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:

Hippo/Mst signalling couples metabolic state and immune function of CD8α+ dendritic cells

Abstract

Dendritic cells orchestrate the crosstalk between innate and adaptive immunity. CD8α+ dendritic cells present antigens to CD8+ T cells and elicit cytotoxic T cell responses to viruses, bacteria and tumours1. Although lineage-specific transcriptional regulators of CD8α+ dendritic cell development have been identified2, the molecular pathways that selectively orchestrate CD8α+ dendritic cell function remain elusive. Moreover, metabolic reprogramming is important for dendritic cell development and activation3,4, but metabolic dependence and regulation of dendritic cell subsets are largely uncharacterized. Here we use a data-driven systems biology algorithm (NetBID) to identify a role of the Hippo pathway kinases Mst1 and Mst2 (Mst1/2) in selectively programming CD8α+ dendritic cell function and metabolism. Our NetBID analysis reveals a marked enrichment of the activities of Hippo pathway kinases in CD8α+ dendritic cells relative to CD8α dendritic cells. Dendritic cell-specific deletion of Mst1/2—but not Lats1 and Lats2 (Lats1/2) or Yap and Taz (Yap/Taz), which mediate canonical Hippo signalling—disrupts homeostasis and function of CD8+ T cells and anti-tumour immunity. Mst1/2-deficient CD8α+ dendritic cells are impaired in presentation of extracellular proteins and cognate peptides to prime CD8+ T cells, while CD8α dendritic cells that lack Mst1/2 have largely normal function. Mechanistically, compared to CD8α dendritic cells, CD8α+ dendritic cells exhibit much stronger oxidative metabolism and critically depend on Mst1/2 signalling to maintain bioenergetic activities and mitochondrial dynamics for their functional capacities. Further, selective expression of IL-12 by CD8α+ dendritic cells depends on Mst1/2 and the crosstalk with non-canonical NF-κB signalling. Our findings identify Mst1/2 as selective drivers of CD8α+ dendritic cell function by integrating metabolic activity and cytokine signalling, and highlight that the interplay between immune signalling and metabolic reprogramming underlies the unique functions of dendritic cell subsets.

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: NetBID identifies Hippo signalling kinases as drivers of CD8α+ DCs, and deletion of Mst1/2 in DCs leads to selective CD8+ T cell homeostatic and functional defects.
Fig. 2: Mst1/2 are selectively required in CD8α+ DCs to orchestrate CD8+ T cell homeostasis and function.
Fig. 3: Unique metabolic state of CD8α+ DCs supports their immune priming function in an Mst1/2-dependent manner.
Fig. 4: Mst1/2 orchestrate selective expression of IL-12 in CD8α+ DCs via crosstalk with non-canonical NF-κB signalling.

Similar content being viewed by others

References

  1. Merad, M., Sathe, P., Helft, J., Miller, J. & Mortha, A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu. Rev. Immunol. 31, 563–604 (2013).

    Article  PubMed  CAS  Google Scholar 

  2. Murphy, K. M. Transcriptional control of dendritic cell development. Annu. Rev. Immunol. 34, 93–119 (2016).

    Article  PubMed  CAS  Google Scholar 

  3. Pearce, E. J. & Everts, B. Dendritic cell metabolism. Nat. Rev. Immunol. 15, 18–29 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. O’Neill, L. A. J. & Pearce, E. J. Immunometabolism governs dendritic cell and macrophage function. J. Exp. Med. 213, 15–23 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Dudziak, D. et al. Differential antigen processing by dendritic cell subsets in vivo. Science 315, 107–111 (2007).

    Article  ADS  PubMed  CAS  Google Scholar 

  6. Piovan, E. et al. direct reversal of glucocorticoid resistance by AKT inhibition in acute lymphoblastic leukemia. Cancer Cell 24, 766–776 (2013).

    Article  PubMed  CAS  Google Scholar 

  7. Rodriguez-Barrueco, R. et al. Inhibition of the autocrine IL-6–JAK2–STAT3–calprotectin axis as targeted therapy for HR/HER2+ breast cancers. Genes Dev. 29, 1631–1648 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Huang, G. et al. Signaling via the kinase p38α programs dendritic cells to drive TH17 differentiation and autoimmune inflammation. Nat. Immunol. 13, 152–161 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Meng, Z., Moroishi, T. & Guan, K.-L. Mechanisms of Hippo pathway regulation. Genes Dev. 30, 1–17 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8α+ dendritic cells in cytotoxic T cell immunity. Science 322, 1097–1100 (2008).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  11. Mashayekhi, M. et al. CD8α+ dendritic cells are the critical source of interleukin-12 that controls acute infection by Toxoplasma gondii tachyzoites. Immunity 35, 249–259 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Naik, S. H. et al. Cutting edge: generation of splenic CD8+ and CD8 dendritic cell equivalents in fms-like tyrosine kinase 3 ligand bone marrow cultures. J. Immunol. 174, 6592–6597 (2005).

