Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Biomedicine & Pharmacotherapy 130 (2020) 110625

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Review

Immune boosting functional foods and their mechanisms: A critical


evaluation of probiotics and prebiotics
Tolulope Joshua Ashaolu a, b, *
a
Smart Agriculture Research and Application Team, Ton Duc Thang University, Ho Chi Minh City, Viet Nam
b
Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Viet Nam

A R T I C L E I N F O A B S T R A C T

Keywords: Comprehensive studies conducted on the link between the gut microbiome and immunity in recent decades have
Functional foods correspondingly led to ever increasing interests in functional foods, especially probiotics and prebiotics. Pro­
Probiotics biotics and prebiotics play crucial roles in managing the intestinal microbiota in order to improve host health,
Prebiotics
even though their influence on other body sites are being investigated. Different colonic bacteria metabolize
Microbiota
dietary prebiotics to produce beneficial metabolites, especially short chain fatty acids (SCFAs) that improve
Short chain fatty acids
luminal contents and intestinal performance, while positively affecting overall host physiology. Thus, this review
provides a general perspective of the immune system, the gut immune system and its microbiota. The review also
evaluates functional foods with critical but comprehensive perspectives into probiotics and prebiotics, their
immune boosting and mechanisms of action. It is recommended that further mechanistic and translational
studies are conducted to promote health, social life and also empower poverty-stricken communities.

1. Introduction though it is not their only pharmacologically important zone of the


body. This review provides an overview of the gut immune system and
There is a trendy demand for various food components that can their microbiota. The definitions and trending knowledge of both pro­
improve immune response. Functional foods can regulate the immune biotic and prebiotic action mechanisms with regards to immunity were
system through immune response enhancement or inhibition, which critically evaluated, while capping it all with prospective outlook, con­
provide host defenses against infection, and suppress allergies and cerns, challenges, and conclusive remarks.
inflammation [1]. The production of these food components such as
prebiotics and probiotics, is ever increasing with the intention of 2. A brief overview of the immune system
fighting chronic health problems like non-alcoholic fatty liver disease
(NAFLD), cancer, obesity, diabetes and cardiovascular diseases. Pres­ The immune system is broadly categorized into innate (non-specific)
ently, the most known functional foods are the probiotics and prebiotics or adaptive (specific). The specific immunity is linked to the immuno­
based on their health promoting properties. globulins (Ig), produced by B cells in response to antigenic attacks, such
Several investigations and in-depth studies have established pro­ as pathogens and allergens. The Igs are further divided into IgA, IgG and
biotics with both clear and unclear mechanisms, and thus it is suggested IgE classes - all produced via isotype switching upon activation. Of them
that they be incorporated into health management as alternative ther­ all, the IgAs are more relevant to the present review, as they can be
apy or complements to pharmaceutical agents, foods and lifestyle [2]. found in saliva, tears, sweat, breast milk and mucosal secretions. The
Prebiotics were known to be selectively degraded by host microorgan­ IgAs also help in controlling commensal bacteria at mucosal sites as well
isms to release beneficial metabolites. Prebiotics are not just enhancers as protecting the body from infection [1]. T-cells are another group of
of bifidobacteria and lactobacilli anymore, they have gained recognition specific/adaptive immune cells found in the small and large intestines.
regarding their physiological and systemic functions [3]. They have receptors that consist of α- and β-chains, and are further
Usually, the gut has been implicated in immune cells activity and classified based on their surface expression of CD4 or CD8 molecules.
overall immunity based on probiotics and prebiotics interventions, even CD4+ T cells are helper T cells (Th) assisting with other immune cells

* Correspondence to: Smart Agriculture Research and Application Team, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Viet Nam.
E-mail address: tolulopeashaolu@tdtu.edu.vn.

https://doi.org/10.1016/j.biopha.2020.110625
Received 3 July 2020; Received in revised form 2 August 2020; Accepted 5 August 2020
Available online 11 August 2020
0753-3322/© 2020 Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
T.J. Ashaolu Biomedicine & Pharmacotherapy 130 (2020) 110625

functions. These helper T cells assist with the production of multifarious prebiotics and probiotics meant to target obesity and metabolic syn­
cytokines such as interferon γ (IFN-γ), interleukin-4 (IL-4), and drome [17], as well as high blood pressure, oxidation, cellular damage
interleukin-7 (IL-17). and other diseases from sources/raw materials like vegetables, cereals,
diary and meat products [15,18,19]. Host health benefits which include
3. The gut immune system and its microbiota reduction of macular degeneration and prostate cancer risks were
associated with certain lutein and lycopene, while flavones, catechins
The gut is divided into the upper and the lower regions. The upper and anthocyanidins have been shown to possess antioxidative properties
region comprises of the mouth, pharynx, esophagus, stomach and duo­ by chelating free radicals and reducing the incidence of certain chronic
denum while the lower part mainly consists of the small and large in­ diseases [15]. Essential minerals and dietary elements were previously
testines. The gut microbiota are predominantly present in the lower included in the production of functional foods such as calcium, iron, zinc
region, and this makes the intestinal immune system quite important. and vitamins [20]; and now, among the most prominent and well
This immune system is composed of gut-associated lymphoid tissue researched functional foods are the probiotics and prebiotics, which
(GALT) and other cells, meant to protect the gut from several types of have the capacity of boosting consumers immune status [21]. Each of
antigens readily supplied from foods, commensal and pathogenic bac­ them has its own immunomodulatory and immune boosting benefits and
teria. Also, many and diverse intestinal cells are responsible for stimu­ they can beneficially interact in a “symbiotic” process to boost the im­
lating IgA isotypes production, which further buffer up the gut munity of the human host.
immunity. The helper T-cells 17 (Th17 cells) are also part of the gut
immune system that keep the mucosal barrier intact while stimulating 5. Probiotics
the intestinal epithelial cells to produce antimicrobial peptides [4].
The human gut is predominantly inoculated at birth, and the diverse The intention of consuming probiotics is to harness their benefits in a
microbiome develop following certain feeding and dietary patterns until symbiotic or commensal relationship with the intestinal microbes.
around 3–5 years of age, which then look similar to the adults [5]. These Initially, probiotics were defined as contributors to host’s intestinal
organisms interfere with the production of several cells and tissues microbial balance [2]. The modern consensus definition of probiotics is
within the gut in order to confer immunity on the system. Aging, diet, that they are living microorganisms, which when administered in
infection or indiscriminate use of medications may pose some challenges adequate amounts can confer health and well-being to the host [22].
to upset the make-up of the microbes as well as their fermentation end Thus, probiotics benefits are beyond the mediation of gut microbiota
products. In fact, a perturbation of the gut microbiota will lead to certain based on associated mechanisms. A paramount criterion for obtaining
acute and chronic disorders such as inflammatory bowel disease (IBD), effective probiotics is their ability to adhere to the gastrointestinal tract
irritable bowel syndrome (IBS) and obesity [6,7]. The fact that gut (intestinal mucosa and epithelial cells), in order to avoid being passed
fermentation and microbial functions largely depend on diet imply that out via gut motility, and thus multiply, colonize and modulate the im­
the consumption of functional foods like probiotics and prebiotics will mune system of the whole body by competitively suppressing pathogens
be a good dietary approach to improve the human gut microbiota for [21,23].
increased health benefits. Bacteria like Bifidobacteria possess special The commonest probiotics used in functional foods and other fer­
transport mechanisms, cellular and extracellular glycosidases, which mented products include Lactobacilli, Enterococci, Bifidobacteria and
actively digest low molecular weight prebiotic carbohydrates, while Leuconostoc spp. Other than these lactic acid bacteria group, yeasts such
Bacteroides break down high molecular weight prebiotic carbohydrates as Saccharomyces spp. are also used as probiotics [24]. In fact, the
[8,9]. However, Ruminococcus spp. will degrade resistant starch into low market for probiotics is ever increasing in size, and consumers’ demands
molecular weight dextrins, capable of being utilized by other colonic have risen spontaneously based on the immune-enhancing benefits of
organisms [121]. These gut microbes are continuously being engineered probiotics. They are already being sold as conventional foods, dietary
into marketable probiotics, which alongside prebiotics are considered supplements, medical foods and drugs in the United States, and are often
among functional foods demanding health claims and other authenti­ administrated in capsules, liquid or powder forms [24]. The immune
cations from relevant regulatory bodies. boosting effects and mechanisms of some probiotics are listed in Table 1.

