Hostname: page-component-7c8c6479df-xxrs7 Total loading time: 0 Render date: 2024-03-27T09:31:46.978Z Has data issue: false hasContentIssue false

Probiotics: a proactive approach to health. A symposium report

Published online by Cambridge University Press:  09 November 2015

Linda V. Thomas*
Affiliation:
Yakult UK Limited, Odyssey Business Park, West End Road, South Ruislip, Middlesex HA4 6QQ, UK
Kaori Suzuki
Affiliation:
Yakult Europe B.V., Schutsluisweg 1, 1332 EN Almere, The Netherlands
Jia Zhao
Affiliation:
Yakult Europe B.V., Schutsluisweg 1, 1332 EN Almere, The Netherlands
*
*Corresponding author: L. V. Thomas, email LThomas@yakult.co.uk
Rights & Permissions [Opens in a new window]

Abstract

This report summarises talks given at the 8th International Yakult Symposium, held on 23–24 April 2015 in Berlin. Two presentations explored different aspects of probiotic intervention: the small intestine as a probiotic target and inclusion of probiotics into integrative approaches to gastroenterology. Probiotic recommendations in gastroenterology guidelines and current data on probiotic efficacy in paediatric patients were reviewed. Updates were given on probiotic and gut microbiota research in obesity and obesity-related diseases, the gut–brain axis and development of psychobiotics, and the protective effects of equol-producing strains for prostate cancer. Recent studies were presented on probiotic benefit for antibiotic-associated diarrhoea and people with HIV, as well as protection against the adverse effects of a short-term high-fat diet. Aspects of probiotic mechanisms of activity were discussed, including immunomodulatory mechanisms and metabolite effects, the anti-inflammatory properties of Faecalibacterium prausnitzii, the relationship between periodontitis, microbial production of butyrate in the oral cavity and ageing, and the pathogenic mechanisms of Campylobacter. Finally, an insight was given on a recent expert meeting, which re-examined the probiotic definition, advised on the appropriate use and scope of the term and outlined different probiotic categories and the prevalence of different mechanisms of activity.

Type
Full Papers
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © The Authors 2015

The 8th International Yakult Symposium, held on 23–24 April 2015 in Berlin, was entitled ‘Probiotics, a proactive approach to health’. The title was chosen for two reasons: to emphasise the importance of taking steps to maintain health throughout life and, because the gut microbiota has a major influence on the whole body (not just on the gut), its modulation by probiotics can be part of a strategy for achieving this. This report summarises the talks given by the panel of international expert speakers, who covered different aspects of microbiology, gastroenterology, immunology as well as metabolic and infectious disease. All speakers approved the manuscript before submission.

Aspects of probiotic intervention

The small intestine: a target for probiotics

The primary message from Professor Michiel Kleerebezem (Wageningen University, The Netherlands) was that the small intestine is pivotal for health(Reference El Aidy, van den Bogert and Kleerebezem1). With its distinct community of commensal microbiota and concentration of immune cells, it is a key target for probiotic intervention. Analysis of its microbiota, however, has been hampered by a lack of non-invasive sampling methods for healthy volunteers: sampling has usually been carried out via naso-ileal catheters. Although luminal microbiota samples are easily obtained from the distal ileum of ileostomy subjects, these are usually inflammatory bowel disease (IBD) or cancer patients.

Newly developed radio-controlled capsules (IntelliCap®) are currently being evaluated for the extraction of small volume samples (100–200 μl) from the small intestine of healthy volunteers, to avoid the use of invasive technologies or causing undue discomfort(Reference van der Schaar, Dijksman and Broekhuizen-de Gast2).

16S rRNA phylogenetic microarray (The Human Intestinal Tract Chip) analysis of the ileostomal, small intestine and faecal samples has found that the gut microbiota of the small intestine is much simpler than that of the colon, with far fewer species(Reference Rajilic-Stojanovic, Heilig and Molenaar3Reference van den Bogert, de Vos and Zoetendal5). Ileostomy samples can serve as models for the microbiota of the proximal small intestine, as the microbial compositions are reasonably similar(Reference Zoetendal, Raes and van den Bogert6). Predominant genera in the small intestine are Streptococcus, Veillonella, Clostridium and Escherichia (Reference van den Bogert, Erkus and Boekhorst7). Metagenomic and metatranscriptomic studies have confirmed that the small intestinal microbiota is strongly focused on the import and fermentation of simple carbohydrates(Reference Zoetendal, Raes and van den Bogert6). Escherichia and Streptococcus spp. seem to be involved in carbohydrate import, using phosphotransferase transport systems to convert carbohydrates to pyruvate by glycolysis, and then converting this intermediate to the fermentation end products such as lactate, acetate, formate and potentially ethanol. The secondary fermentation of lactate and acetate by Veillonella and Clostridium spp. produces propionate, acetate and butyrate. As lactobacilli are not dominant in the small intestine, probiotics have the potential to overcrowd the endogenous microbiota and cause a dramatic, albeit transient, community shift that cannot be achieved in the densely colonised colon.

A pioneering study on healthy men revealed a significant host mucosal transcription response to ingestion of probiotic lactobacilli in the small intestine, with 300–750 genes affected in a strain-specific manner(Reference van Baarlen, Troost and van der Meer8, Reference van Baarlen, Troost and van Hemert9). Network biology approaches for transcriptome data interpretation have now advanced sufficiently to enable a molecular explanation of the clinical outcome of a Lactobacillus intervention. Moreover, the in vivo data also confirmed specific wound healing-accelerating effects of the probiotic Lactobacillus rhamnosus GG (LGG) on mucosal physiology, which are achieved through previously established secretion of the proteins P40 and P75 that modulate epidermal growth factor-receptor signalling(Reference Yan, Cao and Cover10). These analyses may also be employed to predict physiological consequences using comparative transcriptomic analyses, which have established significant correlation of responses to probiotics with responses measured for pharmaceutical drugs(Reference Lamb11). It should be noted, however, that most of these pharmaceuticals work systemically, whereas probiotics work locally in the gut. Nevertheless, some probiotic effects and mechanisms may be similar to those achieved by specific drugs and may provide guidance to future probiotic intervention trials.

There is considerable variation in people’s responses to any probiotic; probiotic-responsive genes also cluster according to individual and not by intervention(Reference van Baarlen, Troost and van der Meer8, Reference Bron, van Baarlen and Kleerebezem12). Volunteers for a probiotic trial may be considered healthy yet actually vary enormously in the molecular makeup of their mucosa, indicating that differential molecular solutions for health are possible and can influence the responsiveness to a probiotic intervention, which may (in part) explain ‘non-responders’ in probiotic trials. How this molecular individuality of human subjects is achieved remains unclear and could include many factors such as genotype, epigenetic imprinting, dietary habits, lifestyle and/or endogenous microbiota. Professor Kleerebezem underlined the importance of a crossover trial design, and for probiotic intervention studies exemplified the potential of subject stratification prior to enrolment, on the basis of their predicted susceptibility to the intervention being tested.

Probiotics as part of an integrative approach to gastroenterology

Professor Jost Langhorst (Kliniken Essen-Mitte, Germany) explained that IBD, irritable bowel syndrome (IBS) and other gastrointestinal (GI) diseases are driven by multiple factors, including genetic predisposition, immune dysregulation, gut dysbiosis and barrier dysfunction. Crohn’s disease (CD) patients, for example, have a low diverse gut microbiota with reduced Faecalibacterium prausnitzii and increased Escherichia coli. Relatives of the patients, with the same CD genetic disposition, also showed some degree of dysbiosis and gut barrier abnormality(Reference Hedin, Stagg and Whelan13). As several microbial-related factors (e.g. dysbiosis and low microbiota diversity, presence/persistence of pathobionts and pathogenic antigens) link a defective mucosal interface to inflammation(Reference Frank, St Amand and Feldman14, Reference Lepage, Hasler and Spehlmann15), the gut microbiota should be a treatment target(Reference Renz, von Mutius and Brandtzaeg16).

Professor Langhorst recommended an integrative approach to chronic GI disorders, combining evidence-based complementary and alternative medicine (CAM) with mainstream medicine and lifestyle modifications. Health-care professionals also need to keep up to date with CAM as patients with chronic GI disorders explore and ask about all available treatments(Reference Hilsden, Verhoef and Rasmussen17). As an example, botanical therapies used in IBD were reviewed by Professor Langhorst. Psyllium, for example, may be as effective as mesalamine in prolonging remission in ulcerative colitis (UC)(Reference Fernandez-Banares, Hinojosa and Sanchez-Lombrana18) and also showed effectiveness for active CD when used in combination with probiotics(Reference Fujimori, Tatsuguchi and Gudis19). Other plants and plant-derived substances investigated are the following: curcumin; frankincense; a combination of myrrh, chamomile and charcoal; bilberries; tormentil; and wormwood(Reference Langhorst, Wulfert and Lauche20). Results on psyllium and curcumin are now positive enough to warrant mention in German UC guidelines(Reference Dignass, Preiss and Aust21). Cannabis, a strongly regulated and controlled substance, is also of interest with IBD, but side effects are frequent, and there may be a higher risk of surgery in CD(Reference Storr, Devlin and Kaplan22). Traditional Chinese medicine is also of interest in IBD: acupuncture has shown benefit(Reference Joos, Wildau and Kohnen23).

In industrialised countries, lifestyle and environmental factors have a strong influence on IBD(Reference Rogler and Vavricka24), and these factors (e.g. smoking and lack of exercise) may also influence the gut microbiota(Reference Clarke, Murphy and O’Sullivan25, Reference Cosnes26). Mind–body interventions are also used in treatment of chronic GI disorders because of the evidence that stress exacerbates IBD symptoms(Reference Levenstein, Prantera and Varvo27, Reference Bernstein, Singh and Graff28). Stress reduction strategies and mind–body therapy have shown benefit in UC(Reference Jedel, Hoffman and Merriman29, Reference Berrill, Sadlier and Hood30), and stress can also change the composition of the commensal microbiota(Reference Bailey, Dowd and Galley31). Such observations are part of the rationale for exploring probiotic benefit for IBD. Certain strains have shown benefit (e.g. E. coli Nissle, lactobacilli, bifidobacteria and streptococci) in maintaining remission in UC and acute pouchitis, but there is no reliable evidence for CD. Patients are interested in another strategy for gut microbiota modulation – faecal microbiota transplant (FMT) – and may even attempt it themselves. FMT capsules or microbial consortium have now been developed, and a stool bank facility has been established in the USA (http://www.openbiome.org/). Although FMT has shown positive effects with recurrent Clostridium difficile infection(Reference van Nood, Vrieze and Nieuwdorp32), results have been more uneven in IBD(Reference Kao, Hotte and Gillevet33, Reference Kump, Grochenig and Lackner34). Two recent randomised studies of patients with active UC have been conducted: one had no effect, and one was effective in inducing remission(Reference Moayyedi, Surette and Kim35, Reference Rossen, Fuentes and van der Spek36).

Guidelines and recommendations for probiotic usage

Gastroenterology guidelines for irritable bowel syndrome

Professor Viola Andresen (Israelitic Hospital Hamburg, Germany) explained that guidelines are a combination of medical science, clinical practice and education intended to help promote good clinical practice and to inform the public. Guidelines also need to ensure cost-effectiveness and support clinical decisions and may even be used in legal disputes. The guidelines in Germany are decided by expert opinion alone or by consensus following the Delphi process(Reference Jones and Hunter37), as well as by evaluation of evidence found by a systematic search and review of the literature, which considers risk of bias, inconsistency, indirectness and imprecision. Recommendations are based on the Grading of Recommendations Assessment, Development and Evaluation approach(Reference Guyatt, Oxman and Schunemann38). Even if no evidence is available, an expert opinion may still be needed.

The rationale for probiotic use in IBS is based on the role of the gut microbiota in many GI functions and the observation of a disturbed microbiota in patients. The difficulty in evaluating efficacy, however, was illustrated by studies with Bifidobacterium longum subsp. infantis 35624. A 2005 trial reported alleviation of symptoms(Reference O’Mahony, McCarthy and Kelly39), yet a later study did not find benefit at this test level (1010 colony-forming units (CFU)) but did with a lower dosage (108 CFU)(Reference Whorwell, Altringer and Morel40). Despite conflicting results, meta-analyses do suggest probiotic benefit, but the data raise several questions(Reference McFarland and Dublin41, Reference Ford, Quigley and Lacy42). More research is needed to determine mechanisms of actions, which probiotics are effective and at what dosage and duration.

Several countries now mention probiotics in their clinical guidance on IBS. Although the American Gastroenterological Association IBS management guidelines do not cover probiotics, the accompanying technical review indicates that probiotics may be beneficial and can be considered on an individual basis(Reference Chang, Lembo and Sultan43). A related and more comprehensive review concluded that, as a whole, probiotics improve global symptoms, bloating and flatulence, but this was a weak recommendation based on low quality of evidence(Reference Ford, Quigley and Lacy42). The UK’s National Institute for Health and Care Excellence does not directly recommend probiotics but does offer advice to people who choose to take them – to take probiotics for at least 4 weeks at the manufacturer’s recommended dose while monitoring the effect(44). German guidelines recommend that selected probiotics can be used for treatment, with the strain selection based on symptoms(Reference Layer, Andresen and Pehl45).

