You are on page 1of 14

Best Practice & Research Clinical Gastroenterology 27 (2013) 59–72

Contents lists available at SciVerse ScienceDirect

Best Practice & Research Clinical


Gastroenterology

Nutrition, the gut microbiome and the metabolic


syndrome
Petia Kovatcheva-Datchary, PhD, Postdoctoral Fellow *,
Tulika Arora, PhD, Postdoctoral Fellow 1
Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory, Department of
Molecular and Clinical Medicine, University of Gothenburg, Bruna Straket 16, 413 45 Gothenburg, Sweden

a b s t r a c t
Keywords:
Gut microbiota Metabolic syndrome is a lifestyle disease, determined by the inter-
Metabolic syndrome play of genetic and environmental factors. Obesity is a significant risk
Obesity factor for development of the metabolic syndrome, and the preva-
Prebiotic
lence of obesity is increasing due to changes in lifestyle and diet.
Probiotic
Recently, the gut microbiota has emerged as an important contrib-
utor to the development of obesity and metabolic disorders, through
its interactions with environmental (e.g. diet) and genetic factors.
Human and animal studies have shown that alterations in intestinal
microbiota composition and shifts in the gut microbiome towards
increased energy harvest are associated with an obese phenotype.
However, the underlying mechanisms by which gut microbiota af-
fects host metabolism still need to be defined.
In this review we discuss the complexity surrounding the in-
teractions between diet and the gut microbiota, and their connec-
tion to obesity. Furthermore, we review the literature on the effects
of probiotics and prebiotics on the gut microbiota and host meta-
bolism, focussing primarily on their anti-obesity potential.
Ó 2013 Elsevier Ltd. All rights reserved.

Introduction

Diet and lifestyle are crucial factors that influence the susceptibility of humans to metabolic dis-
eases. Metabolic syndrome is a modern concept and is defined as a combination of metabolic and
medical disorders such as obesity, elevated fasting glucose, high blood pressure and dyslipidemia [1,2].

* Corresponding author. Tel.: þ46 31 3428672; fax: þ46 31 82 3762.


E-mail addresses: Petia.Kovatcheva@wlab.gu.se (P. Kovatcheva-Datchary), Tulika.Arora@wlab.gu.se (T. Arora).
1
Tel.: þ46 31 3428672; fax: þ46 31 82 3762.

1521-6918/$ – see front matter Ó 2013 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.bpg.2013.03.017
60 P. Kovatcheva-Datchary, T. Arora / Best Practice & Research Clinical Gastroenterology 27 (2013) 59–72

The rates of occurrence of these conditions and interactions with each other differ between sex, age
and ethnicities. However, diagnosis with any component of the metabolic syndrome increases the risk
of developing cardiovascular disease (CVD) and type 2 diabetes mellitus (T2DM) [3].
The recent rise in obesity is regarded as the triggering factor for expansion of the metabolic syn-
drome. Although genetics play a role in the development of obesity, environmental factors are thought
to be responsible for the recent dramatic increase in the prevalence of obesity. Obesity results from an
imbalance between energy intake and expenditure. Growing evidence reveals the importance of the
gut microbiota in the development of obesity (obesity pathogenesis). Thus dietary strategies to
manipulate the gut microbiota, particularly the use of probiotics and prebiotics as therapeutics, are
proposed for obesity and metabolic syndrome management (Fig. 1) [4].
The human gut harbours an immense assemblage of microorganisms, the gut microbiota, which
comprise a population of 1014 cells. Their collective genome, termed the metagenome or microbiome,
outnumbers the human genome by 150-fold [5]. Interestingly, this metabolic repertoire delicately
affects our physiology with functions that we have not to evolve on our own. Gut microbiota possess an
array of activities associated with utilization of non-digestible dietary carbohydrates and host-derived
glycoconjugates (e.g. mucin), deconjugation and dehydroxylation of bile acids, biosynthesis of vitamins
(K and B group) and isoprenoids, and metabolism of amino acids and xenobiotics. Based on this col-
lective metabolic potential, the gut microbiota can be viewed as a separate ‘microbial organ’ [6].
In this review, recent advances in understanding the role of the gut microbiota in the development
of obesity and potential therapeutic applications will be explored.

Nutrition and gut microbiota maturation

The interactions between diet and the gut microbiota in mammals are extremely complex, and any
major change in lifestyle or diet is likely to affect microbial stability. Diet is a primary determinant in
the development of the microbiota colonization pattern from the first stages of life. Exclusive
breastfeeding results in an infant gut microbiota composition that is enriched in bifidobacteria and
lactic acid bacteria, while formula feeding results in a more diverse community that is dominated by

Fig. 1. Interaction between diet and gut microbiota affects host metabolism. Dietary manipulation with probiotics and prebiotics
alters the composition and metabolic capacity of gut microbiota. Dietary manipulation in obesity with prebiotics and probiotics
changes gut microbiota by favouring bacteria beneficial to the host and enhances the production of short chain fatty acids (SCFAs) –
acetate, propionate and butyrate. These result in decreased lipogenesis, reduced inflammation and oxidative stress in liver;
decreased adipogenesis, and reduced adipocyte size and number in adipose tissue; increased production of gut hormones and
intestinal transit in the large intestine; reduced appetite in the brain. GLP-1: Glucagon like peptide-1, PYY: Peptide YY.
P. Kovatcheva-Datchary, T. Arora / Best Practice & Research Clinical Gastroenterology 27 (2013) 59–72 61

bifidobacteria, Bacteroides spp., Clostridium spp., and facultative anaerobes [7,8]. Breast milk is rich in
oligosaccharides, which are known to act as substrates for fermentation in the distal gut and promote
the growth of beneficial microbes as bifidobacteria [7]. The introduction of solid food leads to a large
compositional shift into the intestinal microbiota composition [8,9]. Recently, the impact of diet on the
gut microbiota composition at the stage of infant weaning has been studied. Comparison of the fecal
microbiota by 16S rDNA sequencing of children from Burkina Faso (BF) consuming a rural African diet
with children from Italy consuming a modern Western diet showed no significant differences in the
microbiota composition between the two cohorts during the breastfeeding period [10]. However,
weaning resulted in significant changes in the gut microbiota in both groups. In the BF children,
enrichment in Bacteroidetes, with a significant abundance of bacteria from the genera Prevotella and
Xylanibacter, was observed. These functional groups encode enzymes for hydrolysis of cellulose and
xylan, thus enabling degradation of polysaccharides. In addition, fecal levels of short chain fatty acids
(SCFAs), the main products of polysaccharide fermentation, were higher in BF children. A relative
depletion of Firmicutes was also observed in the BF children. In the Italian children, these features were
completely absent.
Parallel ontogenetic changes of the microbiome among Malawian, Venezuelan and American
populations from infants to adults, associated with differences in diet, culture and lifestyle, have been
reported by Gordon’s group [11]. Greatest interpersonal variation, both in species and genes level, has
been recorded in the infant microbiome, but still bifidobacteria was the dominant microbial group. The
functional repertoire of the infant microbiome of the three populations was shaped with genes
necessary for folate biosynthesis with a later shift to folate metabolism. Genes necessary for synthesis
of vitamins, like B1, B7 and especially B12, were more enriched in the adult microbiome, together with
genes involved in fermentation, methanogenesis, and metabolism of certain amino acids. Interestingly,
the authors observed that the gut microbiomes of the Malawian and Venezuelan populations, which
consume a ‘rural’ diet, were enriched in Prevotella spp., similar to the observation of De Filipo et al [10]
in the Burkina Faso cohort. In the American population, since the diet is higher in protein, the
microbiome is enriched with genes necessary for breaking down amino acids. Moreover, the micro-
biome of the American tends to shift to a more Bacteroides-enriched gut community [11]. These results
support previous findings that carnivore microbiomes are enriched in protein degradation genes, while
herbivore microbiomes are enriched in genes necessary to break down starch [12].
Together, these data confirm that nutrition is a driving factor in shaping gut microbiota composition
and its functional maturation, from birth to adulthood. Nevertheless, more studies are needed to
determine what factors or events during gut microbiota maturation might contribute to the devel-
opment of metabolic disorders.