4. Functional foods 5.1. Immune boosting and mechanisms of action

The definitions ascribed to functional foods are many, and include The action mechanisms of probiotics interfere with the gut epithelial
foods marketed with health claims; food sources with positive physio­ and immune cells composition and functions. Several insights into the
logical properties beyond their nutritional uses of providing essential mechanisms of probiotics are deduced from in vitro, animal, cell culture
nutrients; and natural substances that can be consumed daily meant to or ex vivo human studies, and not all mechanisms have been confirmed
regulate or affect a system of the body upon ingestion [10,11]. The in every probiotic strain or humans because a probiotic cell may exhibit
European Consensus provided the commonest and most recent defini­ several mechanisms simultaneously, further complexed by diverse fac­
tion, which states that ‘a food can be regarded as functional if it is tors [2]. There is a probiotic inherent immunomodulatory property
satisfactorily demonstrated to affect beneficially one or more target which varies from strain to strain. For instance, a probiotic strain
functions in the body, beyond adequate nutritional effects, in a way that significantly reduced sepsis in a study, but upon reformulation allowed
is relevant to either improved stage of health and well-being and/or enterocolitis in infants [43,44]. There is a colonization resistance
reduction of risk of disease’ [12]. A functional food can be natural, concept, which is possibly the sum outcome of the functioning of many
components-added, or components-eliminated foods via biotechnolog­ of the different immune-boosting mechanisms of probiotics action in
ical processes [13]. The ingredients in functional foods based on tech­ concert; it is described as the occupation of host tissues by the native gut
nological factors can help in preventing some diseases or improve microbiota in order to exclude potential pathogenic infections [2,45].
performance and well-being of consumers beyond its nutritional role, The validated and proposed action mechanisms of probiotics are
either whole or specific groups of the population defined by age or ge­ represented in Fig. 1. Probiotics are well known to boost the immunity of
netic differences [14,15]. humans by protecting against gastrointestinal pathogens, thus the
In fact, functional foods have been classified based on food type or mechanisms of action by which they exert their beneficial effects on the
active components added or naturally present in the foods i.e. fibre, host include secretion of antimicrobial substances, competitive exclu­
flavonoids, vitamins, minerals, fatty acids, carotenoids, and so on [16]. sion for adhesion sites and nutritional sources, enhancement of intesti­
Some functional foods are regarded as microbial medicines, i.e. nal barrier function, and immunomodulation [24].

2
T.J. Ashaolu Biomedicine & Pharmacotherapy 130 (2020) 110625

Table 1 Regarding antimicrobials production, probiotics produce diverse


The immune boosting effects and mechanisms of selected probiotics. substances such as organic acids, hydrogen peroxide and bacteriocins,
Probiotic organism Immune boosting Mechanisms Citation which are capable of inhibiting both Gram-positive and Gram-negative
functions bacteria. Several strains of Lactobacillus spp. secrete low molecular
Lactobacillus Detoxify toxins Binding via surface Jebali et al. weight bacteriocins (LMWB) and high molecular weight bacteriocins
plantarum structures [25] (class III), which are less than, and greater than 1000 Da, respectively.
MON03 The LMWB are antimicrobial in activity. The mechanism of action in­
Lactobacillus kefiri Detoxify toxins Adsorption and Taheur et al. volves the destruction of target pathogenic cells via pore formation or
KFLM3 biotransformation [26]
Saccharomyces
cell wall synthesis inhibition [46], as found with nisin, which binds the
cerevisiae precursor lipid II of spore-forming bacilli cell wall in order to prevent its
KFGY7 biosynthesis. This later forms an aggregation to invaginate the antimi­
Acetobacter crobial peptides for pore formation within the bacterial cell membrane,
syzygii KFGM1
thereby making the low molecular weight compounds to leak, weaken
Lactobacillus Detoxify toxins Binding via surface Ismail et al.
helveticus ATCC structures [27] the proton motive force, and consequently, cause cell death [47,48].
12046 Another mechanism under this category of antimicrobial substances
Bacillus Detoxify toxins Enzymic degradation Rao et al. secretion involves the production of acetic and lactic acids for acidifi­
licheniformis [28] cation in order to inhibit the growth of pathogens such as Salmonella spp.
CFR1
Saccharomyces Detoxify toxins Binding via surface Ismail et al.
The intracellular pH become reduced to acidify the cytoplasm, while the
cerevisiae HR structures [27] proton motive force become collapsed. The cumulation of these actions
125a leads to inhibition of nutrients transport and consequently, bactericidal
Lactococcus lactis Detoxify toxins Binding via surface Ismail et al. effects [49,50]. In addition, probiotic bacteria secrete microcins, which
JF 3102, structures [27]
act by binding iron siderophore receptors for cell entry or produce
Lactobacillus
plantarum NRRL harmful substances after gaining entrance into the cell. These processes
B-4496 lead to the inhibition of intracellular enzymes like ATP-synthase, DNA
Streptomyces cacaoi Detoxify toxins Enzymic degradation Harkai et al. gyrase and RNA polymerase, as well as their functions such as mRNA
subsp. Asoensis [29] translation, and then cause pathogen cell death.
K234,
Competitive exclusion mechanism involves adhesion sites and nu­
Streptomyces
luteogriseus trients competition between a probiotic organism and a pathogenic or­
K144, ganism. This enables the probiotic to inhibit the pathogen’s growth. The
Streptomyces probiotics may utilize their surface protein structures to achieve this like
rimosus K145
the case with mucins, or make their environment unfavorable for the
Stimulate dendritic Kikuchi et al.
Secrete IgA
cells to produce IL-6 [30] pathogens via secretion of antimicrobials including organic acids [24].
Stimulate dendritic Gut pathogens and LAB members have similar binding sites to the host
Sakai et al.
Produce IgA cells to produce TGF-
[31]
receptors [51,52], while microbe-associated molecular patterns
β (MAMPs) can bind the host pattern recognition receptors (PRRs) like
Improve natural
Stimulate secretion Takeda et al. toll-like receptors (TLRs) with the aim of out-competing the pathogens
killer (NK) cell
of IL-12 [32] [53]. Other than these means, pathogenic adhesion may be inhibited by
activity
Stimulate secretion Fujiwara probiotics via receptor disruption by probiotic enzymes, biosurfactants
Inhibit Th2 activity
of IL-12 et al. [33] secretion, and receptor analog production [24,54].
Stimulate activated T Kanzato
Several other Inhibit Th2 activity Intestinal epithelial barriers are well maintained by probiotic or­
cells death et al. [34]
Lactobacillus
Aoki-
ganisms to protect against certain intestinal hyper-acidity and diseases
strains Improve oral like obesity, inflammatory bowel diseases, and irritable bowel syn­
Induce Tregs Yoshida
tolerance
et al. [35] drome. Some studies demonstrated that probiotics increase gene
Weaken pro- expression associated with tight junction signaling, thus keeping intes­
inflammatory
tinal barrier integrity intact [24]. Examples include the modulation of
Reduce cytokines and Shimazu
inflammation chemokines by et al. [36] genes that encode adherence junction proteins like E-cadherin and
down-regulation of β-catenin, as well as their phosphorylation and the abundance of protein
TLR-signals kinase C (PKC) isoforms such as PKCδ by probiotic lactobacilli [55].
Chlebicz and
Binding via surface Probiotics do not only maintain the intestinal barriers, they also
Detoxify toxins Śliżewska
structures
[120]
potentiate their reparative process after damage. For instance, Staphy­
Reduce Kawashima lococcus thermophilus and Lactobacillus acidophilus markedly inhibited
Induce of IFN-β
inflammation et al. [37] enteroinvasive Escherichia coli from adherence to, invasion, mainte­
Up-regulate Nakamura nance and enhancement of HT29, and Caco-2 cells up-regulation of
Produce IgA
expression of pIgR et al. [38]
transepithelial resistance, cytoskeletal and tight junction protein phos­
Reduce A20 and
Modulate anti-viral
improve IRF-3, IFN-
Ishizuka phorylation [24]. Over-expression of mucin glycoproteins by probiotics
activity et al. [39]
Several other b, MxA, RNasel also confer immunity on host’s gut by keeping the intestinal barrier
Bifidobacterium
Protect from
Produce acetate and intact. The mucins are found in the epithelial mucus layer above the
strains improve intestinal Fukuda et al. intestinal epithelium and are associated with immune boosting. Mucin
enteropathogenic
defence with [40]
infection
epithelial cells
secretion may be enhanced by probiotics as a mechanistic improvement
Suppress Th2 Iwabuchi to intestinal barrier physiology in order to curb pathogenic adhesion and
Inhibit allergy
chemokines et al. [41] survival [56,57]. Some lactobacilli do increase mucin secretion in
Reduce Miyauchi human intestinal cell lines via adhesion to their monolayers, as found in
Inhibit IL-17
inflammation et al. [42]
the expression of MUC2, MUC3 and MUC5AC in HT29 cells [58].
The defensins, which are small peptides that are active against bac­
teria, fungi and viruses, and can also be produced by probiotic-
stimulated epithelial cells for barrier integrity, function and stability

3
T.J. Ashaolu Biomedicine & Pharmacotherapy 130 (2020) 110625

Fig. 1. Action mechanisms of probiotics.


Caption: Probiotics offer several means of boosting host immunity, ranging from influencing immune and other host cells directly, production of antimicrobial
components and cross-feeding other microbes, to a combination of many mechanisms of action.

