Accepted Manuscript Title: Review article: Bifidobacteria- Insight into clinical outcomes and mechanisms of its probiotic action Author: Amrita Sarkar Santanu Mandal PII: DOI: Reference:
S0944-5013(16)30432-3 http://dx.doi.org/doi:10.1016/j.micres.2016.07.001 MICRES 25919
To appear in: Received date: Revised date: Accepted date:
23-2-2016 12-6-2016 7-7-2016
Please cite this article as: Sarkar Amrita, Mandal Santanu.Review article: Bifidobacteria- Insight into clinical outcomes and mechanisms of its probiotic action.Microbiological Research http://dx.doi.org/10.1016/j.micres.2016.07.001 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Review article: Bifidobacteria- Insight into clinical outcomes and mechanisms of its probiotic action Amrita Sarkar†,* Santanu Mandal† †
School of Chemistry and Manchester Institute of Biotechnology. The University of Manchester, 131 Princess
Street, Manchester, M1 7DN, UK. *
Corresponding author: Amrita Sarkar; Email:
[email protected]
Graphical Abstract
Abstract The invasion of pathogens causes a disruption of the gut homeostasis. Innate immune responses and those triggered by endogenous microbiota form the first line of defence in our body. Pathogens often successfully overcome the resistances offered, calling for therapeutic intervention. Conventional strategy involving
1
antibiotics might eradicate pathogens, but often leave the gut uncolonised and susceptible to recurrences. Probiotic supplements are useful alternatives. Bifidobacterium is one of widely studied probiotic genus, effective in restoring gut homeostasis. Mechanisms of probiotic action of bifidobacteria are several, often with strain-specificity. Analysis of streamlined literature reports reveal that although most studies report the probiotic aspect of bifidobacteria, sporadic documented contradictory results exist, challenging its therapeutic application and prompting studies to unambiguously establish the strain-associated probiotic activity and negate adverse effects prior to its clinical administration. Multi-strain/combinatorial therapy possibly relies on a combination of underlying operating mechanisms, each contributing towards enhanced probiotic efficacy, understanding which could help in developing customised formulations against targeted pathogens. Bifidogenic activity is also mediated by surface-associated structural components such as exopolysaccharides, lipoteichoic acids along with metabolites and bifidocins. This highlights scope for developing advanced structural therapeutic strategy which might be pivotal in replacing intact cell probiotics therapy. Keywords: gut microbiome; pathogen protection; clinical application; adverse effects; immunomodulatory effect; combinatorial therapy
1. Introduction Increasing population of antibiotic resistant virulent pathogens has triggered the search of alternate means of pathogen combat. Photopharmacotherapy and probiotics in combination with prebiotics has become routes of major diversion from conventional use of antibiotics (Velema et al. 2014; Wang et al. 2013). They not only serve as green channels in reducing built-up of active antimicrobial elements in the environment but also lower drug resistivity in deadly pathogens. Since the acceptance by United Nations FAO and WHO in 2001 (Castellazzi et al. 2013), probiotics have found extensive applications. Healthy gut bacteria in humans are acquired during birth, which confer health promoting effects to the host. This neonatal microbial population gradually alters with age serving as a biomarker for health. Reduction in intestinal probiotic community often poses serious risks of pathogenic invasions and colonisation of gut epithelium, intake of probiotic supplements can be particularly useful. Numerous bifidobacterial strains have found to be useful in treating different clinical conditions. Therefore it is likely that different strains operate through a combination of more than one pathway instead of a single mechanistic route. The present communication aims to provide the current perspective of the concepts evolving about the mechanisms of probiotic action of bifidobacteria in humans rather than a comprehensive
2
literature summary. Understanding mechanisms could be helpful in identification of suitable probiotic strains for prevention and customised therapeutic treatment of infections with known pathogenesis and prognosis. Interestingly these apparently benign and mostly probiotic microorganisms have been reported to have adverse effects on human health. Although infrequent, but these reports emphasise on the need for in-depth study prior to developing probiotic formulation for clinical administration.
2. Clinical prospect of bifidobacteria Bifidobacterial flora in neonatal gut is established within the first few days after delivery and constitutes upto almost 80% of the microfloral composition during infancy (Turroni et al. 2012). The gut flora established at birth and weaning, gradually alters with age, dietary, seasonal and geographical variations along with intake of probiotics along with other endogeneous and exogeneous factors (Balamurugan et al. 2008; Lagier et al. 2012; Davenport 2014; Mitsuoka 1990). Clinical study shows that usage of antibiotics on neonates not only causes a delay in developing a normal bifidobacterial intestinal flora, but also increases risk of colonisation by pathogenic bacteria such as Kleibsella, Enterobacteria, Citrobacter, Pseudomonas etc (Hussey et al. 2011). The delayed and disturbed bifidobacterial colonisation during infancy increases the risks of childhood diseases and could also influence the health of the host in future (Marra et al. 2006; Alm et al 2008; Bailey et al 2014). 2.1. Beneficial effects. Breakdown of probiotic barrier against pathogens often leads to gastrointestinal problems such as gastroenteritis, enterocolitis, irritable bowel syndrome (IBS), diarrhoea and food allergies/intolerances (Bermudez-Brito et al. 2012; Guglielmetti et al. 2011; Groschwitz and Hogan 2009). Bifidobacteria shows efficacy in alleviating related symptoms of constipation, abdominal pain, flatulence, bloating by rebalancing the gut flora and preventing abnormal bacterial fermentation of food residues (O’Mahony et al. 2005). Specific strains of B. infantis and B. lactis along with Streptococcus thermophiles, L. acidophilus and L. rhamnosus show efficacy in alleviating symptoms of acute gastroenteritis in infants (Erdogan et
al.
2012;
Vandenplas
and
Hert
2011).
Diarrhoea
often
follows
antibiotic
therapy
or
chemotherapy/radiotherapy in cancer patients (Fox et al. 2015; Demers 2014). Administration of B. lactis BB12, B94, B. longum BB-536 and specific strains of B. Bifidum in combination with L. rhamnosus GG, L. acidophilus La-5, LAC-361 is reported to reduce episodes and symptoms of diarrhoea in infants and adults (Demers et al. 2014; Fox et al. 2015; İşlek et al. 2014; Dinleyici et al. 2013; El-Soud et al. 2015). The increasing trend of IBS among people (~15-20%) raises a major concern about the prevention and treatment strategies. Clinical study shows that administration of B. bifidum MIMBb75 strain as nutritional supplement could
3
substantially relieve patients of IBS symptoms and promote a healthy life compared to those on placebo (Guglielmetti et al 2011). B. bifidum KCTC12199BP, B. lactis KCTC 11904BP, W51, B. longum KCTC12200BP are some of the other strains exhibiting efficacy in IBS therapy (Rossi et al. 2015; Yoon et al. 2014). While B. animalis DN-173010 is particularly found to be helpful in improving faecal colonic transit time in people with long transit times (Picard et al. 2005). In vitro studies using human intestinal HT-29 cell lines also revealed the anti-inflammatory effects of B. animalis subsp. lactis PBS075 (Presti et al. 2015). Similar studies using human colonic microbiota model reveal that B. longum subsp. infantis NCIMB 702205 strain has the ability to reduce the gut-derived lipopolysaccharide which is related to chronic inflammatory and metabolic diseases (Rodes et al. 2013). Specific strains of B. breve and B. lactis along with L. casei have been reported to improve intestinal motility along with lowering of nosocomial sepsis and mortality in neonates and infants diagnosed with necrotising enterocolitis (Braga et al. 2011; Dilli et al. 2015). Bifidobacteria is also reported to exhibit probiotic effects against Crohn’s disease, ulcerative colitis and pouchitis (Shen et al. 2014; Tamaki et al. 2015; Saez-Lara et al. 2015). Other health benefits include antagonistic effect on colorectal cancer and treatment of Helicobacter pylori infections (Khodadad et al. 2013; Chitapanarux et al. 2015; Chong 2014; Yoon et al. 2014; Lee et al. 2008; Bindels et al. 2012). Particularly interesting study recently showed B. bifidum PRL2010 assisting in cholesterol assimilation through upregulation of genes encoding putative transporters and reductases (Zanotti et al. 2015). While double blinded, randomised, placebo controlled trial involving Japanese obese adults showed Bifidobacterium animalis ssp. lactis GCL2505 assisted reduction of visceral fat (Takahashi et al. 2016). Bifidobacteria not only improves lactose utilisation but also helps in stabilisation of gut mucosal barrier (Kailasapathy and Chin 2000). Atopic dermatitis/eczema is a chronic and pruritic skin disorder affecting infants and adults of all age group (Eichenfield et al. 2014; Wollenberg e al. 2016). Although its cause is not clearly understood, may arise due to immunological disorder related to IgE mediated reactions. The first line of treatment includes hydrational and anti-inflammatory topical medications (Wollenberg et al. 2016; Ring et al. 2012) with continuous search for alternative nonpharmacologic therapies. The first role of probiotic bifidobacteria in prevention and therapeutic intervention in such atopic diseases is well established, with B. breve LMG 23729, B. longum BB536, B. lactis Bb-12 being some of the strains with reported efficacy (Enomoto et al. 2014; Meneghin et al. 2012), while B. lactis NCC 2818 is reported to alleviate symptoms of seasonal allergic rhinitis (Singh et al. 2013). Such allergies are possibly related to limited Th1/Th2 levels and delayed development of immune tolerances due to lower exposure to extrinsic allergens such as pollens during infancy. The probiotic microorganism mitigates
4
immune parameters and lowers Th2 cytokine levels (Singh et al. 2013). Similarly double blinded, placebo controlled and randomised clinical trial with B. bifidum G9-1 and B. longum MM-2 in combination with Lactobacillus gasseri KS-13 is reported to improve quality of life in individuals suffering from seasonal allergies (Dennis et al. 2016). Urogenital infection (including bacterial vaginosis, urinary tract infections, fungal and yeast vaginitis) affects women globally. Development of antibiotic resistance and limited usage during pregnancy are some of the drawbacks of antibiotic administration. In vivo experiments involving intravaginal Staphylococcosis in mice triggered by Staphylococcus aureus reported the anti-staphylococcal activity of specific strains of B. animalis VKL and VKB along with Lactobacillus strains (Babenko et al. 2012). The probiotic strains used not only assists in the elimination of S. aureus but also in the establishment of a normal and stable vaginal microflora. Further research should be targeted towards developing such probiotic formulations for clinical applications. Reduction of risks of preterm delivery, obesity and stress management, treatment of oral plaque and improvement of symptoms related to depressive disorders and Schizophrenia (Akkasheh et al. 2015; Minami et al. 2015; Nishihira et al. 2014; Toiviainen et al. 2015; Tomasik et al. 2015; Myhre et al. 2011) are some of the numerous beneficial effects offered by bifidobacteria. Detrimental effects on immune activation, GI tract with inflammation caused by HIV infection are not completely restituted by combined antiretroviral therapy (cART). Recent study documents promising results in improving HIV induced damages by supplementing cART therapy with probiotic B. breve, B. infantis and B. longum strains along with L. acidophilus, L. plantarum, L. casei, L. delbrueckii ssp. bulgaricus and Streptococcus faecium (d’Ettorre et al. 2015). While certain species of bifidobacteria and lactobacilli have demonstrated oxalate degrading abilities thus opening scope for their application in treatment of recurrent calcium oxalate-based kidney stones (Kullin et al. 2016; Kirkali et. al 2015). Although strain specific effects are observed, yet these clinical results indicate boundless prospect of genus Bifidobacterium towards amelioration of human health. 2.2. Adverse effects. Though studies investigating anaerobic microbes in clinical isolates are rare, yet sporadic cases of adverse effects related to bifidobacteria have been reported. Bush et al. (2014) suggested that these apparently probiotic and benign organisms can also have adverse effect on human health. Their case study reports the sepsis of prosthetic joint post total hip arthroplasty. Clinical examination of fluid from joint revealed the presence of Bifidobacterium, possibly from intake of probiotic formulation (containing strains of B. bifidum, B. breve and B. longum along with Lactobacillus and Streptococcus) ingested by the patient. While, B. dentium,
5
B. denticolens, B. inopinatum forms a class of bifidobacteria commonly associated with dental caries (Leahy et al. 2005; Mahlen and Clarridge 2009). For the first time occurrence of sepsis by B. longum in adult has been documented, while bacteremia in preterm infants has also been reported (Ha et al. 1999; Bourne et al. 1978; Weber et al. 2015). Certain strains of Bifidobacterium longum subsp. infantis, originating from administered probiotics have been identified by Bertelli et al. (2015) in a separate study as causative agents in multiple cases of bacteremia in preterm infants. Besides bacteremia, meningitis, endocarditis, peritonitis, intra-abdominal, enterocolitis, gynecologic and pulmonary infections involving Bifidobacterium spp. as causative agent has also been reported (Hata et al. 1988; Bourne et al. 1978; Weber et al. 2015; Wilson et al. 1972). Studies also attribute the occurrence of bacterial vaginosis to the increase in population of bifidobacteria along with other anaerobic microorganisms such as Bacterioides spp. (Schwiertz et al. 2015). Bifidobacteria mediated infections also include abdominal abscesses, obstetric and gynecologic and wounds (Brook and Frazier 1993). While specific B. breve strain has been identified as a causative agent of septicaemia in neonates, administered with probiotic formulations post operative procedures (Ohishi et al. 2010). Although such clinical incidences are often attributed to immune compromise in patients and prematurely born neonates rather than bifidobacteria itself (Margolles et al. 2011; Ohishi et al. 2010), it is nevertheless important to unambiguously establish the probiotic property of a particular strain prior to its clinical use. Report suggest that B. animalis subsp. animalis ATCC 25527T and B. infantis ATCC 15697T strains are capable of inducing duodenal and colonic inflammation in germ free IL10 deficient mice (Moran et al 2009). The authors suggest possible pathogenicity of these bifidobacteria strains in a susceptible host. Hence it is recommended that new probiotic strains for consumption should be identified from the commensal flora of humans devoid of any intrinsic antibiotic resistance, so that any rare probiotic infection could be eliminated (Borriello et al 2003). Although the number of bifidobacteria mediated infections has not raised much compared to its general consumption and clinical usage, a thorough understanding of their risks and benefits is unavoidable. Mechanisms of translocation of probiotic species from gastrointestinal to extraintestinal sites, their bloodstream survival, mucin degradation activity and possible infectivity should be considered carefully prior to its administration, especially to patients with challenged immune systems (Borriello et al 2003; Boyle et al 2006).
