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:

Notch and the pre-TCR coordinate thymocyte proliferation by induction of the SCF subunits Fbxl1 and Fbxl12

Abstract

Proliferation is tightly regulated during T cell development, and is limited to immature CD4CD8 thymocytes. The major proliferative event is initiated at the ‘β-selection’ stage following successful rearrangement of Tcrβ, and is triggered by and dependent on concurrent signaling by Notch and the pre-T cell receptor (TCR); however, it is unclear how these signals cooperate to promote cell proliferation. Here, we found that β-selection-associated proliferation required the combined activity of two Skp-cullin-F-box (SCF) ubiquitin ligase complexes that included as substrate recognition subunits the F-box proteins Fbxl1 or Fbxl12. Both SCF complexes targeted the cyclin-dependent kinase inhibitor Cdkn1b for polyubiquitination and proteasomal degradation. We found that Notch signals induced the transcription of Fbxl1, whereas pre-TCR signals induced the transcription of Fbxl12. Thus, concurrent Notch and pre-TCR signaling induced the expression of two genes, Fbxl1 and Fbxl12, whose products functioned identically but additively to promote degradation of Cdkn1b, cell cycle progression, and proliferation of β-selected thymocytes.

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: SCF-Fbxl12 complexes target Cdkn1b for polyubiquitination and proteasomal degradation.
Fig. 2: Impaired β-selection-associated proliferation in Lck-Cre Fbxl12fl/fl mice.
Fig. 3: Restoration of thymocyte development and β-selection-associated proliferation in Lck-Cre Fbx12fl/fl mice by deletion of Cdkn1b.
Fig. 4: Thymocyte development and β-selection-associated proliferation are strongly impaired in thymocytes lacking Fbxl1 and Fbxl12.
Fig. 5: Fbxl1 and Fbxl12 target the same site on Cdkn1b for K48 polyubiquitination.
Fig. 6: Notch signaling and pre-TCR signaling regulate Fbxl1 and Fbxl12 expression, respectively.
Fig. 7: Fbxl1 and Fbxl12 function interchangeably to promote proliferation, but are not sufficient for β selection.
Fig. 8: Proliferation of immature γδTCR+ thymocytes is mediated primarily by TCR-induced regulation of Fbxl12.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon request.

References

  1. Rodewald, H. R., Ogawa, M., Haller, C., Waskow, C. & DiSanto, J. P. Pro-thymocyte expansion by c-kit and the common cytokine receptor γ chain is essential for repertoire formation. Immunity 6, 265–272 (1997).

    Article  CAS  PubMed  Google Scholar 

  2. Shortman, K., Egerton, M., Spangrude, G. J. & Scollay, R. The generation and fate of thymocytes. Semin. Immunol. 2, 3–12 (1990).

    CAS  PubMed  Google Scholar 

  3. Wong, G. W., Knowles, G. C., Mak, T. W., Ferrando, A. A. & Zuniga-Pflucker, J. C. HES1 opposes a PTEN-dependent check on survival, differentiation, and proliferation of TCRβ-selected mouse thymocytes. Blood 120, 1439–1448 (2012).

    Article  CAS  PubMed  Google Scholar 

  4. Penit, C., Lucas, B. & Vasseur, F. Cell expansion and growth arrest phases during the transition from precursor (CD48) to immature (CD4+8+) thymocytes in normal and genetically modified mice. J. Immunol. 154, 5103–5113 (1995).

    CAS  PubMed  Google Scholar 

  5. Kreslavsky, T. et al. β-Selection-induced proliferation is required for αβ T cell differentiation. Immunity 37, 840–853 (2012).

    Article  CAS  PubMed  Google Scholar 

  6. Ciofani, M. et al. Obligatory role for cooperative signaling by pre-TCR and Notch during thymocyte differentiation. J. Immunol. 172, 5230–5239 (2004).

    Article  CAS  PubMed  Google Scholar 

  7. Hoffman, E. S. et al. Productive T-cell receptor β-chain gene rearrangement: coincident regulation of cell cycle and clonality during development in vivo. Genes Dev. 10, 948–962 (1996).

    Article  CAS  PubMed  Google Scholar 

  8. Maillard, I. et al. The requirement for Notch signaling at the β-selection checkpoint in vivo is absolute and independent of the pre-T cell receptor. J. Exp. Med. 203, 2239–2245 (2006).

