You are on page 1of 10

Journal of Microbiology (2018) Vol. 56, No. 3, pp.

189–198 eISSN 1976-3794


DOI 10.1007/s12275-018-8049-8 pISSN 1225-8873

REVIEW

Current understanding of microbiota- and dietary-therapies for


treating inflammatory bowel disease

factors involved in IBD and studies attempted to treat IBD


Taekil Eom1, Yong Sung Kim2, with probiotics. We also discuss the potential use of micro-
3 4,5,6
Chang Hwan Choi , Michael J. Sadowsky , biota therapies as one promising approach in treating IBD.
1,7*
and Tatsuya Unno As therapies based on the modulation of gut microbiota be-
comes more common, future studies should include indivi-
1
Subtropical/tropical Organism Gene Bank, Jeju National University, dual gut microbiota differences to develop personalized ther-
Jeju 63243, Republic of Korea apy for IBD.
2
Department of Gastroenterology, Wonkwang Digestive Disease Research
Institute, Wonkwang University Sanbon Hospital, Gunpo 15865,
Republic of Korea Keywords: dysbiosis, gut microbiota, inflammatory bowel
3
Department of Internal Medicine, Chung-Ang University College of disease, probiotics, prebiotics, short chain fatty acids
Medicine, Seoul 06974, Republic of Korea
4
BioTechnology Institute, University of Minnesota, St. Paul, Minnesota
55108, USA
5
Department of Soil, Water, and Climate, University of Minnesota, St. Introduction
Paul, Minnesota 55108, USA
6
Department of Plant and Microbial Biology, University of Minnesota, Inflammatory bowel disease (IBD) is a chronic immune-me-
St. Paul, Minnesota 55108, USA
7
Faculty of Biotechnology, School of life sciences, SARI, Jeju National diated inflammatory bowel condition that consists of two
University, Jeju 63243, Republic of Korea main diseases, ulcerative colitis (UC) and Crohn’s disease
(CD). IBD is a widespread disease in the western world. In
(Received Jan 27, 2018 / Revised Feb 6, 2018 / Accepted Feb 11, 2018) Europe, the highest annual incidence of UC and CD were
24.3 and 12.7 per 100,000 person-years, respectively (Molo-
Inflammatory bowel disease (IBD) is a result of chronic in- decky et al., 2012). According to the US Centers for Disease
flammation caused, in some part, by dysbiosis of intestinal Control and Prevention (CDC), IBD reports have been in-
microbiota, mainly commensal bacteria. Gut dysbiosis can creasing in the US, and it has been estimated that 1.3% of
be caused by multiple factors, including abnormal immune U.S. adults (~ 3 million people) reported an IBD diagnosis,
responses which might be related to genetic susceptibility, either as CD or UC. It has also seen rapid increase in Asian
infection, western dietary habits, and administration of anti- countries such as Japan, South Korea and China where wes-
biotics. Consequently, the disease itself is characterized as ternization has progressed (Yang et al., 2008; Ng et al.,
having multiple causes, etiologies, and severities. Recent stu- 2013b). IBD can occur in all ages but is usually diagnosed
dies have identified > 200 IBD risk loci in the host. It has been between 15 and 40 years of age (Johnston and Logan, 2008).
postulated that gut microbiota interact with these risk loci The manifestation of symptoms depends on the disease and
resulting in dysbiosis, and this subsequently leads to the de- the body site involved. For example, UC normally affects
velopment of IBD. Typical gut microbiota in IBD patients only the large intestine and usually presents with a superfi-
are characterized with decrease in species richness and many cial ulcer due to localized inflammatory reaction in the mu-
of the commensal, and beneficial, fecal bacteria such as Fir- cosa and submucosa. Symptoms include bloody diarrhea,
micutes and Bacteroidetes and an increase or bloom of Pro- mucus discharge, and abdominal pain. In patients with pro-
teobacteria. However, at this time, cause and effect relation- ctitis only, disease presentation usually involves bloody stools,
ships have not been rigorously established. While treatments blood mucus, and tenesmus (urgency to void), but abdomi-
of IBD usually includes medications such as corticosteroids, nal pain is rare. In contrast, CD occurs in the entire gastro-
5-aminosalicylates, antibiotics, immunomodulators, and anti- intestinal tract (mouth to anus) and involves the whole in-
TNF agents, restoration of gut dysbiosis seems to be a safer testinal layer. Presentations include deep ulceration, stenosis,
and more sustainable approach. Bacteriotherapies (now called and in some patients the presence of internal and external
microbiota therapies) and dietary interventions are effective fistula. Symptoms include abdominal pain, low grade fever,
way to modulate gut microbiota. In this review, we summarize fatigue, weight loss, diarrhea, and sometimes bloody stools.
Patients with IBD also exhibited extra-intestinal symptoms
*For correspondence. E-mail: tatsu@jejunu.ac.kr; Tel.: +82-64-754-3354; such as arthritis, ankylosing spondylitis, sacroiliitis, conjunc-
Fax: +82-64-756-3351 tivitis, anterior uveitis/iritis, and episcleritis, erythema nodo-
Copyright G2018, The Microbiological Society of Korea sum, pyoderma gangrenosum, and primary sclerosing chol-
190 Eom et al.

Fig. 1. Factors involved in IBD


occurrence.

angitis (Isene et al., 2015). transcription factor family member that is mainly expressed
+
The pathogenesis of IBD is not completely understood. in a subset of CD4 T-cells that suppress the immune system
Figure 1 shows factors involved in IBD. Current hypotheses (Kim, 2009).
are that the homeostasis between microbiota, intestinal ep- Treatment of IBD involves the induction and maintenance
ithelial cells and immune cells are disrupted by genetic fac- of a state of remission. Currently available treatments include
tors and environmental factors such as antibiotics, smok- 5-aminosalicylate agents (5-ASA), corticosteroids, antibiotics,
ing, diets, and stress (Ananthakrishnan, 2015), resulting in nonsteroidal anti-inflammatory drugs (NSAID), immuno-
a chronic state of dysregulated inflammation. IBD appears modulators (azathioprine, 6-mercaptopurine, methotrexate,
to be caused by an abnormal immune response to commensal cyclosporine, tacrolimus), and anti-TNF therapies. Surgical
intestinal bacteria rather than to infectious pathogens (Packey treatment may be required if there is no response to medica-
and Sartor, 2008). To date, many studies have focused on the tions or if there is perforation, obstruction, bleeding, or other
role of innate and adaptive immunity in the intestinal mu- tissue pathologies. While the risk of surgery within 10 years
cosa in IBD patients. In aspect of innate immunity, epithelial after diagnosed CD or UC used to be 46.6 and 15.6%, res-
barrier dysfunction including the disruption of mucin and pectively, this has been decreasing over the past decades
tight junctions, defective expression of antimicrobial pep- (Frolkis et al., 2013).
tides, and abnormal microbial antigen sensing have been
reported to be the cause of IBD. In addition, dysregulation
of autophagy with unfolded protein XBP1 and ORMDL3 is Factors involved in IBD
also considered to be pathogenesis of IBD (Kaser et al., 2010).
The adaptive immune response is a T-cell mediated immune Abnormal immune response associated with IBD
system, which has long-lasting memory against specific tar- High-throughput DNA sequence analysis among IBD pa-
gets, including intra- and extra-cellular pathogens and allergy- tients identified 215 human genetic defects associated with
causing antigens. The T cells include helper (Th) and regu-
IBD in the genome-wide association study (GWAS) (Luo
latory T cells (Tregs), that are differentiated from naive T cells et al., 2017). While most of these genetic defects are yet to be
(T0). In CD, IL-12 stimulates Th-1 cells to secrete IFN-γ, functionally-characterized, there are several IBD risk genes
which induces production of TNF-α by activating mucosal
that have been intensively studied. For example, MUC2 plays
macrophages. In contrast, UC is characterized by a Th2-im- a major role in secretion of mucin from goblet cells, and
mune response and excessive secretion of IL-5 and IL-13. failure in providing mucosal layer allows for the direct con-
However, this idea is not universally accepted and there is -/-
tact of gut bacteria with epithelial cells. MUC2 mice have
a controversial studies about this concept (Di Sabatino et been reported to develop colitis and an increased risk of col-
al., 2012). While CD and UC used to be considered as Th1- orectal cancer (Van der Sluis et al., 2006). In addition, clau-
and Th2-mediated diseases, respectively, recent studies have din, a family of transmembrane proteins, plays a key role in
suggested that Th17 is also involved in IBD (Zenewicz et controlling apical tight junction of epithelial cells. Signifi-
al., 2009). TGF-β promotes differentiation of T0 into Tregs cantly higher expression of claudin 2 has been reported for
by inducing FoxP3 expression, conversely, in the presence UC patients, whereas defects in tight junctions are not exclu-
of IL-6, the production of Tregs is inhibited and TGF-β sive to CD (Ahmad et al., 2014). Whether the dysfunctional
promotes the expression of the nuclear hormone receptors tight junctions are the cause of IBD or the consequence of
RORγt, promoting T0 differentiation into Th17 cells who IBD is still under discussion, nevertheless, improving apical
produce a number of cytokine molecules and amplify the in- tight junctions is a promising approach to treat the symp-
flammatory process (Galvez, 2014). The FoxP3 is a forkhead toms of IBD (Landy et al., 2016). Thus as stated above, a di-
Roles of probiotics in IBD 191

