Download as pdf or txt
Download as pdf or txt
You are on page 1of 37

Accepted Manuscript

Title: Actinobacteria: a relevant minority for the maintenance


of gut homeostasis

Author: Cecilia Binda Loris Riccardo Lopetuso Gianenrico


Rizzatti Giulia Gibiino Vincenzo Cennamo Antonio
Gasbarrini

PII: S1590-8658(18)30210-X
DOI: https://doi.org/doi:10.1016/j.dld.2018.02.012
Reference: YDLD 3676

To appear in: Digestive and Liver Disease

Received date: 18-10-2017


Revised date: 26-1-2018
Accepted date: 19-2-2018

Please cite this article as: Binda C, Lopetuso LR, Rizzatti G, Gibiino
G, Cennamo V, Gasbarrini A, Actinobacteria: a relevant minority for the
maintenance of gut homeostasis, Digestive and Liver Disease (2018),
https://doi.org/10.1016/j.dld.2018.02.012

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.
Title page

ACTINOBACTERIA: A RELEVANT MINORITY FOR THE MAINTENANCE OF GUT

HOMEOSTASIS

Cecilia Binda*, Loris Riccardo Lopetuso*, Gianenrico Rizzatti*, Giulia Gibiino*, Vincenzo

Cennamo^, Antonio Gasbarrini*

t
ip
cr
* Department of Internal Medicine, Gastroenterology and Hepatology, Catholic University of

Sacred Heart of Rome, A. Gemelli Hospital - Italy.

us
^ Unit of Gastroenterology and Digestive Endoscopy, AUSL Bologna Bellaria-Maggiore Hospital,

Bologna - Italy
an
M
Electronic word count: 4120
d

Corresponding Author:
e

Prof. Antonio Gasbarrini


pt

Department of Internal Medicine, Gastroenterology Division,

Catholic University of Rome, Policlinico “A. Gemelli” Hospital


ce

Largo Gemelli, 8
Ac

00168 Rome (ITALY)

Phone/fax number: 0039-06-30156018

E-mail: antonio.gasbarrini@unicatt.it

Page 1 of 36
Abstract

Actinobacteria are one the four major phyla of the gut microbiota and, although they represent only

a small percentage, are pivotal in the maintenance of gut homeostasis. During the last decade many

studies focused the attention on Actinobacteria, especially on their role both in gastrointestinal and

systemic diseases and on their possible therapeutic use. In fact, classes of this phylum, especially

Bifidobacteria, are widely used as probiotic demonstrating beneficial effects in many pathological

t
ip
conditions, even if larger in vivo studies are needed to confirm such encouraging results. This

cr
review aims to explore the current knowledge on their physiological functions and to speculate on

their possible therapeutic role(s) in gastrointestinal and systemic diseases.

us
Key words: gut microbiota, Actinobacteria, Bifidobacteria spp, gut homeostasis, dysbiosis,

probiotic.
an
M
e d
pt
ce
Ac

Page 2 of 36
INTRODUCTION

The gut microbiota is composed by multiple commensal microbial species including 100 trillion

(10^14) bacteria, quadrillion viruses, fungi, parasites, archeas and yeasts, reaching an overall

biomass of about 1 kg and more than 3 million of genes [1- 4]. The different gastrointestinal regions

are characterized by a different bio-compartmentalization with a distinct and stable microbial

community. This is influenced by the acid environment, the presence of bile and pancreatic

t
ip
secretion and the high peristaltic activity in the stomach and small intestine, which do not allow a

cr
stable bacterial colonization, differently from the colon where bacterial colonization is favored by

the low redox potential and the slow transit [5, 6]. The majority of microbes forming the human

us
microbiota can be assigned to four major phyla: Bacteroidetes, Firmicutes, Proteobacteria and

Actinobacteria [7, 8]. Firmicutes and Bacteroidetes represent more than 90% of the relative
an
abundance of the gut microbiome and their relationship plays a pivotal role in the maintenance of
M
gut homeostasis. Actinobacteria and Proteobacteria represent the remaining 10% [7, 8].

Mainly oropharyngeal origin aerobic gram-positive bacteria inhabit stomach, duodenum and
d

jejunum, whereas coliforms and anaerobic species (such as Bacteroides, Bifidobateria, Clostridia
e

and Lactobacilli) are predominant in the ileum and post ileocecal valve, respectively [9, 10].
pt

Gut microbiota is involved in many useful functions, such as energy production from nutrient

biotransformation [11, 12], regulation of lipid metabolism [13], metabolism of vitamins and
ce

absorption of calcium, magnesium and iron [12, 14, 15], maintenance of the intestinal barrier
Ac

function [16, 17], the development of immune system from the first days of life [18- 23].

Many conditions are supposed to be related to quantitative and qualitative changes in gut

microbiota composition and function, such as inflammatory bowel diseases, celiac disease, irritable

bowel syndrome (IBS), obesity, non-alcoholic fatty liver disease (NAFLD) and non-alcoholic

steato-hepatitis (NASH), diabetes, cardiovascular disease, arthritis, psoriasis and psychiatric

disorders. This gut microbiota impairment is known as dysbiosis [24- 30].

Page 3 of 36
Recently, increasing interest has focused on the phyla of Actinobacteria, especially on

Bifidobacteria family. The purpose of this review is to explore the crucial role of Actinobacteria in

the maintenance of gut homeostasis and the critical importance of their future therapeutic

applications.

ACTINOBACTERIA IN THE GASTROINTESTINAL TRACT

t
ip
Actinobacteria are Gram positive, multiple branching rods, non-motile, non-spore-forming and

cr
anaerobic bacteria [31], that include three main anaerobe families (Bifidobacteria, Propionibacteria

and Corynebacteria) and an aerobe family (Streptomyces). The most represented in the human gut

us
are Bifidobacteria. Interestingly, the complete genome sequences are available for certain

Bifidobacterial species, like B. adolescents, B. animalis, B. breve, B. bifidum, B. long and B.

angulatum [31].
an
M
Many factors can impact on the presence of Actinobacteria in the intestine. Although the relation

between phylum diversity and delivery is still unclear, studies demonstrated a higher diversity
d

within Actinobacteria phylum among children that underwent vaginal delivery [32-37]. In fact,
e

phylum diversity within Actinobacteria, (i.e., Bifidobacterium) was significantly lower in infants
pt

delivered by caesarean section compared to vaginally delivered newborns during the first week of

life [33 -35], at the age of 1 month [36], and three months [37]. Other studies did not confirm such
ce

differences or showed a lower effect on Bifidobacteria and Bacteroides colonization at the age of 6
Ac

and 12 month [38, 39]. The lower abundance of Bifidobacteria and Bacteroides in c-section

delivered infants may be explained with a higher mother consumption of antibiotics before, during

and after the delivery. Indeed, postnatal use of antibiotic has been associated with decreased

numbers of Bifidobacterium and Bacteroides and a higher relative abundance of the Clostridium

leptum [40- 42]. Furthermore, a higher abundance of Bifidobacterium genus has been demonstrated

in breastfed infants [43]. Interestingly, it has been demonstrated that human milk is rich in

Page 4 of 36
substances, like human milk oligosaccharides (HMOs), that may act as probiotics and stimulate the

growth of Lactobacillus and Bifidobacterium species [44].

Weaning causes a further change in microbial composition and Bifidobacteria are no longer the

dominant group, with the adult microbiota established after the second year of life [45]. During the

adulthood, the percentage of Actinobacteria remains stable around 8% [46] and constitutes one of

the prevalent commensal bacterial in the distal small and large intestine [47]. Intriguingly, an Italian

t
ip
study on centenarian patients showed a reduction in the total number of anaerobes compared to

cr
young adults, with a reduction of Bifidobacteria and Bacteroides [48]. These evidences have opened

new horizons for the concept of “fragile” microbiota that could deeply affect the delicate health

us
equilibrium in old patients.

ACTINOBACTERIA AND GUT BARRIER


an
M
Gut barrier is a multi-layer system able to afford a daily exposure to pathogens and external

environment. It can be divided into a superficial physical barrier constituted by the epithelial cells,
d

the tight junctions and the mucus; and a functional barrier represented by gut-associated mucosa
e

lymphoid tissue (GALT), peristalsis and antimicrobial substances, able to regulate the
pt

immunological response to pathogens and tolerance to commensal bacteria [16, 49]. Gut microbiota

is pivotal in the maintenance of intestinal barrier functions, increasing tight junctions expression,
ce

regulating mucin biosynthesis and catabolism, providing energy for epithelial cells proliferation and
Ac

stimulating the immune system [17, 49]. In this scenario, Actinobacteria are absolute players in

maintaining gut barrier homeostasis.