    Article  PubMed  CAS  Google Scholar 

  13. Cogliati, S. et al. Mitochondrial cristae shape determines respiratory chain supercomplexes assembly and respiratory efficiency. Cell 155, 160–171 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Buck, M. D. D. et al. Mitochondrial dynamics controls T cell fate through metabolic programming. Cell 166, 63–76 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Mishra, P. & Chan, D. C. Metabolic regulation of mitochondrial dynamics. J. Cell Biol. 212, 379–387 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Valenzuela, J., Schmidt, C. & Mescher, M. The roles of IL-12 in providing a third signal for clonal expansion of naive CD8 T cells. J. Immunol. 169, 6842–6849 (2002).

    Article  PubMed  CAS  Google Scholar 

  17. Sun, S. C. The non-canonical NF-κB pathway in immunity and inflammation. Nat. Rev. Immunol. 17, 545–558 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Katakam, A. K. et al. Dendritic cells require NIK for CD40-dependent cross-priming of CD8+ T cells. Proc. Natl Acad. Sci. USA 112, 14664–14669 (2015).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  19. Sasaki, Y. et al. NIK overexpression amplifies, whereas ablation of its TRAF3-binding domain replaces BAFF:BAFF-R-mediated survival signals in B cells. Proc. Natl Acad. Sci. USA 105, 10883–10888 (2008).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  20. Bachem, A. et al. Superior antigen cross-presentation and XCR1 expression define human CD11c+ CD141+ cells as homologues of mouse CD8+ dendritic cells. J. Exp. Med. 207, 1273–1281 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Lu, L. et al. Hippo signaling is a potent in vivo growth and tumor suppressor pathway in the mammalian liver. Proc. Natl Acad. Sci. USA 107, 1437–1442 (2010).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  22. Xin, M. et al. Hippo pathway effector Yap promotes cardiac regeneration. Proc. Natl Acad. Sci. USA 110, 13839–13844 (2013).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  23. Wang, Y. et al. Transforming growth factor beta-activated kinase 1 (TAK1)-dependent checkpoint in the survival of dendritic cells promotes immune homeostasis and function. Proc. Natl Acad. Sci. USA 109, E343–E352 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Jongbloed, S. L. et al. Human CD141+ (BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens. J. Exp. Med. 207, 1247–1260 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Fan, F. et al. Pharmacological targeting of kinases MST1 and MST2 augments tissue repair and regeneration. Sci. Transl. Med. 8, 352ra108 (2016).

    Article  ADS  PubMed  CAS  Google Scholar 

  26. Wang, D. et al. A small molecule promotes mitochondrial fusion in mammalian cells. Angew. Chem. Int. Ed. 51, 9302–9305 (2012).

    Article  CAS  Google Scholar 

  27. Cassidy-Stone, A. et al. Chemical inhibition of the mitochondrial division dynamin reveals its role in Bax/Bak-dependent mitochondrial outer membrane permeabilization. Dev. Cell 14, 193–204 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Maraskovsky, E. et al. Dramatic increase in the numbers of functionally mature dendritic cells in Flt3 ligand-treated mice: multiple dendritic cell subpopulations identified. J. Exp. Med. 184, 1953–1962 (1996).

    Article  PubMed  CAS  Google Scholar 

  29. den Haan, J. M., Lehar, S. M. & Bevan, M. J. CD8+ but not CD8 dendritic cells cross-prime cytotoxic T cells in vivo. J. Exp. Med. 192, 1685–1696 (2000).

    Article  Google Scholar 

  30. Kretzer, N. M. et al. RAB43 facilitates cross-presentation of cell-associated antigens by CD8α+ dendritic cells. J. Exp. Med. 213, 2871–2883 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Wang, Y. et al. Tuberous sclerosis 1 (Tsc1)-dependent metabolic checkpoint controls development of dendritic cells. Proc. Natl Acad. Sci. USA 110, E4894–E4903 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Liedmann, S. et al. Viral suppressors of the RIG-I-mediated interferon response are pre-packaged in influenza virions. Nat. Commun. 5, 5645 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Huang, B., Wang, W., Bates, M. & Zhuang, X. Three-dimensional super-resolution imaging by stochastic optical reconstruction microscopy. Science 319, 810–813 (2008).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  34. Tan, H. et al. Integrative proteomics and phosphoproteomics profiling reveals dynamic signaling networks and bioenergetics pathways underlying T cell activation. Immunity 46, 488–503 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Wang, H. et al. Systematic optimization of long gradient chromatography mass spectrometry for deep analysis of brain proteome. J. Proteome Res. 14, 829–838 (2015).

