The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics

Seppo Salminen, Maria Carmen Collado, Akihito Endo, Colin Hill, Sarah Lebeer, Eamonn M M Quigley, Mary Ellen Sanders, Raanan Shamir, Jonathan R Swann, Hania Szajewska, Gabriel Vinderola, Seppo Salminen, Maria Carmen Collado, Akihito Endo, Colin Hill, Sarah Lebeer, Eamonn M M Quigley, Mary Ellen Sanders, Raanan Shamir, Jonathan R Swann, Hania Szajewska, Gabriel Vinderola

Abstract

In 2019, the International Scientific Association for Probiotics and Prebiotics (ISAPP) convened a panel of experts specializing in nutrition, microbial physiology, gastroenterology, paediatrics, food science and microbiology to review the definition and scope of postbiotics. The term 'postbiotics' is increasingly found in the scientific literature and on commercial products, yet is inconsistently used and lacks a clear definition. The purpose of this panel was to consider the scientific, commercial and regulatory parameters encompassing this emerging term, propose a useful definition and thereby establish a foundation for future developments. The panel defined a postbiotic as a "preparation of inanimate microorganisms and/or their components that confers a health benefit on the host". Effective postbiotics must contain inactivated microbial cells or cell components, with or without metabolites, that contribute to observed health benefits. The panel also discussed existing evidence of health-promoting effects of postbiotics, potential mechanisms of action, levels of evidence required to meet the stated definition, safety and implications for stakeholders. The panel determined that a definition of postbiotics is useful so that scientists, clinical triallists, industry, regulators and consumers have common ground for future activity in this area. A generally accepted definition will hopefully lead to regulatory clarity and promote innovation and the development of new postbiotic products.

Conflict of interest statement

S.S. serves on the board of ISAPP, is a board member of Yoghurt in Nutrition Initiative and has been a speaker in meetings funded by industry, Nestlé Nutrition Institute and Institute Danone. M.C.C. has participated as a speaker for HIPP, Danone, Nutricia, Nestlé Nutrition Institute and Mead Johnson. A.E. has led industry-sponsored research projects with support from B Food Science and Takanashi Milk Products, and has been a speaker for the companies. C.H. serves on the board of ISAPP, is a consultant to Artugen Therapeutics developing a live biotherapeutic, and has received research grants from several industry partners, including ADARE Pharmaceuticals, manufacturers of Lacteol. S.L. serves on the academic board of ISAPP and has received research grants from several industry partners, such as Yun. She has been compensated for speaking by Yakult. E.M.M.Q. serves on the board of ISAPP, as a consultant to 4D Pharma, Alimentary Health, Allergan, Biocodex, Ironwood, Salix, Takeda and Vibrant, and has research support from 4D Pharma, Biomerica and Vibrant. M.E.S. has been compensated for speaking engagements or for consulting from Associated British Foods, California Dairy Research Foundation, Cargill, Danone Research, Danone USA, Fairlife, General Mills, GlaxoSmithKline, JJ Heimbach, Kellogg, Kerry, Mead Johnson, Medscape, PepsiCo, Pfizer, Probi, Procter & Gamble, Sanofi, Trouw Nutrition, Visalia Dairy Company, Winclove Probiotics and Yakult. R.S. has participated as a clinical investigator, and/or advisory board member, and/or consultant, and/or speaker for Abbott, Danone and Nestlé. J.R.S. has led industry-sponsored research projects with support from AstraZeneca, Danone, Servier and Vitacress. H.S. serves on the board of ISAPP and has participated as a clinical investigator, and/or advisory board member, and/or consultant, and/or speaker for Arla, Biogaia, Biocodex, Ch. Hansen, Danone, Nestlé, Nestlé Nutrition Institute, Nutricia and Merck. G.V. has led industry-sponsored research projects on dairy products and probiotics. These projects were independently carried out and had no influence on the content of this manuscript. He is member of the Argentinian board of the Yoghurt in Nutrition Initiative (YINI Danone Argentina) and serves on the board of ISAPP. He was not a member of ISAPP Board at the time of the meeting, but has been elected as a board member as of June 2020.

© 2021. The Author(s).

Figures

Fig. 1. Total number of mentions in…
Fig. 1. Total number of mentions in the literature of different terms referring to inanimate microorganisms and/or their metabolites.
Several different terms, all defined differently, have been used over the years to refer to some form of inactivated or killed microorganisms in the research literature according to a search of the literature found on PubMed for the period 1 January 2000 to 21 January 2021. Bacterial lysates were not included in the search although they may be considered postbiotics if health benefits are documented and other criteria for postbiotics are met; the isolation of lysates is also a procedure in molecular biology studies that is often used in situations unrelated to postbiotics, so the term could not be used unambiguously in this search. The data that support the plots within Fig. 1 are available from the authors upon reasonable request.
Fig. 2. Increasing use of the term…
Fig. 2. Increasing use of the term postbiotics in the published literature.
Several different terms have been used over the years to refer to some form of inactivated or killed microorganisms in the research literature according to a search of the literature found on PubMed for the period 1 January 2000 to 21 January 2021. During the past 5 years, ‘postbiotics’ has emerged as the most common of these terms. The data that support the plots within Fig. 2 are available from the authors upon reasonable request.
Fig. 3. Scanning electron micrographs of Lacticaseibacillus…
Fig. 3. Scanning electron micrographs of Lacticaseibacillus rhamnosus GG.
Scanning electron micrographs of Lacticaseibacillus rhamnosus GG in live (part a) and processed (part b) form showing that processing steps to obtain postbiotics can have a major effect on the physical and functional properties of the bacteria, even if the overall biomass and rod shape is preserved. Inactivation was performed in this case by spray drying that resulted in a mixture of live, full piliated cells and inactivated cells lacking pili surface appendages (as described in Kiekens et al.). The bacteria were spotted on a gold-coated membrane, which is especially visible after processing. Adapted with permission from ref., Wiley.