Gut microbiome and obesity

The development of obesity is a multifactorial process involving genetic susceptibility and envi-
ronmental factors, such as lifestyle and inappropriate diet. Still the root of obesity aetiology is an
imbalance between food intake and energy expenditure.
Recently, the gut microbiota has been suggested as a driving force in the pathogenesis of metabolic
disease and particularly obesity. Bäckhed and co-workers showed that mice raised in absence of mi-
croorganisms, termed germ-free (GF), had less total body fat than mice that were colonized with a
normal microbiota at birth, termed conventionally raised (CONV-R), despite the fact that GF animals had
higher caloric intake [13]. Colonization of GF mice with cecal content from CONV-R donors (resulting in
‘conventionalized’ (CONV-D) mice) resulted in increased total body fat without any increase in food
consumption or energy expenditure. The CONV-D animals also displayed impaired glucose metabolism,
increased levels of circulating leptin and glucose, and adipocyte hypertrophy after the two-week
colonization period [13]. In a following study, Bäckhed and co-workers demonstrated that GF mice fed a
high-fat, high-carbohydrate Western diet for eight weeks gained significantly less weight than CONV-R
mice and were protected against diet-induced glucose intolerance and insulin resistance [14]. These
findings suggest a relationship between nutrition, gut microbiota and energy homeostasis.
Furthermore, several studies have suggested that the obese microbiome has an altered composition
and functional repertoire. The development of obesity in genetically obese leptin deficient ob/ob mice
62 P. Kovatcheva-Datchary, T. Arora / Best Practice & Research Clinical Gastroenterology 27 (2013) 59–72

has been associated with a reduction in the abundance of Bacteroidetes and a proportional increase in
Firmicutes, two of the major bacterial phyla in the gut microbiota [15]. Similar changes in microbiota
composition have been observed in wild-type mice fed a high-fat/high-sugar Western diet [16].
However, comparisons of the abundance of the two major bacterial phyla in obese vs. lean microbiome
in animals and humans, have produced conflicting results. While several studies have reported similar
increases in the ratio between Firmicutes and Bacteroidetes in obese individuals [17–19], other studies
have not observed a change in the Firmicutes/Bacteroidetes ratio [20–22]. For example, one study
reported reductions in Bifidobacterium spp. and Bacteroidetes and increases in specific members of
Firmicutes (e.g. Staphylococcus) and Proteobacteria (e.g. Enterobacteriaceae) in overweight pregnant
women [19], while another study found enrichment in Prevotellaceae, a group within the Bacteroidetes
phylum, in obese individuals [20]. These disparities highlight the importance of considering factors
such as diet, age, degree of obesity, demographic geography and population size, as well as technique
and methodology used to profile the gut microbiota, when comparing studies.
Nevertheless, an altered functional repertoire has been associated with the obese microbiome.
Turnbaugh and co-workers identified a ‘core microbiome’ of microbial genes shared among individuals
and found that variations from that core are associated with obesity [23]. Further metagenomic and
systems biology-based approaches reached a similar conclusion, indicating that obese microbiomes
have reduced taxonomic richness and a less modular metabolic network in comparison with lean
microbiomes [5,24,25]. Comparable observations have been reported in a study comparing the
microbiomes of pregnant women in the first and third trimester. The microbiomes from women in the
third trimester were characterized by reduced microbial diversity and enrichment in Proteobacteria
and Actinobacteria. Moreover, the transfer of fecal microbiota from women in the third trimester to
germ-free mice induced symptoms of metabolic syndrome in the mice [26]. We might need more
studies focused on identifying important microbial genes instead of on characterizing gut microbiota
composition in order to describe the obese microbiome and further predict its response after thera-
peutics treatment. More mechanistic studies in animals are needed in order to elucidate functions of
specific gut microbes on the host and to better understand their interactions with environmental
factors (e.g. diet).

Dietary manipulation in obesity

As discussed earlier, it is possible to manipulate the gut microbiota composition with diet. The
major dietary ingredients studied in recent years include prebiotics, probiotics and synbiotics. The
interaction of these dietary ingredients with the microbiota is accompanied by physiological changes
in the host. We will focus on the metabolic interaction of probiotics and prebiotics with the host and
their anti-obesity potential (Fig. 1).

Probiotics

Metabolic interactions of probiotics with gut microbiota

Probiotics are dietary supplements consumed in the form of fermented milk products, fermented
foods or as drugs. Probiotics are defined as the live microorganisms that, when administered in
adequate amounts, confer health benefits to the host [27]. Probiotic supplementation results in
enrichment of the probiotic species in intestinal contents. It also results in alterations in the compo-
sition of gut microbiota and microbial metabolites, like SCFAs, in both murine models [28] and humans
[29]. Recent applications of metagenomic and associated techniques have provided insight into the
interaction of probiotics with the host gut microbiome. To reduce the complexity of studying crosstalk
between probiotic species and millions of resident bacteria, GF mice were colonized with a simplified
microbiota or mono- or bi-associated with representative bacterial species. Their interaction with
probiotic bacteria was determined in this simplified setting by either global transcriptomics or global
metabolite analysis as discussed in the following section.
With regard to energy homeostasis, probiotics have been reported to expand the carbohydrate
utilization properties of host microbiota members. In GF mice monocolonized with Bacteroides
P. Kovatcheva-Datchary, T. Arora / Best Practice & Research Clinical Gastroenterology 27 (2013) 59–72 63

thetaiotamicron, co-colonization with Bifidobacterium longum resulted in upregulation of a gene locus


in B. thetaiotamicron involved in the hydrolysis of xylose containing glycans. In contrast, co-
colonization with Lactococcus casei upregulated a different set of genes encoding hexosaminidases
and arabinosidases [30]. Enhanced carbohydrate utilization from non-digestible plant polysaccharides
is known to result in higher production of SCFAs that contribute to total energy in the host. In GF mice
monoassociated with the probiotic strains Lactobacillus plantarum WCFS1 [31] and Lacobacillus john-
sonii NCC533 [32], an upregulation of genes involved in carbohydrate transport and metabolism was
observed. While increased efficiency to hydrolyse different complex sugars in L. plantarum WCFS1
resulted in higher production of fumarate and alcohol [31], it led to a longer colonization period of L.
johnsonii NCC533 in the mouse gut [32]. Consumption of fermented milk containing multiple probiotic
strains led to enrichment of enzymes catalysing carbohydrates into propionate in both monozygotic
twins and gnotobiotic mice containing a model human microbiota community [33].
Global metabolite profiling has also demonstrated that probiotics may affect the gut microbiome. L.
paracasei altered levels of lipid species, creatine and glutathione implicating changes in lipid synthesis,
nutrient absorption and oxidative stress in gnotobiotic mice [34]. L. paracasei and L. rhamnosus
administration decreased the acetate:propionate ratio and increased catabolism of amino acids in
humanized gnotobiotic mice. Changes in bile acid metabolism were observed with L. paracasei
exclusively [35]. Thus, probiotics can alter both gene expression and function of the gut microbiota,
thereby altering energy homeostasis and physiological functions in the host.