3. Modes of probiotic action Bifidogenic effects are largely known, yet the underlying mechanism is poorly understood. Bifidobacteria exerts probiotic effect against conditions ranging from IBS, ulcerative colitis, allergic diseases, Immunoglobulin E
6
(IgE) associated diseases, atopic dermatitis (AD) to oral plaque. Thus it is quite unlikely that all probiotic bifidobacteria strains operate using same single mechanism. Of the numerous literature reports available, Table 1 selectively lists some, highlighting the possible underlying mechanisms of bifidogenic activity with a schematic overview in Fig. 1. Each of these actions involves detailed mechanistic pathways which have been discussed here. 3.1. Adhesion to the gut epithelium followed by colonisation. Adhesion to the intestinal mucosa is the primordial need for bacteria for prolonged persistence to exert their health promoting effects. This may be perceived though several mechanisms exploiting host cell surface glycan and lectin receptor/motifs to bacterial capsular polysaccharides (Esko and Sharon 2009; Fanning et al. 2012; Hidalgo-Cantabrana et al. 2015). Studies indicate strain specific interplay of lectins, lipoproteins, adhesion molecules, pili/appendages or exopolysaccharides assisted multifactorial binding to the gut mucosa (Gleinser et al. 2012; Guglielmetti et al. 2008; Kavanaugh et al. 2013; Roger et al. 2010). Successful sequencing of a large number of bifidobacterial genomes has enabled identification and prediction of genes encoding adhesion factors. Probiotic-mediated modulation of multi-faceted adhesion factors involving transcription, chaperone proteins and glycoside hydrolases have been attributed to the adherence of B. longum subsp. infantis ATCC 15697 to HT-29 and Caco2 intestinal cells (Kavanaugh et al. 2013).
In vitro studies show that cell wall surface lipoprotein, BopA,
functions as adhesion promoter in B. bifidum MIMBb75 to Caco-2 cells with its adhesion directly correlated to the increase in the lipoprotein expression (Gleinser et al. 2012; Guglielmetti et al. 2008). Later, studies readdressed the role of BopA mediator. Antiserum against BopA demonstrated the adhesion of B. bifidum to human colonic mucin to be BopA independent (Kainulainen et al. 2013). Such contradictory results suggest the need for sustained efforts to establish mechanisms involving protein mediated adhesions. Glycolytic enzymes are known to mediate cross-talk between human plasminogen and B. bifidum S16, B. breve BBSF, B. longum S123 and B. lactis BI07. Study show that the enolase contains pattern recognition receptors for the human plasminogen with high binding affinity (Candela et al. 2009). Electron microscopy and mucin binding assays attribute the adhesion of certain B. bifidum strains to human intestinal cell line HT29 to the surface associated Transaldolase (González-Rodríguez et al. 2012). It is suggested that such enzymes could also promote colonisation of GI epithelial tissues post adhesion. Recent study predicts the possibility of fimbrial attachment of B. longum subsp. longum to the GI epithelium. Putative fimbrial His-BL0675 A type protein (among the variants B, C, D and E) showed strong binding affinity to the surface oligosaccharides on porcine colonic mucine (Suzuki et al. 2016). The identified
7
gene cluster in the bacterial genome are designated as major (FimA or FimP) and minor (FimB and/or FimQ) pilin subunit encoding genes (Foroni et al. 2011). Other studies involve identification of gene clusters responsible for pili biosynthesis in specific B. breve and B. bifidum strains (Motherway et al. 2011; Turroni et al. 2013). Type IVb tight adherence (Tad) pilus-encoding gene cluster tad2003 is identified for efficient gut colonisation of B. breve UCC2003 in murines. Transmission electron microscopy and atomic force microscopy also confirms the presence of pili structures on bacterial cells (Foroni et al. 2011; González-Rodríguez et al. 2012). Other than indigenous factors, exogenous contributions from prebiotic and human milk oligosaccharides (HMO) play key role in bacterial adhesion and colonisation (Bouhnik et al. 2004; Chichlowski et al. 2012; Hidalgo-Cantabrana et al. 2014; Kavanaugh et al. 2013; Salazar et al. 2015; Wickramasinghe et al. 2015). They not only serve as substrates for bifidobacterial metabolism, but have been reported to increase the adhesion and efficacy of Bifidobacterium spp (Kailasapathy and Chin 2000; Osman et al. 2006). This is possibly due to exogenous protective effects of the prebiotics, enhancing the survival rate of the probiotic cells within the gastrointestinal tract and stimulating growth and activity of the ingested probiotics along with other species residing within the gut microbiota (Kailasapathy and Chin 2000). Another interesting mechanism involves exopolysaccharide mediated bifidobacterial adhesion onto human intestinal mucosal surface (Inturri et al. 2014; López et al. 2012; Ranadheera et al. 2014; Salazar et al. 2014). Recent studies have identified gene cluster responsible for EPS production from genomes of most Bifidobacterium strains (Hidalgo-Cantabrana et al. 2014). Comparative study between B. breve A28 and B. bifidum A10, shows that the former strongly adheres to the intestinal cell lines due to higher production of EPS than the latter (Alp et al. 2010). 3.2. Production of metabolites and lowering of pH. Metabolism of dietary constituents (such as fructans and undigested carbohydrates) by intestinal probiotic community can generate molecules of physiological importance (Puertollano et al. 2014; Yasmin et al. 2015). Enzymes such as α, β-glucosidase, galactosidases and fructofuranosidases involving breakdown of the dietary carbohydrate residues has been reported to be expressed by probiotic strains in the intestine. Although the activity of the enzymes are dependent upon the carbon source available in the growth medium, yet B. lactis BB-12 is reported to show glucosidase and galactosidase activities irrespective of the media used. However reported diversity exists between different species of Bifidobacterium genus in terms of their inulin metabolic activity (Vuyst and Leroy 2011). The metabolites produced via heterofermentative pathways such as short chain fatty acids (SCFAs), polyunsaturated fatty acid (PUFA) derivatives,
8
hydrogen peroxides, diacetyls and carbon dioxides are known to exert inhibitory effects on the growth of pathogens (Kailasapathy and Chin 2000; Matsuki et al. 2013; Sugahara et al. 2015; Vuyst and Leroy 2011). SCFA includes lactates, formats, acetates, propionates, butyrates and pimelates (Asarat et al. 2015; Sugahara et al. 2015; Tabasco et al. 2014). These and other organic acids play a key role in reducing intestinal pH, preventing the growth and colonisation by acid sensitive and putrefactive pathogens (Fukuda et al. 2011). Murine based in vivo experiments demonstrated the protective role of bifidobacterial acetate against enterohaemorrhagic Escherichia coli O157:H7, while lactate and acetates from B. breve DN-156 007 participate in regulating epithelial proliferation in the gut, contributing towards gut homeostasis (Fukuda et al. 2011; Matsuki et al. 2013). Antimutagenic effects of probiotics such as bifidobacteria have increased the use of such strains for reduction in levels of toxic pro-carcinogenic enzymes and colorectal cancers. The metabolites such as propionates and butyrates are known to exert antagonistic effect especially on colon carcinoma cell proliferation inducing apoptosis of carcinoma cells and transformed cell lines in humans (Bindels et al. 2012; Gioia et al. 2014; Lee et al. 2008). Other biosynthetic pathways of bifidobacteria include synthesis of Vitamin B12 and folates, conferring protection against cancer (Gioia et al. 2014; Rossi et al. 2011). The PUFAs are particularly interesting due to the anti-carcinogenic, anti-inflammatory, anti-atherogenic and anti-diabetic properties (Gioia et al. 2014). Thus fermentation of dietary carbohydrate components by probiotic species confers a range of health beneficial effects. 3.3. Release of bacteriocins. One of the popularly suggested mechanisms of probiotic effect of bifidobacteria is through the release of compounds (molecular masses ranging from ~ 3-130 kDa) known as bacteriocins (Martinez et al. 2013; Poltavska and Kovalenko 2012). These ribosomally synthesised antimicrobial compounds were first reported in 1980 (Martinez et al. 2013). They exhibit strain specific activity against intra-genus (narrow spectrum) or inter-genus (wide range) bacterial strains. Since their identification and characterisation, bacteriocins have been classified into four categories based on their molecular masses, thermal stability and amino acid composition (Klaenhammer 1993). These compounds generally exhibit broad range activity against some species of the genera Bacillus, Enterococcus, E. coli, Salmonella, Clostridium, Shigella, Staphylococcus and Streptococcus (Collado et al. 2005; Martinez et al. 2013). The bacteriocins commonly promote antimicrobial activity through pore formation/permeabilisation leading to cell lysis and preventing cell wall biosynthesis (Bajaj et al. 2015; Liu et al. 2015; Martinez et al. 2013). Their proteinaceous nature makes them resistant to the actions of enzymes such as amylase and lipase but susceptible towards proteinases (Collado et al. 2005). The maximum inhibitory effects of these bacteriocins have been found to be between 8-16 hours of cell
9
culture (Fliss et al. 2010; Poltavska and Kovalenko 2012). This explains the maximum antimicrobial activity of specific bifidobacteria strains in their late logarithmic phase of growth. Bifidocin B from B. bifidum NCFB 1454 exhibits pH dependent absorption onto cell surface receptors of gram positive bacteria, leading to cell death (Klaenhammer 1993). Similarly Bifidocin A isolated from B. animalis BB04 exhibits wide range of bactericidal activity (against gram positive and negative bacteria and some yeasts) through cell lysis of pathogens, while acidocin B isolated from Bifidobacterium sp. inhibits Clostridium sp. in fermented food products (Bali et al. 2014; Liu et al. 2015). Antibiotic sensitive and resistant strains of H. pylori are inhibited by thermally stable bifidobacteria bacteriocins (at high temperatures of 80 and 100 °C) (Collado et al. 2005). Other bacteriocins produced by genus Bifidobacterium include bifidin I (from B. bifidum), bifilact (from B. lactis), thermophilicin (from B. thermophilum), bifilong and bisin (from B. longum) (Liu et al. 2015). These bacteriocins in general possess stability in a wide pH range of 3-10 and also towards the action of weak organic solvents. They have significant thermal stability and are resistant towards refrigeration, freezing, action of salts and enzymes. A more detailed understanding of their mechanism of action could promote them as potential candidates for application in food processing and preservation, heath care, prophylactic and pharmaceutical industries. 3.4. Enhancement of the Epithelial Barrier. Human GI tract is covered with a lining of epithelium decorated with a layer of protruding villi which isolates interior of the body from the external environment. The space in between is sealed with tight junctions which regulate though trans-membrane proteins and actin cytoskeleton (Ulluwishewa et al. 2011). The dynamic yet tight epithelial junctions between the villi prevent the entry of pathogens and undesirable metabolites into the blood stream while allowing essential nutrient to pass through, upon stimulation. However certain pathogens like Vibrio cholerae release endogenous toxins, increasing the permeability of the epithelial junctions (Arrieta et al. 2006). This often leads to serious chronic disorders known as Leaky gut syndrome (LGS) and IBS. Symptoms of LGS include inflammation and irritation allowing uninterrupted influx of pathogenic toxins into the blood stream compromising the endocrine and lymphatic system (Saggioro et al. 2014). Other than pathogen invasions, toxins, pro-inflammatory cytokines, inflammatory mediators, mast cells, dietary conditions, oxidative stress and production of hydrogen peroxide are some of the factors contributing to the lowering of epithelial barrier function of the human gut. Probiotic bifidobacteria operates through strain and dose dependent enhancement of gut epithelial barrier by several different ways. They stimulate the secretion of thick mucus, a layer that is being constantly replaced, thus preventing the strong adhesion of pathogens. Trans-
10