rect linkage between cause and effect is currently unclear have lower abundance of the genus Roseburia which was
or unknown. negatively correlated to the IBD risk scores, suggesting that
In the gastrointestinal tract, the presence of invaders can be IBD risk loci affect gut microbiota regardless the disease
sensed by pattern recognition receptors (PRRs), via a process characteristics. Roseburia is a heritable butyrate producer
called recognizing pathogen associated molecular patterns that resides within mucosal layer (Van den Abbeele et al.,
(PAMPs). The PRRs include Toll-like receptors (TLRs) and 2013; Goodrich et al., 2014). Gevers et al. (2014) investigated
NOD-like receptors (NLRs). Signals from these receptors re- new-onset treatment-naive gut microbiota in CD patients
sult in activation of nuclear factor (NF)-κB and production and reported that intestinal microbial shifts occur with mu-
of pro-inflammatory mediators. The TLRs, which are located cosal microbiota, rather than luminal microbiota, at the early
on cell membranes, play an important function in sensing stages of IBD. These shifts generally include decreased spe-
bacterial cells through recognition of lipopolysaccharide (LPS) cies diversity and the abundances of Bacteroides, Clostridia,
and peptidoglycans, which subsequently enhance the secre- Bifidobacteriaceae, Faecalibacterium, and Ruminococcaceae
tion of immunoglobulin A (IgA) in the intestine (Shang et as well as an increase in abundance of Enterobacteriaceae,
al., 2008). Secretory IgA protects intestinal surface against adherent-invasive E. coli (EHEC), Fusobacteriaceae, Myco-
harmful stimuli. In contrast, NLRs are located in cytoplasmic bacterium avium, and Clostridium difficile. The chronic in-
compartments. The NOD2 gene, one NLR, is the first reported flammation or bowel resection in patients with IBD further
IBD risk gene (Hugot et al., 2001; Ogura et al., 2001). Genetic shifts gut microbiota away from “healthy” gut microbiota
defects in NOD2 lead to the development of ileal CD, by in- (Halfvarson et al., 2017). These studies, however, report cor-
creasing activation of NF-κB. There are, however, contra- relative data and the effects of individual microbiota on CD
dicting reports on the role the NOD2 gene in presentation presentation needs to be tested addressing Koch’s postulate.
of CD (Yamamoto and Ma, 2009). In gut, epithelial cells are covered with two mucus layers
Adaptive immune responses are initiated following inva- made of mucin produced by goblet cells. While the inner mu-
sion of specific bacteria or viruses. There are three patterns cus layer is nearly bacteria free, the outer mucus layer on the
in adaptive immune responses in defense of: i) intracellular large intestine contains mainly mucus degraders and some
invasion by bacteria or viruses; ii) extracellular parasites such commensal bacteria that feed on secondary metabolites pro-
as helminths and Schistosoma; and iii) extracellular invasion duced from mucin degradation (Li et al., 2015). Figure 2
by bacteria or viruses. Under the intracellular invasion, im- shows interactions within mucosal microbiota. Bacteria trap-
mune cells (i.e., macrophages and dendric cells) first produce ped in the outer mucus layers often include mucolytic bac-
IL-12 to cause T0 differentiation into Th1, then Th1 sub- teria, such as members of Akkermansia, Bacteroides, Bifido-
sequently increases the amount of IFN-γ that activates im- bacterium, and Ruminococcus. Mucolytic bacteria degrade
mune cells. It should be noted however, that overexpression mucin oligosaccharides, which can be further utilized by non-
of IFN-γ leads to the development of CD (Powrie et al., mucolytic bacteria, providing different microbial metabolites
1994). In case of extracellular invasion of parasites, T0 is than those produced by luminal microbiota (Ouwerkerk et
differentiated into Th2 under IL-4 and TGF-β. Th2 acti- al., 2013). The intestinal glycobiome transforms mucin gly-
vates eosinophil and mast cells by producing IL-5 and IL-6, cans into short chain fatty acids (SCFAs), which can be fur-
respectively. Th2 also produces immunoglobulin E to attack ther utilized by colonocytes as energy sources. It should be
these parasites. While Th2 is effective in stopping inflamma- noted that the presence of these bacteria does not imply
tion caused by those parasites, dysregulation of Th2 may complete degradation of the mucus layer, and many bacteria
cause over expression of IL-13, which subsequently induces only utilize partial mucus-derived sugars (Marcobal et al.,
UC (Strober and Fuss, 2011). Th17 cells are pro-inflamma- 2013). Mucus-derived sugars include N-acetyl-D-glucosamine
tory T helper cells generated in response to the invasion of (NAG), N-acetylgalactosamine, galactose, fucose, and sialic
extracellular bacteria and viruses. Dysfunctional Th17 causes
autoimmune disorders. The use of anti-IL-23 materials have
been also tested for drug development for inflammatory di-
seases, such as psoriasis (Gaffen et al., 2014). Further studies
are needed to determine whether dysregulation of Th17 is
preferably associated with CD or UC (Zenewicz et al., 2009).
The Tregs, on the other hand, are the suppressor of Th cells
by producing TGF-β and IL-10, which subsequently de-
creases secretion of harmful cytokines. A decrease in the
number of Tregs leads to the weakened anti-inflammatory
effects, causing IBD (Boden and Snapper, 2008).

Gut microbiota associated with IBD


In healthy humans, commensal bacteria maintain a symbiotic
relationship with the host by modulating a number of innate
and adaptive immune systems. Defects in genes involved in
the immune systems could cause failure in maintaining com-
mensal gut microbiota, leading to dysbiosis. Imhann et al.
(2018) reported that 582 healthy humans with IBD risk loci Fig. 2. Interactions of intestinal bacteria with intestinal mucus.
192 Eom et al.

