Daily preventive zinc supplementation increases the antibody response against pathogenic Escherichia coli in children with zinc insufficiency: a randomised controlled trial

Chidchamai Kewcharoenwong, Myint Myint Sein, Arnone Nithichanon, Aranya Khongmee, K Ryan Wessells, Guy-Marino Hinnouho, Maxwell A Barffour, Sengchanh Kounnavong, Sonja Y Hess, Charles B Stephensen, Ganjana Lertmemongkolchai, Chidchamai Kewcharoenwong, Myint Myint Sein, Arnone Nithichanon, Aranya Khongmee, K Ryan Wessells, Guy-Marino Hinnouho, Maxwell A Barffour, Sengchanh Kounnavong, Sonja Y Hess, Charles B Stephensen, Ganjana Lertmemongkolchai

Abstract

Zinc deficiency impairs the antibody-mediated immune response and is common in children from lower-income countries. This study aimed to investigate the impact of different zinc supplementation regimens (7, 10 or 20 mg/day elemental zinc)-therapeutic dispersible zinc tablets (TZ), daily multiple micronutrient powder (MNP), daily preventive zinc tablets (PZ) and placebo powder (control)-and compare between baseline and endline antibody production against pathogenic Escherichia coli in Laotian children (aged 6-23 months). Fifty representative plasma samples of each treatment group were randomly selected from 512 children to determine anti-E. coli IgG antibody levels and avidity. Of the 200 children, 78.5% had zinc deficiency (plasma zinc concentration < 65 µg/dL) and 40% had anaemia before receiving zinc supplementation. aAfter receiving the TZ, MNP or PZ regimen, the plasma anti-E. coli IgG levels were significantly increased compared with baseline; the effect on the antibody level was more pronounced in children with zinc deficiency. Interestingly, there was increased anti-E. coli IgG avidity in the control and PZ groups. This study suggests that PZ might be the optimal zinc supplementation regimen to increase both the quantity and quality of antibody responses in children with zinc deficiency. Clinical trial registration: https://ichgcp.net/clinical-trials-registry/NCT02428647 (NCT02428647, 29/04/2015).

Conflict of interest statement

The spouse of SYH worked for the Bill & Melinda Gates Foundation, which provided part of the financial support. The other authors declare no competing interests.

© 2022. The Author(s).

Figures

Figure 1
Figure 1
Plasma IgG levels in response to pathogenic Escherichia coli of the four intervention groups. (a) Plasma samples from baseline and endline of each intervention group (Control, n = 50; TZ, therapeutic zinc with diarrhoea, n = 48; MNP, zinc-containing micronutrient powder, n = 47; PZ, preventive zinc, n = 50) were used to determine the anti-E. coli IgG levels. The bars represent the median with the interquartile range of each group and each dot represents an individual sample. The P-value was determined with the Kruskal–Wallis test. (b) Changes in the antibody level in each individual. The P-values indicate significant differences between baseline and endline of each intervention group determined by the Wilcoxon matched-pairs signed rank test (**P < 0.01; ***P < 0.001; ns, non-significant). (c) The pie charts show the percentages of children who showed no change or an increase or decrease of at least 10% in antibody levels at endline compared with baseline for each group. (d) Chi-square analysis of the relative risk and P-values of increasing antibody levels for each intervention group compared with the Control group.
Figure 2
Figure 2
Changes in the avidity index of children among the four intervention groups. (a) Plasma samples from baseline and endline of each intervention group (Control, n = 50; TZ, therapeutic zinc with diarrhoea, n = 50; MNP, zinc-containing micronutrient powder, n = 50; PZ, preventive zinc, n = 50) were used to determine the avidity index. Bars represent the median with the interquartile range of each group; each dot represents an individual sample. The P-value was determined with the Kruskal–Wallis test. (b) Changes in the avidity index of each individual. The P-values indicate significant differences between baseline and endline of each intervention group determined by the Wilcoxon matched-pairs signed rank test (**P < 0.01; ***P < 0.001; ns non-significant). (c) The pie charts show the percentages of children who showed no change or an increase or decrease of at least 5% in the avidity index at endline compared with baseline for each group. (d) Chi-square analysis of relative risk and P-values of increasing avidity from each intervention group compared with the Control group.
Figure 3
Figure 3
The plasma IgG level and avidity index of the four intervention groups divided by the baseline zinc status. For each group, the (a) plasma IgG level and (b) avidity index were divided into zinc-sufficient and zinc-deficient sub-groups (C control; TZ therapeutic zinc with diarrhoea; MNP zinc-containing micronutrient powder; PZ preventive zinc). Children who had baseline plasma zinc lower than 65 μg/dL were categorised in the zinc-deficient sub-group. The sample sizes are indicated above each graph. Each dot represents an individual sample. Zn Suf zinc sufficient group; Zn Def zinc deficient group. The P-values indicate significant differences between baseline and endline of each intervention group determined by the Wilcoxon matched-pairs signed rank test. *P < 0.05; **P < 0.01; ***P < 0.001; ns non-significant.

