Bifidobacterium animalis subsp lactis HN019 presents antimicrobial potential against periodontopathogens and modulates the immunological response of oral mucosa in periodontitis patients

Marcos M Invernici, Flávia A C Furlaneto, Sérgio L Salvador, Arthur C Ouwehand, Seppo Salminen, Anastasia Mantziari, Gabriel Vinderola, Edilson Ervolino, Sandro Isaías Santana, Pedro Henrique Felix Silva, Michel R Messora, Marcos M Invernici, Flávia A C Furlaneto, Sérgio L Salvador, Arthur C Ouwehand, Seppo Salminen, Anastasia Mantziari, Gabriel Vinderola, Edilson Ervolino, Sandro Isaías Santana, Pedro Henrique Felix Silva, Michel R Messora

Abstract

Objective: To evaluate the effects of Bifidobacterium animalis subsp. lactis HN019 (HN019) on clinical periodontal parameters (plaque accumulation and gingival bleeding), on immunocompetence of gingival tissues [expression of beta-defensin (BD)-3, toll-like receptor 4 (TLR4), cluster of differentiation(CD)-57 and CD-4], and on immunological properties of saliva (IgA levels) in non-surgical periodontal therapy in generalized chronic periodontitis (GCP) patients. Adhesion to buccal epithelial cells (BEC) and the antimicrobial properties of HN019 were also investigated.

Materials and methods: Thirty patients were recruited and monitored clinically at baseline (before scaling and root planing-SRP) and after 30 and 90 days. Patients were randomly assigned to Test (SRP+Probiotic, n = 15) or Control (SRP+Placebo, n = 15) group. Probiotic lozenges were used for 30 days. Gingival tissues and saliva were immunologically analyzed. The adhesion of HN019 with or without Porphyromonas gingivalis in BEC and its antimicrobial properties were investigated in in vitro assays. Data were statistically analyzed (p<0.05).

Results: Test group presented lower plaque index (30 days) and lower marginal gingival bleeding (90 days) when compared with Control group. Higher BD-3, TLR4 and CD-4 expressions were observed in gingival tissues in Test group than in Control group. HN019 reduced the adhesion of P. gingivalis to BEC and showed antimicrobial potential against periodontopathogens.

Conclusion: Immunological and antimicrobial properties of B. lactis HN019 make it a potential probiotic to be used in non-surgical periodontal therapy of patients with GCP.

Clinical relevance: B. lactis HN019 may be a potential probiotic to improve the effects of non-surgical periodontal therapy. Name of the registry and registration number (ClinicalTrials.gov): "Effects of probiotic therapy in the treatment of periodontitis"-NCT03408548.

Conflict of interest statement

E.I DuPont de Nemours & Co. Danisco Sweeteners Oy provided financial support in the form of a salary for ACO and donated probiotics. There are no patents, products in development or marketed products to declare. This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Flowchart of the study design.
Fig 1. Flowchart of the study design.
Fig 2
Fig 2
IgA levels (A) of Test and Control groups at baseline and at 30 and 90 postoperative days. Mean changes in IgA levels relative to baseline values obtained for the Test and Control groups are shown in (B). Ratio of IgA levels = changes in IgA levels in relation to baseline values. n.s* No significant changes were observed in IgA levels at both 30 and 90 days, compared to baseline, for Test and Control groups. n.s** No significant differences between Test and Control groups at both 30 and 90 days.
Fig 3. Relative frequency values for BD-3…
Fig 3. Relative frequency values for BD-3 and TLR4 immunolabeling scores in Test and Control groups at healthy and diseased sites at baseline and at 30 postoperative days, as well as within-group and between-group comparison results.
*Significant difference (Kruskal-Wallis, Dunn, p<0.05). *Significant difference when compared to Test baseline and Control baseline. **Significant difference when compared to Test baseline. §Significant difference when compared to Control baseline. ¶Significant difference when compared to Test baseline. #Significant difference when compared to Control 30 days.
Fig 4. Relative frequency values for CD-57…
Fig 4. Relative frequency values for CD-57 and CD-4 immunolabeling scores for test and control groups at healthy and diseased sites at baseline and at 30 postoperative days, as well as within-group and between-group comparison results.
*Significant difference (Kruskal-Wallis, Dunn, p