The production of short chain fatty acids (SCFA), such as acetate, propionate and butyrate, from

carbohydrate fermentation is crucial for providing energy to epithelial cells turnover and for their

potent antibacterial activity [16, 17]. In this field, Bifidobacteria have beneficial effects in the

maintenance of gut barrier thanks to their great production of SCFA [50]. In particular, they

produce high concentration of acetate that can protect the host from enteropathogenic infections,

Page 5 of 36
such as entero-hemorrhagic Escherichia coli and Shigella [51]. Moreover it has been demonstrated

that Bifidobacteria have a non-negligible production of lactate, which can be metabolized by a

group of bacteria (“lactate utilizer”) to produce butyrate [52]. In this scenario, in vitro studies

demonstrated that SCFA, especially butyrate, are correlated with an increased expression of the

gene MUC2, a mucin glycoprotein that is one of the major component of the mucous layer and thus

of intestinal barrier [53-55]. The mechanism responsible for the increased production of MUC

t
ip
induced by butyrate is not definitely established. Gaudier et al demonstrated that butyrate could

cr
modify MUC gene expression in goblet cells depending on the energy source available with a

consequent higher expression of MUC2 in conditions of glucose deprivation when the consumption

us
of butyrate by goblet cells is increased [54]. Authors concluded that butyrate metabolism is

involved not only in mucin synthesis but can play a role also in gene transcription. Another in vitro
an
study showed a relationship between SCFA, especially butyrate, the production of prostaglandins
M
(PG) by intestinal myofibroblasts and epithelial mucin expression [53]. In fact, SCFA seemed to

enhance the production of PGE1 in subepithelial myofibroblasts that in turn was able to stimulate
d

epithelial mucin expression, probably through the epithelial differentiation induced by cell-to-cell
e

contact, extracellular matrix and soluble factors such as transforming growth factor β. Other
pt

important elements that contribute to the viscoelastic properties of the mucous layer are the Trefoil

factors (TFFs). These mucin-associated peptides are mainly secreted by goblet cells, are supposed
ce

to be involved in the mucosal maintenance and repair, and seem to reduce the recruitment of
Ac

inflammatory cells [56, 57]. Moreover, butyrate contributes to the colonic defense barrier by

increasing the expression of TFFs [58]. In vitro and in vivo studies and in studies showed a possible

effects of butyrate on the expression of heat shot proteins (HSPs), which contribute to the gut

barrier homeostasis by suppressing the production of inflammatory modulators [59, 60]. A

mechanism for the enhancement of HSPs expression has not been already discerned. So far, only

some hypotheses have been proposed. Indeed, SCFA are able to induce cellular acidification

Page 6 of 36
through a non-ionic diffusion that may induce the production of stress kinases and HSPs. Moreover,

the butyrate seems to inhibit the histone deacetylase, a crucial transcriptional regulator [59].

Finally, a possible role of butyrate on intestinal permeability has been assessed in different in vitro

studies, which demonstrated a butyrate influence on tight junctions expression. Interestingly, these

effects seem to be concentration-dependent, with a concentration up to 2 mM that can decrease the

intestinal permeability and a concentration higher than 8 mM that, on the contrary, is able to induce

t
ip
an increased permeability [56, 61, 62].

cr
ACTINOBACTERIA AND METABOLISM

us
Human intestine does not possess many of the enzymes involved in the degradation and

biotransformation of substances introduced with the diet. Indeed, the majority of enzymes engaged
an
in energy production is provided by the commensal bacteria, especially located in the colon and
M
belonging to the genus Bacteroides, Bifidobacterium, Ruminococcus and Roseburia [63]. In

particular, the pathways involved include the fermentation of large polysaccharides,


d

oligosaccarides, unabsorbed sugars and fibers, that release hydrogen, carbon dioxide and SCFAs,
e

the degradation of proteins, the regulation of lipid metabolism through lipoprotein lipase (LPL), and
pt

the absorption and biosynthesis of vitamin K, B12, iron, calcium and magnesium [13-15, 64-67].

Bifidobacteria are able to produce large quantities of acetate, but do not produce propionate and
ce

butyrate, which are mainly produced by Bacteroides phylium and Clostridium cluster XIVa and IV
Ac

[68, 69]. However, the acetate released by B. longum NCC2705 represents a co-substrate for

butyrate production [70] that constitutes the main energy source for colonocytes [71].

Actinobacteria are also involved in the biodegradation of resistant starch [72 - 74]. Actinobacteria,

in particular Bifidobacteria, through the glycosyl hydrolases (GHs) hydrolyze the glycosidic bond

between two or more sugars and cooperate to the breakdown of plant-derived carbohydrate starch

and polysaccharides, such as FOS, GOS, XOS, inulin or arabinoxilan [75 - 77]. Moreover,

Bifidobacteria are supposed to be involved in the transformation of linoleic acid (LA) into

Page 7 of 36
conjugated linoleic acids (CLA), a group of isomers of LA [78], that have potential health-

promoting properties like anticarcinogenesis, anti-atherosclerosis, anti-diabetes, anti obesity and

enhancement of immune functions [79].

Although it has been largely demonstrated that the diet strongly influences gut microbiota

composition, the relationship between diet and Actinobacteria is still unclear. In fact, energy

restrictions, high fiber diet and dietary components such as fructo-oligosaccharides (FOS), galacto-

t
ip
oligosaccharides (GOS), xylo-oligosaccharides (XOS) are associated to higher microbial diversity

cr
[80]. While some studies showed that Actinobacteria abundance is positively associated with a

high-fat diet and negatively associated with fiber intake [81, 82], an opposite association is

us
suggested by other studies in which a high concentration of Bifidobacterium spp is positively

correlated with lean individuals, high consumption of complex carbohydrates, improvement of


an
glucose homeostasis, reduction of obesity and inflammation [83 – 86]. A study comparing fecal
M
samples of lean and obese women showed different concentration of Bifidobacteria and Clostridium

coccoides between the two groups. In particular, there was a negative correlation between
d

Bifidobacteria and body fat percentage with an inverse correlation with insulin levels and the
e

homeostasis model assessment (HOMA) index [83]. However, in this study there was no difference
pt

in LPS levels between lean and obese subjects and no correlation with body fat percentage, insulin

levels or HOMA-index. Nevertheless, a study by Cani et al [85] demonstrated that mice fed with an
ce

high fat diet enriched with prebiotic (fermentable dietary fibre, oligofructose (OFS)) could restore
Ac

their levels of Bifidobacteria, improve glucose tolerance, insulin secretion, and induce an increase

of proglucagon mRNA precursor. Authors highlighted that all these changes were correlated with a

reduction of endotoxiemia, suggesting a protective role of Bifidobacteria, while no relationship was

evident with other bacterial group [85]. Despite the mechanism of action has not been completely

understood, authors suggested that the product of OFS degradation, the SCFA, could ameliorate the

gut barrier function both through a direct action on colonic cells and by modulating gut microbiota,

especially Bifidobacteria [85]. Furthermore, the use of dextrins derived from maize starch is able to

Page 8 of 36
stimulate the growth of Actinobacteria and Bacteroidetes in both normal-weight and obese children

[87], with possible beneficial effect on the production of SCFA.

A modulation on gut microbiota, and therefore on Actinobacteria phylum, has been shown with the

diet low in fermentable oligosaccharides, disaccharides and polyols, fructo-oligosaccharides and

galacto-oligosaccharides (FODMAPs). FODMAPs are small carbohydrate molecules that are

slowly absorbed in the form of monosaccharides or not absorbed when disaccharides or

t
ip
polysaccharides, lasting for a prolonged period into the intestinal lumen [67]. There, FODMAPs

cr
exert an osmotic action collecting water into the lumen and are fermented by commensal bacteria

releasing SCFA, dioxide, hydrogen, methane and carbon [88, 89]. A study by McIntosh et al

us
compared the impact of high and low FODMAPs diet in IBS patients and demonstrated an

increased richness of Actinobacteria with a decreased number of Bifidobacteria in the low


an
FODMAPs diet group [90]. On the contrary, other studies showed that a low FODMAPs diet could
M
lead to a lower abundance in both Bifidobacteria and Actinobacteria because of their FODMAPs

need for their metabolism [91, 92]. However, not all the patients treated with low FODMAPs
d

experienced an improvement of IBS symptoms [90]. This could probably due to different effects of
e

diverse Bifidobacteria strains. Recent studies demonstrated that the use of Bifidobacterium infantis
pt

and Bifidobacterium animalis has beneficial effects on IBS symptoms [93 - 95]. However, all the

available studies have not examined the microbiome of patients with IBS following the
ce

reintroduction phase, but only after the end of the strict low-FODMAP diet, which is usually only
Ac

recommended for 2 to 6 weeks.

ACTINOBACTERIA AND IMMUNOLOGICAL FUNCTION

The gut microbiota plays a pivotal role in the development of immune system and Bifidobacteria,

are also critical in this field. The stimulation of intraepithelial lymphocytes, the production of

mucosal immunoglobulins and the promotion of a tolerogenic immune response are the main

functions exerted by commensal bacteria [19, 20,22, 23, 96].