    Article  PubMed  CAS  Google Scholar 

  36. Zeng, H. et al. mTORC1 couples immune signals and metabolic programming to establish Treg-cell function. Nature 499, 485–490 (2013).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  37. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Putcha, P. et al. HDAC6 activity is a non-oncogene addiction hub for inflammatory breast cancers. Breast Cancer Res. 17, 149 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Wang, K. et al. Genome-wide identification of post-translational modulators of transcription factor activity in human B cells. Nat. Biotechnol. 27, 829–837 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Alvarez, M. J. et al. Functional characterization of somatic mutations in cancer using network-based inference of protein activity. Nat. Genet. 48, 838–847 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Lefebvre, C. et al. A human B-cell interactome identifies MYB and FOXM1 as master regulators of proliferation in germinal centers. Mol. Syst. Biol. 6, 377 (2010).

    Article  MathSciNet  PubMed  PubMed Central  CAS  Google Scholar 

  42. Margolin, A. A. et al. ARACNE: An algorithm for the reconstruction of gene regulatory networks in a mammalian cellular context. BMC Bioinformatics 7, S7 (2006).

    Article  PubMed  PubMed Central  MathSciNet  CAS  Google Scholar 

  43. Efron, B. & Tibshirani, R. On testing the significance of sets of genes. Ann. Appl. Stat. 1, 107–129 (2007).

    Article  MathSciNet  MATH  Google Scholar 

  44. Irizarry, R. A., Wang, C., Zhou, Y. & Speed, T. P. Gene set enrichment analysis made simple. Stat. Methods Med. Res. 18, 565–575 (2009).

    Article  MathSciNet  PubMed  PubMed Central  Google Scholar 

  45. Gelman, A. et al. Bayesian Data Analysis 3rd edn. (CRC Press, Boca Raton, 2013).

    Google Scholar 

  46. Riley, J. W. et al. The American soldier: adjustment during army life. Am. Sociol. Rev. 14, 557–559 (1949).

    Article  Google Scholar 

  47. 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  ADS  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

The authors acknowledge N. Chapman for critical reading of the manuscript, R. Johnson for Stk4 and Stk3 floxed mice, E. Olson for Yap1 and Taz floxed mice, S.C. Sun for the NIK(Δ78–84) construct, M. Hendren and A. KC for animal work, SJCRH Electron Microscopy core resource for electron microscopy, and Immunology FACS core facility for cell sorting. This work was supported by NIH AI105887, AI101407, CA176624, CA221290, NS064599 (to H.C.), and AG047928 (to J.P.).

Reviewer information

Nature thanks L. O’Neill and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

X.D. designed and performed in vitro and in vivo experiments, and wrote the manuscript; J.W. performed human DC study and immunoprecipitation; Y.W. initiated the project, and together with P.W.F.K. performed Seahorse assays; A.K. performed network inference; H.T., Y.L. and J.P. performed and analysed proteomics assays; C.G. performed STORM assays and helped with electron microscopy experiments; T.-L.M.N. performed the tumour study; Y.D. and G.N. performed functional enrichment analysis of microarrays; J.Y. developed the NetBID algorithm, performed systems biology analysis, wrote the manuscript and supervised overall computational analysis; and H.C. designed experiments, wrote the manuscript, and provided overall direction.

Corresponding authors

Correspondence to Jiyang Yu or Hongbo Chi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 NetBID analysis for the reconstruction of the DC signalling interactome (DCI), network and enrichment analyses of top kinase drivers, and identification and validation of Stk4 (Mst1) regulons.

a, PCA plot of baseline microarray gene expression profiles of total DCs (blue, n = 15; used for de novo DCI reconstruction), CD8α+ and CD8α DCs (green and red, respectively, n = 4 each; used for differential expression analysis) after removal of batch effects. b, Top 36 hub kinases that are differentially activated in CD8α+ DCs relative to CD8α DCs inferred by NetBID. Left, the NetBID panel indicates the significance level (colour coded by z score; labelled values are P values) of the driver network in integrated analysis, transcriptomics (mRNA), whole proteomics (wProtein) and phosphoproteomics (pProtein) data, respectively. Right, differential expression of the drivers (colour coded by z score; labelled values are signed fold changes). The Venn diagram shows the enrichment of Hippo pathway kinases in the top putative kinase drivers. c, Network interactions of top 36 kinase drivers of CD8α+ DCs. d, Top signalling pathways enriched by 36 NetBID-inferred kinase drivers (P < 0.01, number of overlapped genes >2). e, Known kinases in Hippo signalling9 and analysis by NetBID. f, Stk4-mediated gene network (n = 140) from DCI computationally inferred from baseline gene expression profiles of total DCs by NetBID. The width of an edge is proportional to the pairwise mutual information of connected nodes. g, h, Enrichment of predicted Mst1 signalling regulons (as shown in f) in differentially expressed genes between Mst1/2-deficient (Mst1/2∆DC) and wild-type CD8α+ DCs (g) or CD8α DCs (h). Pval.GSEA indicates the P value of GSEA; Pval.Stk4 and FC.Stk4 indicate the P value and signed fold change of Stk4 expression (insert).