    Article  CAS  PubMed  Google Scholar 

  9. Yashiro-Ohtani, Y., Ohtani, T. & Pear, W. S. Notch regulation of early thymocyte development. Semin. Immunol. 22, 261–269 (2010).

    Article  CAS  PubMed  Google Scholar 

  10. Aifantis, I., Mandal, M., Sawai, K., Ferrando, A. & Vilimas, T. Regulation of T-cell progenitor survival and cell-cycle entry by the pre-T-cell receptor. Immunol. Rev. 209, 159–169 (2006).

    Article  CAS  Google Scholar 

  11. Rowell, E. A. & Wells, A. D. The role of cyclin-dependent kinases in T-cell development, proliferation, and function. Crit. Rev. Immunol. 26, 189–212 (2006).

    Article  CAS  Google Scholar 

  12. Deng, C., Zhang, P., Harper, J. W., Elledge, S. J. & Leder, P. Mice lacking p21CIP1/WAF1 undergo normal development, but are defective in G1 checkpoint control. Cell 82, 675–684 (1995).

    Article  CAS  Google Scholar 

  13. Matsumoto, A., Takeishi, S. & Nakayama, K. I. p57 regulates T-cell development and prevents lymphomagenesis by balancing p53 activity and pre-TCR signaling. Blood 123, 3429–3439 (2014).

    Article  CAS  Google Scholar 

  14. Nakayama, K. et al. Mice lackingp27Kip1 display increased body size, multiple organ hyperplasia, retinal dysplasia, and pituitary tumors. Cell 85, 707–720 (1996).

    Article  CAS  Google Scholar 

  15. Kiyokawa, H. et al. Enhanced growth of mice lacking the cyclin-dependent kinase inhibitor function of p27Kip1. Cell 85, 721–732 (1996).

    Article  CAS  Google Scholar 

  16. Fero, M. L. et al. A syndrome of multiorgan hyperplasia with features of gigantism, tumorigenesis, and female sterility in p27Kip1-deficient mice. Cell 85, 733–744 (1996).

    Article  CAS  Google Scholar 

  17. Tsukiyama, T. et al. Down-regulation of p27Kip1 expression is required for development and function of T cells. J. Immunol. 166, 304–312 (2001).

    Article  CAS  Google Scholar 

  18. Carrano, A. C., Eytan, E., Hershko, A. & Pagano, M. SKP2 is required for ubiquitin-mediated degradation of the CDK inhibitor p27. Nat. Cell Biol. 1, 193–199 (1999).

    Article  CAS  Google Scholar 

  19. Nakayama, K. et al. Skp2-mediated degradation of p27 regulates progression into mitosis. Dev. Cell 6, 661–672 (2004).

    Article  CAS  Google Scholar 

  20. Kossatz, U. et al. Skp2-dependent degradation of p27Kip1 is essential for cell cycle progression. Genes Dev. 18, 2602–2607 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. Sarmento, L. M. et al. Notch1 modulates timing of G1–S progression by inducing SKP2 transcription and p27Kip1 degradation. J. Exp. Med. 202, 157–168 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Dohda, T. et al. Notch signaling induces SKP2 expression and promotes reduction of p27Kip1 in T-cell acute lymphoblastic leukemia cell lines. Exp. Cell Res. 313, 3141–3152 (2007).

    Article  CAS  Google Scholar 

  23. Del Debbio, C. B. et al. Notch signaling activates stem cell properties of Müller glia through transcriptional regulation and Skp2-mediated degradation of p27Kip1. PLoS ONE 11, e0152025 (2016).

    Article  PubMed  Google Scholar 

  24. Hristova, N. R., Tagscherer, K. E., Fassl, A., Kopitz, J. & Roth, W. Notch1-dependent regulation of p27 determines cell fate in colorectal cancer. Int. J. Oncol. 43, 1967–1975 (2013).

    Article  CAS  Google Scholar 

  25. Tsvetkov, L. M., Yeh, K. H., Lee, S. J., Sun, H. & Zhang, H. p27Kip1 ubiquitination and degradation is regulated by the SCFSkp2 complex through phosphorylated Thr187 in p27. Curr. Biol. 9, 661–664 (1999).

    Article  CAS  Google Scholar 

  26. Jin, J. et al. Systematic analysis and nomenclature of mammalian F-box proteins. Genes Dev. 18, 2573–2580 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Nishiyama, M., Nita, A., Yumimoto, K. & Nakayama, K. I. FBXL12-mediated degradation of ALDH3 is essential for trophoblast differentiation during placental development. Stem Cells 33, 3327–3340 (2015).

