Nutrients: Probiotics: How Effective Are They in The Fight Against Obesity?
Nutrients: Probiotics: How Effective Are They in The Fight Against Obesity?
Nutrients: Probiotics: How Effective Are They in The Fight Against Obesity?
Review
Probiotics: How Effective Are They in the Fight
against Obesity?
Kiran Mazloom 1,† , Imran Siddiqi 1,† and Mihai Covasa 1,2, *
1 Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health
Sciences, Pomona, CA 91766, USA; [email protected] (K.M.); [email protected] (I.S.)
2 Department of Health and Human Development, University of Suceava, Suceava 720229, Romania
* Correspondence: [email protected]
† Contributed equally to the work.
Received: 14 December 2018; Accepted: 18 January 2019; Published: 24 January 2019
Abstract: Obesity has been associated with structural and functional changes in the gut microbiota.
The abundance in, and diversity of, certain bacteria may favor energy harvest and metabolic pathways
leading to obesity. Therefore, gut microbiota has become a potential target that can be manipulated to
obtain optimal health. Probiotics have been shown to influence the composition of the gut microbiota,
improve gut integrity, and restore the microbial shifts characteristic of obesity. Based on physical
and biochemical parameters, metabolic and inflammatory markers, and alterations in gut microbe
diversity, animal studies revealed beneficial results in obese models whereas the results in humans are
sparse and inconsistent. Thus, the purpose of this review is to present evidence from animal studies
and human clinical trials demonstrating the effects of various probiotic strains and their potential
efficacy in improving obesity and associated metabolic dysfunctions. Furthermore, the review
discusses current gaps in our understanding of how probiotics modulate gut microflora to protect
against obesity. Finally, we propose future studies and methodological approaches that may shed
light on the challenges facing the scientific community in deciphering the host–bacteria interaction
in obesity.
1. Introduction
Obesity is now considered a worldwide pandemic affecting approximately 1 in 3 individuals [1].
Despite significant efforts in the past decade to control the incidence of obesity, progress has been slow
in understanding the etiology and the various mechanisms mediating its development that may lead
to the identification of viable therapeutic approaches for treatment. Among the array of factors and
their complex interactions that play a role in obesity, new, accumulating evidence show that the gut
microbiota is an important contributor. The gut microbiota represents the sum of all bacteria that are
present in the gastrointestinal (GI) tract starting from the oral cavity and increasing in its density along
the small and large intestine [2]. Thus, for the purpose of this review, we will refer to the “microbiota”
as the physical bacteria present in the GI tract and “microbiome” as the set of genes that form the
bacterial strains.
A large body of evidence has described several possible mechanisms by which the gut microbiota
can contribute to, and/or influence, obesity. Although so much is still unknown and debatable,
thus far there is a general consensus that the gut microbiota is implicated in obesity through dietary
carbohydrate fermentation, lipogenesis, excess energy storage, and several other pathways including
a vast array of metabolites, hormones and neurotransmitters, some of which are known to control
food intake and the regulation of energy balance [3–5]. Furthermore, there is convincing literature
demonstrating that the composition and diversity of the gut microbiota is altered in obese rodents
and humans when compared to lean counterparts. For example, gut microbiota composition is
modified in obesity resulting in an enrichment or reduction in the proportions of specific bacterial
groups. Similarly, gut microbiota gene richness is also affected in obesity with studies showing a
20–40% decrease in the diversity of bacteria [6,7]. These findings suggest that restoration of the
compositional profile and richness of the gut microbiota may result in rescuing the obese phenotype
and associated metabolic defects. One way of accomplishing this is through the use of prebiotics,
probiotics, and synbiotics. Therefore, in this review, we will discuss the role of probiotics, their effects
and mechanisms implicated in the modulation of gut microbiota and its subsequent impact on
obesity development using comparative evidence from both preclinical and human clinical studies.
Furthermore, we will present and discuss current gaps in our understanding of how probiotics-induced
changes in gut microbiota profile may mitigate host metabolic dysregulations accompanying obesity.
Finally, we will propose future studies and methodological approaches that may shed light on the
challenges facing the scientific community in deciphering the host–bacteria interaction in obesity.
peptides [15]. The gut is also involved in nutrient sensing, with metabolic products from bacteria
activating enteroendocrine cells (EEC) through paracrine signaling from enterocytes [16]. In vitro and
in vivo studies have demonstrated that SCFAs may be used as main energy source, but they also serve
as signaling molecules that can activate G-protein coupled receptors (GPRs), including GPR43 (also
known as free fatty acid receptor 2) in adipose and intestinal tissues [17]. In adipose tissue, SCFAs
bind to GPR43, thus promoting adipogenesis and increasing energy expenditure [18]. In intestinal
tissue, SCFAs bind to GPR43 leading to secretion of anorexigenic peptides, including glucagon-like
peptide-1 (GLP-1) and peptide YY (PYY), resulting in improved glucose tolerance and increased energy
utilization. Additionally, increased production of selected SCFAs is associated with high levels of
ghrelin and insulin [16]. In particular, butyrate is involved in energy regulation by stimulating L
cells, a subpopulation of EEC, to secrete GLP-1. GLP-1, a peptide involved in satiety and insulin
secretion, has been found in lower quantities in obese compared to lean individuals [19]. Similarly,
PYY, also produced by the intestinal L cells, is important for satiety, increasing in concentration during
the postprandial period [20]. As such, administration of PYY-3-36 in obese individuals results in
a significant reduction of food intake [21]. Thus, GLP-1 and PYY act as appetite suppressants and
are potent mediators of the gut–brain axis, which facilitate important cross-talk regarding energy
homeostasis, digestion, and appetite [22]. They act to decrease intestinal motility, gastric emptying
and regulate glucose homeostasis and energy utilization [23].
The orexigenic gastric peptide hormone, ghrelin, is negatively correlated with Bifidobacterium,
Lactobacillus, and Blautia coccoides/Eubacterium rectale, and is positively correlated with Bacteroides and
Prevotella. Ghrelin has several functions including stimulation of gastric emptying, appetite stimulation,
glucagon secretion, and inhibition of insulin secretion and thermogenesis [16]. It has been shown
that cells which produce ghrelin have GPR43 receptors, but it is not clear yet if gut metabolites
directly stimulate these receptors, resulting in ghrelin secretion. By contrast, the anorexigenic hormone
leptin is positively correlated with Bifidobacterium and Lactobacillus. Although it is not clear whether
these are causal relationships, it is thought that leptin can modulate gut microbiota by stimulating
mucin production, which may favor differential bacterial growth [24]. Lastly, it should be noted
that microbiota composition can affect EEC counts and their respective receptor expressions, as the
Firmicutes to Bacteroidetes ratio was positively correlated with GPR43 expression in obese mice [25].
In summary, SCFAs binding to GPR43 in both adipose and intestinal tissues regulate obesity and
energy accumulation. This mechanism helps maintain energy homeostasis and may be used as a tool
in the treatment of metabolic diseases [17].
The gut microbiota not only enhances lipogenesis, but also reduces levels of fast-induced adipose
factor (FIAF), also called Angiopoietin-like 4 protein (ANGPTL4) a lipoprotein lipase (LPL) inhibitor
produced by the liver, intestine, and adipose tissue and a main regulator of metabolism and adiposity
(see [26,27]). Increased intake of a high carbohydrate and fat diet can lead to dysbiosis and increased
triglyceride deposition in adipose tissue which is associated with decreased FIAF expression [12].