Page 9 of 36
A decreased number of Bifidobacteria is associated to an enhancement of gut permeability [97] that

leads to the translocation of LPS into the serum. This triggers the immune system activation and

sustains chronic inflammatory conditions, such as insulin resistance, diabetes and liver diseases

[98]. The administration of Bifidobacterium pseudocatenulatum CECT 7765 along with high fat

diet in mice is able to down-regulate the inflammation by reducing the production of inflammatory

cytokines and chemokines, especially IL-6 and MCP-1, which are usually increased in obesity and

t
ip
metabolic disorders [99]. The reduction of these inflammatory markers is concomitant with the

cr
improvement of glucose tolerance and with the increasing of IL-4 and IL-13. These cytokines are

able to stimulate the M2 phenotypic differentiation of macrophages, a subtype of adipose tissue

us
macrophages that secretes the anti-inflammatory cytokine IL-10, and to promote the control of

inflammation and normal insulin sensitivity [100]. However, the same study showed that after the
an
administration of B. pseudocatenulatum CECT 7765 there were increased level of IL-10 only in
M
standard diet fed mice and not in high fat diet-fed mice, indicating that probably parallel regulatory

mechanisms are involved. Furthermore, B. pseudocatenulatum CECT 7765 can boost an


d

appropriate inflammatory response to a bacterial stimulus (LPS) that is usually impaired in high fat
e

diet fed mice. In fact, the administration of this probiotic in these mice stimulates the production of
pt

TNF-α by LPS-stimulated macrophages, boosts the oxidative burst and therefore the phagocytosis

function in peritoneal macrophages. Finally, it increases the ability of DCs to present antigens and
ce

prime a T-lymphocyte proliferative response [99]. In this direction, the reduced phagocytic capacity
Ac

and oxidative burst of macrophages and the impaired function of DCs are possible reasons for the

increased susceptibility to infections in obese subjects [101, 102].

Moreover, Actinobacteria, and mainly Bifidobacteria species, can modulate immune-inflammatory

and autoimmune response by inducing regulatory T-cells [103, 104].

Grounding on the evidence that Bifidobacterium longum subspecies infantis (B.infantis) induces

regulatory T cells activity in animal models [103, 105 - 108] and increases the relative proportion of

the same cells in peripheral blood of healthy humans [109], Groeger et al studied the effect of

Page 10 of 36
B.infantis inflammatory mediator production in inflammatory disorders in patients with ulcerative

colitis (UC), psoriasis and chronic fatigue syndrome (CFS) [110]. They found that 6-8 weeks of

treatment with B.infantis significantly reduced plasma C-reactive protein (CRP) levels in all three

group of patients, whereas a statistically significant attenuation of TNF-α was observed for psoriasis

and CFS. In the UC group they observed a trend of reduction in TNF-α levels that however did not

reach the statistical significance, probably because of the shorter treatment period (6 weeks of

t
ip
treatment vs 8 weeks of treatment of the other two groups). Instead, plasma levels of IL-6 were only

cr
marginally decreased after treatment reinforcing the hypothesis of a specific influence on immune

response for each gut microbiota component [110]. The use of Bifidobacterium species has also

us
been studied in respiratory pathology, alone or in combination with prebiotic or other probiotics like

Lactobacilllus. In this field, Bifidobacteria seem to be effective to prevent asthma-like symptoms in


an
infant with atopic dermatitis and to treat the symptoms of allergic rhinitis [111 – 115]. This is
M
probably due to an immunomodulatory effect of Bifidobacteria on Th balance, regulating Th-2

immunoresponse that is usually skewed in atopic patients and is responsible of higher levels of
d

eosinophils and IgE production [113].


e
pt

ACTINOBACTERIA AND GUT-BRAIN AXIS

Recently, the interest on the impact of gut microflora on gut-brain axis is increasing. This could
ce

involve neuroendocrine, immunological and direct neural mechanisms [116 - 118]. Dysbiosis and
Ac

the consequent increased intestinal permeability are associated to an up-regulation of systemic

inflammation that may also involve the central nervous system [119]. Both the gut microbiota

production of neurochemicals (i.e., serotonin, SCFA, dopamine and γ-aminobutyric acid) [116, 120]

and neurotoxic metabolites (i.e., ammonia) [119] by strengthens the possibility of an implication of

the gut-brain axis in depression, anxiety, IBS, IBD, and neurodevelopmental disorders such as

autism [116, 121 - 124]. A direct neural communication between the gut and the brain through the

Page 11 of 36
stimulation of the enteric nervous system and consequently the vagus nerve has also been supposed

[125, 126].

Standing on previous studies that showed a reduction of Bifidobacteria and lactobacilli following a

stressful experience and emotional stress both in animals and humans [127 - 131], Desbonnet et al

conducted a study to assess the potential antidepressant properties of probiotic Bifidobacteria

infantis in rats [132]. They demonstrated that a two-week treatment with the probiotic

t
ip
Bifidobacteria infantis determines an attenuation of pro-inflammatory immune response and an

cr
elevation of tryptophan, a precursor of serotonine that represents a target of antidepressant

treatments. Authors concluded that these results might support the beneficial effect of this probiotic

us
in the treatment of depression, especially when associated to gastrointestinal disorders. Two years

later, these findings were supported by the evidence that the chronic treatment with Bifidobacteria
an
infantis was able to attenuate depression in the rats exposed to maternal separation stress in early
M
life [133].

On the other side, a recent study by Lyte at al [134] showed that mice fed with resistant starch (RS)
d

had increased levels of Bifidobacteria, according also to a previous study by Tachon et al [135].
e

RS-fed mice evidenced a significant increase in anxiety-like behavior that the authors considered to
pt

be induced by the same diet with a concomitant abundance of Bifidobacterium [134].

Furthermore, a role of Actinobacteria, mainly Bifidobacteria, in the deterioration of cognitive


ce

function has been supposed. In fact, preliminary data on the use of the probiotic VSL#3, a mixture
Ac

of 8 different strains of bacteria including three Bifidobacteria species, showed an improvement in

the age-related decrease of long-term potentiation (LTP) in rats [136]. One of the mechanisms

supposed to be involved in the regulation of LTP is the anti-inflammatory effect of VSL#3 on the

modulation of the hippocampal molecules expression, such as CD 68 and CD 11b. These are

markers of microglial activation, and therefore of inflammation. Moreover the authors reported that

VSL#3 is able to influence the expression of several genes in the cortex. In particular, it is able to

attenuate the age-related changes in three genes, PLA2G3, Nid2 and Alox15, which are associated

Page 12 of 36
to inflammation. Changes of genes expression in the brain may be linked to the variation of several

molecules induced by VSL#3. In fact, this study showed that this probiotic regulates specific

mediators involved in synaptic plasticity in the hippocampus of aged rats, especially brain-derived

neurotrophic factor (BDNF), synapsin and syntaxin. The authors suggested that the synaptotrophic

effect of VSL#3 is mainly due to the BDNF, whose expression was increased in those rats treated

with the probiotic. The increase of BDNF levels was associated to higher level of synapsin. Of note,

t
ip
previous studies recognized its fundamental role in sustaining LTP through the induction of

cr
neurogenesis and synaptogenesis [136].

Lastly, the existence and the importance of gut-brain axis have also been suggested in alcohol

us
dependence. Some alcohol dependent subjects develop a high intestinal permeability that is

associated with significantly lower levels of Bifidobacteria [137]. These subjects are characterized
an
by higher levels of depression, anxiety and craving and have a consistent higher risk of persistence
M
of psychological symptoms after detoxification when compared to alcohol dependent subject with a

lower intestinal permeability, that on the contrary recover completely after the abstinence period.
e d

BIFIDOBACTERIA AS PROBIOTIC
pt

Bifidobacteria, together with Lactobacilli, represents the cornerstone of probiotic therapeutic

approach, but only few Bifidobacteria strains have been studied alone. Thus, in the latter cases the
ce

beneficial effects cannot be exclusively related to this probiotic family.