Fig. 4. Postulated mechanisms of postbiotics and…
Fig. 4. Postulated mechanisms of postbiotics and example effector molecules utilized by them.
Five mechanisms of action of postbiotics are postulated: (1) modulation of the resident microbiota; (2) enhancement of epithelial barrier functions; (3) modulation of local and systemic immune responses; (4) modulation of systemic metabolic responses; and (5) systemic signalling via the nervous system. Some examples of microbial effector molecules mediating these mechanisms are shown (non-exhaustive list). Conceptually, the activity of effector molecules could be better retained if the cellular structure of the postbiotics is preserved, for example, through increased avidity in interactions with immune receptors or through increasing the residence time of the active molecules inside the host. The cell wall protects against rapid degradation by digestive enzymes and immune attack inside the host. This aspect is similar to the situation with vaccines, which also function best if cellular structure is preserved, but with the most toxic and/or pathogenic parts being inactivated or deleted. BSH, bile salt hydrolase; EPS, exopolysaccharide; MAMP, microbe-associated molecular pattern; PRR, pattern-recognition receptor; SCFAs, short-chain fatty acids; TCR, T cell receptor; TH cell, T helper cell; Treg cell, regulatory T cell.

References

    1. Selma-Royo M, et al. Shaping microbiota during the first 1000 days of life. Adv. Exp. Med. Biol. 2019;1125:3–24.
    1. Mills S, Stanton C, Lane JA, Smith GJ, Ross RP. Precision nutrition and the microbiome, Part I: current state of the science. Nutrients. 2019;11:923.
    1. Hill C, et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014;11:506–514.
    1. Gibson GR, et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017;14:491–502.
    1. Swanson KS, et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat. Rev. Gastroenterol. Hepatol. 2020;17:687–701.
    1. Fiore W, Arioli S, Guglielmetti S. The neglected microbial components of commercial probiotic formulations. Microorganisms. 2020;8:1177.
    1. Holmes E, et al. Therapeutic modulation of microbiota-host metabolic interactions. Sci. Transl Med. 2012;4:137rv136.
    1. Marco ML, et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on fermented foods. Nat. Rev. Gastroenterol. Hepatol. 2021;18:196–208.
    1. Marco ML, et al. Health benefits of fermented foods: microbiota and beyond. Curr. Opin. Biotechnol. 2017;44:94–102.
    1. Esposito S, et al. Nonspecific immunomodulators for recurrent respiratory tract infections, wheezing and asthma in children: a systematic review of mechanistic and clinical evidence. Curr. Opin. Allergy Clin. Immunol. 2018;18:198–209.
    1. Deshpande G, Athalye-Jape G, Patole S. Para-probiotics for preterm neonates–the next frontier. Nutrients. 2018;10:871.
    1. Kanauchi O, Andoh A, AbuBakar S, Yamamoto N. Probiotics and paraprobiotics in viral infection: clinical application and effects on the innate and acquired immune systems. Curr. Pharm. Des. 2018;24:710–717.
    1. Murata M, et al. Effects of paraprobiotic Lactobacillus paracasei MCC1849 supplementation on symptoms of the common cold and mood states in healthy adults. Benef. Microbes. 2018;9:855–864.
    1. Sugawara T, et al. Regulatory effect of paraprobiotic Lactobacillus gasseri CP2305 on gut environment and function. Microb. Ecol. Health Dis. 2016;27:30259.
    1. Nishida K, et al. Para-psychobiotic Lactobacillus gasseri CP2305 ameliorates stress-related symptoms and sleep quality. J. Appl. Microbiol. 2017;123:1561–1570.
    1. Taverniti V, Guglielmetti S. The immunomodulatory properties of probiotic microorganisms beyond their viability (ghost probiotics: proposal of paraprobiotic concept) Genes Nutr. 2011;6:261–274.
    1. Sharma M, Chandel D, Shukla G. Antigenotoxicity and cytotoxic potentials of metabiotics extracted from isolated probiotic, Lactobacillus rhamnosus MD 14 on Caco-2 and HT-29 human colon cancer cells. Nutr. Cancer. 2020;72:110–119.
    1. Shenderov BA. Metabiotics: novel idea or natural development of probiotic conception. Microb. Ecol. Health Dis. 2013;24:20399.
    1. Pique N, Berlanga M, Minana-Galbis D. Health benefits of heat-killed (tyndallized) probiotics: an overview. Int. J. Mol. Sci. 2019;20:2534.
    1. Lopetuso L, et al. Gelatin tannate and tyndallized probiotics: a novel approach for treatment of diarrhea. Eur. Rev. Med. Pharmacol. Sci. 2017;21:873–883.
    1. Jurkiewicz D, Zielnik-Jurkiewicz B. Bacterial lysates in the prevention of respiratory tract infections. Otolaryngol. Pol. 2018;72:1–8.
    1. Zheng J, et al. A taxonomic note on the genus Lactobacillus: description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int. J. Syst. Evol. Microbiol. 2020;70:2782–2858.
    1. Cicenia A, et al. Postbiotic activities of lactobacilli-derived factors. J. Clin. Gastroenterol. 2014;48(Suppl. 1):S18–S22.
    1. Patel RM, Denning PW. Therapeutic use of prebiotics, probiotics, and postbiotics to prevent necrotizing enterocolitis: what is the current evidence? Clin. Perinatol. 2013;40:11–25.
    1. Andresen V, Gschossmann J, Layer P. Heat-inactivated Bifidobacterium bifidum MIMBb75 (SYN-HI-001) in the treatment of irritable bowel syndrome: a multicentre, randomised, double-blind, placebo-controlled clinical trial. Lancet Gastroenterol. Hepatol. 2020;5:658–666.
    1. Aguilar-Toala JE, et al. In silico prediction and in vitro assessment of multifunctional properties of postbiotics obtained from two probiotic bacteria. Probiotics Antimicrob. Proteins. 2019;12:608–622.