This, in turn, leads to enhanced adipocyte LPL activity resulting in increased uptake of fatty acids,
increased fat storage and ultimately obesity [11]. As a result, FIAF serves as a protective mechanism
against diet-induced obesity. However, whether gut microbiota influences FIAF levels in obesity is
still unclear since high fat diet-induced obesity in germ free mice only increases mRNA expression of
FIAF in the intestine but not in the circulation [28]. In addition, gut microbiota inhibit the activity of
AMP-activated protein kinase (AMPK), an important liver and skeletal muscle enzyme with a role
in cellular energy homeostasis and obesity. When energy expenditure is low, AMPK is decreased
resulting in less activation of enzymes involved in beta oxidation, including acetyl CoA carboxylase
and carnitine palmitoyltransferase I, thus leading to obesity [29]. This explains why germ-free mice fed
a Western-type diet have increased levels of phosphorylated AMPK promoting fatty acid oxidation [29].
Therefore, gut microbiota suppresses AMPK activity leading to heightened cholesterol and triglycerides
synthesis, lipogenesis, excess fat accumulation and obesity [30].
Nutrients 2019, 11, 258 4 of 24
Obesity has also been linked to low-grade inflammation due to the failure of intestinal epithelial
membrane receptor proteins that play a sensory role in the gut [31–33]. This causes increased gut
permeability, reduced expression of tight junction proteins leading to bacterial fragments, such as
lipopolysaccharides (LPS) to diffuse through the gut and into the bloodstream, resulting in metabolic
endotoxemia. LPS then combines with pattern recognition receptor CD14, which, together, are recognized
by toll-like receptor-4 (TLR4), a major component of the innate immune system that maintains intestinal
homeostasis. Individuals who consume a high-fat diet have increased plasma LPS levels [34,35] that
stimulate cells through TLR4 leading to the development of low-grade inflammation observed in
obesity [36,37]. Increased LPS plasma concentration either via high fat feeding or experimentally induced
(i.e., through infusion) results in metabolic changes and systemic inflammation [38]. These changes were
associated with a significant reduction in the population of Lactobacillus spp., Bifidobacterium spp., and
Bacteroides–Prevotella spp in the gut. On the other hand, administration of Bifidobacteria to rodents with
thermal injury improved gut integrity and metabolic endotoxemia [39].
A further link between microbiota and obesity lies in the ability of Firmicutes and Actinobacteria
to produce conjugated linoleic acid (CLA). Altered production of this fatty acid is concerning in
the context of obesity because CLA has been shown to have several anti-obesity effects, including
increased energy metabolism, energy expenditure, and lipolysis as well as decreased adipogenesis and
lipogenesis [147]. Additionally, studies have demonstrated that CLA decreases de novo lipid synthesis
and induces adipocyte apoptosis [41]. Apoptosis of adipose tissue is associated with the induction of
TNF-alpha and uncoupling protein-2. As the name implies, uncoupling protein-2 “uncouples” electron
transfer across the inner mitochondrial membrane, which results in the thermal dissipation of energy
as opposed to conversion to the energy storage molecule adenosine triphosphate (ATP) [42]. It is
thought that CLA mediates some of its effects by displacing arachidonic acid from the phospholipids
contained in cell membranes, thereby decreasing synthesis of eicosanoids like prostaglandins and
leukotrienes, well known players in inflammation. CLA has signaling functions as well, including
activation of transcription factors and peroxisome proliferator-activated receptors (PPARs), which have
downstream effects on lipid metabolism and immune function [43]. Lastly, when administered to mice,
CLA enhanced sympathetic nervous system activity which led to increased energy metabolism and
reduced adipose tissue [44].
and an increased risk of obesity [45]. Other bacteria such as Lactobacillus are also present in high
quantities in obese and overweight children, whereas high levels of Bifidobacterium were found in lean
children [52]. Lastly, a high prevalence of Faecalibacterium prausnitzii, the most abundant Gram-positive
commensal bacteria present in the gut, has been linked with obesity [57]. Therefore, current evidence
shows microbiota compositional differences in obese compared to healthy, non-obese organisms.
The abundance or richness of bacterial genes has also been associated with obesity. For example,
low gene richness or counts (LGC) correlated with increased trunk-fat and obesity. In a large study
involving 61 severely obese women, it was found that 75% of the subjects had low gene counts [55]
compared to only 23–40% when subjects were overweight or moderately obese [6,7]. In addition, certain
metabolites and the proteins involved in their metabolism were associated with low microbial gene
richness (MGR). For example, as trunk fat mass increases, MGR decreases along with the metabolite,
3-methoxyphenylacetic acid. This acid is a product of polyphenol and flavonoid fermentation and may
have beneficial effects on the gut microbiota. As such, a reduction in histidine and enzymes involved
in histidine production and degradation has also been implicated in obesity. Gamma-aminobutyric
acid (GABA), a precursor to histidine production that is linked to downregulation of pro-inflammatory
cytokines, is one such pathway that is negatively affected in obesity leading to further inflammation [55].
the submucosa, triggering an inflammatory response, which is seen in a range of pathologies from
inflammatory bowel disease to obesity [63]. Although not completely understood, the administration of
probiotics has been shown to aid in the functionality of the intestinal barrier [64]. Lactobacilli modulate
expression of numerous genes encoding adherence junction proteins like E-cadherin, B-catenin in a
T84 cell barrier model [65]. Additionally, Escherichia coli Nissle 1917 (EcN1917) not only prevented
derangement of the mucosal barrier by a pathogenic E. coli, but also restored integrity in T84 and
Caco-2 cells. This effect was mediated by increased expression and repositioning of tight junction
proteins of the zonula occludens (ZO-2) and protein kinase C (PKC), which led to the reconstruction of
tight junction complex [66]. Some of the major macromolecular components of the epithelial mucus
include mucin glycoproteins, and may have implications in the development of metabolic syndrome.
Probiotics that can promote mucous secretion may improve gut barrier function and exclude pathogens.
One example is the probiotic VSL3 administered to rats for 7 days, which had a 60-fold increase in
MUC 2 expression as well as an increased secretion of mucin [67]. Also, VSL3 (a mixture of probiotics
and prebiotics) co-defends the epithelial barrier and increases tight junction protein expression through
activation of p38 and extracellular kinase pathways [68].
Lactobacillus
Over the past several years, promising preclinical studies investigating the effects of probiotic
supplementation on the development of obesity have emerged. A majority of the studies reviewed
here focused on intervention with Lactobacillus spp. and have demonstrated considerable anti-obesity
effects and the potential for probiotic based therapies in the treatment and prevention of obesity [82].
One of the early studies conducted over an eight-week period showed that Lactobacillus rhamnosus
PL60 reduced body weight without reducing energy intake, and significantly reduced white adipose
tissue. Signals of apoptosis and uncoupling protein-2 (UCP-2) mRNA levels were increased in adipose
tissue, while fatty acid synthase and serum leptin were simultaneously reduced [83]. The authors
attributed these effects to the production of t10, c12-conjugated linoleic acid by the probiotic [83].
In a follow-up study, t10, c12-CLA-producing L. plantarum PL62 reduced epididymal, inguinal,
mesenteric, and perineal white adipose tissue mass, while also significantly lowering body weight and
blood glucose levels in diet-induced obese mice [72]. In order to investigate the insulin-sensitizing
mechanisms associated with Lactobacillus rhamnosus GG (LGG), mice fed a high-fat diet (HFD) were
given oral LGG for 13 weeks, which resulted in attenuated weight gain, and improved insulin
sensitivity. Concurrently, LGG increased the expression of fatty acid oxidative genes in the liver while
it decreased gluconeogenic genes. This resulted in reductions in lipid accumulation by stimulating
adiponectin secretion and downstream activation of AMPK, an enzyme involved in controlling the
energy status of cells [84]. L. rhamnosus NCDC 17 improved oral glucose tolerance test, biochemical
parameters and oxidative stress in diabetic rats [85]. L. rhamnosus alone or in combination with any of
the herbal preparations (Aloe vera/Gymnema sylvestre powders, 1% w/w) seems to show anti-obesity
and anti-inflammatory properties in mice fed the high-fat diet [86]. Thus, modulation of gut microbiota
by probiotic strains containing Lactobacillus rhamnosus can have beneficial effects on body weight,
glucose and fat metabolism, insulin sensitivity and chronic systemic inflammation.