    Article  CAS  Google Scholar 

  28. Sun, Z. et al. Requirement for RORγ in thymocyte survival and lymphoid organ development. Science 288, 2369–2373 (2000).

    Article  CAS  Google Scholar 

  29. Clague, M. J. & Urbe, S. Ubiquitin: same molecule, different degradation pathways. Cell 143, 682–685 (2010).

    Article  CAS  Google Scholar 

  30. Oshikawa, K., Matsumoto, M., Oyamada, K. & Nakayama, K. I. Proteome-wide identification of ubiquitylation sites by conjugation of engineered lysine-less ubiquitin. J. Proteome Res. 11, 796–807 (2012).

    Article  CAS  PubMed  Google Scholar 

  31. Tussiwand, R. et al. The preTCR-dependent DN3 to DP transition requires Notch signaling, is improved by CXCL12 signaling and is inhibited by IL-7 signaling. Eur. J. Immunol. 41, 3371–3380 (2011).

    Article  CAS  PubMed  Google Scholar 

  32. Garbe, A. I., Krueger, A., Gounari, F., Zuniga-Pflucker, J. C. & von Boehmer, H. Differential synergy of Notch and T cell receptor signaling determines αβ versus γδ lineage fate. J. Exp. Med. 203, 1579–1590 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Schmitt, T. M. & Zuniga-Pflucker, J. C. Induction of T cell development from hematopoietic progenitor cells by Delta-like-1 in vitro. Immunity 17, 749–756 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Shinkai, Y. et al. RAG-2-deficient mice lack mature lymphocytes owing to inability to initiate V(D)J rearrangement. Cell 68, 855–867 (1992).

    Article  CAS  PubMed  Google Scholar 

  35. Levelt, C. N., Mombaerts, P., Iglesias, A., Tonegawa, S. & Eichmann, K. Restoration of early thymocyte differentiation in T-cell receptor beta-chain-deficient mutant mice by transmembrane signaling through CD3 epsilon. Proc. Natl Acad. Sci. USA 90, 11401–11405 (1993).

    Article  CAS  PubMed  Google Scholar 

  36. Shinkai, Y. & Alt, F. W. CD3ε-mediated signals rescue the development of CD4+CD8+ thymocytes in RAG-2−/− mice in the absence of TCR β chain expression. Int. Immunol. 6, 995–1001 (1994).

    Article  CAS  PubMed  Google Scholar 

  37. Wiest, D. L., Kearse, K. P., Shores, E. W. & Singer, A. Developmentally regulated expression of CD3 components independent of clonotypic T cell antigen receptor complexes on immature thymocytes. J. Exp. Med. 180, 1375–1382 (1994).

    Article  CAS  PubMed  Google Scholar 

  38. Taghon, T., Yui, M. A., Pant, R., Diamond, R. A. & Rothenberg, E. V. Developmental and molecular characterization of emerging β- and γδ-selected pre-T cells in the adult mouse thymus. Immunity 24, 53–64 (2006).

    Article  CAS  Google Scholar 

  39. Ciofani, M., Knowles, G. C., Wiest, D. L., von Boehmer, H. & Zuniga-Pflucker, J. C. Stage-specific and differential Notch dependency at the αβ and γδ T lineage bifurcation. Immunity 25, 105–116 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Haks, M. C. et al. Attenuation of γδTCR signaling efficiently diverts thymocytes to the αβ lineage. Immunity 22, 595–606 (2005).

    Article  CAS  Google Scholar 

  41. Hayes, S. M. & Love, P. E. Strength of signal: a fundamental mechanism for cell fate specification. Immunol. Rev. 209, 170–175 (2006).

    Article  Google Scholar 

  42. Rowell, E. A., Walsh, M. C. & Wells, A. D. Opposing roles for the cyclin-dependent kinase inhibitor p27Kip1 in the control of CD4+ T cell proliferation and effector function. J. Immunol. 174, 3359–3368 (2005).

    Article  CAS  Google Scholar 

  43. Jiang, H. et al. Ubiquitylation of RAG-2 by Skp2-SCF links destruction of the V(D)J recombinase to the cell cycle. Mol. Cell 18, 699–709 (2005).

    Article  CAS  Google Scholar 

  44. Tedesco, D., Lukas, J. & Reed, S. I. The pRb-related protein p130 is regulated by phosphorylation-dependent proteolysis via the protein-ubiquitin ligase SCFSkp2. Genes Dev. 16, 2946–2957 (2002).