Extended Data Fig. 2 T cell homeostasis in mice with DC-specific deletion of Hippo pathway genes.

a, b, Total cellularity (a) or T cell numbers (TCRβ+ CD8+ and TCRβ+ CD4+) (b) of the spleen, peripheral lymph nodes (PLN) and mesenteric lymph nodes (MLN) of wild-type and Mst1/2∆DC mice (n = 5 mice per genotype). c, Flow cytometry analysis of splenic CD4+ and CD8+ T cell populations (upper) and frequencies of total CD4+ and CD8+ T cells in spleen, PLN and MLN (lower) of wild-type and Mst1/2∆DC mice (n = 8 mice per genotype). d, CD44 and CD62L expression on splenic CD4+ and CD8+ T cells of wild-type and Mst1/2∆DC mice. e, CD44 and IFNγ expression in splenic CD4+ and CD8+ T cells. fk, Flow cytometry analysis of CD4+ and CD8+ populations, expression of CD44 and CD62L, or CD44 and IFNγ in CD4+ and CD8+ T cells from spleen of wild-type and Lats1/2∆DC (fh) or Yap/Taz∆DC mice (ik). ln, Frequencies of CD44high CD62Llow effector/memory cells (l) and CD44+ IFNγ+ cells (m) in splenic CD4+ and CD8+ T cells and frequencies of splenic CD4+ and CD8+ T cells (n) of wild-type (n = 9), Cd11ccreStk4fl/flStk3+/+ (n = 5), Cd11ccreStk4+/+Stk3fl/fl (n = 4), Cd11ccreStk4fl/+Stk3fl/fl (n = 5) and Cd11ccreStk4fl/flStk3fl/+ (n = 4) mice (Cd11c is also known as Itgax). Numbers in quadrants or gates indicate percentage of cells. Data are mean and s.e.m. NS, not significant; *P < 0.05; **P < 0.01; two-tailed unpaired Student’s t-test in ac; one-way ANOVA in ln. Data summarize two (ik), three (fh), four (a, b, d, e), six (c) or eight (ln) independent experiments.

Source Data.

Extended Data Fig. 3 Analyses of Mst1 and Mst2 deletion in DCs and lymphocytes from Mst1/2∆DC mice and T cell homeostatic status in mixed bone marrow chimaeras.

a, Real-time PCR (upper and middle) and immunoblot (lower) analyses of Stk4 (also known as Mst1) and Stk3 (also known as Mst2) mRNA and protein expression in CD4+ T cells, CD8+ T cells and B cells from wild-type and Mst1/2∆DC mice (n = 3 mice per genotype). b, Real-time PCR analysis of Stk4 and Stk3 mRNA expression in splenic CD8α+ and CD8α DCs from wild-type and Mst1/2∆DC mice (n = 3 for accessing Mst2 expression in Mst1/2-deficient CD8α+ DCs, n = 4 for others). c, d, Bone marrow cells from wild-type or Mst1/2∆DC CD45.2.2+ mice were mixed with cells from CD45.1.2+ (spike) mice at a 1:1 ratio and transferred into lethally irradiated CD45.1.1+ mice. After 6–8 weeks, bone marrow chimaeras were analysed for the expression of CD44, CD62L and IFNγ (c) and frequencies of CD44high CD62Llow effector/memory cells and CD44+ IFNγ+ cells (d) in splenic CD8+ T cells derived from wild-type or Mst1/2∆DC donor bone marrow cells (CD45.2.2+) or spike cells CD45.1.2+ (n = 5 mice per genotype). Numbers in quadrants indicate percentage of cells. Data are shown as mean and s.e.m. **P < 0.01; two-tailed unpaired Student’s t-test in a, b, d. Data summarize two (a, b) or three (c, d) independent experiments.

Source Data.