[59]. These defensins include antimicrobial proteins (AMPs), which act Toll/interleukin-1 (IL-1) receptor (TIR), responsible for several adaptor
as enzymes by degrading cell wall structures or non-enzymatic rupturing molecules interactions like myeloid differentiation protein (MyD88)
of the pathogen’s cell membrane via cationic binding, and thus promote that leads to the activation of both mitogen-activated protein kinase
pathogen death with defensin pores incorporated in its membrane. (MAPK) and NF-κB [53], essential in cytokines and other immune cells
Probiotics are living organisms with immunomodulatory benefits, production.
and some of these immunomodulatory effects are produced when In addition to these, probiotics can induce anti-inflammatory cyto­
attenuated or dead, implying that probiotic effects are not necessarily kines such as IL-10 and TGF-β, by down-regulating the expression of pro-
obtained from live organisms [1]. The enhancement of the antibodies inflammatory cytokines, such as TNF and IFN-γ in order to enhance
such as IgG and IgA by probiotics like LAB and bifidobacteria may be mucosal immune responses [64,66]. However, certain probiotic bacteria
useful in preventing invasion of the mucosal barriers as LAB can act on alter cytokine production via modulation of the signals transduced by
intestinal dendritic and epithelial cells or improve the expression of the cells. They may inhibit I-κB via ubiquitination by blockade of the
polymeric immunoglobulin receptor (pIgR) [30,31,60]. Some LAB proteasome activity or interfere with RelA gene localization in a
members can use TLR2 to stimulate IL-6 or TGF-β production from γ-dependent signal cascade, which is stimulated by proliferative
dendritic cells [38]. Lactic acid bacteria also stimulate IL-12 production peroxisome [67,68]. Upon activation by probiotic bacteria, Th0 are
mediated by polysaccharides in order to augment NK activity, which differentiated into Tregs by DCs to inhibit Th1, Th2 and Th17 responsive
then enhances host defense [1]. actions capable of inflammation [24].
Some probiotics also demonstrate their ability to increase antibody Moreover, cell-surface structures like capsule and fimbriae are used
production for the purpose of improving defences against pathogenic by certain probiotic organisms as mechanistic drivers for their immune
invasions and thereby serve as adjuvant vaccines [61,62]. The amounts boosting activities [2].
of anti-inflammatory cytokines such as tumor necrotic factor (TNF) can Indeed, some studies also indicate that the immunoregulatory effects
be increased by some probiotic strains, which then aid with colon cancer of probiotics may be beneficial in NAFLD treatment as they modulate the
and colitis inhibition or therapy [63]. Probiotic organisms do confer intestinal microbiota; improve epithelial barrier function and strengthen
immune boosting benefits to the gut while providing some counterbal­ the intestinal wall by decreasing its permeability; reduce bacterial
ance to inflammation caused by an invading foreign body [64]. They act translocation and endotoxemia; improve intestinal inflammation; and
with monocytes and lymphocytes, which play important roles in innate reduce oxidative and inflammatory liver damage [69]. Although pro­
and adaptive immunity, as well as epithelial and dendritic cells. biotics and prebiotics have been proposed in the treatment and pre­
Dendritic cells and intestinal epithelial cells (IECs) use their pattern vention of patients with obesity-related NAFLD, their therapeutic use is
recognition receptors (PPRs) such as the TLRs to communicate with not supported by high-quality clinical studies, even though an effective
probiotics and other gut microbes, which then play significant roles in short-term treatment for NAFLD, as part of a multidisciplinary approach,
immune regulation. The TLRs are transmembrane proteins located has been claimed [69]. Being the intestinal mucosal barrier of immune
within endocytic vesicular membranes and other intracellular cell nature, the microbiota may be viewed as part of this system because of
membranes [65]. The TLRs cytoplasm have conservative the mutual influence occurring between the host and the luminal

4
T.J. Ashaolu Biomedicine & Pharmacotherapy 130 (2020) 110625

microorganisms. Alteration of the mucosal barrier function with oligosaccharides [e.g. fructooligosaccharides (FOS), gal­
accompanying increased permeability and/or bacterial translocation actooligosaccharides (GOS), inulin and xylooligosaccharides (XOS)],
has been linked to many conditions [70]. Further investigations into which cannot be digested by enzymic activities, rather used up by gut
several other factors that may relieve or aid a damaged mucosal barrier microbes like Saccharomyces, bifidobacteria, eubacteria and lactobacilli;
healing process for intestinal and systemic immune maintenance, are certain compounds like proteins or peptides, unsaturated fatty acids,
required. flavonoids, micronutrients and macronutrients also demonstrated pre­
biotic properties [73]. Critical observations show that prebiotics im­
6. Prebiotics mune boosting effects are associated with probiotics stimulation and
their metabolites produced such as short chain and branched chain fatty
From the onset, prebiotics were referred to indigestible food in­ acids; and may also be derived from other uncommon microbial taxa
gredients capable of providing health benefits to the host through se­ such as faecalibacteria, eubacteria or roseburia [21,74].
lective stimulation of the activity of colonic bacteria [71]. Subsequent Speculated ecosystemic metabolism of polysaccharides have been
comprehension of the gut microbiota and technological advancements repeatedly presented but their modus operandi within the gastrointes­
such as metagenomics, nutrigenetics and metatranscriptomics have tinal tract is not clear yet [75]. Functional redundancy exists despite the
further elucidated on the intricacies and delicateness of this definition. presence of diverse taxa in gut microbiota. The ecological functions of
For instance, the International Scientific Association of Probiotics and bacteria are specific in different individuals [76], and less vivid
Prebiotics (ISAPP) in London (United Kingdom), has defined a prebiotic comprehension of the functional ecology of the gut microbiota poses a
as “a substrate that is selectively utilized by host microorganisms challenge of describing prebiotics action mechanisms [2]. However,
conferring a health benefit”. This occurred in 2016, and is the most probable mechanisms of prebiotics leading to human health benefits can
recent definition [72]. be postulated as shown in Fig. 2.
Therefore, prebiotics may not necessarily be carbohydrates, and may
also be applied to other atypical “substrates” in the human body apart
6.1. Immune boosting and mechanisms of action
from the gut such as the skin or vaginal tract, capable of being colonized
by the probiotics. This process have to follow stringent microbial
Dietary prebiotics are usually those indigestible fibrous compounds
selectivity. Other than the established prebiotics that are
passed through the upper gut in order to stimulate probiotics growth. A

Fig. 2. Action mechanisms of prebiotics.


Caption: Upon selective utilization of prebiotics in the gut, the microbiota grow to exert various immune boosting functions at species and strain multi-levels. The
bacterial cell wall and other biomass increase immunomodulation and fecal bulking for healthy bowel movement. Metabolites like organic acids reduce luminal pH,
harmful to pathogens but aid solubility and absorbability of minerals such as calcium, as well as positively regulate hormones and epithelial integrity. GLP1:
glucagon like peptide1; M cell: microfold cell; NK: natural killer; PYY: peptide YY; TGFβ: transforming growth factor-β; TH1, TH2: type 1 T helper, type 2 T helper;
Treg: regulatory T; ZO1: zonula occludens 1. Adapted from Sanders et al. [2].

5
T.J. Ashaolu Biomedicine & Pharmacotherapy 130 (2020) 110625

simple case study is indigestible FOS, made of 1-kestose (GF2) and is a strong relationship between SCFAs and G-protein coupled receptors
nystose (GF3), metabolized specifically by bifidobacteria and bacter­ (GPRs) because SCFAs activate GPR41 and GPR43 on intestinal
oides amongst others, as energy sources for replication in the gastroin­ epithelial cells in order to stimulate immune responses via chemokines
testinal tract [1]. This process alters the gut ecosystem based on series of and cytokines swift secretion, reactions already confirmed to help in
the bacterial dynamics and movement of their metabolites, which then tissue inflammation and immunity in mice studies [81].
stimulate IgA secretion, an important intestinal immune system In addition to these mechanisms, certain fatty acid receptors
component [77]. In the same process, the larger GF3 molecule can in­ (FFARs), including FFAR2 and FFAR3 may react with gut fermented
crease intestinal bacteroides in order to stimulate IgA production [1,77]. metabolites like SCFAs for lipolysis enhancement and incretin glucagon-
Prebiotics may also interfere with type 2 T-helper reactions in order like peptide-1 (GLP-1) secretion [82]. Several tissues possess these re­
to alleviate allergy according to some studies in infants. Arslanoglu et al. ceptors indicative of possible action mechanism linkage between
[78] found out that feeding healthy term infants who are at atopy risk immunomodulation and prebiotic fermentation.
with prebiotic-supplemented hypoallergenic formula for 6 months could Regarding appetite regulation, many mechanisms have been impli­
lead to a significant reduction in allergies prevalence even after 5 years cated, such as the interaction between SCFAs and colonic cells to pro­
of the feeding programme. In a similar study, there were reduced duce anorexigenic hormones like peptide YY (PYY) and GLP-1; or
wheezing, atopic dermatitis and urticaria in infants fed with long-chain metabolism escape of SCFAs through the colonic epithelial cells to the
FOS and GOS formula, administered in a double-blind, randomized, liver via the hepatic portal vein in order to create a satiety signal stim­
placebo-controlled trial as compared to infants fed with non-prebiotic ulated by propionate-mediated gluconeogenesis [83]. Leptin induce­
formula [79,80]. In these studies, the exact mechanisms remain unclear. ment can also result from fatty acid receptors interaction with SCFAs in
SCFAs produced via prebiotic degradation by the intestinal micro­ the blood stream, or else cross the blood–brain barrier and enter the
biota also act in defense of the immune system. Tarantino & Finelli [69] hypothalamus to promote anorectic signals [84]. A detailed mechanistic
reported that prebiotic treatment had some varied impacts among illustration of SCFAs production and their consequential pathways upon
NAFLD and obese individuals who had undergone gastric bypass surgery prebiotics fermentation and bacterial cross-feeding is shown in Fig. 3.
with intriguing hypothesis based on molecular and biochemical mech­ From another outlook, model systems have been used to make in vitro
anisms. Processes like minor conversion of indigestible carbohydrates probe into prebiotics defence against pathogens by the inhibition of
into SCFAs or decreased local alcohol production may reduce the risk of pathogen growth via decreased intestinal pH upon organic acids pro­
worsening NAFLD by ameliorating insulin resistance. However, it is duction [85,86].
currently still unclear as to which bacterial groups play a role in the Age-related immune boosting efficacy of prebiotics was investigated
development of obesity in humans [69]. The production of acetic acids in a human study for ten weeks, whereby some elderly individuals were
as bifidobacteria metabolites in the intestinal epithelial cells can prevent fed daily with GOS, and it was found out that their immune cells func­
intestinal enterohemorrhagic Escherichia coli O157 infection [40]. There tions in the likes of natural killer cells were more proactive, and their