Several studies have tested the effects of the gram positive L. plantarum, a widespread member
of the genus Lactobacillus and with the largest genome from the lactic acid bacteria group, in obesity.
For example, Park et al. demonstrated that L. plantarum Q180 administered to mice on HFD reduced
body weight gain, and concurrently decreased triglycerides, serum leptin, aspartate aminotransferase
(AST), and epididymal fat weight [87]. Similarly, a 4-week study with HFD rats supplemented with a
high protein whey beverage containing kimchi-derived Lactobacillus plantarum DK211 prevented body
weight gain, and body fat accumulation. The treatment group also saw a decrease in organ weight, total
cholesterol, LDL cholesterol, triglycerides, blood glucose, and serum insulin, leptin, as well as ghrelin
compared to the HFD group [88]. Similarly, L. plantarum LG42 administered to male C57BL/6J mice on
a HFD for 12 weeks had significant anti-obesity effects such as lowered body weight, with a significant
Nutrients 2019, 11, 258 8 of 24
reduction in epididymal and back fat; decrease in hepatic triglyceride, serum insulin, and leptin
levels; increased mRNA expression of PPARα and CPT-I; decreased levels of acetyl-CoA carboxylase,
SREBP-1, and LXRα compared to control; reduced expression of PPARγ and its downstream genes [89].
Furthermore, when tested for its immuno-modulatory role, L. plantarum TN8 showed protective effects
on lipid, hepatic, and renal profiles in obese rats [90]. This Lactobacillus strain not only improved body
weight, but also had other beneficial anti-obesity effects by increasing IL-10 secretion while decreasing
other pro-inflammatory cytokine production [90]. Along the same lines, administration of another
L. plantarum strain, Ln4, a lactic acid bacteria isolated from fermented foods, reduced weight gain and
epididymal fat mass and lowered plasma triglycerides in mice fed a high-fat diet [91]. Protein levels of
adipokines such as C-reactive protein (CRP), insulin-like growth factor binding proteins-3 (IGFBP-3),
and monocyte chemoattractant protein-1 (MCP-1) were also decreased in white adipose tissue of mice
treated with Ln4. In addition, this probiotic strain induced changes in expression of several hepatic
genes involved in regulation of glucose and lipid metabolism including increased IRS2, Akt2, AMPK,
LPL, and reduced CD36, resulting in reduced insulin resistance, improved glucose tolerance and
insulin response. Together, these results point to Ln4 as a potential therapeutic probiotic agent for
the treatment of metabolic disorders. Finally, L. plantarum strain No. 14 (LP14) given to HFD female
mice reduced body weight gain, mean adipocyte size, white adipose tissue weight, total cholesterol,
and leptin after 11 weeks of administration. However, in a separate experiment, LP14 had no influence
on serum triacylglycerol accumulation following olive oil administration in Triton WR1339-treated
mice, which suggest that dietary fat absorption is unaffected by LP14 and its beneficial effects are
achieved through an alternate mechanism [92]. However, Lactobacillus plantarum (LP625) alone and
in combination with herbs (Aloe vera and Gymnema sylvestre) decreased body and epididymal fat
weight and reduced expression of pro-inflammatory cytokines.in mice fed the high-fat diet [93]. Thus,
based on the studies presented, L. plantarum has proved, at least in the animal studies, an effective
probiotic with anti-inflammatory and anti-obesity effects.
Several other Lactobacillus strains such as Lactobacillus gasseri and paracasei have been studied
for their anti-obesity and anti-inflammatory effects. For example, a 24-week long study found that
administration of Lactobacillus gasseri SBT2055 to mice fed a 10% fat diet resulted in reduced expression
of pro-inflammatory genes like CCL2 and CCR2 in adipose tissue, prevented body weight gain and
fat accumulation, providing evidence that an improved inflammatory state could be mechanistically
responsible for the aforementioned effects [94]. Similarly, rats fed skim milk with L. gasseri SBT2055
had a significant reduction in average adipocyte size, as well as decreased leptin and cholesterol [95].
Another Lactobacillus gasseri strain, BNR17 with promising anti-obesity effects reduced body weight,
white adipose tissue weight, serum leptin, and insulin levels in mice fed a high carbohydrate diet
when administered twice daily for 12 weeks [96]. Upregulation of fatty oxidation genes was also
seen in addition to downregulation of genes involved in fatty acid synthesis, providing insights
into the mechanistic basis for these positive results [97]. In an attempt to elucidate the role of the
autonomic nervous system in the observed anti-obesity effects, L. paracasei ST11 (NCC2461) was
administered intraduodenally. This led to an increase of sympathetic nerve activity in white and brown
adipose tissue and, following intragastric injection, thermogenesis and lipolysis increased in brown
and white adipose tissue, respectively. Overall, these changes indicate that the results of attenuated
weight gain are, in part, due to excitation of the sympathetic nervous system, and a consequent
lypolytic/thermogenic response [98]. In addition to examining the effects of single strains, several
studies have used multiple, mixed bacterial strains. For example, one study assessed the effects of the
prebiotic XOS, probiotic L. paracasei HII01, and synbiotic (1:1 combination of both) in HFD mice for
12 weeks. The results showed that the prebiotics, probiotics, and synbiotics, all significantly improved
insulin sensitivity, and attenuated dyslipidemia [99]. Body weight and visceral fat, however, showed
a significant reduction only in the pre-and synbiotic-treated groups. A significant decrease in LPS
levels was seen equally in the treatment groups, indicating a reduction in metabolic endotoxemia.
Pro-inflammatory cytokines, IL-6 and IL-1B, were also reduced. These benefits of L. paracasei HII01,
Nutrients 2019, 11, 258 9 of 24
XOS, and synbiotic combination could be due to the improvement of gut dysbiosis by lowering the
amount of LPS-containing Enterobacteriaceae within the gut lumen, which mitigates the eventual
LPS translocation into gut tissue and serum. Finally, combination of L. rhamnosus LGG with L. sakei
NR28 for 3 weeks, produced a significant reduction in epididymal fat mass, as well as obesity-related
biomarkers such as acetyl-CoA carboxylase, fatty acid synthase, stearoyl-CoA desaturase-1 in the liver
of HFD mice [100]. Anti-hyperglycemic and hyperlipidemic effects of probiotics are strain-dependent
as well as type of animal models [101]. Together, these preclinical studies show that treatment
with selective probiotics or their combinations can result in amelioration of metabolic dysfunctions
associated with obesity.
7.1. Bifidobacterium
A 2012 study found that Lactobacillus acidophilus NCDC13 administration to diet-induced obese
mice significantly increased Bifidobacterium, a genus that has been studied for its role in the amelioration
of obesity, shifting the gut microbiota balance positively after 8 weeks, although its specific anti-obesity
potential could not be established in mice fed a HFD [104]. Likewise, administration of Bifidobacterium
probiotic strains increased fecal Bifidobacterium pseudocatenulatum SPM 1204, B. longum SPM 1205,
and B. longum SPM 1207 in HFD rats [105]. This was associated with other anti-obesity effects
such as reduced serum total cholesterol, HDL-C, LDL-C, triglycerides, glucose, leptin, AST/ALT,
lipase, and reduction in harmful enzymatic activities done by β-glucosidase, β -glucuronidase, and
tryptophanase [105]. When comparing the effects of B. L66-5 vs. L75-4 versus M13-4 vs. FS31-12
in HFD mice, it was shown that while all four strains reduced serum and liver triglycerides, only B.