Fig 5. Photomicrographs showing BD-3 (A-H) and…

Fig 5. Photomicrographs showing BD-3 (A-H) and TLR4 (I-P) immunolabeling in gingival biopsies at baseline…

Fig 5. Photomicrographs showing BD-3 (A-H) and TLR4 (I-P) immunolabeling in gingival biopsies at baseline (A, C, E, G, I, K, M, O) and 30 days (B, D, F, H, J, L, N, P) from healthy (A-D, I-L) and diseased (E-H, M-P) sites in Test (A, B, E, F, I, J, M, N) and Control (C, D, G, H, K, L, O, P) groups.
Arrows = BD-3 and TLR4-positive cells; ep = Epithelial tissue; ct = Connective tissue. Scale = 25 μm. Counterstaining: Hematoxylin.

Fig 6. Photomicrographs showing CD-57 (A-H) and…

Fig 6. Photomicrographs showing CD-57 (A-H) and CD-4 (I-P) immunolabeling in gingival biopsies (connective tissue)…

Fig 6. Photomicrographs showing CD-57 (A-H) and CD-4 (I-P) immunolabeling in gingival biopsies (connective tissue) at baseline (A, C, E, G, I, K, M, O) and 30 days (B, D, F, H, J, L, N, P) from healthy (A-D, I-L) and diseased (E-H, M-P) sites in Test (A, B, E, F, I, J, M, N) and Control (C, D, G, H, K, L, O, P) groups.
Arrows = CD-57 and CD-4 positive cells. Scale = 25 μm. Counterstaining: Hematoxylin.

Fig 7. Photomicrographs showing buccal epithelial cells…

Fig 7. Photomicrographs showing buccal epithelial cells in adhesion assays using crystal violet and safranin.

Fig 7. Photomicrographs showing buccal epithelial cells in adhesion assays using crystal violet and safranin.
(A) P. gingivalis alone attached in a buccal cell (original magnification x100). (B)–B. lactis HN019 alone attached in a buccal cell (original magnification x100). (C)–Mixture of B. lactis HN019 with P. gingivalis attached in a buccal cell (original magnification x100). Red arrows indicate P. gingivalis (Gram-negative bacteria with shorter rods or coccobacilli morphology). Blue arrows indicate Bifidobacterium (Gram-positive bacteria with long bacilli morphology).

Fig 8. Growth inhibition of Aggregatibacter actinomycetemcomitans…

Fig 8. Growth inhibition of Aggregatibacter actinomycetemcomitans , Porphyromonas gingivalis , Prevotella intermedia , and…

Fig 8. Growth inhibition of Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis, Prevotella intermedia, and Fusobacterium nucleatum by Bifidobacterium animalis subs. Lactis HN019 (agar diffusion method).
(A) Clear areas around the wells indicate zones of inhibition. (B) Means and standard deviations of the zone of inhibition observed in sensitivity of different periodontopathogens to B. lactis HN019.
All figures (8)
Fig 5. Photomicrographs showing BD-3 (A-H) and…
Fig 5. Photomicrographs showing BD-3 (A-H) and TLR4 (I-P) immunolabeling in gingival biopsies at baseline (A, C, E, G, I, K, M, O) and 30 days (B, D, F, H, J, L, N, P) from healthy (A-D, I-L) and diseased (E-H, M-P) sites in Test (A, B, E, F, I, J, M, N) and Control (C, D, G, H, K, L, O, P) groups.
Arrows = BD-3 and TLR4-positive cells; ep = Epithelial tissue; ct = Connective tissue. Scale = 25 μm. Counterstaining: Hematoxylin.
Fig 6. Photomicrographs showing CD-57 (A-H) and…
Fig 6. Photomicrographs showing CD-57 (A-H) and CD-4 (I-P) immunolabeling in gingival biopsies (connective tissue) at baseline (A, C, E, G, I, K, M, O) and 30 days (B, D, F, H, J, L, N, P) from healthy (A-D, I-L) and diseased (E-H, M-P) sites in Test (A, B, E, F, I, J, M, N) and Control (C, D, G, H, K, L, O, P) groups.
Arrows = CD-57 and CD-4 positive cells. Scale = 25 μm. Counterstaining: Hematoxylin.
Fig 7. Photomicrographs showing buccal epithelial cells…
Fig 7. Photomicrographs showing buccal epithelial cells in adhesion assays using crystal violet and safranin.
(A) P. gingivalis alone attached in a buccal cell (original magnification x100). (B)–B. lactis HN019 alone attached in a buccal cell (original magnification x100). (C)–Mixture of B. lactis HN019 with P. gingivalis attached in a buccal cell (original magnification x100). Red arrows indicate P. gingivalis (Gram-negative bacteria with shorter rods or coccobacilli morphology). Blue arrows indicate Bifidobacterium (Gram-positive bacteria with long bacilli morphology).
Fig 8. Growth inhibition of Aggregatibacter actinomycetemcomitans…
Fig 8. Growth inhibition of Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis, Prevotella intermedia, and Fusobacterium nucleatum by Bifidobacterium animalis subs. Lactis HN019 (agar diffusion method).
(A) Clear areas around the wells indicate zones of inhibition. (B) Means and standard deviations of the zone of inhibition observed in sensitivity of different periodontopathogens to B. lactis HN019.