Fig. 3. SCFAs production and their consequential pathways upon prebiotics fermentation and bacterial cross-feeding.
Caption: Organic acids (e.g. CO2 and H2) and short chain fatty acids (SCFAs: lactate, acetate, propionate and butyrate) are the major metabolites produced by gut
microbial fermentation of prebiotics. Acetate is the largest amount of SCFAs produced, and often released by Methanobrevibacter smithii and Blautia hydrogenotrophica
via reductive acetogenesis or pyruvate acetyl-CoA pathways. Firmicutes are involved in butyrate production from two acetate-derived acetyl-CoA molecules, con­
verted via the intermediates to butyryl CoA. This subsequently synthesize butyrate via butyryl CoA: acetate CoA transferase or butyrate kinase pathways. Propionate
has multi-production pathways such as the acrylate pathway, where Firmicutes reduces pyruvate to lactate by the enzymic action of lactoyl-CoA dehydratase to
produce propionate. In succinate pathway, propionate is produced by Firmicutes and Bacteriodetes through succinate decarboxylation. The third pathway involves
the conversion of fucose and rhamnose from propionaldehyde to propionyl-CoA by CoA-dependent propionaldehyde dehydrogenase. Other metabolites such as
lactate are produced in fewer amounts. Then, all these SCFAs modulate transepithelial fluid transport for membrane integrity maintenance, mucosa improvement and
disease relief. Adapted from Louis et al. [119] and Neri-Numa and Pastore [74].

6
T.J. Ashaolu Biomedicine & Pharmacotherapy 130 (2020) 110625

phagocytic activities increased [87]. Once there is a probiotic compe­ some studies whereby young adolescents consumed a mix of FOS and
tition for prebiotic nutrients, stabilized by the growing population of inulin or GOS, absorption and mineralization of calcium into bone was
commensal microbes, lesser available nutrients will be left for patho­ significantly increased, thus reducing the risks associated with osteo­
genic organisms, and thus force them into redundancy, suppression and porosis in later life [89,90]. Although previous studies in animal models
inhibition. supported these research [91], but clinically validated long-term studies
Moreover, on the basis of mineral nutrients route, absorption occur are not available [2].
in the small intestine via active transport mechanisms. The drop in Finally, prebiotics play important roles in the metabolic processes
luminal pH due to the production of SCFAs among other acidic metab­ associated with immunomodulation. Gut barrier dysfunction allows
olites can lead to increased calcium solubility, readily available for movement of various inflammatory mediators like the bacterial lipo­
passive uptake and consequent immune boosting via immune cells polysaccharide (LPS) from the gut into the blood stream, a process
activation or bone strength enhancement among other benefits. How­ known as metabolic endotoxaemia, and shown to be an important factor
ever, the solubility of calcium salts present in many food supplements in the development of obesity and diabetes in mice models [92]. The gut
are pH-dependent and are less available. So, calcium solubility may barrier integrity associated with immunity, can be enhanced when
indeed increase with increased pH based on the starting pH [88]. In prebiotics and subsequent SCFAs release act. GOS has proved this point

Table 2
Prebiotics influences on immune boosting recently demonstrated in in vitro, animal and human studies.
Prebiotics source Experimental design In vitro, Analysed metabolites/bacteria Resultant influences Citation
animal or
human
studies

Orange juice enriched with Ten healthy children and Human Acetate, butyrate, lactobacilli and Up-regulation of insulin, glucose, Lima et al.
hesperidin and naringin women in a controlled non- bifidobacteria triglycerides and total cholesterol. [100]
randomized clinical study for
sixty days.
Galactooligosaccharides (GOS) Caco-2, colon and gingival In vitro Lactate, propionate, butyrate, Regulation and modulation of Perdijk
and sialyllactose epithelial cells lines. bifidobacteria and Bacteroides. genes associated with cell cycle et al. [101]
stages and cyclin-dependent
kinases.
Blackcurrant enriched with Sixten Sprague-Dawley rats fed Animal Butyrate and Bacteroides- Influences on food intake and body Paturi et al.
prebiotic polyphenols for 6 weeks, randomly divided Prevotella-Prophyromonas group weight and disrupted large intestine [102]
into 8 groups. increased while bifidobacteria and biomarkers.
C. perfringens decreased.
Grapeseed flour enriched with Ten Mice in random groups of Animal Propionic acid. Adipogenesis, permeable intestine, Cho et al.
prebiotic polyphenols and kefir- six fed for 9 weeks. systemic inflammation, glucose and [103]
derived lactic acid bacteria insulin resistance.
Aronia juice enriched with A Simulator of the human In vitro Increased propionate, butyrate, Pro-inflammatory markers such as Wu et al.
prebiotic polyphenols intestinal microbial ecosystem, akkermansia and Firmicutes. interleukin-8 and monocytes were [104]
Caco-2 and endothelial cells Decreased bifidobacteria and down-regulated.
Coculture were used for 2 acetate.
weeks.
MSPrebiotic® Eighty-four mid-age and elderly Human Butyrate, bifidobacteria and Lowered insulin and glucose levels. Alfa et al.
adults consumed the prebiotic in proteobacteria [105]
a prospective, randomized,
blinded, placebo-controlled trial
for 12 weeks.
Inulin Caco-2 cells plus fecal batch In vitro Acetate, propionate, butyrate, Improved and enhanced gut barrier Van Den
fermentations. bifidobacteria, actinobacteria and and immune function (nuclear Abbeele
members of Bacteroides and factor kappa b, transepithelial et al. [106]
Lentisphaerae. electrical resistance, and
interleukins-10 and -6.
Hesperidin Eighteen Lewis rats in random Animal Coated caecal bacteria coated with Immune modulation and up- Estruel-
groups of three fed for 4 weeks. increased lactobacilli, enterococci regulation of Immunoglobulin A Amades
and staphylococci. Decreased and lymphocytes. et al. [107]
Clostridium and Eubacterium spp.
B-GOS® Forty-one children with autism Human Veillonellaceae, Lachnospiraceae, Urine and faecal metabolites Grimaldi
spectrum disorder were Faecalibacterium, Bacteroides and changed, reduced bowel movement et al. [108]
randomly divided into 2 groups Bifidobacterium spp. and abdominal pain, and improved
and fed for 6 weeks. anti-social behavior.
Vivinal-GOS® Eighteen crayfish in random Animal Fewer lactic acid bacteria, and Increased innate immunity, stress Nedaei et al.
groups of twelve fed for 97 days. higher number of intestinal resistance and digestion. [109]
heterotrophic bacteria.
Blueberry polyphenols extract Ten mice in random groups of Animal Deferribacteres, Actinobacteria, Modulated leptin levels, reduced Jiao et al.
six fed for 12 weeks. Proteobacteria, Bifidobacterium, cholesterols and body weight. [110]
Desulfovibrio, Prevotella, Improved lipid metabolism by
Adlercreutzia, Helicobacter and modulation of lipid genes.
Flexispira spp.
scGOS/lcFOS plus fermented milk Twenty-one Lewis rats in Animal Streptococcus thermophilus strain Reduced diarrhea symptoms and Rigo-
random groups of three fed with 065 and Bifidobacterium breve strain inhibited rotavirus replication. Adrover
a mixture of FOS,GOS and C50 et al. [111]
fermented Milk for 21 days.
Orafti® inulin-type fructans Two hundred and nine healthy Human Bifidobacteria Had immune boosting effects and Soldi et al.
children, randomized, placebo- reduced antibiotic-induced [112]
controlled, double- blind trial, intestinal microbiota perturbations
fed for 24 weeks.