Anti-obesity potential of probiotics

With advancing knowledge of how probiotics interact with the gut microbiome, there is an
increasing interest in exploring the anti-obesity potential of probiotics. Our main focus is to include the
studies that have reported effects of probiotic supplementation on weight loss, energy intake or
epididymal fat reduction (Table 1).
Conjugated linoleic acid (CLA) is a naturally occurring derivative of linoleic acid found in foods and
dairy products and has been shown to increase metabolic rate in mice [36]. The CLA-producing pro-
biotic strain L. rhamnosus PL60 has been reported to reduce body weight gain and white adipose tissue
mass with no effect on food intake in high-fat diet fed mice. The effect was coupled to higher
expression of uncoupling protein-2 (UCP2), while expression of fatty acid synthase (fas) and serum
leptin and glucose levels were reduced [37]. Another probiotic strain that produces CLA, L. plantarum
PL62, also resulted in reduced body weight gain and glucose levels in diet-induced obese mice [38].
Probiotics have been shown to reduce adipocyte size in different adipose depots [39,40], which is
considered an important parameter in assessing their anti-obesity potential. The putative mechanisms
put forth are increased fecal excretion of neutral sterols and bile acids, decreased lymphatic absorption
of triglycerides, phospholipids and cholesterol [41], or increased lipolysis [42]. In the 3T3-L1 cell line,
incubation with L. plantarum KY1032 cell free extract resulted in reduced adipogenesis [43], and in-
cubation with the insoluble fraction from fermented kefir resulted in reduced adipocyte differentiation
[44]. Administration of L. paracasei NCC2461 to rats increased sympathetic nerve activity in white and
brown adipose tissue, resulting in higher thermogenesis in brown adipose tissue and increased
lipolysis in white adipose tissue [45]. Supplementation with L. paracasei F19 resulted in reduced total
body fat and decreased triglyceride levels in different lipoprotein fractions in mice fed high-fat diet
[46]. In addition, both GF and CONV-R mice supplemented with L. paracasei F19 had increased serum
levels of Angiopoietin-like 4 (ANGPTL4), a microbially regulated lipoprotein lipase inhibitor that reg-
ulates lipid deposition into adipocytes [14,46]. Administration of L. paracasei F19 and L. acidophilus
NCFB1748 to GF mice resulted in enrichment of probiotic strains in the ileum compared to the colon
and upregulation of insulin-sensitizing hormones, adipsin and adiponectin. Decreased expression of
resistin-like b, known to induce insulin resistance, was also reported [47]. Apoe/ mice that were
supplemented with L. reuteri ATCC exhibited reduced body weight gain, reduced adipose and liver
weights, and increased expression of carnitine palmitoyl transferase1a (cpt1a), suggesting higher he-
patic b-oxidation [48].
There are relatively few studies that couple changes in microbiota composition upon probiotic
supplementation with anti-obesity functions. Supplementation of L. rhamnosus GG and L. sakei NR28
Table 1
Anti-obesity effects of probiotic supplementation in animal and human studies.

Animal Diet Probiotic strain Dose Mode of Effects Reference


strain or delivery
human
1. C57/BL6J HFD L. rhamnosus PL60 1  107or PBS YBWG, YAT mass, [37]
mice 1  109 cfu/day No change in FI
2. C57/BL6J HFD L. plantarum PL62 1  107or PBS YBWG, YAT, [38]
mice 1  109 cfu/day No change in FI
3. Sprague– HFD L. gasseri SBT2055 6  107 cfu/g diet Fermented YAdipocyte size, [39]
Dawley milk powder No change in BWG,
rats added to diet AT mass
in 20%
concentration
4. Zucker HFD L. gasseri SBT2055 6  107 cfu/g diet Fermented YAT, YAdipocyte [41]
obese and milk powder size in lean rats,
lean rats added to diet No effect on BWG,
in 20% FI
concentration
5. C57/BL6 HFD L. plantarum 14 1  108 cfu/mouse PBS YAdipocyte size, [40]
mice YAT, No effect on
BWG
6. Sprague– HCD L. gasseri BNR 17 2  109/ml PBS YBWG, YAT, [104]
Dawley rats No effect on FI
7 10
7. db/db Regular L. gasseri BNR 17 10 –10 cfu/day PBS YBWG, YFI [105]
mice diet
9. Sprague– HFD VSL#3 (Lactobacilli, 1.5  109 Water YAT, No change in [66]
Dawley Bifidobacterium cfu/mouse BWG, FI
rats spp. & S. salivarius
ssp. thermophilus)
10. Wistar HFD L. paracasei ST11 5  108/ml Water YBWG, YAT, [45]
rats (NCC2461) No change in FI
11. Sprague– HFD B. pseudocatenulatum 108–109 cfu PBS YBWG, YAT, [106]
Dawley SPM 1204, B. longum No change in FI
rats SPM 1205, and
B. longum SPM 1207
12. Wistar HFD B. longum 2  109/ml Saline YBWG, YAT, [107]
rats Yblood
glucose
14. Sprague– HFD L. plantarum DSM 109 cfu/day Water YBWG, YAT, [108]
Dawley 15313 Yleptin
rats
15. C57/BL6J HFD L. acidophilus 5  107–9  107/ml Yoghurt [Bifidobacterium [50]
mice NCDC13 spp., No change
in FI, BWG, AT
16. C57/BL6J Normal L. sakei NR28 or 108 cfu/10 ml Oral gavage YFirmicutes – [49]
mice Chow L. rhamnosus GG particularly
Clostridium
cluster XIVa,
YBWG, YAT
17. Balb/c Normal L. ingluviei 4  1010 Gastric [Firmicutes, [51]
mice Chow lavage Lactobacillus spp.,
[BWG
18. Apoe/ High-fat L. reuteri ATCC PTA 109 cfu/mouse Drinking YBWG, YAT, Yliver [48]
mice western 4659, DSM 17938 per day water weight
diet – 0.2% (DSM), L6798
cholesterol
19. Healthy, Regular L. gasseri SBT2055 5  1010 cfu/100 g Fermented YVisceral, [52]
overweight diet product subcutaneous fat,
subjects YBWG, YBMI
20. Infants Formula L. rhamnosus GG 107 cfu/g Formula diet [Length, [weight [53]
diet
21. Pregnant Regular L. rhamnosus GG 1  1010 Capsules YBWG in children [54]
women diet

AT: Adipose tissue, BWG: Body weight gain, FI: Food intake, HCD: High-carbohydrate diet, HFD: High-fat diet, PBS: Phosphate
buffered saline.
P. Kovatcheva-Datchary, T. Arora / Best Practice & Research Clinical Gastroenterology 27 (2013) 59–72 65

reduced the relative abundance of both Firmicutes and Clostridium cluster XIVa in the small intestine of
mice and was accompanied by reduced body weight gain, epididymal fat mass and expression of
hepatic stearoyl-CoA desaturase-1 (scd-1), fas and acetyl-CoA carboxylase (acc) enzymes involved in
lipogenesis [49]. However, supplementation with L. acidophilus NCDC13 in diet-induced obese mice
increased total bifidobacteria in caecal contents as well as feces but did not decrease adiposity [50]. In
yet another study, inoculation with L. ingluviei enriched total fecal Lactobacillus spp. and Firmicutes in
mice and, in contrast to other studies, resulted in increased body weight gain, liver weight, and
metabolism [51].
In healthy overweight subjects, administration of L. gasseri SBT2055 resulted in reduction of
abdominal visceral and subcutaneous fat [52]. Supplementation of L. rhamnosus GG in the infant for-
mula for six months resulted in better growth with higher weight gain [53]. However, in a follow up
study, pre- and postnatal administration of L. rhamnosus GG inhibited excessive weight gain in children
[54]. Thus, the physiological effects of probiotics are highly strain-dependant. The variations in out-
comes between different studies appear to be due to choice of probiotic strain, route of administration
and length of study.

Probiotic effect on liver function

Hepatic steatosis is closely linked to metabolic syndrome. It is characterized by aberrant lipid


storage in liver and subsequent hepatic inflammation. Ischemia/reperfusion is a model of acute liver
injury which leads to disruption of gut microbiota composition with increases in Enterococci and
Enterobacteria and concomitant decreases in Lactobacillus spp. and Bifidobacterium spp., eventually
resulting in higher plasma endotoxin levels. In addition, bacterial translocation to the kidney has been
reported [55]. Colonization of GF mice with microbiota from hyperglycaemic mice has been shown to
contribute to the development of fatty liver disease independent of obesity [56]. Probiotics have also
been implicated in the improvement of liver functions. These effects are mediated by the role of
probiotic bacteria in normalization of intestinal microbiota composition, immunomodulation and
maintenance of gut barrier function [57].
L. paracasei F19 supplementation protected rats from ischemia/reperfusion-induced liver injury by
restoring ileal lactobacilli and bifidobacteria numbers and intestinal barrier [58]. Supplementation
with different lactobacilli strains (L. acidophilus NM1; L. rhamnosus ATCC 53103, and L. rhamnosus DSM
6594 þ L. plantarum DSM 9843) [59] or with the combination of L. fermentum and B. catenulatum [60]
prevented bacterial translocation and decreased Enterobacteriaceae in a rat model of acute liver injury.
However, B. animalis increased the bacterial translocation to mesenteric lymph nodes with no effect on
liver damage [59]. L. rhamnosus GG ameliorated alcohol-induced liver disease by reducing inflam-
mation and oxidative stress in both liver and colon [61]. It also reduced alcohol-induced gut leakiness,
increased expression of ileal tight junction proteins maintaining the epithelial integrity [62], and
decreased endotoxemia preserving the gut barrier function [62]. The effects exerted by the probiotic
were found to be dependant on hypoxia-inducible factor [63]. Supplementation with VSL#3, another
multistrain probiotic, mediated a natural killer T cell-dependant improvement in diet-induced stea-
tosis and hepatic insulin signalling, resulting in improved insulin sensitivity [64]. VSL#3 supplemen-
tation also reduced c-jun kinase activity and hepatic lipogenesis in leptin deficient ob/ob mice [65]. The
antioxidative effects of VSL#3, coupled with reductions in inflammatory enzymes like inducible
nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2, have also been shown to protect against liver
damage in mice [66].

