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:

PD1 blockade improves survival and CD8+ cytotoxic capacity, without increasing inflammation, during normal microbial experience in old mice

Abstract

By 2030, individuals 65 years of age or older will make up approximately 20% of the world’s population1. Older individuals are at the highest risk for mortality from infections, largely due to the pro-inflammatory, dysfunctional immune response, which is collectively known as immunosenescence2. During aging, CD8+ T cells acquire an exhausted phenotype, including increased expression of inhibitory receptors, such as programmed cell death 1 (PD1), a decline in effector function and elevated expression of inflammatory factors3,4,5,6,7. PD1 reduces T cell receptor activity via SHP2-dependent dephosphorylation of multiple pathways; accordingly, inhibiting PD1 activity through monoclonal antibodies increases CD8+ T cell effector response in young mice8,9,10,11. Attempts to improve CD8+ T cell responses by blocking inhibitory receptors are attractive; however, they can lead to adverse immune events due to overamplification of T cell receptor signaling and T cell activation12,13. Here we investigated the effect of monoclonal anti-PD1 immunotherapy during normal microbial experience, otherwise known as exposure to dirty mice, to determine whether it either improves exhausted CD8+ T cell responses in old mice or leads to a heightened inflammatory response and increased mortality.

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: Inflammation is increased in old mice after NME.
Fig. 2: CD8+ T cells bearing markers of exhaustion are expanded in old mice after NME.
Fig. 3: PD1 checkpoint blockade improves survival to NME in old mice.
Fig. 4: PD1 checkpoint blockade improves CD8+ T cell cytotoxic capacity.

Similar content being viewed by others

Data availability

All genomics and sequencing data will be publicly available at the Gene Expression Omnibus (GEO) on the date of publication under accession number GSE250165. All other data supporting the findings of this study are available as source data files or from the corresponding author upon reasonable request. Source data are provided with this paper.

References

  1. World Health Organization. Decade of healthy ageing: baseline report. https://www.who.int/publications/i/item/9789240017900 (2021).

  2. Nikolich-Žugich, J. The twilight of immunity: emerging concepts in aging of the immune system. Nat. Immunol. 19, 10–19 (2018).

    Article  PubMed  Google Scholar 

  3. Lee, K. A. et al. Characterization of age-associated exhausted CD8+ T cells defined by increased expression of Tim-3 and PD-1. Aging Cell 15, 291–300 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Decman, V. et al. Defective CD8 T cell responses in aged mice are due to quantitative and qualitative changes in virus-specific precursors. J. Immunol. 188, 1933–1941 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Quinn, K. M. et al. Age-related decline in primary CD8+ T cell responses is associated with the development of senescence in virtual memory CD8+ T cells. Cell Rep. 23, 3512–3524 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Wherry, E. J. & Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 15, 486–499 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wherry, E. J. et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27, 670–684 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Sharpe, A. H. & Pauken, K. E. The diverse functions of the PD1 inhibitory pathway. Nat. Rev. Immunol. 18, 153–167 (2018).

    Article  CAS  PubMed  Google Scholar 

  9. Parry, R. V. et al. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol. Cell. Biol. 25, 9543–9553 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yokosuka, T. et al. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J. Exp. Med. 209, 1201–1217 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hui, E. et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 355, 1428–1433 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Martins, F. et al. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat. Rev. Clin. Oncol. 16, 563–580 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Wang, P. F. et al. Immune-related adverse events associated with anti-PD-1/PD-L1 treatment for malignancies: a meta-analysis. Front. Pharmacol. 8, 730 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Hamilton, S. E. et al. New insights into the immune system using dirty mice. J. Immunol. 205, 3–11 (2020).

    Article  CAS  PubMed  Google Scholar 

  15. Beura, L. K. et al. Normalizing the environment recapitulates adult human immune traits in laboratory mice. Nature 532, 512–516 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Huggins, M. A. et al. Microbial exposure enhances immunity to pathogens recognized by TLR2 but increases susceptibility to cytokine storm through TLR4 sensitization. Cell Rep. 28, 1729–1743 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Camell, C. D. et al. Senolytics reduce coronavirus-related mortality in old mice. Science 373, eabe4832 (2020).

    Article  Google Scholar 

  18. Caputa, G. et al. Intracellular infection and immune system cues rewire adipocytes to acquire immune function. Cell Metab. 34, 747–760 (2022).