    Article  CAS  PubMed  Google Scholar 

  45. Kim, S. Y., Herbst, A., Tworkowski, K. A., Salghetti, S. E. & Tansey, W. P. Skp2 regulates Myc protein stability and activity. Mol. Cell 11, 1177–1188 (2003).

    Article  CAS  Google Scholar 

  46. Li, X., Zhao, Q., Liao, R., Sun, P. & Wu, X. The SCFSkp2 ubiquitin ligase complex interacts with the human replication licensing factor Cdt1 and regulates Cdt1 degradation. J. Biol. Chem. 278, 30854–30858 (2003).

    Article  CAS  Google Scholar 

  47. Passoni, L. et al. Intrathymic δ selection events in γδ cell development. Immunity 7, 83–95 (1997).

    Article  CAS  PubMed  Google Scholar 

  48. Prinz, I. et al. Visualization of the earliest steps of γδ T cell development in the adult thymus. Nat. Immunol. 7, 995–1003 (2006).

    Article  CAS  PubMed  Google Scholar 

  49. Wilson, A., MacDonald, H. R. & Radtke, F. Notch 1-deficient common lymphoid precursors adopt a B cell fate in the thymus. J. Exp. Med. 194, 1003–1012 (2001).

    Article  CAS  PubMed  Google Scholar 

  50. Onoyama, I. et al. Conditional inactivation of Fbxw7 impairs cell-cycle exit during T cell differentiation and results in lymphomatogenesis. J. Exp. Med. 204, 2875–2888 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Nakayama, K. et al. Targeted disruption of Skp2 results in accumulation of cyclin E and p27Kip1, polyploidy and centrosome overduplication. EMBO J. 19, 2069–2081 (2000).

    Article  CAS  PubMed  Google Scholar 

  52. Chien, W. M. et al. Genetic mosaics reveal both cell-autonomous and cell-nonautonomous function of murine p27Kip1. Proc. Natl Acad. Sci. USA 103, 4122–4127 (2006).

    Article  CAS  Google Scholar 

  53. Mohtashami, M., Shah, D. K., Kianizad, K., Awong, G. & Zuniga-Pflucker, J. C. Induction of T-cell development by Delta-like 4-expressing fibroblasts. Int. Immunol. 25, 601–611 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Intramural Research Program of the Eunice Kennedy Shriver National Institute of Child Health and Human Development (project number 1ZIAHD001803-25 to P.E.L.), a grant from the Swedish Society for Medical Research (to B.Z.), the Canadian Institutes of Health Research (FND-154332 to J.C.Z.-P.) and the National Institutes of Health (1P01AI102853-01 to J.C.Z.-P.). J.C.Z.-P. is supported by a Canada Research Chair in Developmental Immunology, and K.Y. is supported by a Canada Graduate Scholarship from the Natural Sciences and Engineering Research Council of Canada. The authors thank R. Bosselut for critical reading of the manuscript, and R. Bosselut, A. Bhandoola, B. J. Fowlkes, N. Taylor and H. Petrie for helpful discussions on the project.

Author information

Authors and Affiliations

Authors

Contributions

B.Z., K.Y., L.L. and J.Y.L. performed the experiments. B.Z., P.E.L. and J.C.Z.-P. designed the experiments. P.E.L. wrote the manuscript.

Corresponding author

Correspondence to Paul E. Love.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information: Ioana Visan was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Integrated supplementary information

Supplementary Figure 1 β-selection-associated proliferation is increased in Lck-Cre Cdkn1bfl/fl mice and decreased in Fbxl1-/- mice, and deletion of Cdkn1b rescues thymocyte development in Fbxl1-/- mice.

(a) Flow cytometry analysis showing the phenotype of thymocytes (left) or splenocytes (right) from mice of the indicated genotype. Thymus: left, CD4 vs CD8 staining of total thymocytes; center, CD44 vs CD25 staining of lineage-negative DN thymocytes. Spleen: CD4 vs CD8 staining of total splenocytes; (b) Immunoblot analysis showing Cdkn1b protein expression in purified lineage-negative DN thymocytes from Lck-Cre or Lck-Cre Cdkn1bfl/fl (Cdkn1b KO) mice. (c) Cell numbers of total thymocytes or the indicated thymocyte subsets and DN3/DN4 ratio (n=4 mice per genotype). (d) Numbers of CD4 SP and CD8 SP cells in the thymus and spleen of indicated genotype (n=4 mice per genotype). For all graphs, horizontal lines indicate the mean and vertical lines indicate the standard deviation (±s.d.), P values were determined by unpaired two-tailed Student’s t-test. NS, not significant (P>0.05), *P<0.05, **P<0.01, ***P<0.005, ****P<0.0001. Data shown in (a) and (b) are representative of four or two independent experiments, respectively.