    1. Martin R, et al. Functional characterization of novel Faecalibacterium prausnitzii strains isolated from healthy volunteers: a step forward in the use of F. prausnitzii as a next-generation probiotic. Front. Microbiol. 2017;8:1226.
    1. Brodmann T, et al. Safety of novel microbes for human consumption: practical examples of assessment in the European Union. Front. Microbiol. 2017;8:1725.
    1. Breyner NM, et al. Microbial anti-inflammatory molecule (MAM) from Faecalibacterium prausnitzii shows a protective effect on DNBS and DSS-induced colitis model in mice through inhibition of NF-κB pathway. Front. Microbiol. 2017;8:114.
    1. Cani PD, de Vos WM. Next-generation beneficial microbes: the case of Akkermansia muciniphila. Front. Microbiol. 2017;8:1765.
    1. European Medicines Agency. Assessment report. Referral under Article 31 of Directive 2001/83/EC. Bacterial lysates-containing medicinal products for respiratory conditions. EMA (2019).
    1. Cardinale F, et al. Epithelial dysfunction, respiratory infections and asthma: the importance of immunomodulation. A focus on OM-85. Expert Rev. Respir. Med. 2020;14:1019–1026.
    1. Huber M, Mossmann H, Bessler WG. Th1-orientated immunological properties of the bacterial extract OM-85-BV. Eur. J. Med. Res. 2005;10:209–217.
    1. Braido F, Tarantini F, Ghiglione V, Melioli G, Canonica GW. Bacterial lysate in the prevention of acute exacerbation of COPD and in respiratory recurrent infections. Int. J. Chron. Obstruct Pulmon Dis. 2007;2:335–345.
    1. Braido F, et al. The bacterial lysate Lantigen B reduces the number of acute episodes in patients with recurrent infections of the respiratory tract: the results of a double blind, placebo controlled, multicenter clinical trial. Immunol. Lett. 2014;162:185–193.
    1. Morandi B, et al. A mixture of bacterial mechanical lysates is more efficient than single strain lysate and of bacterial-derived soluble products for the induction of an activating phenotype in human dendritic cells. Immunol. Lett. 2011;138:86–91.
    1. Lanzilli G, et al. Administration of a polyvalent mechanical bacterial lysate to elderly patients with COPD: effects on circulating T, B and NK cells. Immunol. Lett. 2013;149:62–67.
    1. Rossi GA, et al. Naturally occurring immune response against bacteria commonly involved in upper respiratory tract infections: analysis of the antigen-specific salivary IgA levels. Immunol. Lett. 2003;86:85–91.
    1. Braido F, et al. Sub-lingual administration of a polyvalent mechanical bacterial lysate (PMBL) in patients with moderate, severe, or very severe chronic obstructive pulmonary disease (COPD) according to the GOLD spirometric classification: a multicentre, double-blind, randomised, controlled, phase IV study (AIACE study: Advanced Immunological Approach in COPD Exacerbation) Pulm. Pharmacol. Ther. 2015;33:75–80.
    1. Zarezadeh M, et al. Spirulina supplementation and anthropometric indices: a systematic review and meta-analysis of controlled clinical trials. Phytother Res. 2021;35:577–586.
    1. Mitsuoka, T. Intestinal Flora and Probiotics (Gakkai Shuppan Center, 1998).
    1. Kamioka H, et al. Quality of systematic reviews of the Foods With Function Claims registered at the Consumer Affairs Agency Web site in Japan: a prospective systematic review. Nutr. Res. 2017;40:21–31.
    1. Nakamura F, et al. Effect of fragmented Lactobacillus amylovorus CP1563 on lipid metabolism in overweight and mildly obese individuals: a randomized controlled trial. Microb. Ecol. Health Dis. 2016;27:30312.
    1. Morita Y, Jounai K, Miyake M, Inaba M, Kanauchi O. Effect of heat-killed Lactobacillus paracasei KW3110 ingestion on ocular disorders caused by visual display terminal (VDT) loads: a randomized, double-blind, placebo-controlled parallel-group study. Nutrients. 2018;10:1058.
    1. Maeda-Yamamoto M, Ohtani T. Development of functional agricultural products utilizing the new health claim labeling system in Japan. Biosci. Biotechnol. Biochem. 2018;82:554–563.
    1. Yin J, Xu B, Zeng X, Shen K. Broncho-Vaxom in pediatric recurrent respiratory tract infections: a systematic review and meta-analysis. Int. Immunopharmacol. 2018;54:198–209.
    1. European Food Safety Authority Scientific Opinion on the safety of heat-treated milk products fermented with Bacteroides xylanisolvens DSM 23964 as a novel food. EFSA J. 2015;13:3956.
    1. Council of Europe. Directive 2004/27/EC of the European Parliament and of the Council of 31 March 2004 amending Directive 2001/83/EC on the Community code relating to medicinal products for human use. European Commission (2004).
    1. Council of Europe. European Pharmacopoeia 7.0: 5.1.4. Microbiological quality of non-streile pharmaceutical preparations and substances for pharmaceutical use. Medicinal Genomics (2011).
    1. Council of Europe. Regulation (EU) 2017/745 of the European Parliament and of the Council of 5 April 2017 on medical devices, amending Directive 2001/83/EC, Regulation (EC) No 178/2002 and Regulation (EC) No 1223/2009 and repealing Council Directives 90/385/EEC and 93/42/EEC. (2017).
    1. Binetti, A., Burns, P., Tomei, D., Reinheimer, J. & Vinderola, G. in Lactic Acid Bacteria, Microbiological and Functional Aspects Ch. 39 (eds Vinderola, G., Salminen, S., Ouwehand, A. & von Wright, A.) (CRC, 2019).
    1. Food and Drug Administration. What does FDA regulate? FDA (2021).
    1. Yelin I, et al. Genomic and epidemiological evidence of bacterial transmission from probiotic capsule to blood in ICU patients. Nat. Med. 2019;25:1728–1732.