Extended Data Fig. 4 In vivo T cell responses in wild-type and Mst1/2∆DC mice challenged with tumour, pathogen or cognate antigen.

a, b, Flow cytometry analysis of IFNγ expression (left) and frequencies of IFNγ+ cells (right) in CD8+ (a) and CD4+ (b) T cells from draining lymph node (DLN) and tumour tissues of wild-type and Mst1/2∆DC mice challenged with MC38 tumour cells (n = 7 for wild type, n = 5 for Mst1/2∆DC for DLN; n = 4 for wild type, n = 8 for Mst1/2∆DC for tumour tissues). c, Flow cytometry analysis of PD-1 expression on CD8+ (upper) and CD4+ (lower) T cells from DLN and tumour tissues of wild-type and Mst1/2∆DC mice challenged with MC38 tumour cells. d, Flow cytometry analysis of LAG3 and TIM3 expression of CD8+ (upper) and CD4+ (lower) T cells from DLN of wild-type and Mst1/2∆DC mice challenged with MC38 tumour cells. e, Flow cytometry of H-2Kb-OVA+ CD8+ T cells in blood from wild-type and Mst1/2∆DC mice infected with LM-OVA. f, Flow cytometry (left) and frequencies (right) of IFNγ+ and TNFα+ cells of PMA and ionomycin-stimulated CD8+ T cells in the blood from wild-type and Mst1/2∆DC mice infected with LM-OVA (n = 5 for wild type, n = 4 for Mst1/2∆DC). g, CFSE dilution of donor OT-I T cells in OVA-immunized mice. Numbers in quadrants or gates indicate percentage of cells. Data are shown as mean and s.e.m. *P < 0.05, **P < 0.01; two-tailed unpaired Student’s t-test in a, b, f. Data summarize two (ae, g) independent experiments.

Source Data.

Extended Data Fig. 5 Altered homeostasis of Mst1/2-deficient DCs.

a, Detailed gating strategy for flow cytometry of splenic conventional DCs (cDC), CD8α+ cDC (CD8α+ CD11b), CD8α cDC (CD8α CD11b+) and pDC populations in wild-type and Mst1/2∆DC mice. MHC-II, MHC class II. b, Frequencies and cell numbers of splenic DC populations in wild-type and Mst1/2∆DC mice as gated in a (n = 5 mice per genotype). c, Flow cytometry analysis of CD40, CD70, CD80, CD86, H-2Kb, MHC-II, 4-1BBL, OX40L and PD-L1 expression on splenic CD8α+ and CD8α DCs as gated in a. d, Flow cytometry (left) of CD45.1.2+ bone marrow cell-derived and CD45.2.2+ wild-type or Mst1/2∆DC donor bone marrow cell-derived splenic cDC, CD8α+ cDC, CD8α cDC and pDC populations in mixed chimaeras and frequencies (right) of CD8α+ cDCs in total cDCs and pDCs in spleen derived from wild-type or Mst1/2∆DC donor bone marrow cells in mixed chimaeras (n = 5 mice per genotype). e, Normalized chimaerism for the indicated DC subsets in mixed bone marrow chimaeras. The percentage of indicated DC subsets was normalized by that of B cells from same mice. The chimaerism of the wild type was set as 1 (n = 5 mice per genotype). f, Flow cytometry analysis of donor (wild-type or Mst1/2∆DC, CD45.2.2+) and spike (CD45.1.2+) bone marrow cell percentages in the bone marrow mixture before transfer. Numbers in gates indicate percentage of cells. Data are shown as mean and s.e.m. *P < 0.05, **P < 0.01; two-tailed unpaired Student’s t-test in b, d, e. Data summarize four (ac) or three (d, e) independent experiments.

Source Data.

Extended Data Fig. 6 Homeostasis of DCs after deletion of Hippo pathway genes.

a, Frequencies of splenic cDC, CD8α+ cDC, and pDC populations in wild-type (n = 9), Cd11ccreStk4fl/flStk3+/+ (n = 5), Cd11ccreStk4+/+Stk3fl/fl (n = 4), Cd11ccreStk4fl/+Stk3fl/fl (n = 5) and Cd11ccreStk4fl/flStk3fl/+ (n = 4) mice. b, c, Flow cytometry of splenic cDC, CD8α+ cDC, CD8α cDC and pDC populations in wild-type and Lats1/2∆DC (b), or Yap/Taz∆DC (c) mice. Numbers in gates indicate percentage of cells. Data summarize two (b), three (c) or eight (a) independent experiments.

Source Data.

