Compensatory treatment with the human β3-adrenergic agonist Mirabegron delays MPN in mice. Blood counts of WT mice 8 weeks after transplantation with MPN or control (n = 5) BM cells. Mirabegron (2 mg kg−1, n = 8) or vehicle (n = 7) treatment (i.p., twice a day) was administered the last two weeks. c, BM Nes-GFP+ cells from MPN mice treated with BRL37344 do not express neuroglial genes; mRNA expression of the Schwann cell markers myelin basic protein (Mbp), myelin-associated oligodendrocyte basic protein (Mobp) and pleckstrin homology domain containing, family B (evectins) member 1 (Pleckhb1) in BM CD45CD31Ter119GFP+ cells sorted from Nes-gfp mice treated over 8 weeks with BRL37344 or vehicle, starting 4 weeks after transplantation with MPN or control BM cells (n = 4). Mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001 (unpaired two-tailed t test).

Extended Data Figure 8 Incipient MPN signs are improved by BRL37344 or 4-methylcatechol.

a, b, Haematological parameters of WT mice 8 weeks after transplantation with BM cells from pIpC-treated control and Mx1-cre;JAK2(V617F) mice. The latter received the neuroprotective drug 4-methylcatechol (10 µg kg−1, once daily), BRL37344 (2 mg kg−1) or vehicle injections (twice a day) for the last 4 weeks (i.p.; n = 5). MPN progression was monitored in peripheral blood and mice were killed when they showed only incipient symptoms. a, Blood counts, spleen size, weight and nucleated cell number, BM nucleated cells (limbs and sternum), CD11b+ myeloid, B220+ B-lymphoid and CD3+ T cells. b, BM linsca-1+c-kit+ (LSK) haematopoietic progenitors, Ter119+CD71 mature erythroblasts and CD41+/CD42+ megakaryocyte progenitors. Mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001 (unpaired two-tailed t test).

Extended Data Figure 9 Long-term BRL37344 treatment inhibits JAK2(V617F)+ HSC expansion but it does not compromise normal haematopoiesis.

a, b, Haematopoietic progenitors quantitated by FACS (a) or colony-forming units (CFU-C) (b) in BM and blood of WT mice 12-16 weeks after transplantation with MPN (a) and WT BM cells (b), 16 weeks after BRL37344 (2 mg kg−1) or vehicle injections (twice a day, i.p.; n = 3). Mean ± s.e.m. *P < 0.05 (unpaired two-tailed t test). c, BRL37344 treatment does not change normal HSC BM number. CD45.2 WT mice were transplanted with BM cells from congenic CD45.1 WT mice, together with limiting numbers of BM cells from CD45.2 control mice treated with vehicle/BRL37344 over 4 weeks, starting 11 weeks after the transplant (n = 5). The frequency of mice that failed reconstitution 16 weeks after transplantation is plotted against the number of tested cells. HSC frequencies are indicated; n.s., non-significant; Pearson chi-squared t test.

Extended Data Figure 10 Differentially expressed genes in BM CD45CD31Ter119 GFP+ cells from Nes-gfp;Mx1-cre;JAK2(V617F) mice.

a, b, Top most upregulated (a) and downregulated (b) genes in RNaseq analyses of BM CD45CD31Ter119 GFP+ cells from Nes-gfp;Mx1-cre;JAK2(V617F) mice (red) versus control mice (blue). Plots represent differentially expressed genes in fragments per kilobase of exon per million fragments mapped (FPKM).

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Arranz, L., Sánchez-Aguilera, A., Martín-Pérez, D. et al. Neuropathy of haematopoietic stem cell niche is essential for myeloproliferative neoplasms. Nature 512, 78–81 (2014). https://doi.org/10.1038/nature13383

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13383

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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