Prebiotics

Metabolic interactions of prebiotics with gut microbiota

Prebiotics are defined as dietary ingredients that promote ‘the selective stimulation of growth and/or
activity(ies) of one or a limited number of microbial genus(era)/species in the gut microbiota that
confer(s) health benefits to the host’ [67]. Dietary fibres are known to exhibit prebiotic effects as they are
utilized by specific bacteria, resulting in their proliferation, which are considered beneficial to the host.
66 P. Kovatcheva-Datchary, T. Arora / Best Practice & Research Clinical Gastroenterology 27 (2013) 59–72

The most studied prebiotic supplements are inulin and related variants such as fructooligosaccharides,
which have different degrees of polymerization. Inulin is generally bifidogenic in nature, i.e. it enhances
the growth of Bifidobacterium spp. and some Lactobacillus spp. in different murine models. This effect is
coupled with a reduction in body weight gain and improvements in glucose homeostasis [68–70] and
obesity-related inflammation called metabolic endotoxemia [71,72]. However, pyrosequencing revealed
that oligofructose feeding resulted in changes in more than 100 taxa, of which 16 taxa displayed more
than ten fold change [73]. One of the identified species was Akkermansia muciniphila, which was
negatively correlated with body weight [19]. Other studies have also shown that prebiotic fibre
decreased the Firmicutes to Bacteroidetes ratio in obese rats [74]. Supplementation of fungal chitin
glucan increased the number of bacteria closely related to Clostridium cluster XIVa including Roseburia
spp., which was accompanied by reduced weight gain and fat mass development [75]. Wheat derived
arabinoxylans also restored the levels of Bacteroides–Prevotella spp. and Roseburia spp. and markedly
increased caecal bifidobacteria content, in particular B. animalis lactis, in high-fat diet fed mice [76].
In healthy humans, the consumption of polydextrose and soluble corn fibre led to greater fecal
Clostridiaceae and Veillonellaceae and lower Eubacteriaceae. The abundance of Faecalibacterium, Phas-
colarctobacterium, and Dialister was greater following prebiotic intake [77]. Addition of inulin to the
diet increased Bifidobacterium spp. and Faecalibacterium prausnitzi and decreased Bacteroides intesti-
nalis, Bacteroides vulgatus and Propionibacterium in obese women [78]. Consumption of gal-
actooligosaccharides for 12 weeks induced increases in several lineages of Bifidobacterium spp. at the
expense of Bacteroides in healthy human subjects [79].

Anti-obesity potential of prebiotics

Prebiotics exhibit anti-obesity potential owing to their fermentation in distal gut and impact on gut
microbiota composition, which have different physiological prospects. The underlying mechanisms
driving the response are not clear. However, there are some links associated with production of SCFAs,
decrease in bacterial derived lipopolysaccharides and alteration in gut hormones production. The
major studies reflecting anti-obesity effects of prebiotics in both humans and animal models are
presented in Table 2.
The major effect of inulin supplementation appears to be its influence on production of gastroin-
testinal hormones like glucagon like peptide-1 (GLP-1), peptide YY (PYY), ghrelin and other related
peptide hormones both in rodents [70,74,80] and in humans [81–83]. These hormones modulate
several physiologic functions such as insulin secretion (incretin effect), gastrointestinal motility and
appetite regulation by modulating secretion of neuropeptides in major hypothalamic appetite centers.
These factors may all contribute to the anti-obesity potential of prebiotics. Microbial production of
SCFAs has been proposed to play a role in increasing secretion of gut hormones like GLP-1 [84,85].
Inulin supplementation has also been reported to reduce fat mass accumulation in different adipose
tissue depots and to affect adipose tissue metabolism. It reduces lipolysis in subcutaneous adipose
tissue and blunts the expression of GPR43, a G-protein coupled receptor that regulates adipogenesis in
diet-induced obese mice [86]. A link between the endocannabinoid system (eCB), changes in gut
microbiota composition and adipogenesis has also been suggested. The eCB system is overexpressed in
obesity [87]. Inulin supplementation in ob/ob mice decreased adiposity and was coupled with reduced
anadamide content and cannabinoid receptor-1 expression in adipose tissue [88].
Prebiotic supplementation has been shown to influence not only fat mass but also host energy ho-
meostasis. When gnotobiotic mice were colonized with microbiota from human infants and supple-
mented with L. paracasei and galactooligosaccharides, global metabolites analysis revealed increased
levels of glutamate, glutamine, branched-chain amino acids and alanine in the liver as well as accumu-
lation of hepatic glycogen, suggesting increased gluconeogenesis and glycogenesis [89]. It also increased
SCFAs production and excretion of protein breakdown metabolites [90]. The increased excretion of energy-
rich metabolites upon supplementation of inulin and beta-glucan was also shown in high-fat fed mice [91].
A role for prebiotic fibres in appetite regulation has also been demonstrated. The central hypo-
thalamic brain centers like arcuate nucleus, ventromedial hypothalamus, paraventricular nucleus and
brainstem regions involved in appetite regulation were shown to be differentially activated upon
prebiotic feeding. A satiating effect of resistant starch, supported by reduced neuronal activity in
P. Kovatcheva-Datchary, T. Arora / Best Practice & Research Clinical Gastroenterology 27 (2013) 59–72 67

Table 2
Anti-obesity effects of prebiotic supplementation in animal and human studies.

Animal strain Diet/period Prebiotic Dose Effects Reference


or human
1. Wistar rats, Regular Raftilose P95 100 g/kg diet Improve glycaemia and plasma [70]
male diet/six (OFS based) insulin, [plasma portal GLP-1, YFI
weeks
2. Wistar rats, HFD/five OFS 100 g/kg diet [plasma portal GLP-1, [serum GIP, [80]
male weeks YBWG
3. C57/BL6J mice, HFD/four OFS 100 g/kg diet Yfasted and fed blood glucose, YBWG [68]
male weeks
4. C57/BL6J mice, HFD/14 OFS Not described Restored Bifidobacterium spp. levels, [71]
male weeks improved blood glucose
5. ob/ob C57/BL6J HFD/eight OFS 0.3 g/mouse YFirmicutes, [Bacteroidetes, [73]
mice weeks per day YProteobacteria, [Verrucomicrobia,
[L-cell number, improved glucose
tolerance and lipid metabolism, YLPS
6. C57/BL6J mice, HFD/four AX 100 g/kg diet [Bifidobacterium spp. (e.g. B. animalis [76]
male weeks lactis), restored Roseburia spp. and
Bacteroides–Prevotella spp. levels,
YBWG, Yfat mass development, YAT
7. C57/BL6J mice, HFD/ weeks Chitin-glucan 100 g/kg diet [Firmicutes (Clostridium cluster XIVa – [75]
male fibre Roseburia spp.), YBWG, Yfat mass
development, Yfasting glucose,
improved glucose tolerance, Y hepatic
triglycerides, Y cholesterol
8. C57/BL6J mice, HFD/eight AXOS 75 g/kg diet [Bifidobacterium spp., YLactobacillus [109]
male weeks spp., YBWG, Ycaloric intake, Yfat mass,
[PYY, [GLP-1, YLPS
9. Lean and obese Regular Inulin-OFS 0%, 10% and 20% [Firmicutes, YBacteroidetes, dose [74]
JCR:LA-cp rats, diet/six Inulin–OFS mix dependant [Bifidobacterium spp. and
male weeks Lactobacillus spp., dose dependant
[satiety hormones
10. Men and Regular OFS 2  8.0 g/day [satiety after breakfast and dinner, [110]
women diet/two Yhunger and YFI following dinner
weeks
11. Men and Regular Orafti 2  8.0 g/day Yhunger, [satiety, [breath H2, [81]
women diet/two Synergy1 [GLP-1, [PYY
weeks
12. Men and Regular AXOS 2  5.0 g/day [Bifidobacterium spp., Yp-cresol urine [111]
women diet/three excretion
weeks
13. Men and Regular GOS 0.0 g, 2.5 g, GOS dose dependant [Bifidobacterium [79]
women diet/12 5.0 g, 10.0 g/day spp.
weeks
14. Men and Regular Chitin-glucan 1.5 g/day or YLDL oxidation, YLDL cholesterol [112]
women diet/six fibre 4.5 g/day
weeks
15. Obese Regular Inulin type 16 g/day [Actinobacteria (Bifidobacterium spp.), [78]
women diet/12 fructan [Firmicutes (Clostridium leptum group
weeks – Faecalibacterium prausnitzii),
YBacteroidetes
(B. intestinalis and B. vulgatus),
YPropionibacterium, YLPS
16. Men and Regular AXOS 0.0 g, 2.2 g, Dose dependant [Bifidobacterium spp., [113]
women diet/three 4.8 g/day Dose dependant [ferulic acid
weeks
17. Men and Regular GOS 2  4.0 g/day [Bifidobacterium spp. [114]
women diet/three
weeks