Extended Data Fig. 7 Role of Mst1/2 in selectively programming functions of CD8α+ DCs and CD24high FLT3L-BMDCs to prime CD8 T cells.

a, Frequency of CFSElow cells of donor OT-I T cells in wild-type (n = 5), Mst1/2∆DC (n = 5), Batf3−/−:wild-type (n = 5) or Batf3−/−:Mst1/2∆DC (n = 6) mixed chimaeras immunized with OVA. b, CFSE-labelled OT-I T cells (CD45.1+) were transferred to Β2m−/− mice followed by immunization one day later with OVA-pulsed CD8α+ or CD8α splenic DCs isolated from wild-type or Mst1/2∆DC mice (following DC expansion by FLT3L), and CFSE dilution of OT-I cells was examined three days after DC immunization. Shown are representative flow cytometry histograms of CFSE dilution and frequency of CFSElow cells in donor OT-I T cells (gated on CD45.1+CD45.2) from spleen of B2m−/− mice (n = 4 for Mst1/2-deficient CD8α DCs, n = 3 for all others). c, d, CD8α+ (c) or CD8α DCs (d) from wild-type and Mst1/2∆DC mice were fed with 0 or 50 μg/ml soluble OVA conjugated with FITC for 1 h at 37 °C or 4 °C. Cells were then collected and OVA uptake was evaluated by flow cytometry. Numbers in graphs indicate the mean fluorescence intensity of OVA-FITC. e, Flow cytometry (upper) and quantification (lower, n = 4 per genotype) of apoptotic CD8α+ or CD8α DCs examined by Annexin V (left) and active caspase 3 (right) staining in freshly isolated splenocytes from wild-type and Mst1/2∆DC mice. f, CD8α+ (upper) or CD8α DCs (lower) from wild-type and Mst1/2∆DC mice were cultured overnight for analysis of cell viability by 7-aminoactinomycin D (7-AAD) staining. Quantification of the percentage of 7-AAD-negative live cells is shown (n = 3 per genotype). g, Thymidine incorporation of OT-II T cells cultured with OVA protein- or OVA(323–339) peptide-pulsed CD8α+ or CD8α DCs (n = 13 derived from five mice for the Mst1/2-deficient CD8α+ DC/OVA(323–339) group, n = 15 derived from five mice for all other groups). hj, Thymidine incorporation of OT-I (left) or OT-II (right) T cells cultured with OVA protein-pulsed CD24high FLT3L-BMDCs (h), CD24low FLT3L-BMDCs (i), or GM-CSF-derived BMDCs (j) from wild-type and Mst1/2∆DC mice for 72 h (n = 14 from five mice for the Mst1/2-deficient GM-CSF-BMDC/OT-I group, n = 15 from five mice for all other groups). k, Relative thymidine incorporation of OT-I T cells cultured with OVA protein- or OVA(257–264) peptide-pulsed splenic CD8α+ DCs pre-treated with vehicle or Mst1/2 inhibitor (XMU-MP-1) for 4 h. Thymidine incorporation of OT-I T cells cultured with vehicle-treated DCs in each group was set as 1 (n = 3 per group). l, Relative thymidine incorporation of OT-I T cells cultured with OVA protein- or OVA(257–264) peptide-pulsed CD8α+ DCs from WT and Lats1/2∆DC mice (n = 14 derived from five mice for wild type, n = 15 derived from five mice for Lats1/2∆DC). Thymidine incorporation of OT-I T cells cultured with wild-type DCs in each group was set as 1. m, Relative thymidine incorporation of OT-I T cells cultured with OVA protein-pulsed wild-type or Yap/Taz∆DC splenic CD8α+ DCs that were pre-treated with vehicle or Mst1/2 inhibitor (n = 3 derived from two mice per genotype). Thymidine incorporation of OT-I T cells cultured with vehicle-treated DCs was set as 1. Numbers in gates indicate percentage of cells. Data are shown as mean and s.e.m. *P < 0.05, **P < 0.01; two-tailed unpaired Student’s t-test in a, b, el; two-way ANOVA in m. Data summarize two (ad, gj), three (e, f, k, l) or four (e) independent experiments.

Source Data.

Extended Data Fig. 8 Analysis of mitochondrial profiles of Mst1/2-, Lats1/2- or mTOR-deficient DCs.