References

    1. Marsh PD (2003) Are dental diseases examples of ecological catastrophes? Microbiology 149 (Pt 2):279–294. 10.1099/mic.0.26082-0
    1. Ximenez-Fyvie LA, Haffajee AD, Som S, Thompson M, Torresyap G, Socransky SS (2000) The effect of repeated professional supragingival plaque removal on the composition of the supra- and subgingival microbiota. Journal of clinical periodontology 27 (9):637–647 10.1034/j.1600-051x.2000.027009637.x
    1. Shibli JA, Melo L, Ferrari DS, Figueiredo LC, Faveri M, Feres M (2008) Composition of supra- and subgingival biofilm of subjects with healthy and diseased implants. Clinical oral implants research 19 (10):975–982. 10.1111/j.1600-0501.2008.01566.x
    1. Haffajee AD, Socransky SS, Patel MR, Song X (2008) Microbial complexes in supragingival plaque. Oral microbiology and immunology 23 (3):196–205. 10.1111/j.1399-302X.2007.00411.x
    1. Sanz-Sanchez I, Ortiz-Vigon A, Herrera D, Sanz M (2016) Microbiological effects and recolonization patterns after adjunctive subgingival debridement with Er:YAG laser. Clinical oral investigations 20 (6):1253–1261. 10.1007/s00784-015-1617-y
    1. Lindhe J, Westfelt E, Nyman S, Socransky SS, Heijl L, Bratthall G (1982) Healing following surgical/non-surgical treatment of periodontal disease. A clinical study. Journal of clinical periodontology 9 (2):115–128 10.1111/j.1600-051x.1982.tb01227.x
    1. Magnusson I, Lindhe J, Yoneyama T, Liljenberg B (1984) Recolonization of a subgingival microbiota following scaling in deep pockets. Journal of clinical periodontology 11 (3):193–207 10.1111/j.1600-051x.1984.tb01323.x
    1. Mark Bartold P, Van Dyke TE (2017). Host modulation: controlling the inflammation to control the infection. Periodontology 2000 75 (1):317‐329.
    1. FAOlWHO J (2002) Working Group Report on Drafting Guidelines for the Evaluation of Probiotics in Food London. Ontario, Canada
    1. de Vrese M, Schrezenmeir J (2008) Probiotics, prebiotics, and synbiotics. Advances in biochemical engineering/biotechnology 111:1–66. 10.1007/10_2008_097
    1. Teughels W, Durukan A, Ozcelik O, Pauwels M, Quirynen M, Haytac MC (2013) Clinical and microbiological effects of Lactobacillus reuteri probiotics in the treatment of chronic periodontitis: a randomized placebo-controlled study. Journal of clinical periodontology 40 (11):1025–1035. 10.1111/jcpe.12155
    1. Krasse P, Carlsson B, Dahl C, Paulsson A, Nilsson A, Sinkiewicz G (2006) Decreased gum bleeding and reduced gingivitis by the probiotic Lactobacillus reuteri. Swedish dental journal 30 (2):55–60
    1. Riccia DN, Bizzini F, Perilli MG, Polimeni A, Trinchieri V, Amicosante G, et al. (2007) Anti-inflammatory effects of Lactobacillus brevis (CD2) on periodontal disease. Oral diseases 13 (4):376–385. 10.1111/j.1601-0825.2006.01291.x
    1. Twetman S, Derawi B, Keller M, Ekstrand K, Yucel-Lindberg T, Stecksen-Blicks C (2009) Short-term effect of chewing gums containing probiotic Lactobacillus reuteri on the levels of inflammatory mediators in gingival crevicular fluid. Acta odontologica Scandinavica 67 (1):19–24. 10.1080/00016350802516170
    1. Stensson M, Koch G, Coric S, Abrahamsson TR, Jenmalm MC, Birkhed D, et al. (2014) Oral administration of Lactobacillus reuteri during the first year of life reduces caries prevalence in the primary dentition at 9 years of age. Caries research 48 (2):111–117. 10.1159/000354412