    Article  CAS  PubMed  Google Scholar 

  19. Shen, B. et al. Proteomic and metabolomic characterization of COVID-19 patient sera. Cell 182, 59–72 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Frasca, D., Blomberg, B. B. & Paganelli, R. Aging, obesity, and inflammatory age-related diseases. Front. Immunol. 8, 1745 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Chouchani, E. T. & Kajimura, S. Metabolic adaptation and maladaptation in adipose tissue. Nat Metab. 1, 189–200 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Anderson, K. G. et al. Cutting edge: intravascular staining redefines lung CD8 T cell responses. J. Immunol. 189, 2702–2706 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Tomayko, M. M., Steinel, N. C., Anderson, S. M. & Shlomchik, M. J. Cutting edge: hierarchy of maturity of murine memory B cell subsets. J. Immunol. 185, 7146–7150 (2010).

    Article  CAS  PubMed  Google Scholar 

  24. Blackburn, S. D. et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat. Immunol. 10, 29–37 (2009).

    Article  CAS  PubMed  Google Scholar 

  25. Barber, D. L. et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439, 682–687 (2006).

    Article  CAS  PubMed  Google Scholar 

  26. Patsoukis, N., Wang, Q., Strauss, L. & Boussiotis, V. A. Revisiting the PD-1 pathway. Sci. Adv. 6, eabd2712 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Mogilenko, D. A. et al. Comprehensive profiling of an aging immune system reveals clonal GZMK+ CD8+ T cells as conserved hallmark of inflammaging. Immunity 54, 99–115 (2021).

    Article  CAS  PubMed  Google Scholar 

  28. Beltra, J.-C. et al. Developmental relationships of four exhausted CD8+ T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity 52, 825–841 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hudson, W. H. et al. Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1+ stem-like CD8+ T cells during chronic infection. Immunity 51, 1043–1058 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Renkema, K. R. et al. KLRG1+ memory CD8 T cells combine properties of short-lived effectors and long-lived memory. J. Immunol. 205, 1059–1069 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Blackburn, S. D., Shin, H., Freeman, G. J. & Wherry, E. J. Selective expansion of a subset of exhausted CD8 T cells by αPD-L1 blockade. Proc. Natl Acad. Sci. USA 105, 15016–15021 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Iwai, Y. et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc. Natl Acad. Sci. USA 99, 12293–12297 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Polesso, F. et al. PD-1-specific ‘blocking’ antibodies that deplete PD-1+ T cells present an inconvenient variable in preclinical immunotherapy experiments. Eur. J. Immunol. 51, 1473–1481 (2021).

    Article  CAS  PubMed  Google Scholar 

  35. Martins, C. et al. Distinct antibody clones detect PD-1 checkpoint expression and block PD-L1 interactions on live murine melanoma cells. Sci. Rep. 12, 12491 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Wang, T. W. et al. Blocking PD-L1–PD-1 improves senescence surveillance and ageing phenotypes. Nature 611, 358–364 (2022).

    Article  CAS  PubMed  Google Scholar 

  37. Utzschneider, D. T. et al. T cell factor 1-expressing memory-like CD8+ T cells sustain the immune response to chronic viral infections. Immunity 45, 415–427 (2016).

    Article  CAS  PubMed  Google Scholar 

  38. He, R. et al. Follicular CXCR5-expressing CD8+ T cells curtail chronic viral infection. Nature 537, 412–428 (2016).

    Article  CAS  PubMed  Google Scholar 

  39. Lages, C. S., Lewkowich, I., Sproles, A., Wills-Karp, M. & Chougnet, C. Partial restoration of T-cell function in aged mice by in vitro blockade of the PD-1/ PD-L1 pathway. Aging Cell 9, 785–798 (2010).

    Article  CAS  PubMed  Google Scholar 

  40. Elias, R. et al. Efficacy of PD-1 & PD-L1 inhibitors in older adults: a meta-analysis. J. Immunother. Cancer 6, 26 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Nishijima, T. F., Muss, H. B., Shachar, S. S. & Moschos, S. J. Comparison of efficacy of immune checkpoint inhibitors (ICIs) between younger and older patients: a systematic review and meta-analysis. Cancer Treat. Rev. 45, 30–37 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Russell, J. H. & Ley, T. J. Lymphocyte-mediated cytotoxicity. Annu. Rev. Immunol. 20, 323–370 (2002).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

