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:

Leptin-receptor-expressing bone marrow stromal cells are myofibroblasts in primary myelofibrosis

Abstract

Bone marrow fibrosis is a critical component of primary myelofibrosis (PMF). However, the origin of the myofibroblasts that drive fibrosis is unknown. Using genetic fate mapping we found that bone marrow leptin receptor (Lepr)-expressing mesenchymal stromal lineage cells expanded extensively and were the fibrogenic cells in PMF. These stromal cells downregulated the expression of key haematopoietic-stem-cell-supporting factors and upregulated genes associated with fibrosis and osteogenesis, indicating fibrogenic conversion. Administration of imatinib or conditional deletion of platelet-derived growth factor receptor a (Pdgfra) from Lepr+ stromal cells suppressed their expansion and ameliorated bone marrow fibrosis. Conversely, activation of the PDGFRA pathway in bone marrow Lepr+ cells led to expansion of these cells and extramedullary haematopoiesis, features of PMF. Our data identify Lepr+ stromal lineage cells as the origin of myofibroblasts in PMF and suggest that targeting PDGFRA signalling could be an effective way to treat bone marrow fibrosis.

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: TOE mice develop clinical features of PMF.
Figure 2: HSCs overproliferate and mobilize to the spleen in TOE mice.
Figure 3: Bone marrow mesenchymal stromal cells undergo expansion and fibrotic conversion in PMF.
Figure 4: Bone marrow mesenchymal stromal cells downregulate key HSC maintenance factors, CXCL12 and SCF.
Figure 5: Mesenchymal stromal cells upregulate fibrosis and osteogenesis genes in PMF.
Figure 6: Deletion of Pdgfra from mesenchymal stromal cells or administrating imatinib ameliorates bone marrow fibrosis.
Figure 7: Activation of the PDGFRA pathway in Lepr+ cells leads to mesenchymal stromal cell expansion and HSC mobilization.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Ding, L., Saunders, T. L., Enikolopov, G. & Morrison, S. J. Endothelial and perivascular cells maintain haematopoietic stem cells. Nature 481, 457–462 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ding, L. & Morrison, S. J. Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature 495, 231–235 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Zhou, B. O., Yue, R., Murphy, M. M., Peyer, J. G. & Morrison, S. J. Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. Cell Stem Cell 15, 154–168 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Schepers, K., Campbell, T. B. & Passegue, E. Normal and leukemic stem cell niches: insights and therapeutic opportunities. Cell Stem Cell 16, 254–267 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Abdel-Wahab, O. I. & Levine, R. L. Primary myelofibrosis: update on definition, pathogenesis, and treatment. Annu. Rev. Med. 60, 233–245 (2009).

    Article  CAS  PubMed  Google Scholar 

  6. Tefferi, A. Myeloproliferative neoplasms: a decade of discoveries and treatment advances. Am. J. Hematol. 91, 50–58 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Araki, M. et al. Activation of the thrombopoietin receptor by mutant calreticulin in CALR-mutant myeloproliferative neoplasms. Blood 127, 1307–1316 (2016).

    Article  CAS  PubMed  Google Scholar 

  8. Chachoua, I. et al. Thrombopoietin receptor activation by myeloproliferative neoplasm associated calreticulin mutants. Blood 127, 1325–1335 (2016).

    Article  CAS  PubMed  Google Scholar 

  9. Marty, C. et al. Calreticulin mutants in mice induce an MPL-dependent thrombocytosis with frequent progression to myelofibrosis. Blood 127, 1317–1324 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  12. Rampal, R. et al. Integrated genomic analysis illustrates the central role of JAK-STAT pathway activation in myeloproliferative neoplasm pathogenesis. Blood 123, e123–e133 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Jacobson, R. J., Salo, A. & Fialkow, P. J. Agnogenic myeloid metaplasia: a clonal proliferation of hematopoietic stem cells with secondary myelofibrosis. Blood 51, 189–194 (1978).

