Stool microbiota and vaccine responses of infants

M Nazmul Huda, Zachery Lewis, Karen M Kalanetra, Mamunur Rashid, Shaikh M Ahmad, Rubhana Raqib, Firdausi Qadri, Mark A Underwood, David A Mills, Charles B Stephensen, M Nazmul Huda, Zachery Lewis, Karen M Kalanetra, Mamunur Rashid, Shaikh M Ahmad, Rubhana Raqib, Firdausi Qadri, Mark A Underwood, David A Mills, Charles B Stephensen

Abstract

Objective: Oral vaccine efficacy is low in less-developed countries, perhaps due to intestinal dysbiosis. This study determined if stool microbiota composition predicted infant oral and parenteral vaccine responses.

Methods: The stool microbiota of 48 Bangladeshi infants was characterized at 6, 11, and 15 weeks of age by amplification and sequencing of the 16S ribosomal RNA gene V4 region and by Bifidobacterium-specific, quantitative polymerase chain reaction. Responses to oral polio virus (OPV), bacille Calmette-Guérin (BCG), tetanus toxoid (TT), and hepatitis B virus vaccines were measured at 15 weeks by using vaccine-specific T-cell proliferation for all vaccines, the delayed-type hypersensitivity skin-test response for BCG, and immunoglobulin G responses using the antibody in lymphocyte supernatant method for OPV, TT, and hepatitis B virus. Thymic index (TI) was measured by ultrasound.

Results: Actinobacteria (predominantly Bifidobacterium longum subspecies infantis) dominated the stool microbiota, with Proteobacteria and Bacteroidetes increasing by 15 weeks. Actinobacteria abundance was positively associated with T-cell responses to BCG, OPV, and TT; with the delayed-type hypersensitivity response; with immunoglobulin G responses; and with TI. B longum subspecies infantis correlated positively with TI and several vaccine responses. Bacterial diversity and abundance of Enterobacteriales, Pseudomonadales, and Clostridiales were associated with neutrophilia and lower vaccine responses.

Conclusions: Bifidobacterium predominance may enhance thymic development and responses to both oral and parenteral vaccines early in infancy, whereas deviation from this pattern, resulting in greater bacterial diversity, may cause systemic inflammation (neutrophilia) and lower vaccine responses. Vaccine responsiveness may be improved by promoting intestinal bifidobacteria and minimizing dysbiosis early in infancy.

Keywords: Actinobacteria; Bangladesh; Bifidobacterium; Proteobacteria; T lymphocyte; antibody; hepatitis; intestinal; microbiota; polio; tetanus; tuberculosis; vaccine.

Copyright © 2014 by the American Academy of Pediatrics.