AT: Adipose tissue, AX: Arabinoxylan, AXOS: Arabinoxylan-oligosaccharide, BWG: Body weight gain, FI: Food intake, GIP: Gastric
inhibitory peptide, GLP-1: Glucagon like peptide-1, GOS: Galactooligosaccharide, HFD: High-fat diet, LDL: Low density lipo-
protein, LPS: lipopolysaccharide, OFS: Oligofructose, PYY: Peptide YY.
68 P. Kovatcheva-Datchary, T. Arora / Best Practice & Research Clinical Gastroenterology 27 (2013) 59–72

ventromedial and paraventricular hypothalamic nuclei, was shown [92]. Supplementation with beta-
glucan also resulted in reduced activity in the arcuate nucleus and related appetite centers, which
could explain the reduced weight gain in animals [91]. On the contrary, inulin supplementation was
shown to increase neuronal activity in the arcuate nucleus; however, a reduction in body weight gain
was consistently observed [93].

Prebiotics effect on liver function

Prebiotics may also ameliorate liver diseases by decreasing hepatic de novo lipogenesis [94]. A number
of studies have shown reductions in enzymes like acc, malic enzyme, ATP citrate lipase and fas upon
prebiotic supplementation [95–98]. However, other studies have not found any effect on fatty acid
synthase, but rather on enzymes expression involved in b-oxidation of lipids [99,100]. Reduction in free
fatty acid esterification to triglycerides in hepatocytes has also been reported [101]. Prebiotics may also
contribute in this regard by altering the acetate:propionate ratio [102,103]. As acetate is lipogenic in
nature, production of more propionate compared to acetate may decrease the hepatic lipogenic potential.

Conclusion

The prevalence of obesity and allied disorders such as metabolic syndrome are becoming a great
challenge for health care throughout the world. Diet and lifestyle are crucial factors influencing the
development and progression of obesity. Recent insights have examined obesity aetiology with a new
perspective and found that our own microbiota might be involved in the development of these disor-
ders. The plexus surrounding diet and the microbiome in mammals remains to be unravelled. More
controlled human and animal studies are necessary to clarify these complex interactions. One possibility
is the combination of metagenomic and metabolomic approaches in order to further elucidate the
metabolic interactions between diet, the microbiota and the host and to understand how the micro-
biome is altered in obesity. Those approaches, combined with more mechanistic studies in animal
models, will help to further describe in situ functions of distinct microbial groups or individual species of
the gut microbiota and to understand their roles in human physiology including metabolic diseases.

Practice points

 Nutrition is a driving factor in shaping gut microbiota composition and its functional
maturation from the early stages of life.
 The gut microbiota has been recognized as an important contributor to pathological condi-
tions such as obesity and metabolic disorders.
 Alterations of the composition and functional properties of the gut microbiota have been
associated with obesity.
 Increasing Bifidobacterium spp. by dietary means may have anti-obesity effects.

Research agenda

 The mechanisms by which the gut microbiota contributes to the development or mainte-
nance of obesity are not well understood.
 The underlying mechanisms by which probiotics and prebiotics exert their anti-obesity
effects need to be deduced.
 There is a need for well-controlled studies with regard to gut microbiota modulation, changes
in microbial metabolites and corresponding anti-obesity effects following probiotic
supplementation.
P. Kovatcheva-Datchary, T. Arora / Best Practice & Research Clinical Gastroenterology 27 (2013) 59–72 69

Acknowledgements

We thank Anita Wichmann for editing the manuscript and Anna Hallén for the graphical design. We
thank Fredrik Bäckhed for the helpful comments on the article.

References

[1] Eckel RH, Grundy SM, Zimmet PZ. The metabolic syndrome. The Lancet 2005;365(9468):1415–28.
[2] Tilg H, Kaser A. Gut microbiome, obesity, and metabolic dysfunction. The Journal of Clinical Investigation 2011;121(6):
2126–32.
[3] Semenkovich CF. Insulin resistance and atherosclerosis. The Journal of Clinical Investigation 2006;116(7):1813–22.
[4] Astrup A. Dietary management of obesity. Journal of Parenteral and Enteral Nutrition 2008;32(5):575–7.
[5] Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut microbial gene catalogue established by
metagenomic sequencing. Nature 2010;464(7285):59–65.
[6] O’Hara AM, Shanahan F. The gut flora as a forgotten organ. EMBO Reports 2006;7:688–93.
[7] Wall R, Ross RP, Ryan CA, Hussey S, Murphy B, Fitzgerald GF, et al. Role of gut microbiota in early infant development.
Clinical Medicine Insights: Pediatrics 2009;3:45–54.
[8] Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO. Development of the human infant intestinal microbiota. PLoS
Biology 2007;5(7):e177.
[9] Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R, et al. Succession of microbial consortia in the
developing infant gut microbiome. Proceedings of the National Academy of Sciences 2011;108(Suppl. 1):4578–85.
*[10] De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, et al. Impact of diet in shaping gut microbiota
revealed by a comparative study in children from Europe and rural Africa. Proceedings of the National Academy of
Sciences 2010;107(33):14691–6.
*[11] Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et al. Human gut microbiome viewed
across age and geography. Nature 2012;486(7402):222–7.
[12] Muegge BD, Kuczynski J, Knights D, Clemente JC, González A, Fontana L, et al. Diet drives convergence in gut
microbiome functions across mammalian phylogeny and within humans. Science 2011;332(6032):970–4.
*[13] Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, et al. The gut microbiota as an environmental factor that
regulates fat storage. Proceedings of the National Academy of Sciences 2004;101(44):15718–23.
*[14] Backhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity
in germ-free mice. Proceedings of the National Academy of Sciences 2007;104(3):979–84.
[15] Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proceedings
of the National Academy of Sciences 2005;102(31):11070–5.
[16] Turnbaugh PJ, Bäckhed F, Fulton L, Gordon JI. Diet-induced obesity is linked to marked but reversible alterations in the
mouse distal gut microbiome. Cell Host & Microbe 2008;3(4):213–23.
[17] Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature 2006;
444(7122):1022–3.
[18] Armougom F, Henry M, Vialettes B, Raccah D, Raoult D. Monitoring bacterial community of human gut microbiota
reveals an increase in Lactobacillus in obese patients and Methanogens in anorexic patients. PLoS One 2009;4(9):e7125.
[19] Santacruz A, Collado MC, García-Valdés L, Segura MT, Martín-Lagos JA, Anjos T, et al. Gut microbiota composition is
associated with body weight, weight gain and biochemical parameters in pregnant women. British Journal of
Nutrition 2010;104(01):83–92.
[20] Zhang H, DiBaise JK, Zuccolo A, Kudrna D, Braidotti M, Yu Y, et al. Human gut microbiota in obesity and after gastric
bypass. Proceedings of the National Academy of Sciences 2009;106(7):2365–70.
[21] Duncan SH, Lobley GE, Holtrop G, Ince J, Johnstone AM, Louis P, et al. Human colonic microbiota associated with diet,
obesity and weight loss. International Journal of Obesity 2008;32(11):1720–4.
[22] Schwiertz A, Taras D, Schafer K, Beijer S, Bos NA, Donus C, et al. Microbiota and SCFA in lean and overweight healthy
subjects. Obesity 2009;18(1):190–5.
[23] Turnbaugh PJ, Gordon JI. The core gut microbiome, energy balance and obesity. The Journal of Physiology 2009;
587(17):4153–8.
[24] Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, et al. A core gut microbiome in obese and lean
twins. Nature 2009;457(7228):480–4.
[25] Greenblum S, Turnbaugh PJ, Borenstein E. Metagenomic systems biology of the human gut microbiome reveals to-
pological shifts associated with obesity and inflammatory bowel disease. Proceedings of the National Academy of
Sciences 2012;109(2):594–9.
*[26] Koren O, Goodrich JK, Cullender TC, Spor A, Laitinen K, Kling Bäckhed H, et al. Host remodeling of the gut microbiome
and metabolic changes during pregnancy. Cell 2012;150(3):470–80.
[27] FAO/WHO. Health and nutritional properties of probiotics in food including powder milk with live lactic acid bacteria.
WHO; 2001.
[28] Lan A, Lagadic-Gossmann D, Lemaire C, Brenner C, Jan G. Acidic extracellular pH shifts colorectal cancer cell death
from apoptosis to necrosis upon exposure to propionate and acetate, major end-products of the human probiotic
propionibacteria. Apoptosis 2007;12(3):573–91.
[29] Girard-Pipau F, Pompei A, Schneider S, Nano J, Hebuterne X, Boquet P, et al. Intestinal microflora, short chain and
cellular fatty acids, influence of a probiotic Saccharomyces boulardii. Microbial Ecology in Health and Disease 2002;
14(4):220–7.
[30] Sonnenburg JL, Chen CT, Gordon JI. Genomic and metabolic studies of the impact of probiotics on a model gut
symbiont and host. PLoS Biology 2006;4(12):e413.
70 P. Kovatcheva-Datchary, T. Arora / Best Practice & Research Clinical Gastroenterology 27 (2013) 59–72