acid (Sonnenburg et al., 2010). The NAG and sialic acid were Probiotics intervention to treat IBD
known to repress virulence in Salmonella and enterohe- Several probiotics including Lactobacillus strains (rhamnosus
morrhagic E. coli (EHEC) (Sicard et al., 2017). Fucose, on
GG, acidophilus, reuteri ATCC 55730, johnsonii), Bifidobac-
the other hand, is also provided by the host in the form of terium strains (breve, bifidum, animalis), Saccharomyces bou-
fucosylated-proteins through expression of the fut2 gene to lardii, Escherichia coli Nissle (ECN) 1917, and VSL#3 (a
maintain commensal fucose-utilizers in the outer mucus
probiotic mix of 4 lactobacilli, 3 bifidobacteria and a Strep-
layer (Pickard et al., 2014). These commensal fucose-utilizers tococcus) have been studied for their efficacy in the treatment
inhibit the growth of pro-inflammatory pathogenic bacteria, of IBD (Abraham and Quigley, 2017). In individual studies,
such as Citrobacter rodentium and Salmonella, and defects
treatment with several probiotics was reported to be effective,
in the fut2 gene may lead to inflammation (Goto et al., 2014). but the sample size was small and there were inconsistent
Likewise, administration of antibiotics inhibits commensal results. The meta-analysis showed that only VSL#3 was effec-
bacteria that feed on mucin-derived monosaccharides, which
tive in inducing remission of active UC (Derwa et al., 2017).
allows expansion of antibiotic resistant pro-inflammatory The VSL#3 mixture was also reported, in several clinical tri-
pathogenic bacteria, such as Salmonella enterica serotype als, to be effective in the prevention of pouchitis following
Typhimurium and Clostridium difficile, that feed on liberated
an ileal pouch–anal anastomosis (IPAA) (Gionchetti et al.,
mucin-derived monosaccharides released by the mucin de- 2003; Mimura et al., 2004). The ECN strain1917 appeared
graders (Ng et al., 2013a). Therefore, maintaining an intes- equivalent to 5-ASA in preventing relapse of IBD (Losurdo
tinal-mucosal microbial homeostasis may prevent occur-
et al., 2015). However, the effect of various probiotics on the
rence of IBD, even for those having IBD risk loci. treatment of CD is not clear in terms of induction of remi-
ssion, prevention of recurrence following active disease, or
prevention of post-operative recurrence (Derwa et al., 2017;
Probiotics to treat IBD McIlroy et al., 2018).
It is not clear why the results of clinical studies are disap-
Effects of probiotics on intestinal health
pointing despite the role of intestinal microbiota in the path-
Although there is no legal definition for probiotics, it is ge- ophysiology of IBD and the proven effects of probiotics in
nerally accepted as “live microorganisms that confer a health preclinical studies. The meta-analysis also provides limited,
benefit on the host when administered in adequate amounts” but not conclusive, information since different strains and
(Sanders, 2008). The mechanism of action of probiotics has doses of probiotics were used, with different duration of use,
been dropped from the definition, and the mode of action and with patients showing different disease severity in each
and its beneficial effects differ among probiotics at the strain clinical studies (Derwa et al., 2017). This limitation is similar
level (Whelan and Quigley, 2013). With respect to IBD, the to that for IBS, a disease in which probiotics are more com-
potentially beneficial effects caused by probiotics have been monly used (Mazurak et al., 2015). In addition, it has been
thought to include: i) inhibition of invasion by pathogenic reported that efficacy of probiotics is strain-specific (Whelan
bacteria; ii) improvement of epithelial barrier functions; and Quigley, 2013) and may vary depending on the personal
and iii) immunomodulation (Dos Reis et al., 2017). Some of microbiota (Kolmeder et al., 2016). It was also suggested that
the probiotics reduce intestinal pH through production of orally administrated probiotics may have difficulty in reach-
SCFAs, which have been shown to inhibit intestinal patho- ing the colon (Dos Reis et al., 2017). To date, probiotics are
genic E. coli (Fukuda et al., 2011). Moreover, production of not approved as pharmaceuticals and are marketed as diet-
SCFAs, especially butyrate, feeds colonocytes and enhances ary supplements, therefore, not regulated by authorities. As
epithelial tight junction integrity (Ewaschuk et al., 2008). such, no health improvement can be claimed.
Some probiotics are known to interfere with adhesion of gas- In the case of VSL#3, which showed beneficial effects on
trointestinal pathogens by competitive exclusion and bacte- treating UC, the formulation was recently changed and the
riocin production (Kravtsov et al., 2008). In addition, some product differs depending on the country in which it is sold
species of lactic acid bacteria induce cytokine secretion from (Biagioli et al., 2017). One study showed that the original
monocytes and activate NF-κB (Jensen et al., 2015), the ma- and newfound VSL#3 exhibited different proportion of dead
jor key mediator in inflammation (Liu et al., 2017). It has microbes and in vitro anti-tumoral effects (Cinque et al.,
been reported that the abundance of Bifidobacterium is neg- 2016). Lastly, it is very rare, but the potential risk of probiotic-
atively associated with IBD (Tojo et al., 2014), thus taking induced bacteremia should be considered in immunosup-
probiotics may help regaining gut homeostasis. pressed patients with severe active IBD (Vahabnezhad et al.,
There are conflicting results concerning the impact of pro- 2013). In the future, well designed clinical studies for first and
biotics on gut microbiota (Maldonado-Gomez et al., 2016). next-generation probiotics following strict trial guidelines
In some studies, it has been reported that probiotics may not are needed (Shanahan, 2017).
have impacts on healthy adults’ intestinal microbiota (Kris-
tensen et al., 2016). Sanders (2016) suggested that probiotics
do not directly change microbiota, but rather maintain gut Prebiotics to treat IBD
homeostasis so that gut dysbiosis caused by perturbations
can be minimized or allow faster recovery. Collectively, the Effects of prebiotics on intestinal health
use of probiotics may be a promising approach to treat and Since many of traditional probiotics such as Lactobacillus
prevent IBD, but much more work is needed. do not colonize the gut for long term, the utilization of com-
Roles of probiotics in IBD 193

mensal intestinal microorganisms to treat or prevent IBD which consists of 2 to 8 fructose molecules. Typically, inulin
may provide a sustainable solution to the problem (Sanchez has been reported to improve constipation, prevent intestinal
et al., 2017). The growth of beneficial bacteria is regulated diseases, reduce serum cholesterol, lower blood lipid levels,
by complex non-digestible carbohydrates. Prebiotics are de- and lower blood glucose levels (Flamm et al., 2001). Because
fined as non-digestible carbohydrates that selectively pro- inulin and OF are not degraded by the carbohydrate hydro-
mote the activity and growth of one or more strains of ben- lyzing enzyme secreted from the small intestine, over 80% of
eficial bacteria in the large intestine, thus improving the in- ingested inulin and OF reach the large intestine and are used
testinal environment and benefiting the host (Gibson and in the production of SCFAs by various bacteria (Cherbut,
Roberfroid, 1995). Therefore, the functionality of prebiotics 2002). In the westernized diet model, administration of in-
in the human digestive tract varies significantly depending on ulin and OF plays a role in increasing number of lactic acid
the viability and metabolic activity of beneficial bacteria in bacteria, which consequently reduced intestinal pH (Licht
the intestines. Prebiotics are not degraded by the carbohy- et al., 2006). Furthermore, administration of inulin and OF
drate hydrolyzing enzymes secreted from the digestive tract increased the synthesis of butyrate which inhibited dam-
and are not absorbed in the intestines for use as an energy ages on the intestinal epithelial cells due to inflammatory re-
source. Prebiotics are selectively decomposed by probiotic actions (Videla et al., 2001). Others reported that the use of
strains such as Lactobacillus and Bifidobacterium in the large mesalazine, an IBD therapeutic agent with inulin contain-
intestine. Typical prebiotics include inulin, fructo-oligosac- ing OF, decreased dyspepsia scores and fecal calprotectin in
charide (FOS), galacto-oligosaccharide (GOS), and lactulose, IBD patients more effectively than the use without prebiotics
that are widely available in nature or obtained through en- (Casellas et al., 2007). Studies have reported that the inges-
zymatic synthesis (Mussatto and Mancilha, 2007). Previous tion of inulin and OF reduced the production of aberrant
studies have reported various functionalities in prebiotics, crypt foci (ACF), a precursor to colorectal cancer, and also
including enhancement of intestinal immunity, anti-diabetic reported that the effects of long-chain inulin was more evi-
and anti-obesity activities, and improvement of constipation dent than those with the shorter sugar chain (Verghese et
(Roberfroid, 2000; Slavin, 2013). Furthermore, prebiotics are al., 2005).
utilized by probiotic bacteria and metabolized into SCFAs, GOS is a polymer similar to the oligosaccharide of breast
such as acetate, propionate, and butyrate. Butyrate can be milk, in which the sugar molecule is linked by a β-glycosidic
used by colonocytes as a source of energy, subsequently sti- bond, and has shown similar effects as breast milk on intes-
mulates mucin biosynthesis and enhances intestinal barrier tinal microorganisms (Rivero-Urgell and Santamaria-Orleans,
(Maslowski and Mackay, 2011; Rios-Covian et al., 2016). It 2001). The length of GOS sugar chains is typically in the range
has been reported that various prebiotics and probiotics have from 3 to 10. GOS reaches the large intestine without being
potentials in treating IBD (Zhu et al., 2011). IBD patients affected by saliva, gastric acid, or the digestive juices of the
showed decreased fecal SCFAs (Takaishi et al., 2008). The small intestine. GOS is known to enrich Bifidobacterium and
beneficial effects of SCFAs include: i) inhibiting adhesion of Lactobacillus in the large intestine, prevent pathogenic bac-
pathogenic microorganisms to intestinal cells (Kamada et al., terial infection, reduces pH, increase fecal SCFA contents,
2013); ii) supporting secretion of antimicrobial substances relieve constipation, stimulate immune activity, reduce allergy
from epithelial cells within the colon (Sunkara et al., 2012); and atopy, and prevent colon cancer (Sangwan et al., 2011).
iii) selectively allowing the growth of probiotics over patho- Despite the various effects of GOS in the intestines, its effects
genic Escherichia coli and Bacteroidaceae (Sun and O’Riordan, on IBD prevention have been controversial. In a 2,4,6-tri-
2013); iv) providing anti-inflammatory effects for epithelial nitrobenzenesulfonic acid (TNBS)-induced IBD model, in-
cells through inhibition of activation of NF-kB and PPARγ, gestion of GOS showed an increase of Bifidobacterium in the
and limiting production of IL-1β, IL-6, and TNF-α (Fung intestines but did not reduce inflammation (Holma et al.,
et al., 2012); and v) stimulating epidermal growth factor re- 2002). Conversely, ingestion of GOS showed activation of NK
ceptor (EGFR) to maintain intestinal homeostasis by increa- cells and reduced severity of colitis induced by Helicobacter
sing mucin synthesis (Andrianifahanana et al., 2006). These hepaticus infection in Smad-3 knock-out mice (Gopalakri-
results suggest that prebiotics have potential as therapeutic shnan et al., 2012). Collectively these results suggest that fur-
and preventive agents for IBD, though the exact roles of ther studies are required to determine the effect of GOS on
prebiotics in IBD have yet to be revealed. the prevention and treatment of IBD. Several studies have
convincingly shown that milk glycans have a prebiotic effect
Prebiotics intervention to treat IBD on the microbiome of the infant gut and may be useful to pre-
venting diseases such as necrotizing enterocolitis in preterm
A number of studies on the protective effects of prebiotics in
infants (Pacheco et al., 2015).
IBD treatment have focused on inulin, FOS and GOS, since
β-Glucan is an abundant polysaccharide in nature whose
they can be extracted from plants at low cost. Mushroom and
glucose molecules are linked via β-bonds. This polysaccharide,
seaweeds contain potentially prebiotic products such as β-
along with cellulose, is an important component of plant fiber
glucan, laminarin, and fucoidan, which have been also sug-
and a major constituent of cell walls in seaweed and fungi.
gested as potential prebiotics in treating IBD (O’Sullivan et
While β-glucans are postulated to have immunostimulatory
al., 2010; Daou and Zhang, 2012).
effects (Daou and Zhang, 2012), recent studies have reported
Inulin is a polysaccharide obtained from chicory roots,
that the physiological functions attributed to β-glucan are
which consists of 2 to 60 fructose molecules, with the sugar
not direct effects, but rather due to intestinal microorganisms
chain linked by a β-(2,1) bond. The polysaccharide obtained
whose activities are enhanced by β-glucan (Cloetens et al.,
by enzymatic hydrolysis of inulin is called oligofructose (OF),
194 Eom et al.

