Modulation of immune responses to vaccination by the microbiota: implications and potential mechanisms

David J Lynn, Saoirse C Benson, Miriam A Lynn, Bali Pulendran, David J Lynn, Saoirse C Benson, Miriam A Lynn, Bali Pulendran

Abstract

The need for highly effective vaccines that induce robust and long-lasting immunity has never been more apparent. However, for reasons that are still poorly understood, immune responses to vaccination are highly variable between different individuals and different populations. Furthermore, vaccine immunogenicity is frequently suboptimal in the very populations who are at most risk from infectious disease, including infants, the elderly, and those living in low-income and middle-income countries. Although many factors have the potential to influence vaccine immunogenicity and therefore vaccine effectiveness, increasing evidence from clinical studies and animal models now suggests that the composition and function of the gut microbiota are crucial factors modulating immune responses to vaccination. In this Review, we synthesize this evidence, discuss the immunological mechanisms that potentially mediate these effects and consider the potential of microbiota-targeted interventions to optimize vaccine effectiveness.

Conflict of interest statement

D.J.L. receives funding from GlaxoSmithKline as part of a collaborative research agreement in this area. B.P. serves on the External Immunology Network of GlaxoSmithKline and is on the scientific advisory board of Medicago. S.C.B. and M.A.L. declare no competing interests.

© 2021. Springer Nature Limited.

Figures

Fig. 1. Differences in the composition and…
Fig. 1. Differences in the composition and functional capacity of the gut microbiota between low-income and high-income countries correlate with differences in vaccine immunogenicity.
Highlighted are example studies that have compared vaccine immunogenicity in individuals from low-income and middle-income countries (LMICs; red, orange and yellow) to those living in high-income countries (HICs; purple); see Table 1 for further details. The data for oral vaccines having reduced immunogenicity in LMICs are particularly convincing but further work is required to confirm whether responses to parenteral vaccines are impaired in LMICs as many of the reports so far are based on post hoc analyses of independent cohorts. Intriguingly, reported differences in vaccine immunogenicity correlate with differences in the composition and functional capacity of the gut microbiota between these populations. Classifications of income status are based on data from the World Bank, which within the broad category of LMICs, classifies countries as low income (red), lower-middle income (orange) and upper-middle income (yellow). BCG, Bacillus Calmette-Guérin; DTP–HepB–Hib, diphtheria, tetanus, pertussis–hepatitis B virus–Haemophilus influenzae type B; IFNγ, interferon-γ.
Fig. 2. Factors with the potential to…
Fig. 2. Factors with the potential to influence vaccine immunogenicity and/or efficacy.
A range of intrinsic host factors (such as age, sex, genetics and comorbidities) and extrinsic factors (such as perinatal, nutritional, environmental and behavioural factors) have been suggested to influence vaccine immunogenicity and/or efficacy (reviewed in detail in ref.). The influence of these factors on vaccine immunogenicity is likely mediated indirectly via the effects of these factors on baseline immunity and/or the composition of the microbiota. Vaccine immunogenicity is also, of course, dependent on vaccine-intrinsic factors such as the adjuvant used, and vaccine efficacy may be influenced by factors other than vaccine immunogenicity such as the degree of match between the vaccine and the strains circulating at the time.
Fig. 3. Differences in the gut microbiota…
Fig. 3. Differences in the gut microbiota of infants and the elderly compared with that of young adults correlate with altered immune status and suboptimal vaccine immunogenicity.
a | The composition of the gut microbiota in early life is unstable and has low levels of diversity, with a small number of bacterial families tending to dominate. Over time, the diversity of the gut microbiota increases until an adult-like composition is reached between 2 and 3 years of age. The adult gut microbiota is more complex than in infancy (higher levels of diversity) but is also more homogeneous between individuals and, in the absence of external perturbations (such as antibiotics), is generally quite stable. b | As people age, the diversity and stability of the gut microbiota decline. There is also an increased relative abundance of the inflammation-associated Proteobacteria and a decrease in Actinobacteria. c | The composition of the gut microbiota can strongly influence immune function and the baseline status of the immune system at the time of vaccination. Baseline immune status has been shown to be predictive of responses to vaccination in several studies. An in-depth discussion of other factors that influence changes in immune status with ageing is outside the scope of this Review. d | Compared with healthy adults, vaccine immunogenicity is poorer in infants and in the elderly. Increasing data suggest causal links between these phenomena.