a, Functional annotations of upregulated metabolic pathways according to KEGG and Hallmark databases in CD8α+ DCs (compared to CD8α DCs) profiled using proteomics. b, ECAR of splenic CD8α+ and CD8α DCs. Oligo, oligomycin; FCCP, carbonyl cyanide p-trifluoromethoxyphenylhydrazone. c, Flow cytometry analysis of mitochondrial mass and membrane potential of wild-type splenic CD8α+ and CD8α DCs using Mitotracker and TMRM (Tetramethylrhodamine, methyl ester) staining, respectively. Numbers in graph indicate the mean fluorescence intensity. d, Relative thymidine incorporation of OT-I T cells cultured with OVA protein- or OVA(257–264) peptide-pulsed CD8α+ and CD8α DCs pre-treated with vehicle or metabolic inhibitors. Values after culture with vehicle-treated DCs were set as 1. e, Thymidine incorporation of OT-I T cells cultured with OVA protein- or OVA(257–264) peptide-pulsed splenic CD8α+ DCs from wild-type and mTOR∆DC mice for 72 h (n = 12 from four mice per genotype). f, Flow cytometry analysis of mitochondrial mass and mitochondrial membrane potential of wild-type and Mst1/2∆DC splenic CD8α+ and CD8α DCs by Mitotracker and TMRM staining, respectively. Numbers in graph indicate the mean fluorescence intensity. g, Transmission electron microscopy analysis of mitochondria of splenic CD8α DCs from wild-type or Mst1/2∆DC mice. Arrows indicate mitochondria. h, Immunoblot analysis of expression of NDUFB8 (complex I), SDHB (complex II), UQCRC2 (complex III), MT-CO1 (complex IV) and ATP5A (complex V) in CD8α+ and CD8α DCs. i, Flow cytometry analysis of mitochondrial mass and mitochondrial membrane potential of wild-type and Lats1/2∆DC splenic CD8α+ and CD8α DCs by Mitotracker and TMRM staining, respectively. Numbers in graph indicate the mean fluorescence intensity. j, Immunoblot analysis of p-S6 and c-Myc protein in CD8α+ and CD8α DCs of wild-type and Mst1/2∆DC mice. k, Immunoblot analysis of NDUFB8 (complex I), SDHB (complex II), UQCRC2 (complex III), MT-CO1 (complex IV) and ATP5A (complex V) protein in CD8α+ and CD8α DCs of wild-type and mTOR∆DC mice. Data are shown as mean and s.e.m. *P < 0.05, **P < 0.01; one-way ANOVA in d; two-tailed unpaired Student’s t-test in e. Data summarize two (e, f, hk), three (b, d) or four (c) independent experiments.

Source Data.

Extended Data Fig. 9 Selective regulation of IL-12 signalling and expression by Mst1/2 in CD8α+ DCs.

a, Venn diagram showing the overlap of top 28 upregulated (Mst1/2∆DC versus wild type) pathways by GSEA between CD8α+ and CD8α DCs. Briefly, transcriptional profiles of Mst1/2-deficient CD8α+ and CD8α DCs were compared to their respective wild-type counterparts by GSEA, and then upregulated pathways (FDR < 0.05) were identified in CD8α+ DCs (Mst1/2ΔDC versus wild type) and CD8α DCs (Mst1/2ΔDC versus wild type) and used to generate a Venn diagram. The top 28 upregulated pathways in Mst1/2-deficient CD8α+ and CD8α DCs (compared to their respective wild-type counterparts) were largely shared (24/28) between the two DC subsets. b, List of the significantly upregulated (FDR < 0.05) 24 pathways (out of 28) shared by Mst1/2-deficient CD8α+ and CD8α DCs (compared to their respective wild-type counterparts), as revealed by GSEA. c, List of the significantly downregulated (FDR < 0.05) pathways determined by GSEA (arranged according to FDR values) in Mst1/2-deficient CD8α+ DCs (versus wild-type cells). NES, normalized enrichment score. d, Il12b expression in wild-type CD8α+ and CD8α DCs. e, Relative IL-12 p40 cytokine concentration in the supernatant of lipopolysaccharide (LPS)-treated CD8α+ and CD8α DCs from wild-type and Mst1/2∆DC mice (n = 4 per genotype for CD8α+ DCs, n = 3 per genotype for CD8α DCs). IL-12 p40 cytokine concentration of wild-type DCs was set as 1. f, Real-time PCR analysis of Il12b mRNA expression in wild-type splenic CD8α+ and CD8α DCs treated with vehicle or Mst1/2 inhibitor (XMU-MP-1) (n = 4 per treatment). g, Il12b expression in FLT3L-BMDCs (n = 4 for wild-type CD24high BMDCs, n = 5 for other groups). h, i, Real-time PCR analysis of Il1a, Il1b, Il6, Il10 and Ifnb1 mRNA expression in splenic CD8α+ (h) and CD8α (i) DCs from wild-type and Mst1/2ΔDC mice (n = 4 mice per genotype). j, k, Frequencies of CD44high CD62Llow (j) and CD44+ IFNγ+ (k) cells in splenic CD4+ and CD8+ T cells from wild-type (n = 5), Mst1/2∆DC (n = 5), Il12a−/− (n = 6) and Mst1/2∆DC Il12a−/− (n = 6) mice. Wild-type and Mst1/2ΔDC groups were the same as those shown in Fig. 1d, e. Data are shown as mean and s.e.m. *P < 0.05, **P < 0.01; unpaired Student’s t-test in d, fi; two-tailed paired Student’s t-test in e; one-way ANOVA in j, k. Data summarize two (d, g), three (e), four (j, k) or five (f) independent experiments.