References

    1. Wu D, Lewis ED, Pae M, Meydani SN. Nutritional modulation of immune function: Analysis of evidence, mechanisms, and clinical relevance. Front. Immunol. 2018;9:3160. doi: 10.3389/fimmu.2018.03160.
    1. Maywald M, Wessels I, Rink L. Zinc signals and immunity. Int. J. Mol. Sci. 2017 doi: 10.3390/ijms18102222.
    1. Hojyo S, Fukada T. Roles of zinc signaling in the immune system. J. Immunol. Res. 2016;2016:6762343. doi: 10.1155/2016/6762343.
    1. Allman D, Pillai S. Peripheral B cell subsets. Curr. Opin. Immunol. 2008;20:149–157. doi: 10.1016/j.coi.2008.03.014.
    1. Hojyo S, et al. Zinc transporter SLC39A10/ZIP10 controls humoral immunity by modulating B-cell receptor signal strength. Proc. Natl. Acad. Sci. USA. 2014;111:11786–11791. doi: 10.1073/pnas.1323557111.
    1. Wessells KR, Brown KH. Estimating the global prevalence of zinc deficiency: Results based on zinc availability in national food supplies and the prevalence of stunting. PLoS ONE. 2012;7:e50568. doi: 10.1371/journal.pone.0050568.
    1. Brown KH, Peerson JM, Baker SK, Hess SY. Preventive zinc supplementation among infants, preschoolers, and older prepubertal children. Food Nutr. Bull. 2009;30:S12–40. doi: 10.1177/15648265090301S103.
    1. Mayo-Wilson E, Imdad A, Junior J, Dean S, Bhutta ZA. Preventive zinc supplementation for children, and the effect of additional iron: A systematic review and meta-analysis. BMJ Open. 2014;4:e004647. doi: 10.1136/bmjopen-2013-004647.
    1. WHO. Diarrhoeal Disease. (2017).
    1. Abba K, Sinfield R, Hart CA, Garner P. Pathogens associated with persistent diarrhoea in children in low and middle income countries: Systematic review. BMC Infect. Dis. 2009;9:88. doi: 10.1186/1471-2334-9-88.
    1. Sarmin M, et al. Predictor of death in diarrheal children under 5 years of age having severe sepsis in an urban critical care ward in Bangladesh. Glob. Pediatr. Health. 2019;6:2333794X19862716. doi: 10.1177/2333794X19862716.
    1. Li Y, Frey E, Mackenzie AM, Finlay BB. Human response to Escherichia coli O157:H7 infection: Antibodies to secreted virulence factors. Infect. Immun. 2000;68:5090–5095. doi: 10.1128/IAI.68.9.5090-5095.2000.
    1. Giuntini S, et al. Identification and characterization of human monoclonal antibodies for immunoprophylaxis against enterotoxigenic Escherichia coli infection. Infect. Immun. 2018 doi: 10.1128/IAI.00355-18.
    1. Alam MM, et al. Antigen-specific memory B-cell responses to enterotoxigenic Escherichia coli infection in Bangladeshi adults. PLoS Negl. Trop. Dis. 2014;8:e2822. doi: 10.1371/journal.pntd.0002822.
    1. Alam MM, et al. Antibody avidity in humoral immune responses in Bangladeshi children and adults following administration of an oral killed cholera vaccine. Clin. Vaccine Immunol. 2013;20:1541–1548. doi: 10.1128/CVI.00341-13.
    1. Wessells KR, et al. Comparison of two forms of daily preventive zinc supplementation versus therapeutic zinc supplementation for diarrhea on young children's physical growth and risk of infection: Study design and rationale for a randomized controlled trial. BMC Nutr. 2018;4:39. doi: 10.1186/s40795-018-0247-6.
    1. Kewcharoenwong C, et al. Daily preventive zinc supplementation decreases lymphocyte and eosinophil concentrations in rural Laotian children from communities with a high prevalence of zinc deficiency: Results of a randomized controlled trial. J. Nutr. 2020;150:2204–2213. doi: 10.1093/jn/nxaa037.
    1. Wessells KR, et al. Impact of daily preventive zinc or therapeutic zinc supplementation for diarrhea on plasma biomarkers of environmental enteric dysfunction among rural laotian children: A randomized controlled trial. Am. J. Trop. Med. Hyg. 2020;102:415–426. doi: 10.4269/ajtmh.19-0584.
    1. Barffour MA, et al. Effects of daily zinc, daily multiple micronutrient powder, or therapeutic zinc supplementation for diarrhea prevention on physical growth, anemia, and micronutrient status in rural Laotian children: A randomized controlled trial. J. Pediatr. 2019;207:80–89e82. doi: 10.1016/j.jpeds.2018.11.022.
    1. Barffour MA, et al. Effects of therapeutic zinc supplementation for diarrhea and two preventive zinc supplementation regimens on the incidence and duration of diarrhea and acute respiratory tract infections in rural Laotian children: A randomized controlled trial. J. Glob. Health. 2020;10:010424. doi: 10.7189/jogh.10.010424.
