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:

A dynamic niche provides Kit ligand in a stage-specific manner to the earliest thymocyte progenitors

Abstract

Thymic T cell development is initiated from bone-marrow-derived multi potent thymus-seeding progenitors. During the early stages of thymocyte differentiation, progenitors become T cell restricted. However, the cellular environments supporting these critical initial stages of T cell development within the thymic cortex are not known. Here we use the dependence of early, c-Kit-expressing thymic progenitors on Kit ligand (KitL) to show that CD4CD8c-Kit+CD25 DN1-stage progenitors associate with, and depend on, the membrane-bound form of KitL (mKitL) provided by a cortex-specific KitL-expressing vascular endothelial cell (VEC) population. In contrast, the subsequent CD4CD8c-Kit+CD25+ DN2-stage progenitors associate selectively with cortical thymic epithelial cells (cTECs) and depend on cTEC-presented mKitL. These results show that the dynamic process of early thymic progenitor differentiation is paralleled by migration-dependent change to the supporting niche, and identify VECs as a thymic niche cell, with mKitL as a critical ligand.

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: Distinct patterns of Kit ligand (KitL) expression in thymic stromal cell types.
Figure 2: Localization of KitL-expressing thymic stromal subsets to the cortico-medullary junction and cortex.
Figure 3: Thymic stromal cell subsets show distinct ligand expression profiles.
Figure 4: c-Kit+ thymocyte progenitors home to mKitL-expressing cortical VECs and TECs.
Figure 5: Cortical vascular expression of mKitL is critical for thymocyte progenitor homeostasis.
Figure 6: Effect of thymic epithelial mKitL deletion on thymocyte number and thymic progenitors.
Figure 7: mKitL promotes early thymocyte progenitor survival.
Figure 8: Model of the cellular niches occupied by c-Kit+ thymocyte progenitors.

Similar content being viewed by others

References

  1. Scadden, D. T. Nice neighborhood: emerging concepts of the stem cell niche. Cell 157, 41–50 (2014).

    Article  CAS  Google Scholar 

  2. Scadden, D. T. The stem-cell niche as an entity of action. Nature 441, 1075–1079 (2006).

    Article  CAS  Google Scholar 

  3. Chasis, J. A. & Mohandas, N. Erythroblastic islands: niches for erythropoiesis. Blood 112, 470–478 (2008).

    Article  CAS  Google Scholar 

  4. Zhu, J. et al. Osteoblasts support B-lymphocyte commitment and differentiation from hematopoietic stem cells. Blood 109, 3706–3712 (2007).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  6. Bunting, M. D., Comerford, I. & McColl, S. R. Finding their niche: chemokines directing cell migration in the thymus. Immunol. Cell Biol. 89, 185–196 (2011).

    Article  CAS  Google Scholar 

  7. Love, P. E. & Bhandoola, A. Signal integration and crosstalk during thymocyte migration and emigration. Nat. Rev. Immunol. 11, 469–477 (2011).

    Article  CAS  Google Scholar 

  8. Radtke, F. et al. Deficient T cell fate specification in mice with an induced inactivation of Notch1. Immunity 10, 547–558 (1999).

    Article  CAS  Google Scholar 

  9. Koch, U. et al. Delta-like 4 is the essential, nonredundant ligand for Notch1 during thymic T cell lineage commitment. J. Exp. Med. 205, 2515–2523 (2008).

    Article  CAS  Google Scholar 

  10. Von Freeden-Jeffry, U., Solvason, N., Howard, M. & Murray, R. The earliest T lineage-committed cells depend on IL-7 for Bcl-2 expression and normal cell cycle progression. Immunity 7, 147–154 (1997).

    Article  CAS  Google Scholar 

  11. Sitnicka, E. et al. Critical role of FLT3 ligand in IL-7 receptor independent T lymphopoiesis and regulation of lymphoid-primed multipotent progenitors. Blood 110, 2955–2964 (2007).

    Article  CAS  Google Scholar 

  12. Kwan, J. & Killeen, N. CCR7 directs the migration of thymocytes into the thymic medulla. J. Immunol. 172, 3999–4007 (2004).

    Article  CAS  Google Scholar 

  13. Trampont, P. C. et al. CXCR4 acts as a costimulator during thymic β-selection. Nat. Immunol. 11, 162–170 (2010).

    Article  CAS  Google Scholar 

  14. Shah, D. K. & Zuniga-Pflucker, J. C. An overview of the intrathymic intricacies of T cell development. J. Immunol. 192, 4017–4023 (2014).

