Effect of administration of a probiotic preparation on gut microbiota and immune response in healthy women in India: an open-label, single-arm pilot study

Ankita Singh, Aditya N Sarangi, Amit Goel, Rajni Srivastava, Rajat Bhargava, Priyanka Gaur, Amita Aggarwal, Rakesh Aggarwal, Ankita Singh, Aditya N Sarangi, Amit Goel, Rajni Srivastava, Rajat Bhargava, Priyanka Gaur, Amita Aggarwal, Rakesh Aggarwal

Abstract

Background: Probiotics have been shown to be useful for the treatment of many disease conditions. These beneficial effects are believed to be mediated by change in the composition of gut microbiota and modulation of the host immune responses. However, the available data on the effect of probiotics on these parameters are quite limited.

Methods: We studied the composition of fecal microbiota, using 16S rRNA sequencing, and host immune responses in peripheral blood (plasma cytokine levels, T cell subsets and in vitro cytokine production after stimulation with anti-CD3/CD28 antibody or lipopolysaccharide) in a group of 14 healthy women at three time-points - before and after administration of a probiotic preparation (a capsule of VSL#3, each containing 112.5 billion freeze-dried bacterial cells belonging to 8 species, twice a day for 4 weeks), and 4-weeks after discontinuation of the probiotic administration.

Results: There was no change in the abundance of various bacterial taxa as well as in the alpha diversity of gut microbiota following administration of the probiotic, or following its discontinuation. Probiotic administration led to a reduction in the relative frequency of circulating Th17 cells, and in vitro production of cytokines in whole-blood cultures in response to lipopolysaccharide stimulation. However, it had no effect on the relative frequencies of Th1, Th2 and T regulatory cells among circulating peripheral blood mononuclear cells, on plasma cytokine levels and on in vitro production of cytokines by T cells.

Conclusions: We found that VSL#3 administration did not lead to any changes in gut flora, but led to a reduction in the frequency of Th17 cells and in the production of pro-inflammatory cytokine on lipopolysaccharide stimulation. These findings suggest that the beneficial anti-inflammatory effect of this preparation in patients with autoimmune and allergic disorders may be related to reduced production of monocyte-derived cytokines rather than to changes in the composition of gut microbiota.

Trial registration: NCT03330678 , Date of registration 30th October 2017. Retrospectively registered.

Keywords: Gut microbiome; Healthy women; Immune response; Probiotic; VSL#3.

Conflict of interest statement

Ethics approval and consent to participate

The study was approved by Ethics Committee of the Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow. Each participant provided written informed consent.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

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

Figures

Fig. 1
Fig. 1
CONSORT flow diagram
Fig. 2
Fig. 2
Abundances of various bacterial families (a) and genera (b) in fecal microbiota from healthy women before (blue) and after (red) four weeks of probiotic administration. Data are shown using box-plots and percent values on a log10 scale. The ends of boxes represent 25th to 75th centiles, and any dots to the left or right of the boxes indicate outliers
Fig. 3
Fig. 3
Abundances of bacterial families (a) and genera (b) in fecal microbiota from healthy women after four weeks of probiotic administration (red) and 4-weeks after discontinuation of probiotic administration (green). Data are shown using box-plots and percent values on a log10 scale. The ends of boxes represent 25th to 75th centiles, and any dots to the left or right of the boxes indicate outliers
Fig. 4
Fig. 4
Frequencies of Th1 (a), Th2 (b), Treg (c) and Th17 cells (d) in whole blood cultures from healthy women (n = 14) at baseline, after 4-weeks of probiotic administration and 4-weeks after discontinuation of probiotic administration (8 wk) by flow cytometry (Mann-Whitney U test)
Fig. 5
Fig. 5
Estimation of levels of cytokines (pg/ml): IL-10 (a), IL-6 (b) and TNF (c), predominantly produced by monocytes on LPS stimulation of whole blood in culture supernatant of healthy women (n = 14). The estimation was done at three time-points; baseline, 4-weeks of probiotic administration and 4-weeks after discontinuation of probiotics by BD Cytometric Bead Array (Mann-Whitney U test)