[31] Marco ML, Peters TH, Bongers RS, Molenaar D, van Hemert S, Sonnenburg JL, et al. Lifestyle of Lactobacillus plantarum
in the mouse caecum. Environmental Microbiology 2009;11(10):2747–57.
[32] Denou E, Pridmore RD, Berger B, Panoff JM, Arigoni F, Brussow H. Identification of genes associated with the long-gut-
persistence phenotype of the probiotic Lactobacillus johnsonii strain NCC533 using a combination of genomics and
transcriptome analysis. Journal of Bacteriology 2008;190(9):3161–8.
*[33] McNulty NP, Yatsunenko T, Hsiao A, Faith JJ, Muegge BD, Goodman AL, et al. The impact of a consortium of fermented
milk strains on the gut microbiome of gnotobiotic mice and monozygotic twins. Science Translational Medicine 2011;
3(106). 106ra.
[34] Martin FP, Wang Y, Sprenger N, Holmes E, Lindon JC, Kochhar S, et al. Effects of probiotic Lactobacillus paracasei
treatment on the host gut tissue metabolic profiles probed via magic-angle-spinning NMR spectroscopy. Journal of
Proteome Research 2007;6(4):1471–81.
[35] Martin FP, Wang Y, Sprenger N, Yap IK, Lundstedt T, Lek P, et al. Probiotic modulation of symbiotic gut microbial-host
metabolic interactions in a humanized microbiome mouse model. Molecular Systems Biology 2008;4:157.
[36] West DB, Delany JP, Camet PM, Blohm F, Truett AA, Scimeca J. Effects of conjugated linoleic acid on body fat and energy
metabolism in the mouse. American Journal of Physiology – Regulatory, Integrative and Comparative Physiology 1998;
275(3):R667–72.
[37] Lee HY, Park JH, Seok SH, Baek MW, Kim DJ, Lee KE, et al. Human originated bacteria, Lactobacillus rhamnosus PL60,
produce conjugated linoleic acid and show anti-obesity effects in diet-induced obese mice. Biochimica Biophysica Acta
2006;1761:736–44.
[38] Lee K, Paek K, Lee HY, Park JH, Lee Y. Antiobesity effect of trans-10, cis-12-conjugated linoleic acid-producing
Lactobacillus plantarum PL62 on diet-induced obese mice. Journal of Applied Microbiology 2007;103(4):1140–6.
[39] Sato M, Uzu K, Yoshida T, Hamad EM, Kawakami H, Matsuyama H, et al. Effects of milk fermented by Lactobacillus
gasseri SBT2055 on adipocyte size in rats. British Journal of Nutrition 2008;99(5):1013–7.
[40] Takemura N, Okubo T, Sonoyama K. Lactobacillus plantarum strain No. 14 reduces adipocyte size in mice fed high-fat
diet. Experimental Biology and Medicine 2010;235(7):849–56.
[41] Hamad EM, Sato M, Uzu K, Yoshida T, Higashi S, Kawakami H, et al. Milk fermented by Lactobacillus gasseri SBT2055
influences adipocyte size via inhibition of dietary fat absorption in Zucker rats. British Journal of Nutrition 2009;
101(5):716–24.
[42] Kadooka Y, Ogawa A, Ikuyama K, Sato M. The probiotic Lactobacillus gasseri SBT2055 inhibits enlargement of visceral
adipocytes and upregulation of serum soluble adhesion molecule (sICAM-1) in rats. International Dairy Journal 2011;
21(9):623–7.
[43] Park DY, Ahn YT, Huh CS, Jeon SM, Choi MS. The inhibitory effect of Lactobacillus plantarum KY1032 cell extract on the
adipogenesis of 3T3-L1 Cells. Journal of Medicinal Food 2011;14(6):670–5.
[44] Ho JN, Choi JW, Lim WC, Kim MK, Lee IY, Cho HY. Kefir inhibits 3T3-L1 adipocyte differentiation through down-
regulation of adipogenic transcription factor expression. Journal of The Science of Food and Agriculture 2013;93(3):
485–90.
[45] Tanida M, Shen J, Maeda K, Horii Y, Yamano T, Fukushima Y, et al. High-fat diet-induced obesity is attenuated by
probiotic strain Lactobacillus paracasei ST11 (NCC2461) in rats. Obesity Research & Clinical Practice 2008;2(3):159–69.
[46] Aronsson L, Huang Y, Parini P, Korach-Andre M, Hakansson J, Gustafsson JA, et al. Decreased fat storage by Lactobacillus
paracasei is associated with increased levels of angiopoietin-like 4 protein (ANGPTL4). PloS One 2010;5(9):e13087.
[47] Nerstedt A, Nilsson EC, Ohlson K, Hakansson J, Thomas Svensson L, Lowenadler B, et al. Administration of Lactobacillus
evokes coordinated changes in the intestinal expression profile of genes regulating energy homeostasis and immune
phenotype in mice. British Journal of Nutrition 2007;97(6):1117–27.
[48] Fåk F, Bäckhed F. Lactobacillus reuteri prevents diet-induced obesity, but not atherosclerosis, in a strain dependent
fashion in Apoe/ mice. PLoS One 2012;7(10):e46837.
[49] Ji YS, Kim HN, Park HJ, Lee JE, Yeo SY, Yang JS, et al. Modulation of the murine microbiome with a concomitant anti-
obesity effect by Lactobacillus rhamnosus GG and Lactobacillus sakei NR28. Beneficial Microbes 2012;3(1):13–22.
[50] Arora T, Anastasovska J, Gibson G, Tuohy K, Sharma RK, Bell J, et al. Effect of Lactobacillus acidophilus NCDC 13
supplementation on the progression of obesity in diet-induced obese mice. British Journal of Nutrition 2012;108(8):
1382–9.
[51] Angelakis E, Bastelica D, Ben Amara A, El Filali A, Dutour A, Mege JL, et al. An evaluation of the effects of Lacto-
bacillus ingluviei on body weight, the intestinal microbiome and metabolism in mice. Microbial Pathogenesis 2012;
52(1):61–8.
[52] Kadooka Y, Sato M, Imaizumi K, Ogawa A, Ikuyama K, Akai Y, et al. Regulation of abdominal adiposity by probiotics
(Lactobacillus gasseri SBT2055) in adults with obese tendencies in a randomized controlled trial. European Journal of
Clinical Nutrition 2010;64(6):636–43.
[53] Vendt N, Grünberg H, Tuure T, Malminiemi O, Wuolijoki E, Tillmann V, et al. Growth during the first 6 months of life in
infants using formula enriched with Lactobacillus rhamnosus GG: double-blind, randomized trial. Journal of Human
Nutrition and Dietetics 2006;19(1):51–8.
[54] Luoto R, Kalliomaki M, Laitinen K, Isolauri E. The impact of perinatal probiotic intervention on the development of
overweight and obesity: follow-up study from birth to 10 years. International Journal of Obesity 2010;34(10):1531–7.
[55] Xing HC, Li LJ, Xu KJ, Shen T, Chen YB, Sheng JF, et al. Intestinal microflora in rats with ischemia/reperfusion liver
injury. Journal of Zhejiang University Science B 2005;6(1):14–21.
*[56] Le Roy T, Llopis M, Lepage P, Bruneau A, Rabot S, Bevilacqua C, et al. Intestinal microbiota determines development of
non-alcoholic fatty liver disease in mice. Gut 2012. http://dx.doi.org/10.1136/gutjnl-2012–303816.
[57] Iacono A, Raso GM, Canani RB, Calignano A, Meli R. Probiotics as an emerging therapeutic strategy to treat NAFLD:
focus on molecular and biochemical mechanisms. The Journal of Nutritional Biochemistry 2011;22(8):699–711.
[58] Nardone G, Compare D, Liguori E, Di Mauro V, Rocco A, Barone M, et al. Protective effects of Lactobacillus paracasei F19
in a rat model of oxidative and metabolic hepatic injury. American Journal of Physiology Gastrointestinal and Liver
Physiology 2010;299(3):G669–76.
P. Kovatcheva-Datchary, T. Arora / Best Practice & Research Clinical Gastroenterology 27 (2013) 59–72 71