Fig. 4. Potential mechanisms by which the…
Fig. 4. Potential mechanisms by which the microbiota could modulate vaccine immunogenicity and efficacy.
a | Immunomodulatory molecules produced by the microbiota, such as flagellin and peptidoglycan, have been shown in animal models to modulate vaccine responses by providing natural adjuvants that are sensed by pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs) and NOD2, expressed by antigen-presenting cells. Other immunomodulatory molecules, such as lipopolysaccharide, may also similarly modulate responses. PRRs expressed by T cells and B cells may also sense these molecules directly. b | Dendritic cells (DCs) have a crucial role in immune responses to vaccination by presenting vaccine antigens to T cells and secreting immunomodulatory cytokines. The microbiota regulates the production of type I interferons by plasmacytoid DCs (pDCs), which in turn instruct a specific metabolic and epigenomic state in conventional DCs (cDCs) that enhances T cell priming. c | Immunomodulatory metabolites produced by the microbiota, such as short-chain fatty acids (SCFAs), can enhance B cell metabolism to support the energy demands of antibody production and can increase the expression of genes involved in plasma cell differentiation and class switching, potentially altering responses to vaccination. d | Increasing data suggest that the microbiota can encode epitopes that are cross-reactive with pathogen-encoded or vaccine-encoded epitopes. The presence of cross-reactive B cells or T cells could potentially alter the responses to vaccination.

References

    1. Andre FE, et al. Vaccination greatly reduces disease, disability, death and inequity worldwide. Bull. World Health Organ. 2008;86:140–146. doi: 10.2471/BLT.07.040089.
    1. Lewnard JA, Lo NC, Arinaminpathy N, Frost I, Laxminarayan R. Childhood vaccines and antibiotic use in low- and middle-income countries. Nature. 2020;581:94–99. doi: 10.1038/s41586-020-2238-4.
    1. Siegrist, C. A. Vaccine immunology. in Vaccines (eds Plotkin, S. A., Orenstein, W. A. & Offit, P. A.) 17–36 (Elsevier Inc, 2008).
    1. Zimmermann P, Curtis N. Factors that influence the immune response to vaccination. Clin. Microbiol. Rev. 2019;32:e00084–18. doi: 10.1128/CMR.00084-18.
    1. PrabhuDas M, et al. Challenges in infant immunity: implications for responses to infection and vaccines. Nat. Immunol. 2011;12:189–194. doi: 10.1038/ni0311-189.
    1. Ciabattini A, et al. Vaccination in the elderly: the challenge of immune changes with aging. Semin. Immunol. 2018;40:83–94. doi: 10.1016/j.smim.2018.10.010.
    1. DeJong EN, Surette MG, Bowdish DME. The gut microbiota and unhealthy aging: disentangling cause from consequence. Cell Host Microbe. 2020;28:180–189. doi: 10.1016/j.chom.2020.07.013.
    1. Backhed F, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe. 2015;17:690–703. doi: 10.1016/j.chom.2015.04.004.
    1. Pasolli E, et al. Extensive unexplored human microbiome diversity revealed by over 150,000 genomes from metagenomes spanning age, geography, and lifestyle. Cell. 2019;176:649–662. doi: 10.1016/j.cell.2019.01.001.
    1. Lynn DJ, Pulendran B. The potential of the microbiota to influence vaccine responses. J. Leukoc. Biol. 2017;103:225–231. doi: 10.1189/jlb.5MR0617-216R.
    1. Parker EP, et al. Causes of impaired oral vaccine efficacy in developing countries. Future Microbiol. 2018;13:97–118. doi: 10.2217/fmb-2017-0128.
    1. Patel M, et al. A systematic review of anti-rotavirus serum IgA antibody titer as a potential correlate of rotavirus vaccine efficacy. J. Infect. Dis. 2013;208:284–294. doi: 10.1093/infdis/jit166.