L66-5 reduced body weight [106]. In studies using another strain, B. adolescentis supplementation
reduced body and fat weight of mice fed a high-fat diet [107]. B. adolescentis was further shown
to reduce body weight gain, improve diet-induced nonalcoholic steatohepatitis, and decrease liver
damage associated with the inhibition of lipid peroxidation, NFκB activation, and inflammation [108].
When comparing several Bifidobacterium animalis subsp. lactis I-2494 (BA), L. paracasei CNCM I-4270
(LC), and L. rhamnosus I-3690 (LR) Wang et al. showed that all individually attenuated weight gain
and macrophage infiltration into epididymal adipose tissue, while also improving glucose-insulin
homeostasis and hepatic steatosis of HFD mice after 12 weeks. Overall, all three strains shifted the
HFD-disrupted gut back toward that of lean mice fed a chow diet, but found that while LC and LR
increased cecal acetate, they did not affect lipopolysaccharide-binding protein in circulation. In contrast,
BA did not increase acetate but decreased adipose and hepatic TNF-α expression, suggesting that
Lactobacillus and Bifidobacterium attenuate obesity through a strain-specific, differential manner, and
highlighted the importance of understanding specific mechanistic differences between probiotics [109].
1 (SCD1), and PPARγ) [110]. Similarly, Cano et al. studied strains from the IATA-CSIC and Spanish
Culture Collection (CECT) including Bacteroides uniformis CECT 7771 that was investigated for its utility
in HFD-induced obesity. Supplementation resulted in decreased body weight gain, liver steatosis,
triglycerides, and reduced dietary fat absorption. Additionally, this treatment ameliorated immune
dysfunction seen in obese mice, which was evidenced by restored capacity of dendritic cells to cause
T-cell proliferation response and was associated with partial restoration of microbiota composition [111].
Pouthidis et al. expanded upon the immune dysfunction associated with obesity, feeding a “fast food
diet” that resulted in CD4+ Th17 biased immunity, changes in microbial composition, and abdominal
obesity in mice. However, intervention with yogurt containing a mixture of S. thermophiles,
L. bulgaricus, L. acidophilus, B. bifidus, L. casei, and L rhamnosus inhibited age-associated weight
gain, while L. reuteri ATCC 6475 administered in drinking water prevented abdominal fat pathology
independent of baseline diet. These effects were conferred with purified CD4+ T cells, and depended
on active immune tolerance by the induction of Foxp3+ regulatory T cells and IL-10. [112]. These
studies suggest that supplementation with L. reuteri restores a beneficial balance in the Th17/Treg
host immunity, even in those individuals who have a pro-inflammatory immune state and chronic
inflammation in association with a Westernized “fast food” diet. In mice fed a high fat and sucrose
diet, epididymal fat mass and adipocyte size were significantly reduced with no difference in body
weight [101]. As noted above, obesity is a disease characterized by disrupted microbiota, inflammation,
and gut barrier alterations. The Gram-negative, strictly anaerobic bacteria, Akkermansia muciniphila,
widely studied for its anti-inflammatory effects, has been shown to be in close proximity to the
intestinal epithelium, supporting the hypothesis that A. muciniphila plays a role in mutualistic
interactions between the gut microbiota and host that controls gut barrier functions as well as other
physiological processes that occur in obesity [113]. A. muciniphila is a mucin-degrading bacterium
that resides in the mucus layer of the gut, and is the dominant human bacterium that colonizes this
nutrient-rich environment, representing anywhere from 3–5% of the microbial community in healthy
individuals [114]. The abundance of this bacterium is inversely correlated with body weight in both
humans and rodents. Everard et al. observed that viable A. muciniphila treatment for only four weeks
reversed HFD-induced metabolic abnormalities including fat mass gain, metabolic endotoxemia,
adipose tissue inflammation, and insulin resistance [115].
biologically-significant compounds that have synergistic effects [118]. Likewise, Alard and colleagues
studied the combination of L. rhamnosus LMG S-28148 and B. animalis subsp. Lactis LMG P-28149 (mix)
and found that HFD mice given the probiotic cocktail had significantly reduced body weight gain and
adiposity. The authors noted that the observed reduced insulin resistance, and dyslipidemia could be
explained by adipose tissue immune cell-remodeling, primarily affecting macrophages. At the gut
level, there were changes in the uptake of fatty acids, restored expression of the short chain fatty acid
receptor GPR43, and microbiotal changes which included recovered levels of Akkermansia muciniphila,
increased Rickenellacea, and decreased Lactobacillaceae. When B. animalis subsp lactis LMG P-28149 was
given alone, its efficiency was comparable to the mixture in regards to reduction in body weight,
adipose tissue mass, and serum leptin levels [119]. Furthermore, administration of B. longum alone
or mixed with L. casei Shirota reduced weight and triglycerides in HFD rats. Surprisingly, B. longum
alone was better at modulating leptin level, fat mass, adipocyte size, lipoprotein lipase and PPAR-γ
expression, and increasing adiponectin [120]. A recent study showed that a mixture of B. lactis Bi1,
B. breve Bbr8, and B. breve BL10 (B. mix) was the best at ameliorating obesity in HFD mice compared to
administration of single strains of LGG, L acidophilus LA1/K8, or alternative mixtures like L. bulgaricus
LB2 with S. termophilus Z57. B. mix reduced weight gain, adipose tissue fat accumulation, adipocyte
size, and macrophage/CD4+ T cell infiltration. Lastly, improvements in lipid profile and regulation of
leptin and cytokine secretion were seen [121]. Overall, these studies showed that selective mixtures
of probiotic strains could be more effective than single strains in ameliorating metabolic parameters
associated with obesity.
and strengthen the intestinal barrier. When Lactobacillus reuteri NCIMB 30242 was administered twice
a day for six weeks to healthy, hypercholesterolemic adult men and women, there was no significant
effect on BMI or body weight [127]. On the other hand, Lactobacillus reuteri JBD30I administered
to overweight and obese individuals for 12 weeks had a positive effect by reducing dietary fat
and free fatty acid absorption in the small intestine leading to increased excretion of the free fatty
acids [128]. Although this may prove helpful in the treatment of obesity, thus far there are no clear
and definitive results that support this theory. Several other Lactobacillus strains have been tested in
humans with various outcomes (Table 1). For example, experiments where Lactobacillus plantarum
A7 containing soy milk or regular soy milk was administered for eight weeks to patients with type
2 diabetes showed no significant differences in BMI or waist to hip ratio between the probiotic and
placebo groups [129]. On the other hand, cheese-containing Lactobacillus plantarum TENSIA given
to obese hypertensive patients for three weeks resulted in significant reduction of body weight,
BMI, and fat mass of the subjects compared to subjects who consumed the control cheese [130].
Thus Lactobacillus plantarum TENSIA is among the few probiotics that has been shown to reduce
body weight, BMI, and fat mass correlated to obesity in humans. Another Lactobacillus probiotic
present in the gastrointestinal tract and tested for the treatment of obesity is Lactobacillus salivarus Ls-33.