References

    1. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett C, Knight R, Gordon JI. The human microbiome project: exploring the microbial part of ourselves in a changing world. Nature. 2007;449:804–810. doi: 10.1038/nature06244.
    1. Vernocchi P, Del Chierico F, Putignani L. Gut microbiota profiling: metabolomics based approach to unravel compounds affecting human health. Front Microbiol. 2016;7:1144. doi: 10.3389/fmicb.2016.01144.
    1. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, et al. A core gut microbiome in obese and lean twins. Nature. 2009;457:480–484. doi: 10.1038/nature07540.
    1. Blandino G, Inturri R, Lazzara F, Di Rosa M, Malaguarnera L. Impact of gut microbiota on diabetes mellitus. Diabetes Metab. 2016;42:303–315. doi: 10.1016/j.diabet.2016.04.004.
    1. Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A. 2007;104:13780–13785. doi: 10.1073/pnas.0706625104.
    1. Bedaiwi MK, Inman RD. Microbiome and probiotics: link to arthritis. Curr Opin Rheumatol. 2014;26:410–415. doi: 10.1097/BOR.0000000000000075.
    1. Chen J, Chia N, Kalari KR, Yao JZ, Novotna M, Soldan MMP, Luckey DH, Marietta EV, et al. Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls. SciRep. 2016;6:28484.
    1. Nadal I, Donat E, Ribes-Koninckx C, Calabuig M, Sanz Y. Imbalance in the composition of the duodenal microbiota of children with coeliac disease. J Med Microbiol. 2007;56:1669–1674. doi: 10.1099/jmm.0.47410-0.
    1. Chen Y, Yang F, Lu H, Wang B, Chen Y, Lei D, Wang Y, Zhu B, et al. Characterization of fecal microbial communities in patients with liver cirrhosis. Hepatology. 2011;54:562–572. doi: 10.1002/hep.24423.
    1. Lee YK, Mazmanian SK. Has the microbiota played a critical role in the evolution of the adaptive immune system? Science. 2010;330:1768–1773. doi: 10.1126/science.1195568.
    1. Round JL, O'Connell RM, Mazmanian SK. Coordination of tolerogenic immune responses by the commensal microbiota. J Autoimmun. 2010;34:J220–J225. doi: 10.1016/j.jaut.2009.11.007.
    1. Preidis GA, Versalovic J. Targeting the human microbiome with antibiotics, probiotics, and prebiotics: gastroenterology enters the metagenomics era. Gastroenterology. 2009;136:2015–2031. doi: 10.1053/j.gastro.2009.01.072.
    1. Gilliland SE, Morelli L, and Reid G. Report of a joint FAO/WHO expert consultation on evaluation of health and nutritional properties of probiotics in food including powder milk with live lactic acid bacteria; 2001 Oct 1–4; Córdoba, Argentina. Available from: .
    1. Tamaki H, Nakase H, Inoue S, Kawanami C, Itani T, Ohana M, Kusaka T, Uose S, Hisatsune H, Tojo M, Noda T, Arasawa S, Izuta M, Kubo A, Ogawa C, Matsunaka T, Shibatouge M. Efficacy of probiotic treatment with Bifidobacterium longum 536 for induction of remission in active ulcerative colitis: a randomized, double-blinded, placebo-controlled multicenter trial. Dig Endosc. 2016;28:67–74. doi: 10.1111/den.12553.
    1. Shukla A, Gaur P, Aggarwal A. Effect of probiotics on clinical and immune parameters in enthesitis-related-arthritis category of juvenile idiopathic arthritis. Clin Expl Immunol. 2016;195:301–308. doi: 10.1111/cei.12818.
    1. Zamani B, Golkar HR, Farshbaf S, Emadi-Baygi M, Tajabadi-Ebrahimi M, Jafari P, Akhavan R, Taghizadeh M, Memarzadeh MR, Asemi Z. Clinical and metabolic response to probiotic supplementation in patients with rheumatoid arthritis: a randomized, double-blind, placebo-controlled trial. Int J Rheum Dis. 2016;19:869–879. doi: 10.1111/1756-185X.12888.
    1. Mangalam A, Shahi SK, Luckey D, Karau M, Marietta E, Luo N, Choung RS, Ju J, Sompallae R, Gibson-Corley K, Patel R, Rodriguez M, David C, Taneja V, Murray J. Human gut-derived commensal Bacteria suppress CNS inflammatory and demyelinating disease. Cell Rep. 2017;20:1269–1277. doi: 10.1016/j.celrep.2017.07.031.