2012). Numerous studies on the prevention of IBD through Laminarin extracted from Laminaria reduced the expre-
the ingestion of β-glucan have been conducted using dextran ssion of cytokines and receptors which are related to the
sulfate sodium (DSS)- and TNBS-induced bowel inflamma- Th17 and also decreased the abundance of E. coli, showing
tion models. It has been reported that oral administration the possibility for the prevention of IBD (O’Shea et al., 2016).
of glucan derived from the mushroom Pleurotus pulmona- The use of prebiotics is not free of health risks. While a num-
rius alleviated symptoms of intestinal inflammation induced ber of studies have reported beneficial effects of prebiotics,
by DSS and inhibited the formation of colon cancer caused they should be used with cautions since they may cause ab-
by colitis (Lavi et al., 2010). Moreover, it has been reported dominal pain, indigestion, diarrhea, and bloating or flatu-
that a β-glucan polymer extracted from Shitake mushrooms lence (Marteau and Flourie, 2001). Excessive ingestion of
(Lentinus edodes) inhibits MAPK and PPARγ phosphory- prebiotics could induce diarrhea due to osmotic retention of
lation, resulting in reduction of the inflammatory cytokines fluid in the intestine (Swennen et al., 2006). FOS ingestion
and oxidative stress induced by DSS (Shi et al., 2016). The in patients with gastroesophageal reflux disease increased
efficacy of β-glucan for the prevention of colitis has further gastroesophageal reflux, but how this occurs is not known
been verified in clinical trials. One study reported that the (Piche et al., 2003). Inulin and FOS have been reported to in-
administration of β-glucan extracted from Agaricus to IBD crease Lactobacillus and Bifidobacterium in the intestines,
patients reduced the level of inflammatory cytokines in the but also increased colonization of Salmonella and in cecal
blood and decreased the level of calprotectin, a fecal marker contents and mucosa (Ten Bruggencate et al., 2004). There-
of IBD (Forland et al., 2011). Similarly, administration of the fore, further studies are required to determine the protective
β-glucan extracted from the chaga mushroom to IBD pa- mechanisms of prebiotics against inflammation.
tients inhibited inflammatory response-induced damage to
lymphocytes in blood (Najafzadeh et al., 2007).
Lastly, several varieties of seaweed are also potential pre- Other methods to treat IBD
biotics (O’Sullivan et al., 2010) which have long been con-
sumed as food in Korea, China, Japan, and Southeast Asian Fecal microbiota transplantation (FMT) is a method used
countries. In contrast, seaweeds are mainly used for pharma- to transplant microbiota obtained from healthy donors to
ceuticals, cosmetics, and food additives in western countries. patients most presenting with recurrent Clostridium difficile
Seaweeds are classified into three groups; brown algae (Pha- infection (rCDI). This technology has remarkable cure rates
ophyceae, about 1,750 species), red alage (Rhodophyceae, for rCDI, approaching 96% (Sadowsky et al., 2017). Unlike
about 6,000 species), and green algae (Chlorophyceae, about medical drugs including antibiotics and immunosuppre-
1,500 species) (Kılınç et al., 2013). Seaweeds are abundant ssants, FMT increases the number and diversity of fecal bac-
in protein and carbohydrates and also contain omega-3 fatty terial populations in recipients, and engraftment lasts for
acids, carotenoids, vitamins, and minerals (Wells et al., 2017). long time periods (Weingarden et al., 2015). This suggests
Seaweeds are particularly abundant in non-digestible poly- that FMT may be a promising therapy for IBD. However,
saccharides (approximately 75% of their dry weight), typi- FMT has shown rather disappointing success rate in treating
cally including alginates, laminarins, and fucoidans in brown IBD, when compared to treating CDI (Pigneur and Sokol,
algae, agars, and carrageenans in red algae, and ulvan in 2016). Systematical review of previous cases done to treat UC
green algae (Senthilkumar et al., 2013). Seaweed-derived poly- with FMT suggested that there was only slightly higher rate
saccharides studied for the prevention and inhibition of IBD of clinical remission and response compared to placebo (19%
include brown algae alginates, laminarins, and fucoidans. and 21%, respectively) (Costello et al., 2017). The efficacy of
It has been reported that ingestion of alginate was linked to FMT may vary depending on the fecal microbiota of the do-
an alleviation of the inflammatory response in the colon and nor, method used to prepare the microbiota, dose and fre-
prevention of injury to the goblet cells in animal models of quency, the patient’s condition, and delivery routes. The re-
DSS- and TNBS-induced colitis with no association of the lationship between FMT and clinical resolution of IBD ap-
adverse effects such as potential weight loss or spleen reduc- pears complex, perhaps due to differing etiologies of UC and
tion, seen with prednisolone treatments (Yamamoto et al., CD and severity of disease systems. In a recent study, Kho-
2013). Fucoidan, a sulfated polysaccharide mainly composed ruts and colleagues (Khoruts et al., 2016) reported that IBD
of fucose and abundant in brown algae, has been reported influenced success of FMT for treatment of CDI. More-
to have various physiological functions, such as immuno- over, while FMT did clear CDI in nearly 74.5% of 272 pa-
modulation, anticancer activity, anti-obesity effects (Ale et tients, those without IBD were cured at a greater rate (92%)
al., 2011), and, more recently, prebiotic effects (Okolie et al., and nearly 25% of patients with IBD had a significant flare
2017). In the DSS-induced IBD model, ingestion of fucoi- following FMT. More importantly however, there was an
dan improved inflammation-related indicators such as in- increase in failure rate due to IBD. Interestingly, while some
flammatory cytokine levels, weight loss, diarrhea, and hema- patients showed noteworthy improvements in their IBD after
tochezia, while further inhibiting the reduction of crypt and FMT, a few had modest improvements and even less con-
goblet cells in the large intestine (Lean et al., 2015). In an- tinued to struggle with their underlying IBD despite clear-
other IBD model, ingestion of fucoidan extracted from mo- ing of CDI. As FMT is a relatively new approach, further
zuku seaweed (Cladosiphon okamuranus) decreased inflam- study is needed to improve FMT-based treatment of patients
matory cytokines such as IFNγ and IL-6, and increased in- with IBD.
flammation-inhibiting IL-10 and TGF-ß (Matsumoto et al.,
2004). Laminarin is a β-glucan derived from brown algae.
Roles of probiotics in IBD 195

Acknowledgements

We are grateful to Sustainable Agriculture Research Institute


(SARI) at Jeju National University for providing the experi-
mental facilities. This research was supported, in part, by the
Basic Science Research Program through the National Re-
search Foundation of Korea (NRF) funded by the Ministry
of Education (2016R1A6A1A03012862) and by the Tradi-
tional Culture Convergence Research Program through the
NRF, and funded by the Ministry of Science, ICT & Future
Planning (2016M3C1B5907205) to (TU). We also thank the
Minnesota Agricultural Experiment Station for support (to
MJS).