Professor Andresen finished by noting the strain-specific nature of probiotics and the heterogeneous nature of available studies. She also discussed whether probiotics should be classified as food or medicine, and their acceptance by experts. Finally, she underlined that the potential success of gut microbiota modulation for IBS was underlined by a case report of a patient who had suffered post-infectious IBS for 2 years, which was refractory to any conventional treatment. The patient became symptom-free within hours of undergoing FMT and remained healthy 14 months later.

Probiotic efficacy in paediatrics: a review of the evidence

After noting the problem of the strain-specific nature of probiotic benefit for meta-analyses and systematic analyses, Professor Hania Szajewska (The Medical University of Warsaw, Poland) gave a comprehensive update of this area.

  • Treatment of acute gastroenteritis: in 2014, the European Society for Paediatric Gastroenterology Hepatology and Nutrition published an evidence-based position paper, concluding that use of probiotics with documented efficacy may be considered(Reference Szajewska, Guarino and Hojsak46). The use of the following probiotics may be considered as adjuncts to standard oral rehydration therapy for reducing diarrhoea duration: LGG or the yeast species Saccharomyces boulardii (low quality of evidence for both strains; strong recommendation); and Lactobacillus reuteri DSM 17938 (low quality of evidence; weak recommendation). There was insufficient evidence to recommend any of the many other probiotics that have been studied.

  • Prevention of nosocomial diarrhoea: some, but not all, probiotics have shown efficacy(Reference Saavedra, Bauman and Oung47, Reference Szajewska, Wanke and Patro48), particularly for GI infections. A recent study with Bifidobacterium animalis subsp. lactis failed to show prevention of common infections in hospitalised children(Reference Hojsak, Tokic Pivac and Mocic Pavic49).

  • Prevention of antibiotic-associated diarrhoea (AAD): although there was no evidence to support probiotics as a treatment, a recent meta-analysis that identified sixteen randomised-controlled trials (RCT) in children concluded an overall relative risk (RR) of 0·55 (95 % CI 0·38, 0·8) for AAD prevention, with a number needed to treat (NNT) of 12(Reference Hempel, Newberry and Maher50).

  • Prevention of necrotising enterocolitis (NEC): a recent Cochrane review calculated an RR of 0·43 (95 % CI 0·33, 0·56) for probiotics in preventing NEC, and an RR of 0·65 (95 % CI 0·52, 0·81) for death(Reference AlFaleh and Anabrees51). The NNT was approximately 33. The evidence is sufficiently strong enough to support a change in practice. Probiotics with documented efficacy may be considered for prevention of NEC, particularly where incidence is high. More information is needed to establish as to which products are effective, as well as their recommended dosages and duration of use.

  • Infantile colic: there is evidence that L. reuteri DSM 17938 has benefit for infantile colic(Reference Chau, Lau and Greenberg52, Reference Mi, Zhao and Qiao53). (One study reported no benefit, possibly due to participants’ heterogeneity(Reference Sung, Hiscock and Tang54).) A recent meta-analysis of three RCT concluded that the probiotic is likely to reduce crying time by about 43 min (a welcome benefit for parents), especially in exclusively or predominantly exclusively breast-fed infants(Reference Urbanska and Szajewska55). A recent RCT suggested that L. reuteri DSM 17938 is also effective in preventing infantile colic; the probiotic reduced crying time from 71 to 38 min(Reference Indrio, Di Mauro and Riezzo56).

  • Prevention of allergic disease: in 2015, the World Allergy Organization published new evidence-based guidelines relating to probiotic use(Reference Fiocchi, Pawankar and Cuello-Garcia57). Current evidence does not indicate that probiotics reduce the risk of children developing allergy, but, despite very poor quality of evidence, it was recommended that there is a likely net benefit from probiotic use in pregnant women who are at high risk of having an allergic child, in women who breast-feed infants that are at high risk of allergy and in infants who are at high risk of developing allergy. However, it remains unclear which probiotic(s) should be used.

Professor Szajewska concluded that the composition of the gut microbiota plays a significant role in the development of a number of disorders affecting children. Although there is huge potential for probiotics, many questions still need to be answered.

Probiotic benefits for other conditions: an update on the evidence

Obesity and obesity-related disease

Professor Mauro Serafini (CRA-NUT, Italy) explained how a Western high-fat diet induces a sustained postprandial state of hyperlipidaemia and hyperglycaemia. This triggers cellular inflammation as well as oxidative stress due to the generation of reactive oxygen species. In turn, these cause endothelial damage and insulin resistance, which increases the risk for diseases such as the metabolic syndrome, atherosclerosis, type 2 diabetes (T2D), hypertension and stroke(Reference Aljada, Mohanty and Ghanim58Reference Miglio, Peluso and Raguzzini60).

A recent placebo-controlled study on healthy overweight people by Professor Serafini’s group demonstrated that a dietary intervention in the form of fruit juice prevented an increase in plasma levels of pro-inflammatory cytokines (TNF-α, IL-6 and IL-17) that occurred after eating a high-fat meal(Reference Peluso, Raguzzini and Villano61). Further studies have shown that eating high-fat meals increases plasma levels of uric acid and thiols; this also can be prevented by simultaneous consumption of fruit juice(Reference Miglio, Peluso and Raguzzini60, Reference Miglio, Peluso and Raguzzini62). These effects may be due to the presence of flavonoid in the fruit juice; these plant metabolites are found in many foods(Reference Peluso, Miglio and Morabito63). Flavonoid bioavailability in vivo (i.e. their antioxidant capability within the human body) can vary enormously in different types of food and can also differ significantly from antioxidant levels detected by in vitro testing of the food. Polyphenol flavonoids found in chocolate, for instance, have high antioxidant levels, but the interference of milk with their absorption in vivo explains why dark chocolate has a far greater antioxidant benefit in vivo (Reference Serafini, Bugianesi and Maiani64). After ingestion, as well as being digested by host enzymes, dietary polyphenols are metabolised by the gut microbiota to produce a range of bioactive derivatives that can be absorbed by the intestinal cells(Reference Marin, Miguelez and Villar65, Reference Chen, Zheng and Li66). The profile of bacterial species present in any individual’s gut microbiota determines what bioactive metabolites will be derived from dietary polyphenols.

There is potential for probiotics to enhance microbial production of bioactive flavonoid metabolites in the gut(Reference Nagpal, Yadav and Marotta67). Several animal studies have also indicated that certain Lactobacillus and Bifidobacterium probiotic strains can reduce oxidative stress via mechanisms such as reducing hydrogen peroxide and hydroxyl radicals(Reference Ishii, Sugimoto and Izawa68Reference Sengul, Isik and Aslim72). The one human study conducted to date showed a similar positive trend: consumption of a probiotic yogurt improved the antioxidant status of people with T2D(Reference Ejtahed, Mohtadi-Nia and Homayouni-Rad73).

Professor Nathalie Delzenne (Catholic University Louvain, Belgium) suggested the gut dysbiosis as a target for management of obesity and related disorders(Reference Tilg and Moschen74). In obese and overweight people, a gut microbiota of low diversity has been linked to these people’s propensity for metabolic disturbances and chronic low-grade inflammation(Reference Le Chatelier, Nielsen and Qin75). Animal models show that the aberrant microbiota, often with lower levels of bifidobacteria and Akkermansia muciniphila, triggers disruption to the gut barrier, increasing permeability and promoting translocation of bacteria lipopolysaccharide (LPS) to cause endotoxaemia(Reference Cani, Osto and Geurts76, Reference Everard, Belzer and Geurts77). In diet-induced obese mice, live A. muciniphila administration decreased serum LPS, reduced adipose tissue inflammation and increased expression of the antimicrobial peptide RegIIIγ in the colon. This intervention also reverses the thinning of the mucus layer observed in obesity(Reference Everard, Belzer and Geurts77). Obesity-related disease is also associated with an altered profile of microbial metabolites, such as lower levels of SCFA and bile salt hydrolases, and raised numbers of sulphide-reducing bacteria. Recent animal studies also underline the ability of the gut microbiota to metabolise dietary PUFA(Reference Druart, Bindels and Schmaltz78).

In diet-induced obese animals, probiotic administration results in loss of body weight and/or fat mass and may also improve blood lipid levels, glycaemia or other metabolic disorders(Reference Kang, Yun and Park79Reference Chen, Wang and Li82). In mice fed a high-fat diet, probiotics shifted the aberrant gut microbiota profile towards that of lean mice on a normal diet. Strains differentially attenuated the induced obesity, inflammation and the metabolic syndrome(Reference Wang, Tang and Zhang83). Human studies, however, have not been as convincing. Only a few have shown probiotic benefit, with only minor effects on the body weight and fat mass but better effects on blood lipids and metabolic response to diet(Reference Kadooka, Sato and Ogawa84Reference Druart, Alligier and Salazar86). A recent study with Lactobacillus casei Shirota has indicated the potential of certain probiotics in a preventative strategy for overfeeding-induced insulin resistance(Reference Hulston, Churnside and Venables87); however, Professor Delzenne recommended development of novel commensal strains as well as butyrate-producing(Reference Weng, Endo and Li88) and CLA-producing probiotics(Reference Coakley, Ross and Nordgren89).

Prebiotics affect not just bifidobacteria numbers but also the overall richness and diversity of microbial functions and species in the gut. Obesity studies in mice have shown that prebiotics changed the ratio of Firmicutes:Bacteroidetes and modified over 100 microbial taxa(Reference Everard, Lazarevic and Derrien90). High-fat diet and prebiotic intervention also influence host antimicrobial peptides(Reference Everard, Lazarevic and Gaia91) and gut endocrine function(Reference Geurts, Neyrinck and Delzenne92). A 2013 review, which identified six prebiotic trials in overweight or obese people and three in non-alcoholic steatohepatitis or T2D, concluded that prebiotics increased feelings of satiety and reduced postprandial plasma glucose and insulin concentrations(Reference Kellow, Coughlan and Reid93). Effects of prebiotics on endotoxaemia, fat mass, gut hormones, cardiovascular and hepatic health were also reported, which may be linked to microbial changes. A study on obese women found that an inulin-type fructans changed the gut microbiota by increasing levels of Bifidobacterium and F. prausnitzii (which correlated with reduced serum LPS) and reducing Bacteroides intestinalis, Bacteroides vulgatus and Propionibacterium (which correlated with slight decreases in fat mass, and plasma lactate and phosphatidylcholine levels)(Reference Dewulf, Cani and Claus94). Another study on obese women showed levels of B. longum and A. muciniphila correlated with reductions in plasma LPS, and Bifidobacterium adolescentis with decreases in fat mass. Perhaps surprisingly, the prebiotic also decreased SCFA levels (raised SCFA could be a metabolic risk factor for obese women)(Reference Salazar, Dewulf and Neyrinck95). The observed link between some PUFA-derived bacterial metabolites (e.g. CLA), certain gut species and host metabolism is not yet fully understood(Reference Druart, Dewulf and Cani96).

Dr Carl Hulston (Loughborough University, UK) continued the theme of the gut microbiota’s influence on obesity and T2D(Reference Larsen, Vogensen and van den Berg97, Reference Cani, Possemiers and Van de Wiele98) in a short talk on his recent study into the effects of a probiotic on health risks associated with poor diet(Reference Hulston, Churnside and Venables87). In an RCT, a test group drank an L. casei Shirota fermented milk twice daily for 4 weeks, whereas a control group received no supplementation. All ate their normal diet for 3 weeks and then consumed a high-fat high-energy diet for 1 week. Compared with baseline, the week of overfeeding resulted in an increase of glucose AUC in an oral glucose tolerance test and elevated fasting plasma glucose, as well as a decrease in insulin sensitivity for the control group, whereas all these parameters were maintained in the probiotic group. Fasting serum insulin did not change in either group. These results suggest that probiotics may be useful in preventing diet-induced metabolic diseases but need to be confirmed in a larger study; the mechanism of activity also needs to be investigated.

Manipulation of the gut–brain axis: the emergence of psychobiotic therapy

Professor John Cryan (University College Cork, Ireland) reviewed research into the microbiota-gut–brain axis and explained that the gut microbiota is considered ‘the conductor of the orchestra of immune-neuroendocrine communication’, essential for normal stress, antidepressant and anxiety responses(Reference El Aidy, Dinan and Cryan99, Reference Stilling, Bordenstein and Dinan100). Unfortunately, the majority of evidence in this field is still from animal studies: either germ-free, subjected to early-life modulation of the gut microbiota, or exposed to specific pathogens, probiotics or antibiotics(Reference Cryan and Dinan101). These studies have shown that stress in early life not only alters behaviour but also causes key changes in the immune system and the gut microbiota, which are themselves linked to changes in colonic transit time and morphology, intestinal permeability and mucosal inflammation(Reference O’Mahony, Marchesi and Scully102, Reference O’Malley, Julio-Pieper and Gibney103). Perturbation of the postnatal microbial colonisation process affects neurodevelopment, with possible consequences for later mental health(Reference Borre, O’Keeffe and Clarke104).