Figures

FIGURE 1
FIGURE 1
A, Relative abundance of microbiota by 16S rRNA gene sequence analysis in each of the 48 subjects (numbered from highest to lowest abundance of Actinobacteria at 6 weeks) at all time points. For each subject, bars are ordered by age (6, 11, and 15 weeks) from left to right. B, Mean relative abundance at 6, 11, and 15 weeks of age.
FIGURE 2
FIGURE 2
Positive association of vaccine responses with stool Actinobacteria at 15 weeks of age. The relative abundance of stool microbiota was determined by sequence analysis of the 16S rRNA gene. Vaccine responses were characterized by the CD4 T-cell SI in response to vaccine antigens for OPV vaccine (A), BCG vaccine using PPD antigen (B), TT vaccine (C), HBV vaccine (D), and SEB (E), the positive control for T-cell stimulation. Box plots (A–D) show the median; 10th, 25th, 75th, and 90th percentiles; and individual outliers. P values (Wilcoxon rank-sum test) for comparisons between groups were as follows: A, .046; B, .036; C, .019; and D, 0.78. E, The scatterplot association was determined by Spearman correlation (R): R = 0.331, P = .034.
FIGURE 3
FIGURE 3
Positive association of vaccine responses with stool Bifidobacterium, B longum, B longum subspecies infantis, and B longum subspecies longum at 15 weeks of age. Infant vaccine responses were characterized as being above (“high”) or below (“low”) the median vaccine responses for the CD4 SI for OPV (A), the CD8 SI for OPV (B), the CD4 SI for PPD as an index of response to the BCG vaccine (C), and for the positive control for polyclonal T-cell proliferation, the CD4 SI for SEB (D). Differences between those above and below the median were identified by the Wilcoxon rank-sum test as indicated by the superscript letters: aP < .10, bP < .05, cP < .01. The Bifidobacterium genus was identified by PCR, B longum species by T-RFLP, and B longum subspecies infantis and B longum subspecies longum as described in Methods.
FIGURE 4
FIGURE 4
Positive association of vaccine responses with relative abundance of stool Bifidobacterium determined by sequence analysis of the 16S rRNA gene. Vaccine responses were characterized by CD4 stimulation index response to TT (A), CD4 stimulation index in response to the polyclonal T-cell mitogen SEB (B), the DTH skin-test response to PPD antigen as an index of the response to BCG immunization (C), and the IgG response to the OPV vaccine (D). Spearman correlation coefficients (R) and P values were as follows: A, R = 0.389, P = .013; B, R = 0.315, P = .045; C, R = 0.337, P = .020; and D, R = 0.301, P = .047.
FIGURE 5
FIGURE 5
Positive association of vaccine responses with stool Bifidobacteriales, Actinomycetales, and Coriobacteriales (from the class Actinobacteria) at 15 weeks of age. The relative abundance of stool microbiota at the order level in the class Actinobacteria was determined by sequence analysis of the 16S rRNA gene at 15 weeks of age. A and B, The response to BCG vaccination was determined by using the DTH skin-test response to PPD antigen. C, The response to TT vaccination was determined by using the antibody in lymphocyte supernatant assay to measure TT-specific IgG levels (TT IgG). D, The CD4 T-cell SI in response to PPD antigen. A, The scatterplot association was determined by Spearman correlation (R): R = 0.333, P = .025. Box plots (B, C, and D) show the median; 10th, 25th, 75th, and 90th percentiles; and individual outliers. P values (Wilcoxon rank-sum test) for comparisons between groups were as follows: B, .018; C, .041; and D, .034.
FIGURE 6
FIGURE 6
Negative association of stool Pseudomonadales (A), Enterobacteriales (B), and Shannon Diversity Index (measured at the phylum level) (C) with CD4 T-cell SI response to TT vaccine at 15 weeks of age. The relative abundance of stool microbiota was determined by sequence analysis of the 16S rRNA gene. Box plots show the median; 10th, 25th, 75th, and 90th percentiles; and individual outliers. P values (Wilcoxon rank-sum test) for comparisons between groups were as follows: A, 0.022; B, 0.047. C, The Spearman correlation coefficient was R = −0.490 with P = .0013.
FIGURE 7
FIGURE 7
Correlation of Bifidobacterium longum subspecies infantis in stool determined by qPCR with TI (A) and relative abundance of Escherichia/Shigella genera in stool determined by sequence analysis of the 16S rRNA gene with blood neutrophil levels (B). Spearman correlation coefficients (R) and P values were as follows: A, R = 0.389, P = .0083; B, R = 0.431, P = .0028.