References

Abraham, B.P. and Quigley, E.M.M. 2017. Probiotics in inflamma-


Fig. 3. Factors affecting gut microbiota. tory bowel disease. Gastroenterol. Clin. North Am. 46, 769–782.
Ahmad, R., Chaturvedi, R., Olivares-Villagomez, D., Habib, T., Asim,
M., Shivesh, P., Polk, D.B., Wilson, K.T., Washington, M.K., Van
Conclusion Kaer, L., et al. 2014. Targeted colonic claudin-2 expression renders
resistance to epithelial injury, induces immune suppression, and
As gut microbiota is strongly associated with IBD, under- protects from colitis. Mucosal Immunol. 7, 1340–1353.
standing how a patient’s gut microbiota respond to treat- Ale, M.T., Mikkelsen, J.D., and Meyer, A.S. 2011. Important deter-
ments has become important. Recent studies indicated the minants for fucoidan bioactivity: a critical review of structure-
importance of mucosal microbiota, where bacteria have clo- function relations and extraction methods for fucose-containing
sulfated polysaccharides from brown seaweeds. Mar. Drugs 9,
ser interaction with host cells than those in the lumen. Since 2106–2130.
modulating luminal bacteria indirectly affects mucosal mi- Ananthakrishnan, A.N. 2015. Environmental risk factors for in-
crobiota, diet- and microbiota-therapies, are the important flammatory bowel diseases: a review. Dig. Dis. Sci. 60, 290–298.
factors in managing IBD (Fig. 3). Few studies have investi- Andrianifahanana, M., Moniaux, N., and Batra, S.K. 2006. Regulation
gated roles of mucosal microbiota compared to that of lumi- of mucin expression: mechanistic aspects and implications for
nal microbiota. The use of DSS-treated mice for IBD study cancer and inflammatory diseases. Biochim. Biophys. Acta 1765,
makes it difficult to obtain purely separated mucosal micro- 189–222.
biota from the ones in lumen, thus further animal model IBD Biagioli, M., Cipriani, S., Carino, A., Distrutti, E., Marchianò, S., and
studies may require larger subjects such as rats. Comparison Fiorucci, S. 2017. Variability in industrial production affects pro-
biotics activity: identification of batches of probiotic VSL#3 that
of microbiome between IBD biopsy and fecal samples may
increases intestinal permeability and worsens colitis in rodents.
also help us understand roles of mucosal microbiota. Gastroenterology 152, S969.
The efficacy of the treatments developed for IBD usually Boden, E.K. and Snapper, S.B. 2008. Regulatory T cells in inflam-
vary due to interindividual differences in gut microbiota. For matory bowel disease. Curr. Opin. Gastroenterol. 24, 733–741.
example, results expected from dietary intervention may vary Casellas, F., Borruel, N., Torrejon, A., Varela, E., Antolin, M., Guar-
depending on how it was digested and absorbed by hosts, ner, F., and Malagelada, J.R. 2007. Oral oligofructose-enriched
which is mostly determined by types of gut microbiota. There- inulin supplementation in acute ulcerative colitis is well tolerated
fore, personalized treatments are needed, especially for di- and associated with lowered faecal calprotectin. Aliment. Phar-
sease associated with gut dysbiosis. While FMT may replace macol. Ther. 25, 1061–1067.
Cherbut, C. 2002. Inulin and oligofructose in the dietary fibre con-
a majority of gut microbiota lost to toxin- and antibiotic-
cept. Br. J. Nutr. 87 Suppl 2, S159–S162.
induced dysbiosis, it gradually becomes personalized likely Cinque, B., La Torre, C., Lombardi, F., Palumbo, P., Van der Rest, M.,
due to host genetic differences (Staley et al., 2016). To de- and Cifone, M.G. 2016. Production conditions affect the in vitro
velop sustainable IBD treatments, personalized treatments anti-tumoral effects of a high concentration multi-strain probiotic
should be chosen based on characteristics of a patient’s gut preparation. PLoS One 11, e0163216.
microbiota and genetics as well as dietary habits and life style Cloetens, L., Ulmius, M., Johansson-Persson, A., Akesson, B., and On-
information. Recent development of next generation sequen- ning, G. 2012. Role of dietary beta-glucans in the prevention of
cing technology and bioinformatics, coupled with compu- the metabolic syndrome. Nutr. Rev. 70, 444–458.
tational infrastructure, may lead to the development of per- Costello, S.P., Soo, W., Bryant, R.V., Jairath, V., Hart, A.L., and And-
sonalized IBD treatment through big data analysis and a rews, J.M. 2017. Systematic review with meta-analysis: faecal mi-
crobiota transplantation for the induction of remission for active
better understanding of host-microbe interactions in the gut ulcerative colitis. Aliment. Pharmacol. Ther. 46, 213–224.
environment. Daou, C. and Zhang, H. 2012. Oat beta-glucan: Its role in health pro-
motion and prevention of diseases. Compr. Rev. Food Sci. F. 11,
355–365.
Derwa, Y., Gracie, D.J., Hamlin, P.J., and Ford, A.C. 2017. Systematic
review with meta-analysis: the efficacy of probiotics in inflamma-
196 Eom et al.