    Article  CAS  Google Scholar 

  15. Tsai, P. T., Lee, R. A. & Wu, H. BMP4 acts upstream of FGF in modulating thymic stroma and regulating thymopoiesis. Blood 102, 3947–3953 (2003).

    Article  CAS  Google Scholar 

  16. Heinonen, K. M. et al. Wnt4 regulates thymic cellularity through the expansion of thymic epithelial cells and early thymic progenitors. Blood 118, 5163–5173 (2011).

    Article  CAS  Google Scholar 

  17. Hager-Theodorides, A. L. et al. Bone morphogenetic protein 2/4 signaling regulates early thymocyte differentiation. J. Immunol. 169, 5496–5504 (2002).

    Article  CAS  Google Scholar 

  18. Takahama, Y. Journey through the thymus: stromal guides for T-cell development and selection. Nat. Rev. Immunol. 6, 127–135 (2006).

    Article  CAS  Google Scholar 

  19. Rodewald, H. R., Kretzschmar, K., Swat, W. & Takeda, S. Intrathymically expressed c-kit ligand (stem cell factor) is a major factor driving expansion of very immature thymocytes in vivo. Immunity 3, 313–319 (1995).

    Article  CAS  Google Scholar 

  20. Lennartsson, J. & Ronnstrand, L. Stem cell factor receptor/c-Kit: from basic science to clinical implications. Phys. Rev. 92, 1619–1649 (2012).

    CAS  Google Scholar 

  21. Gray, D. H., Chidgey, A. P. & Boyd, R. L. Analysis of thymic stromal cell populations using flow cytometry. J. Immunol. Methods 260, 15–28 (2002).

    Article  CAS  Google Scholar 

  22. Red-Horse, K., Ueno, H., Weissman, I. L. & Krasnow, M. A. Coronary arteries form by developmental reprogramming of venous cells. Nature 464, 549–553 (2010).

    Article  CAS  Google Scholar 

  23. Brannan, C. I. et al. Steel-Dickie mutation encodes a c-kit ligand lacking transmembrane and cytoplasmic domains. Proc. Natl Acad. Sci. USA 88, 4671–4674 (1991).

    Article  CAS  Google Scholar 

  24. Flanagan, J. G., Chan, D. C. & Leder, P. Transmembrane form of the kit ligand growth factor is determined by alternative splicing and is missing in the Sld mutant. Cell 64, 1025–1035 (1991).

    Article  CAS  Google Scholar 

  25. Gu, Y., Runyan, C., Shoemaker, A., Surani, M. A. & Wylie, C. Membrane-bound Steel factor maintains a high local concentration for mouse primordial germ cell motility, and defines the region of their migration. PLoS ONE 6, e25984 (2011).

    Article  CAS  Google Scholar 

  26. Mikkelsen, H. B., Malysz, J., Huizinga, J. D. & Thuneberg, L. Action potential generation, Kit receptor immunohistochemistry and morphology of Steel-Dickie (Sl/Sld) mutant mouse small intestine. Neurogastroenterol. Motil. 10, 11–26 (1998).

    Article  CAS  Google Scholar 

  27. Kapur, R. et al. Signaling through the interaction of membrane-restricted stem cell factor and c-kit receptor tyrosine kinase: genetic evidence for a differential role in erythropoiesis. Blood 91, 879–889 (1998).

    CAS  PubMed  Google Scholar 

  28. Russell, E. S. Hereditary anemias of the mouse: a review for geneticists. Adv. Genet. 20, 357–459 (1979).

    Article  CAS  Google Scholar 

  29. Kisanuki, Y. Y. et al. Tie2-Cre transgenic mice: a new model for endothelial cell-lineage analysis in vivo. Dev. Biol. 230, 230–242 (2001).

    Article  CAS  Google Scholar 

  30. Claxton, S. et al. Efficient, inducible Cre-recombinase activation in vascular endothelium. Genesis 46, 74–80 (2008).

    Article  CAS  Google Scholar 

  31. Srinivas, S. et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev. Biol. 1, 4 (2001).

    Article  CAS  Google Scholar 

  32. Gordon, J. et al. Specific expression of lacZ and cre recombinase in fetal thymic epithelial cells by multiplex gene targeting at the Foxn1 locus. BMC Dev. Biol. 7, 69 (2007).

    Article  Google Scholar 

  33. Calderon, L. & Boehm, T. Synergistic, context-dependent, and hierarchical functions of epithelial components in thymic microenvironments. Cell 149, 159–172 (2012).