The gut microbiota influences the brain and nervous system via several routes, including the vagus nerve, the spinal cord, and the immune and neuroendocrine systems. Gut bacteria produce several neuroactive molecules – for example, γ-aminobutyric acid, serotonin, catecholamines and acetylcholine(Reference Wall, Cryan and Ross105) – and may also affect the early development of the hypothalamic–pituitary–adrenal reaction to stress(Reference Sudo, Chida and Aiba106). The importance of the gut microbiota for brain development and behaviour is clearly shown by germ-free animals, which display less anxiety and neurochemical differences(Reference Diaz Heijtz, Wang and Anuar107, Reference Neufeld, Kang and Bienenstock108). Male germ-free animals show changes in the hippocampal serotonergic system, not reversible with restoration of the gut microbiota(Reference Clarke, Grenham and Scully109). Animal models have also shown that the gut microbiota influences regulation of neurogenesis in the adult hippocampus(Reference Ogbonnaya, Clarke and Shanahan110) and blood–brain barrier permeability (reversible with the introduction of butyrate-producing bacteria)(Reference Braniste, Al-Asmakh and Kowal111). It may also act as an epigenetic regulator of brain function, which may have a bearing on neurodevelopmental disorders such as autism. SCFA such as butyrate can alter the function of enzymes that modify histone proteins, thus changing gene expression(Reference Stilling, Dinan and Cryan112).

Development of probiotics with psychotrophic effects would be facilitated by charting developmental events in the brain through life and relating this to parallel changes in the gut microbiota and its metabolites(Reference Borre, O’Keeffe and Clarke104). Animal studies indicate the benefit of lactobacilli and bifidobacteria probiotics, with the vagus nerve as the key route for effects(Reference Bravo, Forsythe and Chew113, Reference Savignac, Tramullas and Kiely114).

Dysbiosis may be an underlying factor in the gut problems frequently experienced by people with autism spectrum disorder (ASD). The gut microbiota is essential for normal social development in mice(Reference Desbonnet, Clarke and Shanahan115), and alterations in the gut microbiota and function were associated with autism-like behaviour in a murine ASD model, particularly in males(Reference de Theije, Wopereis and Ramadan116). This has promoted interest in probiotics for ameliorating both gut and behavioural symptoms(Reference Hsiao, McBride and Hsien117, Reference Gilbert, Krajmalnik-Brown and Porazinska118). FMT has also been shown to change brain chemistry and behaviour in germ-free recipient mice(Reference Collins, Kassam and Bercik119). One outcome from transplanting from mice on a high-fat diet to those on normal feed was a negative change in neurocognitive behaviour(Reference Bruce-Keller, Salbaum and Luo120).

IBS is considered a disorder of the gut–brain axis, and microbiome research confirms this. Transient dysbiosis early in life affects visceral sensitivity, increasing the risk for later developing IBS(Reference O’Mahony, Felice and Nally121). The gut microbiota profile also correlates with the clinical phenotype of patients(Reference Jeffery, O’Toole and Ohman122). Probiotic benefit has been demonstrated in several animal studies(Reference McKernan, Fitzgerald and Dinan123), and human study evidence is accumulating, particularly with lactic acid bacteria(Reference Clarke, Cryan and Dinan124). IBS patients also appear to have a subtle deficit in cognitive behaviour linked to changes in cortisol levels; perhaps this is another target for probiotics(Reference Kennedy, Clarke and O’Neill125).

Finally, Professor Cryan introduced the term ‘psychobiotics’ for probiotics aimed at psychiatric illness(Reference Dinan, Stanton and Cryan126). Since a first report in 1910(Reference Phillips127), data are now accumulating on improvements in mental health parameters associated with lactobacilli and bifidobacteria(Reference Benton, Williams and Brown128Reference Mohammadi, Jazayeri and Khosravi-Darani131).

Prostate cancer

Professor Hideyuki Akaza (The University of Tokyo, Japan) began by outlining the geographical differences in prostate cancer (PC) incidence. Although PC is much lower in Asian countries compared with Western countries(Reference Center, Jemal and Lortet-Tieulent132), Japanese migrants in Hawaii have an increased PC incidence compared with Japanese people still living in Japan. This observation suggested that lifestyle factors, and not just diet, may be involved in determining disease risk(Reference Kolonel, Altshuler and Henderson133). As this cancer progresses very slowly (up to 27 years), preventive interventions are worth pursuing(Reference Cuzick, Thorat and Andriole134). It may not be cost-effective or even safe for drugs such as 5α-reductase inhibitors(Reference Lacy and Kyprianou135) to be taken over long periods, making dietary interventions a better option.

In 1998, a detailed examination of UN data for fifty-nine countries on PC-related mortality, dietary intake and lifestyle factors highlighted the particular benefit of soya consumption in protecting against the disease(Reference Hebert, Hurley and Olendzki136), a finding confirmed by later meta-analysis(Reference Yan and Spitznagel137). The active ingredients in soya are isoflavones, such as genistein, daidzein and equol(Reference Mahmoud, Yang and Bosland138). The anti-androgen activity of equol is explained by its ability to bind to oestrogen receptor β and the sex hormone-binding globulin (SHBG), thus inhibiting the growth of sex hormone-dependent tumours such as PC(Reference Lund, Munson and Haldy139).

Certain bacterial species in the colon are able to metabolise daidzein to produce equol, but these species are only carried by about 30 % of people consuming a Western diet, which is low in soya(Reference Rafii140). Professor Akaza’s group conducted a study on healthy Japanese people (both equol producers and non-producers), given 60 mg soya isoflavones daily for 3 months, which caused an increase in serum levels of equol and SHBG and a decrease in free testosterone and dihydrotestosterone(Reference Tanaka, Fujimoto and Chihara141). The results also indicated that prolonged consumption of soya isoflavone might convert non-equol producers to equol producers; this may be an important finding as the lack of ability to metabolise daidzein to equol in the colon appears to be a PC risk factor(Reference Sugiyama, Masumori and Fukuta142). Numbers of equol producers are low in Western countries, where PC risk is higher compared with countries such as Japan and Korea. Worryingly, the ability to produce equol appears to be dropping in the Japanese population, which could be linked to a parallel rise in PC incidence(Reference Fujimoto, Tanaka and Hirao143). A phase II placebo-controlled pilot trial in men with rising prostate-specific antigen showed that dietary supplementation of isoflavone for 12 months significantly increased serum levels of equol in subjects known to produce this metabolite. In subjects aged 65 years or older, the incidence of PC was also lower in the test group compared with the placebo group(Reference Miyanaga, Akaza and Hinotsu144).

A novel Slackia spp. strain NATTS – a gram-positive bacterium belonging to the phylum Actinobacteria and isolated from Japanese adults – can produce high levels of equol(Reference Tsuji, Moriyama and Nomoto145). Research is now focusing on understanding the global distribution of equol-producing strains and comparing this with PC risk. Large-scale intervention studies are also planned, which will investigate men at high risk of PC and the potential benefit of intervention with soya foods, equol supplements and/or foods containing the Slackia strain or its daidzein-metabolising enzymes. The realisation of a link between components of the intestinal microbiota and cancer risk reduction will support the development of a new research field(Reference Akaza146).

Antibiotic-associated diarrhoea in spinal cord injury patients

People with spinal cord injury (SCI) face a range of health problems, explained Dr Samford Wong (Stoke Mandeville Hospital, UK), which is why quality of life is a key research focus. Patients are encouraged to participate in sport and supported into re-employment but complications, such as malnutrition, affect clinical outcome(Reference Wong, Derry and Jamous147). Use of catheters increases risk of urinary tract infection as well as diarrhoea resulting from antibiotic treatment delays rehabilitation, which affects the patient and increases their health-care costs. This is why cost-effective, reliable and simple measures are needed. Probiotics have potential for preventing AAD(Reference Issa and Moucari148) and in fact are already being used in some SCI centres. However, medical staff are not always aware that efficacy can be strain-, dose- and disease-specific.

Recently an RCT in SCI patients conducted by Dr Wong demonstrated that a fermented milk drink containing L. casei Shirota significantly prevented AAD, which developed in 54·9 % of patients in the control group compared with 17·1 % given the probiotic(Reference Wong, Jamous and O’Driscoll149). To fully evaluate probiotic potential for SCI patients, Dr Wong proposed a three-step approach: (i) to evaluate current AAD and C. difficile-AD (CDAD) practices in UK and European SCI centres; (ii) to conduct a systematic review and meta-analysis on their effectiveness in preventing AAD/CDAD in SCI patients; and (iii) to conduct a multicentre, double-blind, placebo-controlled trial to confirm the benefit of L. casei Shirota.

HIV

HIV/AIDS is characterised by a progressive depletion of CD4+ T-cells and a severe impairment of the immune system. This is often accompanied by an alteration of the gut mucosal barrier, which allows translocation of microbes and their products, and provokes a chronic state of inflammation(Reference Brenchley, Price and Schacker150). In view of the immunomodulatory effects reported for probiotics in non-immunocompetent and HIV+ subjects(Reference Cunningham-Rundles, Ahrne and Johann-Liang151, Reference Dong, Rowland and Thomas152), Dr D’Angelo’s (‘G. d’Annunzio’ University of Chieti-Pescara, Italy) group conducted a pilot study on the effects of L. casei Shirota (at 1·3×1010 CFU/d for 4 weeks) on clinically stable HIV+ patients on antiretroviral therapy(Reference Falasca, Vecchiet and Ucciferri153). Blood samples were taken before and after the intervention. Haematological (lymphocyte subsets), immunological (circulating cytokine levels and mRNA expression in peripheral blood mononuclear cells) and other parameters (LPS and cystatin C) were measured. The probiotic was associated with slightly reduced plasma LPS, significantly increased CD56+ and significantly reduced mRNA levels of IL-1β, IL-10, IL-12 and transforming growth factor-β in peripheral blood mononuclear cells, while plasma IL-23 increased. An observed decrease of cystatin C also indicated that cardiovascular risk might be reduced. Dr D’Angelo concluded that this preliminary evidence of an improvement in systemic inflammatory cytokines suggests that this probiotic could improve the management of antiretroviral therapy-treated HIV+ patients and warrants further investigation.

Probiotic mechanisms: immune modulation and effects of metabolites

Faecalibacterium prausnitzii

This bacterium, the subject of a talk by Professor Philippe Langella (INRA, France), is an ubiquitous commensal species comprising >3·5 % of the gut microbiota, which is extremely oxygen sensitive and a butyrate producer. It belongs to the gram-positive Firmicutes phylum and Clostridium leptum group cluster IV(Reference Miquel, Martin and Bridonneau154Reference Qin, Li and Raes156). Reduced abundance and diversity of Firmicutes occur in CD patients. A landmark study by Sokol et al.(Reference Sokol, Pigneur and Watterlot157) in 2008 also demonstrated an association between reduced F. prausnitzii numbers and increased risk for postoperative recurrence of ileal CD. The species was also shown to have anti-inflammatory effects in cellular and animal models of gut inflammation(Reference Sokol, Pigneur and Watterlot157). A different strategy to find new probiotic strains was proposed: identify candidate species whose presence/absence is linked to health in diseases linked to microbial dysbiosis. One such species is F. prausnitizii. Having been linked to dysbiosis in CD, UC, colorectal cancer and IBS, it could almost be an indicator of intestinal health(Reference Cao, Shen and Ran158Reference Rajilic-Stojanovic, Biagi and Heilig160). A novel, chronic dinitrobenzene sulphonic acid (DNBS)-induced acute and chronic colitis model in mice developed by Professor Langella’s group showed that F. prausnitzii and its supernatant protected the gut epithelium during episodes of chronic colitis and its reactivation(Reference Martin, Chain and Miquel161).

The same DNBS-induced mouse model was adapted to better mimic IBS, with cycles of low-grade, sub-clinical inflammation and then a period of recovery followed by reactivation of inflammation. The IBS mice had raised levels of serotonin and inflammatory cytokines in the colon (IL-6, interferon-γ (IFN-γ), IL-4 and IL-22), as well as impaired gut permeability, but these effects were reversed following treatment with F. prausnitzii or its supernatant(Reference Martin, Miquel and Chain162). The species was also tested in a neonatal maternal separation model that induces colonic hypersensitivity and increases gut permeability. In this pain model, F. prausnitzii strain A2–165 (now patented) demonstrated antinociceptive effects and restored gut permeability.

A further F. prausnitzii gnotobiotic model has been developed, where dual colonisation with E. coli achieves more effective and stable colonisation by F. prausnitzii. When colitis was induced in this model, disease activity and other parameters were improved by the presence of F. prausnitzii. The species was also shown to be very metabolically active, producing several compounds with beneficial effects, such as shikimic and salicylic acids. Both have anti-pain activity; the latter is also a precursor of 5-aminosalicylic acid, the anti-inflammatory drug used to treat IBD(Reference Miquel, Leclerc and Martin163).

The next stage is to develop F. prausnitzii as a human probiotic, but first a few obstacles have to be overcome. The species is considered to be a novel food; hence, full toxicology assays and characterisation of the strain are needed for regulatory approval. The freeze-drying process for commercial preparation of the strain also needs to be optimised. Work is continuing to fully understand the species, its mode of action and its bioactive metabolites.

