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:

Fasting-induced FOXO4 blunts human CD4+ T helper cell responsiveness

Abstract

Intermittent fasting blunts inflammation in asthma1 and rheumatoid arthritis2, suggesting that fasting may be exploited as an immune-modulatory intervention. However, the mechanisms underpinning the anti-inflammatory effects of fasting are poorly characterized3,4,5. Here, we show that fasting in humans is sufficient to blunt CD4+ T helper cell responsiveness. RNA sequencing and flow cytometry immunophenotyping of peripheral blood mononuclear cells from volunteers subjected to overnight or 24-h fasting and 3 h of refeeding suggest that fasting blunts CD4+ T helper cell activation and differentiation. Transcriptomic analysis reveals that longer fasting has a more robust effect on CD4+ T-cell biology. Through bioinformatics analyses, we identify the transcription factor FOXO4 and its canonical target FK506-binding protein 5 (FKBP5) as a potential fasting-responsive regulatory axis. Genetic gain- or loss-of-function of FOXO4 and FKBP5 is sufficient to modulate TH1 and TH17 cytokine production. Moreover, we find that fasting-induced or genetic overexpression of FOXO4 and FKBP5 is sufficient to downregulate mammalian target of rapamycin complex 1 signalling and suppress signal transducer and activator of transcription 1/3 activation. Our results identify FOXO4–FKBP5 as a new fasting-induced, signal transducer and activator of transcription–mediated regulatory pathway to blunt human CD4+ T helper cell responsiveness.

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: RNA-seq analysis shows distinct separation of the three caloric load conditions.
Fig. 2: Fasting/feeding differentially regulate CD4+ T-cell activation and differentiation.
Fig. 3: Fasting-induced FOXO4 effects.
Fig. 4: FKBP5 induction mimics FOXO4 overexpression and fasting effects on T-cell activation.

Similar content being viewed by others

Data availability

The mRNA-seq datasets and data linked to the participants’ body mass index and sex can be accessed at the GEO database under accession number GSE165149 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE165149). All code used in the study can be found at https://github.com/NHLBI-BCB/PTNA. Source data are provided with this paper.

Code availability

All code used in the study can be found at https://github.com/NHLBI-BCB/PTNA. Please also remove this text from the Data availability section.

References

  1. Johnson, J. B. et al. Alternate day calorie restriction improves clinical findings and reduces markers of oxidative stress and inflammation in overweight adults with moderate asthma. Free Radic. Biol. Med. 42, 665–674 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Fraser, D. A. et al. Reduction in serum leptin and IGF-1 but preserved T-lymphocyte numbers and activation after a ketogenic diet in rheumatoid arthritis patients. Clin. Exp. Rheumatol. 18, 209–214 (2000).

    CAS  PubMed  Google Scholar 

  3. Traba, J. et al. Fasting and refeeding differentially regulate NLRP3 inflammasome activation in human subjects. J. Clin. Invest. 125, 4592–4600 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Meydani, S. N. et al. Long-term moderate calorie restriction inhibits inflammation without impairing cell-mediated immunity: a randomized controlled trial in non-obese humans. Aging (Albany NY) 8, 1416–1431 (2016).

    Article  CAS  Google Scholar 

  5. Sack, M. N. Mitochondrial fidelity and metabolic agility control immune cell fate and function. J. Clin. Invest. 128, 3651–3661 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Han, K. et al. A pilot study to investigate the immune-modulatory effects of fasting in steroid-naive mild asthmatics. J. Immunol. 201, 1382–1388 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Husebye, H. et al. Endocytic pathways regulate Toll-like receptor 4 signaling and link innate and adaptive immunity. EMBO J. 25, 683–692 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Crotzer, V. L. & Blum, J. S. Cytosol to lysosome transport of intracellular antigens during immune surveillance. Traffic 9, 10–16 (2008).

    Article  CAS  PubMed  Google Scholar 

  9. Pasquale, E. B. Eph-ephrin bidirectional signaling in physiology and disease. Cell 133, 38–52 (2008).

    Article  CAS  PubMed  Google Scholar 

  10. Finak, G. et al. Standardizing flow cytometry immunophenotyping analysis from the Human ImmunoPhenotyping Consortium. Sci. Rep. 6, 20686 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008).