[59] Adawi D, Ahrne S, Molin G. Effects of different probiotic strains of Lactobacillus and Bifidobacterium on bacterial
translocation and liver injury in an acute liver injury model. International Journal of Food Microbiology 2001;70(3):
213–20.
[60] Xing HC, Li LJ, Xu KJ, Shen T, Chen YB, Sheng JF, et al. Protective role of supplement with foreign Bifidobacterium and
Lactobacillus in experimental hepatic ischemia-reperfusion injury. Journal of Gastroenterology and Hepatology 2006;
21(4):647–56.
[61] Forsyth CB, Farhadi A, Jakate SM, Tang Y, Shaikh M, Keshavarzian A. Lactobacillus GG treatment ameliorates alcohol-
induced intestinal oxidative stress, gut leakiness, and liver injury in a rat model of alcoholic steatohepatitis. Alcohol
2009;43(2):163–72.
[62] Wang Y, Liu Y, Sidhu A, Ma Z, McClain C, Feng W. Lactobacillus rhamnosus GG culture supernatant ameliorates acute
alcohol-induced intestinal permeability and liver injury. American Journal of Physiology Gastrointestinal and Liver
Physiology 2012;303(1):G32–41.
[63] Wang Y, Kirpich I, Liu Y, Ma Z, Barve S, McClain CJ, et al. Lactobacillus rhamnosus GG treatment potentiates intestinal
hypoxia-inducible factor, promotes intestinal integrity and ameliorates alcohol-induced liver injury. The American
Journal of Pathology 2011;179(6):2866–75.
[64] Ma X, Hua J, Li Z. Probiotics improve high fat diet-induced hepatic steatosis and insulin resistance by increasing
hepatic NKT cells. Journal of Hepatology 2008;49(5):821–30.
[65] Li Z, Yang S, Lin H, Huang J, Watkins PA, Moser AB, et al. Probiotics and antibodies to TNF inhibit inflammatory activity
and improve nonalcoholic fatty liver disease. Hepatology 2003;37(2):343–50.
[66] Esposito E, Iacono A, Bianco G, Autore G, Cuzzocrea S, Vajro P, et al. Probiotics reduce the inflammatory response
induced by a high-fat diet in the liver of young rats. The Journal of Nutrition 2009;139(5):905–11.
[67] Roberfroid M, Gibson GR, Hoyles L, McCartney AL, Rastall R, Rowland I, et al. Prebiotic effects: metabolic and health
benefits. British Journal of Nutrition 2010;104(Suppl. S2):S1–63.
[68] Cani PD, Knauf C, Iglesias MA, Drucker DJ, Delzenne NM, Burcelin RG. Improvement of glucose tolerance and hepatic
insulin sensitivity by oligofructose requires a functional glucagon-like peptide 1 receptor. Diabetes 2006;55(5):1484–90.
[69] Delmée E, Cani PD, Gual G, Knauf C, Burcelin R, Maton N, et al. Relation between colonic proglucagon expression and
metabolic response to oligofructose in high fat diet-fed mice. Life Sciences 2006;79(10):1007–13.
[70] Cani PD, Daubioul CA, Reusens B, Remacle C, Catillon G, Delzenne NM. Involvement of endogenous glucagon-like
peptide-1(7–36) amide on glycaemia-lowering effect of oligofructose in streptozotocin-treated rats. Journal of
Endocrinology 2005;185(3):457–65.
[71] Cani PD, Neyrinck AM, Fava F, Knauf C, Burcelin RG, Tuohy KM, et al. Selective increases of bifidobacteria in gut
microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia.
Diabetologia 2007;50(11):2374–83.
[72] Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O, 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(8):1091–103.
[73] Everard A, Lazarevic V, Derrien M, Girard M, Muccioli GG, Neyrinck AM, et al. Responses of gut microbiota and glucose and
lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes 2011;60(11):2775–86.
[74] Parnell JA, Reimer RA. Prebiotic fibres dose-dependently increase satiety hormones and alter Bacteroidetes and Fir-
micutes in lean and obese JCR:LA-cp rats. British Journal of Nutrition 2012;107(04):601–13.
[75] Neyrinck AM, Possemiers S, Verstraete W, De Backer F, Cani PD, Delzenne NM. Dietary modulation of Clostridial cluster
XIVa gut bacteria (Roseburia spp.) by chitin-glucan fiber improves host metabolic alterations induced by high-fat diet
in mice. The Journal of Nutritional Biochemistry 2012;23(1):51–9.
[76] Neyrinck AM, Possemiers S, Druart C, Van de Wiele T, De Backer F, Cani PD, et al. Prebiotic effects of wheat arabi-
noxylan related to the increase in bifidobacteria, Roseburia and Bacteroides/Prevotella in diet-induced obese mice. PloS
One 2011;6(6):e20944.
[77] Hooda S, Boler BMV, Serao MCR, Brulc JM, Staeger MA, Boileau TW, et al. 454 pyrosequencing reveals a shift in fecal
microbiota of healthy adult men consuming polydextrose or soluble corn fiber. The Journal of Nutrition 2012;142(7):
1259–65.
[78] Dewulf EM, Cani PD, Claus SP, Fuentes S, Puylaert PGB, Neyrinck AM, et al. Insight into the prebiotic concept: lessons
from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut 2012. http://dx.
doi.org/10.1136/gutjnl-2012–303304.
[79] Davis LMG, Martínez I, Walter J, Goin C, Hutkins RW. Barcoded pyrosequencing reveals that consumption of gal-
actooligosaccharides results in a highly specific bifidogenic response in humans. PLoS One 2011;6(9):e25200.
[80] Cani PD, Neyrinck AM, Maton N, Delzenne NM. Oligofructose promotes satiety in rats fed a high-fat diet: involvement
of glucagon-like peptide-1. Obesity 2005;13(6):1000–7.
*[81] Cani PD, Lecourt E, Dewulf EM, Sohet FM, Pachikian BD, Naslain D, et al. Gut microbiota fermentation of prebiotics
increases satietogenic and incretin gut peptide production with consequences for appetite sensation and glucose
response after a meal. The American Journal of Clinical Nutrition 2009;90(5):1236–43.
[82] Parnell JA, Reimer RA. Weight loss during oligofructose supplementation is associated with decreased ghrelin and
increased peptide YY in overweight and obese adults. The American Journal of Clinical Nutrition 2009;89(6):1751–9.
[83] Verhoef SPM, Meyer D, Westerterp KR. Effects of oligofructose on appetite profile, glucagon-like peptide 1 and peptide
YY3-36 concentrations and energy intake. British Journal of Nutrition 2011;106(11):1757–62.
*[84] Tolhurst G, Heffron H, Lam YS, Parker HE, Habib AM, Diakogiannaki E, et al. Short-chain fatty acids stimulate glucagon-
like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 2012;61(2):364–71.
[85] Cani PD, Hoste S, Guiot Y, Delzenne NM. Dietary non-digestible carbohydrates promote L-cell differentiation in the
proximal colon of rats. British Journal of Nutrition 2007;98(01):32–7.
[86] Dewulf EM, Cani PD, Neyrinck AM, Possemiers S, Holle AV, Muccioli GG, et al. Inulin-type fructans with prebiotic
properties counteract GPR43 overexpression and PPARg-related adipogenesis in the white adipose tissue of high-fat
diet-fed mice. The Journal of Nutritional Biochemistry 2011;22(8):712–22.
72 P. Kovatcheva-Datchary, T. Arora / Best Practice & Research Clinical Gastroenterology 27 (2013) 59–72