Immunomodulatory mechanisms of probiotics

Dr Liam O’Mahony (Swiss Institute of Allergy and Asthma Research, Switzerland) explained that immune tolerance in the gut evolved in order to minimise the impact on the host of dealing with an invader and to allow the commensal microbiota to colonise the gut, where it provides nutrients to and protection for the host(Reference Read, Graham and Raberg164, Reference Maloy and Powrie165). In conditions of gut dysbiosis, however, even commensal bacteria can trigger an inappropriate inflammatory response(Reference Round and Mazmanian166). For example, recognition of segmented filamentous bacteria by the mucosal immune system can, in the wrong circumstances, lead to T-helper 17 cell-mediated inflammation(Reference Atarashi, Umesaki and Honda167). Gut microbiota colonisation in early-life influences the risk for immune disorders; allergy development in germ-free mice, for instance, depends on what age the mice are colonised with bacteria(Reference Olszak, An and Zeissig168). Mice studies show that food allergy is associated with a particular profile of gut microbiota; this triggered allergy and anaphylaxis when transplanted to germ-free mice(Reference Noval Rivas, Burton and Wise169).

Dr O’Mahony’s group is elucidating the cellular and molecular effects of an immunoregulatory probiotic, B. longum subsp. infantis 35624(Reference Konieczna, Akdis and Quigley170). In mice, its consumption resulted in induction of T-regulatory cells and attenuation of NF-κB activation, preventing excessive inflammation induced by Salmonella infection(Reference O’Mahony, Scully and O’Mahony171). Induction of T-regulatory cells by the strain has also been shown in humans, as well as reduction of systemic pro-inflammatory biomarkers in patients with psoriasis, IBS, chronic fatigue syndrome or UC(Reference Groeger, O’Mahony and Murphy172). In the peripheral blood, the strain stimulates dendritic cell-induced T-regulatory cells to produce IL-10 and also enhances Foxp3 expression. It also reduces production of IL-12 and TNF-α, effects which all appear to be strain-specific(Reference Konieczna, Groeger and Ziegler173, Reference Lyons, O’Mahony and O’Brien174).

Bacterial metabolites can also exert immunological effects. The biogenic amine histamine, which is found at particularly high levels in the gut mucosa of IBS and IBD patients, can promote either pro- or anti-inflammatory effects depending on which of its four receptors are activated(Reference O’Mahony, Akdis and Akdis175, Reference Frei, Ferstl and Konieczna176). Not all commensal bacteria, however, express histidine decarboxylase (the enzyme needed to convert histidine to histamine). The isolate Lactobacillus saerimneri strain 30a produces high levels of biologically active histamine(Reference Ferstl, Frei and Schiavi177). Feeding this strain to wild-type mice and mice lacking histamine receptor 2, for example, resulted in a deterioration in health. Current research is investigating the ability of the gut microbiota to produce histamine, as this may have health implications.

SCFA are other types of bacterial metabolites with immunoregulatory effects. Dr O’Mahony’s group has used an ovalbumin-challenge respiratory allergy model to show that butyrate reduces pro-inflammatory cytokines such as IL-17, which can be raised with asthma. Dr O’Mahony stressed that the timing of any intervention relating to immune benefit may be critical; the greatest effects are likely to be early in life(Reference Roduit, Frei and Loss178).

Campylobacter infection

Although Campylobacter is the most common bacterial cause of enteritis in industrialised countries, Professor Stefan Bereswill (Charité – University Medicine Berlin, Germany) explained that its pathogenicity is still not fully understood. Although Campylobacter jejuni can asymptomatically colonise animals such as chickens and cows, it causes inflammation and acute diarrhoea in humans, and rare but serious post-infectious immune-related disorders such as Guillain–Barré syndrome(Reference Young, Davis and Dirita179). Mice with a normal gut microbiota display a strong colonisation resistance to C. jejuni; hence, finding a good animal model for this infection has proved a challenge for researchers. Wild-type and GM mice models have now been developed, where the commensal microbiota has been eradicated or modified (e.g. to one resembling a human gut microbiota)(Reference Bereswill, Fischer and Plickert180). Infant mice harbouring a conventional gut microbiota have much higher counts of commensal E. coli compared with adult animals, and overgrowth of the murine gut microbiota with E. coli abrogates colonisation resistance against C. jejuni (Reference Heimesaat, Plickert and Fischer181, Reference Haag, Fischer and Otto182). The pathogen readily colonises antibiotic-treated gnotobiotic mice with a knockout for IL-10 expression (i.e. IL-10−/−), causing enterocolitis and the development of LPS-mediated inflammation and specific T-cell responses that are characteristic of campylobacteriosis in humans(Reference Bereswill, Fischer and Plickert180, Reference Heimesaat and Bereswill183). Studies on these mice have shown that C. jejuni is a potent activator of the innate immune response via toll-like receptor (TLR) 4 and, to a lesser extent, TLR2 (these receptors detect LPS and lipoprotein molecules on the bacterial surface). TLR4 and TLR2 also appear to be involved in the ability of the pathogen to induce inflammation and apoptosis of enterocytes(Reference Haag, Fischer and Otto184). The addition of a sialic acid derivative to the surface LPS of C. jejuni also aggravates its interaction with TLR, increasing the invasion, translocation and inflammatory potential of the pathogen and thereby raising the risk of post-infectious complications(Reference Mortensen, Kuijf and Ang185). Several cytokines have been identified as important in protection against Campylobacter. Investigations using human biopsy material, for example, have shown that the innate and adaptive T-cell immune responses to C. jejuni are associated with production of IFN-γ, IL-22 and IL-17A(Reference Edwards, Nistala and Mills186). The Gastrointestinal Microbiology Research Group at Charité (headed by Markus Heimesaat and Stefan Bereswill) has shown that IL-22 and IL-23 are essential for maintaining the composition of the intestinal microbiota; this then helps maintain colonisation resistance. Current work is aimed at analysing the pro- and anti-inflammatory T-cell subsets in murine infections and in IL-22−/− and IL-23−/− mice. C. jejuni infection in 3-week-old mice results in a self-limiting illness, but neutrophil infiltration can be observed in the colon, liver, lungs and kidneys, and this can lead to a state of chronic inflammation in asymptomatic carriers. The similarity of this pathology to the human disease with its serious postoperative complications means that these novel animal models will be extremely useful tools for C. jejuni research and perhaps development of probiotics to help prevent this infection.

Butyric acid, the oral microbiota and ageing

A complex association exists between periodontal disease and certain systemic disorders such as arteriosclerosis, T2D, pneumonia, heart disease and premature childbirth(Reference Kholy, Genco and Van Dyke187Reference Olsen189). Professor Kuniyasu Ochiai’s (Nihon University School of Dentistry, Japan) group are researching into this, focusing on the role of butyric acid (BA), which is produced at high levels by periodontopathic plaque species such as Porphyromonas gingivalis, Fusobacterium nucleatum and Eubacterium (Reference Niederman, Buyle-Bodin and Lu190, Reference Kurita-Ochiai, Fukushima and Ochiai191). BA accumulates in gingival crevices at higher levels in periodontitis cases compared with the healthy(Reference Niederman, Buyle-Bodin and Lu190) and could serve as a promoter of periodontitis(Reference Yoneda, Kawarai and Narisawa192, Reference Arai, Ochiai and Senpuku193). At low concentrations, BA stimulates cell growth, but at higher concentrations it induces apoptosis in neutrophils, T-cells and macrophages(Reference Kurita-Ochiai, Fukushima and Ochiai191, Reference Kurita-Ochiai, Ochiai and Fukushima194, Reference Kurita-Ochiai, Amano and Fukushima195). T-cells survive by adhering to fibroblasts(Reference Kurita-Ochiai, Seto and Ochiai196) as a result of increased expression of adhesion molecules plus increased production of pro-inflammatory cytokines IL-6 and IL-11 by the gingival fibroblasts. Although fibroblasts are usually resistant to BA-induced apoptosis, cells from periodontitis patients are highly sensitive(Reference Kurita-Ochiai, Seto and Suzuki197).

The complex signalling network associated with oral BA shows how it may elicit systemic effects, which could influence ageing and latent infections. Animal studies have shown that BA-induced primary signalling affects total haem, NADP and hydrogen peroxide(Reference Cueno, Imai and Tamura198). These compounds are linked to the free radical theory of ageing(Reference Cueno, Tamura and Ochiai199), reactivation of latent infections such as HIV(Reference Imai, Yamada and Tamura200), periodontal disease(Reference Cueno, Imai and Matsukawa201) and promotion of P. gingivalis (Reference Cueno, Tamura and Ohya202). Secondary signalling affects cytosolic and serum levels of Ca (factors associated with osteoporosis and Alzheimer’s disease)(Reference Cornelius, Koverech and Crupi203, Reference Cueno, Kamio and Seki204) and caspases (proteases important for apoptosis and inflammation)(Reference Shalini, Dorstyn and Dawar205). The late network caused by BA affects the oxidised form of NAD, sirtuin1 and the mammalian target of rapamycin, and is linked to the ageing processes(Reference Cueno, Imai and Tamura198, Reference Cueno, Tamura and Ochiai199).

In HIV-infected patients, degree of periodontitis correlates to HIV-RNA load in gingival crevicular fluid(Reference Maticic, Poljak and Kramar206). Professor Ochiai’s group found that the BA produced by P. gingivalis acts as an epigenetic regulator, inhibiting the histone deacetylases that maintain HIV latency(Reference Imai, Ochiai and Okamoto207). Severe periodontitis could trigger a synergy between the local effects of BA in gingival tissue and systemic effects of induced TNF-α. An epidemiological study has started in Japan to confirm whether periodontal disease is a risk factor for HIV reactivation. Epstein–Barr virus (EBV), another latent infection, has been isolated from patients with periodontitis(Reference Saygun, Nizam and Keskiner208, Reference Konstantinidis, Sakellari and Papa209), and BA in P. gingivalis culture supernatant can also reactivate EBV in vitro by a mechanism involving histone modification and chromatin remodelling(Reference Imai, Inoue and Tamura210).

These findings show BA in a new light. Although it is considered beneficial in the gut, Professor Ochiai debated whether we need to understand the effects of high levels of SCFA on gut tissues. His final message was very clear: brush your teeth!

A re-examination of the probiotic definition and category

‘Probiotic’ means different things to different people depending on their particular interest, explained Professor Colin Hill (University College Cork, Ireland). Some health conditions (e.g. AAD) respond to several different probiotics(Reference Hempel, Newberry and Maher50), yet some probiotics are associated with several health benefits(Reference Segers and Lebeer211), and a further complication is that some effects are very strain-specific(Reference Elazab, Mendy and Gasana212). In response to concerns about regulatory developments in Europe as well as misuse of the term ‘probiotic’ and confusion of general public about this category, the International Scientific Association for Probiotics and Prebiotics organised a meeting in 2013 to re-examine the probiotic concept. The panel comprised clinical and scientific experts, including members of an original FAO/WHO expert panel, who in 2001 had agreed what has been a widely accepted definition for probiotics(213). Professor Hill gave an insight into these discussions and the resulting consensus statement, published in 2014(Reference Hill, Guarner and Reid214).

The panel endorsed the 2001 probiotic definition, with just a small change to improve its grammar to: ‘Live micro-organisms that, when administered in adequate amounts, confer a health benefit on the host’. They recommended that ‘probiotic’ should only apply to products containing a suitable number of live cells of well-defined and safe microbial strains and with a reasonable level of scientific evidence of health benefit from a body of research that included well-conducted human studies. The panel agreed that the process of assessment of scientific evidence for food probiotics should differ from the assessment for pharmaceutical drugs, and the process should be consistent with other foods that have approved health claims (e.g. vitamins). The panel agreed that changes are needed by regulators in jurisdictions such as the USA and Europe(Reference Binnendijk and Rijkers215).

The panel identified three categories of probiotics:

  • Microbial species, used in a food or food supplement, without a specific health claim

    • with reasonable evidence of a general benefit for humans associated with the species itself and/or its function, structure, activity or end product;

    • claim: ‘contains probiotics’.

  • Microbial strains, used in/as a food or food supplement, with a specific health claim

    • based on convincing evidence specific to the constituent strain(s) to satisfy the appropriate regulatory authority.

  • Microbial strains, used as probiotic drugs

    • sufficient trial evidence to meet appropriate regulatory standards for drugs.

A very broad range of bacterial species have been associated with a robust gut microbiota; therefore, it is very likely that the probiotic framework could be extended to include new well-defined beneficial microbes, such as A. muciniphila and F. prausnitzii, but this could only happen when/if there is sufficient evidence of the benefit and understanding of mechanism of activity.

Dead microbes or microbial products are not probiotic. Live microbial cultures with no evidence of benefit that are traditionally associated with fermented foods also fall outside the probiotic framework and should be labelled as ‘containing live and active cultures’. FMT cannot be considered probiotic, because this involves an undefined mixture of micro-organisms. FMT preparations containing defined mixtures of microbes, such as RePOOPulate, however, do meet the probiotic criteria(Reference Petrof, Gloor and Vanner216).

