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:

Diet switch pre-vaccination improves immune response and metabolic status in formerly obese mice

Abstract

Metabolic disease is epidemiologically linked to severe complications upon influenza virus infection, thus vaccination is a priority in this high-risk population. Yet, vaccine responses are less effective in these same hosts. Here we examined how the timing of diet switching from a high-fat diet to a control diet affected influenza vaccine efficacy in diet-induced obese mice. Our results demonstrate that the systemic meta-inflammation generated by high-fat diet exposure limited T cell maturation to the memory compartment at the time of vaccination, impacting the recall of effector memory T cells upon viral challenge. This was not improved with a diet switch post-vaccination. However, the metabolic dysfunction of T cells was reversed if weight loss occurred 4 weeks before vaccination, restoring a functional recall response. This corresponded with changes in the systemic obesity-related biomarkers leptin and adiponectin, highlighting the systemic and specific effects of diet on influenza vaccine immunogenicity.

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: Short-term weight loss after vaccination does not improve survival upon viral challenge.
Fig. 2: Viral spread is reduced despite heightened markers of inflammation and blunted seroconversion in formerly obese mice.
Fig. 3: Diet at vaccination defines generation of memory response.
Fig. 4: Obesity-related metabolic biomarkers flux due to diet status.
Fig. 5: Weight loss before vaccination improves survival outcomes.
Fig. 6: HFD-related impairment of T cell functionality is partially rescued by diet switch before vaccination.

Similar content being viewed by others

Data availability

The data supporting the findings of this study are available within the Article and its supplementary materials. Per NIAID contract requirements, the datasets from these studies have been uploaded to the Statistical, Data Management and Coordination Center (SDMCC) where it will be released to the public upon publication. Source data are provided with this paper.

References

  1. Bluher, M. Obesity: global epidemiology and pathogenesis. Nat. Rev. Endocrinol. 15, 288–298 (2019).

    Article  PubMed  Google Scholar 

  2. Honce, R. & Schultz-Cherry, S. Impact of obesity on influenza A virus pathogenesis, immune response, and evolution. Front. Immunol. 10, 1071 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Zlotnikov, N. et al. Infection with the Lyme disease pathogen suppresses innate immunity in mice with diet-induced obesity. Cell. Microbiol. 19, e12689 (2017).

    Article  PubMed  Google Scholar 

  4. Weger-Lucarelli, J. et al. Host nutritional status affects alphavirus virulence, transmission, and evolution. PLoS Pathog. 15, e1008089 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Chuong, C. et al. Nutritional status impacts dengue virus infection in mice. BMC Biol. 18, 106 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Van Kerkhove, M. D. et al. Risk factors for severe outcomes following 2009 influenza A (H1N1) infection: a global pooled analysis. PLoS Med. 8, e1001053 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Dietz, W. & Santos-Burgoa, C. Obesity and its implications for COVID-19 mortality. Obesity 28, 1005 (2020).

    Article  CAS  PubMed  Google Scholar 

  8. Pausé, C., Parker, G. & Gray, L. Resisting the problematisation of fatness in COVID-19: in pursuit of health justice. Int. J. Disaster Risk Reduct. 54, 102021 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Kim, Y. H. et al. Diet-induced obesity dramatically reduces the efficacy of a 2009 pandemic H1N1 vaccine in a mouse model. J. Infect. Dis. 205, 244–251 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Karlsson, E. A. et al. Obesity outweighs protection conferred by adjuvanted influenza vaccination. mBio 7, e01144-6 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Neidich, S. D. et al. Increased risk of influenza among vaccinated adults who are obese. Int. J. Obes. 41, 1324–1330 (2017).

    Article  CAS  Google Scholar 

  12. Honce, R. et al. Obesity-related microenvironment promotes emergence of virulent influenza virus strains. mBio 11, e03341-19 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Klein, S. L. & Flanagan, K. L. Sex differences in immune responses. Nat. Rev. Immunol. 16, 626–638 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Varghese, M., Griffin, C. & Singer, K. The role of sex and sex hormones in regulating obesity-induced inflammation. Adv. Exp. Med. Biol. 1043, 65–86 (2017).