When administered to obese adolescents for twelve weeks L. salivarius was ineffective in exerting any
impact on SCFA concentrations, a strong marker correlated to obesity or on the metabolic syndrome
associated parameters. However, adolescents that consumed the probiotic L. salivarius had an increased
ratio of the Bacteroides-Prevotella-Poryphyromonas group compared to the Firmicutes belonging bacterial
group suggesting a modulatory role of L. salivarius on fecal microbiota composition [131–133]. However,
supplementation with VSL#3® in obese Latino adolescents increased adiposity with no significant
detectable changes in gut microbiota, gut appetite-regulating hormones, liver fat and fibrosis and
dietary intake [134]. Lastly, administration of the plant-derived lactic acid bacterium Pediococcus
pentosaceus (LP28) that was heat-killed significantly lowered BMI, body fat, and waist circumference,
whereas live LP28 and placebo exhibited no significant differences. Therefore, heat-killed LP28 may be
helpful in the treatment of obesity, although more work needs to evaluate its efficacy as a valuable
treatment option for obesity [135].
disease (NAFLD) [139] and to overweight and obese women [140] had no significant effect on body
weight and fat percentage compared to the control group. Other studies utilizing a mixture of
Lactobacillus and Bifidobacterium have been more successful. For example, in a recent experiment,
Gomes et al. administered L. acidophilus, L. casei, L. lactis, B. bifidum, and B. lactis to overweight and
obese women on a dietary intervention for 8 weeks. Overall, supplementation with the probiotic
mixture resulted in reduced abdominal adiposity, and increased antioxidant activity. Additionally,
the dietary intervention plus probiotic supplementation produced a significant reduction in waist
circumference, and plasma polyunsaturated fatty acids [141]. These results are consistent with
a 2018 study demonstrating beneficial effects of administering multispecies probiotics from the
genus Bifidobacterium and Lactobacillus (see table below) [142]. In this study involving 81 obese
postmenopausal women, the probiotic mixture at both low and high doses positively modified
glucose metabolism, lipid profile, waist circumference, visceral fat, serum uric acid levels, and LPS
concentration. Additionally, serum total cholesterol, triglycerides, LDL cholesterol, glucose, insulin,
and insulin resistance index were improved in the high dose group. Both groups had significant
differences in fat percentage and visceral fat. Taken together these studies show that although some
combination on Lactobacillus and Bifidobacteria probiotic strains may be more effective than others in
ameliorating obesity-related metabolic defects, by and large, the effectiveness of probiotics on human
obesity and their mechanisms of action are yet to be fully elucidated.
lipid that stimulates secretion of GLPs. Although the data from A. muciniphila is encouraging,
more work is required to elucidate the key mechanisms involved in its beneficial effects in human
obesity [149].
Another next-generation probiotic, Roseburia intestinalis, metabolizes dietary fiber, is a major SCFA
producer, provides energy for enterocytes, and has anti-inflammatory effects [150]. Cross-feeding
chains have been established between Bifidobacteria, F. prausnitzii, and R. intestinalis, which enhance
SCFA butyrate production in the colon. More work on the metabolic capabilities and cross-feeding
chains in the gut could open novel avenues into the modulation of the composition and action of
the microbiota. This knowledge coupled with more affordable profiling of individual gut microbial
differences could lead to tailored efficacious therapies in the future [150].
The long-term benefits of probiotics in relation to the gut microbiome and obesity are poorly
studied and their results are inconsistent. For example, in one study, pregnant women were fed a
probiotic mixture containing B. bifidum W23, B. lactis W52, and Lc. Lactis W58 during their last six
weeks of pregnancy. The composition of the microbiota in the children of these women was examined
for the first six years of life, and the study showed that there were no long-lasting differences [151].
However, another study examined the long-term safety and benefits of perinatal administration of
certain probiotics. The results showed that overall prenatal probiotic intervention was safe in the
long term. In addition, children who regularly consumed probiotics had a decreased risk of being
overweight in a long-term follow-up [152]. Thus, although long-term efficacy of probiotics are still
controversial and more research is needed to establish their effects, probiotic products have been
proven to be safe for human use.
Table 1. Cont.
minute interactions between the host and flora, including genetic material exchange, may hold the
key to meaningful clinical translation. Further understanding of this complex cross talk will aid in the
development of more tailored and targeted implementation of probiotic therapies. Additionally,
most of the aforementioned studies have been performed in tightly controlled animal models,
which limit their potential application to human subjects, who will likely need to be stratified based on
specific markers that take into account lifestyle, age, genetics, and other environmental influences on
microbiota composition. Elucidation of the metagenomic relationship between changing microbiota
and probiotic species under different diets/nutritional states will also be needed. Furthermore,
most of the research in this dynamic field has been conducted using Lactobacillus and Bifidobacterium
strains, thus creating the necessity for identifying new bacterial candidates along with their potential
mechanistic effects on obesity.
Thus far, clinical cohorts consisted of relatively small sample size, and focused on short-term
physical parameters, or inflammatory markers, making long-term follow up studies highly desired in
future work. Additional randomized placebo-controlled trials will help develop clinical guidelines for
the use of probiotic therapy in obesity, and formulate nutritional recommendations and address safety
concerns regarding functional foods that contain probiotics like fermented dairy products and kimchi.
Questions about specific bacterial strains’ effect on microbial composition, duration of treatment,
and appropriate dose are still left unanswered. Notwithstanding these shortcomings, there is no doubt
that probiotic therapy represents an exciting new frontier in the treatment of obesity and associated
metabolic dysfunctions.
Author Contributions: All authors contributed to the conceptualization, design, drafting and revision of the
manuscript. All authors approved the submitted version of the manuscript.
Funding: The work was supported by the project titled “The analysis of interrelationship between gut microbiota
and the host with applications in the prevention and control of type 2 diabetes” co-financed by the European Regional
Development Fund through Competitiveness Operational Program under the contract number 120/16.09.2016.
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Agha, M.; Agha, R. The rising prevalence of obesity: Part A: Impact on public health. Int. J. Surg. Oncol. (N. Y.)
2017, 2, e17. [CrossRef]
2. O’Hara, A.M.; Shanahan, F. The gut flora as a forgotten organ. EMBO Rep. 2006, 7, 688–693. [CrossRef]
3. Shahid, S.U.; Irfan, U. The gut microbiota and its potential role in obesity. Future Microbiol. 2018, 13, 589–603.
4. Wolever, T.; Brighenti, F.; Royall, D.; Jenkins, A.L.; Jenkins, D.J. Effect of rectal infusion of short chain fatty
acids in human subjects. Am. J. Gastroenterol. 1989, 84, 1027–1033.
5. Wolever, T.M.S.; Spadafora, P.; Eshuis, H. Interaction between colonic acetate and propionate in humans.
Am. J. Clin. Nutr. 1991, 53, 681–687. [CrossRef]
6. Cotillard, A.; Kennedy, S.P.; Kong, L.C.; Prifti, E.; Pons, N.; Le Chatelier, E.; Almeida, M.; Quinquis, B.;
Levenez, F.; Galleron, N.; et al. Dietary intervention impact on gut microbial gene richness. Nature 2013, 500,
585–588. [CrossRef]
7. Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.;
Batto, J.M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature
2013, 500, 541–546. [CrossRef]
8. Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.;
Relman, D.A. Diversity of the human intestinal microbial flora. Science 2005, 308, 1635–1638. [CrossRef]