    1. Tekce M, Ince G, Gursoy H, Dirikan Ipci S, Cakar G, Kadir T, et al. (2015) Clinical and microbiological effects of probiotic lozenges in the treatment of chronic periodontitis: a 1-year follow-up study. Journal of clinical periodontology 42 (4):363–372. 10.1111/jcpe.12387
    1. Ince G, Gursoy H, Ipci SD, Cakar G, Emekli-Alturfan E, Yilmaz S (2015) Clinical and Biochemical Evaluation of Lozenges Containing Lactobacillus reuteri as an Adjunct to Non-Surgical Periodontal Therapy in Chronic Periodontitis. Journal of periodontology 86 (6):746–754. 10.1902/jop.2015.140612
    1. Szkaradkiewicz AK, Stopa J, Karpiński TM (2014) Effect of oral administration involving a probiotic strain of Lactobacillus reuteri on pro-inflammatory cytokine response in patients with chronic periodontitis. Archivum immunologiae et therapiae experimentalis 62 (6):495–500. 10.1007/s00005-014-0277-y
    1. Kuru BE, Laleman I, Yalnizoglu T, Kuru L, Teughels W (2017) The Influence of a Bifidobacterium animalis Probiotic on Gingival Health: A Randomized Controlled Clinical Trial. Journal of periodontology 88 (11):1115–1123. 10.1902/jop.2017.170213
    1. Penala S, Kalakonda B, Pathakota KR, Jayakumar A, Koppolu P, Lakshmi BV, et al. (2016) Efficacy of local use of probiotics as an adjunct to scaling and root planing in chronic periodontitis and halitosis: A randomized controlled trial. Journal of research in pharmacy practice 5 (2):86–93. 10.4103/2279-042X.179568
    1. Nissen L, Sgorbati B, Biavati B, Belibasakis GN (2014) Lactobacillus salivarius and L. gasseri down-regulate Aggregatibacter actinomycetemcomitans exotoxins expression. Annals of microbiology 64:611–617. 10.1007/s13213-013-0694-x
    1. Haukioja A, Yli-Knuuttila H, Loimaranta V, Kari K, Ouwehand AC, Meurman JH, et al. (2006) Oral adhesion and survival of probiotic and other lactobacilli and bifidobacteria in vitro. Oral microbiology and immunology 21 (5):326–332. 10.1111/j.1399-302X.2006.00299.x
    1. Corthesy B, Gaskins HR, Mercenier A (2007) Cross-talk between probiotic bacteria and the host immune system. The Journal of nutrition 137 (3 Suppl 2):781S–790S 10.1093/jn/137.3.781S
    1. Menard S, Candalh C, Bambou JC, Terpend K, Cerf-Bensussan N, Heyman M (2004) Lactic acid bacteria secrete metabolites retaining anti-inflammatory properties after intestinal transport. Gut 53 (6):821–828 10.1136/gut.2003.026252
    1. Wilson M (2005) Microbial inhabitants of humans: their ecology and role in health and disease. Cambridge University Press.
    1. Becker MR, Paster BJ, Leys EJ, Moeschberger ML, Kenyon SG, Galvin JL, et al. (2002) Molecular analysis of bacterial species associated with childhood caries. Journal of clinical microbiology 40 (3):1001–1009 10.1128/jcm.40.3.1001-1009.2002
    1. Fuller R, Gibson GR (1997) Modification of the intestinal microflora using probiotics and prebiotics. Scandinavian journal of gastroenterology Supplement 222:28–31. 10.1080/00365521.1997.11720714
    1. Ricoldi MST, Furlaneto FAC, Oliveira LFF, Teixeira GC, Pischiotini JP, Moreira ALG, et al. (2017) Effects of the probiotic Bifidobacterium animalis subsp. lactis on the non-surgical treatment of periodontitis. A histomorphometric, microtomographic and immunohistochemical study in rats. PloS one 12 (6):e0179946 10.1371/journal.pone.0179946