    Article  CAS  Google Scholar 

  34. Krause, D. S. & Scadden, D. T. Deconstructing the complexity of a microenvironmental niche. Cell 149, 16–17 (2012).

    Article  CAS  Google Scholar 

  35. Ramasamy, S. K., Kusumbe, A. P. & Adams, R. H. Regulation of tissue morphogenesis by endothelial cell-derived signals. Trends Cell Biol. 25, 148–157 (2014).

    Article  Google Scholar 

  36. Sasaki, T. et al. Regulation of hematopoietic cell clusters in the placental niche through SCF/Kit signaling in embryonic mouse. Development 137, 3941–3952 (2010).

    Article  CAS  Google Scholar 

  37. 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  Google Scholar 

  38. Badea, T. C., Wang, Y. & Nathans, J. A noninvasive genetic/pharmacologic strategy for visualizing cell morphology and clonal relationships in the mouse. J. Neurosci. 23, 2314–2322 (2003).

    Article  CAS  Google Scholar 

  39. Moreira, P. N., Pozueta, J., Giraldo, P., Gutierrez-Adan, A. & Montoliu, L. Generation of yeast artificial chromosome transgenic mice by intracytoplasmic sperm injection. Methods Mol. Biol. 349, 151–161 (2006).

    PubMed  Google Scholar 

  40. Farley, F. W., Soriano, P., Steffen, L. S. & Dymecki, S. M. Widespread recombinase expression using FLPeR (flipper) mice. Genesis 28, 106–110 (2000).

    Article  CAS  Google Scholar 

  41. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  Google Scholar 

  42. Ramskold, D. et al. Full-length mRNA-Seq from single-cell levels of RNA and individual circulating tumor cells. Nat. Biotechnol. 30, 777–782 (2012).

    Article  Google Scholar 

  43. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  Google Scholar 

  44. Grover, A. et al. Erythropoietin guides multipotent hematopoietic progenitor cells toward an erythroid fate. J. Exp. Med. 211, 181–188 (2014).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank C. Blackburn (Institute for Stem Cell Research, UK), J. Nathans (Johns Hopkins Medical School, Maryland, USA) and M. Fruttiger (National Institute of Medical Research, UK) for providing Foxn1–Cre, Rosa26–CreERT2 and Pdgfb–CreERT2 mice, respectively, A. Samraj for screening of targeted embryonic stem cells, and the Wolfson Imaging Centre for image analysis. This work was supported by an MRC Strategic Award (G0701761) and MRC Program Grant (G0900892) to C.N., by an MRC Program Grant to S.E.W.J., by Cancer Research UK and by Leukemia and Lymphoma Research. A.J.M. is the recipient of an MRC Senior Clinical fellowship.

Author information

Authors and Affiliations

Authors

Contributions

M.B. carried out mouse genetic experiments and imaging, and associated data analysis; R.F. and S.M. generated Sl/Sld andΔEx7 mice and measured KitL levels and activity; S.T. analysed RNASeq data; T.C.L. provided protocols for single-cell profiling; A.G. generated RNAseq libraries; D.W. performed image analysis; P.B. provided ICKitL-specific antibody; A.J.M. provided protocols for single-cell profiling; S.E.W.J. and C.N. conceived the project and wrote the manuscript with M.B.

Corresponding authors

Correspondence to Sten Eirik W. Jacobsen or Claus Nerlov.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 5 Heat maps of stromal cell gene expression.

(a) Heat map of gene expression data from Fig. 3a–g. RPKM values were normalized for each individual gene across all cell types. (b) mRNA expression in single KT+ VECs, KT VECs (h) and cTECs and mTECs (i) using microfluidics-based qPCR. Expression values are normalized to B2m for each gene, and subsequently to the average value for each individual gene across all cell types. Each column represents a single cell.

Supplementary Figure 6 Lack of detectable sKitL expression from Steel-Dickie allele.