[87] Lambert DM, Muccioli GG. Endocannabinoids and related N-acylethanolamines in the control of appetite and energy
metabolism: emergence of new molecular players. Current Opinion in Clinical Nutrition and Metabolic Care 2007;
10(6):735–44.
[88] Muccioli GG, Naslain D, Backhed F, Reigstad CS, Lambert DM, Delzenne NM, et al. The endocannabinoid system links
gut microbiota to adipogenesis. Molecular System Biology 2010;6:392.
[89] Martin F-PJ, Wang Y, Sprenger N, Yap IKS, Rezzi S, Ramadan Z, et al. Top-down systems biology integration of con-
ditional prebiotic modulated transgenomic interactions in a humanized microbiome mouse model. Molecular System
Biology 2008;4:205.
[90] Martin FP, Sprenger N, Montoliu I, Rezzi S, Kochhar S, Nicholson JK. Dietary modulation of gut functional ecology
studied by fecal metabonomics. Journal of Proteome Research 2010;9(10):5284–95.
[91] Arora T, Loo RL, Anastasovska J, Gibson GR, Tuohy KM, Sharma RK, et al. Differential effects of two fermentable car-
bohydrates on central appetite regulation and body composition. PLoS One 2012;7(8):e43263.
[92] So PW, Yu WS, Kuo YT, Wasserfall C, Goldstone AP, Bell JD, et al. Impact of resistant starch on body fat patterning and
central appetite regulation. PLoS One 2007;2(12):e1309.
[93] Anastasovska J, Arora T, Sanchez Canon GJ, Parkinson JRC, Touhy K, Gibson GR, et al. Fermentable carbohydrate alters
hypothalamic neuronal activity and protects against the obesogenic environment. Obesity 2012;20(5):1016–23.
[94] Parnell JA, Raman M, Rioux KP, Reimer RA. The potential role of prebiotic fibre for treatment and management of non-
alcoholic fatty liver disease and associated obesity and insulin resistance. Liver International: Official Journal of the
International Association for the Study of the Liver 2012;32(5):701–11.
[95] Agheli N, Kabir M, Berni-Canani S, Petitjean E, Boussairi A, Luo J, et al. Plasma lipids and fatty acid synthase activity are
regulated by short-chain fructo-öligosaccharides in sucrose-fed insulin-resistant rats. The Journal of Nutrition 1998;
128(8):1283–8.
[96] Delzenne NM, Kok NN. Biochemical basis of oligofructose-induced hypolipidemia in animal models. The Journal of
Nutrition 1999;129(7):1467S–70S.
[97] Kok N, Roberfroid M, Robert A, Delzenne N. Involvement of lipogenesis in the lower VLDL secretion induced by oli-
gofructose in rats. British Journal of Nutrition 1996;76(06):881–90.
[98] Sugatani J, Osabe M, Wada T, Yamakawa K, Yamazaki Y, Takahashi T, et al. Comparison of enzymatically synthesized
inulin, resistant maltodextrin and clofibrate effects on biomarkers of metabolic disease in rats fed a high-fat and high-
sucrose (cafeteria) diet. European Journal of Nutrition 2008;47(4):192–200.
[99] Daubioul CA, Taper HS, De Wispelaere LD, Delzenne NM. Dietary oligofructose lessens hepatic steatosis, but does not
prevent hypertriglyceridemia in obese zucker rats. The Journal of Nutrition 2000;130(5):1314–9.
[100] Parnell JA, Reimer RA. Effect of prebiotic fibre supplementation on hepatic gene expression and serum lipids: a dose-
response study in JCR:LA-cp rats. British Journal of Nutrition 2010;103(11):1577–84.
[101] Fiordaliso M, Kok N, Desager JP, Goethals F, Deboyser D, Roberfroid M, et al. Dietary oligofructose lowers triglycerides,
phospholipids and cholesterol in serum and very low density lipoproteins of rats. Lipids 1995;30(2):163–7.
[102] Busserolles J, Gueux E, Rock E, Demigné C, Mazur A, Rayssiguier Y. Oligofructose protects against the hyper-
triglyceridemic and pro-oxidative effects of a high fructose diet in rats. The Journal of Nutrition 2003;133(6):1903–8.
[103] Daubioul CA, Rousseau N, Demeure R, Gallez B, Taper H, Declerck B, et al. Dietary fructans, but not cellulose, decrease
triglyceride accumulation in the liver of obese zucker fa/fa rats. The Journal of Nutrition 2002;132(5):967–73.
[104] Kang J-H, Yun S-I, Park H- O. Effects of Lactobacillus gasseri BNR17 on body weight and adipose tissue mass in diet-
induced overweight rats. Journal of Microbiology 2010;48(5):712–4.
[105] Yun SI, Park HO, Kang JH. Effect of Lactobacillus gasseri BNR17 on blood glucose levels and body weight in a mouse
model of type 2 diabetes. Journal of Applied Microbiology 2009;107(5):1681–6.
[106] An H, Park S, Lee D, Kim J, Cha M, Lee S, et al. Antiobesity and lipid-lowering effects of Bifidobacterium spp. in high fat
diet-induced obese rats. Lipids in Health and Disease 2011;10(1):116.
[107] Chen JJ, Wang R, Li X-f, Wang R-l. Bifidobacterium longum supplementation improved high-fat-fed-induced metabolic
syndrome and promoted intestinal Reg I gene expression. Experimental Biology and Medicine 2011;236(7):823–31.
[108] Karlsson CLJ, Molin G, Fåk F, Johansson Hagslätt M-L, Jakesevic M, Håkansson Å, et al. Effects on weight gain and gut
microbiota in rats given bacterial supplements and a high-energy-dense diet from fetal life through to 6 months of
age. British Journal of Nutrition 2011;106(06):887–95.
[109] Neyrinck AM, Van Hee VF, Piront N, De Backer F, Toussaint O, Cani PD, et al. Wheat-derived arabinoxylan oligosac-
charides with prebiotic effect increase satietogenic gut peptides and reduce metabolic endotoxemia in diet-induced
obese mice. Nutrition and Diabetes 2012;2:e28.
[110] Cani PD, Joly E, Horsmans Y, Delzenne NM. Oligofructose promotes satiety in healthy human: a pilot study. European
Journal of Clinical Nutrition 2005;60(5):567–72.
[111] Cloetens L, Broekaert WF, Delaedt Y, Ollevier F, Courtin CM, Delcour JA, et al. Tolerance of arabinoxylan-
oligosaccharides and their prebiotic activity in healthy subjects: a randomised, placebo-controlled cross-over study.
British Journal of Nutrition 2010;103(05):703–13.
[112] Bays HE, Evans JL, Maki KC, Evans M, Maquet V, Cooper R, et al. Chitin-glucan fiber effects on oxidized low-density
lipoprotein: a randomized controlled trial. European Journal of Clinical Nutrition 2013;67:2–7.
[113] Maki KC, Gibson GR, Dickmann RS, Kendall CWC, Chen CYO, Costabile A, et al. Digestive and physiologic effects of a
wheat bran extract, arabino-xylan-oligosaccharide, in breakfast cereal. Nutrition 2012;28(11–12):1115–21.
[114] Walton GE, van den Heuvel EGHM, Kosters MHW, Rastall RA, Tuohy KM, Gibson GR. A randomised crossover study
investigating the effects of galacto-oligosaccharides on the faecal microbiota in men and women over 50 years of age.
British Journal of Nutrition 2012;107(10):1466–75.

You might also like