    PubMed  PubMed Central  Google Scholar 

  12. Shoemaker, J. E. et al. CTen: a web-based platform for identifying enriched cell types from heterogeneous microarray data. BMC Genomics 13, 460 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lee, G. R., Fields, P. E. & Flavell, R. A. Regulation of IL-4 gene expression by distal regulatory elements and GATA-3 at the chromatin level. Immunity 14, 447–459 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Xiao, H. et al. HDAC5 controls the functions of Foxp3+ T-regulatory and CD8+ T cells. Int. J. Cancer 138, 2477–2486 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Klemann, C. et al. Clinical and immunological phenotype of patients with primary immunodeficiency due to damaging mutations in NFKB2. Front. Immunol. 10, 297 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Grant, C. E., Bailey, T. L. & Noble, W. S. FIMO: scanning for occurrences of a given motif. Bioinformatics 27, 1017–1018 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Howell, J. J. & Manning, B. D. mTOR couples cellular nutrient sensing to organismal metabolic homeostasis. Trends Endocrinol. Metab. 22, 94–102 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Durant, L. et al. Diverse targets of the transcription factor STAT3 contribute to T cell pathogenicity and homeostasis. Immunity 32, 605–615 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Traba, J. et al. Prolonged fasting suppresses mitochondrial NLRP3 inflammasome assembly and activation via SIRT3-mediated activation of superoxide dismutase 2. J. Biol. Chem. 292, 12153–12164 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ciofani, M. et al. A validated regulatory network for Th17 cell specification. Cell 151, 289–303 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Holloway, A. F., Rao, S. & Shannon, M. F. Regulation of cytokine gene transcription in the immune system. Mol. Immunol. 38, 567–580 (2002).

    Article  CAS  PubMed  Google Scholar 

  22. Vallabhapurapu, S. & Karin, M. Regulation and function of NF-κB transcription factors in the immune system. Annu. Rev. Immunol. 27, 693–733 (2009).

    Article  CAS  PubMed  Google Scholar 

  23. Smale, S. T. Transcriptional regulation in the immune system: a status report. Trends Immunol. 35, 190–194 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Coffer, P. J. & Burgering, B. M. T. Forkhead-box transcription factors and their role in the immune system. Nat. Rev. Immunol. 4, 889–899 (2004).

    Article  CAS  PubMed  Google Scholar 

  25. Hori, S., Nomura, T. & Sakaguchi, S. Control of regulatory T cell development by the transcription factor Foxp3. Science 299, 1057–1061 (2003).

    Article  CAS  PubMed  Google Scholar 

  26. Zhou, W. et al. FoxO4 inhibits NF-κB and protects mice against colonic injury and inflammation. Gastroenterology 137, 1403–1414 (2009).

    Article  CAS  PubMed  Google Scholar 

  27. Baughman, G., Wiederrecht, G. J., Campbell, N. F., Martin, M. M. & Bourgeois, S. FKBP51, a novel T-cell-specific immunophilin capable of calcineurin inhibition. Mol. Cell. Biol. 15, 4395–4402 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zannas, A. S. et al. Epigenetic upregulation of FKBP5 by aging and stress contributes to NF-κB-driven inflammation and cardiovascular risk. Proc. Natl Acad. Sci. USA 116, 11370–11379 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Stechschulte, L. A. et al. FKBP51 reciprocally regulates GRα and PPARγ activation via the Akt-p38 pathway. Mol. Endocrinol. 28, 1254–1264 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Pertea, M., Kim, D., Pertea, G. M., Leek, J. T. & Salzberg, S. L. Transcript-level expression analysis of RNA-seq experiments with HISAT, StringTie and Ballgown. Nat. Protoc. 11, 1650–1667 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Pertea, M. et al. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat. Biotechnol. 33, 290–295 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Frazee, A. C. et al. Ballgown bridges the gap between transcriptome assembly and expression analysis. Nat. Biotechnol. 33, 243–246 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Warde-Farley, D. et al. The GeneMANIA prediction server: biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 38, W214–W220 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lopes, C. T. et al. Cytoscape Web: an interactive web-based network browser. Bioinformatics 26, 2347–2348 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Taylor, A., Steinberg, J., Andrews, T. S. & Webber, C. GeneNet Toolbox for MATLAB: a flexible platform for the analysis of gene connectivity in biological networks. Bioinformatics 31, 442–444 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the Intramural Research Program of the NHLBI (NIH). We thank J. Mann and R. Shi in the Center of Human Immunology, National Institute of Allergy and Infectious Diseases for the assistance with project management and sample collection, respectively. We thank and acknowledge the assistance of the NHLBI DNA Sequencing and Genomics Core in performing the RNA library sequencing. This trial is registered at ClinicalTrials.gov (NCT02719899, NCT01143454) and NIH Clinical Center blood bank (NCT00001846). This study was funded by the Division of Intramural Research of the NHLBI (NIH; HL005102, HL005199 and HL006199) (M.N.S.).