    Article  CAS  PubMed  Google Scholar 

  15. Namkoong, H. et al. Obesity worsens the outcome of influenza virus infection associated with impaired type I interferon induction in mice. Biochem. Biophys. Res. Commun. 513, 405–411 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Baaten, B. J., Li, C.-R. & Bradley, L. M. Multifaceted regulation of T cells by CD44. Commun. Integr. Biol. 3, 508–512 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Kiran, S., Kumar, V., Murphy, E. A., Enos, R. T. & Singh, U. P. High fat diet-induced CD8+ T cells in adipose tissue mediate macrophages to sustain low-grade chronic inflammation. Front. Immunol. 12, 680944 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Myers, M. A. et al. Dynamically linking influenza virus infection kinetics, lung injury, inflammation, and disease severity. eLife 10, e68864 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Rebeles, J. et al. Obesity-induced changes in T-cell metabolism are associated with impaired memory T-cell response to influenza and are not reversed with weight loss. J. Infect. Dis. 219, 1652–1661 (2019).

    Article  CAS  PubMed  Google Scholar 

  20. Srikanthan, K., Feyh, A., Visweshwar, H., Shapiro, J. I. & Sodhi, K. Systematic review of metabolic syndrome biomarkers: a panel for early detection, management, and risk stratification in the West Virginian population. Int. J. Med. Sci. 13, 25–38 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Frühbeck, G., Catalán, V., Rodríguez, A. & Gómez-Ambrosi, J. Adiponectin–leptin ratio: a promising index to estimate adipose tissue dysfunction. Relation with obesity-associated cardiometabolic risk. Adipocyte 7, 57–62 (2018).

    Article  PubMed  Google Scholar 

  22. Pérez-Pérez, A., Sánchez-Jiménez, F., Vilariño-García, T. & Sánchez-Margalet, V. Role of leptin in inflammation and vice versa. Int. J. Mol. Sci. 21, 5887 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  23. van Niekerk, G., Christowitz, C., Conradie, D. & Engelbrecht, A.-M. Insulin as an immunomodulatory hormone. Cytokine Growth Factor Rev. 52, 34–44 (2020).

    Article  PubMed  Google Scholar 

  24. Zhang, A. J. et al. Leptin mediates the pathogenesis of severe 2009 pandemic influenza A(H1N1) infection associated with cytokine dysregulation in mice with diet-induced obesity. J. Infect. Dis. 207, 1270–1280 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. Cho, W. J. et al. Diet-induced obesity reduces the production of influenza vaccine-induced antibodies via impaired macrophage function. Acta Virol. 60, 298–306 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Sheridan, P. A. et al. Obesity is associated with impaired immune response to influenza vaccination in humans. Int. J. Obes. 36, 1072–1077 (2012).

    Article  CAS  Google Scholar 

  27. Paich, H. A. et al. Overweight and obese adult humans have a defective cellular immune response to pandemic H1N1 influenza A virus. Obesity 21, 2377–2386 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Kalil, A. C. & Thomas, P. G. Influenza virus-related critical illness: pathophysiology and epidemiology. Crit. Care 23, 258 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  29. O’Brien, A. et al. Obesity reduces mTORC1 activity in mucosal-associated invariant T cells, driving defective metabolic and functional responses. J. Immunol. 202, 3404–3411 (2019).

    Article  PubMed  Google Scholar 

  30. Andersen, C. J., Murphy, K. E. & Fernandez, M. L. Impact of obesity and metabolic syndrome on immunity. Adv. Nutr. 7, 66–75 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Frasca, D. & Blomberg, B. B. The impact of obesity and metabolic syndrome on vaccination success. Interdiscip. Top. Gerontol. Geriatr. 43, 86–97 (2020).