    Article  CAS  PubMed  Google Scholar 

  14. Lundberg, P. et al. Myeloproliferative neoplasms can be initiated from a single hematopoietic stem cell expressing JAK2-V617F. J. Exp. Med. 211, 2213–2230 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Papadantonakis, N., Matsuura, S. & Ravid, K. Megakaryocyte pathology and bone marrow fibrosis: the lysyl oxidase connection. Blood 120, 1774–1781 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Yan, X. Q. et al. Chronic exposure to retroviral vector encoded MGDF (mpl-ligand) induces lineage-specific growth and differentiation of megakaryocytes in mice. Blood 86, 4025–4033 (1995).

    Article  CAS  PubMed  Google Scholar 

  17. Villeval, J. L. et al. High thrombopoietin production by hematopoietic cells induces a fatal myeloproliferative syndrome in mice. Blood 90, 4369–4383 (1997).

    Article  CAS  PubMed  Google Scholar 

  18. Shivdasani, R. A., Fujiwara, Y., McDevitt, M. A. & Orkin, S. H. A lineage-selective knockout establishes the critical role of transcription factor GATA-1 in megakaryocyte growth and platelet development. EMBO J. 16, 3965–3973 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Vannucchi, A. M. et al. Development of myelofibrosis in mice genetically impaired for GATA-1 expression (GATA-1(low) mice). Blood 100, 1123–1132 (2002).

    Article  CAS  PubMed  Google Scholar 

  20. Jeremy Wen, Q. et al. Targeting megakaryocytic-induced fibrosis in myeloproliferative neoplasms by AURKA inhibition. Nat. Med. 21, 1473–1480 (2015).

    Article  CAS  Google Scholar 

  21. Bonner, J. C. Regulation of PDGF and its receptors in fibrotic diseases. Cytokine Growth Factor Rev. 15, 255–273 (2004).

    Article  CAS  PubMed  Google Scholar 

  22. Olson, L. E. & Soriano, P. Increased PDGFRα activation disrupts connective tissue development and drives systemic fibrosis. Dev. Cell 16, 303–313 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Iwayama, T. et al. PDGFRα signaling drives adipose tissue fibrosis by targeting progenitor cell plasticity. Genes Dev. 29, 1106–1119 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Gersuk, G. M., Carmel, R. & Pattengale, P. K. Platelet-derived growth factor concentrations in platelet-poor plasma and urine from patients with myeloproliferative disorders. Blood 74, 2330–2334 (1989).

    Article  CAS  PubMed  Google Scholar 

  25. Tefferi, A. Pathogenesis of myelofibrosis with myeloid metaplasia. J. Clin. Oncol. 23, 8520–8530 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Morikawa, S. et al. Prospective identification, isolation, and systemic transplantation of multipotent mesenchymal stem cells in murine bone marrow. J. Exp. Med. 206, 2483–2496 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Komada, Y. et al. Origins and properties of dental, thymic, and bone marrow mesenchymal cells and their stem cells. PLoS ONE 7, e46436 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kunisaki, Y. et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature 502, 637–643 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Pinho, S. et al. PDGFRα and CD51 mark human nestin+ sphere-forming mesenchymal stem cells capable of hematopoietic progenitor cell expansion. J. Exp. Med. 210, 1351–1367 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. 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  PubMed  PubMed Central  Google Scholar 

  32. Arranz, L. et al. Neuropathy of haematopoietic stem cell niche is essential for myeloproliferative neoplasms. Nature 512, 78–81 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Reilly, J. T. et al. Guideline for the diagnosis and management of myelofibrosis. Br. J. Haematol. 158, 453–471 (2012).

    Article  PubMed  Google Scholar 

  34. Yata, Y. et al. DNase I-hypersensitive sites enhance α1(I) collagen gene expression in hepatic stellate cells. Hepatology 37, 267–276 (2003).

    Article  CAS  PubMed  Google Scholar 

  35. Lin, S. L., Kisseleva, T., Brenner, D. A. & Duffield, J. S. Pericytes and perivascular fibroblasts are the primary source of collagen-producing cells in obstructive fibrosis of the kidney. Am. J. Pathol. 173, 1617–1627 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mederacke, I. et al. Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat. Commun. 4, 2823 (2013).