Author information

Authors and Affiliations

Authors

Contributions

K.H., K.S. and M.N.S. conceived the project. M.N.S. secured the funding. K.H., K.S., R.D.H., K.E.R.S., M.P. and M.N.S. designed the experiments. K.H., K.S., M.J.R., A.N., S.H., K.W., R.D.H., A.S., P.K.D., A.B. and H.L.T. carried out the experiments. K.H., K.S., F.S., P.K.D., A.S., J.C. and H.L.T. analysed the data. K.H., K.S. and M.N.S. wrote the manuscript. M.J.R., J.P.M., K.E.R.S., N.N.M. and M.P. edited the manuscript.

Corresponding author

Correspondence to Michael N. Sack.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks Koji Hase and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: George Caputa.

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

Extended data

Extended Data Fig. 1 Initial analysis of RNA-seq data acquired from the 3 nutritional-load conditions.

a, Individual points (symbols) and means ± SEM (lines) of glucose, insulin and growth hormone levels following 24-hr. fasting and 3-hrs. following a fixed caloric meal. Dot plots represent mean ± SEM with value of each subject (n = 26–28 subjects). The values represent average of duplicates. Two-sided, paired Student’s t-tests. Sera glucose (n = 28), Sera insulin (n = 26), Sera growth hormone (n = 26). b, Table showing number of DE genes (p < 0.05) identified in the indicated comparisons. PBMCs’ RNA from 21 subjects used to generate RNA-seq data. c, Unsupervised principal component analysis (PCA) performed on DE genes (p < 0.05) for indicated comparisons (n = 21 subjects). d, Combined PCA analysis of the top 1000 DE genes (p < 0.05) from all 3 groups. e, Top 10 pathways in which the DE genes (p < 0.05) from the indicated comparisons. The q values (p values adjusted for false discovery rate (FDR)) from the enrichment result are represented by negative log10 scale (x axis). The most significant pathways predominantly align with lymphocyte and T cell differentiation and activation comparison with refed state. f, g, Box and whisker plots show range of fold change of the subset of 114 DE genes (p < 0.05, RNA-seq analysis) that were either downregulated (f) or upregulated (g) to a greater degree following the 24-hr. fast vs. refeeding than baseline (overnight fast) vs. refeeding. The box and whiskers plots show median and upper/lower quartile of the relative gene expression. The whiskers show Turkey distribution and the outlier levels are shown as individual genes (n = 21 subjects). h, Pathway enrichment analysis of 844 DE genes exclusively identified in the 24-hr. fasted versus refed state shown with q values (p value adjusted for FDR) for each pathway represented by negative log 10 scale (x axis). Statistical Source Data of Extended Data Fig. 1.

Source data

Extended Data Fig. 2 Flow cytometry using PBMCs exhibit differential fasting and refeeding cell-surface receptor expression levels.

a, Nutrient-load dependent CD45+ PBMC flow cytometry distribution. Schematic representation of cytometric labelling to distinguish cell type distributions at baseline, following 24-hr. fasting and refeeding (n = 19 subjects). b, Cytometric plots and gating strategies to measure cell surface markers on T helper cells. c-j, Dot and line plots show cell populations of each subject (Wilcoxon two-sided, paired analysis to compare groups, n = 19 subjects). c, Flow plots of activated classical monocytes from representative subject comparing 3 nutrient conditions. The plots show relative cell population frequencies of classical monocytes (CD14highCD16) and median fluorescence intensity (MFI) of activated classical monocytes (HLADR+ in CD14highCD16). d, Representative flow plots showing gating and quantitation of activated CD8+ T cells. Plots show expression of activated CD8+ T cells (CD38+HLADR+ in CD8+) showing significant increases in refed samples compared to baseline and fasting. e, Dot and line plots showing significant blunting in refed samples compared to baseline and fasting in activated DCs (HLADR+ in CD16CD56), myeloid DCs (CD11c+CD123) and plasmacytoid cells (CD11cCD123+). f, Representative flow plots showing gating and quantitation of regulatory T cells and plots show no difference in Treg cells (FOXP3+). g, Follicular helper T cell (CD4+CXCR5+) levels show no difference in three caloric-load conditions. h, Plots showing no difference in activated NK cells (HLADR+ in CD16CD56+, CD16+CD56+, and CD16+CD56). i, Plots showing no difference in immature B cells (CD19+CD20) and mature B cells (CD19+CD20+). j, Quantifying relative cell population frequencies with specific CD8+ T (Tc) markers. Cell populations of Tc1 (CXCR3+CCR6) and Tc17 (CXCR3CCR6+) cells show no change in three caloric-load conditions. The antibody information of BD lyoplate and 18-color panel and gating strategy of flow cytometry is shown in Supplementary Tables 1 and 2 and Supplementary Data 3.