    1. Clark A, et al. Efficacy of live oral rotavirus vaccines by duration of follow-up: a meta-regression of randomised controlled trials. Lancet Infect. Dis. 2019;19:717–727. doi: 10.1016/S1473-3099(19)30126-4.
    1. Hallander HO, et al. Calibrated serological techniques demonstrate significant different serum response rates to an oral killed cholera vaccine between Swedish and Nicaraguan children. Vaccine. 2002;21:138–145. doi: 10.1016/S0264-410X(02)00348-1.
    1. Patriarca PA, Wright PF, John TJ. Factors affecting the immunogenicity of oral poliovirus vaccine in developing countries: review. Rev. Infect. Dis. 1991;13:926–939. doi: 10.1093/clinids/13.5.926.
    1. Levine MM, Kotloff KL, Barry EM, Pasetti MF, Sztein MB. Clinical trials of Shigella vaccines: two steps forward and one step back on a long, hard road. Nat. Rev. Microbiol. 2007;5:540–553. doi: 10.1038/nrmicro1662.
    1. Rusmil K, et al. The immunogenicity, safety, and consistency of an Indonesia combined DTP-HB-Hib vaccine in expanded program on immunization schedule. BMC Pediatr. 2015;15:219. doi: 10.1186/s12887-015-0525-2.
    1. Dbaibo G, et al. Quadrivalent influenza vaccine prevents illness and reduces healthcare utilization across diverse geographic regions during five influenza seasons: a randomized clinical trial. Pediatr. Infect. Dis. J. 2020;39:e1–e10. doi: 10.1097/INF.0000000000002504.
    1. Rts SCTP. Efficacy and safety of the RTS,S/AS01 malaria vaccine during 18 months after vaccination: a phase 3 randomized, controlled trial in children and young infants at 11 African sites. PLoS Med. 2014;11:e1001685. doi: 10.1371/journal.pmed.1001685.
    1. Muyanja E, et al. Immune activation alters cellular and humoral responses to yellow fever 17D vaccine. J. Clin. Invest. 2014;124:3147–3158. doi: 10.1172/JCI75429.
    1. Pasin C, et al. Dynamics of the humoral immune response to a prime-boost Ebola vaccine: quantification and sources of variation. J. Virol. 2019;93:e00579–19. doi: 10.1128/JVI.00579-19.
    1. Choe YJ, Blatt DB, Lee HJ, Choi EH. Associations between geographic region and immune response variations to pneumococcal conjugate vaccines in clinical trials: A systematic review and meta-analysis. Int. J. Infect. Dis. 2020;92:261–268. doi: 10.1016/j.ijid.2019.12.021.
    1. Lalor MK, et al. Population differences in immune responses to Bacille Calmette-Guerin vaccination in infancy. J. Infect. Dis. 2009;199:795–800. doi: 10.1086/597069.
    1. Baden LR, et al. Assessment of the safety and immunogenicity of 2 novel vaccine platforms for HIV-1 prevention: a randomized trial. Ann. Intern. Med. 2016;164:313–322. doi: 10.7326/M15-0880.
    1. Cox RJ, Brokstad KA. Not just antibodies: B cells and T cells mediate immunity to COVID-19. Nat. Rev. Immunol. 2020;20:581–582. doi: 10.1038/s41577-020-00436-4.
    1. Osterholm MT, Kelley NS, Sommer A, Belongia EA. Efficacy and effectiveness of influenza vaccines: a systematic review and meta-analysis. Lancet Infect. Dis. 2012;12:36–44. doi: 10.1016/S1473-3099(11)70295-X.
    1. Tsang JS, et al. Improving vaccine-induced immunity: can baseline predict outcome? Trends Immunol. 2020;41:457–465. doi: 10.1016/j.it.2020.04.001.
    1. Simon AK, Hollander GA, McMichael A. Evolution of the immune system in humans from infancy to old age. Proc. Biol. Sci. 2015;282:20143085.
    1. Hill DL, et al. Immune system development varies according to age, location, and anemia in African children. Sci. Transl Med. 2020;12:eaaw9522. doi: 10.1126/scitranslmed.aaw9522.