Ac

For example, the probiotic formulation VSL#3, a mixture of lyophilized four Lactobacilli (L. casei,

L. plantarium, L acidophilus, L. bulgaricus), three Bifidobacteria strains (B. longuum, B. breve, B.

infantis) and Streptococcus salivaris subspecies termophilus demonstrated to be effective in several

conditions. Its efficacy has been mainly assessed in the prevention of pouchitis relapse in patients

with UC [138, 139], with promising results in maintaining remission and treating mild-to-moderate

UC both in adult and children [140 - 143], in metabolic disorders, in non-alcoholic steato-hepatitis

(NASH) [144, 145] and in the prevention of antibiotic-associated diarrhea [146]. An amelioration of

Page 13 of 36
the pro-inflammatory status in patients affected by UC has been demonstrated with the use of

Bifidobacterium infantis alone [110], as mentioned above, and Bifidobacterium breve in association

with GOS [147]. The use of Bifidobacteria, alone or in association with other probiotics is also

effective in the alleviation of symptoms in patients with IBS [93 - 95, 148 - 150], constipation [151,

152] and in the reduction of antibiotic-associated diarrhea in infants [153]. Moreover, a recent study

showed that a supplementation of B.breve in association with breast feeding is able to reduce the

t
ip
incidence of necrotizing enterocolitis (NEC) in preterm neonates born between 28 and 34 weeks of

cr
gestation. However, further studies are needed for neonates < 28 weeks where the reduction of NEC

didn’t reach a statistical significance [154].

us
Furthermore, in vitro and murine models studies highlighted how Bifidobacteria, especially B.

longum and B. lactis strains, seem to be active against rotavirus infection [155] and could protect
an
against colorectal cancer (CRC) [156, 157]. Grounding on a previous study where the use of a
M
combination of RS and B. lactis had significantly increased the acute apoptotic response to a

genotoxic carcinogen (AARGC) in rat colon [158], Le Leu et al confirmed his protective action
d

against CRC in rats model, supporting the importance of this synbiotic in cancer prevention. The
e

mechanism(s) of actions that sustain(s) apoptosis with this symbiotic remain(s) unclear, but authors
pt

supposed the presence of immunomodulating properties derived by the interaction between B.lactis

and RS resulting in butyrate production [156]. A recent randomized, double-blind, placebo-


ce

controlled study on humans affected by colorectal polyps (no CRC) or CRC [157] highlighted how
Ac

the dietary symbiotic, B. lactis, L. Rhamnosus and oligofructose-enriched inulin, was able to reduce

colon cell proliferation and cell DNA-damage. However, the two groups responded differently to

the synbiotic, maybe because of an increased resistance to qualitative and quantitative changes of

cancer patients intestinal microflora [157].

In addition, the effects of Bifidobacteria are not limited to the gastrointestinal tract. In fact, a

possible therapeutic role of such probiotic has also been supposed in the treatment of respiratory

Page 14 of 36
pathologies [112-115], psoriasis, chronic fatigue syndrome [110], depression [132, 133] and in the

deterioration of cognitive functions [136].

CONCLUSIONS

This review shows how Actinobacteria phylum, despite it represents a minority group of

commensal bacteria, plays a pivotal role in the development and maintenance of gut homeostasis

t
ip
(Figure 1). Its involvement has been supposed in the modulation of gut permeability, immune

cr
system, metabolism and gut-brain axis. An unbalanced abundance has been evidenced in several

pathological conditions. For this reason, the interest in the use of Actinobacteria, especially of

us
Bifidobacteria family, as probiotic is constantly increasing. This group may represent a significant

part of the next-generation of probiotics with a potential efficacy both in gut diseases and extra-

intestinal disorders (Table 1).


an
M
However, further studies are needed to deeply understand the interaction between Actinobacteria

and the host and consequently their therapeutic implications in the prevention and treatment of
d

several systemic disorders with the end goal of promoting gut health.
e
pt
ce
Ac

Page 15 of 36
REFERENCES

1. Mondot S, de Wouters T, Doré J, Lepage P. The human gut microbiome and its dysfunctions.

Dig Dis 2013; 31(3-4):278-85

2. Leser TD, Molbach L: Better living through microbial action: the benefits of the mammalian

gastrointestinal microbiota on the host. Envirol Microbiol 2009; 11:2194-2206

t
ip
3. Neish AS. Microbes in gastrointestinal health and disease. Gastroenterology 2009; 136: 65-80.

cr
4. Lopetuso LR, Ianiro G, Scaldaferri F, Cammarota G, Gasbarrini A. Gut Virome and

Inflammatory Bowel Disease. Inflamm Bowel Dis 2016;22:1708-12

us
5. Donaldson GP, Lee SM, Mazmanian SK. Gut biogeography of the bacterial microbiota. Nat Rev

Microbiol 2016; 14(1):20-32


an
6. Guarner F, Malagelada JR. Gut flora in health and disease. Lancet 2003; 361:512-9
M
7. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR et al. Enterotypes of the

human gut microbiome. Nature 2011; 473: 174-180.


d

8. Segata N, Haake SK, Mannon P, Lemon KP, Waldron L, Gevers D et al. Composition of the
e

adult digestive tract bacterial microbiome based on seven mouth surfaces, tonsils, throat and
pt

stool samples. Genome Biol 2012; 13:R42.

9. D'Aversa F, Tortora A, Ianiro G, Ponziani FR, Annicchiarico BE, Gasbarrini A.Gut microbiota
ce

and metabolic syndrome. Intern Emerg Med 2013; 8 Suppl1:S11-5


Ac

10. Gallo A, Passaro G, Gasbarrini A, Landolfi R, Montalto M. Modulation of microbiota as

treatment for intestinal inflammatory disorders: An uptodate. World J Gastroenterol 2016;

22:7186-202

11. Round JL, O'Connell RM, Mazmanian SK. Coordination of tolerogenic immuneresponses by

the commensal microbiota. J Autoimmun 2010; 34:J220-5

12. Sekirov I, Russel SL, Antunes LC, Finlay BB. Gut microbiota in health and disease. Physiol

Rev 2010;90:859-904.

Page 16 of 36
13. Backhed G, Crawford PA. Coordinated regulation of the metabolome and lipidome at the host-

microbial interface. Biochem Biophys Acta 2010; 1801:240-245

14. Conly JM, Stein K, Worobetz L, Rutledge-Harding S. The contribution of vitaminK2

(menaquinones) produced by the intestinal microflora to human nutritional requirements for

vitamin K. Am J Gastroenterol 1994; 89:915-23

15. Younes H, Coudray C, Bellanger J, Demigné C, Rayssiguier Y, Rémésy C. Effects of two

t
ip
fermentable carbohydrates (inulin and resistant starch) and their combination on calcium and

cr
magnesium balance in rats. Br J Nutr 2001; 86:479-85

16. Scaldaferri F, Pizzoferrato M, Gerardi V, Lopetuso L, Gasbarrini A. The gut barrier: new

us
acquisition and therapeutic approaches. J Clin Gastroenterol 2012; 46(Suppl):S12-S17.

17. Purchiaroni F, Tortora A, Gabrielli M, Bertucci F, Gigante G, Ianiro G, OjettiV, Scarpellini E,


an
Gasbarrini A. The role of intestinal microbiota and the immune system. Eur Rev Med
M
Pharmacol Sci 2013; 17:323-33

18. Allen CA, Torres AG. Host-microbe communication within the GI tract. Adv Expt Med Biol
d

2008;635:93-101.
e

19. Maynard CL, Elson CO, Hatton RD, Weaver CT. Reciprocal interactions of theintestinal
pt

microbiota and immune system. Nature 2012; 489:231-41

20. Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H et al. Treg induction bya
ce

rationally selected mixture of Clostridia strains from the human microbiota.Nature 2013;
Ac

500:232-6

21. Galdean CM, Perdigon G. The probiotic bacterium Lactobacillus casei induces activation of the

gut mucosal immune system through innate immunity. Colin Vaccine Immunol 2006;13:219-

226.

22. Sun M, He C, Cong Y, Liu Z. Regulatory immune cells in regulation of intestinal inflammatory

response to microbiota. Mucosal Immunol 2015; 8:969-78

Page 17 of 36
23. McCarville JL, Caminero A, Verdu EF. Novel perspectives on therapeutic modulation of the gut

microbiota. Therap Adv Gastroenterol 2016; 9:580-93

24. Sartor RB. Microbial influences in inflammatory bowel disease. Gastroenterology 2008;

134:577-594.

25. De Palma G, Blennerhassett P, Lu J, Deng Y, Park AJ, Green W, et al. Microbiota and host

determinants of behavioural phenotype in maternally separated mice. Nat Commun 2015;

t
ip
6:7735

cr
26. Hill DA, Artis D. Intestinal bacteria and the regulation of immune cell homeostasis. Annu Rev

Immunol 2010; 28:623-667.

us
27. Cox A, West N and Cripps A. Obesity, inflammation, and the gut microbiota. Lancet Diabetes

Endocrinol 2015; 3:207-215


an
28. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, et al. Intestinal
M
microbiotametabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med

2013; 19:576-85
d

29. Sun J, Furio L, Mecheri R, van der Does AM, Lundeberg E, Saveanu L, et al. Pancreatic β-Cells
e

Limit Autoimmune Diabetes via an Immunoregulatory Antimicrobial Peptide Expressed under


pt

the Influence ofthe Gut Microbiota. Immunity 2015; 43:304-17

30. Scher JU, Ubeda C, Artacho A, Attur M, Isaac S, Reddy SM, et al. Decreasedbacterial diversity
ce

characterizes the altered gut microbiota in patients withpsoriatic arthritis, resembling dysbiosis
Ac

in inflammatory bowel disease. Arthritis Rheumatol 2015; 67:128-39.