epithelial electrical resistance measurements show prevention of tumor necrosis factor (TNF-α) mediated disruption of epithelial barrier by certain bifidobacteria species (Hsieh et al. 2015; Ulluwishewa et al. 2011). Studies also report bifidobacteria mediated wound repair of Caco-2 cell monolayer pre-treated with TNF-α (Sultana et al. 2013). The enhancement of the barrier with increase in tight junction function is also attributed to the modulation of associated trans-membrane proteins (Hsieh et al. 2015). 3.5. Immunomodulatory effects. Many of the immunomodulatory bioactive signalling molecules of probiotics are microbe-associated molecular patterns (MAMPs) which interact with trans-membrane host pattern recognition receptors (PRRs) like toll-like receptors (TLRs) (Tomoda et al. 2015). The exact mechanism of interaction in bifidobacteria is still largely unknown, however may be mediated CHAP domain of TgaA or by MAMP-TLR2 mapping (Guglielmetti et al. 2014; Ventura et al. 2014). Bifidobacteria is reported to exhibit not just species but strain specific immunomodulatory potency. B. adolescentis DB-2458 and B. longum subsp. infantis GB-1496 isolated from human breast milk exerts strong immunomodulatory effects through Th1/Th2 associated cytokine regulation (Chiu et al. 2014). This increases the scope of their incorporation in probiotic formulation for infants. A dose dependent regulation of Th1/Th2 cytokine levels is observed in mitogenstimulated human peripheral blood mononuclear cells (PBMCs) by B. bifidus and B. infantis strains in combinations with L. acidophilus (Li et al. 2011). Dendritic cells (DCs) form the gateway to our immune system. B. breve CNCM1-4035 and its culture supernatant is capable of improving innate immune responses of human intestinal DCs against pathogenic Salmonella enterica serovar Typhi through TLR signalling pathways (Bermudez-Brito et al. 2013). The increase in anti-inflammatory cytokine IL-10 compared to pro-inflammatory cytokines and chemokines, TNF-α, is crucial in maintaining homeostasis against Salmonella inflammation. Upregulation of TLR9 gene transcription by both intact live cells and cell free culture supernatant of B. breve indicate that TLR9 signalling is a pathway for anti-inflammatory effects (Bermudez-Brito et al. 2013). While B. animalis subsp. lactis BB-12 is shown to down-regulate the expression of TLR-2 on PBMC and corresponding pro-inflammatory cytokine secretion in adults (Meng et al. 2015). Modulation of immune responses in vitro by induction of IL-10 production has also been reported for some members of B. adolescentis, B. longum and B. bifidum species (Vieira et al. 2015; Vitaliti et al. 2014). A combination of L. rhamnosus and B. breve M-16V is reported to supress the immune responses induced by cigarette smoke. In vitro expression of IL-1β, IL-6, IL-10, IL-23, TNFα, CXCL-8 and HMGB1 release in human THP-1 macrophages along with cigarette smoke-induced activation of TLR4, TLR9 and NF-κB signalling pathways were reported to be suppressed by the probiotic microorganisms (Mortaz et al. 2015).
11
Conjointly B. breve BR03 and L. salivarius LS01 show clinical efficacy in alleviating symptoms of allergic asthma, improving the functioning of lung. They increase the release of IL-10, an inhibitor of proinflammatory cytokines IL-13, IL-17 and TGF-β and rebalance Th1/Th2 levels (Drago et al. 2015). Serine protease forms an important class of effector molecule encoded by genes found in several bifidobacterial species. They inhibit the action of human proteases like neutrophils and pancreatic elastases, which are found at the site of intestinal inflammation (Ventura et al. 2014). 3.6. Competitive exclusion of pathogens. The gut microbiome is a natural environment harbouring a diverse collection of microbes. The key to existence of individual species is space for growth, binding receptor sites and access to nutritional resources within the GI tract. Hence, competition between individual species determines the homeostatic composition of the gut microbiota. Probiotic bacteria operate through strain specific antagonistic mechanisms for competitive exclusion of pathogens (Gueimonde et al. 2007). B. breve CNCM I-4035 is found to exhibit inhibitory effects on the growth of enterotoxigenic (ETEC) and enteropathogenic (EPEC) bacteria, while other species were found to inhibit adhesion of pathogens onto the intestinal mucus (Collado et al. 2006; Muñoz-Quezada et al. 2013; Uraipan et al. 2015). MAMP-PRR interactions play key role in association of pathogens with the intestinal epithelia. Probiotics also express molecular patterns which recognise the same trans-membrane receptors as the pathogens, thereby blocking the sites for pathogenic access by competitive exclusion and sometimes displacing already attached pathogens (Madsen 2012). Prebiotics often assists the competitive exclusion of enteric pathogen by competing for pathogen binding sites (Becerra et al. 2015; Chichlowski et al. 2012). The ability of bifidobacteria to efficiently sequester iron in the gut is a mechanism of competition for nutrients, causing iron starvation in EPEC bacteria (Vazquez-Gutierrez et al. 2015).
4. Roles of surface associated structural components. Research interest at present focuses on understanding the probiotic activity of cell wall components, to devise structural component based advanced therapeutics, replacing the use of intact cells. However the molecular basis of the strain-specific probiotic actions mediated by these effector molecules need to be thoroughly investigated. Of the different components associated, notable interest lies with exopolysaccharides, peptidoglycans and lipoteichoic acids. 4.1. Exopolysaccharides and biofilms. Densely packed communities of microbes surround themselves in slimy exopolymeric matrices known as biofilms. Polysaccharides being a major constituent in these matrices, referred to as exopolysacharides (EPS) have thus gained extensive attention. In this regard, works by Ruas-Madiedo,
12
Reyes-Gavilán and Sinderen are particularly noteworthy (Fanning et al. 2012; Hidalgo-Cantabrana et al. 2013 and 2014; Ruas-Madiedo et al. 2009; Salazar et al. 2008). Bifidobacteria strains are popularly associated with such exopolymeric substances, forming an interfacial layer separating the bacteria from its surrounding environment. Mechanistic investigation of mode of action of intact bifidobacteria cells has convincingly shown the assistance of exopolysaccharides in its probiotic activity. Thus efforts have been made to sequence genomes and identify genes related to EPS biosynthesis. Chemical identification of monosaccharide composition of purified EPS has been achieved for several Bifidobacterium spp. strains. These studies indicate variability in structural constitution and related gene clusters among species/subspecies within Bifidobacterium genus. Primordial challenge faced in devising probiotic supplement is the viability of the microorganism under physiological conditions of GI tract and retaining its probiotic property. Inspite of strain specific viability of bacteria, presence of EPS layer is attributed to its enhanced survival in human gut. Numerous direct and indirect experiments show that presence of EPS provides long term resistance to bifidobacteria towards the action of bile salts and acids (Alp et al. 2010; Fanning et al. 2012; Sánchez et al. 2014; Wickramasinghe et al. 2015). In fact reports suggest that bile induces/affects the production of EPS in certain strains of B. animalis and B. longum (Alp et al. 2010; Hidalgo-Cantabrana et al. 2013; Salazar et al. 2008; Sánchez et al. 2014). Recently through combined transcriptome and physiological approach, it was showed that enhanced acid tolerance of B. breve BB8dpH compared to its parent B. breve BB8 is related to expression of genes involved in biosynthesis of cell wall components, namely, peptidoglycan and EPS, along with those involved in carbohydrate transport, metabolism and energy production pathways (Yang et al. 2015). Similarly upregulation of several genes related to EPS synthesis in B. longum subsp. longum BBMN 68 was observed post acid adaptation (Jin et al. 2015). Along with EPS induced adhesion of bifidobacteria to intestinal epithelia (Inturri et al. 2014; López et al. 2012; Ventura et al. 2014), large number of studies have been aimed at its contribution in inhibition/exclusion of pathogens. EPS isolated from B. bifidum WBIN03, facilitates the growth of lactobacilli along with other anaerobic bacteria while simultaneously inhibits the growth of enterobacteria, enterococci and Bacteroides fragilis. 80μg/ml of EPS from B. longum BCRC 14634 is reported to inhibit food spoilage and pathogenic bacteria (Wu et al. 2010). The probiotic activity of EPS is attributed to its interference with the cell division process of the pathogens rather than cell lysis. While B. bifidum PRL2010 isolated from infant faeces were found to adhere onto human intestinal Caco-2 and HT-29 cell monolayers, thus preventing the adhesion of pathogenic E. coli and Cronobacter sakazakii (Serafini et al. 2013). It was also shown that EPS in EPS+B. breve
13
UCC2003 is responsible for its persistence in murine gut and colon and assisted in reducing colonisation by pathogenic C. rodentium (Fanning et al. 2012). Although no specific host receptors have been identified in bifidobacterial EPS recognition, EPS has been suggested as an effector in cross-talk between probiotic microorganism and host immune system (Hidalgo-Cantabrana et al. 2014; Turroni et al. 2014). EPS+B. breve UCC2003 treated mice when infected with C. rodentium shows lower levels of pro-inflammatory cytokines compared to the ones treated with EPS-B. breve UCC2003 strains, which has higher levels of IL-12, IFN-γ TNFα (Fanning et al. 2012). While treatment of murine macrophage cell line J77A.1 enhances the IL-10 levels and suppresses lipopolysaccharide (LPS) activity and TNF-α levels (Wu et al. 2010). An increase in suppressorregulatory TGF-β cytokine level is observed in Wistar rats fed with EPS producing B. animalis subsp. lactis IPLA R1 and B. longum IPLA E44 strains (Salazar et al.2014). 4.2. Petidoglycan. These biopolymers are one of the major components of the bifidobacteria cell walls and have been associated to its acid tolerance and immunomodulatory effects. In fact resistance of Bifidobacterium to acid stress is known to improve after pre-stressing cells at low pH. The acid tolerance response (ATR) at significantly low pH has been shown to increase the protein abundance related in peptidoglycan synthesis and other metabolic processes of B. longum subsp. longum BBMN68 (Jin et al. 2015; Zhang et al. 2015). The changes in EPS, peptidoglycan and cyclopropane fatty acid content of bifidobacteria induced by acid adaptation results in the strengthening of the cell wall, thereby blocking the entry of H + (Jin et al. 2012; Sánchez et al. 2014). A3α-peptidoglycan from Bifidobacterium cell wall is seen to enhance immune response promoting health in sea cucumber, Apostichopus japonicas, against Vibrio splendidus infection (Zhang et al. 2015). While peptidoglycan from B. bifidum induces apoptosis in colorectal carcinoma cells in mice (Li-sheng et al. 1999). 4.3. Lipoteichoic acids and lipoglycans. Cell surface associated lipoteichoic acid (LTA) often serves as major virulence factors in gram positive bacteria. These complex glycolipid structures (lipofuranan linked to monoglycerophospate groups with D-alanine substitutions) mediate interaction with host receptors, triggering signalling pathways, resulting in probiotic/pathogenic effects (Lebeer et al. 2010; Lee et al. 2013; Lynch et al. 2004). Along with LTA, closely resembling structures of macroamphiphilic lipoglycans have been reported in bifidobacteria, having L-alanine substituted single glycerol phosphate monomeric side chains attached to the glycan backbone by phosphodiester linkages (Lebeer et al. 2012; Fischer et al. 1987). These lipoglycans are also classified as type V LTA (Schneewind et al. 2014). Immunomodulatory role of LTA in probiotic Lactobacillus through TLR2 recognition is well established. Although these biopolymers have long been identified in bifidobacteria, their roles in PRR mediated interaction in host have not yet been properly studied. However Guo
14
et al. (2015) to our best knowledge first showed bifidobacterial LTA in combination with 5-fluorouracil to exhibit anti-tumor effects which alleviate chemotherapeutic side effects by immune modulatory activity (Xie et al. 2012). Nevertheless, there is clear need for more research in this direction along with in vivo demonstration of LTA mediated biological activities. As seen from discussion, clinical reports show that administration of bifidobacteria along with bacteria belonging to other probiotic genera to exhibit combinatorial effects in a host (Guo et al 2015; Isidro et al. 2015; Mortaz et al 2015). Bifidobacteria have been widely reported to exhibit strain specific probiotic activity through immune modulation, pathogen exclusion, production of metabolites and bacteriocins and surface associated biopolymers. Such differential behaviour could be ascribed to the strain-dependent molecular mechanism of operation. Enhanced efficacy of multi-strain/combinatorial therapy reported in pathogen combat possibly arises due to a combination of several probiotic mechanisms, with each strain making their own contributions. Although such therapy has been widely put to use, underlying mechanisms attributed to each component has not been much analysed. Careful selection of strains based on their probiotic mechanism could help in developing targeted formulations against different pathogens.