    1. Wargo JA. Modulating gut microbes. Science. 2020;369:1302–1303. doi: 10.1126/science.abc3965.
    1. Harris VC, et al. The infant gut microbiome correlates significantly with rotavirus vaccine response in rural Ghana. J. Infect. Dis. 2017;215:34–41. doi: 10.1093/infdis/jiw518.
    1. Harris V, et al. Rotavirus vaccine response correlates with the infant gut microbiota composition in Pakistan. Gut Microbes. 2018;9:93–101. doi: 10.1080/19490976.2017.1376162.
    1. Parker EPK, et al. Influence of the intestinal microbiota on the immunogenicity of oral rotavirus vaccine given to infants in south India. Vaccine. 2018;36:264–272. doi: 10.1016/j.vaccine.2017.11.031.
    1. Fix J, et al. Association between gut microbiome composition and rotavirus vaccine response among Nicaraguan infants. Am. J. Trop. Med. Hyg. 2020;102:213–219. doi: 10.4269/ajtmh.19-0355.
    1. Zhao T, et al. Influence of gut microbiota on mucosal IgA antibody response to the polio vaccine. NPJ Vaccines. 2020;5:47. doi: 10.1038/s41541-020-0194-5.
    1. Praharaj I, et al. Influence of nonpolio enteroviruses and the bacterial gut microbiota on oral poliovirus vaccine response: a study from South India. J. Infect. Dis. 2019;219:1178–1186. doi: 10.1093/infdis/jiy568.
    1. Huda MN, et al. Bifidobacterium abundance in early infancy and vaccine response at 2 years of age. Pediatrics. 2019;143:e20181489. doi: 10.1542/peds.2018-1489.
    1. Zimmermann P, et al. Biological sex influences antibody responses to routine vaccinations in the first year of life. Acta. Paediatr. 2020;109:147–157. doi: 10.1111/apa.14932.
    1. Grassly NC, et al. The effect of azithromycin on the immunogenicity of oral poliovirus vaccine: a double-blind randomised placebo-controlled trial in seronegative Indian infants. Lancet Infect. Dis. 2016;16:905–914. doi: 10.1016/S1473-3099(16)30023-8.
    1. Gilmartin AA, Petri WA., Jr. Exploring the role of environmental enteropathy in malnutrition, infant development and oral vaccine response. Philos. Trans. R Soc. Lond. B Biol. Sci. 2015;370:20140143. doi: 10.1098/rstb.2014.0143.
    1. Harris VC, et al. Effect of antibiotic-mediated microbiome modulation on rotavirus vaccine immunogenicity: a human, randomized-control proof-of-concept trial. Cell Host Microbe. 2018;24:197–207. doi: 10.1016/j.chom.2018.07.005.
    1. Hagan T, et al. Antibiotics-driven gut microbiome perturbation alters immunity to vaccines in humans. Cell. 2019;178:1313–1328. doi: 10.1016/j.cell.2019.08.010.
    1. Nakaya HI, et al. Systems analysis of immunity to influenza vaccination across multiple years and in diverse populations reveals shared molecular signatures. Immunity. 2015;43:1186–1198. doi: 10.1016/j.immuni.2015.11.012.
    1. Lynn MA, et al. Early-life antibiotic-driven dysbiosis leads to dysregulated vaccine immune responses in mice. Cell Host Microbe. 2018;23:653–660. doi: 10.1016/j.chom.2018.04.009.
    1. The Australian New Zealand Clinical Trials Registry (ANZCTR). A clinical study to determine whether antibiotic-driven dysbiosis is associated with impaired vaccine responses in infants. ANZCTR (2017).
    1. Uchiyama R, Chassaing B, Zhang B, Gewirtz AT. Antibiotic treatment suppresses rotavirus infection and enhances specific humoral immunity. J. Infect. Dis. 2014;210:171–182. doi: 10.1093/infdis/jiu037.
    1. Lamousé-Smith ES, Tzeng A, Starnbach MN. The intestinal flora is required to support antibody responses to systemic immunization in infant and germ free mice. PLoS ONE. 2011;6:e27662. doi: 10.1371/journal.pone.0027662.