7
T.J. Ashaolu Biomedicine & Pharmacotherapy 130 (2020) 110625

in an in vivo study as it improved the barrier function [93]. There is a health-promoting foods. The consumption of probiotics such as lacto­
common ground for the acceptance of prebiotics metabolic effects that bacilli and bidobacteria may proffer an option to antibiotics for pre­
they are positive regulators of glucose balance, inflammatory response vention and treatment of microbial infections based on certain
and blood lipid profile in humans following similar hypotheses mechanisms including antimicrobial toxins production, maintenance of
described earlier, even though the results have never been the same intestinal barrier integrity, competing for nutrients and adhesion, as
[94–96]. Mice models indicated that glycemic index could be improved well as modulation of the immune system. This is not dissimilar to ef­
by inulin because of its direct inhibitory effect on the intestinal iso­ fects and mechanisms of prebiotics, which are entangled in synergistic
maltase–sucrase enzyme complex [97]. In epithelial cell lines, the in processes with the beneficial probiotic organisms. Thus, better
vitro fermentation of GOS induced tight junction proteins expression and comprehension of the mechanisms involved are required, as well as
decreased transepithelial flux [98,99], a mechanism not yet clear in vivo extensive human studies to validate excellent potent probiotic species/
and a bit daunting as a task. Table 2 shows prebiotics influences on strains and effective amount of doses meant for particular diseases.
immune boosting recently demonstrated in in vitro, animal and human Generally, the results of these interventions may be valuable for target
studies. populations. Researchers are bestowed with the responsibility of
considering the importance of their findings to consumers, as well as
7. Further considerations their fundamentality to overall conclusions on products’ efficacy.
Moreover, more investigations and translational research on probiotics
Most consumers embrace the idea of health promoting foods and and prebiotics meant to help developing countries’ consumers are
food ingredients. Inclusion of probiotics and prebiotics in their foods or recommended.
direct consumption of these functional food products as dietary sup­
plements have their own challenges too. Upon consumption, probiotic Author contributions
organisms should be stable and remain viable when passed through the
digestive tract in order to perform the expected activity and confer the TJA conceptualized, researched, compiled and analyzed data, as well
needed benefits. And once in the colon, they should be able to metab­ as drafted and revised the manuscript.
olize their counterpart prebiotics for synergistic and robust beneficial
outcomes.
Declaration of Competing Interest
It appears that safety is the most paramount measure required when
developing probiotic microorganisms, but interestingly most of them are
The authors report no declarations of interest.
safe [113]. Newer biotechnological developments and better compre­
hension of the organisms’ action mechanisms have led to engineering
newer strains with much better strengths than their progenitors, thus Acknowledgments
manufacturing an entirely different probiotics [114]. Nevertheless,
presuming that the newer species and strains of probiotics are equally The work was self-supported. The author also expresses his profound
safe as their progenitors will be preposterous. In fact, at the molecular gratitude to the Smart Agriculture Research and Application Team,
level, the genes may be mutated and may be unsafe. Therefore, how safe Faculty of Applied Sciences, Ton Duc Thang University, HCMC,
and efficacious the novel probiotic organisms will be before commer­ Vietnam.
cialization is a question to be meticulously pondered on.
Another consideration is the neutral, antagonistic or synergistic ef­ References
fects of probiotic bacteria on several other substances and products like
[1] S. Hachimura, M. Totsuka, A. Hosono, Immunomodulation by food: impact on gut
the antibiotics. The concern that some probiotics may transfer resistance immunity and immune cell function, Biosci. Biotechnol. Biochem. 82 (4) (2018)
genes to pathogenic bacteria require strain resistance testing to a wide 584–599.
variety of common classes of antibiotics such as tetracyclines, quino­ [2] M.E. Sanders, D.J. Merenstein, G. Reid, G.R. Gibson, R.A. Rastall, Probiotics and
prebiotics in intestinal health and disease: from biology to the clinic, Nat. Rev.
lones and macrolides. The transmission of drug resistance genes to the
Gastroenterol. Hepatol. (2019) 1–12.
pathogenic bacteria should also be ascertained [24]. Next generation [3] S.L. Collins, A. McMillan, S. Seney, C. van der Veer, R. Kort, M.W. Sumarah,
“omics”-based technologies will ease the necessary tests needed to do G. Reid, Promising prebiotic candidate established by evaluation of lactitol,
this, and thus help ascertain how safe and quality the identified pro­ lactulose, raffinose, and oligofructose for maintenance of a lactobacillus-
dominated vaginal microbiota, Appl. Environ. Microbiol. 84 (5) (2018)
biotic strain is. e02200–17.
Prebiotics are largely carbohydrates but some are non-carbohydrates [4] K. Atarashi, Y. Umesaki, K. Honda, Microbiotal influence on T cell subset
and serve as novel food ingredients with health promoting functions. development, in: Seminars in Immunology, vol. 23, Academic Press, 2011,
pp. 146–153. April, No. 2.
How these ingredients interact with the gut microorganisms or [5] J.M. Rodríguez, K. Murphy, C. Stanton, R.P. Ross, O.I. Kober, N. Juge, et al., The
consumed probiotics, with their widely diversified structures are how­ composition of the gut microbiota throughout life, with an emphasis on early life,
ever yet to be completely comprehended [74]. Microb. Ecol. Health Dis. 26 (1) (2015) 26050.
[6] N. Yoshida, T. Yamashita, K.I. Hirata, Gut microbiome and cardiovascular
Finally, the availability of probiotic organisms in less developed re­ diseases, Diseases 6 (3) (2018) 56.
gions of the world cannot be compared with the developed regions. The [7] G.K. John, L. Wang, J. Nanavati, C. Twose, R. Singh, G. Mullin, Dietary alteration
potentials of beneficial organisms and local food sources to improve of the gut microbiome and its impact on weight and fat mass: a systematic review
and meta-analysis, Genes 9 (3) (2018) 167.
consumers overall well-being in developing countries cannot be over­ [8] B.R. Hamaker, Y.E. Tuncil, A perspective on the complexity of dietary fiber
emphasized just as their affordability and accessibility to many con­ structures and their potential effect on the gut microbiota, J. Mol. Biol. 426 (23)
sumers in developed nations should not be undermined. However, in a (2014) 3838–3850.
[9] A. Rivière, M. Selak, A. Geirnaert, P. Van den Abbeele, L. De Vuyst,
recent programme, probiotics L. rhamnosus (GR-1 or Yoba 2012) and
Complementary mechanisms for degradation of inulin-type fructans and
plus S. thermophilus C106 were introduced in sachets for the production arabinoxylan oligosaccharides among bifidobacterial strains suggest bacterial
of multifarious fermented foods such as yoghurt, millet, cereals and cooperation, Appl. Environ. Microbiol. 84 (9) (2018) e02893–17.
juices, which help to bolster consumers’ health and improve the social [10] G. Rivera, V. Bocanegra-García, A. Monge, Traditional plants as source of
functional foods: a review Plantas tradicionales como fuente de alimentos
status and well-being of poverty-stricken societies [2,115]. funcionales: una revisión, CyTA–J. Food 8 (2) (2010) 159–167.
[11] A. Maroyi, Utilization of Bridelia mollis as herbal medicine, nutraceutical and
8. Conclusions functional food in southern Africa: a review, Trop. J. Pharm. Res. 18 (1) (2019)
203–209.
[12] M.B. Roberfroid, A European consensus of scientific concepts of functional foods,
Functional foods such as probiotics and prebiotics are the future of Nutrition (Burbank, Los Angeles County, Calif.) 16 (7–8) (2000) 689–691.