7. Conclusion The increased studies aimed at establishing probiotic mechanisms and determining clinical efficacy in the past decade indicates the promising future of bifidobacteria in replacing partially/completely antibiotics in the treatment of several pathological conditions. Probiotics have bestowed beneficial health effects in long term therapies especially in immune-mediated and atopic diseases almost in all age groups. Simultaneously infrequent studies report contradictory findings challenge the use of the probiotic microorganism, prompting the need for in-depth study prior to their administration to patients. Successful combinatorial therapy calls for analysis of strain-dependent probiotic mechanisms involved. Such efforts could help in selecting strains for customised formulations with synergistic effects against specific pathogens. Intake of intact cells along with beneficial health effects might pose risks of mutations and intrinsic resistance leading to antibiotic resistant phenotypes. The advancement in the field of probiotics has begun the quest for developing new era probiotics which aim to replace the use of live intact microorganisms by their heat inactivated counterparts or structural components responsible for their probiotic activity. Purified cell-surface components such as EPS, LTA along with metabolites and bifidocins might play a pivotal role in replacing intact cell therapy. Inspite of the promising prospect of bifidobacteria, corroborative studies are needed for
15
understanding the role of bifidobacteria in altering gut microbial populations and associated physiological effects in humans.
Conflict of Interest: There is no conflict of authorship and both the authors approve the final version of the manuscript.
Acknowledgement. The authors would like to express their gratitude towards The University of Manchester. Both the authors are currently Research Associates at The University of Manchester. Dr. Amrita Sarkar is the submission’s guarantor. The topic for the review is conceived by her. She has been involved in the major literature study, data streamlining, graphic designing and manuscript writing. Dr. Santanu Mandal has assisted in scientific discussions and editing the manuscript.
References Akkasheh G, Kashani-Poor Z, Tajadadi-Ebrahimi M, et al. (2016) Clinical and metabolic response to probiotic administration in patients with major depressive disorder: a randomized, double-blind, placebo-controlled trial. Nutrition 32, 315-320. Alm B, Erdes L, Möllborg P, et al. (2008) Neonatal Antibiotic Treatment Is a Risk Factor for Early Wheezing. Pediatrics 121, 697-702. Alp G, Aslim B, Suludere Z, Akca G. (2010) The role of hemagglutination and effect of exopolysaccharide production on bifidobacteria adhesion to Caco-2 cells in vitro. Microbiol Immunol 54, 658-665. Alp G, Aslim B. (2010) Relationship between the resistance to bile salts and low pH with exopolysaccharide (EPS) production of Bifidobacterium spp. isolated from infants feces and breast milk. Anaerobe 16, 101-105. Arrieta MC, Bistritz L, Meddings JB. (2006) Alterations in intestinal permeability. Gut; 55, 1512-1520. Asarat M, Apostolopoulos V, Vasiljevic T, Donkor O. (2015) Short-chain fatty acids produced by synbiotic mixtures in skim milk differentially regulate proliferation and cytokine production in peripheral blood mononuclear cells, on in peripheral blood mononuclear cells. Int J Food Sci Nutr 66, 755-765. Babenko LP, Lazarenko LM, Shynkarenko LM, Mokrozub VV, Pidgorskyi VS, Spivak MJ. (2012) The effect of lacto- and bifidobacteria compositions on the vaginal microflora in cases of intravaginal staphylococcosis. Mikrobiol Z 74, 116-125.
16
Bailey LC, Forrest CB, Zhang P, et al. (2014) Association of Antibiotics in Infancy With Early Childhood Obesity. JAMA Pediatr 186, 1063-1069. Bajaj BK, Claes IJJ, Lebeer S. (2015) Functional mechanisms of probiotics. J Microbiol Biotech Food Sci 4, 321-327. Balamurugan R, Janardhan HP, George S, Chittaranjan SP, Ramakrishna BS. (2008) Bacterial succession in the colon during childhood and adolescence: molecular studies in a southern Indian village. Am J Clin Nutr 88, 1643-1647. Bali V, Panesar PS, Bera MB, Kennedy JF. (2014) Bacteriocins: Recent Trends and Potential Applications. Crit Rev Food Sci Nutr DOI: 10.1080/10408398.2012.729231 Becerra JE, Coll-Marqués JM, Rodríguez-Díaz J, Monedero V, Yebra MJ. (2015) Preparative scale purification of fucosyl-N-acetylglucosamine disaccharides and their evaluation as potential prebiotics and antiadhesins. Appl Microbiol Biotechnol 99, 7165-7176. Bermudez-Brito M, Muñoz-Quezada S, Gomez-Llorente C, et al. (2013) Cell-Free Culture Supernatant of Bifidobacterium breve CNCM I-4035 Decreases Pro-Inflammatory Cytokines in Human Dendritic Cells Challenged with Salmonella typhi through TLR Activation. PLOS ONE 8, 1-8. Bermudez-Brito M, Plaza-Díaz J, Muñoz-Quezada S, Gómez-Llorente C, Gil A. (2012) Probiotic Mechanisms of Action. Ann Nutr Metab 61,160-174. Bertelli C, Pillonel T, Torregrossa A, et al. (2015) Bifidobacterium longum Bacteremia in Preterm Infants Receiving Probiotics. Clin Infect Dis 60, 924-927. Bindels LB, Porporato P, Dewulf EM, et al. (2012) Gut microbiota-derived propionate reduces cancer cell proliferation in the liver. Br J Cancer 107, 1337-1344. Borriello SP, Hammes WP, Holzapfel W, et al. (2003) Safety of Probiotics That Contain Lactobacilli or Bifidobacteria. Clin Infect Dis 36, 775-780. Bouhnik Y, Attar A, Joly FA, Riottot M, Dyard F, Flourie´ B. (2004) Lactulose ingestion increases faecal bifidobacterial counts: A randomised double-blind study in healthy humans. Eur J Clin Nutr 58, 462-466. Bourne KA, Beebe JL, Lue YA, et al. (1978) Bacteremia due to Bifidobacterium, Eubacterium or Lactobacillus; twenty-one cases and review of the literature. Yale J Biol Med 51, 505-512. Boyle RJ, Robins-Browne RM, Tang MLK. (2006) Probiotic use in clinical practice: what are the risks? Am J Clin Nutr 83, 1256-1264.
17
Braga TD, da Silva GAP, de Lira PIC, Lima MDC. (2011) Efficacy of Bifidobacterium breve and Lactobacillus casei oral supplementation on necrotizing enterocolitis in very-low-birth-weight preterm infants: a double-blind, randomized, controlled trial. Am J Clin Nutr 93, 81-86. Brook I, Frazier EH. (1993) Significant recovery of nonsporulating anaerobic rods from clinical specimens. Clin Infect Dis 16, 476-480. Bush LM, De Almeida KNF, Martin G, Perez MT. (2014) Probiotic-Associated Bifidobacterium Septic Prosthetic Joint Arthritis. Infect Dis Clin Pract 22, e39-e41. Candela M, Biagi E, Centanni M, et al. (2009) Bifidobacterial enolase, a cell surface receptor for human plasminogen involved in the interaction with the host. Microbiology 155, 3294-3303. Castellazzi AM, Valsecchi C, Caimmi S, et al. (2013) Probiotics and food allergy. Ital J Pediatr 39, 47-56. Cheikhyoussef A, Pogori N, Chen H, et al. (2009) Antimicrobial activity and partial characterization of bacteriocin-like inhibitory substances (BLIS) produced by Bifidobacterium infantis BCRC 14602. Food Control 20, 553-559. Chenoll E, Casinos B, Bataller E, et al. (2011) Novel Probiotic Bifidobacterium bifidum CECT 7366 Strain Active against the Pathogenic Bacterium Helicobacter pylori. Appl Environ Microbiol 77, 1335-1343. Chichlowski M, De Lartigue G, German JB, Raybould HE, Mills DA. (2012) Bifidobacteria isolated from infants and cultured on human milk oligosaccharides affect intestinal epithelial function. J. Pediatr Gastroenterol Nutr 55, 321-327. Chiu Y-H, Tsai J-J, Lin S-L, Chotirosvakin C, Lin M-Y. (2014) Characterisation of bifidobacteria with immunomodulatory properties isolated from human breast milk. J Funct Foods 7, 700-708. Chong ESL. (2014) A potential role of probiotics in colorectal cancer prevention: review of possible mechanisms of action. World J Microbiol Biotechnol 30, 351-374. Collado MC, González A, González R, Hernández M, Ferrús MA, Sanz Y. (2005) Antimicrobial peptides are among the antagonistic metabolites produced by Bifidobacterium against Helicobacter pylori. Int J Antimicrob Ag 25, 385-391. Collado MC, Gueimonde M, Sanz Y, Salminen S. (2006) Adhesion properties and competitive pathogen exclusion ability of bifidobacteria with acquired acid resistance. J Food Prot 69, 1675-1679. Collado MC, Hernández M, Sanz Y. (2005) Production of bacteriocin-like inhibitory compounds by human fecal Bifidobacterium strains. J Food Prot 68, 1034-1040.