    Article  PubMed  Google Scholar 

  32. Corrado, M. & Pearce, E. L. Targeting memory T cell metabolism to improve immunity. J. Clin. Invest. 132, e148546 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Xu, H. C. et al. Type I interferon protects antiviral CD8+ T cells from NK cell cytotoxicity. Immunity 40, 949–960 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Le, C. T. et al. PD-1 blockade reverses obesity-mediated T cell priming impairment. Front. Immunol. 11, 590568 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Herati, R. S. et al. PD-1 directed immunotherapy alters Tfh and humoral immune responses to seasonal influenza vaccine. Nat. Immunol. 23, 1183–1192 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Jin, J. et al. Activation of mTORC1 at late endosomes misdirects T cell fate decision in older individuals. Sci. Immunol. 6, eabg0791 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kiernan, K., Nichols, A. G., Alwarawrah, Y. & MacIver, N. J. Effects of T cell leptin signaling on systemic glucose tolerance and T cell responses in obesity. PLoS ONE 18, e0286470 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zani, F. et al. The dietary sweetener sucralose is a negative modulator of T cell-mediated responses. Nature 615, 705–711 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Mauro, C. et al. Obesity-induced metabolic stress leads to biased effector memory CD4+ T cell differentiation via PI3K p110δ-Akt-mediated signals. Cell Metab. 25, 593–609 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Messaoudi, I. et al. Long-lasting effect of obesity on skeletal muscle transcriptome. BMC Genomics 18, 411 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Rossi, E. L. et al. Obesity-associated alterations in inflammation, epigenetics, and mammary tumor growth persist in formerly obese mice. Cancer Prev. Res. 9, 339–348 (2016).

    Article  CAS  Google Scholar 

  42. Frasca, D., Diaz, A., Romero, M. & Blomberg, B. B. Leptin induces immunosenescence in human B cells. Cell. Immunol. 348, 103994 (2020).

    Article  CAS  PubMed  Google Scholar 

  43. Green, W. D. et al. Inflammation and metabolism of influenza-stimulated peripheral blood mononuclear cells from adults with obesity following bariatric surgery. J. Infect. Dis. 227, 92–102 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Sipe, L. M. et al. Response to immune checkpoint blockade improved in pre-clinical model of breast cancer after bariatric surgery. eLife 11, e79143 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pingili, A. K. et al. Immune checkpoint blockade reprograms systemic immune landscape and tumor microenvironment in obesity-associated breast cancer. Cell Rep. 35, 109285 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Pearce, E. L. et al. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature 460, 103–107 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. van der Windt, G. J. W. et al. Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity 36, 68–78 (2012).

    Article  PubMed  Google Scholar 

  48. Hany, M. et al. Impact of bariatric surgery on the effectiveness of serological response after COVID-19 vaccination. Langenbecks Arch. Surg. 407, 2337–2346 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Tylka, T. L. et al. The weight-inclusive versus weight-normative approach to health: evaluating the evidence for prioritizing well-being over weight loss. J. Obes. 2014, 983495 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Aminian, A. et al. Association of weight loss achieved through metabolic surgery with risk and severity of COVID-19 infection. JAMA Surg. https://doi.org/10.1001/jamasurg.2021.6496 (2021).

    Article  PubMed Central  Google Scholar 

  51. Williams, L. M. et al. The development of diet-induced obesity and glucose intolerance in C57BL/6 mice on a high-fat diet consists of distinct phases. PLoS ONE 9, e106159 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Skinner, R. C., Warren, D. C., Lateef, S. N., Benedito, V. A. & Tou, J. C. Apple pomace consumption favorably alters hepatic lipid metabolism in young female Sprague-Dawley rats fed a western diet. Nutrients 10, E1882 (2018).

    Article  Google Scholar 

  53. Reber, A. & Katz, J. Immunological assessment of influenza vaccines and immune correlates of protection. Expert Rev. Vaccines 12, 519–536 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Rowe, T. et al. Detection of antibody to avian influenza A (H5N1) virus in human serum by using a combination of serologic assays. J. Clin. Microbiol. 37, 937–943 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Anderson, K. G. et al. Intravascular staining for discrimination of vascular and tissue leukocytes. Nat. Protoc. 9, 209–222 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. de Brito Monteiro, L., Davanzo, G. G., de Aguiar, C. F. & Moraes-Vieira, P. M. M. Using flow cytometry for mitochondrial assays. MethodsX 7, 100938 (2020).

    Article  Google Scholar 

  57. Pengcheng, L., Lu, J. L. & Koestler, D. pwr2: Power and Sample Size Analysis for One-Way and Two-Way ANOVA Models https://cran.r-project.org/web/packages/pwr2/pwr2.pdf (2017).

  58. Ben-Shachar, M., Lüdecke, D. & Makowski, D. Effectsize: estimation of effect size indices and standardized parameters. J. Open Source Softw. 5, 2815 (2020).