9. Azad, M.A.K.; Sarker, M.; Li, T.; Yin, J. Probiotic species in the modulation of gut microbiota: An overview.
BioMed Res. Int. 2018, 2018, 9478630. [CrossRef]
10. Hentges, D.J. The anaerobic microflora of the human body. Clin. Infect. Dis. J. 1993, 16, S175–S180. [CrossRef]
11. Kobyliak, N.; Virchenko, O.; Falalyeyeva, T. Pathophysiological role of host microbiota in the development
of obesity. Nutr. J. 2016, 15, 43. [CrossRef]
Nutrients 2019, 11, 258 17 of 24
12. Backhed, F.; Ding, H.; Wang, T.; Hooper, L.V.; Koh, G.Y.; Nagy, A.; Semenkovich, C.F.; Gordon, J.I. The gut
microbiota as an environmental factor that regulates fat storage. Proc. Natl. Acad. Sci. USA 2004, 101,
15718–15723. [CrossRef]
13. Flint, H.J.; Bayer, E.A.; Rincon, M.T.; Lamed, R.; White, B.A. Polysaccharide utilization by gut bacteria:
Potential for new insights from genomic analysis. Nat. Rev. Microbiol. 2008, 6, 121–131. [CrossRef]
14. Samuel, B.S.; Gordon, J.I. A humanized gnotobiotic mouse model of host–archaeal–bacterial mutualism.
Proc. Natl. Acad. Sci. USA 2006, 103, 10011–10016. [CrossRef]
15. Schele, E.; Grahnemo, L.; Anesten, F.; Hallen, A.; Backhed, F.; Jansson, J.O. The gut microbiota reduces
leptin sensitivity and the expression of the obesity-suppressing neuropeptides proglucagon (Gcg) and
brain-derived neurotrophic factor (Bdnf) in the central nervous system. Endocrinology 2013, 154, 3643–3651.
[CrossRef]
16. Queipo-Ortuno, M.I.; Seoane, L.M.; Murri, M.; Pardo, M.; Gomez-Zumaquero, J.M.; Cardona, F.;
Casanueva, F.; Tinahones, F.J. Gut microbiota composition in male rat models under different nutritional
status and physical activity and its association with serum leptin and ghrelin levels. PLoS ONE 2013, 8,
e65465. [CrossRef]
17. Kimura, I.; Inoue, D.; Hirano, K.; Tsujimoto, G. The SCFA receptor GPR43 and energy metabolism.
Front. Endocrinol. (Lausanne) 2014, 5, 85. [CrossRef]
18. Hong, Y.-H.; Nishimura, Y.; Hishikawa, D.; Tsuzuki, H.; Miyahara, H.; Gotoh, C.; Choi, K.-C.; Feng, D.D.;
Chen, C.; Lee, H.-G.; et al. Acetate and propionate short chain fatty acids stimulate adipogenesis via GPCR43.
Endocrinology 2005, 146, 5092–5099. [CrossRef]
19. Samuel, B.S.; Shaito, A.; Motoike, T.; Rey, F.E.; Backhed, F.; Manchester, J.K.; Hammer, R.E.; Williams, S.C.;
Crowley, J.; Yanagisawa, M.; et al. Effects of the gut microbiota on host adiposity are modulated by the
short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc. Natl. Acad. Sci. USA 2008, 105,
16767–16772. [CrossRef]
20. Grandt, D.; Schimiczek, M.; Beglinger, C.; Layer, P.; Goebell, H.; Eysselein, V.E.; Reeve, J.R., Jr. Two
molecular forms of peptide YY (PYY) are abundant in human blood: Characterization of a radioimmunoassay
recognizing PYY 1-36 and PYY 3-36. Regul. Pept. 1994, 51, 151–159. [CrossRef]
21. Batterham, R.L.; Cohen, M.A.; Ellis, S.M.; Le Roux, C.W.; Withers, D.J.; Frost, G.S.; Ghatei, M.A.; Bloom, S.R.
Inhibition of food intake in obese subjects by peptide YY3-36. N. Engl. J. Med. 2003, 349, 941–948. [CrossRef]
[PubMed]
22. Bauer, P.V.; Hamr, S.C.; Duca, F.A. Regulation of energy balance by a gut-brain axis and involvement of the
gut microbiota. Cell. Mol. Life Sci. 2016, 73, 737–755. [CrossRef] [PubMed]
23. Holst, J.J. Incretin hormones and the satiation signal. Int. J. Obes. (Lond.) 2013, 37, 1161–1168. [CrossRef]
[PubMed]
24. El Homsi, M.; Ducroc, R.; Claustre, J.; Jourdan, G.; Gertler, A.; Estienne, M.; Bado, A.; Scoazec, J.Y.;
Plaisancie, P. Leptin modulates the expression of secreted and membrane-associated mucins in colonic
epithelial cells by targeting PKC, PI3K, and MAPK pathways. Am. J. Physiol. Gastrointest. Liver Physiol. 2007,
293, G365–G373. [CrossRef] [PubMed]
25. Lu, Y.; Fan, C.; Li, P.; Lu, Y.; Chang, X.; Qi, K. Short chain fatty acids prevent high-fat-diet-induced obesity in
mice by regulating G protein-coupled receptors and gut microbiota. Sci. Rep. 2016, 6, 37589. [CrossRef]
26. Dahiya, D.K.; Renuka; Puniya, M.; Shandilya, U.K.; Dhewa, T.; Kumar, N.; Kumar, S.; Puniya, A.K.; Shukla, P.
Gut microbiota modulation and its relationship with obesity using prebiotic fibers and probiotics: A review.
Front. Microbiol. 2017, 8, 563. [CrossRef]
27. Yoon, J.C.; Chickering, T.W.; Rosen, E.D.; Dussault, B.; Qin, Y.; Soukas, A.; Friedman, J.M.; Holmes, W.E.;
Spiegelman, B.M. Peroxisome proliferator-activated receptor gamma target gene encoding a novel
angiopoietin-related protein associated with adipose differentiation. Mol. Cell. Biol. 2000, 20, 5343–5349.
[CrossRef]
28. Fleissner, C.K.; Huebel, N.; Abd El-Bary, M.M.; Loh, G.; Klaus, S.; Blaut, M. Absence of intestinal microbiota
does not protect mice from diet-induced obesity. Br. J. Nutr. 2010, 104, 919–929. [CrossRef]
29. Bäckhed, F.; Manchester, J.K.; Semenkovich, C.F.; Gordon, J.I. Mechanisms underlying the resistance to
diet-induced obesity in germ-free mice. Proc. Natl. Acad. Sci. USA 2007, 104, 979–984. [CrossRef]
30. Boulange, C.L.; Neves, A.L.; Chilloux, J.; Nicholson, J.K.; Dumas, M.E. Impact of the gut microbiota on
inflammation, obesity, and metabolic disease. Genome Med. 2016, 8, 42. [CrossRef]
Nutrients 2019, 11, 258 18 of 24
31. Kuwahara, A. Contributions of colonic short-chain fatty acid receptors in energy homeostasis.
Front. Endocrinol. (Lausanne) 2014, 5, 144. [CrossRef] [PubMed]
32. Boroni Moreira, A.P.; Fiche Salles Teixeira, T.; Gouveia Peluzio, M.d.C.; Cássia Gonçalves Alfenas, R.d.
Gut microbiota and the development of obesity. Nutrición Hospitalaria 2012, 27, 1408–1414. [PubMed]
33. Tilg, H.; Kaser, A. Gut microbiome, obesity, and metabolic dysfunction. J. Clin. Investig. 2011, 121, 2126–2132.
[CrossRef] [PubMed]
34. Amar, J.; Burcelin, R.; Ruidavets, J.B.; Cani, P.D.; Fauvel, J.; Alessi, M.C.; Chamontin, B.; Ferrieres, J. Energy
intake is associated with endotoxemia in apparently healthy men. Am. J. Clin. Nutr. 2008, 87, 1219–1223.