18
Davenport ER, Mizrahi-Man O, Michelini K, et al. (2014) Seasonal Variation in Human Gut Microbiome Composition, PLoS ONE 9, 1-10. Delgado S, O’Sullivan E, Fitzgerald G, Mayo B. (2008) In vitro evaluation of the probiotic properties of human intestinal Bifidobacterium species and selection of new probiotic candidates. J Appl Microbiol 104: 1119-1127. Demers M, Dagnault A, Desjardins J. (2014) A randomized double-blind controlled trial: Impact of probiotics on diarrhea in patients treated with pelvic radiation. Clinical Nutrition 33, 761-767. Dennis JC, Culpepper T, Nieves CJ. et al. (2016) Probiotics (Lactobacillus gasseri KS-13, Bifidobacterium bifidum G9-1, and Bifidobacterium longum MM-2) and Health-Related Quality of Life in Individuals with Seasonal Allergies. FASEB J 30, 916-917. d’Ettorre G, Ceccarelli G, Giustini N, et al. (2015) Probiotics Reduce Inflammation in Antiretroviral Treated, HIV-Infected Individuals: Results of the “Probio-HIV” Clinical Trial. PLoS ONE 10, 1-15. Dilli D, Aydin B, Fettah ND. (2015) The Pro Pre-Save Study: Effects of Probiotics and Prebiotics Alone or Combined on Necrotizing Enterocolitis in Very Low Birth Weight Infants. J Pediatr 166, 545-551. Dinleyici EC, Dalgic N, Guven S, et al. (2013) The effect of a multispecies synbiotic mixture on the duration of diarrhea and length of hospital stay in children with acute diarrhea in Turkey: Single blinded randomized study. Eur J Pediatr 172, 459-464. Drago L, Vecchi ED, Gabrieli A, Grandi RD, Toscano M. (2015) Immunomodulatory Effects of Lactobacillus salivarius LS01 and Bifidobacterium breve BR03, Alone and in Combination, on Peripheral Blood Mononuclear Cells of Allergic Asthmatics. Allergy Asthma Immunol Res 7, 409-413. Eichenfield LF, Tom WL, Berger TG, et al. (2014) Guidelines of care for the management of atopic dermatitis: Section 2. Management and treatment of atopic dermatitis with topical therapies. J Am Acad Dermatol 71, 116132. El-Soud NHA, Said RN, Mosallam DS, Baraka NAM, Sabry MA. (2015) Bifidobacterium lactis in Treatment of Children with Acute Diarrhea. A Randomized Double Blind Controlled Trial. J Med Sci 3, 403-407. Enomoto T, Sowa M, Nishimori K, et al. (2014) Effects of bifidobacterial supplementation to pregnant women and infants in the prevention of allergy development in infants and on fecal microbiota. Allergol Int 63, 575585. Erdogan O, Tanyeri B, Torun E, et al. (2012) The Comparition of the Efficacy of Two Different Probiotics in Rotavirus Gastroenteritis in Children. J Trop Med 2012, 1-5.
19
Esko JD, Sharon N. (2009) Essentials of Glycobiology. Chapter 34: Microbial Lectins: Hemagglutinins, Adhesins, and Toxins. Varki A, Cummings RD, Esko JD, et al. (Eds). 2nd Edition. Cold Spring Harbor (NY): Cold Spring Harbor Laboratory Press. Fanning S, Hall LJ, Cronin M, et al. (2012) Bifidobacterial surface-exopolysaccharide facilitates commensalhost interaction through immune modulation and pathogen protection. PNAS 109, 2108-2113. Fischer W. (1987) ‘Lipoteichoic acid’ of Bifidobacterium bifidum subspecies pennsylvanicum DSM 20239. Eur J Biochem 165, 639-646. Fliss I, Ouwehand AC, Kheadr E, Lahtinen S, Davids SJ. (2010) Bifidobacteria: Genomics and Molecular Aspects, Chapter 7: Antimirobial Activity of the Genus Bifidobacterium. Mayo B, van Sinderen D. (Eds). Caister Academic Press, Norfolk, UK. Foroni E, Serafini F, Amidani D, et al. (2011) Genetic analysis and morphological identification of pilus-like structures in members of the genus Bifidobacterium. Microb Cell Fact 10 (Suppl 1), 1-13. Fox MJ, Ahuja KDK, Robertson IK, Ball MJ, Eri RD. (2015) Can probiotic yogurt prevent diarrhoea in children on antibiotics? A double-blind, randomised, placebo-controlled study. BMJ Open 5, 1-6. Fukuda S, Toh H, Hase K, et al. (2011) Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 469, 543-547. Gioia DD, Gaggia F, Baffoni L, Stenico V. (2014) Beneficial Microbes in Fermented and Functional Foods. Chapter 16: Role of Bifidobacteria in the production of bioactive compounds and detoxification of harmful compounds. Rai VR, Bai JA. (Eds), CRC Press, NewYork. Gleinser M, Grimm V, Zhurina D, Yuan J, Riede U. (2012) Improved adhesive properties of recombinant bifidobacteria expressing the Bifidobacterium bifidum-specific lipoprotein BopA. Microb Cell Fact 11, 1-14. González-Rodríguez
I, Sánchez
B, Ruiz
L, et
al.
(2012)
Role
of
Extracellular
Transaldolase
from Bifidobacterium bifidum in Mucin Adhesion and Aggregation. Appl Environ Microbiol 78, 3992-3998. Groschwitz KR, Hogan SP. (2009) Intestinal barrier function: Molecular regulation and disease pathogenesis. J Allergy Clin Immunol 124, 3-20. Gueimonde M, Margolles A, de los Reyes-Gavilán CG, Salminen S. (2007) Competitive exclusion of enteropathogens from human intestinal mucus by Bifidobacterium strains with acquired resistance to bile-A preliminary study. Int J Food Microbiol 113, 228-232.
20
Guglielmetti S, Mora D, Gschwender M, Popp K. (2011) Randomised clinical trial: Bifidobacterium bifidum MIMBb75 significantly alleviates irritable bowel syndrome and improves quality of life-a double-blind, placebo-controlled study. Aliment Pharmacol Ther 33, 1123-1132. Guglielmetti S, Tamagnini I, Mora D, et al. (2008) Implication of an Outer Surface Lipoprotein in Adhesion of Bifidobacterium bifidum to Caco-2 Cells. Appl Environ Microbiol 74, 4695-4702. Guglielmetti S, Zanoni I, Balzaretti S, et al. (2014) Murein Lytic Enzyme TgaA of Bifidobacterium bifidum MIMBb75 Modulates Dendritic Cell Maturation through Its Cysteine- and Histidine-Dependent Amidohydrolase/Peptidase (CHAP) Amidase Domain. Appl Environ Microbiol 80, 5170-5177. Guo B, Xie N, Wang Y. (2015) Cooperative effect of Bifidobacteria lipoteichoic acid combined with 5fluorouracil on hepatoma-22 cells growth and apoptosis. Bulletin du Cancer 102, 204-212. Guo S, Guo Y, Ergun A, Lu L, Walker WA, Ganguli K. (2015) Secreted Metabolites of Bifidobacterium infantis and Lactobacillus acidophilus Protect Immature Human Enterocytes from IL-1β-Induced Inflammation: A Transcription Profiling Analysis. PLoS ONE 10, 1-19. Ha GY, Yang CH, Kim H, Chong Y. (1999) Case of Sepsis Caused by Bifidobacterium longum. J Clin Microbiol 37, 1227-1228. Hata D, Yoshida A, Ohkubo H, et al. Meningitis caused by Bifidobacterium in an infant. (1988) Pediatr Infect Dis J 7, 669-671. Hidalgo-Cantabrana C, Nikolic M, López P, et al. (2014) Exopolysaccharide-producing Bifidobacterium animalis subsp. lactis strains and their polymers elicit different responses on immune cells from blood and gut associated lymphoid tissue. Anaerobe 26, 24-30. Hidalgo-Cantabrana C, Sánchez B, Álvarez-Martín P, et al. (2015) A single mutation in the gene responsible for the mucoid phenotype of Bifidobacterium animalis subsp. lactis confers surface and functional characteristics. Appl Environ Microbiol 81, 7960-7968. Hidalgo-Cantabrana C, Sánchez B, Milani C, Ventura M, Margolles A, Ruas-Madiedo P. (2014) Genomic overview and biological functions of exopolysaccharide biosynthesis in Bifidobacterium spp. Appl Environ Microbiol 80, 9-18. Hidalgo-Cantabrana C, Sánchez B, Moine D, et al. (2013) Insights into the Ropy Phenotype of the Exopolysaccharide-Producing Strain Bifidobacterium animalis subsp. lactis A1dOxR. Appl Environ Microbiol; 79, 3870-3874.
21
Hsieh C-Y, Osaka T, Moriyama E, Date Y, Kikuchi J, Tsuneda S. (2015) Strengthening of the intestinal epithelial tight junction by Bifidobacterium bifidum. Physiol Rep 3, 1-17. Hussey S, Wall R, Gruffman E, et al. (2011) Parenteral Antibiotics Reduce Bifidobacteria Colonization and Diversity in Neonates. Int J Microbiol 2011, 1-6. Inturri R, Stivala A, Sinatra F, Morrone R, Blandino G. (2014) Scanning Electron Microscopy Observation of Adhesion Properties of Bifidobacterium longum W11 and Chromatographic Analysis of Its Exopolysaccaride. Food Nutr Sci 5, 1787-1792. Isidro RA, Bonilla FJ, Pagan H, et al.( 2015) The Probiotic Mixture VSL#3 Alters the Morphology and Secretion Profile of Both Polarized and Unpolarized Human Macrophages in a Polarization-Dependent Manner. J Clin Cell Immunol 5, 1-20. İşlek A, SayarE, Yilmaz A, Baysan BÖ, Mutlu D, Artan R. (2014) The role of Bifidobacterium lactis B94 plus inulin in the treatment of acute infectious diarrhea in children. Turk J Gastroenterol 25, 628-633. Jeon SG, Kayama H, Ueda Y, et al. (2012) Probiotic Bifidobacterium breve Induces IL-10-Producing Tr1 Cells in the Colon. PLoS Pathogens 8, 1-15. Jin J, Zhang B, Guo H, et al. (2015) Effect of Pre-Stressing on the Acid-Stress Response in Bifidobacterium Revealed Using Proteomic and Physiological Approaches. PLoS ONE 10, 1-14. Jin J, Zhang B, Guo H, et al. (2012) Mechanism Analysis of Acid Tolerance Response of Bifidobacterium longum subsp. longum BBMN 68 by Gene Expression Profile Using RNA-Sequencing. PLoS ONE 7, 1-18. Kailasapathy K, Chin J. (2000) Survival and therapeutic potential of probiotic organisms with reference to Lactobacillus acidophilus and Bifidobacterium spp. Immunol Cell Biol 78, 80-88. Kainulainen V, Reunanen J, Hiippala K, et al. (2013) BopA Does Not Have a Major Role in the Adhesion of Bifidobacterium bifidum to Intestinal Epithelial Cells, Extracellular Matrix Proteins, and Mucus. Appl Environ Microbiol 79, 6989-6997. Kavanaugh DW, O’Callaghan J, Buttó LF, et al. (2013) Exposure of Bifidobacterium longum subsp. infantis to Milk Oligosaccharides Increases Adhesion to Epithelial Cells and Induces a Substantial Transcriptional Response. PLoS ONE 8, 1-13. Kirkali Z, Rasooly R, Star RA, Rodgers GP. (2015) Urinary Stone Disease: Progress, Status, and Needs. Urology 86, 651-653. Klaenhammer TR. (1993) Genetics of bacteriocins produced by lactic acid bacteria. FEMS Microbiol Rev 12, 39-86.