tory bowel disease. Aliment. Pharmacol. Ther. 46, 389–400. Holma, R., Juvonen, P., Asmawi, M.Z., Vapaatalo, H., and Korpela,
Di Sabatino, A., Biancheri, P., Rovedatti, L., MacDonald, T.T., and R. 2002. Galacto-oligosaccharides stimulate the growth of bifido-
Corazza, G.R. 2012. New pathogenic paradigms in inflammatory bacteria but fail to attenuate inflammation in experimental colitis
bowel disease. Inflamm. Bowel Dis. 18, 368–371. in rats. Scand. J. Gastroenterol. 37, 1042–1047.
Dos Reis, S.A., da Conceicao, L.L., Siqueira, N.P., Rosa, D.D., da Silva, Hugot, J.P., Chamaillard, M., Zouali, H., Lesage, S., Cezard, J.P.,
L.L., and Peluzio, M.D. 2017. Review of the mechanisms of pro- Belaiche, J., Almer, S., Tysk, C., O’Morain, C.A., Gassull, M., et al.
biotic actions in the prevention of colorectal cancer. Nutr. Res. 2001. Association of NOD2 leucine-rich repeat variants with sus-
37, 1–19. ceptibility to Crohn’s disease. Nature 411, 599–603.
Ewaschuk, J.B., Diaz, H., Meddings, L., Diederichs, B., Dmytrash, Imhann, F., Vich Vila, A., Bonder, M.J., Fu, J., Gevers, D., Vissche-
A., Backer, J., Looijer-van Langen, M., and Madsen, K.L. 2008. dijk, M.C., Spekhorst, L.M., Alberts, R., Franke, L., van Dullemen,
Secreted bioactive factors from Bifidobacterium infantis enhance H.M., et al. 2018. Interplay of host genetics and gut microbiota
epithelial cell barrier function. Am. J. Physiol. Gastrointest. Liver underlying the onset and clinical presentation of inflammatory
Physiol. 295, G1025–G1034. bowel disease. Gut 67, 108–119.
Flamm, G., Glinsmann, W., Kritchevsky, D., Prosky, L., and Rober- Isene, R., Bernklev, T., Hoie, O., Munkholm, P., Tsianos, E., Stock-
froid, M. 2001. Inulin and oligofructose as dietary fiber: a review brugger, R., Odes, S., Palm, O., Smastuen, M., Moum, B., et al.
of the evidence. Crit. Rev. Food Sci. Nutr. 41, 353–362. 2015. Extraintestinal manifestations in Crohn’s disease and ul-
Forland, D.T., Johnson, E., Saetre, L., Lyberg, T., Lygren, I., and Het- cerative colitis: results from a prospective, population-based Euro-
land, G. 2011. Effect of an extract based on the medicinal mush- pean inception cohort. Scand. J. Gastroenterol. 50, 300–305.
room Agaricus blazei murill on expression of cytokines and cal- Jensen, H., Dromtorp, S.M., Axelsson, L., and Grimmer, S. 2015.
protectin in patients with ulcerative colitis and Crohn’s disease. Immunomodulation of monocytes by probiotic and selected lac-
Scand. J. Immunol. 73, 66–75. tic acid bacteria. Probiotics Antimicrob. Proteins 7, 14–23.
Frolkis, A.D., Dykeman, J., Negron, M.E., Debruyn, J., Jette, N., Johnston, R.D. and Logan, R.F. 2008. What is the peak age for on-
Fiest, K.M., Frolkis, T., Barkema, H.W., Rioux, K.P., Panaccione, set of IBD? Inflamm. Bowel Dis. 14 Suppl 2, S4–S5.
R., et al. 2013. Risk of surgery for inflammatory bowel diseases Kamada, N., Chen, G.Y., Inohara, N., and Nunez, G. 2013. Control
has decreased over time: a systematic review and meta-analysis of pathogens and pathobionts by the gut microbiota. Nat. Im-
of population-based studies. Gastroenterology 145, 996–1006. munol. 14, 685–690.
Fukuda, S., Toh, H., Hase, K., Oshima, K., Nakanishi, Y., Yoshimura, Kaser, A., Martinez-Naves, E., and Blumberg, R.S. 2010. Endoplasmic
K., Tobe, T., Clarke, J.M., Topping, D.L., Suzuki, T., et al. 2011. reticulum stress: implications for inflammatory bowel disease
Bifidobacteria can protect from enteropathogenic infection through pathogenesis. Curr. Opin. Gastroenterol. 26, 318–326.
production of acetate. Nature 469, 543–547. Khoruts, A., Rank, K.M., Newman, K.M., Viskocil, K., Vaughn, B.P.,
Fung, K.Y., Cosgrove, L., Lockett, T., Head, R., and Topping, D.L. Hamilton, M.J., and Sadowsky, M.J. 2016. Inflammatory bowel
2012. A review of the potential mechanisms for the lowering of disease affects the outcome of fecal microbiota transplantation
colorectal oncogenesis by butyrate. Br. J. Nutr. 108, 820–831. for recurrent Clostridium difficile infection. Clin. Gastroenterol.
Gaffen, S.L., Jain, R., Garg, A.V., and Cua, D.J. 2014. The IL-23- Hepatol. 14, 1433–1438.
IL-17 immune axis: from mechanisms to therapeutic testing. Kim, C.H. 2009. FOXP3 and its role in the immune system. Adv.
Nat. Rev. Immunol. 14, 585–600. Exp. Med. Biol. 665, 17–29.
Galvez, J. 2014. Role of Th17 cells in the pathogenesis of human Kılınç, B., Cirik, S., Turan, G., Tekogul, H., and Koru, E. 2013. Sea-
IBD. ISRN Inflamm. 2014, 928461. weed for food and industrial applications, In Muzzalupo, I. (ed.),
Gevers, D., Kugathasan, S., Denson, L.A., Vazquez-Baeza, Y., Van Food Industry. InTech, DOI: 10.5772/53172.
Treuren, W., Ren, B., Schwager, E., Knights, D., Song, S.J., Yassour, Kolmeder, C.A., Salojarvi, J., Ritari, J., de Been, M., Raes, J., Falony,
M., et al. 2014. The treatment-naive microbiome in new-onset G., Vieira-Silva, S., Kekkonen, R.A., Corthals, G.L., Palva, A., et
Crohn’s disease. Cell Host Microbe 15, 382–392. al. 2016. Faecal metaproteomic analysis reveals a personalized
Gibson, G.R. and Roberfroid, M.B. 1995. Dietary modulation of the and stable functional microbiome and limited effects of a pro-
human colonic microbiota: introducing the concept of prebiotics. biotic intervention in adults. PLoS One 11, e0153294.
J. Nutr. 125, 1401–1412. Kravtsov, E.G., Yermolayev, A.V., Anokhina, I.V., Yashina, N.V.,
Gionchetti, P., Rizzello, F., Helwig, U., Venturi, A., Lammers, K.M., Chesnokova, V.L., and Dalin, M.V. 2008. Adhesion characteri-
Brigidi, P., Vitali, B., Poggioli, G., Miglioli, M., and Campieri, stics of lactobacillus is a criterion of the probiotic choice. Bull.
M. 2003. Prophylaxis of pouchitis onset with probiotic therapy: Exp. Biol. Med. 145, 232–234.
a double-blind, placebo-controlled trial. Gastroenterology 124, Kristensen, N.B., Bryrup, T., Allin, K.H., Nielsen, T., Hansen, T.H.,
1202–1209. and Pedersen, O. 2016. Alterations in fecal microbiota compo-
Goodrich, J.K., Waters, J.L., Poole, A.C., Sutter, J.L., Koren, O., sition by probiotic supplementation in healthy adults: a system-
Blekhman, R., Beaumont, M., Van Treuren, W., Knight, R., Bell, atic review of randomized controlled trials. Genome Med. 8, 52.
J.T., et al. 2014. Human genetics shape the gut microbiome. Cell Landy, J., Ronde, E., English, N., Clark, S.K., Hart, A.L., Knight, S.C.,
159, 789–799. Ciclitira, P.J., and Al-Hassi, H.O. 2016. Tight junctions in inflam-
Gopalakrishnan, A., Clinthorne, J.F., Rondini, E.A., McCaskey, S.J., matory bowel diseases and inflammatory bowel disease asso-
Gurzell, E.A., Langohr, I.M., Gardner, E.M., and Fenton, J.I. 2012. ciated colorectal cancer. World J. Gastroenterol. 22, 3117–3126.
Supplementation with galacto-oligosaccharides increases the per- Lavi, I., Levinson, D., Peri, I., Nimri, L., Hadar, Y., and Schwartz, B.
centage of NK cells and reduces colitis severity in Smad3-defi- 2010. Orally administered glucans from the edible mushroom Pleu-
cient mice. J. Nutr. 142, 1336–1342. rotus pulmonarius reduce acute inflammation in dextran sulfate
Goto, Y., Obata, T., Kunisawa, J., Sato, S., Ivanov, I.I., Lamichhane, sodium-induced experimental colitis. Br. J. Nutr. 103, 393–402.
A., Takeyama, N., Kamioka, M., Sakamoto, M., Matsuki, T., et al. Lean, Q.Y., Eri, R.D., Fitton, J.H., Patel, R.P., and Gueven, N. 2015.
2014. Innate lymphoid cells regulate intestinal epithelial cell gly- Fucoidan extracts ameliorate acute colitis. PLoS One 10, e0128453.
cosylation. Science 345, 1254009. Li, H., Limenitakis, J.P., Fuhrer, T., Geuking, M.B., Lawson, M.A.,
Halfvarson, J., Brislawn, C.J., Lamendella, R., Vazquez-Baeza, Y., Wal- Wyss, M., Brugiroux, S., Keller, I., Macpherson, J.A., Rupp, S.,
ters, W.A., Bramer, L.M., D’Amato, M., Bonfiglio, F., McDonald, et al. 2015. The outer mucus layer hosts a distinct intestinal mi-
D., Gonzalez, A., et al. 2017. Dynamics of the human gut micro- crobial niche. Nat. Commun. 6, 8292.
biome in inflammatory bowel disease. Nat. Microbiol. 2, 17004. Licht, T.R., Hansen, M., Poulsen, M., and Dragsted, L.O. 2006. Die-
Roles of probiotics in IBD 197