    1. Periti P, Mazzei T. Antibiotic-induced release of bacterial cell wall components in the pathogenesis of sepsis and septic shock: a review. J. Chemother. 1998;10:427–448.
    1. Yıkmış S. New approaches in non-thermal processes in the food industry. Int. J. Nutr. Food Sci. 2016;5:344–351.
    1. Barba FJ, Kouba M, do Prado-Silvac L, Orlien V, Sant’Anac ADS. Mild processing applied to the inactivation of the main foodborne bacterial pathogens: a review. Trends Food Sci. Technol. 2017;66:20–35.
    1. Charoux CMG, et al. Effect of non-thermal plasma technology on microbial inactivation and total phenolic content of a model liquid food system and black pepper grains. LWT. 2020;118:108716.
    1. Fu N, Huang S, Xiao J, Chen XD. Producing powders containing active dry probiotics with the aid of spray drying. Adv. Food Nutr. Res. 2018;85:211–262.
    1. Szajewska H, Skorka A, Piescik-Lech M. Fermented infant formulas without live bacteria: a systematic review. Eur. J. Pediatr. 2015;174:1413–1420.
    1. Broeckx G, Vandenheuvel D, Claes IJ, Lebeer S, Kiekens F. Drying techniques of probiotic bacteria as an important step towards the development of novel pharmabiotics. Int. J. Pharm. 2016;505:303–318.
    1. Roohinejad, S., Koubaa, M., Sant’Ana, A. S. & Greiner, R. in Innovative Technologies for Food Preservation (eds Orlien, V., Sant’Ana, A. S., Barba, F. J., & Koubaa, M.) 111–132 (2018).
    1. Iaconelli C, et al. Drying process strongly affects probiotics viability and functionalities. J. Biotechnol. 2015;214:17–26.
    1. Tabanelli G, et al. Effect of a non-lethal high pressure homogenization treatment on the in vivo response of probiotic lactobacilli. Food Microbiol. 2012;32:302–307.
    1. Lebeer S, et al. Functional analysis of Lactobacillus rhamnosus GG pili in relation to adhesion and immunomodulatory interactions with intestinal epithelial cells. Appl. Env. Microbiol. 2012;78:185–193.
    1. Verma T, Chaves BD, Howell T, Jr., Subbiah J. Thermal inactivation kinetics of Salmonella and Enterococcus faecium NRRL B-2354 on dried basil leaves. Food Microbiol. 2021;96:103710.
    1. Ruiling L, Donghong L, Jianwei Z. Bacterial spore inactivation by non-thermal technologies: resistance and inactivation mechanisms. Curr. Opin. Food Sci. 2021;42:31–36.
    1. Jambrak AR, Vukušić T, Donsi F, Paniwnyk L, Djekic I. Three pillars of novel nonthermal food technologies: food safety, quality, and environment. J. Food Quality. 2018;2018:8619707.
    1. Chiron C, Tompkins TA, Burguiere P. Flow cytometry: a versatile technology for specific quantification and viability assessment of micro-organisms in multistrain probiotic products. J. Appl. Microbiol. 2018;124:572–584.
    1. Savijoki K, et al. Growth mode and carbon source impact the surfaceome dynamics of Lactobacillus rhamnosus GG. Front. Microbiol. 2019;10:1272.
    1. Kramer M, Obermajer N, Bogovic Matijasic B, Rogelj I, Kmetec V. Quantification of live and dead probiotic bacteria in lyophilised product by real-time PCR and by flow cytometry. Appl. Microbiol. Biotechnol. 2009;84:1137–1147.
    1. Gobert G, et al. Droplet digital PCR improves absolute quantification of viable lactic acid bacteria in faecal samples. J. Microbiol. Methods. 2018;148:64–73.
    1. Hansen SJZ, et al. Droplet digital PCR is an improved alternative method for high-quality enumeration of viable probiotic strains. Front. Microbiol. 2019;10:3025.
    1. Himmelreich U, Sorrell TC, Daniel HM. Nuclear magnetic resonance spectroscopy-based identification of yeast. Methods Mol. Biol. 2017;1508:289–304.
    1. Francius G, et al. Stretching polysaccharides on live cells using single molecule force spectroscopy. Nat. Protoc. 2009;4:939–946.
    1. Kiekens S, et al. Impact of spray-drying on the pili of Lactobacillus rhamnosus GG. Microb. Biotechnol. 2019;12:849–855.
    1. Sankarganesh P, Joseph B. Fourier transform infrared spectroscopy as a tool for identification of crude microbial extracts with anti-malarial potential. Acta Parasitol. 2016;61:98–101.
    1. Rault A, Bouix M, Béala C. Cryotolerance of Lactobacillus delbrueckii subsp. bulgaricus CFL1 is influenced by the physiological state during fermentation. Int. Dairy. J. 2010;20:792–799.
    1. Lebeer S, et al. Identification of probiotic effector molecules: present state and future perspectives. Curr. Opin. Biotechnol. 2018;49:217–223.
    1. Sun Z, et al. Expanding the biotechnology potential of lactobacilli through comparative genomics of 213 strains and associated genera. Nat. Commun. 2015;6:8322.
    1. Corr SC, et al. Bacteriocin production as a mechanism for the antiinfective activity of Lactobacillus salivarius UCC118. Proc. Natl Acad. Sci. USA. 2007;104:7617–7621.
    1. Grandclement C, Tannieres M, Morera S, Dessaux Y, Faure D. Quorum quenching: role in nature and applied developments. FEMS Microbiol. Rev. 2016;40:86–116.
    1. Laverde Gomez JA, et al. Formate cross-feeding and cooperative metabolic interactions revealed by transcriptomics in co-cultures of acetogenic and amylolytic human colonic bacteria. Env. Microbiol. 2019;21:259–271.