22
Kullin BR, Reid SJ, Abratt VR. (2016) The Role of Bacteria in Urology. Chapter 7: The Use of Probiotic Bacteria to Treat Recurrent Calcium Oxalate Kidney Stone Disease. Lange D, Chew B. (Eds). Springer International Publishing, Switzerland. Lagier J-C, Million M, Hugon P, Armougom F, Raoult D. (2012) Human gut microbiota: repertoire and variations. Front Cell Infect Microbiol 2, 1-19. Leahy SC, Higgins DG, Fitzgerald GF, van Sinderen D. (2005) Getting better with bifidobacteria. J Appl Microbiol 98, 1303-1315. Lee DK, Jang S, Kim MJ, et al. (2008) Anti-proliferative effects of Bifidobacterium adolescentisSPM0212 extract on human colon cancer cell lines. BMC Cancer 8, 310-318. Lee I-C, Tomita S, Kleerebezem M, Bron PA. (2013) The quest for probiotic effector molecules-unraveling strain specificity at the molecular level. Pharmacol Res 69, 61-74. Lebeer S, Vanderleyden J, De Keersmaecker SC. (2010) Host interactions of probiotic bacterial surface molecules: comparison with commensals and pathogens. Nat Rev Microbiol 8, 171-184. Lebeer S, Claes IJJ, Vanderleyden J. (2012) Anti-inflammatory potential of probiotics: lipoteichoic acid makes a difference. Trends Microbiol 20, 5-10. Li C-Y, Lin H-C, Lai C-H, Lu JJ-Y, Wu S-F, Fang S-H. (2011) Immunomodulatory Effects of Lactobacillus and Bifidobacterium on Both Murine and Human Mitogen-Activated T Cells. Int Arch Allergy Immunol 156, 128-136. Li-sheng W, Ling-jia P, Li S, Yong S, Ya-li Z, Dian-yuan Z. (1999) The apoptosis of experimental colorectal carcinoma cells induced by peptidoglycan of bifidobacterium and the expression of apoptotic regulating genes. Chinese J Cancer Res 11, 184-187. Liu G, Ren L, Song Z, Wang C, Sun B. (2015) Purification and characteristics of bifidocin A, a novel bacteriocin produced by Bifidobacterium animals BB04 from centenarians' intestine. Food Control 50, 889-895. López P, Monteserín DC, Gueimonde M, et al. (2012) Exopolysaccharide-producing Bifidobacterium strains elicit different in vitro responses upon interaction with human cells. Food Res Int 46, 99-107. Lynch NJ, Roscher S, Hartung T, et al. (2004) L-Ficolin Specifically Binds to Lipoteichoic Acid, a Cell Wall Constituent of Gram-Positive Bacteria, and Activates the Lectin Pathway of Complement. J Immunol 172: 1198-1202. Madsen KL. (2012) Enhancement of Epithelial Barrier Function by Probiotics. J Epithel Biol Pharmacol 5 (Supplement 1-M8), 55-59.
23
Mahlen SD, Clarridge JE. (2009) Site and Clinical Significance of Alloscardovia omnicolens and Bifidobacterium Species Isolated in the Clinical Laboratory. J Clin Microbiol 47, 3289-3293. Margolles A, Ruas-Madiedo P, de los Reyes-Gavilán CG, Sánchez B, Gueimonde M. (2011) Molecular Detection of Human Bacterial Pathogens. Chapter 5: Bifidobacterium. Liu D (Ed). CRC Press, Taylor & Francis. Marra F, Lynd L, Coombes M, et al. (2006) Does Antibiotic Exposure During Infancy Lead to Development of Asthma?: A Systematic Review and Metaanalysis. Chest 129, 610-618. Martinez FA, Balciunas EM, Converti A, Cotter PD, de Souza ORP (2013) Bacteriocin production by Bifidobacterium spp. A review. Biotechnol Adv 31, 482-488. Matsuki T, Pédron T, Regnault B, Mulet C, Hara T, Sansonetti PJ. (2013) Epithelial Cell Proliferation Arrest Induced by Lactate and Acetate from Lactobacillus casei and Bifidobacterium breve. PLoS One 8, 1-8. Meneghin F, Fabiano V, Mameli C, Zuccotti GV. (2012) Probiotics and Atopic Dermatitis in Children. Pharmaceuticals (Basel) 5, 727-744. Meng H, Ba Z, Lee Y, et al. (2015) Consumption of Bifidobacterium animalis subsp. lactis BB-12 in yogurt reduced expression of TLR-2 on peripheral blood-derived monocytes and pro-inflammatory cytokine secretion in young adults. Eur J Nutr 1-13. DOI 10.1007/s00394-015-1109-5 Minami J-i, Kondo S, Yanagisawa N, et al. (2015) Oral administration of Bifidobacterium breve B-3 modifies metabolic functions in adults with obese tendencies in a randomised controlled trial. J Nutri Sci 4, 1-7. Mitsuoka T. (1990) Bifidobacteria and their role in human health. J Ind Microbiol 6, 263-268. Mortaz E, Adcock IM, Ricciardolo FLM, et al. (2015) Anti-Inflammatory Effects of Lactobacillus Rahmnosus and Bifidobacterium Breve on Cigarette Smoke Activated Human Macrophages. PLoS ONE 10, 117. Motherway MOC, Zomer A, Leahy SC et al. (2011) Functional genome analysis of Bifidobacterium breve UCC2003 reveals type IVb tight adherence (Tad) pili as an essential and conserved host-colonization factor. PNAS 108, 11217-11222. Moran JP, Walter J, Tannock GW, Tonkonogy SL, Sartor RB. Bifidobacterium animalis causes extensive duodenitis and mild colonic inflammation in monoassociated interleukin-10-deficient mice. Inflamm Bowel Dis 15, 1022-1031. Muñoz-Quezada S, Bermudez-Brito M, Chenoll E, et al. (2013) Competitive inhibition of three novel bacteria isolated from faeces of breast-fed infants against selected enteropathogens. Br J Nutr 109, S63-S69.
24
Myhre R, Brantsæter AL, Myking S, et al. (2011) Intake of probiotic food and risk of spontaneous preterm delivery. Am J Clin Nutr 93, 151-157. Ohishi A, Takahashi S, Ito Y, et al. (2010) Bifidobacterium septicemia associated with postoperative probiotic therapy in a neonate with omphalocele. J Pediatr 156 679-681. 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, 541-551. Nishihira J, Kagami-Katsuyama H, Tanaka A, Nishimura M, Kobayashi T, Kawasaki Y. (2014) Elevation of natural killer cell activity and alleviation of mental stress by the consumption of yogurt containing Lactobacillus gasseri SBT2055 and Bifidobacterium longum SBT2928 in a double-blind, placebo-controlled clinical trial. J Funct Foods 11, 261-268. Osman N, Adawi D, Molin G, Ahrne S, Berggren A, Jeppsson B. (2006) Bifidobacterium infantis strains with and without a combination of Oligofructose and Inulin (OFI) attenuate inflammation in DSS-induced colitis in rats. BMC Gastroenterology 6, 1-10. Picard C, Fioramonti J, Francois A, Robinson T, Neant F, Matuchansky C. (2005) Review article: bifidobacteria as probiotic agents – physiological effects and clinical benefits, Aliment Pharmacol Ther 22, 495-512. Poltavska OA, Kovalenko NK. (2012) Antimicrobial activity of bifidobacterial bacteriocin-like substances. Mikrobiol Z 74, 32-42. Presti I, D'Orazio G, Labra M, et al. (2015) Evaluation of the probiotic properties of new Lactobacillus and Bifidobacterium strains and their in vitro effect. Appl Microbiol Biotechnol 99, 5613-5626. Puertollano E, Kolida S, Yaqoob P. (2014) Biological significance of short-chain fatty acid metabolism by the intestinal microbiome. Curr Opin Clin Nutr Metab Care 17, 139-144. Ranadheera CS, Evans CA, Adams MC, Baines SK. (2014) Effect of dairy probiotic combinations on in vitro gastrointestinal tolerance, intestinal epithelial cell adhesion and cytokine secretion. J Funct Foods 8C, 18-25. Ring J, Alomar A, Bieber T, et al. (2012) Guidelines for treatment of atopic eczema (atopic dermatitis) Part I. J Eur Acad Dermatol Venereol 26, 1045-1060. Rodes L, Khan A, Paul A, et al. (2013) Effect of Probiotics Lactobacillus and Bifidobacterium on Gut-Derived Lipopolysaccharides and Inflammatory Cytokines: An In Vitro Study Using a Human Colonic Microbiota Model. J Microbiol Biotechnol 23, 518-526. Roger LC, Costabile A, Holland DT, Hoyles L. McCartney AL. (2010) Examination of faecal Bifidobacterium populations in breast- and formula-fed infants during the first 18 months of life. Microbiology 156, 3329-3341.
25
Rossi M, Amaretti A, Raimondi S. (2011) Folate Production by Probiotic Bacteria. Nutrients 3, 118-134. Rossi R, Rossi L, Fassio F. (2015) Clinical Follow-up of 96 Patients Affected by Irritable Bowel Syndrome Treated with a Novel Multi-strain Symbiotic. J Contemp Immunol 2, 49-58. Ruiz L, Margolles A, Sánchez B. (2013) Bile resistance mechanisms in Lactobacillus and Bifidobacterium. Front Microbiol 4, 1-8. Ruas-Madiedo P, Gueimonde M, Arigoni F, de los Reyes-Gavilán CG, Margolles A. (2009) Bile Affects the Synthesis of Exopolysaccharides by Bifidobacterium animalis. Appl Environ Microbiol 75, 1204-1207. Saez-Lara MJ, Gomez-Llorente C, Plaza-Diaz J, Gil A. (2015) The Role of Probiotic Lactic Acid Bacteria and Bifidobacteria in the Prevention and Treatment of Inflammatory Bowel Disease and Other Related Diseases: A Systematic Review of Randomized Human Clinical Trials. BioMed Res Int 2015, 1-15. Saggioro A. (2014) Leaky Gut, Microbiota, and Cancer: An Incoming Hypothesis. J Clin Gastroenterol 48, S62-S66. 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. Clinical Nutrition 34, 501-507. Salazar N., Gueimonde M., Hernandez-Barranco AM., Ruas-Madiedo P, de los Reyes-Gávilan CG. (2008) Exopolysaccharides Produced by Intestinal BifidobacteriumStrains Act as Fermentable Substrates for Human Intestinal Bacteria. Appl Environ Microbiol 74, 4737-4745. Salazar N, López P, Garrido P, et al. (2014) Immune Modulating Capability of Two ExopolysaccharideProducing Bifidobacterium Strains in a Wistar Rat Model. BioMed Res Int 2014: 1-9. Salazar N, Ruas-Madiedo P, Prieto A, Calle LP, de los Reyes-Gávilan CG. (2012) Characterization of Exopolysaccharides Produced by Bifidobacterium longum NB667 and Its Cholate-Resistant Derivative Strain IPLA B667dCo. J Agric Food Chem 60, 1028-1035. Sánchez B, González-Rodríguez I, de los Reyes Gavilán CG, Ruas-Madiedo P, Margolles A. (2014) Interactive Probiotics. Chapter 5: Bifidobacteria and their Interaction with the Gastrointestinal Environment. Pessione E. (Ed). CRC Press, New York. Schneewind O, Missiakas D. (2014) Lipoteichoic Acids, Phosphate-Containing Polymers in the Envelope of Gram-Positive Bacteria. J Bacteriol 196, 1133-1142. Schwiertz A, Knauf M, Pohl U, Hackel B, Mueller H-J. (2015) Effectiveness and Tolerability of a Synbiotic Vaginal Suppository for the Treatment of Bacterial Vaginosis. Gynecol Obstet 5 1-6.