    1. Oliveira LF, Salvador SL, Silva PH, Furlaneto FA, Figueiredo L, Casarin R, et al. (2017) Benefits of Bifidobacterium animalis subsp. lactis Probiotic in Experimental Periodontitis. Journal of periodontology 88 (2):197–208. 10.1902/jop.2016.160217
    1. Invernici MM, Salvador SL, Silva PHF, Soares MSM, Casarin R, Palioto DB, et al. (2018) Effects of Bifidobacterium probiotic on the treatment of chronic periodontitis: A randomized clinical trial. Journal of clinical periodontology 45 (10):1198–1210. 10.1111/jcpe.12995
    1. Armitage GC (1999) Development of a classification system for periodontal diseases and conditions. Annals of periodontology 4 (1):1–6. 10.1902/annals.1999.4.1.1
    1. Zhu Y, Xiao L, Shen D, Hao Y (2010) Competition between yogurt probiotics and periodontal pathogens in vitro. Acta odontologica Scandinavica 68 (5):261–268. 10.3109/00016357.2010.492235
    1. Jasberg H, Soderling E, Endo A, Beighton D, Haukioja A (2016) Bifidobacteria inhibit the growth of Porphyromonas gingivalis but not of Streptococcus mutans in an in vitro biofilm model. European journal of oral sciences 124 (3):251–258. 10.1111/eos.12266
    1. Rosier BT, Marsh PD, Mira A (2017) Resilience of the Oral Microbiota in Health: Mechanisms That Prevent Dysbiosis. Journal of dental research:22034517742139. 10.1177/0022034517742139
    1. Lee JK, Kim SJ, Ko SH, Ouwehand AC, Ma DS (2015) Modulation of the host response by probiotic Lactobacillus brevis CD2 in experimental gingivitis. Oral diseases 21 (6):705–712. 10.1111/odi.12332
    1. Mendi A, Kose S, Uckan D, Akca G, Yilmaz D, Aral L, et al. (2016) Lactobacillus rhamnosus could inhibit Porphyromonas gingivalis derived CXCL8 attenuation. Journal of applied oral science: revista FOB 24 (1):67–75. 10.1590/1678-775720150145
    1. Wang P, Duan D, Zhou X, Li X, Yang J, Deng M, et al. (2015) Relationship between expression of human gingival beta-defensins and levels of periodontopathogens in subgingival plaque. Journal of periodontal research 50 (1):113–122. 10.1111/jre.12187
    1. Krisanaprakornkit S, Kimball JR, Weinberg A, Darveau RP, Bainbridge BW, Dale BA (2000) Inducible expression of human beta-defensin 2 by Fusobacterium nucleatum in oral epithelial cells: multiple signaling pathways and role of commensal bacteria in innate immunity and the epithelial barrier. Infection and immunity 68 (5):2907–2915 10.1128/iai.68.5.2907-2915.2000
    1. Harder J, Bartels J, Christophers E, Schroder JM (2001) Isolation and characterization of human beta -defensin-3, a novel human inducible peptide antibiotic. The Journal of biological chemistry 276 (8):5707–5713. 10.1074/jbc.M008557200
    1. Jia HP, Schutte BC, Schudy A, Linzmeier R, Guthmiller JM, Johnson GK, et al. (2001) Discovery of new human beta-defensins using a genomics-based approach. Gene 263 (1–2):211–218 10.1016/s0378-1119(00)00569-2
    1. Vardar-Sengul S, Demirci T, Sen BH, Erkizan V, Kurulgan E, Baylas H (2007) Human beta defensin-1 and -2 expression in the gingiva of patients with specific periodontal diseases. Journal of periodontal research 42 (5):429–437. 10.1111/j.1600-0765.2006.00964.x