31. Belizario JE, Napolitano M. Human microbes and their roles in dysbiosis, common disease, and

novel therapeutic approaches. Front Microbiol 2015; 6: 1050.

32. Adlerberth I, Carlsson B, de Man P, Jalil F, Khan SR, Larsson P, Mellander L, Svanborg C,

Wold AE, Hanson LA. Intestinal colonization with Enterobacteriaceae in Pakistani and Swedish

hospital-delivered infants. Acta Paediatr Scand. 1991;80(6–7):602–10.

Page 18 of 36
33. Hesla HM, Stenius F, Jaderlund L, Nelson R, Engstrand L, Alm J, Dicksved J. Impact of

lifestyle on the gut microbiota of healthy infants and their mothers - the ALADDIN birth cohort.

FEMS Microbiol Ecol. 2014;90(3):791–801.

34. Mitsou EK, Kirtzalidou E, Oikonomou I, Liosis G, Kyriacou A. Fecal microflora of Greek

healthy neonates. Anaerobe. 2008;14(2):94–101.

35. Dogra S, Sakwinska O, Soh SE, Ngom-Bru C, Bruck WM, Berger B, Brussow H, Lee YS, Yap

t
ip
F, Chong YS et al.: Dynamics of infant gut microbiota are influenced by delivery mode and

cr
gestational duration and are associated with subsequent adiposity. MBio 2015, 6(1).doi:

10.1128/mBio.02419-14.

us
36. Huurre A, Kalliomaki M, Rautava S, Rinne M, Salminen S, Isolauri E. Mode of delivery -

Effects on gut microbiota and humoral immunity. Neonatology 2008;93(4):236–40.


an
37. Kabeerdoss J, Ferdous S, Balamurugan R, Mechenro J, Vidya R, Santhanam S, Jana AK,
M
Ramakrishna BS. Development of the gut microbiota in southern Indian infants from birth to 6

months: a molecular analysis. J Nutr Sci 2013;2:e18.


d

38. Gronlund MM, Lehtonen OP, Eerola E, Kero P. Fecal microflora in healthy
e

infants born by different methods of delivery: permanent changes in intestinal flora after
pt

cesarean delivery. J Pediatr Gastroenterol Nutr. 1999; 28(1):19–25.

39. Bezirtzoglou E. The intestinal microflora during the first weeks of life. Anaerobe. 1997;3(2–
ce

3):173–7.
Ac

40. Yap GC, Chee KK, Hong PY, Lay C, Satria CD, Sumadiono AA, Soenarto Y, Haksari EL, Aw

M, Shek LPC, et al. Evaluation of stool microbiota signatures in two cohorts of Asian

(Singapore and Indonesia) newborns at risk of atopy. Bmc Microbiol. 2011;11:193.

41. Dethlefsen L, Huse S, Sogin ML, Relman DA. The pervasive effects of an antibiotic on the

human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008;6(11):e280.

Page 19 of 36
42. Penders J, Thijs C, Vink C, Stelma FF, Snijders B, Kummeling I, van den Brandt PA,

Stobberingh EE. Factors influencing the composition of the intestinal microbiota in early

infancy. Pediatrics. 2006;118(2):511–21.

43. Bezirtzoglou E, Tsiotsias A, Welling GW. Microbiota profile in feces of breast- and formula-fed

newborns by using fluorescence in situ hybridization (FISH). Anaerobe. 2011;17(6):478–82.

44. Underwood MA, German JB, Lebrilla CB, Mills DA. Bifidobacterium longum subspecies

t
ip
infantis: champion colonizer of the infant gut. Pediatr Res. 2015; 77(1–2):229–35.

cr
45. Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial

diversity in the human intestine. Cell 2006; 124:837-848.

us
46. D’Argenio V, Salvatore F. The role of the gut microbiome in the healthy adult status. Clin Chim

Acta 2015; 451(PtA):97-102.


an
47. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill SR, Nelson KE,
M
Relman DA. Diversity of the human intestinal microbial flora. Science. 2005; 308:1635-16388.

48. Drago L, Toscano M, Rodighiero V, De Vecchi E, Mogna G. Cultivable and pyrosequenced


d

fecal microflora in centenarians and young subjects. J Clin Gastroenterol 2012;46(suppl):S81-


e

84.
pt

49. Ashida H, Ogawa M, Kim M, Mimuro H, Sasakawa C. Bacterial and host interactions in the gut

epithelial barrier. Nature 2012; 489:231-241.


ce

50. Hardy H, Harris J, Lyon E, Beal J, Foey AD. Probiotics, prebiotics and immunomodulation of
Ac

gut mucosal defences: homeostasis and immunopathology.Nutrients 2013; 5:1869-912.

51. Fukuda S, Toh H, Taylor TD, Ohno H, Hattori M. Acetate-producing bifidobacteria protect the

host from enteropathogenic infection via carbohydrate transporters. Gut Microbes. 2012; 3:449-

54.

52. Scott KP, Martin JC, Duncan SH, Flint HJ. Prebiotic stimulation of human colonic butyrate-

producing bacteria and bifidobacteria, in vitro. FEMS Microbial Ecol 2014;87:30-40

Page 20 of 36
53. Willemsem LE, Koetsier MA, van Deventer SJ et al. Short chain fatty acids stimulate epithelial

mucin 2 expression through differential effects on prostaglandin E(1) and E(2) production by

intestinal myofibroblasts. Gut 2003;52:1442-47

54. Gaudier E, Jarry A, Blottiere HM et al. Butyrate specifically modulates MUC gene expression in

intestinal epithelial goblet cells deprived of glucose. Am J Physiol Gastrointest Liver Physiol

2004;287:G1168-74

t
ip
55. Gendler SJ,Spicer AP. Epithelial mucin genes. Annu Rev Physiol 1995; 57:607-34

cr
56. Hamer HM, Jonkers D, Venema K, Vanhoutviny S, Troost FJ.Review article: the role of

butyrate on colonic function. Aliment Pharmacy There 2008; 27:104-119.

us
57. Barrett KE. A new twist on trefoils. Focus on "TFF3 modulates NF-{kappa}B and a novel

regulatory molecule of NF-{kappa}B in intestinal epithelial cells via a mechanism distinct from
an
TNF-{alpha}". Am J Physiol Cell Physiol 2005; 289:C1069-71
M
58. Song M, Xia B, Li J. Effects of topical treatment of sodium butyrate and 5-aminosalicylic acid

on expression of trefoil factor 3, interleukin 1beta, and nuclear factor kappaB in trinitrobenzene
d

sulphonic acid induced colitis in rats. Postgrad Med J 2006; 82:130-5


e

59. Arvans DL, Vavricka SR, Ren H, et al. Luminal bacterial flora determines physiological
pt

expression of intestinal epithelial cytoprotective heat shock proteins 25 and 72. Am J Physiol

Gastrointest Liver Physiol 2005; 288: G696–704


ce

60. Ren H, Musch MW, Kojima K, et al. Short-chain fatty acids induce intestinal epithelial heat
Ac

shock protein 25 expression in rats and IEC 18 cells. Gastroenterology 2001; 121: 631–9

61. Peng L, He Z, Chen W, et al. Effects of butyrate on intestinal barrier function in a caco-2 cell

monolayer model of intestinal barrier. Pediatr Res 2007; 61: 37–41

62. Mariadason JM, Kilias D, Catto-Smith A, et al. Effect of butyrate on paracellular permeability

in rat distal colonic mucosa ex vivo. J Gastroenterol Hepa- tol 1999; 14: 873–9

Page 21 of 36
63. Davila AM, Blachier F, Gotteland M, Andriamihaja M, Benetti PH, Sanz Y, Tomé D. Intestinal

luminal nitrogen metabolism: Role of the gut microbiota and consequences for the host.

Pharmacol Res 2012; 68:95-107.

64. Bernalier-Donadille A. Fermentative metabolism by the human gut microbiota. Gastroenterol

Clin Biol 2010; 34 Suppl 1:S16-22

65. CummingsJand MacfarlaneG. The control and consequences of bacterial fermentation in the

t
ip
human colon. J. Appl Microbiol 1991; 70:443–459

cr
66. Hamer HM, De Preter V, Windey Kand Verbeke K. Functional analysis of colonic bacterial

metabolism: relevant to health? Am J Physiol Gastrointest Liver Physiol 2012; 302, G1–9.

us
67. Ponziani FR, Binda C, Gasbarrini A. How to modulate gut microbiota:diet, pre-probiotic or

antibiotics? Minerva Gastroenterol Dietol 2017; 8:1026-1025.


an
68. Bindels LB, Delzenne NM, Cani PD, Walter J. Towards a more comprehensive concept for
M
prebiotics. Nat Rev Gastroenterol Hepatol 2015; 12:303-310.