8
T.J. Ashaolu Biomedicine & Pharmacotherapy 130 (2020) 110625

[13] M.T. Ukeyima, V.N. Enujiugha, T.A. Sanni, Current applications of probiotic [39] T. Ishizuka, P. Kanmani, H. Kobayashi, A. Miyazaki, J. Soma, Y. Suda, et al.,
foods in Africa, Afr. J. Biotechnol. 9 (4) (2010). Immunobiotic bifidobacteria strains modulate rotavirus immune response in
[14] I.S. Arvanitoyannis, M. Van Houwelingen-Koukaliaroglou, Functional foods: a porcine intestinal epitheliocytes via pattern recognition receptor signaling, PLoS
survey of health claims, pros and cons, and current legislation, Crit. Rev. Food Sci. One 11 (3) (2016).
Nutr. 45 (5) (2005) 385–404. [40] S. Fukuda, H. Toh, K. Hase, K. Oshima, Y. Nakanishi, K. Yoshimura, et al.,
[15] M.A. Silva, T.G. Albuquerque, R.C. Alves, M.B.P. Oliveira, H.S. Costa, Melon Bifidobacteria can protect from enteropathogenic infection through production of
(Cucumis melo L.) by-products: potential food ingredients for novel functional acetate, Nature 469 (7331) (2011) 543–547.
foods? Trends Food Sci. Technol. 98 (2020) 181–189. [41] N. Iwabuchi, N. Takahashi, J.Z. Xiao, S. Yonezawa, T. Yaeshima, K. Iwatsuki,
[16] T.J. Ashaolu, A review on selection of fermentative microorganisms for functional S. Hachimura, Suppressive effects of Bifidobacterium longum on the production
foods and beverages: the production and future perspectives, Int. J. Food Sci. of Th2-attracting chemokines induced with T cell–antigen-presenting cell
Technol. 54 (8) (2019) 2511–2519. interactions, FEMS Immunol. Med. Microbiol. 55 (3) (2009) 324–334.
[17] M. Green, K. Arora, S. Prakash, Microbial medicine: prebiotic and probiotic [42] E. Miyauchi, T. Ogita, J. Miyamoto, S. Kawamoto, H. Morita, H. Ohno, et al.,
functional foods to target obesity and metabolic syndrome, Int. J. Mol. Sci. 21 (8) Bifidobacterium longum alleviates dextran sulfate sodium-induced colitis by
(2020) 2890. suppressing IL-17A response: involvement of intestinal epithelial costimulatory
[18] T.J. Ashaolu, Antioxidative peptides derived from plants for human nutrition: molecules, PLoS One 8 (11) (2013).
their production, mechanisms and applications, Eur. Food Res. Technol. (2020) [43] K. Costeloe, P. Hardy, E. Juszczak, M. Wilks, M.R. Millar, Bifidobacterium breve
1–13. BBG-001 in very preterm infants: a randomised controlled phase 3 trial, Lancet
[19] A.K. Das, P.K. Nanda, P. Madane, S. Biswas, A. Das, W. Zhang, J.M. Lorenzo, 387 (10019) (2016) 649–660.
A comprehensive review on antioxidant dietary fibre enriched meat-based [44] P. Panigrahi, S. Parida, N.C. Nanda, R. Satpathy, L. Pradhan, D.S. Chandel, et al.,
functional foods, Trends Food Sci. Technol. 99 (2020) 323–326. A randomized synbiotic trial to prevent sepsis among infants in rural India,
[20] C.J. Ziemer, G.R. Gibson, An overview of probiotics, prebiotics and synbiotics in Nature 548 (7668) (2017) 407–412.
the functional food concept: perspectives and future strategies, Int. Dairy J. 8 [45] L. Chiu, T. Bazin, M.E. Truchetet, T. Schaeverbeke, L. Delhaes, T. Pradeu,
(5–6) (1998) 473–479. Protective microbiota: from localized to long-reaching co-immunity, Front.
[21] J.T. Guimarães, C.F. Balthazar, R. Silva, R.S. Rocha, J.S. Graça, E.A. Esmerino, et Immunol. 8 (2017) 1678.
al., Impact of probiotics and prebiotics on food texture, Curr. Opin. Food Sci. 33 [46] M. Hassan, M. Kjos, I.F. Nes, D.B. Diep, F. Lotfipour, Natural antimicrobial
(2020) 38–44. peptides from bacteria: characteristics and potential applications to fight against
[22] C. Hill, F. Guarner, G. Reid, G.R. Gibson, D.J. Merenstein, B. Pot, et al., Expert antibiotic resistance, J. Appl. Microbiol. 113 (4) (2012) 723–736.
consensus document: The International Scientific Association for probiotics and [47] T.J. Montville, M.E.C. Bruno, Evidence that dissipation of proton motive force is a
prebiotics consensus statement on the scope and appropriate use of the term common mechanism of action for bacteriocins and other antimicrobial proteins,
probiotic, Nat. Rev. Gastroenterol. Hepatol. 11 (8) (2014) 506–514. Int. J. Food Microbiol. 24 (1–2) (1994) 53–74.
[23] J.D. Schepper, R. Irwin, J. Kang, K. Dagenais, T. Lemon, A. Shinouskis, et al., [48] G. Bierbaum, H.G. Sahl, Lantibiotics: mode of action, biosynthesis and
Probiotics in gut-bone signaling. Understanding the Gut-Bone Signaling Axis, bioengineering, Current Pharm. Biotechnol. 10 (1) (2009) 2–18.
Springer, Cham, 2017, pp. 225–247. [49] J.B. Russell, F. Diez-Gonzalez, The effects of fermentation acids on bacterial
[24] M.L.Y. Wan, S.J. Forsythe, H. El-Nezami, Probiotics interaction with foodborne growth, in: Advances in Microbial Physiology, vol. 39, Academic Press, 1997,
pathogens: a potential alternative to antibiotics and future challenges, Crit. Rev. pp. 205–234.
Food Sci. Nutr. 59 (20) (2019) 3320–3333. [50] S.C. De Keersmaecker, T.L. Verhoeven, J. Desair, K. Marchal, J. Vanderleyden,
[25] R. Jebali, S. Abbes, J.B. Salah-Abbès, R.B. Younes, Z. Haous, R. Oueslati, Ability I. Nagy, Strong antimicrobial activity of Lactobacillus rhamnosus GG against
of Lactobacillus plantarum MON03 to mitigate aflatoxins (B1 and M1) Salmonella typhimurium is due to accumulation of lactic acid, FEMS Microbiol.
immunotoxicities in mice, J. Immunotoxicol. 12 (3) (2015) 290–299. Lett. 259 (1) (2006) 89–96.
[26] F.B. Taheur, K. Fedhila, K. Chaieb, B. Kouidhi, A. Bakhrouf, L. Abrunhosa, [51] T. Mukai, T. Asasaka, E. Sato, K. Mori, M. Matsumoto, H. Ohori, Inhibition of
Adsorption of aflatoxin B1, zearalenone and ochratoxin a by microorganisms binding of Helicobacter pylori to the glycolipid receptors by probiotic
isolated from Kefir grains, Int. J. Food Microbiol. 251 (2017) 1–7. Lactobacillus reuteri, FEMS Immunol. Med. Microbiol. 32 (2) (2002) 105–110.
[27] A. Ismail, R.E. Levin, M. Riaz, S. Akhtar, Y.Y. Gong, C.A. de Oliveira, Effect of [52] V.D. Valeriano, B.B. Bagon, M.P. Balolong, D.K. Kang, Carbohydrate-binding
different microbial concentrations on binding of aflatoxin M1 and stability specificities of potential probiotic Lactobacillus strains in porcine jejunal (IPEC-
testing, Food Control 73 (2017) 492–496. J2) cells and porcine mucin, J. Microbiol. 54 (7) (2016) 510–519.
[28] K.R. Rao, A.V. Vipin, P. Hariprasad, K.A. Appaiah, G.J.F.C. Venkateswaran, [53] T. Kawai, S. Akira, The role of pattern-recognition receptors in innate immunity:
Biological detoxification of Aflatoxin B1 by Bacillus licheniformis CFR1, Food update on Toll-like receptors, Nat. Immunol. 11 (5) (2010) 373.
Control 71 (2017) 234–241. [54] T.A. Oelschlaeger, Mechanisms of probiotic actions–a review, Int. J. Med.
[29] P. Harkai, I. Szabó, M. Cserháti, C. Krifaton, A. Risa, J. Radó, et al., Microbiol. 300 (1) (2010) 57–62.
Biodegradation of aflatoxin-B1 and zearalenone by Streptomyces sp. collection, [55] S. Hummel, K. Veltman, C. Cichon, U. Sonnenborn, M.A. Schmidt, Differential
Int. Biodeterior. Biodegrad. 108 (2016) 48–56. targeting of the E-cadherin/β-catenin complex by Gram-positive probiotic
[30] Y. Kikuchi, A. Kunitoh-Asari, K. Hayakawa, S. Imai, K. Kasuya, K. Abe, et al., Oral lactobacilli improves epithelial barrier function, Appl. Environ. Microbiol. 78 (4)
administration of Lactobacillus plantarum strain AYA enhances IgA secretion and (2012) 1140–1147.
provides survival protection against influenza virus infection in mice, PLoS One 9 [56] A. Mattar, D.H. Teitelbaum, R. Drongowski, F. Yongyi, C. Harmon, A. Coran,
(1) (2014). Probiotics up-regulate MUC-2 mucin gene expression in a Caco-2 cell-culture
[31] F. Sakai, T. Hosoya, A. Ono-Ohmachi, K. Ukibe, A. Ogawa, T. Moriya, et al., model, Pediatric Surg. Int. 18 (7) (2002) 586–590.
Lactobacillus gasseri SBT2055 induces TGF-β expression in dendritic cells and [57] M.C. Collado, E. Isolauri, S. Salminen, Specific probiotic strains and their
activates TLR2 signal to produce IgA in the small intestine, PLoS One 9 (8) combinations counteract adhesion of Enterobacter sakazakii to intestinal mucus,
(2014). FEMS Microbiol. Lett. 285 (1) (2008) 58–64.
[32] K. Takeda, T. Suzuki, S.I. Shimada, K. Shida, M. Nanno, K. Okumura, Interleukin- [58] J.M. Otte, D.K. Podolsky, Functional modulation of enterocytes by gram-positive
12 is involved in the enhancement of human natural killer cell activity by and gram-negative microorganisms, Am. J. Physiol.-Gastrointest. Liver Physiol.
Lactobacillus casei Shirota, Clin. Exp. Immunol. 146 (1) (2006) 109–115. 286 (4) (2004) G613–G626.
[33] D. Fujiwara, S. Inoue, H. Wakabayashi, T. Fujii, The anti-allergic effects of lactic [59] E. Furrie, S. Macfarlane, A. Kennedy, J.H. Cummings, S.V. Walsh, D.A. O’neil, G.
acid bacteria are strain dependent and mediated by effects on both Th1/Th2 T. Macfarlane, Synbiotic therapy (Bifidobacterium longum/Synergy 1) initiates
cytokine expression and balance, Int. Arch. Allergy Immunol. 135 (3) (2004) resolution of inflammation in patients with active ulcerative colitis: a randomised
205–215. controlled pilot trial, Gut 54 (2) (2005) 242–249.
[34] H. Kanzato, S. Fujiwara, W. Ise, S. Kaminogawa, R. Sato, S. Hachimura, [60] Y. Kadooka, K. Tominari, F. Sakai, H. Yasui, Prevention of rotavirus-induced
Lactobacillus acidophilus strain L-92 induces apoptosis of antigen-stimulated T diarrhea by preferential secretion of IgA in breast milk via maternal
cells by modulating dendritic cell function, Immunobiology 213 (5) (2008) administration of Lactobacillus gasseri SBT2055, J. Pediatr. Gastroenterol. Nutr.
399–408. 55 (1) (2012) 66–71.
[35] A. Aoki-Yoshida, K. Yamada, S. Hachimura, T. Sashihara, S. Ikegami, M. Shimizu, [61] A. Przemska-Kosicka, C.E. Childs, S. Enani, C. Maidens, H. Dong, I.B. Dayel, et al.,
M. Totsuka, Enhancement of oral tolerance induction in DO11. 10 mice by Effect of a synbiotic on the response to seasonal influenza vaccination is strongly
Lactobacillus gasseri OLL2809 via increase of effector regulatory T cells, PLoS influenced by degree of immunosenescence, Immun. Ageing 13 (1) (2016) 6.
One 11 (7) (2016). [62] L. Vitetta, E.T. Saltzman, M. Thomsen, T. Nikov, S. Hall, Adjuvant probiotics and
[36] T. Shimazu, J. Villena, M. Tohno, H. Fujie, S. Hosoya, T. Shimosato, et al., the intestinal microbiome: enhancing vaccines and immunotherapy outcomes,
Immunobiotic Lactobacillus jensenii elicits anti-inflammatory activity in porcine Vaccines 5 (4) (2017) 50.
intestinal epithelial cells by modulating negative regulators of the Toll-like [63] T.R. Klaenhammer, M. Kleerebezem, M.V. Kopp, M. Rescigno, The impact of
receptor signaling pathway, Infect. Immun. 80 (1) (2012) 276–288. probiotics and prebiotics on the immune system, Nat. Rev. Immunol. 12 (10)
[37] T. Kawashima, A. Kosaka, H. Yan, Z. Guo, R. Uchiyama, R. Fukui, et al., Double- (2012) 728–734.
stranded RNA of intestinal commensal but not pathogenic bacteria triggers [64] S.C. Corr, Y. Li, C.U. Riedel, P.W. O’Toole, C. Hill, C.G. Gahan, Bacteriocin
production of protective interferon-β, Immunity 38 (6) (2013) 1187–1197. production as a mechanism for the antiinfective activity of Lactobacillus
[38] Y. Nakamura, M. Terahara, T. Iwamoto, K. Yamada, M. Asano, S. Kakuta, et al., salivarius UCC118, Proc. Natl. Acad. Sci. U. S. A. 104 (18) (2007) 7617–7621.
Upregulation of polymeric immunoglobulin receptor expression by the heat- [65] C. Gómez-Llorente, S. Munoz, A. Gil, Role of Toll-like receptors in the
inactivated potential probiotic bifidobacterium bifidum OLB6378 in a mouse development of immunotolerance mediated by probiotics, Proc. Nutr. Soc. 69 (3)
intestinal explant model, Scand. J. Immunol. 75 (2) (2012) 176–183. (2010) 381–389.