[CrossRef] [PubMed]
35. Erridge, C.; Attina, T.; Spickett, C.M.; Webb, D.J. A high-fat meal induces low-grade endotoxemia: Evidence
of a novel mechanism of postprandial inflammation. Am. J. Clin. Nutr. 2007, 86, 1286–1292. [CrossRef]
[PubMed]
36. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in
gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and
diabetes in mice. Diabetes 2008, 57, 1470–1481. [CrossRef] [PubMed]
37. Rakoff-Nahoum, S.; Paglino, J.; Eslami-Varzaneh, F.; Edberg, S.; Medzhitov, R. Recognition of commensal
microflora by toll-like receptors is required for intestinal homeostasis. Cell 2004, 118, 229–241. [CrossRef]
38. Cani, P.D. Human gut microbiome: Hopes, threats and promises. Gut 2018, 67, 1716–1725. [CrossRef]
39. Wang, Z.; Xiao, G.; Yao, Y.; Guo, S.; Lu, K.; Sheng, Z. The role of bifidobacteria in gut barrier function after
thermal injury in rats. J. Trauma 2006, 61, 650–657. [CrossRef]
40. Kennedy, A.; Martinez, K.; Schmidt, S.; Mandrup, S.; LaPoint, K.; McIntosh, M. Antiobesity mechanisms of
action of conjugated linoleic acid. J. Nutr. Biochem. 2010, 21, 171–179. [CrossRef] [PubMed]
41. Obsen, T.; Faergeman, N.J.; Chung, S.; Martinez, K.; Gobern, S.; Loreau, O.; Wabitsch, M.; Mandrup, S.;
McIntosh, M. Trans-10, cis-12 conjugated linoleic acid decreases de novo lipid synthesis in human adipocytes.
J. Nutr. Biochem. 2012, 23, 580–590. [CrossRef] [PubMed]
42. Ryder, J.W.; Portocarrero, C.P.; Song, X.M.; Cui, L.; Yu, M.; Combatsiaris, T.; Galuska, D.; Bauman, D.E.;
Barbano, D.M.; Charron, M.J.; et al. Isomer-specific antidiabetic properties of conjugated linoleic acid.
Improved glucose tolerance, skeletal muscle insulin action, and UCP-2 gene expression. Diabetes 2001, 50,
1149–1157. [CrossRef] [PubMed]
43. Belury, M.A. Dietary conjugated linoleic acid in health: Physiological effects and mechanisms of action.
Annu. Rev. Nutr. 2002, 22, 505–531. [CrossRef] [PubMed]
44. Ohnuki, K.; Haramizu, S.; Oki, K.; Ishihara, K.; Fushiki, T. A single oral administration of conjugated linoleic
acid enhanced energy metabolism in mice. Lipids 2001, 36, 583–587. [CrossRef] [PubMed]
45. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut
microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [CrossRef] [PubMed]
46. Pryde, S.E.; Duncan, S.H.; Hold, G.L.; Stewart, C.S.; Flint, H.J. The microbiology of butyrate formation in the
human colon. FEMS Microbiol. Lett. 2002, 217, 133–139. [CrossRef] [PubMed]
47. Duncan, S.H.; Hold, G.L.; Barcenilla, A.; Stewart, C.S.; Flint, H.J. Roseburia intestinalis sp. nov., a novel
saccharolytic, butyrate-producing bacterium from human faeces. Int. J. Syst. Evol. Microbiol. 2002, 52,
1615–1620.
48. Kasai, C.; Sugimoto, K.; Moritani, I.; Tanaka, J.; Oya, Y.; Inoue, H.; Tameda, M.; Shiraki, K.; Ito, M.;
Takei, Y.; et al. Comparison of the gut microbiota composition between obese and non-obese individuals in a
Japanese population, as analyzed by terminal restriction fragment length polymorphism and next-generation
sequencing. BMC Gastroenterol. 2015, 15, 100. [CrossRef]
49. Koliada, A.; Syzenko, G.; Moseiko, V.; Budovska, L.; Puchkov, K.; Perederiy, V.; Gavalko, Y.; Dorofeyev, A.;
Romanenko, M.; Tkach, S.; et al. Association between body mass index and Firmicutes/Bacteroidetes ratio
in an adult Ukrainian population. BMC Microbiol. 2017, 17, 120. [CrossRef]
50. Turnbaugh, P.J.; Hamady, M.; Yatsunenko, T.; Cantarel, B.L.; Duncan, A.; Ley, R.E.; Sogin, M.L.; Jones, W.J.;
Roe, B.A.; Affourtit, J.P.; et al. A core gut microbiome in obese and lean twins. Nature 2009, 457, 480–484.
[CrossRef]
51. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Human gut microbes associated with obesity. Nature 2006,
444, 1022–1023. [CrossRef] [PubMed]
Nutrients 2019, 11, 258 19 of 24
52. Ignacio, A.; Fernandes, M.R.; Rodrigues, V.A.; Groppo, F.C.; Cardoso, A.L.; Avila-Campos, M.J.; Nakano, V.
Correlation between body mass index and faecal microbiota from children. Clin. Microbiol. Infect. 2016, 22,
258.e1–258.e8. [CrossRef] [PubMed]
53. Haro, C.; Rangel-Zuniga, O.A.; Alcala-Diaz, J.F.; Gomez-Delgado, F.; Perez-Martinez, P.; Delgado-Lista, J.;
Quintana-Navarro, G.M.; Landa, B.B.; Navas-Cortes, J.A.; Tena-Sempere, M.; et al. Intestinal microbiota is
influenced by gender and body mass index. PLoS ONE 2016, 11, e0154090. [CrossRef] [PubMed]
54. Hu, H.J.; Park, S.G.; Jang, H.B.; Choi, M.K.; Park, K.H.; Kang, J.H.; Park, S.I.; Lee, H.J.; Cho, S.H. Obesity
alters the microbial community profile in Korean adolescents. PLoS ONE 2015, 10, e0134333. [CrossRef]
[PubMed]
55. Aron-Wisnewsky, J.; Prifti, E.; Belda, E.; Ichou, F.; Kayser, B.D.; Dao, M.C.; Verger, E.O.; Hedjazi, L.;
Bouillot, J.-L.; Chevallier, J.-M.; et al. Major microbiota dysbiosis in severe obesity: Fate after bariatric surgery.
Gut 2018, 68, 70–82. [CrossRef] [PubMed]
56. Million, M.; Maraninchi, M.; Henry, M.; Armougom, F.; Richet, H.; Carrieri, P.; Valero, R.; Raccah, D.;
Vialettes, B.; Raoult, D. Obesity-associated gut microbiota is enriched in Lactobacillus reuteri and depleted
in Bifidobacterium animalis and Methanobrevibacter smithii. Int. J. Obes. (Lond.) 2012, 36, 817–825. [CrossRef]
[PubMed]
57. Balamurugan, R.; George, G.; Kabeerdoss, J.; Hepsiba, J.; Chandragunasekaran, A.M.; Ramakrishna, B.S.
Quantitative differences in intestinal Faecalibacterium prausnitzii in obese Indian children. Br. J. Nutr. 2010,
103, 335–338. [CrossRef]
58. Fuller, R. Probiotics in man and animals. J. Appl. Bacteriol. 1989, 66, 365–378.
59. Schrezenmeir, J.; de Vrese, M. Probiotics, prebiotics, and synbiotics—Approaching a definition. Am. J.
Clin. Nutr. 2001, 73, 361S–364S. [CrossRef]
60. He, M.; Shi, B. Gut microbiota as a potential target of metabolic syndrome: The role of probiotics and
prebiotics. Cell Biosci. 2017, 7, 54. [CrossRef]
61. Selle, K.; Klaenhammer, T.R. Genomic and phenotypic evidence for probiotic influences of Lactobacillus
gasseri on human health. FEMS Microbiol. Rev. 2013, 37, 915–935. [CrossRef] [PubMed]
62. Ohland, C.L.; Macnaughton, W.K. Probiotic bacteria and intestinal epithelial barrier function. Am. J. Physiol.
Gastrointest. Liver Physiol. 2010, 298, G807–G819. [CrossRef] [PubMed]
63. Hooper, L.V.; Wong, M.H.; Thelin, A.; Hansson, L.; Falk, P.G.; Gordon, J.I. Molecular analysis of commensal
host-microbial relationships in the intestine. Science 2001, 291, 881–884. [CrossRef] [PubMed]
64. Anderson, R.C.; Cookson, A.L.; McNabb, W.C.; Park, Z.; McCann, M.J.; Kelly, W.J.; Roy, N.C. Lactobacillus
plantarum MB452 enhances the function of the intestinal barrier by increasing the expression levels of genes
involved in tight junction formation. BMC Microbiol. 2010, 10, 316. [CrossRef] [PubMed]
65. Hummel, S.; Veltman, K.; Cichon, C.; Sonnenborn, U.; Schmidt, M.A. Differential targeting of the
e-cadherin/beta-catenin complex by gram-positive probiotic lactobacilli improves epithelial barrier function.