69. O’Callaghan A, van Sinderen D. Bifidobacteria and their role as members of the human gut
d

microbiota. Front Microbiol 2016; 7:925.


e

70. Rivière A, Gagnon M, Weckx S, Roy D, De Vuyst L. Mutual cross-feeding interactions between
pt

Bifidobacterium longum NCC2705 and eubacterium rectale ATCC336656 explain the

bifidogenic and butyrogenic effects of arabinoxylan-oligosaccharides. Appl Environ Microbiol


ce

2015; 80:204-217.
Ac

71. Clausen MR, Mortensen PB: Kinetic studies on colonocyte metabolism of short chain fatty acids

and glucose in ulcerative colitis. Gut 1995; 37:684-689.

72. Macfarlane GT, Englyst HN. Starch utilization by the human large intestinal microflora. J Appl

Bacteriol 1986;60:195–201.

73. Ryan SM, Fitzgerald GF, van Sinderen D. Screening for and identification of starch-,

amylopectin-, and pullulan-degrading activities in bifidobacterial strains. Appl Environ

Microbiol. 2006;72(8):5289–96.

Page 22 of 36
74. Salyers AA, West SE, Vercellotti JR, Wilkins TD. Fermentation of mucins and plant

polysaccharides by anaerobic bacteria from the human colon. Appl Environ Microbiol.

1977;34(5):529–33.

75. Pokusaeva K, Fitzgerald GF and Van Sinderen D. Carbohydrate metabolism in Bifidobacteria.

Genes Nutr 2011; 6:285-306.

76. El Kaoutari A, Armougom F, Gordon JI, Raoult D, Henrissat B. The abundance and variety of

t
ip
carbohydrate-active enzymes in the human gut microbiota. Nat Rev Microbol 2013; 11:497-504.

cr
77. Milani C, Lugli AG, Duranti S, Turroni F, Mancabelli L, Ferrario C et al. Bifidobacteria exhibit

social behaviour through carbohydrate resource sharing in the gut. Sci Rep 2015; 5:15782.

us
78. Raimondi S, Amaretti A, Leonardi L, Quartieri A, Gozzoli C, Rossi M. Conjugated linoileic

acid production by Bifidobacteria: screening, kinetic, and composition. Biomed Res Int 2016;

2016:8654317.
an
M
79. Kim JH, Kim Y, Kim YJ, Park Y. Conjugated linoleic acids: potential health benefits as a

functional food ingredient. Annu Rev Food and Sci Techol 2016; 7:221-244.
d

80. Cotillard A, Kennedy SP, Kong LC, Prifti E, Pons N, Le Chatelier E et al. Dietary intervention
e

impact on gut microbial gene richness. Nature 2013; 500:585-8


pt

81. TurnbaughPJ, BäckhedF, FultonLand Gordon JI. Diet-induced obesity is linked to marked but

reversible alterations in the mouse distal gut microbiome. Cell Host Microbe 2008; 3:213–223
ce

82. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Bewtra M, Knights D,
Ac

Walters WA, Knight R, et al. Linking long-term dietary patterns with gutmicrobial enterotypes.

Science 2011; 334:105–108.

83. Teixeira TFS, Grze_skowiak ŁM, Salminen S, Laitinen K, Bressan J, Gouveia Peluzio Mdo C.

Faecal levels of Bifidobacterium and Clostridium coccoides but not plasma lipopolysaccharide

are inversely related to insulin andHOMA index in women. Clin Nutr 2013; 32:1017–1022.

Page 23 of 36
84. Zimmer J, Lange B, Frick JS, Sauer H, Zimmermann K, Schwiertz A, Rusch K, et al. A vegan

or vegetarian diet substantially alters the human colonic faecal microbiota. Eur J Clin Nutr 2012;

66:53–60.

85. Cani PD, Neyrinck AM, Fava F, Knauf C, Burcelin RG, Tuohy KM, Gibson GR, Delzenne NM.

Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in

mice through a mechanism associated with endotoxaemia. Diabetologia 2007; 50:2374–2383.

t
ip
86. Geurts L, Neyrinck AM, Delzenne NM, Knauf C, Cani PD. Gut microbiota controls adipose

cr
tissue expansion, gut barrier and glucose metabolism: novel insights into molecular targets and

interventions using prebiotics. Benef Microbes 2014; 5:3–17.

us
87. Barczynska R, Kapusniak J, Litwin M, Slizewska K, Szalecky M. Dextrins from maize starch as

substances activating the growth of Bacteroidetes and Actinobacteria simultaneosly inhibiting


an
the growth of Firmicutes, responsible for the occurrence of obesity. Plant Foods Hum Nutr
M
2016; 71:190-196.

88. Barrett JS, Gearry RB, Muir JG, Irving PM, Rose R, Rosella O, et al. Dietary poorly absorbed,
d

short-chain carbohydrates increase delivery of water and fermentable substrates to the proximal
e

colon. Aliment Pharmacol Ther. 2010;31:874-82


pt

89. De Giorgio R, Volta U, Gibson PR. Sensitivity to wheat, gluten and FODMAPs in IBS: facts or

fiction? Gut. 2016;65:169-78


ce

90. McIntosh K, Reed DE, Schneider T, Dang F, Keshteli AH, De Palma G, Madsen K, Bercik P,
Ac

Vanner S. FODMAPs alter symptoms and the metabolome of patients with IBS: a randomised

controlled trial. Gut. 2017 Jul;66(7):1241-1251.

91. Staudacher HM, Lomer MC, Anderson JL, et al. Fermentable carbohydrate restriction reduces

luminal bifidobacteria and gastrointestinal symptoms in patients with irritable bowel syndrome.

J Nutr 2012;142:1510–18.

Page 24 of 36
92. Bennet SMP, Böhn L, Störsrud S, Liljebo T, Collin L, Lindfors P, Törnblom H, Öhman L,

Simrén M. Multivariate modelling of faecal bacterial profiles of patients with IBS predicts

responsiveness to a diet low in FODMAPs. Gut 2017. [Epub ahead of print]

93. O’Mahony L, McCarthy J, Kelly P, et al. Lactobacillus and Bifidobacterium in irritable bowel

syndrome: Symptom responses and relationship to cytokine profiles. Gastroenterology

2005;128:541–51.

t
ip
94. Whorwell PJ, Altringer L, Morel J, et al. Efficacy of an encapsulated probiotic Bifidobacterium

cr
infantis 35624 in women with irritable bowel syndrome. Am J Gastroenterol 2006;101:1581–90.

95. Guyonnet D, Chassany O, Ducrotte P, et al. Effect of a fermented milk containing

us
Bifidobacterium animalis DN-173 010 on the health-related quality of life and symptoms in

irritable bowel syndrome in adults in primary care: a multicentre, randomized, double-blind,


an
controlled trial. Aliment Pharmacol Ther 2007;26:475–86.
M
96. Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal

Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science.


d

2015;350:1084-9.
e

97. Duca FA, Sakar Y, Covasa M. The modulatory role of high fat feeding on gastrointestinal
pt

signals in obesity. J Nutr Biochem 2013; 24:1663-77.

98. Scarpellini E, Tack J. Obesity and metabolic syndrome: an inflammatory condition. Dig Dis
ce

2012; 30:148-153.
Ac

99. Gauffin Cano P, Santacruz A, Trejo MF, Sanz Y. Bifidobacterium CECT 7765 improves

metabolic and immunological alterations associated with obesity in high-fat diet-fed mice.

Obesity 2013; 21:2310-21.

100. Cani PD, Possemiers S, Van de Wiele T et al. Changes in gut microbiota control

inflammation in obese mice through a mechanism involving GLP-2 driven improvement of gut

permeability. Gut 2009; 58:1091-1103.

Page 25 of 36
101. Zhou Q, Leeman SE, Amar S. Signaling mechanism involved in altered function of

macrophages from diet-induced obese mice affect immune responces. Proc Natl Acad Sci USA

2009; 106:10740-10745.

102. Macia L, Declare M, Abboud G et al. Impairment of dendritic cell functionally and steady-

state number in obese mice. J Immunol 2006; 177: 5997-6006.

103. O’Mahony C, Scully P, O’Mahony D, Murphy S, O’Brien F, Lyons A et al. Commensal-

t
ip
induced regulatory T cells mediate protection against pathogen-stimulated NF-kappaB

cr
activation. PLoS Pathog 2008; 4:e1000112.

104. Lyons A, O’Mahony D, O’Brien F, MacSharry J, Sheil B, Ceddia M et al. Bacterial strain-

us
specific induction of Foxp3+ T regulatory cells is protective in murine allergy models. Clin Exp

Allergy 2010; 40:811-19.