26
Serafini F, Strati F, Ruas-Madiedo P, et al. (2013) Evaluation of adhesion properties and antibacterial activities of the infant gut commensal Bifidobacterium bifidum PRL2010. Anaerobe 21, 9-17. [17] Shen J, Zuo Z-X, Mao A-P. (2014) Effect of Probiotics on Inducing Remission and Maintaining Therapy in Ulcerative Colitis, Crohn’s Disease, and Pouchitis: Meta-analysis of Randomized Controlled Trials. Inflamm Bowel Dis 20, 21-35. Singh A, Hacini-Rachinel F, Gosoniu ML, et al. (2013) NCC2818 in individuals suffering from seasonal allergic rhinitis to grass pollen: an exploratory, randomized, placebo-controlled clinical trial. Eur J Clin Nutr 67, 161-167. Sugahara H, Odamaki T, Fukuda S, et al. (2015) Probiotic Bifidobacterium longum alters gut luminal metabolism through modification of the gut microbial community. Scientific Reports 5, 1-11. Sultana R, McBain AJ, O'Neill CA. (2013) Strain-Dependent Augmentation of Tight-Junction Barrier Function in Human Primary Epidermal Keratinocytes by Lactobacillus and Bifidobacterium Lysates. Appl Environ Microbiol 79, 4887-4894. Suzuki K, Nishiyama K, Miyajima H, Osawa R, Yamamoto Y, Muka T. (2016) Adhesion properties of a putative polymorphic fimbrial subunit protein from Bifidobacterium longum subsp. longum, Biosci Microbiota Food Health 35, 19-27. Tabasco R, Palencia PFD, Fontecha J, Peláez C, Requena T. (2014) Competition mechanisms of lactic acid bacteria and bifidobacteria: Fermentative metabolism and colonization. LWT - Food Sci Technol 55, 680-684. Takahashi S, Anzawa D, Takami K et al. (2016) Effect of Bifidobacterium animalis ssp. lactis GCL2505 on visceral fat accumulation in healthy Japanese adults: a randomized controlled trial. Biosci Microbiota Food Health http://doi.org/10.12938/bmfh.2016-002 Tamaki H, Nakase H, Inoue S, et al. (2016) Efficacy of probiotic treatment with Bifidobacterium longum 536 for induction of remission in active ulcerative colitis: a randomized, double-blinded, placebo-controlled multicenter trial. Dig Endosc 28, 67-74. Tejero-Sariñena S, Barlow J, Costabile A, Gibson GR, Rowland I. (2012) In vitro evaluation of the antimicrobial activity of a range of probiotics against pathogens: Evidence for the effects of organic acids. Anaerobe 18, 530-538. Toiviainen A, Jalasvuori H, Lahti E, et al. (2015) Impact of orally administered lozenges with Lactobacillus rhamnosus GG and Bifidobacterium animalis subsp. lactis BB-12 on the number of salivary mutans
27
streptococci, amount of plaque, gingival inflammation and the oral microbiome in healthy adults. Clin Oral Invest 19, 77-83. Tomasik J, Yolken RH, Bahn S, Dickerson FB. (2015) Immunomodulatory Effects of Probiotic Supplementation in Schizophrenia Patients: A Randomized, Placebo Controlled Trial. Biomarker Insights 10, 47-54. Tomoda A, Kamiya S, Suzuki H. (2015) Helicobacter pylori and Pathogenesis. BioMed Res Int 2015, 1-2. Turroni F, Peano C, Pass DA. et al. (2012) Diversity of Bifidobacteria within the Infant Gut Microbiota. PLoS ONE 7, 1-12. Turroni F, Serafini F Foroni, E, et al. (2013) Role of sortase-dependent pili of Bifidobacterium bifidum PRL2010 in modulating bacterium-host interactions. PNAS 110, 11151-11156. Turroni F, Taverniti V, Ruas-Madiedo P, et al. (2014) Bifidobacterium bifidum PRL2010 Modulates the Host Innate Immune Response. Appl Environ Microbiol 80, 730-740. Uraipan S, Hongpattarakere T. (2015) Antagonistic Characteristics Against Food-borne Pathogenic Bacteria of Lactic Acid Bacteria and Bifidobacteria Isolated from Feces of Healthy Thai Infants. Jundishapur J Microbiol 8, 1-9. Ulluwishewa D, Anderson RC, McNabb WC, Moughan PJ, Wells JM, Roy NC. (2011) Regulation of Tight Junction Permeability by Intestinal Bacteria and Dietary Components. J Nutr 141, 769-776. Vandenplas Y, Hert SGD. (2011) Probiotical-study group, Randomised clinical trial: the synbiotic food supplement Probiotical vs. placebo for acute gastroenteritis in children. Aliment Pharmacol Ther 34, 862-867. Vazquez-Gutierrez P, Lacroix C, Jaeggi T, Zeder C, Zimmerman MB, Chassard C. (2015) Bifidobacteria strains isolated from stools of iron deficient infants can efficiently sequester iron. BMC Microbiol 15, 1-10. Velema WA, Szymanski W, Feringa BL. (2014) Photopharmacology: Beyond Proof of Principle. J Am Chem Soc 136, 2178-2191. Ventura M, Turroni F, van Sinderen D. (2014) Diet-Microbe Interactions in the Gut: Effects on Human Health and Disease. Chapter 4: Bifidobacteria of the Human Gut: Our Special friends. Tuohy K, Rio DD. (Eds). Academic Press, Elsevier, USA. Vieira AT, Rocha VM, Tavares L, et al. (2015) Control of Klebsiella pneumoniae pulmonary infection and immunomodulation by oral treatment with the commensal probiotic Bifidobacterium longum 51A. Microb Infect; 1-10. doi:10.1016/j.micinf.2015.10.008
28
Vitaliti G, Pavone P, Guglielmo F, Spataro G, Falsaperla R. (2014) The immunomodulatory effect of probiotics beyond atopy: an update. J Asthma 51, 320-332. Vuyst LD, Leroy F. (2011) Cross-feeding between bifidobacteria and butyrate-producing colon bacteria explains bifidobacterial competitiveness, butyrate production, and gas production. Int J Food Microbiol 149, 7380. Wang H, Edwards MA, Falkinham JO, Pruden A. (2013) Probiotic Approach to Pathogen Control in Premise Plumbing Systems? A Review. Environ Sci Technol 47, 10117-10128. Weber E, Reynaud Q, Suy F, et al. (2015) Bifidobacterium Species Bacteremia: Risk Factors in Adults and Infants. Clin Infect Dis 61, 482-484. Wickramasinghe S, Pacheco AR, Lemay DG, Mills DA. (2015) Bifidobacteria grown on human milk oligosaccharides downregulate the expression of inflammation-related genes in Caco-2 cells. BMC Microbiology 15, 172-184. Wilson RW, Martin WJ, Wilkowske CJ, et al. (1972) Anaerobic bacteremia. Mayo Clin Proc 47, 639-646. Wollenberg A, Oranje A, Deleuran M, et al. (2016) ETFAD/EADV Eczema task force 2015 position paper on diagnosis and treatment of atopic dermatitis in adult and paediatric patients. J Eur Acad Dermatol Venereol 30, 729-747. Wu M-H, Pan T-M, Wu Y-J, Chang S-J, Chang M-S, Hu C-Y. (2010) Exopolysaccharide activities from probiotic bifidobacterium: Immunomodulatory effects (on J774A.1 macrophages) and antimicrobial properties. Int J Food Microbiol 144, 104-110. Xie N, Wang Y, Wang Q, Li F-r, Guo B. (2012) Lipoteichoic acid of Bifidobacterium in combination with 5fluorouracil inhibit tumor growth and relieve the immunosuppression. Bulletin du Cancer 99, E55-E63. Yang X, Hang X, Tan J, Yang H. (2015) Differences in acid tolerance between Bifidobacterium breveBB8 and its acid-resistant derivative B. breve BB8dpH, revealed by RNA-sequencing and physiological analysis. Anaerobe 33, 76-84. Yasmin A, Butt MS, Afzaal M, Baak MV, Nadeem MT, Shahid MZ. (2015) Prebiotics, gut microbiota and metabolic risks: Unveiling the relationship. J Funct Foods 17, 189-201. Yoon JS, Sohn W, Lee OY, et al. (2014) Effect of multispecies probiotics on irritable bowel syndrome: A randomized, double-blind, placebo-controlled trial. J Gastroenterol Hepatol 29, 52-59. Zanotti I, Turroni F, Piemontese A, et al. (2015) Evidence for cholesterol-lowering activity by Bifidobacterium bifidum PRL2010 through gut microbiota modulation. Appl Microbiol Biotechnol 99, 6813-6829.
29
Zhang CY, Chen GF, Wang YY, Xu Z, Wang
YG, Song XL. (2015) A3α-peptidoglycan extracted
from Bifidobacterium sp. could enhance immunological ability of Apostichopus japonicas. Aquacult Nutr 21, 679-689. Zihler A, Blay GL, Chassard C, Braegger CP, Lacroix C. (2014) Bifidobacterium thermophilum RBL67 Inhibits Salmonella enterica Serovar Typhimurium in an In vitro Intestinal Fermentation Model. J Food Nutr Disor S1003, 1-10.
30
Fig. 1. Schematic representation of protective mechanism of bifidobacteria displayed against pathogen invasion and in immunomodulation in host intestinal epithelium.
31
Table 1. Selective list of studies reporting different mechanisms of probiotic action of bifidobacteria documented through in vitro and in vivo experiments. Bacteria strains
Analysis
Mechanism of probiotic action
Ref.
Probiotic: (180 isolates analysed) B.
Intact cells, in vitro,
7 strains (B. longum L25, B. pseudocatenulatum
longum, B. pseudocatenulatum, B. bifidum,
Caco-2 cells, agar spot
C63, H34, B. catenulatum L21, L48, L51, B.
et al
B. adolescentis, B. catenulatum, B. dentium,
test
animalis E43) were chosen on basis of bile, acid,
2008
B. animalis
antibiotic resistance, enzymatic activities and
Pathogenic: E. coli wt 555, Salmonella
growth in culture medium.
enterica var. Typhimurium LT2UK1,
(1) Bile (45% of 180 isolates) and low pH (20% of
Staphylococcus aureus ATCC 1448, Listeria
total isolates, pH 3.5) tolerant
monocytogenes 1078, Bacillus cereus NCIB
(2) Strain dependent adhesion to Caco-2 intestinal
1771
cells lines, with L48 and L51 showing better
Delgado
adhesion compared L21. L25 and H43 showed lowest adhesion (3) All 7 strains inhibited pathogenic attachment to Caco-2 cell line (except Bacillus cereus) (4) E43 weakly inhibited only E. coli and L. monocytogenes.
Probiotic: B. breve UCC2003
Intact, in vivo (mice
(1) B. breve-EPS related (a) increased tolerance to
Fanning
Pathogenic: Citrobacter rodentium ICC180
model), flow
adverse GI tract conditions (b) increased
et al.
cytometry,
persistence in gut and (c) reduced colonisation of
2012
bioluminescent
pathogen (2) Reduction of levels of pro-
imaging, agglutination
inflammatory cytokines (3) IFN-γ–, TNF-α–, IL-
assay
12–positive T cells, and IL-12–positive B cells were significantly attenuated.