References

    1. Matamoros S, Gras-Leguen C, Le Vacon F, Potel G, de La Cochetiere MF. Development of intestinal microbiota in infants and its impact on health. Trends Microbiol. 2013;21(4):167–173
    1. Lee YK, Mazmanian SK. Has the microbiota played a critical role in the evolution of the adaptive immune system? Science. 2010;330(6012):1768–1773
    1. Torrazza RM, Ukhanova M, Wang X, et al. . Intestinal microbial ecology and environmental factors affecting necrotizing enterocolitis. PLoS ONE. 2013;8(12):e83304.
    1. Bisgaard H, Li N, Bonnelykke K, et al. Reduced diversity of the intestinal microbiota during infancy is associated with increased risk of allergic disease at school age. J Allergy Clin Immunol. 2011;128(3):646–652, e641–e645
    1. Johnson CL, Versalovic J. The human microbiome and its potential importance to pediatrics. Pediatrics. 2012;129(5):950–960
    1. Isolauri E. Development of healthy gut microbiota early in life. J Paediatr Child Health. 2012;48(suppl 3):1–6
    1. Honda K, Littman DR. The microbiome in infectious disease and inflammation. Annu Rev Immunol. 2012;30:759–795
    1. Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune system. Science. 2012;336(6086):1268–1273
    1. Okwo-Bele JM, Cherian T. The expanded programme on immunization: a lasting legacy of smallpox eradication. Vaccine. 2011;29(suppl 4):D74–D79
    1. Qadri F, Bhuiyan TR, Sack DA, Svennerholm AM. Immune responses and protection in children in developing countries induced by oral vaccines. Vaccine. 2013;31(3):452–460
    1. Kuss SK, Best GT, Etheredge CA, et al. . Intestinal microbiota promote enteric virus replication and systemic pathogenesis. Science. 2011;334(6053):249–252
    1. Maynard CL, Elson CO, Hatton RD, Weaver CT. Reciprocal interactions of the intestinal microbiota and immune system. Nature. 2012;489(7415):231–241
    1. de Vrese M, Rautenberg P, Laue C, Koopmans M, Herremans T, Schrezenmeir J. Probiotic bacteria stimulate virus-specific neutralizing antibodies following a booster polio vaccination. Eur J Nutr. 2005;44(7):406–413
    1. Brisbin JT, Gong J, Orouji S, et al. . Oral treatment of chickens with lactobacilli influences elicitation of immune responses. Clin Vaccine Immunol. 2011;18(9):1447–1455
    1. Maidens C, Childs C, Przemska A, Dayel IB, Yaqoob P. Modulation of vaccine response by concomitant probiotic administration. Br J Clin Pharmacol. 2013;75(3):663–670
    1. Matsuda F, Chowdhury MI, Saha A, et al. . Evaluation of a probiotics, Bifidobacterium breve BBG-01, for enhancement of immunogenicity of an oral inactivated cholera vaccine and safety: a randomized, double-blind, placebo-controlled trial in Bangladeshi children under 5 years of age. Vaccine. 2011;29(10):1855–1858
    1. Fraher MH, O’Toole PW, Quigley EM. Techniques used to characterize the gut microbiota: a guide for the clinician. Nat Rev Gastroenterol Hepatol. 2012;9(6):312–322
    1. Biasucci G, Rubini M, Riboni S, Morelli L, Bessi E, Retetangos C. Mode of delivery affects the bacterial community in the newborn gut. Early Hum Dev. 2010;86(suppl 1):13–15
    1. Karlsson CL, Molin G, Cilio CM, Ahrné S. The pioneer gut microbiota in human neonates vaginally born at term—a pilot study. Pediatr Res. 2011;70(3):282–286
    1. Tsuji H, Oozeer R, Matsuda K, et al. . Molecular monitoring of the development of intestinal microbiota in Japanese infants. Benef Microbes. 2012;3(2):113–125
    1. Echarri PP, Graciá CM, Berruezo GR, et al. . Assessment of intestinal microbiota of full-term breast-fed infants from two different geographical locations. Early Hum Dev. 2011;87(7):511–513
    1. Fallani M, Young D, Scott J, et al. Members of the INFABIO Team . Intestinal microbiota of 6-week-old infants across Europe: geographic influence beyond delivery mode, breast-feeding, and antibiotics. J Pediatr Gastroenterol Nutr. 2010;51(1):77–84