105.
an
O'Mahony L, Feeney M, O'Halloran S, Murphy L, Kiely B, Fitzgibbon J, Lee G, O'Sullivan
M
G, Shanahan F, Collins JK. Probiotic impact on microbial flora, inflammation and tumour

development in IL-10 knockout mice. Aliment Pharmacol Ther 2001; 15:1219-25.


d

106. Konieczna P, Akdis CA, Quigley EM, Shanahan F, O'Mahony L. Portrait of an


e

immunoregulatory Bifidobacterium. Gut Microbes 2012; 3:261-6.


pt

107. McCarthy J, O'Mahony L, O'Callaghan L, Sheil B, Vaughan EE, Fitzsimons N, Fitzgibbon

J, O'Sullivan GC, Kiely B, Collins JK, Shanahan F. Double blind, placebo controlled trial of two
ce

probiotic strains in interleukin 10 knockout mice and mechanistic link with cytokine balance.
Ac

Gut 2003; 52:975-80.

108. McKernan DP, Fitzgerald P, Dinan TG, Cryan JF. The probiotic Bifidobacterium infantis

35624 displays visceral antinociceptive effects in the rat. Neurogastroenterol Motil 2010;

22:1029-35, e268.

109. Konieczna P, Groeger D, Ziegler M, Frei R, Ferstl R, Shanahan F, Quigley EM, Kiely B,

Akdis CA, O'Mahony L. Bifidobacterium infantis 35624 administration induces Foxp3 T

Page 26 of 36
regulatory cells in human peripheral blood: potential role for myeloid and plasmacytoid

dendritic cells. Gut 2012; 61:354-66.

110. Groeger D, O'Mahony L, Murphy EF, Bourke JF, Dinan TG, Kiely B, Shanahan F, Quigley

EM. Bifidobacterium infantis 35624 modulates host inflammatory processes beyond the gut.

Gut Microbes 2013; 4:325-39.

111. Tojo R, Suárez A, Clemente MG, de los Reyes-Gavilán CG, Margolles A, Gueimonde M,

t
ip
Ruas-Madiedo P. Intestinal microbiota in health and disease: role of bifidobacteria in gut

cr
homeostasis. World J Gastroenterol 2014; 20:15163-76.

112. van der Aa LB, van Aalderen WM, Heymans HS, Henk Sillevis Smitt J, Nauta AJ, Knippels

us
LM, Ben Amor K, Sprikkelman AB; Synbad Study Group. Synbiotics prevent asthma-like

symptoms in infants with atopic dermatitis. Allergy 2011;66:170-7.

113.
an
Xiao JZ, Kondo S, Yanagisawa N, Takahashi N, Odamaki T, Iwabuchi N, Miyaji K,
M
Iwatsuki K, Togashi H, Enomoto K, Enomoto T. Probiotics in the treatment of Japanese cedar

pollinosis: a double-blind placebo-controlled trial. Clin Exp Allergy 2006 Nov;36(11):1425-35.


d

114. Singh A, Hacini-Rachinel F, Gosoniu ML, Bourdeau T, Holvoet S, Doucet-Ladeveze R,


e

Beaumont M, Mercenier A, Nutten S. Immune-modulatory effect of probiotic Bifidobacterium


pt

lactis NCC2818 in individuals suffering from seasonal allergic rhinitis to grass pollen: an

exploratory, randomized, placebo-controlled clinical trial. Eur J Clin Nutr 2013; 67:161-7.
ce

115. Odamaki T, Xiao JZ, Iwabuchi N, Sakamoto M, Takahashi N, Kondo S, Miyaji K, Iwatsuki
Ac

K, Togashi H, Enomoto T, Benno Y. Influence of Bifidobacterium longum BB536 intake on

faecal microbiota in individuals with Japanese cedar pollinosis during the pollen season. J Med

Microbiol 2007; 56:1301-8.

116. Mayer EA, Tillisch K, Gupta A. Gut/brain axis and the microbiota. J Clin Invest 2015; 125:

926-938.

117. Grenham S, Clarke G, Cryan JF, Dinan TG. Brain-gut-microbe communication in health

and disease. Front Physiol 2011; 2: 94.

Page 27 of 36
118. Rhee SH, Pothoulakis C, Mayer EA. Principles and clinical implications of the brain-gut-

enteric microbiota axis. Nat Rev Gastroenterol Hepatol 2009; 6: 306-314.

119. Galland L. The gut microbiome and the brain. J Med Food 2014; 17: 1261-1272.

120. Lyte M. Microbial endocrinology: Host-microbiota neuroendocrine interactions influencing

brain and behavior. Gut Microbes 2014; 5: 381-389.

121. Borre YE, O’Keeffe GW, Clarke G, Stanton C, Dinan TG, Cryan JF. Microbiota and

t
ip
neurodevelopmental windows: implications for brain disorders. Trends Mol Med 2014; 20: 509-

cr
518.

122. Dinan TG, Cryan JF. Melancholic microbes: a link between gut microbiota and depression?

us
Neurogastroenterol Motil 2013; 25: 713-719.

123. Bonaz BL, Bernstein CN. Brain-gut interactions in inflammatory bowel disease.

Gastroenterology 2013; 144: 36-49.


an
M
124. Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER, McCue T, Codelli JA, Chow J,

Reisman SE, Petrosino JF, Patterson PH, Mazmanian SK. Microbiota modulate behavioral and
d

physiological abnormalities associated with neurodevelopmental disorders.Cell 2013; 155:


e

1451-1463.
pt

125. Forsythe P, Bienenstock J, Kunze WA. Vagal pathways for microbiome-brain-gut axis

communication. Adv Exp Med Biol 2014; 817: 115-133.


ce

126. Mulak A, Bonaz Bruno. Brain-gut-microbiota axis in Parkinson’s disease. World J


Ac

Gastroenterol 2015; 21:10609-10620.

127. Bailey MT, Coe CL. Maternal separation disrupts the integrity of the intestinal microflora in

infant rhesus monkeys. Dev Psychobiol 1999;35:146-55.

128. Bailey MT, Lubach GR, Coe CL. Prenatal stress alters bacterial colonization of the gut in

infant monkeys. J Pediatr Gastroenterol Nutr 2004; 38:414–421.

129. Tannock GW, Savage DC. Influences of dietary and environmental stress on microbial

populations in the murine gastrointestinal tract. Infect Immun 1974; 9:591-8.

Page 28 of 36
130. Suzuki K, Harasawa R, Yoshitake Y, Mitsuoka T. Effects of crowding and heat stress on

intestinal flora, body weight gain, and feed efficiency of growing rats and chicks. Nippon

Juigaku Zasshi 1983; 45:331–338.

131. Goncharova GI, Liz’ko NN, Liannaia AM, Shilov VM, Spitsa TI. Bifidobacterium flora

status of cosmonauts before and after com- pleting space flights. Kosm Biol Aviakosm Med

1981; 15:14–18.

t
ip
132. Desbonnet L, Garrett L, Clarke G, Bienenstock J, Dinan TG. The probiotic Bifidobacteria

cr
infantis: an assessment of potential antidepressant properties in the rat. J Psych Res 2009;

43:164-174.

us
133. Desbonnet L, Garrett L, Clarke G, Kiely B, Cryan JF, Dinan TG. Effects of the probiotic

bifidobacterium infantis in the maternal separation model of depression. Neuroscience 2010;

170:1179-1188.
an
M
134. Lyte M, Chapel A, Lyte JM, Ai Y, Proctor A, Jane JL, Phillips GJ. Resistant starch alters

the microbiota-gut-brain axis: implications for dietary modulation of behavior. PloS ONE 2016;
d

11:e0146406.
e

135. Tachon S, Zhou J, Keenan M, Martin R, Marco ML. The intestinal microbiota in aged mice
pt

is modulated by dietary resistant starch and correlated with improvements in host responses.

FEMS Microbiol Ecol. 2013; 83:299–309.


ce

136. Distrutti E, O’Reilly J-A, McDonald C, Cipriani S, Renga B, et al. Modulation of Intestinal
Ac

Microbiota by the Probiotic VSL#3 Resets Brain Gene Expression and Ameliorates the Age-

Related Deficit in LTP. PLoS ONE 2014; 9: e106503.

137. Leclercq S, Matamoros S, Cani PD, Neyrinck AM, Jamar F, Stärkel P, Windey K,Tremaroli

V, Bäckhed F, Verbeke K, de Timary P, Delzenne NM. Intestinalpermeability, gut-bacterial

dysbiosis, and behavioral markers of alcohol-dependence severity. Proc Natl Acad Sci U S A

2014;111:e4485-93.

Page 29 of 36
138. Gionchetti P, Rizzello F, Helwig U, Venturi A, Lammers KM, Brigidi P, et al. Prophylaxis

of pouchitis onset withprobiotic therapy: a double-blind, placebo-controlled trial.