The panel supported the concept that certain well-studied species could impart general benefits, particularly for gut health but not for immune health. This has been concluded in meta-analyses(Reference Ritchie and Romanuk217) and acknowledged in Canada where non-strain-specific claims are allowed for various species of Lactobacillus and Bifidobacterium (218). There were more tentative conclusions regarding mechanisms of activity: some (such as colonisation resistance and SCFA production) were considered to be widespread among studied probiotics; others were considered to occur frequently within the same species (such as vitamin synthesis and gut barrier reinforcement); and others were considered to be rare and strain-specific (such as neurological and immunological effects).

The consensus statement is valuable in providing clear guidelines for all stakeholders in the probiotics sector, be they researchers, regulators, health-care professionals, manufacturers or consumers.

Acknowledgements

The 8th International Yakult Symposium was financially supported by Yakult Europe B.V., Almere, The Netherlands. The present report was commissioned by Yakult Europe B.V.

L. V. T., K. S. and J. Z. contributed to the writing of the report.

None of the authors has any conflicts of interest to declare.

References

1.El Aidy, S, van den Bogert, B & Kleerebezem, M (2015) The small intestine microbiota, nutritional modulation and relevance for health. Curr Opin Biotechnol 32, 1420.CrossRefGoogle ScholarPubMed
2.van der Schaar, PJ, Dijksman, JF, Broekhuizen-de Gast, H, et al. (2013) A novel ingestible electronic drug delivery and monitoring device. Gastrointest Endosc 78, 520528.CrossRefGoogle ScholarPubMed
3.Rajilic-Stojanovic, M, Heilig, HG, Molenaar, D, et al. (2009) Development and application of the human intestinal tract chip, a phylogenetic microarray: analysis of universally conserved phylotypes in the abundant microbiota of young and elderly adults. Environ Microbiol 11, 17361751.CrossRefGoogle Scholar
4.Booijink, CC, El-Aidy, S, Rajilic-Stojanovic, M, et al. (2010) High temporal and inter-individual variation detected in the human ileal microbiota. Environ Microbiol 12, 32133227.CrossRefGoogle ScholarPubMed
5.van den Bogert, B, de Vos, WM, Zoetendal, EG, et al. (2011) Microarray analysis and barcoded pyrosequencing provide consistent microbial profiles depending on the source of human intestinal samples. Appl Environ Microbiol 77, 20712080.CrossRefGoogle ScholarPubMed
6.Zoetendal, EG, Raes, J, van den Bogert, B, et al. (2012) The human small intestinal microbiota is driven by rapid uptake and conversion of simple carbohydrates. ISME J 6, 14151426.CrossRefGoogle ScholarPubMed
7.van den Bogert, B, Erkus, O, Boekhorst, J, et al. (2013) Diversity of human small intestinal Streptococcus and Veillonella populations. FEMS Microbiol Ecol 85, 376388.CrossRefGoogle ScholarPubMed
8.van Baarlen, P, Troost, F, van der Meer, C, et al. (2011) Human mucosal in vivo transcriptome responses to three lactobacilli indicate how probiotics may modulate human cellular pathways. Proc Natl Acad Sci U S A 108, Suppl. 1, 45624569.CrossRefGoogle ScholarPubMed
9.van Baarlen, P, Troost, FJ, van Hemert, S, et al. (2009) Differential NF-kappaB pathways induction by Lactobacillus plantarum in the duodenum of healthy humans correlating with immune tolerance. Proc Natl Acad Sci U S A 106, 23712376.CrossRefGoogle ScholarPubMed
10.Yan, F, Cao, H, Cover, TL, et al. (2007) Soluble proteins produced by probiotic bacteria regulate intestinal epithelial cell survival and growth. Gastroenterology 132, 562575.CrossRefGoogle ScholarPubMed
11.Lamb, J (2007) The connectivity map: a new tool for biomedical research. Nat Rev Cancer 7, 5460.CrossRefGoogle ScholarPubMed
12.Bron, PA, van Baarlen, P & Kleerebezem, M (2012) Emerging molecular insights into the interaction between probiotics and the host intestinal mucosa. Nat Rev Microbiol 10, 6678.CrossRefGoogle Scholar
13.Hedin, CR, Stagg, AJ, Whelan, K, et al. (2012) Family studies in Crohn’s disease: new horizons in understanding disease pathogenesis, risk and prevention. Gut 61, 311318.CrossRefGoogle ScholarPubMed
14.Frank, DN, St Amand, AL, Feldman, RA, et al. (2007) Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A 104, 1378013785.CrossRefGoogle ScholarPubMed
15.Lepage, P, Hasler, R, Spehlmann, ME, et al. (2011) Twin study indicates loss of interaction between microbiota and mucosa of patients with ulcerative colitis. Gastroenterology 141, 227236.CrossRefGoogle ScholarPubMed
16.Renz, H, von Mutius, E, Brandtzaeg, P, et al. (2011) Gene-environment interactions in chronic inflammatory disease. Nat Immunol 12, 273277.CrossRefGoogle ScholarPubMed
17.Hilsden, RJ, Verhoef, MJ, Rasmussen, H, et al. (2011) Use of complementary and alternative medicine by patients with inflammatory bowel disease. Inflamm Bowel Dis 17, 655662.CrossRefGoogle ScholarPubMed
18.Fernandez-Banares, F, Hinojosa, J, Sanchez-Lombrana, JL, et al. (1999) Randomized clinical trial of Plantago ovata seeds (dietary fiber) as compared with mesalamine in maintaining remission in ulcerative colitis. Spanish Group for the Study of Crohn’s Disease and Ulcerative Colitis (GETECCU). Am J Gastroenterol 94, 427433.CrossRefGoogle Scholar
19.Fujimori, S, Tatsuguchi, A, Gudis, K, et al. (2007) High dose probiotic and prebiotic cotherapy for remission induction of active Crohn’s disease. J Gastroenterol Hepatol 22, 11991204.CrossRefGoogle ScholarPubMed
20.Langhorst, J, Wulfert, H, Lauche, R, et al. (2015) Systematic review of complementary and alternative medicine treatments in inflammatory bowel diseases. J Crohns Colitis 9, 86106.CrossRefGoogle ScholarPubMed
21.Dignass, A, Preiss, JC, Aust, DE, et al. (2011) [Updated German guideline on diagnosis and treatment of ulcerative colitis, 2011]. Z Gastroenterol 49, 12761341.CrossRefGoogle ScholarPubMed
22.Storr, M, Devlin, S, Kaplan, GG, et al. (2014) Cannabis use provides symptom relief in patients with inflammatory bowel disease but is associated with worse disease prognosis in patients with Crohn’s disease. Inflamm Bowel Dis 20, 472480.CrossRefGoogle ScholarPubMed
23.Joos, S, Wildau, N, Kohnen, R, et al. (2006) Acupuncture and moxibustion in the treatment of ulcerative colitis: a randomized controlled study. Scand J Gastroenterol 41, 10561063.CrossRefGoogle ScholarPubMed
24.Rogler, G & Vavricka, S (2015) Exposome in IBD: recent insights in environmental factors that influence the onset and course of IBD. Inflamm Bowel Dis 21, 400408.CrossRefGoogle ScholarPubMed
25.Clarke, SF, Murphy, EF, O’Sullivan, O, et al. (2014) Exercise and associated dietary extremes impact on gut microbial diversity. Gut 63, 19131920.CrossRefGoogle ScholarPubMed
26.Cosnes, J (2010) Smoking, physical activity, nutrition and lifestyle: environmental factors and their impact on IBD. Dig Dis 28, 411417.CrossRefGoogle ScholarPubMed
27.Levenstein, S, Prantera, C, Varvo, V, et al. (2000) Stress and exacerbation in ulcerative colitis: a prospective study of patients enrolled in remission. Am J Gastroenterol 95, 12131220.CrossRefGoogle ScholarPubMed
28.Bernstein, CN, Singh, S, Graff, LA, et al. (2010) A prospective population-based study of triggers of symptomatic flares in IBD. Am J Gastroenterol 105, 19942002.CrossRefGoogle ScholarPubMed
29.Jedel, S, Hoffman, A, Merriman, P, et al. (2014) A randomized controlled trial of mindfulness-based stress reduction to prevent flare-up in patients with inactive ulcerative colitis. Digestion 89, 142155.CrossRefGoogle ScholarPubMed
30.Berrill, JW, Sadlier, M, Hood, K, et al. (2014) Mindfulness-based therapy for inflammatory bowel disease patients with functional abdominal symptoms or high perceived stress levels. J Crohns Colitis 8, 945955.CrossRefGoogle ScholarPubMed
31.Bailey, MT, Dowd, SE, Galley, JD, et al. (2011) Exposure to a social stressor alters the structure of the intestinal microbiota: implications for stressor-induced immunomodulation. Brain Behav Immun 25, 397407.CrossRefGoogle ScholarPubMed
32.van Nood, E, Vrieze, A, Nieuwdorp, M, et al. (2013) Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med 368, 407415.CrossRefGoogle ScholarPubMed
33.Kao, D, Hotte, N, Gillevet, P, et al. (2014) Fecal microbiota transplantation inducing remission in Crohn’s colitis and the associated changes in fecal microbial profile. J Clin Gastroenterol 48, 625628.CrossRefGoogle ScholarPubMed
34.Kump, PK, Grochenig, HP, Lackner, S, et al. (2013) Alteration of intestinal dysbiosis by fecal microbiota transplantation does not induce remission in patients with chronic active ulcerative colitis. Inflamm Bowel Dis 19, 21552165.CrossRefGoogle Scholar
35.Moayyedi, P, Surette, MG, Kim, PT, et al. (2015) Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized, controlled trial. Gastroenterology 149, 102109.CrossRefGoogle ScholarPubMed
36.Rossen, NG, Fuentes, S, van der Spek, MJ, et al. (2015) Findings from a randomized controlled trial of fecal transplantation for patients with ulcerative colitis. Gastroenterology 149, 110118.CrossRefGoogle ScholarPubMed
37.Jones, J & Hunter, D (1995) Consensus methods for medical and health services research. BMJ 311, 376380.CrossRefGoogle ScholarPubMed
38.Guyatt, GH, Oxman, AD, Schunemann, HJ, et al. (2011) GRADE guidelines: a new series of articles in the Journal of Clinical Epidemiology. J Clin Epidemiol 64, 380382.CrossRefGoogle ScholarPubMed
39.O’Mahony, L, McCarthy, J, Kelly, P, et al. (2005) Lactobacillus and Bifidobacterium in irritable bowel syndrome: symptom responses and relationship to cytokine profiles. Gastroenterology 128, 541551.CrossRefGoogle ScholarPubMed
40.Whorwell, PJ, Altringer, L, Morel, J, et al. (2006) Efficacy of an encapsulated probiotic Bifidobacterium infantis 35624 in women with irritable bowel syndrome. Am J Gastroenterol 101, 15811590.CrossRefGoogle ScholarPubMed
41.McFarland, LV & Dublin, S (2008) Meta-analysis of probiotics for the treatment of irritable bowel syndrome. World J Gastroenterol 14, 26502661.CrossRefGoogle ScholarPubMed
42.Ford, AC, Quigley, EM, Lacy, BE, et al. (2014) Efficacy of prebiotics, probiotics, and synbiotics in irritable bowel syndrome and chronic idiopathic constipation: systematic review and meta-analysis. Am J Gastroenterol 109, 15471561 quiz 1546, 1562.CrossRefGoogle ScholarPubMed
43.Chang, L, Lembo, A & Sultan, S (2014) American Gastroenterological Association Institute Technical Review on the pharmacological management of irritable bowel syndrome. Gastroenterology 147, 11491172.CrossRefGoogle ScholarPubMed
44.National Institute for Health and Care Excellence (2015) Irritable bowel syndrome in adults: diagnosis and management of irritable bowel syndrome in primary care. NICE Guidelines CG61. https://www.nice.org.uk/guidance/cg61/chapter/1-guidance (accessed August 2015).Google Scholar
45.Layer, P, Andresen, V, Pehl, C, et al. (2011) [Irritable bowel syndrome: German consensus guidelines on definition, pathophysiology and management]. Z Gastroenterol 49, 237293.CrossRefGoogle ScholarPubMed
46.Szajewska, H, Guarino, A, Hojsak, I, et al. (2014) Use of probiotics for management of acute gastroenteritis: a position paper by the ESPGHAN Working Group for Probiotics and Prebiotics. J Pediatr Gastroenterol Nutr 58, 531539.CrossRefGoogle Scholar
47.Saavedra, JM, Bauman, NA, Oung, I, et al. (1994) Feeding of Bifidobacterium bifidum and Streptococcus thermophilus to infants in hospital for prevention of diarrhoea and shedding of rotavirus. Lancet 344, 10461049.CrossRefGoogle ScholarPubMed