    1. Savino W, Dardenne M. Nutritional imbalances and infections affect the thymus: consequences on T-cell-mediated immune responses. Proc Nutr Soc. 2010;69(4):636–643
    1. Lewis ZT, Bokulich NA, Kalanetra KM, Ruiz-Moyano S, Underwood MA, Mills DA. Use of bifidobacterial specific terminal restriction fragment length polymorphisms to complement next generation sequence profiling of infant gut communities. Anaerobe. 2013;19:62–69
    1. Kamada N, Seo SU, Chen GY, Núñez G. Role of the gut microbiota in immunity and inflammatory disease. Nat Rev Immunol. 2013;13(5):321–335
    1. Chung H, Pamp SJ, Hill JA, et al. . Gut immune maturation depends on colonization with a host-specific microbiota. Cell. 2012;149(7):1578–1593
    1. Smith K, McCoy KD, Macpherson AJ. Use of axenic animals in studying the adaptation of mammals to their commensal intestinal microbiota. Semin Immunol. 2007;19(2):59–69
    1. Ivanov II, Atarashi K, Manel N, et al. . Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139(3):485–498
    1. Atarashi K, Tanoue T, Shima T, et al. . Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331(6015):337–341
    1. Savino W, Dardenne M, Velloso LA, Dayse Silva-Barbosa S. The thymus is a common target in malnutrition and infection. Br J Nutr. 2007;98(suppl 1):S11–S16
    1. Frei R, Lauener RP, Crameri R, O’Mahony L. Microbiota and dietary interactions: an update to the hygiene hypothesis? Allergy. 2012;67(4):451–461
    1. Kuitunen M. Probiotics and prebiotics in preventing food allergy and eczema. Curr Opin Allergy Clin Immunol. 2013;13(3):280–286
    1. Licciardi PV, Tang ML. Vaccine adjuvant properties of probiotic bacteria. Discov Med. 2011;12(67):525–533
    1. Holscher HD, Czerkies LA, Cekola P, et al. . Bifidobacterium lactis Bb12 enhances intestinal antibody response in formula-fed infants: a randomized, double-blind, controlled trial. JPEN J Parenter Enteral Nutr. 2012;36(1 suppl):106S–117S
    1. Grönlund MM, Arvilommi H, Kero P, Lehtonen OP, Isolauri E. Importance of intestinal colonisation in the maturation of humoral immunity in early infancy: a prospective follow up study of healthy infants aged 0-6 months. Arch Dis Child Fetal Neonatal Ed. 2000;83(3):F186–F192
    1. Lundell AC, Björnsson V, Ljung A, et al. . Infant B cell memory differentiation and early gut bacterial colonization. J Immunol. 2012;188(9):4315–4322
    1. Sjögren YM, Tomicic S, Lundberg A, et al. . Influence of early gut microbiota on the maturation of childhood mucosal and systemic immune responses. Clin Exp Allergy. 2009;39(12):1842–1851
    1. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489(7415):220–230
    1. Mussap M, Noto A, Cibecchini F, Fanos V. The importance of biomarkers in neonatology. Semin Fetal Neonatal Med. 2013;18(1):56–64
    1. German JB, Freeman SL, Lebrilla CB, Mills DA. Human milk oligosaccharides: evolution, structures and bioselectivity as substrates for intestinal bacteria. Nestle Nutr Workshop Ser Pediatr Program. 2008;62:205–218; discussion 218–222
    1. Krstic A, Mojsilovic S, Jovcic G, Bugarski D. The potential of interleukin-17 to mediate hematopoietic response. Immunol Res. 2012;52(1–2):34–41
    1. Jiang W, Lederman MM, Hunt P, et al. . Plasma levels of bacterial DNA correlate with immune activation and the magnitude of immune restoration in persons with antiretroviral-treated HIV infection. J Infect Dis. 2009;199(8):1177–1185
    1. Kamal SM. Preference for institutional delivery and caesarean sections in Bangladesh. J Health Popul Nutr. 2013;31(1):96–109
    1. Moingeon P. Adjuvants for allergy vaccines. Hum Vaccin Immunother. 2012;8(10):1492–1498

Source: PubMed

3
Abonner