Source Data.

Extended Data Fig. 10 Mst/Hippo signalling integrates mitochondrial metabolism and non-canonical NF-κB/IL-12 signalling in CD8α+ DCs.

a, Relative thymidine incorporation of OT-I T cells cultured with OVA protein- or OVA(257–264) peptide-pulsed splenic CD8α+ DCs (left) or CD8α DCs (right) from wild-type and Il12a−/− mice (n = 9 derived from three mice for wild type, n = 12 derived from four mice for Il12a−/−). Thymidine incorporation of OT-I T cells cultured with wild-type DCs in each group was set as 1. b, Real-time PCR analysis of Nfkb2 (left) or Relb (right) mRNA expression in splenic CD8α+ DCs from wild-type and Mst1/2∆DC mice (n = 3 mice per population). c, NF-κB2 and RelB expression in control- or NIK(Δ78–84)-transduced wild-type CD11c+ B220 FLT3L-BMDCs. d, Flow cytometry analysis of mitochondrial membrane potential of wild-type or Mst1/2-deficient CD24high FLT3L-BMDCs transduced with control (left) or NIK(∆78–84) (right) virus. Numbers in graph indicate the mean fluorescence intensity. e, Thymidine incorporation of OT-I T cells cultured with OVA protein-pulsed wild-type or Mst1/2-deficient CD24high FLT3L-BMDCs transduced with control or NIK(∆78–84) virus (n = 6 derived from three mice for wild-type/control group, n = 8 derived from three mice for Mst1/2∆DC/control group, n = 4 derived from three mice per genotype for NIK(∆78–84) group). f, FLT3L-expanded splenic CD8α+ DC lysate was immunoprecipitated with anti-Mst1 antibody and blotted with anti-Traf3. Mst1 blot was from the same experiment as Fig. 3f. g, Immunoblot analysis of Traf3 protein in splenic CD8α+ and CD8α DCs from wild-type and Mst1/2∆DC mice. h, Real-time PCR analysis of Il12b expression in wild-type CD8α+ DCs treated with vehicle, metformin or oligomycin as indicated in figures (n = 3 per group). Il12b expression of vehicle-treated DCs was set as 1. i, Flow cytometry analysis of mitochondrial membrane potential (upper) and mitochondrial mass (lower) of different concentrations of IL-12-treated wild-type splenic CD8α+ DCs by TMRM (tetramethylrhodamine, methyl ester) and Mitotracker staining. Numbers in graph indicate the mean fluorescence intensity. j, Flow cytometry analysis of mitochondrial membrane potential and mitochondrial mass of wild-type and Il12a−/− splenic CD8α+ DCs by TMRM and Mitotracker staining. k, Flow cytometry analysis of mitochondrial membrane potential and mitochondrial mass of wild-type and NIK-deficient (Map3k14−/−) splenic CD8α+ DCs by TMRM and Mitotracker staining. l, Immunoblot analysis of p-Lats1/2, p-Yap and p-Mst1/2 proteins in wild-type splenic CD8α+ DCs treated with FLT3L for the indicated times. m, CD8α+ DC number from spleen of wild-type and Mst1/2∆DC mice treated with or without FLT3L for 10 days. n, Flow cytometry analysis of mitochondrial mass of human CD141+ DCs treated with vehicle or Mst1/2 inhibitor (XMU-MP-1) by Mitotracker staining. o, Real-time PCR analysis of Il12b mRNA expression in human CD141+ (equivalent to mouse CD8α+ DCs) and CD1c+ DCs (equivalent to mouse CD8α DCs) treated with vehicle or Mst1/2 inhibitor (XMU-MP-1) (n = 5 for CD141+ DC, n = 4 for CD1c+ DC). Data are shown as mean and s.e.m. *P < 0.05, **P < 0.01; two-tailed unpaired Student’s t-test in a, b, m, o; two-way ANOVA in e. Data summarize two (a, dg, i, j, l, n, o) or three (h, k, m) independent experiments. p, Brief schematics of non-canonical Hippo signalling in orchestrating CD8α+ DC function. Mst/Hippo signalling integrates metabolic and IL-12 cytokine signalling in CD8α+ DCs through controlling mitochondrial dynamics and non-canonical NF-κB signalling. This regulation is independent of canonical Hippo signalling in organ size control and tumour suppression.

Source Data.

Supplementary information

Supplementary Figure 1

The uncropped western blot images with size marker indications.

Reporting Summary

Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Du, X., Wen, J., Wang, Y. et al. Hippo/Mst signalling couples metabolic state and immune function of CD8α+ dendritic cells. Nature 558, 141–145 (2018). https://doi.org/10.1038/s41586-018-0177-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-018-0177-0

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

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