    Article  Google Scholar 

  59. Meliopoulos, V. A. et al. An epithelial integrin regulates the amplitude of protective lung interferon responses against multiple respiratory pathogens. PLoS Pathog. 12, e1005804 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank P. Freiden, K. Wiggins and B. Clark for expert technical assistance and scientific discussions, and P. Vogel for his pathological expertise (all of St Jude Children’s Research Hospital); D. Warren, C. Oudomvilay and C. Roberts (University of the Ozarks) for assisting in sample and assay preparation; the National Institutes of Health (NIH) Tetramer Core Facility (contract number 75N93020D00005) for providing the Influenza A NP tetramer; and the St Jude Animal Resource Center and Veterinary Pathology Core for helpful experimental assistance. This work was supported by ALSAC (American Lebanese Syrian Associated Charities), the National Institute of Allergy and Infectious Diseases (NIAID) at NIH under Department of Health and Human Services contract HHSN27220140006C for the St Jude Center of Excellence for Influenza Research and Surveillance (CEIRS), by the NIAID Collaborative Influenza Vaccine Innovation Centers (CIVIC) contracts 75N93019C00052 and 75N93021C00016 for the St Jude Center of Excellence for Influenza Research and Response (CEIRR) to S.S.-C. and P.G.T., NIAID NIH Award Number F31AI161986 to A.V.-P. and NIAID R01 AI140766-03 to S.S.-C. A.V.-P. and B.A.W. were supported by the St Jude Graduate School for Biomedical Sciences and A.H.M. was supported by NIAID T32AI106700-07. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. 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

R.H., A.V.-P. and S.S.-C. conceptualized the project. R.H., A.V.-P., S.S.-C. and P.G.T. developed the methodology. R.H., A.V.-P. and S.S.-C. performed validation. R.H., A.V.-P. and R.C.S. conducted formal analysis. R.H., A.V.-P., B.L., S.C., A.H.M., B.A.W., P.H.B., V.H., B.S., E.K.R., L.-A.V.d.V. and R.C.S. conducted investigations. R.H., A.V.-P., B.L., B.S., L.-A.V.d.V., P.G.T. and S.S.-C. procured resources. R.H. and A.V.-P. curated data. R.H., A.V.-P. and R.C.S. wrote the original draft. All authors reviewed and edited the paper. R.H. and A.V.-P. performed visualization. S.S.-C. supervised the project. R.H., A.V.-P and S.S.-C. administered the project. M.A.M., P.G.T. and S.S.-C. acquired funding.

Corresponding author

Correspondence to Stacey Schultz-Cherry.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Reviewer Recognition

Nature Microbiology thanks Kai Dallmeier, Amelia Pinto 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 Long-term weight loss after vaccination does not improve survival upon viral challenge.

(a) Timeline of diet administration, vaccination, and challenge for 12-week long-term diet protocol with diet composition breakdown by percent fat/protein/carbohydrate. (b) Weights of mice through short-term diet protocol and at (c) vaccination and challenge compared via one-way ANOVA (vaccination always lean p < 0.0001; H1N1 always lean p < 0.0001, mice that were formerly obese p < 0.0001). (d–f) Morbidity and mortality in vaccinated mice compared to mock vaccinated mice with n = 10 mice/group. (d) Weight curves post-challenge with statistical comparisons made via mixed-effects model. Grey dotted line indicates a 0% deviation from weight at baseline. (e) Clinical scores post-challenge with statistical comparisons made using a two-way ANOVA. (f) Survival post-challenge with statistical comparisons made using Mantel-Cox log-rank analysis. (g) Hemagglutination inhibition titers at indicated timepoints with n = 4 mice per diet group. Data in (be) are representative of two independent experiments and are graphed as means ± standard error and in (f) as surviving proportions with censored or event animals indicated by their respective symbols and in (g) as geometric means ± standard deviation with x indicating no mice surviving to time point. Statistical comparisons test between mice with obesity and indicated diet group (n = 10 mice/group) in (b, c) and between mock and vaccinated animals within a single diet group in (d–f).

Source data

Extended Data Fig. 2 Weight gain post-vaccination has modest impacts on survival at challenge.