    1. Oh JZ, et al. TLR5-mediated sensing of gut microbiota is necessary for antibody responses to seasonal influenza vaccination. Immunity. 2014;41:478–492. doi: 10.1016/j.immuni.2014.08.009.
    1. Klein EY, et al. Global increase and geographic convergence in antibiotic consumption between 2000 and 2015. Proc. Natl Acad. Sci. USA. 2018;115:E3463–E3470. doi: 10.1073/pnas.1717295115.
    1. Al Nabhani Z, et al. A weaning reaction to microbiota is required for resistance to immunopathologies in the adult. Immunity. 2019;50:1276–1288. doi: 10.1016/j.immuni.2019.02.014.
    1. New JS, et al. Neonatal exposure to commensal-bacteria-derived antigens directs polysaccharide-specific B-1 B cell repertoire development. Immunity. 2020;53:172–186. doi: 10.1016/j.immuni.2020.06.006.
    1. Li H, et al. Mucosal or systemic microbiota exposures shape the B cell repertoire. Nature. 2020;584:274–278. doi: 10.1038/s41586-020-2564-6.
    1. Kageyama T, Matsuo T, Kurakake R, Sano T. Relationship between T cells and microbiota in health and disease. Prog. Mol. Biol. Transl Sci. 2020;171:95–129. doi: 10.1016/bs.pmbts.2020.03.007.
    1. Abt MC, et al. Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity. 2012;37:158–170. doi: 10.1016/j.immuni.2012.04.011.
    1. Ichinohe T, et al. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc. Natl Acad. Sci. USA. 2011;108:5354–5359. doi: 10.1073/pnas.1019378108.
    1. Georg P, Sander LE. Innate sensors that regulate vaccine responses. Curr. Opin. Immunol. 2019;59:31–41. doi: 10.1016/j.coi.2019.02.006.
    1. Nakaya HI, et al. Systems biology of vaccination for seasonal influenza in humans. Nat. Immunol. 2011;12:786–795. doi: 10.1038/ni.2067.
    1. Kim D, et al. Nod2-mediated recognition of the microbiota is critical for mucosal adjuvant activity of cholera toxin. Nat. Med. 2016;22:524–530. doi: 10.1038/nm.4075.
    1. Kasturi SP, et al. Programming the magnitude and persistence of antibody responses with innate immunity. Nature. 2011;470:543–547. doi: 10.1038/nature09737.
    1. Vatanen T, et al. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell. 2016;165:842–853. doi: 10.1016/j.cell.2016.04.007.
    1. Zeng MY, Inohara N, Nunez G. Mechanisms of inflammation-driven bacterial dysbiosis in the gut. Mucosal Immunol. 2017;10:18–26. doi: 10.1038/mi.2016.75.
    1. Mu Q, Kirby J, Reilly CM, Luo XM. Leaky gut as a danger signal for autoimmune diseases. Front. Immunol. 2017;8:598. doi: 10.3389/fimmu.2017.00598.
    1. Singh R, et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat. Commun. 2019;10:89. doi: 10.1038/s41467-018-07859-7.
    1. Ruane D, et al. Microbiota regulate the ability of lung dendritic cells to induce IgA class-switch recombination and generate protective gastrointestinal immune responses. J. Exp. Med. 2016;213:53–73. doi: 10.1084/jem.20150567.
    1. Robak OH, et al. Antibiotic treatment-induced secondary IgA deficiency enhances susceptibility to Pseudomonas aeruginosa pneumonia. J. Clin. Invest. 2018;128:3535–3545. doi: 10.1172/JCI97065.
    1. Schaupp L, et al. Microbiota-induced type I interferons instruct a poised basal state of dendritic cells. Cell. 2020;181:1080–1096. doi: 10.1016/j.cell.2020.04.022.
    1. Koyama M, et al. MHC Class II antigen presentation by the intestinal epithelium initiates graft-versus-host disease and is influenced by the microbiota. Immunity. 2019;51:885–898. doi: 10.1016/j.immuni.2019.08.011.
    1. Bittinger K, et al. Bacterial colonization reprograms the neonatal gut metabolome. Nat. Microbiol. 2020;5:838–847. doi: 10.1038/s41564-020-0694-0.