tary carbohydrate source influences molecular fingerprints of the and Sweeney, T. 2016. The effect of algal polysaccharides lami-
rat faecal microbiota. BMC Microbiol. 6, 98. narin and fucoidan on colonic pathology, cytokine gene expre-
Liu, T., Zhang, L., Joo, D., and Sun, S.C. 2017. NF-kappaB signaling ssion and Enterobacteriaceae in a dextran sodium sulfate-chal-
in inflammation. Signal. Transduct. Target. Ther. 2, 17023. lenged porcine model. J. Nutr. Sci. 5, e15.
Losurdo, G., Iannone, A., Contaldo, A., Ierardi, E., Di Leo, A., and O’Sullivan, L., Murphy, B., McLoughlin, P., Duggan, P., Lawlor, P.G.,
Principi, M. 2015. Escherichia coli nissle 1917 in ulcerative colitis Hughes, H., and Gardiner, G.E. 2010. Prebiotics from marine
treatment: systematic review and meta-analysis. J. Gastrointestin. macroalgae for human and animal health applications. Mar. Drugs
Liver Dis. 24, 499–505. 8, 2038–2064.
Luo, Y., de Lange, K.M., Jostins, L., Moutsianas, L., Randall, J., Ken- Ogura, Y., Bonen, D.K., Inohara, N., Nicolae, D.L., Chen, F.F., Ra-
nedy, N.A., Lamb, C.A., McCarthy, S., Ahmad, T., Edwards, C., mos, R., Britton, H., Moran, T., Karaliuskas, R., Duerr, R.H., et al.
et al. 2017. Exploring the genetic architecture of inflammatory 2001. A frameshift mutation in NOD2 associated with suscepti-
bowel disease by whole-genome sequencing identifies associa- bility to Crohn’s disease. Nature 411, 603–606.
tion at ADCY7. Nat. Genet. 49, 186–192. Okolie, C.L., Rajendran, S.R.C.K., Udenigwe, C.C., Aryee, A.N.A.,
Maldonado-Gomez, M.X., Martinez, I., Bottacini, F., O’Callaghan, A., and Mason, B. 2017. Prospects of brown seaweed polysaccharides
Ventura, M., van Sinderen, D., Hillmann, B., Vangay, P., Knights, (BSP) as prebiotics and potential immunomodulators. J. Food
D., Hutkins, R.W., et al. 2016. Stable engraftment of Bifidobac- Biochem. 41, e12392.
terium iongum AH1206 in the human gut depends on individu- Ouwerkerk, J.P., de Vos, W.M., and Belzer, C. 2013. Glycobiome:
alized features of the resident microbiome. Cell Host Microbe bacteria and mucus at the epithelial interface. Best Pract. Res.
20, 515–526. Clin. Gastroenterol. 27, 25–38.
Marcobal, A., Southwick, A.M., Earle, K.A., and Sonnenburg, J.L. Pacheco, A.R., Barile, D., Underwood, M.A., and Mills, D.A. 2015.
2013. A refined palate: bacterial consumption of host glycans in The impact of the milk glycobiome on the neonate gut micro-
the gut. Glycobiology 23, 1038–1046. biota. Annu. Rev. Anim. Biosci. 3, 419–445.
Marteau, P. and Flourie, B. 2001. Tolerance to low-digestible car- Packey, C.D. and Sartor, R.B. 2008. Interplay of commensal and
bohydrates: symptomatology and methods. Br. J. Nutr. 85 Suppl pathogenic bacteria, genetic mutations, and immunoregulatory
1, S17–S21. defects in the pathogenesis of inflammatory bowel diseases. J.
Maslowski, K.M. and Mackay, C.R. 2011. Diet, gut microbiota and Intern. Med. 263, 597–606.
immune responses. Nat. Immunol. 12, 5–9. Piche, T., des Varannes, S.B., Sacher-Huvelin, S., Holst, J.J., Cuber,
Matsumoto, S., Nagaoka, M., Hara, T., Kimura-Takagi, I., Mistuyama, J.C., and Galmiche, J.P. 2003. Colonic fermentation influences
K., and Ueyama, S. 2004. Fucoidan derived from Cladosiphon oka- lower esophageal sphincter function in gastroesophageal reflux
muranus Tokida ameliorates murine chronic colitis through the disease. Gastroenterology 124, 894–902.
down-regulation of interleukin-6 production on colonic epithelial Pickard, J.M., Maurice, C.F., Kinnebrew, M.A., Abt, M.C., Schenten,
cells. Clin. Exp. Immunol. 136, 432–439. D., Golovkina, T.V., Bogatyrev, S.R., Ismagilov, R.F., Pamer, E.G.,
Mazurak, N., Broelz, E., Storr, M., and Enck, P. 2015. Probiotic the- Turnbaugh, P.J., et al. 2014. Rapid fucosylation of intestinal epi-
rapy of the irritable bowel syndrome: Why is the evidence still thelium sustains host-commensal symbiosis in sickness. Nature
poor and what can be done about it? J. Neurogastroenterol. Motil. 514, 638–641.
21, 471–485. Pigneur, B. and Sokol, H. 2016. Fecal microbiota transplantation in
McIlroy, J., Ianiro, G., Mukhopadhya, I., Hansen, R., and Hold, G.L. inflammatory bowel disease: the quest for the holy grail. Mucosal.
2018. Review article: the gut microbiome in inflammatory bowel Immunol. 9, 1360–1365.
disease-avenues for microbial management. Aliment. Pharmacol. Powrie, F., Leach, M.W., Mauze, S., Menon, S., Caddle, L.B., and
Ther. 47, 26–42. Coffman, R.L. 1994. Inhibition of Th1 responses prevents inflam-
Mimura, T., Rizzello, F., Helwig, U., Poggioli, G., Schreiber, S., Talbot, matory bowel disease in scid mice reconstituted with CD45RBhi
+
I.C., Nicholls, R.J., Gionchetti, P., Campieri, M., and Kamm, M.A. CD4 T cells. Immunity 1, 553–562.
2004. Once daily high dose probiotic therapy (VSL#3) for main- Rios-Covian, D., Ruas-Madiedo, P., Margolles, A., Gueimonde, M.,
taining remission in recurrent or refractory pouchitis. Gut 53, de Los Reyes-Gavilan, C.G., and Salazar, N. 2016. Intestinal short
108–114. chain fatty acids and their link with diet and human health. Front.
Molodecky, N.A., Soon, I.S., Rabi, D.M., Ghali, W.A., Ferris, M., Microbiol. 7, 185.
Chernoff, G., Benchimol, E.I., Panaccione, R., Ghosh, S., Barkema, Rivero-Urgell, M. and Santamaria-Orleans, A. 2001. Oligosaccha-
H.W., et al. 2012. Increasing incidence and prevalence of the in- rides: application in infant food. Early Hum. Dev. 65 Suppl,
flammatory bowel diseases with time, based on systematic review. S43–S52.
Gastroenterology 142, 46–54 e42; quiz e30. Roberfroid, M.B. 2000. Prebiotics and probiotics: are they functional
Mussatto, S.I. and Mancilha, I.M. 2007. Non-digestible oligosac- foods? Am. J. Clin. Nutr. 71, S1682–S1687; discussion S1688–
charides: A review. Carbohydrate Polymers 68, 587–597. S1690.
Najafzadeh, M., Reynolds, P.D., Baumgartner, A., Jerwood, D., and Sadowsky, M.J., Staley, C., Heiner, C., Hall, R., Kelly, C.R., Brandt,
Anderson, D. 2007. Chaga mushroom extract inhibits oxidative L., and Khoruts, A. 2017. Analysis of gut microbiota - An ever
DNA damage in lymphocytes of patients with inflammatory bowel changing landscape. Gut Microbes 8, 268–275.
disease. Biofactors 31, 191–200. Sanchez, B., Delgado, S., Blanco-Miguez, A., Lourenco, A., Gueimonde,
Ng, K.M., Ferreyra, J.A., Higginbottom, S.K., Lynch, J.B., Kashyap, M., and Margolles, A. 2017. Probiotics, gut microbiota, and their
P.C., Gopinath, S., Naidu, N., Choudhury, B., Weimer, B.C., influence on host health and disease. Mol. Nutr. Food Res. 61,
Monack, D.M., et al. 2013a. Microbiota-liberated host sugars fa- 1600240.
cilitate post-antibiotic expansion of enteric pathogens. Nature Sanders, M.E. 2008. Probiotics: definition, sources, selection, and
502, 96–99. uses. Clin Infect Dis 46 Suppl 2, S58–S61; discussion S144–S151.
Ng, S.C., Tang, W., Ching, J.Y., Wong, M., Chow, C.M., Hui, A.J., Sanders, M.E. 2016. Probiotics and microbiota composition. BMC
Wong, T.C., Leung, V.K., Tsang, S.W., Yu, H.H., et al. 2013b. Med. 14, 82.
Incidence and phenotype of inflammatory bowel disease based Sangwan, V., Tomar, S.K., Singh, R.R., Singh, A.K., and Ali, B. 2011.
on results from the Asia-pacific Crohn’s and colitis epidemiology Galactooligosaccharides: novel components of designer foods.
study. Gastroenterology 145, 158–165.e2. J. Food Sci. 76, R103–R111.
O’Shea, C.J., O’Doherty, J.V., Callanan, J.J., Doyle, D., Thornton, K., Senthilkumar, K., Manivasagan, P., Venkatesan, J., and Kim, S.K.
198 Eom et al.