48.Szajewska, H, Wanke, M & Patro, B (2011) Meta-analysis: the effects of Lactobacillus rhamnosus GG supplementation for the prevention of healthcare-associated diarrhoea in children. Aliment Pharmacol Ther 34, 10791087.CrossRefGoogle ScholarPubMed
49.Hojsak, I, Tokic Pivac, V, Mocic Pavic, A, et al. (2015) Bifidobacterium animalis subsp. lactis fails to prevent common infections in hospitalized children: a randomized, double-blind, placebo-controlled study. Am J Clin Nutr 101, 680684.CrossRefGoogle ScholarPubMed
50.Hempel, S, Newberry, SJ, Maher, AR, et al. (2012) Probiotics for the prevention and treatment of antibiotic-associated diarrhea: a systematic review and meta-analysis. JAMA 307, 19591969.Google ScholarPubMed
51.AlFaleh, K & Anabrees, J (2014) Probiotics for prevention of necrotizing enterocolitis in preterm infants. Evid Based Child Health 9, 584671.CrossRefGoogle ScholarPubMed
52.Chau, K, Lau, E, Greenberg, S, et al. (2015) Probiotics for infantile colic: a randomized, double-blind, placebo-controlled trial investigating Lactobacillus reuteri DSM 17938. J Pediatr 166, 7478.CrossRefGoogle ScholarPubMed
53.Mi, GL, Zhao, L, Qiao, DD, et al. (2015) Effectiveness of Lactobacillus reuteri in infantile colic and colicky induced maternal depression: a prospective single blind randomized trial. Antonie Van Leeuwenhoek 107, 15471553.CrossRefGoogle ScholarPubMed
54.Sung, V, Hiscock, H, Tang, ML, et al. (2014) Treating infant colic with the probiotic Lactobacillus reuteri: double blind, placebo controlled randomised trial. BMJ 348, g2107.CrossRefGoogle ScholarPubMed
55.Urbanska, M & Szajewska, H (2014) The efficacy of Lactobacillus reuteri DSM 17938 in infants and children: a review of the current evidence. Eur J Pediatr 173, 13271337.CrossRefGoogle ScholarPubMed
56.Indrio, F, Di Mauro, A, Riezzo, G, et al. (2014) Prophylactic use of a probiotic in the prevention of colic, regurgitation, and functional constipation: a randomized clinical trial. JAMA Pediatr 168, 228233.CrossRefGoogle ScholarPubMed
57.Fiocchi, A, Pawankar, R, Cuello-Garcia, C, et al. (2015) World Allergy Organization-McMaster University guidelines for allergic disease prevention (GLAD-P): probiotics. World Allergy Organ J 8, 4.CrossRefGoogle ScholarPubMed
58.Aljada, A, Mohanty, P, Ghanim, H, et al. (2004) Increase in intranuclear nuclear factor kappaB and decrease in inhibitor kappaB in mononuclear cells after a mixed meal: evidence for a proinflammatory effect. Am J Clin Nutr 79, 682690.CrossRefGoogle ScholarPubMed
59.Nappo, F, Esposito, K, Cioffi, M, et al. (2002) Postprandial endothelial activation in healthy subjects and in type 2 diabetic patients: role of fat and carbohydrate meals. J Am Coll Cardiol 39, 11451150.CrossRefGoogle ScholarPubMed
60.Miglio, C, Peluso, I, Raguzzini, A, et al. (2013) Antioxidant and inflammatory response following high-fat meal consumption in overweight subjects. Eur J Nutr 52, 11071114.CrossRefGoogle ScholarPubMed
61.Peluso, I, Raguzzini, A, Villano, DV, et al. (2012) High fat meal increase of IL-17 is prevented by ingestion of fruit juice drink in healthy overweight subjects. Curr Pharm Des 18, 8590.CrossRefGoogle ScholarPubMed
62.Miglio, C, Peluso, I, Raguzzini, A, et al. (2014) Fruit juice drinks prevent endogenous antioxidant response to high-fat meal ingestion. Br J Nutr 111, 294300.CrossRefGoogle ScholarPubMed
63.Peluso, I, Miglio, C, Morabito, G, et al. (2015) Flavonoids and immune function in human: a systematic review. Crit Rev Food Sci Nutr 55, 383395.CrossRefGoogle ScholarPubMed
64.Serafini, M, Bugianesi, R, Maiani, G, et al. (2003) Plasma antioxidants from chocolate. Nature 424, 1013.CrossRefGoogle ScholarPubMed
65.Marin, L, Miguelez, EM, Villar, CJ, et al. (2015) Bioavailability of dietary polyphenols and gut microbiota metabolism: antimicrobial properties. Biomed Res Int 2015, 905215.CrossRefGoogle ScholarPubMed
66.Chen, Z, Zheng, S, Li, L, et al. (2014) Metabolism of flavonoids in human: a comprehensive review. Curr Drug Metab 15, 4861.CrossRefGoogle ScholarPubMed
67.Nagpal, R, Yadav, H & Marotta, F (2014) Gut microbiota: the next-gen frontier in preventive and therapeutic medicine? Front Med (Lausanne) 1, 15.Google ScholarPubMed
68.Ishii, Y, Sugimoto, S, Izawa, N, et al. (2014) Oral administration of Bifidobacterium breve attenuates UV-induced barrier perturbation and oxidative stress in hairless mice skin. Arch Dermatol Res 306, 467473.CrossRefGoogle ScholarPubMed
69.Wang, AN, Yi, XW, Yu, HF, et al. (2009) Free radical scavenging activity of Lactobacillus fermentum in vitro and its antioxidative effect on growing-finishing pigs. J Appl Microbiol 107, 11401148.CrossRefGoogle ScholarPubMed
70.Kaushal, D & Kansal, VK (2014) Dahi containing Lactobacillus acidophilus and Bifidobacterium bifidum improves phagocytic potential of macrophages in aged mice. J Food Sci Technol 51, 11471153.CrossRefGoogle ScholarPubMed
71.Kumar, RS, Kanmani, P, Yuvaraj, N, et al. (2012) Lactobacillus plantarum AS1 isolated from south Indian fermented food Kallappam suppress 1,2-dimethyl hydrazine (DMH)-induced colorectal cancer in male Wistar rats. Appl Biochem Biotechnol 166, 620631.CrossRefGoogle ScholarPubMed
72.Sengul, N, Isik, S, Aslim, B, et al. (2011) The effect of exopolysaccharide-producing probiotic strains on gut oxidative damage in experimental colitis. Dig Dis Sci 56, 707714.CrossRefGoogle ScholarPubMed
73.Ejtahed, HS, Mohtadi-Nia, J, Homayouni-Rad, A, et al. (2012) Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition 28, 539543.CrossRefGoogle ScholarPubMed
74.Tilg, H & Moschen, AR (2014) Microbiota and diabetes: an evolving relationship. Gut 63, 15131521.CrossRefGoogle ScholarPubMed
75.Le Chatelier, E, Nielsen, T, Qin, J, et al. (2013) Richness of human gut microbiome correlates with metabolic markers. Nature 500, 541546.CrossRefGoogle ScholarPubMed
76.Cani, PD, Osto, M, Geurts, L, et al. (2012) Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity. Gut Microbes 3, 279288.CrossRefGoogle ScholarPubMed
77.Everard, A, Belzer, C, Geurts, L, et al. (2013) Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci U S A 110, 90669071.CrossRefGoogle ScholarPubMed
78.Druart, C, Bindels, LB, Schmaltz, R, et al. (2015) Ability of the gut microbiota to produce PUFA-derived bacterial metabolites: proof of concept in germ-free versus conventionalized mice. Mol Nutr Food Res 59, 16031613.CrossRefGoogle ScholarPubMed
79.Kang, JH, Yun, SI & Park, HO (2010) Effects of Lactobacillus gasseri BNR17 on body weight and adipose tissue mass in diet-induced overweight rats. J Microbiol 48, 712714.CrossRefGoogle ScholarPubMed
80.Karlsson, CL, Molin, G, Fak, F, et al. (2011) Effects on weight gain and gut microbiota in rats given bacterial supplements and a high-energy-dense diet from fetal life through to 6 months of age. Br J Nutr 106, 887895.CrossRefGoogle Scholar
81.Esposito, E, Iacono, A, Bianco, G, et al. (2009) Probiotics reduce the inflammatory response induced by a high-fat diet in the liver of young rats. J Nutr 139, 905911.CrossRefGoogle ScholarPubMed
82.Chen, J, Wang, R, Li, XF, et al. (2012) Bifidobacterium adolescentis supplementation ameliorates visceral fat accumulation and insulin sensitivity in an experimental model of the metabolic syndrome. Br J Nutr 107, 14291434.CrossRefGoogle Scholar
83.Wang, J, Tang, H, Zhang, C, et al. (2015) Modulation of gut microbiota during probiotic-mediated attenuation of metabolic syndrome in high fat diet-fed mice. ISME J 9, 115.CrossRefGoogle ScholarPubMed
84.Kadooka, Y, Sato, M, Ogawa, A, et al. (2013) Effect of Lactobacillus gasseri SBT2055 in fermented milk on abdominal adiposity in adults in a randomised controlled trial. Br J Nutr 110, 16961703.CrossRefGoogle Scholar
85.Woodard, GA, Encarnacion, B, Downey, JR, et al. (2009) Probiotics improve outcomes after Roux-en-Y gastric bypass surgery: a prospective randomized trial. J Gastrointest Surg 13, 11981204.CrossRefGoogle ScholarPubMed
86.Druart, C, Alligier, M, Salazar, N, et al. (2014) Modulation of the gut microbiota by nutrients with prebiotic and probiotic properties. Adv Nutr 5, 624S633S.CrossRefGoogle ScholarPubMed
87.Hulston, CJ, Churnside, AA & Venables, MC (2015) Probiotic supplementation prevents high-fat, overfeeding-induced insulin resistance in human subjects. Br J Nutr 113, 596602.CrossRefGoogle ScholarPubMed
88.Weng, H, Endo, K, Li, J, et al. (2015) Induction of peroxisomes by butyrate-producing probiotics. PLOS ONE 10, e0117851.CrossRefGoogle ScholarPubMed
89.Coakley, M, Ross, RP, Nordgren, M, et al. (2003) Conjugated linoleic acid biosynthesis by human-derived Bifidobacterium species. J Appl Microbiol 94, 138145.CrossRefGoogle ScholarPubMed
90.Everard, A, Lazarevic, V, Derrien, M, et al. (2011) Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes 60, 27752786.CrossRefGoogle ScholarPubMed
91.Everard, A, Lazarevic, V, Gaia, N, et al. (2014) Microbiome of prebiotic-treated mice reveals novel targets involved in host response during obesity. ISME J 8, 21162130.CrossRefGoogle ScholarPubMed
92.Geurts, L, Neyrinck, AM, Delzenne, NM, et al. (2014) Gut microbiota controls adipose tissue expansion, gut barrier and glucose metabolism: novel insights into molecular targets and interventions using prebiotics. Benef Microbes 5, 317.CrossRefGoogle ScholarPubMed
93.Kellow, NJ, Coughlan, MT & Reid, CM (2014) Metabolic benefits of dietary prebiotics in human subjects: a systematic review of randomised controlled trials. Br J Nutr 111, 11471161.CrossRefGoogle ScholarPubMed
94.Dewulf, EM, Cani, PD, Claus, SP, et al. (2013) Insight into the prebiotic concept: lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut 62, 11121121.CrossRefGoogle ScholarPubMed
95.Salazar, N, Dewulf, EM, Neyrinck, AM, et al. (2015) Inulin-type fructans modulate intestinal Bifidobacterium species populations and decrease fecal short-chain fatty acids in obese women. Clin Nutr 34, 501507.CrossRefGoogle ScholarPubMed
96.Druart, C, Dewulf, EM, Cani, PD, et al. (2014) Gut microbial metabolites of polyunsaturated fatty acids correlate with specific fecal bacteria and serum markers of metabolic syndrome in obese women. Lipids 49, 397402.CrossRefGoogle ScholarPubMed
97.Larsen, N, Vogensen, FK, van den Berg, FW, et al. (2010) Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE 5, e9085.CrossRefGoogle ScholarPubMed
98.Cani, PD, Possemiers, S, Van de Wiele, T, et al. (2009) Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 58, 10911103.CrossRefGoogle ScholarPubMed
99.El Aidy, SE, Dinan, TG & Cryan, JF (2015) Gut microbiota: the conductor in the orchestra of immune-neuroendocrine communication. Clin Ther 37, 954967.CrossRefGoogle ScholarPubMed
100.Stilling, RM, Bordenstein, SR, Dinan, TG, et al. (2014) Friends with social benefits: host-microbe interactions as a driver of brain evolution and development? Front Cell Infect Microbiol 4, 147.CrossRefGoogle ScholarPubMed
101.Cryan, JF & Dinan, TG (2012) Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci 13, 701712.CrossRefGoogle ScholarPubMed
102.O’Mahony, SM, Marchesi, JR, Scully, P, et al. (2009) Early life stress alters behavior, immunity, and microbiota in rats: implications for irritable bowel syndrome and psychiatric illnesses. Biol Psychiatry 65, 263267.CrossRefGoogle ScholarPubMed