    1. Kim M, Qie Y, Park J, Kim CH. Gut microbial metabolites fuel host antibody responses. Cell Host Microbe. 2016;20:202–214. doi: 10.1016/j.chom.2016.07.001.
    1. Sanchez HN, et al. B cell-intrinsic epigenetic modulation of antibody responses by dietary fiber-derived short-chain fatty acids. Nat. Commun. 2020;11:60. doi: 10.1038/s41467-019-13603-6.
    1. Scott SA, Fu J, Chang PV. Microbial tryptophan metabolites regulate gut barrier function via the aryl hydrocarbon receptor. Proc. Natl Acad. Sci. USA. 2020;117:19376–19387. doi: 10.1073/pnas.2000047117.
    1. Garrett WS. Immune recognition of microbial metabolites. Nat. Rev. Immunol. 2020;20:91–92. doi: 10.1038/s41577-019-0252-2.
    1. Su LF, Kidd BA, Han A, Kotzin JJ, Davis MM. Virus-specific CD4+ memory-phenotype T cells are abundant in unexposed adults. Immunity. 2013;38:373–383. doi: 10.1016/j.immuni.2012.10.021.
    1. Hegazy AN, et al. Circulating and tissue-resident CD4+ T cells with reactivity to intestinal microbiota are abundant in healthy individuals and function is altered during inflammation. Gastroenterology. 2017;153:1320–1337. doi: 10.1053/j.gastro.2017.07.047.
    1. Bremel RD, Homan EJ. Extensive T-cell epitope repertoire sharing among human proteome, gastrointestinal microbiome, and pathogenic bacteria: implications for the definition of self. Front. Immunol. 2015;6:538. doi: 10.3389/fimmu.2015.00538.
    1. Williams WB, et al. HIV-1 VACCINES. Diversion of HIV-1 vaccine-induced immunity by gp41-microbiota cross-reactive antibodies. Science. 2015;349:aab1253. doi: 10.1126/science.aab1253.
    1. Carrasco Pro S, et al. Microbiota epitope similarity either dampens or enhances the immunogenicity of disease-associated antigenic epitopes. PLoS ONE. 2018;13:e0196551. doi: 10.1371/journal.pone.0196551.
    1. Wilkinson TM, et al. Preexisting influenza-specific CD4+ T cells correlate with disease protection against influenza challenge in humans. Nat. Med. 2012;18:274–280. doi: 10.1038/nm.2612.
    1. Fluckiger A, et al. Cross-reactivity between tumor MHC class I-restricted antigens and an enterococcal bacteriophage. Science. 2020;369:936–942. doi: 10.1126/science.aax0701.
    1. Cully M. Microbiome therapeutics go small molecule. Nat. Rev. Drug Discov. 2019;18:569–572. doi: 10.1038/d41573-019-00122-8.
    1. Sanders ME, Merenstein DJ, Reid G, Gibson GR, Rastall RA. Probiotics and prebiotics in intestinal health and disease: from biology to the clinic. Nat. Rev. Gastroenterol. Hepatol. 2019;16:605–616. doi: 10.1038/s41575-019-0173-3.
    1. AlFaleh K, Anabrees J. Probiotics for prevention of necrotizing enterocolitis in preterm infants. Evid. Based Child. Health. 2014;9:584–671. doi: 10.1002/ebch.1976.
    1. Szajewska H, et al. Systematic review with meta-analysis: Lactobacillus rhamnosus GG for treating acute gastroenteritis in children – a 2019 update. Aliment. Pharmacol. Ther. 2019;49:1376–1384. doi: 10.1111/apt.15267.
    1. Panigrahi P, et al. A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature. 2017;548:407–412. doi: 10.1038/nature23480.
    1. Di Luccia B, et al. Combined prebiotic and microbial intervention improves oral cholera vaccination responses in a mouse model of childhood undernutrition. Cell Host Microbe. 2020;27:899–908. doi: 10.1016/j.chom.2020.04.008.
    1. Zimmermann P, Curtis N. The influence of probiotics on vaccine responses – a systematic review. Vaccine. 2018;36:207–213. doi: 10.1016/j.vaccine.2017.08.069.