9
T.J. Ashaolu Biomedicine & Pharmacotherapy 130 (2020) 110625

[66] C. Di Giacinto, M. Marinaro, M. Sanchez, W. Strober, M. Boirivant, Probiotics bifidobacteria in young girls: a double-blind cross-over trial, Br. J. Nutr. 110 (7)
ameliorate recurrent Th1-mediated murine colitis by inducing IL-10 and IL-10- (2013) 1292–1303.
dependent TGF-β-bearing regulatory cells, J. Immunol. 174 (6) (2005) [91] O. Chonan, K. Matsumoto, M. Watanuki, Effect of galactooligosaccharides on
3237–3246. calcium absorption and preventing bone loss in ovariectomized rats, Biosci.
[67] E.O. Petrof, K. Kojima, M.J. Ropeleski, M.W. Musch, Y. Tao, C. De Simone, E. Biotechnol. Biochem. 59 (2) (1995) 236–239.
B. Chang, Probiotics inhibit nuclear factor-κB and induce heat shock proteins in [92] P.D. Cani, S. Possemiers, T. Van de Wiele, Y. Guiot, A. Everard, O. Rottier, et al.,
colonic epithelial cells through proteasome inhibition, Gastroenterology 127 (5) Changes in gut microbiota control inflammation in obese mice through a
(2004) 1474–1487. mechanism involving GLP-2-driven improvement of gut permeability, Gut 58 (8)
[68] D. Kelly, J.I. Campbell, T.P. King, G. Grant, E.A. Jansson, A.G. Coutts, et al., (2009) 1091–1103.
Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear- [93] J.A. Krumbeck, H.E. Rasmussen, R.W. Hutkins, J. Clarke, K. Shawron,
cytoplasmic shuttling of PPAR-γ and RelA, Nat. Immunol. 5 (1) (2004) 104–112. A. Keshavarzian, J. Walter, Probiotic Bifidobacterium strains and
[69] G. Tarantino, C. Finelli, Systematic review on intervention with prebiotics/ galactooligosaccharides improve intestinal barrier function in obese adults but
probiotics in patients with obesity-related nonalcoholic fatty liver disease, Future show no synergism when used together as synbiotics, Microbiome 6 (1) (2018)
Microbiol. 10 (5) (2015) 889–902. 121.
[70] F. Sánchez de Medina, I. Romero-Calvo, C. Mascaraque, O. Martínez-Augustin, [94] N.J. Kellow, M.T. Coughlan, C.M. Reid, Metabolic benefits of dietary prebiotics in
Intestinal inflammation and mucosal barrier function, Inflamm. Bowel Dis. 20 human subjects: a systematic review of randomised controlled trials, Br. J. Nutr.
(12) (2014) 2394–2404. 111 (7) (2014) 1147–1161.
[71] G.R. Gibson, M.B. Roberfroid, Dietary modulation of the human colonic [95] B.T. Beserra, R. Fernandes, V.A. do Rosario, M.C. Mocellin, M.G. Kuntz, E.
microbiota: introducing the concept of prebiotics, J. Nutr. 125 (6) (1995) B. Trindade, A systematic review and meta-analysis of the prebiotics and
1401–1412. synbiotics effects on glycaemia, insulin concentrations and lipid parameters in
[72] G.R. Gibson, R. Hutkins, M.E. Sanders, S.L. Prescott, R.A. Reimer, S.J. Salminen, adult patients with overweight or obesity, Clin. Nutr. 34 (5) (2015) 845–858.
et al., Expert consensus document: the International scientific association for [96] F. Liu, M. Prabhakar, J. Ju, H. Long, H.W. Zhou, Effect of inulin-type fructans on
probiotics and prebiotics (ISAPP) consensus statement on the definition and scope blood lipid profile and glucose level: a systematic review and meta-analysis of
of prebiotics, Nat. Rev. Gastroenterol. Hepatol. 14 (8) (2017) 491. randomized controlled trials, Eur. J. Clin. Nutr. 71 (1) (2017) 9–20.
[73] T.J. Ashaolu, B. Saibandith, C.T. Yupanqui, S. Wichienchot, Human colonic [97] A.M. Neyrinck, B. Pachikian, B. Taminiau, G. Daube, R. Frederick, P.D. Cani, et
microbiota modulation and branched chain fatty acids production affected by soy al., Intestinal sucrase as a novel target contributing to the regulation of glycemia
protein hydrolysate, Int. J. Food Sci. Technol. 54 (1) (2019) 141–148. by prebiotics, PLoS one 11 (8) (2016).
[74] I.A. Neri-Numa, G.M. Pastore, Novel insights into prebiotic properties on human [98] S. Bhatia, P.N. Prabhu, A.C. Benefiel, M.J. Miller, J. Chow, S.R. Davis, H.
health: a review, Food Res. Int. 131 (2020) 108973, https://doi.org/10.1016/j. R. Gaskins, Galacto-oligosaccharides may directly enhance intestinal barrier
foodres.2019.108973. function through the modulation of goblet cells, Mol. Nutr. Food Res. 59 (3)
[75] K.P. Scott, J.C. Martin, S.H. Duncan, H.J. Flint, Prebiotic stimulation of human (2015) 566–573.
colonic butyrate-producing bacteria and bifidobacteria, in vitro, FEMS Microbiol. [99] P. Akbari, J. Fink-Gremmels, R.H. Willems, E. Difilippo, H.A. Schols, M.
Ecol. 87 (1) (2014) 30–40. H. Schoterman, et al., Characterizing microbiota-independent effects of
[76] A. Moya, M. Ferrer, Functional redundancy-induced stability of gut microbiota oligosaccharides on intestinal epithelial cells: insight into the role of structure and
subjected to disturbance, Trends Microbiol. 24 (5) (2016) 402–413. size, Eur. J. Nutr. 56 (5) (2017) 1919–1930.
[77] Y. Nakanishi, K. Murashima, H. Ohara, T. Suzuki, H. Hayashi, M. Sakamoto, et al., [100] A.C.D. Lima, C. Cecatti, M.P. Fidélix, M.A.T. Adorno, I.K. Sakamoto, T.B. Cesar,
Increase in terminal restriction fragments of Bacteroidetes-derived 16S rRNA K. Sivieri, Effect of daily consumption of orange juice on the levels of blood
genes after administration of short-chain fructooligosaccharides, Appl. Environ. glucose, lipids, and gut microbiota metabolites: controlled clinical trials, J. Med.
Microbiol. 72 (9) (2006) 6271–6276. Food 22 (2) (2019) 202–210.
[78] S. Arslanoglu, G.E. Moro, G. Boehm, F. Wienz, B. Stahl, E. Bertino, Early Neutral [101] O. Perdijk, P. Van Baarlen, M.M. Fernandez-Gutierrez, E. Van Den Brink, F.
Prebiotic Oligosaccharide Supplentation reduces the incidence of some allergic H. Schuren, S. Brugman, et al., Sialyllactose and galactooligosaccharides promote
manifestations in the first 5 years of life, J. Biol. Regul. Homeost. Agents 26 epithelial barrier functioning and distinctly modulate microbiota composition
(2012) 49–59. and short chain fatty acid production, Front. Immunol. 10 (2019) 94.
[79] G. Moro, S. Arslanoglu, B. Stahl, J. Jelinek, U. Wahn, G. Boehm, A mixture of [102] G. Paturi, C.A. Butts, J.A. Monro, D. Hedderley, Effects of blackcurrant and
prebiotic oligosaccharides reduces the incidence of atopic dermatitis during the dietary fibers on large intestinal health biomarkers in rats, Plant Foods Hum.