    Article  PubMed  CAS  Google Scholar 

  37. Oguro, H., Ding, L. & Morrison, S. J. SLAM family markers resolve functionally distinct subpopulations of hematopoietic stem cells and multipotent progenitors. Cell Stem Cell 13, 102–116 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Greenbaum, A. et al. CXCL12 in early mesenchymal progenitors is required for haematopoietic stem-cell maintenance. Nature 495, 227–230 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. De Minicis, S. et al. Gene expression profiles during hepatic stellate cell activation in culture and in vivo. Gastroenterology 132, 1937–1946 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Lataillade, J. J. et al. Does primary myelofibrosis involve a defective stem cell niche? From concept to evidence. Blood 112, 3026–3035 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Yue, R., Zhou, B. O., Shimada, I. S., Zhao, Z. & Morrison, S. J. Leptin receptor promotes adipogenesis and reduces osteogenesis by regulating mesenchymal stromal cells in adult bone marrow. Cell Stem Cell 18, 782–796 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Lydon, N. B. & Druker, B. J. Lessons learned from the development of imatinib. Leuk. Res. 28, S29–S38 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Worthley, D. L. et al. Gremlin 1 identifies a skeletal stem cell with bone, cartilage, and reticular stromal potential. Cell 160, 269–284 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kramann, R. et al. Perivascular Gli1+ progenitors are key contributors to injury-induced organ fibrosis. Cell Stem Cell 16, 51–66 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Inra, C. N. et al. A perisinusoidal niche for extramedullary haematopoiesis in the spleen. Nature 527, 466–471 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Tefferi, A. et al. Phase 2 trial of imatinib mesylate in myelofibrosis with myeloid metaplasia. Blood 99, 3854–3856 (2002).

    Article  CAS  PubMed  Google Scholar 

  47. Hasselbalch, H. C. et al. Imatinib mesylate in idiopathic and postpolycythemic myelofibrosis. Am. J. Hematol. 74, 238–242 (2003).

    Article  CAS  PubMed  Google Scholar 

  48. DeFalco, J. et al. Virus-assisted mapping of neural inputs to a feeding center in the hypothalamus. Science 291, 2608–2613 (2001).

    Article  CAS  PubMed  Google Scholar 

  49. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  50. Tallquist, M. D. & Soriano, P. Cell autonomous requirement for PDGFRα in populations of cranial and cardiac neural crest cells. Development 130, 507–518 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. Sharov, A. A., Dudekula, D. B. & Ko, M. S. A web-based tool for principal component and significance analysis of microarray data. Bioinformatics 21, 2548–2549 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. Huang da, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  CAS  PubMed  Google Scholar 

  53. Huang da, W., Sherman, B. T. & Lempicki, R. A. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37, 1–13 (2009).

    Article  PubMed  CAS  Google Scholar 

  54. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the MPN Research Foundation. L.D. and J.L. were supported by the Rita Allen Foundation and the National Heart, Lung and Blood Institute (1R01HL132074). Flow cytometry was partly supported by the NIH (S10RR027050 and S10OD020056). We thank R. Schwabe at Columbia and D. Brenner at UC San Diego for providing Col-gfp mice. We thank L. Olson at Oklahoma Medical Research Foundation and P. Soriano at Icahn School of Medicine at Mount Sinai for providing PdgfraD842V mice. We thank S. Weyn-Vanhentenryck, C. Zhang and R. Schwabe at Columbia for help in analysing gene expression data. We thank S. Ho and A. Figueroa for help with flow cytometry.

Author information

Authors and Affiliations

Authors

Contributions

G.W., M.D., L.M.-M., Y.L. and L.D. carried out all experiments with the help of Q.L. and J.L. M.D., L.M.-M., G.W. and L.D. designed the experiments, interpreted the results and wrote the manuscript.

Corresponding author

Correspondence to Lei Ding.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 2 HSCs progressively mobilize to the spleen as PMF advances. (Related to Fig. 2).