    1. Maruyama M, et al. The effects of non-viable Lactobacillus on immune function in the elderly: a randomised, double-blind, placebo-controlled study. Int. J. Food. Sci. Nutr. 2016;67:67–73. doi: 10.3109/09637486.2015.1126564.
    1. Akatsu H, et al. Lactobacillus in jelly enhances the effect of influenza vaccination in elderly individuals. J. Am. Geriatr. Soc. 2013;61:1828–1830. doi: 10.1111/jgs.12474.
    1. Bunout D, et al. Effects of a nutritional supplement on the immune response and cytokine production in free-living Chilean elderly. JPEN J. Parenter. Enteral Nutr. 2004;28:348–354. doi: 10.1177/0148607104028005348.
    1. Boge T, et al. A probiotic fermented dairy drink improves antibody response to influenza vaccination in the elderly in two randomised controlled trials. Vaccine. 2009;27:5677–5684. doi: 10.1016/j.vaccine.2009.06.094.
    1. Kotler E, et al. Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell. 2018;174:1406–1423. doi: 10.1016/j.cell.2018.08.047.
    1. Ojima MN, et al. Bifidobacterium bifidum suppresses gut inflammation caused by repeated antibiotic disturbance without recovering gut microbiome diversity in mice. Front. Microbiol. 2020;11:1349–1349. doi: 10.3389/fmicb.2020.01349.
    1. Pulendran B. Immunology taught by vaccines. Science. 2019;366:1074–1075. doi: 10.1126/science.aau6975.
    1. Geva-Zatorsky N, et al. Mining the human gut microbiota for immunomodulatory organisms. Cell. 2017;168:928–943. doi: 10.1016/j.cell.2017.01.022.
    1. Painter SD, Ovsyannikova IG, Poland GA. The weight of obesity on the human immune response to vaccination. Vaccine. 2015;33:4422–4429. doi: 10.1016/j.vaccine.2015.06.101.
    1. Ledford H. How obesity could create problems for a COVID vaccine. Nature. 2020;586:488–489. doi: 10.1038/d41586-020-02946-6.
    1. Salk HM, et al. Taxa of the nasal microbiome are associated with influenza-specific IgA response to live attenuated influenza vaccine. PLoS ONE. 2016;11:e0162803. doi: 10.1371/journal.pone.0162803.
    1. Chen YE, Fischbach MA, Belkaid Y. Skin microbiota-host interactions. Nature. 2018;553:427–436. doi: 10.1038/nature25177.
    1. Kim E, et al. Microneedle array delivered recombinant coronavirus vaccines: Immunogenicity and rapid translational development. EBioMedicine. 2020;55:102743. doi: 10.1016/j.ebiom.2020.102743.
    1. Yeung F, et al. Altered immunity of laboratory mice in the natural environment is associated with fungal colonization. Cell Host Microbe. 2020;27:809–822. doi: 10.1016/j.chom.2020.02.015.
    1. Aaby P, et al. The non-specific and sex-differential effects of vaccines. Nat. Rev. Immunol. 2020;20:464–470. doi: 10.1038/s41577-020-0338-x.
    1. Segata N. Gut microbiome: westernization and the disappearance of intestinal diversity. Curr. Biol. 2015;25:R611–R613. doi: 10.1016/j.cub.2015.05.040.
    1. Smits SA, et al. Seasonal cycling in the gut microbiome of the Hadza hunter-gatherers of Tanzania. Science. 2017;357:802–806. doi: 10.1126/science.aan4834.
    1. De Filippo C, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl Acad. Sci. USA. 2010;107:14691–14696. doi: 10.1073/pnas.1005963107.
    1. Brushett S, Sinha T, Reijneveld SA, de Kroon MLA, Zhernakova A. The effects of urbanization on the infant gut microbiota and health outcomes. Front. Pediatr. 2020;8:408. doi: 10.3389/fped.2020.00408.
    1. Vangay P, et al. US immigration westernizes the human gut microbiome. Cell. 2018;175:962–972. doi: 10.1016/j.cell.2018.10.029.

Source: PubMed

3
구독하다