first six months of age, Arch. Dis. Child. 91 (10) (2006) 814–819. Nutr. 73 (1) (2018) 54–60.
[80] O.S. Ivakhnenko, S.L. Nyankovskyy, Effect of the specific infant formula mixture [103] Y.J. Cho, H.G. Lee, K.H. Seo, W. Yokoyama, H. Kim, Antiobesity effect of prebiotic
of oligosaccharides on local immunity and development of allergic and infectious polyphenol-rich grape seed flour supplemented with probiotic kefir-derived lactic
disease in young children: randomized study, Pediatria Polska 88 (5) (2013) acid bacteria, J. Agric. Food. Chem. 66 (47) (2018) 12498–12511.
398–404. [104] T. Wu, C. Grootaert, J. Pitart, N.K. Vidovic, S. Kamiloglu, S. Possemiers, et al.,
[81] M.H. Kim, S.G. Kang, J.H. Park, M. Yanagisawa, C.H. Kim, Short-chain fatty acids Aronia (Aronia melanocarpa) Polyphenols Modulate the Microbial Community in
activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory a Simulator of the Human Intestinal Microbial Ecosystem (SHIME) and decrease
responses in mice, Gastroenterology 145 (2) (2013) 396–406. secretion of proinflammatory markers in a Caco-2/endothelial cell coculture
[82] L.A. Stoddart, N.J. Smith, G. Milligan, International Union of Pharmacology. model, Mol. Nutr. Food Res. 62 (22) (2018), 1800607.
LXXI. Free fatty acid receptors FFA1,-2, and-3: pharmacology and [105] M.J. Alfa, D. Strang, P.S. Tappia, M. Graham, G. Van Domselaar, J.D. Forbes, et
pathophysiological functions, Pharmacol. Rev. 60 (4) (2008) 405–417. al., A randomized trial to determine the impact of a digestion resistant starch
[83] E.S. Chambers, D.J. Morrison, G. Frost, Control of appetite and energy intake by composition on the gut microbiome in older and mid-age adults, Clin. Nutr. 37 (3)
SCFA: what are the potential underlying mechanisms? Proc. Nutr. Soc. 74 (3) (2018) 797–807.
(2015) 328–336. [106] P. Van den Abbeele, B. Taminiau, I. Pinheiro, C. Duysburgh, H. Jacobs, L. Pijls,
[84] G. Frost, M.L. Sleeth, M. Sahuri-Arisoylu, B. Lizarbe, S. Cerdan, L. Brody, et al., M. Marzorati, Arabinoxylo-oligosaccharides and inulin impact inter-individual
The short-chain fatty acid acetate reduces appetite via a central homeostatic variation on microbial metabolism and composition, which immunomodulates
mechanism, Nat. Commun. 5 (1) (2014) 1–11. human cells, J. Agric. Food. Chem. 66 (5) (2018) 1121–1130.
[85] L.J. Fooks, G.R. Gibson, In vitro investigations of the effect of probiotics and [107] S. Estruel-Amades, M. Massot-Cladera, F.J. Pérez-Cano, À. Franch, M. Castell,
prebiotics on selected human intestinal pathogens, FEMS Microbiol. Ecol. 39 (1) M. Camps-Bossacoma, Hesperidin effects on gut microbiota and gut-associated
(2002) 67–75. lymphoid tissue in healthy rats, Nutrients 11 (2) (2019) 324.
[86] G. Tzortzis, M.L. Baillon, G.R. Gibson, R.A. Rastall, Modulation of anti-pathogenic [108] R. Grimaldi, G.R. Gibson, J. Vulevic, N. Giallourou, J.L. Castro-Mejía, L.
activity in canine-derived Lactobacillus species by carbohydrate growth H. Hansen, et al., A prebiotic intervention study in children with autism spectrum
substrate, J. Appl. Microbiol. 96 (3) (2004) 552–559. disorders (ASDs), Microbiome 6 (1) (2018) 133.
[87] J. Vulevic, A. Juric, G.E. Walton, S.P. Claus, G. Tzortzis, R.E. Toward, G. [109] S. Nedaei, A. Noori, A. Valipour, A.A. Khanipour, S.H. Hoseinifar, Effects of
R. Gibson, Influence of galacto-oligosaccharide mixture (B-GOS) on gut dietary galactooligosaccharide enriched commercial prebiotic on growth
microbiota, immune parameters and metabonomics in elderly persons, Br. J. performance, innate immune response, stress resistance, intestinal microbiota and
Nutr. 114 (4) (2015) 586–595. digestive enzyme activity in Narrow clawed crayfish (Astacus leptodactylus
[88] S.L. Goss, K.A. Lemons, J.E. Kerstetter, R.H. Bogner, Determination of calcium Eschscholtz, 1823), Aquaculture 499 (2019) 80–89.
salt solubility with changes in pH and PCO2, simulating varying gastrointestinal [110] X. Jiao, Y. Wang, Y. Lin, Y. Lang, E. Li, X. Zhang, et al., Blueberry polyphenols
environments, J. Pharm. Pharmacol. 59 (11) (2007) 1485–1492. extract as a potential prebiotic with anti-obesity effects on C57BL/6 J mice by
[89] S.A. Abrams, I.J. Griffin, K.M. Hawthorne, K.J. Ellis, Effect of prebiotic modulating the gut microbiota, J. Nutr. Biochem. 64 (2019) 88–100.
supplementation and calcium intake on body mass index, J. Pediatr. 151 (3) [111] M.D.M. Rigo-Adrover, K. Knipping, J. Garssen, K. van Limpt, J. Knol, À. Franch, et
(2007) 293–298. al., Prevention of rotavirus diarrhea in suckling rats by a specific fermented milk
[90] C.M. Whisner, B.R. Martin, M.H. Schoterman, C.H. Nakatsu, L.D. McCabe, G. concentrate with prebiotic mixture, Nutrients 11 (1) (2019) 189.
P. McCabe, et al., Galacto-oligosaccharides increase calcium absorption and gut [112] S. Soldi, S. Vasileiadis, S. Lohner, F. Uggeri, E. Puglisi, P. Molinari, et al., Prebiotic
supplementation over a cold season and during antibiotic treatment specifically

10
T.J. Ashaolu Biomedicine & Pharmacotherapy 130 (2020) 110625

modulates the gut microbiota composition of 3-6 year-old children, Benefic. [119] P. Louis, G.L. Hold, H.J. Flint, The gut microbiota, bacterial metabolites and
Microbes 10 (3) (2019) 253–263. colorectal cancer, Nat. Rev. Microbiol. 12 (10) (2014) 661–672.
[113] P. Courvalin, Antibiotic resistance: the pros and cons of probiotics, Dig. Liver Dis. [120] A. Chlebicz, K. Śliżewska, In vitro detoxification of aflatoxin B 1, deoxynivalenol,
38 (2006) S261–S265. fumonisins, T-2 toxin and zearalenone by probiotic bacteria from genus
[114] J. Cummins, M.W. Ho, Genetically modified probiotics should be banned, Microb. lactobacillus and saccharomyces cerevisiae yeast, Probiot. Antimicrob. Proteins
Ecol. Health Dis. 17 (2) (2005) 66–68. 12 (1) (2020) 289–301.
[115] G. Reid, R. Kort, S. Alvarez, R. Bourdet-Sicard, V. Benoit, M. Cunningham, et al., [121] X. Ze, S.H. Duncan, P. Louis, H.J. Flint, Ruminococcus bromii is a keystone
Expanding the reach of probiotics through social enterprises, Benefic. Microbes 9 species for the degradation of resistant starch in the human colon, The ISME J . 6
(5) (2018) 707–715. (8) (2012) 1535–1543.

11

You might also like