    1. Bartram AK, Lynch MD, Stearns JC, Moreno-Hagelsieb G, Neufeld JD. Generation of multimillion-sequence 16S rRNA gene libraries from complex microbial communities by assembling paired-end illumina reads. Appl Environ Microbiol. 2011;77:3846–3852. doi: 10.1128/AEM.02772-10.
    1. Masella AP, Bartram AK, Truszkowski JM, Brown DG, Neufeld JD. PANDAseq: paired-end assembler for illumina sequences. BMC Bioinformatics. 2012;13:31. doi: 10.1186/1471-2105-13-31.
    1. Kuczynski J, Stombaugh J, Walters WA, González A, Caporaso JG, Knight R. Using QIIME to analyze 16S rRNA gene sequences from microbial communities. Curr Protoc Microbiol 2012;1E. 5.1-1E. 5.20.
    1. Price MN, Dehal PS, Arkin AP. FastTree 2--approximately maximum-likelihood trees for large alignments. PLoS One. 2010;5:e9490. doi: 10.1371/journal.pone.0009490.
    1. McMurdie PJ, Holmes S. Phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One. 2013;8:e61217. doi: 10.1371/journal.pone.0061217.
    1. Gotelli NJ, Chao A. Measuring and estimating species richness, species diversity, and biotic similarity from sampling data. In: Levin S, editor. Encyclopedia of biodiversity. 2. 2013. pp. 195–211.
    1. Bercovici N, Duffour MT, Agrawal S, Salcedo M, Abastado JP. New methods for assessing T-cell responses. Clin Diagn Lab Immunol. 2000;7:859–864.
    1. Gerards AH, de Lathouder S, de Groot ER, Dijkmans BA, Aarden LA. Inhibition of cytokine production by methotrexate. Studies in healthy volunteers and patients with rheumatoid arthritis. Rheumatology (Oxford) 2003;42:1189–1196. doi: 10.1093/rheumatology/keg323.
    1. Stearns JC, Lynch MD, Senadheera DB, Tenenbaum HC, Goldberg MB, Cvitkovitch DG, Croitoru K, Moreno-Hagelsieb G, et al. Bacterial biogeography of the human digestive tract. Sci Rep. 2011;1:170. doi: 10.1038/srep00170.
    1. Lin A, Bik EM, Costello EK, Dethlefsen L, Haque R, Relman DA, Singh U. Distinct distal gut microbiome diversity and composition in healthy children from Bangladesh and the United States. PLoS One. 2013;8:e53838. doi: 10.1371/journal.pone.0053838.
    1. Nam YD, Jung MJ, Roh SW, Kim MS, Bae JW. Comparative analysis of Korean human gut microbiota by barcoded pyrosequencing. PLoS One. 2011;6:e22109. doi: 10.1371/journal.pone.0022109.
    1. Bhute S, Pande P, Shetty SA, Shelar R, Mane S, Kumbhare SV, Gawali A, Makhani H, et al. Molecular characterization and meta-analysis of gut microbial communities illustrate enrichment of prevotella and megasphaera in Indian subjects. Front Microbiol. 2016;7:660. doi: 10.3389/fmicb.2016.00660.
    1. McFarland LV. Use of probiotics to correct dysbiosis of normal microbiota following disease or disruptive events: a systematic review. BMJ Open. 2014;4:e005047. doi: 10.1136/bmjopen-2014-005047.
    1. Bibiloni R, Fedorak RN, Tannock GW, Madsen KL, Gionchetti P, Campieri M, De Simone C, Sartor RB. VSL# 3 probiotic-mixture induces remission in patients with active ulcerative colitis. Am J Gastroenterol. 2005;100:1539–1546. doi: 10.1111/j.1572-0241.2005.41794.x.
    1. Kuehbacher T, Ott SJ, Helwig U, Mimura T, Rizzello F, Kleessen B, Gionchetti P, Blaut M, et al. Bacterial and fungal microbiota in relation to probiotic therapy (VSL# 3) in pouchitis. Gut. 2006;55:833–841. doi: 10.1136/gut.2005.078303.
    1. Ng SC, Lam EF, Lam TT, Chan Y, Law W, Tse PC, Kamm MA, Sung JJ, et al. Effect of probiotic bacteria on the intestinal microbiota in irritable bowel syndrome. J Gastroenterol Hepatol. 2013;28:1624–1631.
    1. Valentini L, Pinto A, Bourdel-Marchasson I, Ostan R, Brigidi P, Turroni S, Hrelia S, Hrelia P, et al. Impact of personalized diet and probiotic supplementation on inflammation, nutritional parameters and intestinal microbiota - the "RISTOMED project": randomized controlled trial in healthy older people. Clin Nutr. 2015;34:593–602. doi: 10.1016/j.clnu.2014.09.023.