(a) Timeline of diet administration, vaccination, and challenge for 4-week short-term or 12-week long-term reverse diet switch protocol with diet composition breakdown by percent fat/protein/carbohydrate. (b) Weights of formerly lean mice on short- and long-term reverse diet switch protocols at (c) vaccination and challenge compared via one-way ANOVA (short-term p < 0.0001, long-term p < 0.0010). Grey dotted lines represent the mean weight of mice with obesity at corresponding timepoints. (d-e) Morbidity and mortality in vaccinated mice compared to mock vaccinated mice with n = 10 mice/group for (d) short-term and (e) long-term reverse diet switch protocols. Weight curves post-challenge with statistical comparisons made via mixed-effects model. Grey dotted line indicates a 0% deviation from weight at baseline. Clinical scores post-challenge with statistical comparisons made using a two-way ANOVA. Survival post-challenge with statistical comparisons made using Mantel-Cox log-rank analysis. (f) Effector antibody levels in sera collected and quantified by hemagglutination inhibition (HAI). Grey dotted line represents lower limit of detection. Mice assayed longitudinally with n = 5 per group except for the day 21 post-influenza challenge timepoint in which only surviving mice were assayed. Data are representative of three independent experiments and are graphed as means ± standard error and, for survival graphs, as surviving proportions with censored or event animals indicated by their respective symbols. Statistical comparisons test between mice with obesity and indicated diet groups (n = 10 mice/group) in (b, c) and between mock and vaccinated animals within a single diet group in (d, e). These studies were done concurrently with data shown in Fig. 1 and Extended Data Fig. 1.

Source data

Extended Data Fig. 3 Similar responses to challenge post-diet switch in female mice.

(a) Timeline of diet administration, vaccination, and challenge in short-term diet and weight gain protocols with diet composition breakdown by percent fat/protein/carbohydrate. (b) Weights of female mice on diet protocols at (c) vaccination and challenge compared via one-way ANOVA (vaccination always lean p < 0.0001, formerly lean p < 0.0001; H1N1 always lean p < 0.0001, mice that were formerly obese p < 0.0001, formerly lean p < 0.0001). (d–f) Morbidity and mortality in vaccinated mice compared to mock vaccinated mice. (d) Weight curves post-challenge with statistical comparisons made via mixed-effects model. Grey dotted lines represent the mean weight of mice with obesity at corresponding timepoints. (e) Clinical scores post-challenge with statistical comparisons made using a two-way ANOVA. (f) Survival post-challenge with statistical comparisons made using Mantel-Cox log-rank analysis. Data in (b-e) are represented as means ± standard error and in (f) as surviving proportions with censored or event animals indicated by their respective symbols. (g) Effector antibody levels in sera collected and quantified by hemagglutination inhibition (HAI). Grey dotted line represents lower limit of detection. Mice assayed longitudinally with n = 5 per group except for the day 21 post-influenza challenge timepoint in which only surviving mice were assayed. Data displayed as geometric means ± standard deviation with x indicating no mice surviving to time point. Statistical comparisons test between mice with obesity and indicated diet group (n = 10 mice/group) in (b, c, g) and between mock and vaccinated animals within a single diet group in (d-f).

Source data

Extended Data Fig. 4 Frequency plots of primary responses to vaccination prior to diet switch.

Spleens were harvested and homogenized into single cell suspensions and 1 × 106 cells were stained for CD4+ T cells, CD8+ T cells, and B220+ B cells at 21 days post-vaccination and quantified via flow-cytometry. Frequency of (a) CD4+ (CD45i.v. CD45+ CD3+ CD4+, p = 0.0021), (b) CD4+ effector memory (TEFM, CD45i.v. CD45+ CD3+ CD4+ CD62L+ CD44,) and (c) CD4+ central memory (TCM, CD45i.v. CD3+ CD4+ CD62L+ CD44+, p = 0.0007) cells. Frequency of (d) CD8+ (CD45i.v. CD45+ CD3+ CD8+), (e) CD8+ TEFM (CD45i.v. CD45+ CD3+ CD8+ CD62L+ CD44, p = 0.004), and (g) CD8+ TCM (CD45i.v. CD3+ CD8+ CD62L+ CD44+) cells. Frequency of (g) B cells (CD3 B220+). Data are represented as boxplots as frequency of live singlets and analyzed with a two-tailed t-test used to compare groups. Note that samples were collected prior to diet switch. Corresponds to Fig. 4.

Source data

Extended Data Fig. 5 Recall responses are altered by HFD exposure at time of vaccination.