Supplementary Figure 2 β-selection-associated proliferation is increased in Lck-Cre Cdkn1bfl/fl mice and decreased in Fbxl1-/- mice and Fbxl1 is a subunit of an SCF ubiquitin ligase complex that targets Cdkn1b.

(a) Cell cycle analysis of thymocyte subsets showing the percentage of cells in S/G2/M phase. (n=4 mice per genotype). (b) Percentage of annexin V positive cells within the indicated thymocyte subsets from Fbxl1+/+ or Fbxl1-/- mice. (n=3 mice per genotype). (c) Immunoprecipitation and immunoblot analysis showing the interaction of Fbxl1 and Cul1 in HEK-293T cells transfected with plasmids encoding Myc-Fbxl1 and Flag-Cul1. (d) Immunoprecipitation and immunoblot analysis showing the interaction of Fbxl1 and Cdkn1b in HEK-293T cells transfected with plasmids encoding Myc-Fbxl1 and Flag-Cdkn1b. (e) Immunoblot analysis showing degradation of Cdkn1b by SCF-Fbxl1 complexes and its dependence on the Fbxl1 F-box motif in HEK-293T cells transfected with plasmids encoding Myc-Fbxl1 or Myc-Fbxl1ΔF (Myc epitope tagged Fbxl1 lacking the F-box motif) and Flag-Cdkn1b. (f) Immunoblot analysis showing Fbxl1 and Cdkn1b protein expression in purified DN thymocytes from Fbxl1+/+ or Fbxl1-/- mice. For all graphs, small horizontal lines indicate the mean and vertical lines indicate the standard deviation (±s.d.), P values were determined by unpaired two-tailed Student’s t-test. NS, not significant (P>0.05), *P<0.05, **P<0.01, ***P<0.005, ****P<0.0001. Data shown in (c-f) are representative of three independent experiments.

Supplementary Figure 3 Analysis of Fbxl12 expression in human and mouse tissues and generation of Fbxl12fl/fl mice.

(a) BioGPS database plot showing expression of Fbxl12 mRNA in human tissues evaluated by microarray. (http://www.biogps.org). (b) Immgen plot showing expression of Fbxl12 mRNA in mouse tissues and thymocyte subsets detected by microarray. (http://www.immgen.org). (c) Immunoblot analysis showing Fbxl12 and Fbxl1 expression in wild type (B6) thymocyte subsets. (d) Schematic showing gene targeting scheme for generation of Fbxl12 conditional knockout mice. (e) PCR analysis of genomic DNA purified from the tail snips of wild-type (+/+), heterozygous (fl/+) and homozygous(fl/fl) Fbxl12 floxed mice showing WT and Floxed Fbxl12 alleles. (f) Immunoblot analysis showing Fbxl12, Cdkn1b, and β-actin in total thymocytes from Lck-Cre, and Lck-Cre Fbxl12fl/fl mice. Data shown in (c,e,f) are representative of 3 independent experiments.

Supplementary Figure 4 Impaired β-selection-associated cell cycling and proliferation in Lck-Cre Fbxl12fl/fl mice.

(a) Cell cycle analysis of thymocyte subsets from mice of the indicated genotype showing percentage of S/G2/M in the indicated thymocyte subsets. (b) Numbers of CD4 SP and CD8 SP cells in the thymus and spleen of mice of the indicated genotype (n=6 mice per genotype). (c) Percentage of annexin V positive cells within thymocyte subsets from mice of the indicated genotype (n=3 mice per genotype). For all graphs horizontal lines indicate the mean and vertical lines indicate standard deviation (±s.d.), P values were determined by unpaired two-tailed Student’s t-test. NS, not significant (P>0.05), *P<0.05, **P<0.01, ***P<0.005, ****P<0.0001. Data shown in (a) are one representative of five experiments.

Supplementary Figure 5 Analysis of RORγt expression in Lck-Cre Fbxl12fl/fl mice and phenotype of AND TCR transgenic (tg) Lck-Cre Fbxl12fl/fl mice.