(a) Serum KItL levels in 3-week-old wild-type (+/+; n = 18), Sl/+ (n = 7), Sld/+ (n = 7) and Sl/Sld (n = 4) mice measured by ELISA. Error bars show standard errors. Student’s t-test P values are shown. For source data see Supplementary Table 5. (b) Schematic of the exon 7 floxed Kitl allele. (c) Concentration of sKItL in supernatants of HEK293 cells transfected with the empty MSCV-IRES-GFP vector (N = 6) or MSCV-IRES-GFP vectors encoding Sld KitL (N = 3) and ΔEx7 KitL (N = 3), respectively. Error bars show standard errors. For source data see Supplementary Table 5. (d) Bioassay measuring proliferation of the KitL-dependent MC/9 cell line exposed to the supernatants from (c). Values are relative to MC/9 cells grown under optimal proliferation conditions. Error bars show standard errors. N = 3 for each condition. For source data see Supplementary Table 5. (e) Serum KItL levels in 3 week old control (+/+; n = 6) and KitlΔEx7/ΔEx7 (n = 4) mice measured by ELISA. Error bars show standard errors. Student’s t-test P value is shown. For source data see Supplementary Table 5. (f) Total thymic cellularity of 4–5 weeks old +/+ mice (n = 3) and their Sl/+ littermates (n = 3). Error bars show standard errors. The difference between +/+ and Sl/+ genotypes is not statistically significant (P = 0.71). For source data see Supplementary Table 5. (g) Absolute number of phenotypically defined Flt3 + DN1, Flt3–DN1 and DN2 cells in thymi of mice from (f). Error bars show standard errors. No statistical significant differences were observed (P = 0.21, P = 0.48 and P = 0.35, respectively). For source data see Supplementary Table 5. (h) Total bone marrow cells in mice from (f). Error bars show standard errors. The difference between +/+ and Sl/+ is not statistically significant (P = 0.206). For source data see Supplementary Table 5. (i) Number of cells in the indicated phenotypically defined lymphoid progenitor populations in mice from (f). Values are group averages; error bars show standard errors. P-values relative to the control group are shown. For source data see Supplementary Table 5.

Supplementary Figure 7 Specificity of Cre lines used.

(a) Schematic of the Tie2-Cre lineage tracing using the Rosa26-EYFP Cre reporter. (b) Flow cytometric analysis of thymic stromal cell subsets from Tie2-Cre; Rosa26-EYFP mice (green lines) and Rosa26-EYFP control mice (blue lines). Percentage of YFP + cells observed in the VEC (right plot) and combined MCs and TECs populations (left plot) is shown. (c) Schematic of the Pdgfb − CreERT2-IRES-iGFP reporter. (d) Flow cytometric analysis of thymic stromal cell subsets from PdgfbCreERT2-IRES-iGFP mice (green lines) and non-transgenic control mice (blue lines). Percentage of GFP + cells detected in the VECs (right plot) and the combined MC and TEC populations (left plot) is shown. (e) Schematic of Foxn1-Cre lineage tracing using the Rosa26-EYFP Cre reporter. (f) Flow cytometry analysis of thymic stromal cell subsets in the Foxn1-Cre/Rosa26-EYFP mouse model. Percentage of EYFP + cells in the TEC (right plot) and MC plus VEC lineages (left plot) is displayed. (g) Total thymic cells from 6 weeks old controls (n = 3 biological replicates pooled over 2 independent experiments) and Pdgfb-CreERT2 transgenic (n = 3) littermates, treated with tamoxifen and analysed one week after the last injection. The Cre driver is shown below the x-axis. Values are group averages. Error bars show standard errors. No statistical significant differences were observed. For source data see Supplementary Table 5. (h) Absolute number of phenotypic Flt3 + DN1, Flt3- DN1 and DN2 thymocytes from the mice in (g). Error bars show standard errors. No statistical significant differences were observed. For source data see Supplementary Table 5. (i) Number of total bone marrow cells from the mice analysed in (g). Error bars show standard errors. No statistical significant differences were observed. For source data see Supplementary Table 5. (j) Absolute number of phenotypic bone marrow lymphoid progenitor populations from the mice in (g). Error bars show standard errors. No statistically significant differences were observed. For source data see Supplementary Table 5. (k) Absolute number of phenotypic DAPIlineageB220CD4CD8 (DN), DAPICD4 + CD8 + (DP), DAPICD4+CD8 (SP4), DAPICD4CD8+ (SP8) from 4–5 weeks old uninjected (n = 6) and tamoxifen injected (n = 6) control mice pooled from 3 independent experiments. Error bars show standard errors. No statistically significant differences were observed.

Supplementary Figure 8 Loss of mKitL does not affect thymic architecture or morphology.

Immunofluorescence analysis of thymic sections by using antibodies against CD31 (green), Ly51 (red), UEA-1 (blue) to assess the overall thymus architecture in 4–5 weeks old control (ad) (KitlLEx7/LEx7) and FoxN1ΔEx7 mice (eh). Scale bar: 300 μm. Immunofluorescence analysis of thymic sections by using antibodies against CD31 (green), Ly51 (red), UEA-1 (blue) in 4–5 weeks old, tamoxifen injected control (il) (KitlLEx7/LEx7), Tie2/PdgfbΔEx7 mice (mp) and Tie2/Pdgfb/FoxN1ΔEx7 mice. Scale bar: 300 μm.

Supplementary Figure 9 Impact of mKitL depletion on thymic stromal cell and thymic progenitor populations.