    1. Tytgat HL, et al. Lactobacillus rhamnosus GG outcompetes Enterococcus faecium via mucus-binding pili: evidence for a novel and heterospecific probiotic mechanism. Appl. Env. Microbiol. 2016;82:5756–5762.
    1. Petrova MI, et al. Lectin-like molecules of Lactobacillus rhamnosus GG inhibit pathogenic Escherichia coli and Salmonella biofilm formation. PLoS ONE. 2016;11:e0161337.
    1. Yan F, et al. A Lactobacillus rhamnosus GG-derived soluble protein, p40, stimulates ligand release from intestinal epithelial cells to transactivate epidermal growth factor receptor. J. Biol. Chem. 2013;288:30742–30751.
    1. Gao J, et al. A novel postbiotic from Lactobacillus rhamnosus GG with a beneficial effect on intestinal barrier function. Front. Microbiol. 2019;10:477.
    1. Schiavi E, et al. The surface-associated exopolysaccharide of Bifidobacterium longum 35624 plays an essential role in dampening host proinflammatory responses and repressing local TH17 responses. Appl. Env. Microbiol. 2016;82:7185–7196.
    1. Engevik MA, et al. Bifidobacterium dentium fortifies the intestinal mucus layer via autophagy and calcium signaling pathways. mBio. 2019;10:e01087-19.
    1. Feng Y, Wang Y, Wang P, Huang Y, Wang F. Short-chain fatty acids manifest stimulative and protective effects on intestinal barrier function through the inhibition of NLRP3 inflammasome and autophagy. Cell Physiol. Biochem. 2018;49:190–205.
    1. Ohata A, Usami M, Miyoshi M. Short-chain fatty acids alter tight junction permeability in intestinal monolayer cells via lipoxygenase activation. Nutrition. 2005;21:838–847.
    1. Lebeer S, Vanderleyden J, De Keersmaecker SC. Host interactions of probiotic bacterial surface molecules: comparison with commensals and pathogens. Nat. Rev. Microbiol. 2010;8:171–184.
    1. Mohamadzadeh M, et al. Regulation of induced colonic inflammation by Lactobacillus acidophilus deficient in lipoteichoic acid. Proc. Natl Acad. Sci. USA. 2011;108(Suppl. 1):4623–4630.
    1. Macho Fernandez E, et al. Anti-inflammatory capacity of selected lactobacilli in experimental colitis is driven by NOD2-mediated recognition of a specific peptidoglycan-derived muropeptide. Gut. 2011;60:1050–1059.
    1. Sabharwal H, Cichon C, Olschlager TA, Sonnenborn U, Schmidt MA. Interleukin-8, CXCL1, and microRNA miR-146a responses to probiotic Escherichia coli Nissle 1917 and enteropathogenic E. coli in human intestinal epithelial T84 and monocytic THP-1 cells after apical or basolateral infection. Infect. Immun. 2016;84:2482–2492.
    1. Zhong Y, Huang J, Tang W, Chen B, Cai W. Effects of probiotics, probiotic DNA and the CpG oligodeoxynucleotides on ovalbumin-sensitized brown-Norway rats via TLR9/NF-κB pathway. FEMS Immunol. Med. Microbiol. 2012;66:71–82.
    1. Basic M, et al. Loss of CD14 leads to disturbed epithelial-B cell crosstalk and impairment of the intestinal barrier after E. coli Nissle monoassociation. Sci. Rep. 2018;8:719.
    1. Jin X, Zhang M, Yang YF. Saccharomyces cerevisiae β-glucan-induced SBD-1 expression in ovine ruminal epithelial cells is mediated through the TLR-2-MyD88-NF-κB/MAPK pathway. Vet. Res. Commun. 2019;43:77–89.
    1. Sturm A, et al. Escherichia coli Nissle 1917 distinctively modulates T-cell cycling and expansion via toll-like receptor 2 signaling. Infect. Immun. 2005;73:1452–1465.
    1. Luan H, et al. OM85-BV induced the productions of IL-1β, IL-6, and TNF-α via TLR4- and TLR2-mediated ERK1/2/NF-κB pathway in RAW264.7 cells. J. Interferon Cytokine Res. 2014;34:526–536.
    1. Morita N, et al. GPR31-dependent dendrite protrusion of intestinal CX3CR1(+) cells by bacterial metabolites. Nature. 2019;566:110–114.
    1. Cervantes-Barragan L, et al. Lactobacillus reuteri induces gut intraepithelial CD4(+)CD8αα(+) T cells. Science. 2017;357:806–810.
    1. Thomas CM, et al. Histamine derived from probiotic Lactobacillus reuteri suppresses TNF via modulation of PKA and ERK signaling. PLoS ONE. 2012;7:e31951.
    1. Thangaraju M, et al. GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer Res. 2009;69:2826–2832.
    1. Long SL, Gahan CGM, Joyce SA. Interactions between gut bacteria and bile in health and disease. Mol. Asp. Med. 2017;56:54–65.
    1. Travers MA, et al. Deconjugated bile salts produced by extracellular bile-salt hydrolase-like activities from the probiotic Lactobacillus johnsonii La1 inhibit Giardia duodenalis in vitro growth. Front. Microbiol. 2016;7:1453.
    1. Mullish BH, et al. Microbial bile salt hydrolases mediate the efficacy of faecal microbiota transplant in the treatment of recurrent Clostridioides difficile infection. Gut. 2019;68:1791–1800.
    1. De Vadder F, et al. Microbiota-produced succinate improves glucose homeostasis via intestinal gluconeogenesis. Cell Metab. 2016;24:151–157.
    1. Wolever TM, Fernandes J, Rao AV. Serum acetate:propionate ratio is related to serum cholesterol in men but not women. J. Nutr. 1996;126:2790–2797.
    1. Hamer HM, et al. Butyrate modulates oxidative stress in the colonic mucosa of healthy humans. Clin. Nutr. 2009;28:88–93.