Probiotic: B. infantis BCRC 14602 Pathogenic and other strains: B. infantis BCRC 14602, B. adolescentis BCRC 14606,
Agar well diffusion,
(1) Antimicrobial activity of B. infantis BCRC
Cheikhy
cell free culture
14602 by production of proteinaceous bacteriocins
oussef et
supernatant (CFS),
(~ 3.0 kDa) against Salmonella, Shigella, E. coli,
al. 2009
32
B. bifidum BCRC 14615, B. longum BCRC
kinetic study of
Clostridium, Bacillus, Staphylococcus certain
14634, L. acidophilus SBDC, L. plantarum
bacteriocins
Lactobacillus and intragenus strains (except
BCRC 11697, isolate from Chinese Tibet
production, Bradford
producer strain) (2) Bacteriocin activity was
Koumiss, L. delbrueckii subsp. delbrueckii
assay, Tricine-SDS-
inactivated upon treatment with proteolytic
MLCC, L. paracasei subsp. paracasei
PAGE
enzymes, but not with catalase, α-amylase and
MLCC, L. casi W2 SBDC, L. fermentum
lipase (3) Bacteriocin exhibited high temperature
ATCC 9338, L. helveticus BCRC 14097, L.
stability up to 121 C for 15 min pH stability in the
lactis ATCC 11454, Pediococcus
range of 4-10
pentosaceus ATCC 33316, Salmonella sp. MLCC, Salmonella enteritidis CGMCC (B) 50041, Salmonella typhimurium ATCC 29631, Salmonella enterica ssp. enterica ATCC 13076, E. coli TG1 MLCC, E. coli GH5α MLCC, E. coli AS 1.543 MLCC, Staphylococcus aureus AS 1.72 MLCC, Bacillus subtilis MLCC, Bacillus cereus ATCC 14574, Clostridium butyricum MLCC, Shigella dysenteriae CGMCC (B) 51387, Sacchromyces cerevisiae ATCC 40075
Probiotic: B. thermophilum RBL67
In vitro intestinal
(1) Growth in intestinal environment (2) Inhibition
Zihler et
Pathogenic: Salmonella enterica subsp.
fermentation system
of pathogen colonisation (3) Bacteriocin-like
al. 2014
enterica serovar Typhimurium M557
with immobilized
inhibitory compounds (4) Rebalancing metabolic
faecal microbiota, plate
activity of gut microbiota post antibiotic therapy (5)
counts, FISH flow
Possible competition for nutrient and growth in co-
cytometry
cultures leading to competitive exclusion and antagonism by organic acid
Probiotic: B. bifidum CECT 7366
Intact cells and culture
(1) Resistance to adverse GI tract conditions (2)
Chenoll
Pathogenic: Helicobacter pylori NCTC
supernatant, in vitro
Pathogen inhibition upto 81.94% for supernatant
et al.
33
11637, NCTC 11638 and 6 human isolates
and in vivo (mice
and 94.77% for purified supernatant (3) Adhesion
model), agar diffusion
to intestinal mucus (4) Partial relief of pathogenic
assay, broth inhibition
damage to gastric tissues (5) Proteinaceous
assay
antagonistic compounds in supernatant exhibiting
2011
pathogen inhibition
Probiotic: B. bifidum NCIMB 30179, B.
Intact cells, culture
(1) B. breve NCIMB 30180 showed highest
Tejero-
breve NCIMB 30180, B. infantis NCIMB
supernatants, in vitro,
antimicrobial activity while B. infantis NCIMB
Sariñena
30181, B. longum NCIMB 30182 along with
agar spot method, agar
30181 showed almost no effect. (2) B. breve
et al.
strains belonging to Lactobacillus,
diffusion assay
showed highest inhibition of pathogen (3)
2012
Lactococcus, Streptococcus and Bacillus
Production of organic acids (mainly lactic and
genera
acetic acids) from glucose fermentation and
Pathogenic: Escherichia coli NCFB 1989,
consequent lowering of culture pH is the main
S. Typhimurium LT2, Staphylococcus
inhibitory mechanism. B. longum NCIMB 30182
aureus ssp. aureus NCTC 8532,
produced highest amount of lactic acid while B.
Enterococcus faecalis NCTC 775,
infantis NCIMB 30181 produced greater amount of
Clostridium difficile ATCC 43594
acetic acid (4) The observed antimicrobial activity is mainly genus specific with significant various among species.
Probiotic: B. longum BCRC 14634
Intact cells, EPS, LPS,
(1) BCRC 14634-EPS is a mild immune modulator
Pathogenic: E. coli BCRC 10239, S.
in vitro (murine
for macrophages inducing IL-10 secretion in
Typhimurium ATCC 14028, Staphylococcus
macrophage cell line),
J774A.1 cells. EPS also induced lower levels of
aureus ATCC 6538P, Bacillus subtilis
J774A.1 cells,
TNF-α secretion and slight changes in the
ATCC 21778, Pseudomonas aeruginosa
sandwich enzyme
morphology of J774A.1 cells (2) EPS pre-treatment
IFO 3898, Vibrio parhaemolyticus ATCC
immunoassay, phase-
prevented LPS-induced release of TNF-α from
17802, Bacillus cereus ATCC 10361
contrast microscope,
J774A.1 cells. EPS may act as an LPS blocker
Petroff-Hausser
(both have similar structure and might interact with
counting chamber
same receptors) (3) Pathogens were inhibited by 80 ppm EPS (4) EPS treatment did not lyse the pathogens, but rather impaired their division
34
Wu et al. 2010
Probiotic: B. longum BB536, B. animalis
In vitro, metabolites,
(1) SCFA metabolites butyrate and propionate
Asarat et
subsp. lactis BB12, B. lactis LAFTI B94,
LPS, Pour plate
produced in addition to lactate and acetate after
al. 2015
along with 3 strains belonging to
technique, HPLC
culturing in skim milk containing inulin, hi-maize
Lactobacillus genus
or β-glucan, with BB12 producing high amounts of
Pathogenic: LPS component from
SCFAs (2) SCFA show regulatory effects on
Escherichia coli O111:B4
cytokines released by LPS stimulated human PBMCs. The addition of 2 mM butyrate with LPS to PBMCs significantly inhibited release of proinflammatory cytokines (TNF-a, IL-12, TGF-b1 and IFN-g) (3) Butyrate and acetate induce higher production of anti-inflammatory cytokines (IL-4 and IL-10) than propionate at the same concentration
Probiotic: 2 isogenic strains of B.
Intact cells, EPS, in
(1) Mucoid EPS provides a higher resistance to GI
Hidalgo-
animalis ssp. lactis DSM10140 differing in
vitro, ex vivo,
conditions and increased adhesion to human
Cantabra
their EPS-producing phenotype
immunohistochemistry
enterocytes (epithelial intestinal cell line HT29) (2)
na et al.
Pathogenic: none
assay, Transmission
Cytokine profile of human PBMCs and ex
Electron Microscopy,
vivo colon tissues, suggests that EPS producing
Cryo-Scanning
strain could have a higher anti-inflammatory
Electron Microscopy,
activity
2015
human colonic mucosa organ culture
Probiotic: 313 Lactic acid bacteria and 17
Intact cells, cell free
(1) NIF7AN3, NIF7AN5, NIF7AN10 showed
Bifidobacterium (5 suitable strains identified
culture supernatant, in
strong adhesion to mucin. (2) The 5 strains studied
et al.
for study) B. longum subsp. longum
vitro, antibacterial
showed tolerance towards gastric acid and bile
2015
NIF3AN3, NIF7AN1, B. bifidum NIF7AN2,
activity assay, adhesion
salts. NIF7AN3, NIF7AN5, NIF7AN10 showed
NIF7AN5, NIF7AN10
assay using porcine
increase in number post treatment with acid and
Pathogenic: E. coli TISTR 780,
gastric mucin type III
bile. Possibly due to set of adaptive mechanism in
Staphylococcus aureus TISTR 1466,
response to various environmental stresses (3) All
35
Uraipan
Shigella sonnei, S. flexneri, Salmonella
showed high competitive exclusion of food borne
enterica ssp. enteric serovar Typhimurium
pathogens
SA2093, S. paratyphi A
Probiotic: B. bifidum (S17, PRL2010,
Intact cells, in vitro,
(1) B. bifidum showed highest adhesion to all tested
NCIMB41171), B. longum ssp. longum
adhesion assay,
intestinal epithelial cell (IEC) lines. (2)
et al.
(NCC2705, DJO10A, BBMN68, F8,
Northern blot, Southern
Proteinaceous cell surface component mediated
2012
JCM1217, JDM301, KACC 91563), B.
blot, Western blot
adhesion of B. bifidum (S17) to IECs. (3) B.
longum ssp. infantis (157 F, ATCC
bifidum-specific protein, BopA, mediated adhesion
15697), B. breve (UCC2003, DSM20213,
to IECs. B. bifidum, B. longum and B. infantis
ACS-071-V-Sch8b), B. adolescentis
strains overexpressing bopA showed enhanced
(ATCC15703 , L2-32), B. dentium (Bd1,
adhesion to IECs.
Gleinser
ATCC 27678, ATCC 27679, JCVIHMP022), B. animalis ssp. lactis (AD011, BB-12, BLC1, Bl-04, DSM10140, V9), B. pseudocatenulatum DSM20438, B. catenulatum DSM16992, B. angulatum DSM20098, B. gallicum DSM20093 Pathogenic: none
Probiotic: B. bifidum PRL2010 (with
Intact cells, in vitro and
(1) Host induced upregulation of genes cluster that
Turroni
Lactococcus lactis (NZ9000)-pil2 and L.
in vivo (mice model),
encode sortase-dependent pili (2) Initial adhesion of
et al.
lactis-pil3 clones)
Western blot, Cloning
B. bifidum cells to the enterocytes by extending
2013
Pathogenic: none
Pili-encoding B.
their pili toward the apical surface of the host
bifidum genes in L.
epithelial cells (3) L. lactis-pil3 clones induced
lactis, adhesion assay
higher TNF-α response in mouse model, compared
using Caco-2 cell line,
with non-piliated L. lactis-pil3 clones, indicating B.
adhesion inhibition
bifidum pili to influence mucosal immune
assay using anti-pili
responses.
antibodies, immunoassay human
36
macrophage-like cell line U937, AFM, light microscopy
Probiotic: B. breve CNCM I-4035
Intact cells, cell free
(1) Live CNCM I-4035 and CFS induces innate
Bermude
Pathogenic: Salmonella enterica
CFS, in vitro,
immune responses by activating TLR signalling
z-Brito et
serovar Typhi CECT 725
Immunoassay, Reverse
pathway (2) Intact CNCM I-4035 cells and CFS
al. 2013
transcriptase reaction
upregulated TLR9 gene transcription in presence of
and polymerase chain
S. typhi, with CFS having more potential (3)
reaction
CNCM I-4035 affects intestinal immune response, whereas its supernatant exerts anti-inflammatory effects via DCs (4) In presence of S. typhi, CFS decreased pro-inflammatory cytokines, chemokines and restored TGF-β levels in human intestinal DCs challenged while CNCM I-4035 induced proinflammatory TNF-α, IL-8 and RANTES, as well as anti-inflammatory IL-10
Probiotic: B. breve Yakult strain, B. bifidum
Intact cells, in vitro, in
(1) B. breve activates intestinal CD103+ DCs to
Jeon et
Yakult strain YIT10347, B. adolescentis
vivo (mice model),
produce IL-10 and IL-27 via TLR2/MyD88
al. 2012
ATCC15703, B. longum ATCC 15707 and
Flow cytometry,
signalling pathway thus inducing IL-10-producing
L. casei strain Shirota
intracellular cytokine
Tr1 cells in the large intestine (2) B. breve helps in
Pathogenic: none
staining, real time
maintaining homeostasis by inducing intestinal IL-
polymerase chain
10-producing Tr1 cells. (3) B. longum activated
reaction, T-cell-
DCs moderately induces IL-10-producing T cells
mediated colitis model,
(4) While B. adolescentis or B. bifidum did not
histopathological
induce IL-10 production from co-cultured CD4+ T
analysis
cells
37