(a) Bone marrow multipotent progenitors (n = 17 mice for control, n = 5 mice for intermediate TOE, n = 10 mice for advanced TOE) and LSK hematopoietic progenitors (n = 12 mice for control, n = 4 mice for intermediate TOE, n = 6 mice for advanced TOE) increased in intermediate then decreased in advanced stage of PMF. (b) Representative flow cytometry plots for quantifying BrdU incorporation in HSCs after a 5-day BrdU pulse. (c) Spleen HSC frequency gradually increased from intermediate to advanced PMF mice (n = 22 mice for control, n = 15 mice for intermediate TOE, n = 10 mice for advanced TOE). (d) Spleen cellularity gradually increased from intermediate to advanced PMF mice (n = 15 mice for control, n = 6 mice for intermediate TOE, n = 7 mice for advanced TOE). (e,f) Spleen HSC number (n = 13 mice for control, n = 6 mice for intermediate TOE, n = 7 mice for advanced TOE), MPP number (n = 9 mice for control, n = 4 mice for intermediate TOE, n = 7 mice for advanced TOE) and HPC (LSK) number (n = 5 mice for control, n = 3 mice for intermediate TOE, n = 3 mice for advanced TOE) gradually increased from intermediate to advanced PMF mice. (g) An increased total HSC number (bone marrow plus spleen) in intermediate PMF mice was followed by a reduction in advanced PMF mice (n = 12 mice for control, n = 6 mice for intermediate TOE, n = 7 mice for advanced TOE). Data represent mean ± s.e.m. Two-tailed student’s t-tests were used to assess statistical significance: P < 0.05, P < 0.001.

Supplementary Figure 3 Bone marrow mesenchymal stromal lineage cells expand and assume a fibrotic cell fate. (Related to Fig. 3).

(a) Bone marrow CD45/Ter119-CD140a+ mesenchymal stromal cell frequency increased as PMF developed from intermediate to advanced stages (n = 17 mice for control, n = 12 mice for intermediate TOE, n = 8 mice for advanced TOE). (b) Bone marrow mesenchymal stromal lineage cells expanded 6.5 fold as quantified by counting TdTomato + cells on bone marrow sections from vector control and TOE mice (n = 4 images for control and TOE each, data represent mean ± s.e.m. Two-tailed student’s t-tests were used to assess statistical significance: P < 0.001). (c,d) Lepr-cre-expressing mesenchymal stromal cells expanded extensively and displayed elongated fibroblast-like stromal cell morphology. Arrow heads point to elongated fibroblast-like cells. (e) In Lepr-cre; loxptdTomato; Col-gfp control or TOE mice, all tdTomato+ cells are GFP+. (f) In Lepr-cre; loxptdTomato; Col-gfp control or TOE mice, nearly all GFP+ cells are tdTomato+. Images are representative of at least 3 biological replicates.

Supplementary Figure 4 Bone marrow mesenchymal stromal cells down-regulate CXCL12 and SCF, and display a fibrotic cell morphology. (Related to Fig. 4).

(a,b) Representative images showing normal Cxcl12-DsRed expression in Cxcl12DsRed/+ mice transplanted with control virus-infected bone marrow cells. (c,d) Images showing Cxcl12-DsRed expression in TOE mice. Arrow heads point to elongated fibroblast-like stromal cells. (e) qPCR analysis showing downregulation of Scf and Cxcl12 in bone marrow mesenchymal stromal cells from intermediate stage to advanced stage PMF mice (n = 4 mice for control, n = 4 mice for intermediate PMF and n = 4 mice for advanced PMF). Data represent mean ± s.d. Two-tailed student’s t-tests were used to assess statistical significance. (f) Representative flow cytometry plots showing CD45/Ter119Scf-GFP+ stromal cells. Images are representative of at least 3 biological replicates.

Supplementary Figure 5 Gene expression profiling analysis of mesenchymal stromal cells from PMF mice. (Related to Fig. 5).

(a) Representative flow cytometric plots showing the gates to sort CD45/Ter119/CD31Cxcl12-DsRed+ stromal cells from PMF and control bone marrow. A total of three freshly double-sorted aliquots of cells ( 5,000) from PMF (from 5 mice) and control (from 3 mice) Cxcl12DsRed/+ mice were used for gene expression analysis. (b) Normalized expression levels of mesenchymal cell markers and HSC niche factors by CD45/Ter119/CD31Cxcl12-DsRed+ stromal cells from PMF and control bone marrow. Values represent mean ± s.d. from three biological replicates. (c) List of fibrosis genes used to performed GSEA in Fig. 5c. (d) List of osteogenic genes used to performed GSEA in Fig. 5d.