BioRender was used to generate schematics under the following agreement numbers: KB26KBLQB7, MN26KBLQDJ, LB26KBLQFA, FT26KBLQH9, DG26KBLQIS, EY26KBLQMN, AY26KBLQOD, JC26KBLQQ6, VS26KBLQRO, XG26KBLQTV and FT26KBLQVN. We also acknowledge and thank the University of Minnesota Flow Cytometry Resource, the University of Minnesota Genomic Center and J. E. Abrahante Lloréns for the use of their machines and expertise. This work was supported by National Institutes of Health grants R00AG058800 (C.D.C.), R21AG078638 (C.D.C.), R01AG079913-01 (C.D.C.) and R01AI155468 (S.E.H.) and T32AG029796 (K.J.V.D.); the Fesler-Lampert Chair in Aging Studies (C.D.C.); the Glenn Foundation for Medical Research/AFAR Grants for Junior Faculty (C.D.C.); the McKnight Land-Grant (C.D.C.); the Irene Diamond Fund/AFAR Postdoctoral Transition Award (M.J.Y.); the Medical Discovery Team on the Biology of Aging (C.D.C.); and a Longevity Impetus Grant from the Norn Group (K.J.V.D.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

K.J.V.D. carried out most experiments. M.A.H., M.J.Y., S.H.C., M.P., D.M.S. and C.D.C. assisted with experiments. C.S-P. performed bioinformatics analysis. S.E.H. provided insight into experimental design and interpretation of data. K.J.V.D. and C.D.C. conceived the project, analyzed the data and wrote the paper. All authors read, edited and approved the final version of the paper.

Corresponding author

Correspondence to Christina D. Camell.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Nicole La Gruta, Janet Lord and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Inflammation is increased in old mice after NME.

(a-b) Experiment 2. a. Enrichment plots of selected upregulated pathways (IL6 JAK STAT3 signaling, Inflammatory response, TNFA signaling via NFKB and Interferon Gamma Response). b. Enrichment plots of selected downregulated pathways (Bile acid metabolism and Fatty Acid Metabolism). (c) Experiment 4. c. Survival curve measured for 30 days after initiation of NME. (1-month NCD n = 8, 1-month HFD n = 8). P-value: P > 0.9999(ns). (d) Experiment 5. d. Survival curve measured for 30 days after initiation of NME. (5-month NCD n = 8, 5-month HFD n = 8). P-value: P > 0.9999(ns). (e) Experiment 6. e. Survival curve measured for 30 days after initiation of NME. (6-month NCD n = 8, 6-month HFD n = 5). P-value: P = 0.7519(ns). Statistical significance was determined with Log-rank (Mantel Cox) test (c-e). All data are presented as Means ± SEM. All p-values can be found in source data.

Source data

Extended Data Fig. 2 CD8+ T-cells bearing markers of exhaustion are expanded in old mice after NME.

(a-e,i-j) Experiment 2. a. CD45+ Live and IV gating in liver, lung, spleen, and mLN. b. IV cells as a percentage of CD45+ Live cells in the lung, spleen, and mLN. P-values: Lung: young SPF vs old SPF P = 4.766E-06(****), young SPF vs old NME P = 0.0003(***), old SPF vs young NME P = 4.569E-07(****), young NME vs old NME P = 1.5592E-05(****). c. IV leukocyte subsets in mLN. d. CD73+ IgM cells as a percentage of IV- B220+ cells in the liver, lung, mLN, and spleen. P-values: Liver: young SPF vs old NME P = 0.0175(*), old SPF vs old NME P = 0.0199(*), young NME vs old NME P = 3.4536E-06(****); Lung: young SPF vs old NME P = 0.0216(*), old SPF vs old NME P = 0.0499(*), young NME vs old NME P = 9.0894E-05(****); Spleen: young SPF vs old NME P = 0.0001(***), old SPF vs old NME P = 0.0005(***), young NME vs old NME P = 1.2504E-09(****). e. Memory cell subsets as a frequency of IV- CD8+ PD1+ cells. f. Representative contour plots of PD1 spleen expression by IV CD45+ CD4+ CD44+ cells. Numbers are average frequencies. (g-h) Experiment 9. g. PD1+ cells as a percentage of NK cells or h. NKT cells in the liver, lung, mLN, and spleen. P-values: Liver: old SPF vs young NME P = 2.750E-06(****), young NME vs old NME P = 0.0015(**). i. CXCR5+ cells or j. KLRG1+ cells as a percentage of CD8+ IV CD45+ live cells in the liver, lung, mLN, and spleen. P-values: Lung: young NME vs old NME P = 0.0018(**); mLN: young SPF vs young NME P = 0.0440(*). Statistical significance was determined with two-way ANOVA with Tukey’s (b,d,i,j) or Šidák multiple comparisons test (c,g,h). All data are presented as Means ± SEM. All p-values can be found in source data.

Source data

Extended Data Fig. 3 PD1 checkpoint blockade improves survival to NME in old mice.