Extended Data Fig. 3 Weighted Gene Co-expression Network Analysis (WGCNA) identifies distinct and coordinate gene expression patterns in the fasting and refed states.

a, Modules of genes with correlated expression patterns were clustered using WGCNA. The modules, with their distinct color designations using the fasted and refed data are shown on the x and y axes. Numerical assignment aligned to module colors represent number of genes per module. The correlated modules from fasting and refed were aligned to determine gene overlap (significance of the overlap determined by Fisher’s exact test where red shading denotes significance - darker shade > significance). b, Representative cell type enrichment analysis (CTen) result of a fasting cluster showing enrichment of CD8+ and CD4+ T cells encoding genes (p-value depicted by red geometric plot extending from the center of the figure towards enriched cell types – representing -log10 Benjamini-Hochberg adjusted P. c, GeneMANIA derived protein-protein interaction (PPI) networks. Significant DE genes fold change information where blue circles represent increased, and red circles decreased expression during fasting compared to refeeding. d, qRT-PCR validation of selected network genes. Bar graph represent mean ± SEM with value of each subject (n = 14 subjects following 3-4 replicates using two-sided paired Student’s t-test). e, Relative FOXO4 RNA expression in CD4+ T cells in response to siRNA (Bar graph represent mean ± SEM, n = 6 health volunteers, two-sided, paired Student’s t-test). f, Representative protein blot show FOXO4 expression in FOXO4-siRNA treated CD4+ T cells from healthy subjects (n = 3), 3 days following TCR activation (+). g-h, Representative immunoblots showing expression levels of (g) FOXO4- and (h) FKBP5-overexpression in CD4+ T cells from healthy subjects (n = 3). Open arrowheads - endogenous protein bands, overexpressed tagged-proteins - solid arrowheads. Source Data Extended Data Fig. 3: Unprocessed immunoblots.

Source data

Extended Data Fig. 4 Fasting and refed modules identified by WGCNA analysis.

*CTen, Cell type enrichment; †PPI, Protein-protein interactions; §TF, Transcription factors.

Extended Data Fig. 5 Evaluating FKBP5 effect on T cell activation.

a, Top 5 variable importance in prediction (VIP) genes by partial least squares discrimination analysis (PLS-DA) of RNA-seq data from three caloric-load conditions. b, Representative protein blots and quantitative changes normalized by Actin of canonical TFs of Th1 (TBX21), Th2 (GATA3), and Th17 (RORC) in FKBP5-overexpression (OE) in CD4+ T cells isolated from healthy volunteers, 3 days following TCR activation. Bar graph represent mean ± SEM with data point of each health volunteer (n = 8 subjects, Wilcoxon two-sided, paired analysis). c-d, CD4+ T cells were isolated from healthy volunteers and transfected with FKBP5 siRNA and scrambled controls. c, Relative FKBP5 RNA expression in CD4+ T cells (Bar graph represent mean ± SEM with normalized value to scrambled control, n = 6 subjects, values represent average of quadruplicates, two-sided, paired Student’s t-test). d, Cytokine release of IFNγ, IL-4 and IL-17 following FKBP5 knockdown (KD) in CD4+ T cells isolated from healthy volunteers, 3 days following TCR activation. The dot and line graphs represent mean value of each subject (n = 6 biologically independent subjects, values represent average of duplicates, two-sided, ratio paired Student’s t-tests). Source Data Extended Data Fig. 5: Unprocessed immunoblots.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2, and Data 1–3

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Han, K., Singh, K., Rodman, M.J. et al. Fasting-induced FOXO4 blunts human CD4+ T helper cell responsiveness. Nat Metab 3, 318–326 (2021). https://doi.org/10.1038/s42255-021-00356-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00356-0

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research