Lungs were harvested and homogenized into single cell suspensions and 1 × 106 cells were stained for tissue resident and circulating CD4+ and CD8+ T cells at 7 days post-challenge and quantified via flow-cytometry. (a) Absolute number or (b) frequency of tissue resident CD4+ T (CD45i.v. CD45+ CD3+, CD4+), T effector memory (TEFM, CD45i.v. CD45+ CD3+ CD4+ CD62L+ CD44) and T central memory (TCM, CD45i.v. CD45+ CD3+ CD4+ CD62L+ CD44+) cells or CD8+ T (CD45i.v. CD45+ CD3+ CD8+), TEFM (CD45i.v. CD45+ CD3+ CD4+ CD62L+ CD44), and TCM (CD45i.v. CD45+ CD3+ CD4+ CD62L+ CD44+) cells. (c) Absolute number or (d) frequency of circulating CD4+ T (CD45i.v.+ CD45 CD3+ CD4+), T effector memory (TEFM, CD45i.v.+ CD45 CD3+ CD4+ CD62L+ CD44) and T central memory (TCM, CD45i.v.+ CD45 CD3+ CD4+ CD62L+ CD44+) cells or CD8+ T (CD45i.v.+ CD45 CD3+ CD8+), TEFM (CD45i.v.+ CD45 CD3+ CD4+ CD62L+ CD44), and TCM (CD45i.v.+ CD45 CD3+ CD4+ CD62L+ CD44+) cells. Data include mice with obesity, always lean, and mice that were formerly obese (n = 2, n = 6, n = 4 for each group, respectively) and graphed as boxplots with comparisons made between always lean and formerly obese groups via one-way ANOVA. Frequency values are calculated as compared to live singlets.

Source data

Extended Data Fig. 6 Increased survival in phenotypic obese and mice that were formerly obese with 8-week HFD exposure.

(a) Timeline of diet administration, vaccination, and challenge in half-term diet switch studies. (b) Weights of mice through a half-term diet switch studies (n = 5–10 mice/group) at (c) vaccination and challenge (vaccination always lean p < 0.0001; H1N1 always lean p < 0.0001, formerly obese p < 0.0001). (d–f) Morbidity and mortality upon H1N1 virus challenge with n = 5–10 mice/group. (d) Weight curves with grey dotted line indicate a 0% deviation from baseline weight, (e) clinical scores and (f) survival. (g) Hemagglutination inhibition titers at indicated timepoints. The grey dotted line indicates the lower limit of detection. Data are represented in (b-e) as means ± standard error, in (f) as surviving proportions with censored or event animals indicated by their respective symbols, and in (g) as geometric mean ± standard deviation with x indicating no mice surviving to time point. Statistical comparisons test between mice with obesity and indicated diet groups (n = 10 mice/group) in (c).

Source data

Extended Data Fig. 7 Phenotyping of T cell subsets in pre-vaccination diet switch cohorts.

Mean fluorescent intensity (MFI) of splenic CD98, MitoTracker Red (MTR CMXRos) and MitoTracker Green (MTG) in (a) CD4+ effector memory (TEFM) T cells, (b) CD4+ central memory (TCM) T cells, (c) CD8+ TEFM, and (d) CD8+ TCM cells. Data include mice with obesity, always lean, and mice that were formerly obese (n = 7, n = 7, n = 8 for each group, respectively) and graphed as boxplots with comparisons made between via one-way ANOVA with CD4+ TEFM MTG formerly obese p = 0.0148; and CD8+ TEFM cells MTR formerly obese p = 0.0172, MTG formerly obese p = 0.0034.

Source data

Extended Data Fig. 8 Improved elicitation of memory T cell responses upon weight loss pre-vaccination.