(a) Experiments 9 and 11. a. Anti-PD1 intervention and prevention day 7 experimental schematic. (b-e) Experiment 17. b. Young anti-PD1 prevention survival experimental schematic. c. Survival curve measured for 30 days after initiation of NME. P-value: P = 0.5064(ns). d. GzmB+ cells as a percentage of CD8+ CD44+ PD1+ cells in young isotype or prevention anti-PD1 treated mice exposed to NME through cohousing. P-value: P = 0.9130(ns). e. Quantification of GzmB+ cells as a percentage of CD8+ CD44+ PD1 cells in young isotype or prevention anti-PD1 treated mice exposed to NME through cohousing. P-value: P = 0.5179(ns). Statistical significance was determined with Log-rank (Mantel Cox) test (c) or unpaired two-tailed t-test with 95% confidence (d-e). All data are presented as Means ± SEM. All p-values can be found in source data.

Source data

Extended Data Fig. 4 PD1 checkpoint blockade improves CD8+ T-cell cytotoxic capacity.

(a) Experiment 12. a. Representative flow cytometry contour plots in spleen and liver on day 7 of NME indicating depletion efficacy. (b-c) Experiment 13. b. IFNγ and TNFα expression as a percentage of CD8+ CD44+ PD1+ cells or c. PD1 cells after stimulation with PMA+ ionomycin. (d) Experiment 15. d. GzmB+ cells as a frequency of CD8+ PD1+ cells from blood. P-value: old anti-PD1 lived P = 0.0047(**). (e-g) Experiment 14. e. Anti-PD1 SPF day 7 experimental schematic. f. GzmB+ cells as a percentage of CD8+ CD44+ PD1+ cells from old SPF mice. P-value: P = 0.9081(ns). g. GzmB+ cells as a percentage of CD8+ CD44+ PD1 cells from old SPF mice. P-value: P = 0.4928(ns). (h-i) Experiment 18. h. GzmB+ cells as a percentage of IV CD8+ CD44+ CD62L PD1+ cells in young and old mice after fisetin treatment or vehicle control. P-values: Liver: young vehicle vs old fisetin P = 0.0014(**), young fisetin vs old fisetin P = 0.0358(*), old vehicle vs old fisetin P = 0.4195(ns); spleen: old vehicle vs old fisetin P = 0.9922(ns). i. GzmB+ cells as a percentage of IV+ CD8+ CD44+ CD62L PD1+cells in young and old mice after fisetin treatment or vehicle control. P-values: Liver: young vehicle vs old fisetin P = 0.0432(*), young fisetin vs old vehicle P = 0.0147(*), young fisetin vs old fisetin P = 0.0094(**), old vehicle vs old fisetin P = 0.9974(ns); spleen: old vehicle vs old fisetin P = 0.7303(ns). Statistical significance was determined with multiple unpaired two-tailed t-tests with 95% confidence (b-c) or RM two-way ANOVA with Šidák multiple comparisons test (d) or unpaired two-tailed t-test with 95% confidence (f-g) or two-way ANOVA with Tukey’s multiple comparisons test (h-i). All data are presented as Means ± SEM. All p-values can be found in source data.

Source data

Extended Data Fig. 5 Representative gating strategies 1.

a. Representative flow cytometry pseudocolor plots for flow cytometry data shown in Fig. 2 and Extended Data Fig. 2.

Extended Data Fig. 6 Representative gating strategies 2.

a. Representative flow cytometry pseudocolor plots for flow cytometry data shown in Figs. 24 and Extended Data Figs. 24.

Supplementary information

Supplementary Information

Supplementary Table 1.

Reporting Summary

Supplementary Table 2

Liver GSEA upregulated genes.

Supplementary Table 3

Liver GSEA downregulated genes.

Source data

Source Data Fig. 1

Raw data, statistics and P values for Fig. 1.

Source Data Fig. 2

Raw data, statistics and P values for Fig. 2.

Source Data Fig. 3

Raw data, statistics and P values for Fig. 3.

Source Data Fig. 4

Raw data, statistics and P values for Fig. 4.

Source Data Extended Data Fig. 1

Raw data, statistics and P values for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Raw data, statistics and P values for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Raw data, statistics and P values for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Raw data, statistics and P values for Extended Data Fig. 4.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dahlquist, K.J.V., Huggins, M.A., Yousefzadeh, M.J. et al. PD1 blockade improves survival and CD8+ cytotoxic capacity, without increasing inflammation, during normal microbial experience in old mice. Nat Aging (2024). https://doi.org/10.1038/s43587-024-00620-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s43587-024-00620-4

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