    1. Hess SY, et al. Iron status and inherited haemoglobin disorders modify the effects of micronutrient powders on linear growth and morbidity among young Lao children in a double-blind randomised trial. Br. J. Nutr. 2019;122:895–909. doi: 10.1017/S0007114519001715.
    1. Hinnouho GM, et al. Impact of Two forms of daily preventive zinc or therapeutic zinc supplementation for diarrhea on hair cortisol concentrations among rural Laotian children: A randomized controlled trial. Nutrients. 2018 doi: 10.3390/nu11010047.
    1. Hinnouho GM, et al. Impact of different strategies for delivering supplemental zinc on selected fecal markers of environmental enteric dysfunction among young Laotian children: A randomized controlled trial. Am. J. Trop. Med. Hyg. 2020;103:1416–1426. doi: 10.4269/ajtmh.20-0106.
    1. International Zinc Nutrition Consultative, G. et al. International Zinc Nutrition Consultative Group (IZiNCG) technical document #1. Assessment of the risk of zinc deficiency in populations and options for its control. Food Nutr. Bull.25, S99–S203 (2004).
    1. Garvin, J. H. Principles of Gender-Specific Medicine. 2nd edn. (ed. Legato, M. J.). 51–61 (Academic Press, 2010).
    1. Karlsen TH, et al. Intestinal and systemic immune responses to an oral cholera toxoid B subunit whole-cell vaccine administered during zinc supplementation. Infect. Immun. 2003;71:3909–3913. doi: 10.1128/IAI.71.7.3909-3913.2003.
    1. Albert MJ, et al. Supplementation with zinc, but not vitamin A, improves seroconversion to vibriocidal antibody in children given an oral cholera vaccine. J. Infect. Dis. 2003;187:909–913. doi: 10.1086/368132.
    1. Raqib R, et al. Effect of zinc supplementation on immune and inflammatory responses in pediatric patients with shigellosis. Am. J. Clin. Nutr. 2004;79:444–450. doi: 10.1093/ajcn/79.3.444.
    1. Rahman MJ, et al. Effects of zinc supplementation as adjunct therapy on the systemic immune responses in shigellosis. Am. J. Clin. Nutr. 2005;81:495–502. doi: 10.1093/ajcn.81.2.495.
    1. Kontio M, Jokinen S, Paunio M, Peltola H, Davidkin I. Waning antibody levels and avidity: implications for MMR vaccine-induced protection. J. Infect. Dis. 2012;206:1542–1548. doi: 10.1093/infdis/jis568.
    1. Brokstad KA, Cox RJ, Major D, Wood JM, Haaheim LR. Cross-reaction but no avidity change of the serum antibody response after influenza vaccination. Vaccine. 1995;13:1522–1528. doi: 10.1016/0264-410x(95)00095-i.
    1. Struck F, et al. Incomplete IgG avidity maturation after seasonal coronavirus infections. J. Med. Virol. 2022;94:186–196. doi: 10.1002/jmv.27291.
    1. Re MC, et al. Incomplete IgG response to HIV-1 proteins and low avidity levels in recently converted HIV patients treated with early antiretroviral therapy. Int. J. Infect. Dis. 2010;14:e1008–1012. doi: 10.1016/j.ijid.2010.06.015.
    1. Arias-Bouda LM, et al. Changes in avidity and level of immunoglobulin G antibodies to Mycobacterium tuberculosis in sera of patients undergoing treatment for pulmonary tuberculosis. Clin. Diagn. Lab. Immunol. 2003;10:702–709. doi: 10.1128/cdli.10.4.702-709.2003.
    1. Viant C, et al. Antibody affinity shapes the choice between memory and germinal center B cell fates. Cell. 2020;183:1298–1311.e11. doi: 10.1016/j.cell.2020.09.063.
    1. Killilea DW, Ames BN. Magnesium deficiency accelerates cellular senescence in cultured human fibroblasts. Proc. Natl. Acad. Sci. USA. 2008;105:5768–5773. doi: 10.1073/pnas.0712401105.
    1. Capelli R, et al. BPSL1626: Reverse and structural vaccinology reveal a novel candidate for vaccine design against Burkholderia pseudomallei. Antibodies (Basel) 2018 doi: 10.3390/antib7030026.
    1. Gourlay LJ, et al. Exploiting the Burkholderia pseudomallei acute phase antigen BPSL2765 for structure-based epitope discovery/design in structural vaccinology. Chem. Biol. 2013;20:1147–1156. doi: 10.1016/j.chembiol.2013.07.010.
    1. Nithichanon A, et al. Sequence- and structure-based immunoreactive epitope discovery for Burkholderia pseudomallei flagellin. PLoS Negl. Trop. Dis. 2015;9:e0003917. doi: 10.1371/journal.pntd.0003917.
    1. Saenwongsa W, et al. Metformin-induced suppression of IFN-alpha via mTORC1 signalling following seasonal vaccination is associated with impaired antibody responses in type 2 diabetes. Sci. Rep. 2020;10:3229. doi: 10.1038/s41598-020-60213-0.
    1. Nithichanon A, et al. Boosting of post-exposure human T-cell and B-cell recall responses in vivo by Burkholderia pseudomallei-related proteins. Immunology. 2017;151:98–109. doi: 10.1111/imm.12709.

Source: PubMed

3
Abonneren