Gastroenterology 2003; 124:1202-9.

139. Mimura T, Rizzello F, Helwig U, Poggioli G, Schreiber S, Talbot IC, et al. Once daily high

dose probiotic therapy(VSL#3) for maintaining remission in recurrent or refractory pouchitis.

Gut 2004; 53:108-14.

t
ip
140. Sood A, Midha V, Makharia GK, Ahuja V, Singal D, Goswami P, Tandon RK. The

cr
probiotic preparation, VSL#3 induces remission in patients with mild-to-moderately active

ulcerative colitis. Clin Gastroenterol Hepatol 2009; 7:1202-9.

us
141. Mardini HE, Grigorian AY. Probiotic mix VSL#3 is effective adjunctive therapy for mild to

moderately active ulcerative colitis: a meta-analysis. Inflamm Bowel Dis 2014; 20:1562-7.

142.
an
Tursi A, Brandimarte G, Papa A, Giglio A, Elisei W, Giorgetti GM, Forti G, Morini S,
M
Hassan C, Pistoia MA, Modeo ME, Rodino' S, D'Amico T, Sebkova L, Sacca’ N, Di Giulio E,

Luzza F, Imeneo M, Larussa T, Di Rosa S, Annese V, Danese S, Gasbarrini A. Treatment of


d

relapsing mild-to-moderate ulcerative colitis with the probiotic VSL#3 as adjunctive to a


e

standard pharmaceutical treatment: a double-blind, randomized, placebo-controlled study. Am J


pt

Gastroenterol 2010; 105:2218-27.

143. Miele E, Pascarella F, Giannetti E, Quaglietta L, Baldassano RN, Staiano A. Effect of a


ce

probiotic preparation (VSL#3) on induction and maintenance of remission in children with


Ac

ulcerative colitis. Am J Gastroenterol 2009;104: 437-43.

144. Abu-Shanab A, Quigley EM. The role of the gut microbiota in nonalcoholic fatty liver

disease. Nat Rev Gastroenterol Hepatol 2010; 7:691-701.

145. Machado MV, Cortez-Pinto H. Gut microbiota and nonalcoholic fatty liverdisease. Ann

Hepatol 2012; 11:440-9.

Page 30 of 36
146. Selinger CP, Bell A, Cairns A, Lockett M, Sebastian S, Haslam N. Probiotic VSL#3

prevents antibiotic-associated diarrhoea in a double-blind, randomized, placebo-controlled

clinical trial. J Hosp Infect 2013; 84:159-65.

147. Ishikawa H, Matsumoto S, Ohashi Y, Imaoka A, Setoyama H, Umesaki Y, Tanaka R, Otani

T. Beneficial effects of probiotic bifidobacterium and galacto-oligosaccharide in patients with

ulcerative colitis: a randomized controlled study. Digestion. 2011;84:128-33.

t
ip
148. Kajander K, Hatakka K, Poussa T, Färkkilä M, Korpela R. A probiotic mixture alleviates

cr
symptoms in irritable bowel syndrome patients: a controlled 6-month intervention. Aliment

Pharmacol Ther 2005; 22:387-94.

us
149. Kajander K, Myllyluoma E, Rajilić-Stojanović M, Kyrönpalo S, Rasmussen M, Järvenpää

S, Zoetendal EG, de Vos WM, Vapaatalo H, Korpela R. Clinical trial: multispecies probiotic
an
supplementation alleviates the symptoms of irritable bowel syndrome and stabilizes intestinal
M
microbiota. Aliment Pharmacol Ther 2008; 27:48-57.

150. Guandalini S, Magazzù G, Chiaro A, La Balestra V, Di Nardo G, Gopalan S, Sibal A,


d

Romano C, Canani RB, Lionetti P, Setty M. VSL#3 improves symptoms in children with
e

irritable bowel syndrome: a multicenter, randomized, placebo-controlled, double-blind,


pt

crossover study. J Pediatr Gastroenterol Nutr 2010; 51:24-30.

151. Kleessen B, Sykura B, Zunft HJ, Blaut M. Effects of inulin and lactose on fecal microflora,
ce

microbial activity, and bowel habit in elderly constipated persons. Am J Clin Nutr 1997;
Ac

65:1397-402.

152. Leahy SC, Higgins DG, Fitzgerald GF, van Sinderen D. Getting better with bifidobacteria. J

Appl Microbiol 2005; 98:1303-15.

153. Corrêa NB, Péret Filho LA, Penna FJ, Lima FM, Nicoli JR. A randomized formula

controlled trial of Bifidobacterium lactis and Streptococcus thermophilus for prevention of

antibiotic-associated diarrhea in infants. J Clin Gastroenterol 2005; 39:385-9.

Page 31 of 36
154. Patole SK, Rao SC, Keil AD, Nathan EA, Doherty DA, Simmer KN. Benefits of

Bifidobacterium breve M-16V Supplementation in Preterm Neonates - A Retrospective Cohort

Study. PLoS One. 2016;11:e0150775.

155. Chenoll E, Rivero M, Codoñer FM, Martinez-Blanch JF, Ramón D, Genovés S, Moreno

Muñoz JA. Complete Genome Sequence of Bifidobacterium longum subsp. infantis Strain

CECT 7210, a Probiotic Strain Active against Rotavirus Infections. Genome Announc 2015

t
ip
;3(2). pii: e00105-15.

cr
156. Le Leu RK, Hu Y, Brown IL, Woodman RJ, Young GP. Synbiotic intervention of

Bifidobacterium lactis and resistant starch protects against colorectal cancer development in

us
rats. Carcinogenesis. 2010; 31:246-51.

157. Rafter J, Bennett M, Caderni G, Clune Y, Hughes R, Karlsson PC, Klinder A, O'Riordan M,
an
O'Sullivan GC, Pool-Zobel B, Rechkemmer G, Roller M, Rowland I, Salvadori M, Thijs H, Van
M
Loo J, Watzl B, Collins JK. Dietary synbiotics reduce cancer risk factors in polypectomized and

colon cancer patients. Am J Clin Nutr 2007;85:488-96.


d

158. Le Leu,R.K. et al. (2005) A synbiotic combination of resistant starch and Bifidobacterium
e

lactis facilitates apoptotic deletion of carcinogen-damaged cells in rat colon. J. Nutr., 135, 996–
pt

1001
ce
Ac

Page 32 of 36
FIGURE LEGEND

Figure 1

Actinobacteria phylum groups four main families: Bifidobacteria, Propionibacteria, Corynebacteria,

Stremptomyces. Actinobacteria exert crucial physiological functions in the gut. FOS: fructo-

oligosaccharides, GOS: galacto-oligosaccharides, XOS: xylo-oligosaccharides, BDNF: brain-

derived neurotrophic factor.

t
ip
cr
us
an
M
e d
pt
ce
Ac

Page 33 of 36
Table 1

Potential modulatory and therapeutic role of Actinobacteria in intestinal and extra-intestinal

diseases.

INTESTINAL DISEASES EXTRA-INTESTINAL DISEASES


UC: prevention of pouchitis relapse, NASH

t
maintenance of remission and

ip
treatment of mild-to-moderate UC
IBS RESPIRATORY PATHOLOGIES

cr
CONSTIPATION PSORIASIS

us
ANTIBIOTIC-ASSOCIATED CHRONIC FATIGUE SYNDROME
DIARHEA
NEC DEPRESSION
COLORECTAL CANCER
an
COGNITIVE DETERIORATION

RCU/ UC: ulcerative colitis, IBS: irritable bowel syndrome, NEC: necrotizing entero-colitis,
M
NASH: non-alcoholic steato-hepatitis.
e d
pt
ce
Ac

Page 34 of 36
CONFLICT OF INTEREST

Authors declare no conflict of interest.

t
ip
cr
us
an
M
e d
pt
ce
Ac

Page 35 of 36
Figure 1

i
cr
PHYSIOLOGICAL FUNCTIONS

us
an
Immunological function:
Gut Barrier: -maintenance of gut permeability
-production of acetate
-down-regulation of inflammation by

M
-production of lactate that is used by
production of IL-4 and IL-13
“lactate-utilizer” for the production of
-induction of regulatory T-cells
butyrate

ed
-regulation of Th-2 immunoresponse
-maintenance of gut permeability

pt
ce
Metabolism: Gut-Brain Axis:
-production of acetate -potential anti-depressant effects with the
elevation of tryptophan levels
Ac

-biodegradation of resistant starch with


production of FOS, GOS, XOS, inulin, -regulation of mediators involved in
arabinoxilan synaptic plasticity like BDNF, synapsin,
-formation of conjugated linoleic acids Actinobacteria syntaxin
-modulation of hippocampal molecules such
as CD68 and CD11b
Bifidobacteria Propionibacteria

Page 36 of 36
Corynebacteria Streptomyces

You might also like