    1. Bissell J, Joly S, Johnson GK, Organ CC, Dawson D, McCray PB Jr., et al. (2004) Expression of beta-defensins in gingival health and in periodontal disease. Journal of oral pathology & medicine: official publication of the International Association of Oral Pathologists and the American Academy of Oral Pathology 33 (5):278–285. 10.1111/j.0904-2512.2004.00143.x
    1. Kuula H, Salo T, Pirila E, Hagstrom J, Luomanen M, Gutierrez-Fernandez A, et al. (2008) Human beta-defensin-1 and -2 and matrix metalloproteinase-25 and -26 expression in chronic and aggressive periodontitis and in peri-implantitis. Archives of oral biology 53 (2):175–186. 10.1016/j.archoralbio.2007.09.010
    1. Medzhitov R, Preston-Hurlburt P, Janeway CA Jr. (1997) A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature 388 (6640):394–397. 10.1038/41131
    1. Castillo NA, Perdigon G, de Moreno de Leblanc A (2011) Oral administration of a probiotic Lactobacillus modulates cytokine production and TLR expression improving the immune response against Salmonella enterica serovar Typhimurium infection in mice. BMC microbiology 11:177 10.1186/1471-2180-11-177
    1. Yokoyama WM, Kim S, French AR (2004) The dynamic life of natural killer cells. Annual review of immunology 22:405–29. 10.1146/annurev.immunol.22.012703.104711
    1. Kopp W (1988) Density and localization of lymphocytes with natural-killer (NK) cell activity in periodontal biopsy specimens from patients with severe periodontitis. Journal of clinical periodontology 15 (10):595–600. 10.1111/j.1600-051x.1988.tb02257.x
    1. Wynne SE, Walsh LJ, Seymour GJ, Powell RN (1996) In situ demonstration of natural killer (NK) cells in human gingival tissue. Journal of periodontology 57 (11):699–702.
    1. Fujita S, Takahashi H, Okabe H, Ozaki Y, Hara Y, Kato I (1992) Distribution of natural killer cells in periodontal diseases: an immunohistochemical study. Journal of periodontology 63 (8):686–9. 10.1902/jop.1992.63.8.686
    1. Stelin S, Ramakrishan H, Talwar A, Arun KV, Kumar TS (2009) Immunohistological analysis of CD1a langerhans cells and CD57 natural killer cells in healthy and diseased human gingival tissue: A comparative study. Journal of indian society of periodontology 13 (3):150–154. 10.4103/0972-124X.60228
    1. Cobb CM, Singla O, Feil PH, Theisen FC, Schultz RE (1989) Comparison of NK-cell (Leu-7+ and Leu-11b+) populations in clinically healthy gingiva, chronic gingivitis and chronic adult periodontitis. Journal of periodontal research 24 (1):1–7. 10.1111/j.1600-0765.1989.tb00851.x
    1. Palomar MM, Maldonado Galdeano C, Perdigón G (2014) Influence of a probiotic lactobacillus strain on the intestinal ecosystem in a stress model mouse. Brain, behavior, and immunity 35:77–85. 10.1016/j.bbi.2013.08.015
    1. Miller H, Ferris R, Phelps BR (2016) The effect of probiotics on CD4 counts among people living with HIV: a systematic review. Beneficial microbes 7 (3):345–351. 10.3920/BM2015.0163
    1. Romagnani S (1997) The Th1/Th2 paradigm. Immunology today 8 (6):263–266.
    1. Patil PB, Patil BR (2011) Saliva: A diagnostic biomarker of periodontal diseases. Journal of Indian Society of Periodontology 15 (4):310 10.4103/0972-124X.92560