    1. Caspani G, Swann J. Small talk: microbial metabolites involved in the signaling from microbiota to brain. Curr. Opin. Pharmacol. 2019;48:99–106.
    1. O’Mahony SM, Clarke G, Borre YE, Dinan TG, Cryan JF. Serotonin, tryptophan metabolism and the brain–gut–microbiome axis. Behav. Brain Res. 2015;277:32–48.
    1. Iwasaki M, Akiba Y, Kaunitz JD. Duodenal chemosensing of short-chain fatty acids: implications for GI diseases. Curr. Gastroenterol. Rep. 2019;21:35.
    1. Engevik MA, et al. Human-derived Bifidobacterium dentium modulates the mammalian serotonergic system and gut–brain axis. Cell Mol. Gastroenterol. Hepatol. 2021;11:221–248.
    1. Byrne CS, Chambers ES, Morrison DJ, Frost G. The role of short chain fatty acids in appetite regulation and energy homeostasis. Int. J. Obes. 2015;39:1331–1338.
    1. Chambers ES, et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut. 2015;64:1744–1754.
    1. Frost G, et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat. Commun. 2014;5:3611.
    1. Mohedano ML, et al. Real-time detection of riboflavin production by Lactobacillus plantarum strains and tracking of their gastrointestinal survival and functionality in vitro and in vivo using mCherry labeling. Front. Microbiol. 2019;10:1748.
    1. Kennedy DO. Vitamins and the brain: mechanisms, dose and efficacy–a review. Nutrients. 2016;8:68.
    1. Parkins DA, Lashmar UT. The formulation of biopharmaceutical products. Pharm. Sci. Technol. Today. 2000;3:129–137.
    1. Warda AK, et al. Heat-killed lactobacilli alter both microbiota composition and behaviour. Behav. Brain Res. 2019;362:213–223.
    1. Warda AK, et al. Oral administration of heat-treated lactobacilli modifies the murine microbiome and reduces Citrobacter induced colitis. Front. Microbiol. 2020;11:69.
    1. Canducci F, et al. A lyophilized and inactivated culture of Lactobacillus acidophilus increases Helicobacter pylori eradication rates. Aliment. Pharmacol. Ther. 2000;14:1625–1629.
    1. Tarrerias AL, et al. The effect of inactivated Lactobacillus LB fermented culture medium on symptom severity: observational investigation in 297 patients with diarrhea-predominant irritable bowel syndrome. Dig. Dis. 2011;29:588–591.
    1. Xiao SD, et al. Multicenter, randomized, controlled trial of heat-killed Lactobacillus acidophilus LB in patients with chronic diarrhea. Adv. Ther. 2003;20:253–260.
    1. Nishida K, Sawada D, Kuwano Y, Tanaka H, Rokutan K. Health benefits of Lactobacillus gasseri CP2305 tablets in young adults exposed to chronic stress: a randomized, double-blind, placebo-controlled study. Nutrients. 2019;11:1859.
    1. Jones C, et al. Modulation of gut barrier function in patients with obstructive jaundice using probiotic LP299v. Eur. J. Gastroenterol. Hepatol. 2013;25:1424–1430.
    1. Hoffman JR, et al. The effect of 2 weeks of inactivated probiotic Bacillus coagulans on endocrine, inflammatory, and performance responses during self-defense training in soldiers. J. Strength. Cond. Res. 2019;33:2330–2337.
    1. Montane E, et al. Pilot, double-blind, randomized, placebo-controlled clinical trial of the supplement food Nyaditum resae® in adults with or without latent TB infection: safety and immunogenicity. PLoS ONE. 2017;12:e0171294.
    1. Zhang J, Guo S, Li C, Jiang X. Therapeutic effects of inhaled inactivated Mycobacterium phlei in adult patients with moderate persistent asthma. Immunotherapy. 2012;4:383–387.
    1. Tandon MK, et al. Oral immunotherapy with inactivated nontypeable Haemophilus influenzae reduces severity of acute exacerbations in severe COPD. Chest. 2010;137:805–811.
    1. Frank MG, et al. Immunization with Mycobacterium vaccae induces an anti-inflammatory milieu in the CNS: attenuation of stress-induced microglial priming, alarmins and anxiety-like behavior. Brain Behav. Immun. 2018;73:352–363.
    1. Mohammedsaeed W, Cruickshank S, McBain AJ, O’Neill CA. Lactobacillus rhamnosus GG lysate increases re-epithelialization of keratinocyte scratch assays by promoting migration. Sci. Rep. 2015;5:16147.
    1. Rinaldi F, Trink A, Pinto D. Efficacy of postbiotics in a PRP-like cosmetic product for the treatment of alopecia area Celsi: a randomized double-blinded parallel-group study. Dermatol. Ther. 2020;10:483–493.
    1. Fernandez L, Delgado S, Herrero H, Maldonado A, Rodriguez JM. The bacteriocin nisin, an effective agent for the treatment of staphylococcal mastitis during lactation. J. Hum. Lact. 2008;24:311–316.
    1. Kang BS, et al. Antimicrobial activity of enterocins from Enterococcus faecalis SL-5 against Propionibacterium acnes, the causative agent in acne vulgaris, and its therapeutic effect. J. Microbiol. 2009;47:101–109.
    1. Puccetti M, Giovagnoli S, Zelante T, Romani L, Ricci M. Development of novel indole-3-aldehyde-loaded gastro-resistant spray-dried microparticles for postbiotic small intestine local delivery. J. Pharm. Sci. 2018;107:2341–2353.
    1. Scheppach W, et al. Effect of butyrate enemas on the colonic mucosa in distal ulcerative colitis. Gastroenterology. 1992;103:51–56.
    1. Steinhart AH, Brzezinski A, Baker JP. Treatment of refractory ulcerative proctosigmoiditis with butyrate enemas. Am. J. Gastroenterol. 1994;89:179–183.