2013. Brown seaweed fucoidan: biological activity and apoptosis, gut homeostasis. World J. Gastroenterol. 20, 15163–15176.
growth signaling mechanism in cancer. Int. J. Biol. Macromol. Vahabnezhad, E., Mochon, A.B., Wozniak, L.J., and Ziring, D.A. 2013.
60, 366–374. Lactobacillus bacteremia associated with probiotic use in a pedi-
Shanahan, F. 2017. Editorial: probiotics in inflammatory bowel atric patient with ulcerative colitis. J. Clin. Gastroenterol. 47,
disease-wrong organisms, wrong disease, or flawed concepts? 437–439.
Aliment. Pharmacol. Ther. 46, 632–633. Van den Abbeele, P., Belzer, C., Goossens, M., Kleerebezem, M., De
Shang, L., Fukata, M., Thirunarayanan, N., Martin, A.P., Arnaboldi, Vos, W.M., Thas, O., De Weirdt, R., Kerckhof, F.M., and Van de
P., Maussang, D., Berin, C., Unkeless, J.C., Mayer, L., Abreu, M.T., Wiele, T. 2013. Butyrate-producing Clostridium cluster XIVa
et al. 2008. Toll-like receptor signaling in small intestinal epithe- species specifically colonize mucins in an in vitro gut model.
lium promotes B-cell recruitment and IgA production in lamina ISME J. 7, 949–961.
propria. Gastroenterology 135, 529–538. Van der Sluis, M., De Koning, B.A., De Bruijn, A.C., Velcich, A.,
Shi, L., Lin, Q., Yang, T., Nie, Y., Li, X., Liu, B., Shen, J., Liang, Y., Meijerink, J.P., Van Goudoever, J.B., Buller, H.A., Dekker, J.,
Tang, Y., and Luo, F. 2016. Oral administration of Lentinus edodes Van Seuningen, I., Renes, I.B., et al. 2006. Muc2-deficient mice
beta-glucans ameliorates DSS-induced ulcerative colitis in mice spontaneously develop colitis, indicating that MUC2 is critical
via MAPK-Elk-1 and MAPK-PPARgamma pathways. Food Funct. for colonic protection. Gastroenterology 131, 117–129.
7, 4614–4627. Verghese, M., Walker, L.T., Shackelford, L., and Chawan, C.B. 2005.
Sicard, J.F., Le Bihan, G., Vogeleer, P., Jacques, M., and Harel, J. 2017. Inhibitory effects of nondigestible carbohydrates of different chain
Interactions of intestinal bacteria with components of the in- lengths on azoxymethane-induced aberrant crypt foci in Fisher
testinal mucus. Front. Cell. Infect. Microbiol. 7, 387. 344 rats. Nutr. Res. 25, 859–868.
Slavin, J. 2013. Fiber and prebiotics: mechanisms and health benefits. Videla, S., Vilaseca, J., Antolin, M., Garcia-Lafuente, A., Guarner, F.,
Nutrients 5, 1417–1435. Crespo, E., Casalots, J., Salas, A., and Malagelada, J.R. 2001. Die-
Sonnenburg, E.D., Zheng, H., Joglekar, P., Higginbottom, S.K., Fir- tary inulin improves distal colitis induced by dextran sodium
bank, S.J., Bolam, D.N., and Sonnenburg, J.L. 2010. Specificity of sulfate in the rat. Am. J. Gastroenterol. 96, 1486–1493.
polysaccharide use in intestinal bacteroides species determines Weingarden, A., Gonzalez, A., Vazquez-Baeza, Y., Weiss, S., Hum-
diet-induced microbiota alterations. Cell 141, 1241–1252. phry, G., Berg-Lyons, D., Knights, D., Unno, T., Bobr, A., Kang, J.,
Staley, C., Kelly, C.R., Brandt, L.J., Khoruts, A., and Sadowsky, M.J. et al. 2015. Dynamic changes in short- and long-term bacterial
2016. Complete microbiota engraftment is not essential for re- composition following fecal microbiota transplantation for re-
covery from recurrent Clostridium difficile infection following current Clostridium difficile infection. Microbiome 3, 10.
fecal microbiota transplantation. MBio 7, e01965-16. Wells, M.L., Potin, P., Craigie, J.S., Raven, J.A., Merchant, S.S., Helli-
Strober, W. and Fuss, I.J. 2011. Proinflammatory cytokines in the well, K.E., Smith, A.G., Camire, M.E., and Brawley, S.H. 2017.
pathogenesis of inflammatory bowel diseases. Gastroenterology Algae as nutritional and functional food sources: revisiting our
140, 1756–1767. understanding. J. Appl. Phycol. 29, 949–982.
Sun, Y. and O’Riordan, M.X. 2013. Regulation of bacterial patho- Whelan, K. and Quigley, E.M. 2013. Probiotics in the management
genesis by intestinal short-chain fatty acids. Adv. Appl. Microbiol. of irritable bowel syndrome and inflammatory bowel disease.
85, 93–118. Curr. Opin. Gastroenterol. 29, 184–189.
Sunkara, L.T., Jiang, W., and Zhang, G. 2012. Modulation of anti- Yamamoto, S. and Ma, X. 2009. Role of Nod2 in the development
microbial host defense peptide gene expression by free fatty acids. of Crohn’s disease. Microbes Infect. 11, 912–918.
PLoS One 7, e49558. Yamamoto, A., Itoh, T., Nasu, R., and Nishida, R. 2013. Effect of
Swennen, K., Courtin, C.M., and Delcour, J.A. 2006. Non-digestible sodium alginate on dextran sulfate sodium- and 2,4,6-trinitro-
oligosaccharides with prebiotic properties. Crit. Rev. Food Sci. benzene sulfonic acid-induced experimental colitis in mice. Phar-
Nutr. 46, 459–471. macology 92, 108–116.
Takaishi, H., Matsuki, T., Nakazawa, A., Takada, T., Kado, S., Asa- Yang, S.K., Yun, S., Kim, J.H., Park, J.Y., Kim, H.Y., Kim, Y.H., Chang,
hara, T., Kamada, N., Sakuraba, A., Yajima, T., Higuchi, H., et al. D.K., Kim, J.S., Song, I.S., Park, J.B., et al. 2008. Epidemiology
2008. Imbalance in intestinal microflora constitution could be of inflammatory bowel disease in the Songpa-Kangdong district,
involved in the pathogenesis of inflammatory bowel disease. Int. Seoul, Korea, 1986-2005: a KASID study. Inflamm. Bowel Dis.
J. Med. Microbiol. 298, 463–472. 14, 542–549.
Ten Bruggencate, S.J., Bovee-Oudenhoven, I.M., Lettink-Wissink, Zenewicz, L.A., Antov, A., and Flavell, R.A. 2009. CD4 T-cell dif-
M.L., Katan, M.B., and Van Der Meer, R. 2004. Dietary fructo- ferentiation and inflammatory bowel disease. Trends Mol. Med.
oligosaccharides and inulin decrease resistance of rats to sal- 15, 199–207.
monella: protective role of calcium. Gut 53, 530–535. Zhu, Y., Michelle Luo, T., Jobin, C., and Young, H.A. 2011. Gut mi-
Tojo, R., Suarez, A., Clemente, M.G., de los Reyes-Gavilan, C.G., crobiota and probiotics in colon tumorigenesis. Cancer Lett.
Margolles, A., Gueimonde, M., and Ruas-Madiedo, P. 2014. Intes- 309, 119–127.
tinal microbiota in health and disease: role of bifidobacteria in

You might also like