103.O’Malley, D, Julio-Pieper, M, Gibney, SM, et al. (2010) Distinct alterations in colonic morphology and physiology in two rat models of enhanced stress-induced anxiety and depression-like behaviour. Stress 13, 114122.CrossRefGoogle ScholarPubMed
104.Borre, YE, O’Keeffe, GW, Clarke, G, et al. (2014) Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med 20, 509518.CrossRefGoogle ScholarPubMed
105.Wall, R, Cryan, JF, Ross, RP, et al. (2014) Bacterial neuroactive compounds produced by psychobiotics. Adv Exp Med Biol 817, 221239.CrossRefGoogle ScholarPubMed
106.Sudo, N, Chida, Y, Aiba, Y, et al. (2004) Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol 558, 263275.CrossRefGoogle ScholarPubMed
107.Diaz Heijtz, R, Wang, S, Anuar, F, et al. (2011) Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A 108, 30473052.CrossRefGoogle ScholarPubMed
108.Neufeld, KM, Kang, N, Bienenstock, J, et al. (2011) Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol Motil 23, 255264.CrossRefGoogle ScholarPubMed
109.Clarke, G, Grenham, S, Scully, P, et al. (2013) The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry 18, 666673.CrossRefGoogle Scholar
110.Ogbonnaya, ES, Clarke, G, Shanahan, F, et al. (2015) Adult hippocampal neurogenesis is regulated by the microbiome. Biol Psychiatry 78, e7e9.CrossRefGoogle ScholarPubMed
111.Braniste, V, Al-Asmakh, M, Kowal, C, et al. (2014) The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med 6, 263ra158.CrossRefGoogle ScholarPubMed
112.Stilling, RM, Dinan, TG & Cryan, JF (2014) Microbial genes, brain & behaviour – epigenetic regulation of the gut-brain axis. Genes Brain Behav 13, 6986.CrossRefGoogle ScholarPubMed
113.Bravo, JA, Forsythe, P, Chew, MV, et al. (2011) Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A 108, 1605016055.CrossRefGoogle Scholar
114.Savignac, HM, Tramullas, M, Kiely, B, et al. (2015) Bifidobacteria modulate cognitive processes in an anxious mouse strain. Behav Brain Res 287, 5972.CrossRefGoogle Scholar
115.Desbonnet, L, Clarke, G, Shanahan, F, et al. (2014) Microbiota is essential for social development in the mouse. Mol Psychiatry 19, 146148.CrossRefGoogle ScholarPubMed
116.de Theije, CG, Wopereis, H, Ramadan, M, et al. (2014) Altered gut microbiota and activity in a murine model of autism spectrum disorders. Brain Behav Immun 37, 197206.CrossRefGoogle Scholar
117.Hsiao, EY, McBride, SW, Hsien, S, et al. (2013) Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 155, 14511463.CrossRefGoogle ScholarPubMed
118.Gilbert, JA, Krajmalnik-Brown, R, Porazinska, DL, et al. (2013) Toward effective probiotics for autism and other neurodevelopmental disorders. Cell 155, 14461448.CrossRefGoogle ScholarPubMed
119.Collins, SM, Kassam, Z & Bercik, P (2013) The adoptive transfer of behavioral phenotype via the intestinal microbiota: experimental evidence and clinical implications. Curr Opin Microbiol 16, 240245.CrossRefGoogle ScholarPubMed
120.Bruce-Keller, AJ, Salbaum, JM, Luo, M, et al. (2015) Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity. Biol Psychiatry 77, 607615.CrossRefGoogle ScholarPubMed
121.O’Mahony, SM, Felice, VD, Nally, K, et al. (2014) Disturbance of the gut microbiota in early-life selectively affects visceral pain in adulthood without impacting cognitive or anxiety-related behaviors in male rats. Neuroscience 277, 885901.CrossRefGoogle ScholarPubMed
122.Jeffery, IB, O’Toole, PW, Ohman, L, et al. (2012) An irritable bowel syndrome subtype defined by species-specific alterations in faecal microbiota. Gut 61, 9971006.CrossRefGoogle ScholarPubMed
123.McKernan, DP, Fitzgerald, P, Dinan, TG, et al. (2010) The probiotic Bifidobacterium infantis 35624 displays visceral antinociceptive effects in the rat. Neurogastroenterol Motil 22, 10291035.CrossRefGoogle ScholarPubMed
124.Clarke, G, Cryan, JF, Dinan, TG, et al. (2012) Review article: probiotics for the treatment of irritable bowel syndrome-focus on lactic acid bacteria. Aliment Pharmacol Ther 35, 403413.CrossRefGoogle ScholarPubMed
125.Kennedy, PJ, Clarke, G, O’Neill, A, et al. (2014) Cognitive performance in irritable bowel syndrome: evidence of a stress-related impairment in visuospatial memory. Psychol Med 44, 15531566.CrossRefGoogle ScholarPubMed
126.Dinan, TG, Stanton, C & Cryan, JF (2013) Psychobiotics: a novel class of psychotropic. Biol Psychiatry 74, 720726.CrossRefGoogle ScholarPubMed
127.Phillips, JGP (1910) The treatment of melancholia by the lactic acid bacillus. J Mental Sci 56, 422431.CrossRefGoogle Scholar
128.Benton, D, Williams, C & Brown, A (2007) Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur J Clin Nutr 61, 355361.CrossRefGoogle ScholarPubMed
129.Rao, AV, Bested, AC, Beaulne, TM, et al. (2009) A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathog 1, 6.CrossRefGoogle ScholarPubMed
130.Steenbergen, L, Sellaro, R, van Hemert, S, et al. (2015) A randomized controlled trial to test the effect of multispecies probiotics on cognitive reactivity to sad mood. Brain Behav Immun 48, 258264.CrossRefGoogle ScholarPubMed
131.Mohammadi, AA, Jazayeri, S, Khosravi-Darani, K, et al. (2015) The effects of probiotics on mental health and hypothalamic-pituitary-adrenal axis: a randomized, double-blind, placebo-controlled trial in petrochemical workers. Nutr Neurosci (Epublication ahead of print version 16 April 2015).Google Scholar
132.Center, MM, Jemal, A, Lortet-Tieulent, J, et al. (2012) International variation in prostate cancer incidence and mortality rates. Eur Urol 61, 10791092.CrossRefGoogle ScholarPubMed
133.Kolonel, LN, Altshuler, D & Henderson, BE (2004) The multiethnic cohort study: exploring genes, lifestyle and cancer risk. Nat Rev Cancer 4, 519527.CrossRefGoogle ScholarPubMed
134.Cuzick, J, Thorat, MA, Andriole, G, et al. (2014) Prevention and early detection of prostate cancer. Lancet Oncol 15, e484e492.CrossRefGoogle ScholarPubMed
135.Lacy, JM & Kyprianou, N (2014) A tale of two trials: the impact of 5alpha-reductase inhibition on prostate cancer (review). Oncol Lett 8, 13911396.CrossRefGoogle ScholarPubMed
136.Hebert, JR, Hurley, TG, Olendzki, BC, et al. (1998) Nutritional and socioeconomic factors in relation to prostate cancer mortality: a cross-national study. J Natl Cancer Inst 90, 16371647.CrossRefGoogle ScholarPubMed
137.Yan, L & Spitznagel, EL (2009) Soy consumption and prostate cancer risk in men: a revisit of a meta-analysis. Am J Clin Nutr 89, 11551163.CrossRefGoogle Scholar
138.Mahmoud, AM, Yang, W & Bosland, MC (2014) Soy isoflavones and prostate cancer: a review of molecular mechanisms. J Steroid Biochem Mol Biol 140, 116132.CrossRefGoogle ScholarPubMed
139.Lund, TD, Munson, DJ, Haldy, ME, et al. (2004) Equol is a novel anti-androgen that inhibits prostate growth and hormone feedback. Biol Reprod 70, 11881195.CrossRefGoogle ScholarPubMed
140.Rafii, F (2015) The role of colonic bacteria in the metabolism of the natural isoflavone daidzein to equol. Metabolites 5, 5673.CrossRefGoogle Scholar
141.Tanaka, M, Fujimoto, K, Chihara, Y, et al. (2009) Isoflavone supplements stimulated the production of serum equol and decreased the serum dihydrotestosterone levels in healthy male volunteers. Prostate Cancer Prostatic Dis 12, 247252.CrossRefGoogle ScholarPubMed
142.Sugiyama, Y, Masumori, N, Fukuta, F, et al. (2013) Influence of isoflavone intake and equol-producing intestinal flora on prostate cancer risk. Asian Pac J Cancer Prev 14, 14.CrossRefGoogle ScholarPubMed
143.Fujimoto, K, Tanaka, M, Hirao, Y, et al. (2008) Age-stratified serum levels of isoflavones and proportion of equol producers in Japanese and Korean healthy men. Prostate Cancer Prostatic Dis 11, 252257.CrossRefGoogle ScholarPubMed
144.Miyanaga, N, Akaza, H, Hinotsu, S, et al. (2012) Prostate cancer chemoprevention study: an investigative randomized control study using purified isoflavones in men with rising prostate-specific antigen. Cancer Sci 103, 125130.CrossRefGoogle ScholarPubMed
145.Tsuji, H, Moriyama, K, Nomoto, K, et al. (2012) Identification of an enzyme system for daidzein-to-equol conversion in Slackia sp. strain NATTS. Appl Environ Microbiol 78, 12281236.CrossRefGoogle ScholarPubMed
146.Akaza, H (2012) Prostate cancer chemoprevention by soy isoflavones: role of intestinal bacteria as the ‘second human genome’. Cancer Sci 103, 969975.CrossRefGoogle ScholarPubMed
147.Wong, S, Derry, F, Jamous, A, et al. (2014) Is undernutrition risk associated with an adverse clinical outcome in spinal cord-injured patients admitted to a spinal centre? Eur J Clin Nutr 68, 125130.CrossRefGoogle ScholarPubMed
148.Issa, I & Moucari, R (2014) Probiotics for antibiotic-associated diarrhea: do we have a verdict? World J Gastroenterol 20, 1778817795.CrossRefGoogle ScholarPubMed
149.Wong, S, Jamous, A, O’Driscoll, J, et al. (2014) A Lactobacillus casei Shirota probiotic drink reduces antibiotic-associated diarrhoea in patients with spinal cord injuries: a randomised controlled trial. Br J Nutr 111, 672678.CrossRefGoogle ScholarPubMed
150.Brenchley, JM, Price, DA, Schacker, TW, et al. (2006) Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med 12, 13651371.CrossRefGoogle ScholarPubMed
151.Cunningham-Rundles, S, Ahrne, S, Johann-Liang, R, et al. (2011) Effect of probiotic bacteria on microbial host defense, growth, and immune function in human immunodeficiency virus type-1 infection. Nutrients 3, 10421070.CrossRefGoogle ScholarPubMed
152.Dong, H, Rowland, I, Thomas, LV, et al. (2013) Immunomodulatory effects of a probiotic drink containing Lactobacillus casei Shirota in healthy older volunteers. Eur J Nutr 52, 18531863.CrossRefGoogle ScholarPubMed
153.Falasca, K, Vecchiet, J, Ucciferri, C, et al. (2015) Effect of probiotic supplement on cytokine levels in HIV-infected individuals: A preliminary study. Nutrients 7(1), 83358347.CrossRefGoogle Scholar
154.Miquel, S, Martin, R, Bridonneau, C, et al. (2014) Ecology and metabolism of the beneficial intestinal commensal bacterium Faecalibacterium prausnitzii. Gut Microbes 5, 146151.CrossRefGoogle ScholarPubMed
155.Miquel, S, Martin, R, Rossi, O, et al. (2013) Faecalibacterium prausnitzii and human intestinal health. Curr Opin Microbiol 16, 255261.CrossRefGoogle ScholarPubMed
156.Qin, J, Li, R, Raes, J, et al. (2010) A human gut microbial gene catalogue established by metagenomic sequencing. Nature 464, 5965.CrossRefGoogle ScholarPubMed
157.Sokol, H, Pigneur, B, Watterlot, L, et al. (2008) Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A 105, 1673116736.CrossRefGoogle ScholarPubMed
158.Cao, Y, Shen, J & Ran, ZH (2014) Association between Faecalibacterium prausnitzii reduction and inflammatory bowel disease: a meta-analysis and systematic review of the literature. Gastroenterol Res Pract 2014, 872725.CrossRefGoogle ScholarPubMed
159.Balamurugan, R, Rajendiran, E, George, S, et al. (2008) Real-time polymerase chain reaction quantification of specific butyrate-producing bacteria, Desulfovibrio and Enterococcus faecalis in the feces of patients with colorectal cancer. J Gastroenterol Hepatol 23, 12981303.CrossRefGoogle ScholarPubMed
160.Rajilic-Stojanovic, M, Biagi, E, Heilig, HG, et al. (2011) Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology 141, 17921801.CrossRefGoogle ScholarPubMed
161.Martin, R, Chain, F, Miquel, S, et al. (2014) The commensal bacterium Faecalibacterium prausnitzii is protective in DNBS-induced chronic moderate and severe colitis models. Inflamm Bowel Dis 20, 417430.CrossRefGoogle ScholarPubMed