    1. Vernia P, et al. Short-chain fatty acid topical treatment in distal ulcerative colitis. Aliment. Pharmacol. Ther. 1995;9:309–313.
    1. Patz J, Jacobsohn WZ, Gottschalk-Sabag S, Zeides S, Braverman DZ. Treatment of refractory distal ulcerative colitis with short chain fatty acid enemas. Am. J. Gastroenterol. 1996;91:731–734.
    1. Steinhart AH, Hiruki T, Brzezinski A, Baker JP. Treatment of left-sided ulcerative colitis with butyrate enemas: a controlled trial. Aliment. Pharmacol. Ther. 1996;10:729–736.
    1. Scheppach W. Treatment of distal ulcerative colitis with short-chain fatty acid enemas. A placebo-controlled trial. German–Austrian SCFA Study Group. Dig. Dis. Sci. 1996;41:2254–2259.
    1. Talley NA, Chen F, King D, Jones M, Talley NJ. Short-chain fatty acids in the treatment of radiation proctitis: a randomized, double-blind, placebo-controlled, cross-over pilot trial. Dis. Colon. Rectum. 1997;40:1046–1050.
    1. Pinto A, et al. Short chain fatty acids are effective in short-term treatment of chronic radiation proctitis: randomized, double-blind, controlled trial. Dis. Colon. Rectum. 1999;42:788–795.
    1. Vernia P, et al. Topical butyrate for acute radiation proctitis: randomised, crossover trial. Lancet. 2000;356:1232–1235.
    1. Maggio A, et al. Daily sodium butyrate enema for the prevention of radiation proctitis in prostate cancer patients undergoing radical radiation therapy: results of a multicenter randomized placebo-controlled dose-finding phase 2 study. Int. J. Radiat. Oncol. Biol. Phys. 2014;89:518–524.
    1. Vanhoutvin SA, et al. The effects of butyrate enemas on visceral perception in healthy volunteers. Neurogastroenterol. Motil. 2009;21:952–e76.
    1. Guillemot F, et al. Treatment of diversion colitis by short-chain fatty acids. Prospective double-blind study. Dis. Colon. Rectum. 1991;34:861–864.
    1. Luceri C, et al. Effect of butyrate enemas on gene expression profiles and endoscopic/histopathological scores of diverted colorectal mucosa: a randomized trial. Dig. Liver Dis. 2016;48:27–33.
    1. Tominaga K, et al. Diversion colitis and pouchitis: a mini-review. World J. Gastroenterol. 2018;24:1734–1747.
    1. Caluwaerts S, et al. AG013, a mouth rinse formulation of Lactococcus lactis secreting human Trefoil Factor 1, provides a safe and efficacious therapeutic tool for treating oral mucositis. Oral. Oncol. 2010;46:564–570.
    1. Kurtz CB, et al. An engineered E. coli Nissle improves hyperammonemia and survival in mice and shows dose-dependent exposure in healthy humans. Sci. Transl Med. 2019;11:eaau7975.
    1. Kurtz C, et al. Translational development of microbiome-based therapeutics: kinetics of E. coli Nissle and engineered strains in humans and nonhuman primates. Clin. Transl Sci. 2018;11:200–207.
    1. Robert S, Steidler L. Recombinant Lactococcus lactis can make the difference in antigen-specific immune tolerance induction, the type 1 diabetes case. Microb. Cell Fact. 2014;13(Suppl 1):11.
    1. Takiishi T, et al. Reversal of autoimmune diabetes by restoration of antigen-specific tolerance using genetically modified Lactococcus lactis in mice. J. Clin. Invest. 2012;122:1717–1725.
    1. Vandenbroucke K, et al. Orally administered L. lactis secreting an anti-TNF Nanobody demonstrate efficacy in chronic colitis. Mucosal Immunol. 2010;3:49–56.
    1. Frossard CP, Steidler L, Eigenmann PA. Oral administration of an IL-10-secreting Lactococcus lactis strain prevents food-induced IgE sensitization. J. Allergy Clin. Immunol. 2007;119:952–959.
    1. Reardon S. Genetically modified bacteria enlisted in fight against disease. Nature. 2018;558:497–498.
    1. EFSA Panel on Additives and Products or Substances used in Animal Feed (FEEDAP) et al. Safety and nutritional value of a dried killed bacterial biomass from Escherichia coli (FERM BP-10941) (PL73 (LM)) as a feed material for pigs, ruminants and salmonids. EFSA J. 2017;15:e04935.
    1. Agostoni C, et al. Fermented infant formulae without live bacteria. J. Pediatr. Gastroenterol. Nutr. 2007;44:392–397.
    1. Campeotto F, et al. A fermented formula in pre-term infants: clinical tolerance, gut microbiota, down-regulation of faecal calprotectin and up-regulation of faecal secretory IgA. Br. J. Nutr. 2011;105:1843–1851.
    1. Szajewska H, Ruszczynski M, Kolacek S. Meta-analysis shows limited evidence for using Lactobacillus acidophilus LB to treat acute gastroenteritis in children. Acta Paediatr. 2014;103:249–255.
    1. Kaila M, Isolauri E, Saxelin M, Arvilommi H, Vesikari T. Viable versus inactivated lactobacillus strain GG in acute rotavirus diarrhoea. Arch. Dis. Child. 1995;72:51–53.
    1. Mantziari A, Salminen S, Szajewska H, Malagon-Rojas JN. Postbiotics against pathogens commonly involved in pediatric infectious diseases. Microorganisms. 2020;8:1510.
    1. Nocerino R, et al. Cow’s milk and rice fermented with Lactobacillus paracasei CBA L74 prevent infectious diseases in children: a randomized controlled trial. Clin. Nutr. 2017;36:118–125.
    1. Thibault H, Aubert-Jacquin C, Goulet O. Effects of long-term consumption of a fermented infant formula (with Bifidobacterium breve c50 and Streptococcus thermophilus 065) on acute diarrhea in healthy infants. J. Pediatr. Gastroenterol. Nutr. 2004;39:147–152.