(a) Immunoblot analysis showing RORγt and β-actin loading control in thymocytes of the indicated genotype. (b) Flow cytometry analysis of thymocytes from AND TCR transgenic (tg) Fbxl12fl/fl and Lck-Cre Fbxl12fl/fl mice. Shown are: left, CD4 vs CD8 staining of total thymocytes; center, histogram of Vα11 staining of total thymocytes; right, CD44 vs CD25 staining of DN thymocytes. (c) Total thymocyte numbers and ratio of DN3 stage cells to DN4 stage cells (n=4 mice per genotype) in AND-tg Fbxl12fl/fl and AND-tg Lck-Cre Fbxl12fl/fl mice. For all graphs, horizontal lines indicate the mean and vertical lines indicate standard deviation (±s.d.), P values were determined by unpaired two-tailed Student’s t-test. NS, not significant (P>0.05), *P<0.05, **P<0.01, ***P<0.005, ****P<0.0001. Data shown in (a) and (b) are one representative of three experiments.

Supplementary Figure 6 β-selection-associated proliferation is strongly impaired in the absence of Fbxl1 and Fbxl12.

(a) Cell cycle analysis of thymocyte subsets from mice of the indicated genotype showing percentage of S/G2/M cells. (b) Numbers of CD4 SP and CD8 SP cells in the thymus and spleen of mice of the indicated genotype (n=5 mice per genotype). (c) Percentage of annexin V positive cells within the thymocyte subsets of the indicated genotype (n=3 mice per genotype). For all graphs, horizontal lines indicate the mean and vertical lines indicate standard deviation (±s.d.), P values were determined by unpaired two-tailed Student’s t-test. NS, not significant (P>0.05), *P<0.05, **P<0.01, ***P<0.005, ****P<0.0001. Data shown in (a) are representative of four independent experiments.

Supplementary Figure 7 β-selection associated proliferation is impaired in Fbxl1+/-Fbxl12+/- compound heterozygous thymocytes.

(a) Flow cytometry of cells from Thymus (left) or Spleen (right) from mice of the indicated genotype. Thymus: CD4 vs CD8 staining of total thymocytes; center, CD44 vs CD25 staining of lineage-negative DN thymocytes. Spleen: CD4 vs CD8 staining of total splenocytes. (b) Immunoblot analysis showing Fbxl1, Fbxl12, Cdkn1b and β-actin in purified lineage-negative DN thymocytes from mice of the indicated genotype. (c) Cell numbers and DN3/DN4 ratio of thymocytes from mice of the indicated genotype (n=4 mice per genotype). (d) Percentage of cycling S/G2/M stage cells in the indicated thymocyte subsets (n=4 mice per genotype). For all graphs, horizontal lines indicate the mean and vertical lines indicate standard deviation (±s.d.), P values were determined by unpaired two-tailed Student’s t-test. NS, not significant (P>0.05), *P<0.05, **P<0.01, ***P<0.005, ****P<0.0001. Data shown in (a) and (b) are representative of three or two independent experiments, respectively.

Supplementary Figure 8 Time-course of Fbxl1 and Fbxl12 induction and cell cycle analysis of thymocyte subsets.

(a) Real-time PCR analysis showing Fbxl1 and Fbxl12 mRNA expression in Rag2-/- thymocytes plated on OP9-DL1 cells for the indicated times. mRNA expression is relative to Day 0. Data shown are combined results of three experiments. (b) Real-time PCR analysis showing Fbxl1 and Fbxl12 mRNA expression in thymocytes of Rag2-/- mice that were injected (IP) with anti-CD3 antibody and harvested at the indicated times. mRNA expression is relative to Day 0. Data shown are combined results of three experiments. (c) Cell cycle analysis of thymocyte subsets from mice of the indicated genotype in Fig. 7a showing percentage of cells in S/G2/M phase. For all graphs, horizontal lines indicate the mean and vertical lines indicate the standard deviation (±s.d.), P values were determined by unpaired two-tailed Student’s t-test. NS, not significant (P>0.05), *P<0.05, **P<0.01, ***P<0.005, ****P<0.0001. Data shown in (c) are representative of three independent experiments.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, B., Yoganathan, K., Li, L. et al. Notch and the pre-TCR coordinate thymocyte proliferation by induction of the SCF subunits Fbxl1 and Fbxl12. Nat Immunol 20, 1381–1392 (2019). https://doi.org/10.1038/s41590-019-0469-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-019-0469-z

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