    1. Aggarwal A, Sarangi AN, Gaur P, Shukla A, Aggarwal R. Gut microbiome in children with enthesitis-related arthritis in a developing country and the effect of probiotic administration. Clin Exp Immunol. 2017;187:480–489. doi: 10.1111/cei.12900.
    1. Harbige LS, Pinto E, Allgrove J, Thomas LV. Immune response of healthy adults to the ingested probiotic Lactobacillus casei Shirota. Scand J Immunol. 2016;84:353–364. doi: 10.1111/sji.12495.
    1. Schultz M, Linde HJ, Lehn N, Zimmermann K, Grossmann J, Falk W, Scholmerich J. Immunomodulatory consequences of oral administration of Lactobacillus rhamnosus strain GG in healthy volunteers. J Dairy Res. 2003;70:165–173. doi: 10.1017/S0022029903006034.
    1. Kekkonen RA, Lummela N, Karjalainen H, Latvala S, Tynkkynen S, Jarvenpaa S, Kautiainen H, Julkunen I, et al. Probiotic intervention has strain-specific anti-inflammatory effects in healthy adults. World J Gastroenterol. 2008;14:2029. doi: 10.3748/wjg.14.2029.
    1. Meyer AL, Micksche M, Herbacek I, Elmadfa I. Daily intake of probiotic as well as conventional yogurt has a stimulating effect on cellular immunity in young healthy women. Ann Nutri Metab. 2006;50:282–289. doi: 10.1159/000091687.
    1. Hart AL, Lammers K, Brigidi P, Vitali B, Rizzello F, Gionchetti P, Campieri M, Kamm MA, et al. Modulation of human dendritic cell phenotype and function by probiotic bacteria. Gut. 2004;53:1602–1609. doi: 10.1136/gut.2003.037325.
    1. Latvala S, Miettinen M, Kekkonen RA, Korpela R, Julkunen I. Lactobacillus rhamnosus GG and Streptococcus thermophilus induce suppressor of cytokine signalling 3 (SOCS3) gene expression directly and indirectly via interleukin-10 in human primary macrophages. Clin Exp Immunol. 2011;165:94–103. doi: 10.1111/j.1365-2249.2011.04408.x.
    1. Dong H, Rowland I, Tuohy KM, Thomas LV, Yaqoob P. Selective effects of Lactobacillus casei Shirota on T cell activation, natural killer cell activity and cytokine production. Clin Exp Immunol. 2010;161:378–388.
    1. Shadnoush M, Shaker Hosseini R, Mehrabi Y, Delpisheh A, Alipoor E, Faghfoori Z, Mohammadpour N, Zaringhalam Moghadam J. Probiotic yogurt affects pro- and anti-inflammatory factors in patients with inflammatory bowel disease. Iran J Pharm Res. 2013;12:929–936.
    1. Olivares M, Paz Díaz-Ropero M, Gómez N, Sierra S, Lara-Villoslada F, Martín R, Miguel Rodríguez J, Xaus J. Dietary deprivation of fermented foods causes a fall in innate immune response. Lactic acid bacteria can counteract the immunological effect of this deprivation. J Dairy Res. 2006;73:492–498. doi: 10.1017/S0022029906002068.
    1. Christensen HR, Frokiaer H, Pestka JJ. Lactobacilli differentially modulate expression of cytokines and maturation surface markers in murine dendritic cells. J Immunol. 2002;168:171–178. doi: 10.4049/jimmunol.168.1.171.
    1. Schiavi E, Barletta B, Butteroni C, Corinti S, Boirivant M, Di Felice G. Oral therapeutic administration of a probiotic mixture suppresses established Th2 responses and systemic anaphylaxis in a murine model of food allergy. Allergy. 2011;66:499–508. doi: 10.1111/j.1398-9995.2010.02501.x.
    1. Chen L, Zou Y, Peng J, Lu F, Yin Y, Li F, Yang J. Lactobacillus acidophilus suppresses colitis-associated activation of the IL-23/Th17 axis. J Immunol Res 2015; 2015:909514.
    1. Sarra M, Pallone F, Macdonald TT, Monteleone G. IL-23/IL-17 axis in IBD. Inflamm Bowel Dis. 2010;16:1808–1813. doi: 10.1002/ibd.21248.
    1. Zuo L, Yuan KT, Yu L, Meng QH, Chung PC, Yang DH. Bifidobacterium infantis attenuates colitis by regulating T cell subset responses. World J Gastroenterol. 2014;20:18316–18329. doi: 10.3748/wjg.v20.i48.18316.
    1. Cortes-Perez NG, Lozano-Ojalvo D, Maiga MA, Hazebrouck S, Adel-Patient K. Intragastric administration of Lactobacillus casei BL23 induces regulatory FoxP3+RORgammat+ T cells subset in mice. Benef Microbes. 2017;8:433–438. doi: 10.3920/BM2016.0174.

Source: PubMed

3
订阅