Supplementary Figure 6 Lepr-cre; Pdgfrafl/− mice have normal HSC function and Lepr-cre; Pdgfrafl/fl TOE mice fail to develop bone marrow fibrosis (Related to Fig. 6).

(a) Flow cytometry plots showing efficient deletion of PDGFRa. (b) A competitive reconstitution assay for Lepr-cre; Pdgfrafl/− and control mice. 5 × 105 donor bone marrow cells from Lepr-cre; Pdgfrafl/− adult mice or control Lepr-cre; Pdgfrafl+ mice were competitively transplanted with 5 × 105 recipient bone marrow cells into irradiated recipient mice. The percentages of donor-derived Mac-1+ myeloid, CD3+ T, and B220+ B cells in the blood were analyzed for 16 weeks after transplantation (n = 5 recipient mice for each genotype). Data represent mean ± s.d. Two-tailed student’s t-tests were used to assess statistical significance. (c) Lepr-cre; Pdgfrafl/fl TOE mice displayed enlarged spleens and imatinib-treated TOE mice showed normal sized spleens. (d) Representative reticulin staining on spleen sections from Lepr-cre; Pdgfrafl/fl TOE mice revealed excessive deposition of reticulin fibers, similar to control TOE mice. (e) Confocal images showing similar levels of megakaryocyte hyperplasia in the bone marrow from Lepr-cre; Pdgfrafl/fl and control TOE mice. CD41 is a marker for megakaryocytes (red). Nuclei were stained with DAPI (blue). (f) Bone marrow sections from Lepr-cre; Pdgfrafl/fl and control TOE mice were subjected to reticulin staining. While control TOE mice robustly developed bone marrow fibrosis, none of the Lepr-cre; Pdgfrafl/fl TOE mice had bone marrow fibrosis. (g) Representative flow cytometry plots showing effective suppression of bone marrow stromal cell expansion in Lepr-cre; Pdgfrafl/fl TOE and imatinib-treated TOE mice. (h) Spleen sections from control and TOE + imatinib were subjected to reticulin staining. Images are representative of at least 3 biological replicates.

Supplementary Figure 7 Lepr-cre; PdgfraD842V/+ mice have bone marrow stromal fibrotic conversion and HSC mobilization (Related to Fig. 7).

(a) Flow cytometry analysis revealed normal bone marrow stromal cell frequency from Lepr-cre; PdgfraD842V/+ mice (n = 3 mice for control, n = 4 mice for D842V KI). (b) Representative reticulin staining on bone marrow sections from Lepr-cre; PdgfraD842V/+ and control mice. (c) Normal bone marrow cellularity (n = 4 mice for control, n = 5 mice for D842V KI) and HPC frequency of Lepr-cre; PdgfraD842V/+ mice (n = 5 mice for control, n = 6 mice for D842V KI). (d) HSC frequency in livers from Lepr-cre; PdgfraD842V/+ mice (n = 5 mice for control, n = 6 mice for D842V KI). Data represent mean ± s.d. Two-tailed student’s t-tests were used to assess statistical significance. Images are representative of at least 3 biological replicates.

Supplementary Table 1 A list of genes that were significantly up-regulated in mesenchymal stromal cells from PMF mice compared with controls. P values were determined by ANOVA test via an online NIA Array Analysis software (www.lgsun.grc.nia.nih.gov/ANOVA). TOE, Tpo-overexpressing. Con, control vector virus.

Supplementary information

Supplementary information

Supplementary information (PDF 9451 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Decker, M., Martinez-Morentin, L., Wang, G. et al. Leptin-receptor-expressing bone marrow stromal cells are myofibroblasts in primary myelofibrosis. Nat Cell Biol 19, 677–688 (2017). https://doi.org/10.1038/ncb3530

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3530

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