    1. Jorgensen MR, Keller MK, Kragelund C, Hamberg K, Ericson D, Nielsen CH, et al. (2016) Lactobacillus reuteri supplements do not affect salivary IgA or cytokine levels in healthy subjects: A randomized, double-blind, placebo-controlled, cross-over trial. Acta odontologica Scandinavica 74 (5):399–404. 10.3109/00016357.2016.1169439
    1. Paineau D, Carcano D, Leyer G, Darquy S, Alyanakian MA, Simoneau G, et al. (2008) Effects of seven potential probiotic strains on specific immune responses in healthy adults: a double-blind, randomized, controlled trial. FEMS immunology and medical microbiology 53 (1):107–113. 10.1111/j.1574-695X.2008.00413.x
    1. Gleeson M, Bishop NC, Oliveira M, McCauley T, Tauler P, Lawrence C (2012) Effects of a Lactobacillus salivarius probiotic intervention on infection, cold symptom duration and severity, and mucosal immunity in endurance athletes. International journal of sport nutrition and exercise metabolism 22 (4):235–242 10.1123/ijsnem.22.4.235
    1. Kekkonen RA, Lummela N, Karjalainen H, Latvala S, Tynkkynen S, Jarvenpaa S, et al. (2008) Probiotic intervention has strain-specific anti-inflammatory effects in healthy adults. World journal of gastroenterology 14 (13):2029–2036 10.3748/wjg.14.2029
    1. Tiollier E, Chennaoui M, Gomez-Merino D, Drogou C, Filaire E, Guezennec CY (2007) Effect of a probiotics supplementation on respiratory infections and immune and hormonal parameters during intense military training. Military medicine 172 (9):1006–1011 10.7205/milmed.172.9.1006
    1. Dong H, Rowland I, Thomas LV, Yaqoob P (2013) Immunomodulatory effects of a probiotic drink containing Lactobacillus casei Shirota in healthy older volunteers. European journal of nutrition 52 (8):1853–1863. 10.1007/s00394-012-0487-1
    1. Surono IS, Koestomo FP, Novitasari N, Zakaria FR, Yulianasari, Koesnandar (2011) Novel probiotic Enterococcus faecium IS-27526 supplementation increased total salivary sIgA level and bodyweight of pre-school children: a pilot study. Anaerobe 17 (6):496–500. 10.1016/j.anaerobe.2011.06.003
    1. Rizzardini G, Eskesen D, Calder PC, Capetti A, Jespersen L, Clerici M (2012) Evaluation of the immune benefits of two probiotic strains Bifidobacterium animalis ssp. lactis, BB-12(R) and Lactobacillus paracasei ssp. paracasei, L. casei 431(R) in an influenza vaccination model: a randomised, double-blind, placebo-controlled study. The British journal of nutrition 107 (6):876–884. 10.1017/S000711451100420X
    1. Shimizu K, Sato H, Suga Y, Yamahira S, Toba M, Hamuro K, et al. (2014) The effects of Lactobacillus pentosus strain b240 and appropriate physical training on salivary secretory IgA levels in elderly adults with low physical fitness: a randomized, double-blind, placebo-controlled trial. Journal of clinical biochemistry and nutrition 54 (1):61–66. 10.3164/jcbn.13-62
    1. Kotani Y, Shinkai S, Okamatsu H, Toba M, Ogawa K, Yoshida H, et al. (2010) Oral intake of Lactobacillus pentosus strain b240 accelerates salivary immunoglobulin A secretion in the elderly: A randomized, placebo-controlled, double-blind trial. Immunity & ageing: I & A 7:11 10.1186/1742-4933-7-11
    1. Asama T, Arima TH, Gomi T, Keishi T, Tani H, Kimura Y, et al. (2015) Lactobacillus kunkeei YB38 from honeybee products enhances IgA production in healthy adults. Journal of applied microbiology 119 (3):818–826. 10.1111/jam.12889

Source: PubMed

3
Abonner