(a) Relative percentage of phenotypic TECs, VECs and MCs from 4–5 weeks old control, Foxn1ΔEx7 and Tie2/PdgfbΔEx7 mice analyzed as in Fig. 5a. Number of biological replicates displayed. Error bars show standard errors. No statistically significant differences were observed. For source data see Supplementary Table 5. (b) Absolute number of DN, DP, SP4 and SP8 from 4–5 week old control (n = 7) and Foxn1ΔEx7 mice (n = 3). Error bars show standard errors. P-values relative to the control group are shown. For source data see Supplementary Table 5. (c) Absolute number of phenotypic DN, DP, SP4 and SP8 from 4–5 weeks old control (n = 5), Tie2/PdgfbΔEx7 (n = 4) and Tie2/Pdgfb/FoxN1ΔEx7 (n = 5), analyzed as in Fig. 5a. Error bars show standard errors. P-values relative to the control group are shown. For source data see Supplementary Table 5.

Supplementary Figure 10 Effect of stromal mKitL deletion on bone marrow lymphopoiesis.

(a) Number of total bone marrow cells from KitlLEx7/LEx7 (n = 4) and Tie2/PdgfbΔEx7 (n = 4) mice from 2 separate experiments, treated with tamoxifen and analysed as in Fig. 5a. Error bars show standard errors. No statistically significant differences were observed. (b) Number of phenotypic bone marrow lymphoid progenitor populations in mice from (a). Error bars show standard errors. No statistically significant differences were observed. (c) Number of total bone marrow cells from KitlLEx7/LEx7 (n = 7) and Foxn1ΔEx7 (n = 7) mice from 3 separate experiments. Error bars show standard errors. P-values relative to the control group are shown (Student’s t-test). (d) Number of phenotypic bone marrow lymphoid progenitor populations in mice from (c). Error bars show standard errors. No statistically significant differences were observed. (e) Number of total bone marrow cells from KitlLEx7/LEx7 (n = 6) and Tie2/Pdgfb/Foxn1ΔEx7 (n = 5) mice analyzed as in (a) from 4 separate experiments. Error bars show standard errors. No statistically significant differences were observed. (f) Number of phenotypic bone marrow lymphoid progenitor populations in mice from (e). Error bars show standard errors. No statistically significant differences were observed.

Supplementary Figure 11 Thymic stromal cells FACS analysis.

Gating strategy used to define thymic stromal cell populations within the total thymic live (DAPI −) singlet cell population: VECs: LinEpCAMCD31+; MCs LinEpCAMCD31,; cTECs: LinEpCAM+Ly51+UEA-1; mTECs: LinCD31EpCAM+Ly51UEA-1+. Lin is CD45Ter119.

Supplementary Figure 12 Thymic T cell progenitors and bone marrow lymphoid progenitors FACS analysis.

(a) Gating strategy used to define Flt3 + DN1 (Lin − CD4 − CD8α − CD25 − c-KithiFlt3 +), Flt3–DN1 (Lin − CD4 − CD8α − CD25 − c-KithiFlt3 −) and DN2 (Lin − CD4 − CD8α − CD25 + c-KithiFlt3 −) thymocytes in young adult mice (4–6 weeks). (b) Gating strategy used to define lymphoid progenitor populations within the bone marrow live (DAPI −) singlet cell population. The following populations were analysed: NK/T progenitors (LIn–B220 − CD49b − NK1.1 − CD122 +), Lymphoid-biased multi-potent progenitors (LMPPs: Lin − B220 − CD49b − NK1.1 − CD122 − Flt3 + CD127 − c-KithiSca1hi), All Lymphoid Progenitors (ALPs: Lin − B220 − CD49b − NK1.1 − CD122 − Flt3 + CD127 + Ly6D − c-Kitlo), B-Lymphoid Progenitors (BLPs: Lin − B220 − CD49b − NK1.1 − CD122 − Flt3 + CD127 + Ly6D + c-Kit −), Common Lymphoid Progenitors (CLPs: Lin − B220 − CD49b − NK1.1 − CD122 − Flt3 + CD127 + Ly6D − c-KitloSca1lo).

Supplementary Table 1 Genotyping information.
Supplementary Table 2 Antibodies used for immunocytochemistry.
Supplementary Table 3 Antibodies used for flow cytometry.
Supplementary Table 4 TaqMan probes used for qPCR.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1517 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Buono, M., Facchini, R., Matsuoka, S. et al. A dynamic niche provides Kit ligand in a stage-specific manner to the earliest thymocyte progenitors. Nat Cell Biol 18, 157–167 (2016). https://doi.org/10.1038/ncb3299

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3299

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