    1. Sharieff W, Bhutta Z, Schauer C, Tomlinson G, Zlotkin S. Micronutrients (including zinc) reduce diarrhoea in children: the Pakistan Sprinkles Diarrhoea Study. Arch. Dis. Child. 2006;91:573–579.
    1. Corsello G, et al. Preventive effect of cow’s milk fermented with Lactobacillus paracasei CBA L74 on common infectious diseases in children: a multicenter randomized controlled trial. Nutrients. 2017;9:669.
    1. Kirjavainen PV, Salminen SJ, Isolauri E. Probiotic bacteria in the management of atopic disease: underscoring the importance of viability. J. Pediatr. Gastroenterol. Nutr. 2003;36:223–227.
    1. Bodemer C, et al. Adjuvant treatment with the bacterial lysate (OM-85) improves management of atopic dermatitis: a randomized study. PLoS ONE. 2017;12:e0161555.
    1. Lau S, et al. Oral application of bacterial lysate in infancy decreases the risk of atopic dermatitis in children with 1 atopic parent in a randomized, placebo-controlled trial. J. Allergy Clin. Immunol. 2012;129:1040–1047.
    1. Indrio F, Ladisa G, Mautone A, Montagna O. Effect of a fermented formula on thymus size and stool pH in healthy term infants. Pediatr. Res. 2007;62:98–100.
    1. Mullie C, et al. Increased poliovirus-specific intestinal antibody response coincides with promotion of Bifidobacterium longum-infantis and Bifidobacterium breve in infants: a randomized, double-blind, placebo-controlled trial. Pediatr. Res. 2004;56:791–795.
    1. Berni Canani R, et al. Specific signatures of the gut microbiota and increased levels of butyrate in children treated with fermented cow’s milk containing heat-killed Lactobacillus paracasei CBA L74. Appl. Environ. Microbiol. 2017;83:e01206-17.
    1. Campeotto F, et al. High faecal calprotectin concentrations in newborn infants. Arch. Dis. Child. Fetal Neonatal Ed. 2004;89:F353–355.
    1. Tsilingiri K, Rescigno M. Postbiotics: what else? Benef. Microbes. 2013;4:101–107.
    1. Aguilar-Toalá JE, et al. Postbiotics: an evolving term within the functional foods field. Trends Food Sci. Technol. 2018;75:105–114.
    1. Collado MC, Vinderola G, Salminen S. Postbiotics: facts and open questions. A position paper on the need for a consensus definition. Benef. Microbes. 2019;10:711–719.
    1. Foo, H. L., Loh, T. C., Mutalib, N. E. A. & Rahim, R. A. in Microbiome and Metabolome in Diagnosis, Therapy, and other Strategic Applications Ch. 21 (eds Faintuch, J. & Faintuch, S.) 201–211 (Academic, 2019).
    1. Johnson CN, et al. Administration of a postbiotic causes immunomodulatory responses in broiler gut and reduces disease pathogenesis following challenge. Microorganisms. 2019;7:268.
    1. Wegh CAM, Geerlings SY, Knol J, Roeselers G, Belzer C. Postbiotics and their potential applications in early life nutrition and beyond. Int. J. Mol. Sci. 2019;20:4673.
    1. Zagolski O, Strek P, Kasprowicz A, Bialecka A. Effectiveness of polyvalent bacterial lysate and autovaccines against upper respiratory tract bacterial colonization by potential pathogens: a randomized study. Med. Sci. Monit. 2015;21:2997–3002.
    1. Krusteva E, et al. Clinical study of the effect of the preparation DEODAN on leukopenia, induced by cytostatics. Int. J. Immunopharmacol. 1997;19:487–492.
    1. Morisset M, Aubert-Jacquin C, Soulaines P, Moneret-Vautrin DA, Dupont C. A non-hydrolyzed, fermented milk formula reduces digestive and respiratory events in infants at high risk of allergy. Eur. J. Clin. Nutr. 2011;65:175–183.
    1. Roy P, Aubert-Jacquin C, Avart C, Gontier C. Benefits of a thickened infant formula with lactase activity in the management of benign digestive disorders in newborns. Arch. Pediatr. 2004;11:1546–1554.
    1. Boulloche J, Mouterde O, Mallet E. Management of acute diarrhea in infants and toddlers – controlled study of the antidiarrheal efficacy of killed Lactobacillus acidophilus (LB strain) versus a placebo and a reference agent (loperamide) Ann. Pediatr. 1994;41:457–463.
    1. Lievin-Le Moal V, Sarrazin-Davila LE, Servin AL. An experimental study and a randomized, double-blind, placebo-controlled clinical trial to evaluate the antisecretory activity of Lactobacillus acidophilus strain LB against nonrotavirus diarrhea. Pediatrics. 2007;120:e795–803.
    1. Salazar-Lindo E, et al. Effectiveness and safety of Lactobacillus LB in the treatment of mild acute diarrhea in children. J. Pediatr. Gastroenterol. Nutr. 2007;44:571–576.
    1. Simakachorn N, et al. Clinical evaluation of the addition of lyophilized, heat-killed Lactobacillus acidophilus LB to oral rehydration therapy in the treatment of acute diarrhea in children. J. Pediatr. Gastroenterol. Nutr. 2000;30:68–72.
    1. Peng GC, Hsu CH. The efficacy and safety of heat-killed Lactobacillus paracasei for treatment of perennial allergic rhinitis induced by house-dust mite. Pediatr. Allergy Immunol. 2005;16:433–438.
    1. Rampengan NH, Manoppo J, Warouw SM. Comparison of efficacies between live and killed probiotics in children with lactose malabsorption. Southeast. Asian J. Trop. Med. Public. Health. 2010;41:474–481.

Source: PubMed

3
Iratkozz fel