Lungs were harvested and homogenized into single cell suspensions and 1 × 106 cells were stained for CD4+ and CD8+ T cells at 7 days post-challenge and quantified via flow-cytometry. (a) Absolute number or (b) frequency of tissue resident CD4+ T (CD45i.v. CD45+ CD3+ CD4+), T effector memory (TEFM, CD45i.v. CD45+ CD3+ CD4+ CD62L+ CD44) and T central memory (TCM, CD45i.v. CD45+ CD3+ CD4+ CD62L+ CD44+) cells or CD8+ T (CD45i.v. CD45+ CD3+ CD8+, always lean p = 0.0473), TEFM (CD45i.v. CD45+ CD3+ CD4+ CD62L+ CD44), and TCM (CD45i.v. CD45+ CD3+ CD4+ CD62L+ CD44+, formerly obese p = 0.0171) cells. (c) Absolute number or (d) frequency of circulating CD4+ T (CD45i.v.+ CD45 CD3+ CD4+, always lean p = 0.0253), T effector memory (TEFM, CD45i.v.+ CD45 CD3+ CD4+ CD62L+ CD44, always lean p = 0.0069) and T central memory (TCM, CD45i.v.+ CD45 CD3+ CD4+ CD62L+ CD44+, always lean p = 0.0028, p = 0.0004; formerly obese p < 0.0001) cells or CD8+ T (CD45i.v.+ CD45 CD3+ CD8+, always lean p = 0.0038, formerly obese p = 0.0292), TEFM (CD45i.v.+ CD45 CD3+ CD4+ CD62L+ CD44, always lean p = 0.0017), and TCM (CD45i.v.+ CD45 CD3+ CD4+ CD62L+ CD44+, always lean p = 0.0045, p = 0.0019; formerly obese p < 0.0001) cells. Data include always obese, always lean, and mice that were formerly obese (n = 8, n = 7, n = 7 for each group, respectively) and graphed in as boxplots with comparisons made between diet groups via one-way ANOVA. Frequency values are calculated as compared to live singlets.

Source data

Extended Data Fig. 9 Assessment of T-cell surface phenotype in lungs upon distinct diet regimens post-vaccination and post-influenza challenge.

At 4 weeks post-vaccination, mice were challenged with H1N1 virus. Lungs were harvested and homogenized into single-cell suspensions and 1 × 106 cells were stained for CD4+ and CD8+ T cells. (a) Mean fluorescent intensity (MFI) of CD98+ staining on CD4+ T, T effector memory (TEFM), and T central memory (TCM, always lean p = 0.0285) cells. (b) MFI of CD98+ staining on CD8+ TEFM (always lean p = 0.0391, formerly obese p = 0.0391) and TCM cells (always lean p = 0.0504, formerly obese p = 0.0280). (c) Frequency of PD1+ CD4+ T cells (CD44+ CD4+ CD98+, always lean p = 0.0015). MitoTracker Green (MTG) and MitoTracker Red (MTR CMXRos) staining in (d) CD4+ T cells, (e) CD4+ TEFM cells, (f) CD4+ TCM cells, (g) CD8+ TEFM cells (formerly obese MTG p = 0.0428, MTR p = 0.0025), (g) CD8+ TCM (always lean MTG p = 0.0121, formerly obese MTR p < 0.0001) cells. Data include mice with obesity, always lean, and mice that were formerly obese (n = 4, n = 8, n = 10 for each group, respectively) and graphed as boxplots with comparisons made between diet groups via one-way ANOVA. Corresponds to Fig. 6.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2.

Reporting Summary

Source data

Source Data Fig. 1

Raw data and statistical test data.

Source Data Fig. 2

Raw data and statistical test data.

Source Data Fig. 3

Raw data and statistical test data.

Source Data Fig. 4

Raw data and statistical test data.

Source Data Fig. 5

Raw data and statistical test data.

Source Data Fig. 6

Raw data and statistical test data.

Source Data Extended Data Fig. 1

Raw data and statistical test data.

Source Data Extended Data Fig. 2

Raw data and statistical test data.

Source Data Extended Data Fig. 3

Raw data and statistical test data.

Source Data Extended Data Fig. 4

Raw data and statistical test data.

Source Data Extended Data Fig. 5

Raw data and statistical test data.

Source Data Extended Data Fig. 6

Raw data and statistical test data.

Source Data Extended Data Fig. 7

Raw data and statistical test data.

Source Data Extended Data Fig. 8

Raw data and statistical test data.

Source Data Extended Data Fig. 9

Raw data and statistical test data.

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

Honce, R., Vazquez-Pagan, A., Livingston, B. et al. Diet switch pre-vaccination improves immune response and metabolic status in formerly obese mice. Nat Microbiol (2024). https://doi.org/10.1038/s41564-024-01677-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41564-024-01677-y

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: Microbiology