162.Martin, R, Miquel, S, Chain, F, et al. (2015) Faecalibacterium prausnitzii prevents physiological damages in a chronic low-grade inflammation murine model. BMC Microbiol 15, 67.CrossRefGoogle Scholar
163.Miquel, S, Leclerc, M, Martin, R, et al. (2015) Identification of metabolic signatures linked to anti-inflammatory effects of Faecalibacterium prausnitzii. MBio 6, e00300e00315.CrossRefGoogle ScholarPubMed
164.Read, AF, Graham, AL & Raberg, L (2008) Animal defenses against infectious agents: is damage control more important than pathogen control. PLoS Biol 6, e4.CrossRefGoogle ScholarPubMed
165.Maloy, KJ & Powrie, F (2011) Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature 474, 298306.CrossRefGoogle ScholarPubMed
166.Round, JL & Mazmanian, SK (2009) The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol 9, 313323.CrossRefGoogle ScholarPubMed
167.Atarashi, K, Umesaki, Y & Honda, K (2011) Microbiotal influence on T cell subset development. Semin Immunol 23, 146153.CrossRefGoogle ScholarPubMed
168.Olszak, T, An, D, Zeissig, S, et al. (2012) Microbial exposure during early life has persistent effects on natural killer T cell function. Science 336, 489493.CrossRefGoogle ScholarPubMed
169.Noval Rivas, M, Burton, OT, Wise, P, et al. (2013) A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis. J Allergy Clin Immunol 131, 201212.CrossRefGoogle ScholarPubMed
170.Konieczna, P, Akdis, CA, Quigley, EM, et al. (2012) Portrait of an immunoregulatory Bifidobacterium. Gut Microbes 3, 261266.CrossRefGoogle ScholarPubMed
171.O’Mahony, C, Scully, P, O’Mahony, D, et al. (2008) Commensal-induced regulatory T cells mediate protection against pathogen-stimulated NF-kappaB activation. PLoS Pathog 4, e1000112.CrossRefGoogle ScholarPubMed
172.Groeger, D, O’Mahony, L, Murphy, EF, et al. (2013) Bifidobacterium infantis 35624 modulates host inflammatory processes beyond the gut. Gut Microbes 4, 325339.CrossRefGoogle ScholarPubMed
173.Konieczna, P, Groeger, D, Ziegler, M, et al. (2012) Bifidobacterium infantis 35624 administration induces Foxp3 T regulatory cells in human peripheral blood: potential role for myeloid and plasmacytoid dendritic cells. Gut 61, 354366.CrossRefGoogle ScholarPubMed
174.Lyons, A, O’Mahony, D, O’Brien, F, et al. (2010) Bacterial strain-specific induction of Foxp3+ T regulatory cells is protective in murine allergy models. Clin Exp Allergy 40, 811819.CrossRefGoogle ScholarPubMed
175.O’Mahony, L, Akdis, M & Akdis, CA (2011) Regulation of the immune response and inflammation by histamine and histamine receptors. J Allergy Clin Immunol 128, 11531162.CrossRefGoogle ScholarPubMed
176.Frei, R, Ferstl, R, Konieczna, P, et al. (2013) Histamine receptor 2 modifies dendritic cell responses to microbial ligands. J Allergy Clin Immunol 132, 194204.CrossRefGoogle ScholarPubMed
177.Ferstl, R, Frei, R, Schiavi, E, et al. (2014) Histamine receptor 2 is a key influence in immune responses to intestinal histamine-secreting microbes. J Allergy Clin Immunol 134, 744746.CrossRefGoogle ScholarPubMed
178.Roduit, C, Frei, R, Loss, G, et al. (2012) Development of atopic dermatitis according to age of onset and association with early-life exposures. J Allergy Clin Immunol 130, 130136.CrossRefGoogle ScholarPubMed
179.Young, KT, Davis, LM & Dirita, VJ (2007) Campylobacter jejuni: molecular biology and pathogenesis. Nat Rev Microbiol 5, 665679.CrossRefGoogle ScholarPubMed
180.Bereswill, S, Fischer, A, Plickert, R, et al. (2011) Novel murine infection models provide deep insights into the ‘menage a trois’ of Campylobacter jejuni, microbiota and host innate immunity. PLoS ONE 6, e20953.CrossRefGoogle Scholar
181.Heimesaat, MM, Plickert, R, Fischer, A, et al. (2013) Can microbiota transplantation abrogate murine colonization resistance against Campylobacter jejuni? Eur J Microbiol Immunol (Bp) 3, 3643.CrossRefGoogle ScholarPubMed
182.Haag, LM, Fischer, A, Otto, B, et al. (2012) Intestinal microbiota shifts towards elevated commensal Escherichia coli loads abrogate colonization resistance against Campylobacter jejuni in mice. PLOS ONE 7, e35988.CrossRefGoogle ScholarPubMed
183.Heimesaat, MM & Bereswill, S (2015) Murine infection models for the investigation of Campylobacter jejuni – host interactions and pathogenicity. Berl Munch Tierarztl Wochenschr 128, 98103.Google ScholarPubMed
184.Haag, LM, Fischer, A, Otto, B, et al. (2012) Campylobacter jejuni induces acute enterocolitis in gnotobiotic IL-10-/- mice via toll-like-receptor-2 and -4 signaling. PLOS ONE 7, e40761.CrossRefGoogle Scholar
185.Mortensen, NP, Kuijf, ML, Ang, CW, et al. (2009) Sialylation of Campylobacter jejuni lipo-oligosaccharides is associated with severe gastro-enteritis and reactive arthritis. Microbes Infect 11, 988994.CrossRefGoogle ScholarPubMed
186.Edwards, LA, Nistala, K, Mills, DC, et al. (2010) Delineation of the innate and adaptive T-cell immune outcome in the human host in response to Campylobacter jejuni infection. PLoS ONE 5, e15398.CrossRefGoogle ScholarPubMed
187.Kholy, KE, Genco, RJ & Van Dyke, TE (2015) Oral infections and cardiovascular disease. Trends Endocrinol Metab 26, 315321.CrossRefGoogle ScholarPubMed
188.Hajishengallis, G (2015) Periodontitis: from microbial immune subversion to systemic inflammation. Nat Rev Immunol 15, 3044.CrossRefGoogle ScholarPubMed
189.Olsen, I (2015) From the acta prize lecture 2014: the periodontal-systemic connection seen from a microbiological standpoint. Acta Odontol Scand 73, 563568.CrossRefGoogle ScholarPubMed
190.Niederman, R, Buyle-Bodin, Y, Lu, BY, et al. (1997) Short-chain carboxylic acid concentration in human gingival crevicular fluid. J Dent Res 76, 575579.CrossRefGoogle ScholarPubMed
191.Kurita-Ochiai, T, Fukushima, K & Ochiai, K (1995) Volatile fatty acids, metabolic by-products of periodontopathic bacteria, inhibit lymphocyte proliferation and cytokine production. J Dent Res 74, 13671373.CrossRefGoogle ScholarPubMed
192.Yoneda, S, Kawarai, T, Narisawa, N, et al. (2013) Effects of short-chain fatty acids on Actinomyces naeslundii biofilm formation. Mol Oral Microbiol 28, 354365.CrossRefGoogle ScholarPubMed
193.Arai, T, Ochiai, K & Senpuku, H (2015) Actinomyces naeslundii GroEL-dependent initial attachment and biofilm formation in a flow cell system. J Microbiol Methods 109, 160166.CrossRefGoogle Scholar
194.Kurita-Ochiai, T, Ochiai, K & Fukushima, K (2000) Butyric-acid-induced apoptosis in murine thymocytes and splenic T- and B-cells occurs in the absence of p53. J Dent Res 79, 19481954.CrossRefGoogle ScholarPubMed
195.Kurita-Ochiai, T, Amano, S, Fukushima, K, et al. (2003) Cellular events involved in butyric acid-induced T cell apoptosis. J Immunol 171, 35763584.CrossRefGoogle ScholarPubMed
196.Kurita-Ochiai, T, Seto, S & Ochiai, K (2004) Role of cell-cell communication in inhibiting butyric acid-induced T-cell apoptosis. Infect Immun 72, 59475954.CrossRefGoogle ScholarPubMed
197.Kurita-Ochiai, T, Seto, S, Suzuki, N, et al. (2008) Butyric acid induces apoptosis in inflamed fibroblasts. J Dent Res 87, 5155.CrossRefGoogle ScholarPubMed
198.Cueno, ME, Imai, K, Tamura, M, et al. (2014) Butyric acid-induced rat jugular blood cytosolic oxidative stress is associated with SIRT1 decrease. Cell Stress Chaperones 19, 295298.CrossRefGoogle ScholarPubMed
199.Cueno, ME, Tamura, M & Ochiai, K (2015) Middle-aged rats orally supplemented with gel-encapsulated catechin favorably increases blood cytosolic NADPH levels. Phytomedicine 22, 425430.CrossRefGoogle ScholarPubMed
200.Imai, K, Yamada, K, Tamura, M, et al. (2012) Reactivation of latent HIV-1 by a wide variety of butyric acid-producing bacteria. Cell Mol Life Sci 69, 25832592.CrossRefGoogle ScholarPubMed
201.Cueno, ME, Imai, K, Matsukawa, N, et al. (2013) Butyric acid retention in gingival tissue induces oxidative stress in jugular blood mitochondria. Cell Stress Chaperones 18, 661665.CrossRefGoogle ScholarPubMed
202.Cueno, ME, Tamura, M, Ohya, M, et al. (2014) Similar physiological effects in Porphyromonas gingivalis ATCC 33277 under hemin-excess and hemin-limited concentrations are putatively associated to different hydrogen peroxide function. Anaerobe 28, 178181.CrossRefGoogle ScholarPubMed
203.Cornelius, C, Koverech, G, Crupi, R, et al. (2014) Osteoporosis and Alzheimer pathology: role of cellular stress response and hormetic redox signaling in aging and bone remodeling. Front Pharmacol 5, 120.CrossRefGoogle ScholarPubMed
204.Cueno, ME, Kamio, N, Seki, K, et al. (2015) High butyric acid amounts induce oxidative stress, alter calcium homeostasis, and cause neurite retraction in nerve growth factor-treated PC12 cells. Cell Stress Chaperones 20, 709713.CrossRefGoogle ScholarPubMed
205.Shalini, S, Dorstyn, L, Dawar, S, et al. (2015) Old, new and emerging functions of caspases. Cell Death Differ 22, 526539.CrossRefGoogle ScholarPubMed
206.Maticic, M, Poljak, M, Kramar, B, et al. (2000) Proviral HIV-1 DNA in gingival crevicular fluid of HIV-1-infected patients in various stages of HIV disease. J Dent Res 79, 14961501.CrossRefGoogle ScholarPubMed
207.Imai, K, Ochiai, K & Okamoto, T (2009) Reactivation of latent HIV-1 infection by the periodontopathic bacterium Porphyromonas gingivalis involves histone modification. J Immunol 182, 36883695.CrossRefGoogle ScholarPubMed
208.Saygun, I, Nizam, N, Keskiner, I, et al. (2011) Salivary infectious agents and periodontal disease status. J Periodontal Res 46, 235239.CrossRefGoogle ScholarPubMed
209.Konstantinidis, A, Sakellari, D, Papa, A, et al. (2005) Real-time polymerase chain reaction quantification of Epstein-Barr virus in chronic periodontitis patients. J Periodontal Res 40, 294298.CrossRefGoogle ScholarPubMed
210.Imai, K, Inoue, H, Tamura, M, et al. (2012) The periodontal pathogen Porphyromonas gingivalis induces the Epstein-Barr virus lytic switch transactivator ZEBRA by histone modification. Biochimie 94, 839846.CrossRefGoogle ScholarPubMed
211.Segers, ME & Lebeer, S (2014) Towards a better understanding of Lactobacillus rhamnosus GG – host interactions. Microb Cell Fact 13, Suppl. 1, S7.CrossRefGoogle ScholarPubMed
212.Elazab, N, Mendy, A, Gasana, J, et al. (2013) Probiotic administration in early life, atopy, and asthma: a meta-analysis of clinical trials. Pediatrics 132, e666e676.CrossRefGoogle ScholarPubMed
213.World Health Organization, Food and Agricultural Organization of the United Nations (2006) Probiotics in Food. Health and Nutritional Properties and Guidelines for Evaluation. FAO Nutrition Paper, 85. Rome: FAO.Google Scholar
214.Hill, C, Guarner, F, Reid, G, et al. (2014) 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 11, 506514.CrossRefGoogle ScholarPubMed
215.Binnendijk, KH & Rijkers, GT (2013) What is a health benefit? An evaluation of EFSA opinions on health benefits with reference to probiotics. Benef Microbes 4, 223230.CrossRefGoogle ScholarPubMed
216.Petrof, EO, Gloor, GB, Vanner, SJ, et al. (2013) Stool substitute transplant therapy for the eradication of Clostridium difficile infection: ‘RePOOPulating’ the gut. Microbiome 1, 3.CrossRefGoogle ScholarPubMed
217.Ritchie, ML & Romanuk, TN (2012) A meta-analysis of probiotic efficacy for gastrointestinal diseases. PLOS ONE 7, e34938.CrossRefGoogle ScholarPubMed
218.Canadian Food Inspection Agency (2015) Probiotic claims. Acceptable non-strain specific claims. http://inspection.gc.ca/food/labelling/food-labelling-for-industry/health-claims/eng/1392834838383/1392834887794?chap=9#s21c9 (accessed July 2015).Google Scholar