Appl. Environ. Microbiol. 2012, 78, 1140–1147. [CrossRef] [PubMed]
66. Zyrek, A.A.; Cichon, C.; Helms, S.; Enders, C.; Sonnenborn, U.; Schmidt, M.A. Molecular mechanisms
underlying the probiotic effects of Escherichia coli nissle 1917 involve ZO-2 and PKCzeta redistribution
resulting in tight junction and epithelial barrier repair. Cell Microbiol 2007, 9, 804–816. [CrossRef]
67. Caballero-Franco, C.; Keller, K.; De Simone, C.; Chadee, K. The VSL#3 probiotic formula induces mucin
gene expression and secretion in colonic epithelial cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2007, 292,
G315–G322.
68. Dai, C.; Zhao, D.H.; Jiang, M. Vsl#3 probiotics regulate the intestinal epithelial barrier in vivo and in vitro
via the p38 and erk signaling pathways. Int. J. Mol. Med. 2012, 29, 202–208.
69. Juntunen, M.; Kirjavainen, P.V.; Ouwehand, A.C.; Salminen, S.J.; Isolauri, E. Adherence of probiotic bacteria
to human intestinal mucus in healthy infants and during rotavirus infection. Clin. Diagn. Lab. Immunol. 2001,
8, 293–296. [CrossRef]
70. Buck, B.L.; Altermann, E.; Svingerud, T.; Klaenhammer, T.R. Functional analysis of putative adhesion factors
in Lactobacillus acidophilus NCFM. Appl. Environ. Microbiol. 2005, 71, 8344–8351. [CrossRef]
71. Candela, M.; Bergmann, S.; Vici, M.; Vitali, B.; Turroni, S.; Eikmanns, B.J.; Hammerschmidt, S.; Brigidi, P.
Binding of human plasminogen to bifidobacterium. J. Bacteriol. 2007, 189, 5929–5936. [CrossRef]
Nutrients 2019, 11, 258 20 of 24
72. Lee, K.; Paek, K.; Lee, H.Y.; Park, J.H.; Lee, Y. Antiobesity effect of trans-10,cis-12-conjugated linoleic
acid-producing Lactobacillus plantarum PL62 on diet-induced obese mice. J. Appl. Microbiol. 2007, 103,
1140–1146. [CrossRef] [PubMed]
73. O’Shea, E.F.; Cotter, P.D.; Stanton, C.; Ross, R.P.; Hill, C. Production of bioactive substances by intestinal
bacteria as a basis for explaining probiotic mechanisms: Bacteriocins and conjugated linoleic acid. Int. J.
Food Microbiol. 2012, 152, 189–205. [CrossRef] [PubMed]
74. Nielsen, D.S.; Cho, G.S.; Hanak, A.; Huch, M.; Franz, C.M.; Arneborg, N. The effect of bacteriocin-producing
lactobacillus plantarum strains on the intracellular pH of sessile and planktonic listeria monocytogenes
single cells. Int. J. Food Microbiol. 2010, 141 (Suppl. 1), S53–S59. [CrossRef] [PubMed]
75. Callaway, T.R.; Edrington, T.S.; Anderson, R.C.; Harvey, R.B.; Genovese, K.J.; Kennedy, C.N.; Venn, D.W.;
Nisbet, D.J. Probiotics, prebiotics and competitive exclusion for prophylaxis against bacterial disease.
Anim. Health Res. Rev. 2008, 9, 217–225. [CrossRef] [PubMed]
76. Chenoll, E.; Casinos, B.; Bataller, E.; Astals, P.; Echevarria, J.; Iglesias, J.R.; Balbarie, P.; Ramon, D.; Genoves, S.
Novel probiotic Bifidobacterium bifidum cect 7366 strain active against the pathogenic bacterium helicobacter
pylori. Appl. Environ. Microbiol. 2011, 77, 1335–1343. [CrossRef] [PubMed]
77. Coconnier, M.H.; Bernet, M.F.; Chauviere, G.; Servin, A.L. Adhering heat-killed human Lactobacillus
acidophilus, strain LB, inhibits the process of pathogenicity of diarrhoeagenic bacteria in cultured human
intestinal cells. J. Diarrhoeal Dis. Res. 1993, 11, 235–242.
78. Shokryazdan, P.; Sieo, C.C.; Kalavathy, R.; Liang, J.B.; Alitheen, N.B.; Faseleh Jahromi, M.; Ho, Y.W.
Probiotic potential of lactobacillus strains with antimicrobial activity against some human pathogenic
strains. BioMed Res. Int. 2014, 2014, 927268. [CrossRef]
79. Yan, F.; Polk, D.B. Probiotics and immune health. Curr. Opin. Gastroenterol. 2011, 27, 496–501. [CrossRef]
80. van Baarlen, P.; Wells, J.M.; Kleerebezem, M. Regulation of intestinal homeostasis and immunity with
probiotic lactobacilli. Trends Immunol. 2013, 34, 208–215. [CrossRef]
81. Wells, J.M. Immunomodulatory mechanisms of lactobacilli. Microb. Cell Fact. 2011, 10 (Suppl. 1), S17.
[CrossRef] [PubMed]
82. Kang, Y.; Cai, Y. The development of probiotics therapy to obesity: A therapy that has gained considerable
momentum. Hormones (Athens) 2018, 17, 141–151. [CrossRef]
83. Lee, H.Y.; Park, J.H.; Seok, S.H.; Baek, M.W.; Kim, D.J.; Lee, K.E.; Paek, K.S.; Lee, Y.; Park, J.H. Human
originated bacteria, Lactobacillus rhamnosus PL60, produce conjugated linoleic acid and show anti-obesity
effects in diet-induced obese mice. Biochim. Biophys. Acta 2006, 1761, 736–744. [CrossRef] [PubMed]
84. Kim, S.W.; Park, K.Y.; Kim, B.; Kim, E.; Hyun, C.K. Lactobacillus rhamnosus gg improves insulin sensitivity and
reduces adiposity in high-fat diet-fed mice through enhancement of adiponectin production. Biochem. Biophys.
Res. Commun. 2013, 431, 258–263. [CrossRef]
85. Singh, S.; Sharma, R.K.; Malhotra, S.; Pothuraju, R.; Shandilya, U.K. Lactobacillus rhamnosus ncdc17
ameliorates type-2 diabetes by improving gut function, oxidative stress and inflammation in high-fat-diet
fed and streptozotocintreated rats. Benef. Microbes 2017, 8, 243–255. [CrossRef] [PubMed]
86. Pothuraju, R.; Sharma, R.K.; Chagalamarri, J.; Kavadi, P.K.; Jangra, S. Influence of milk fermented with
Lactobacillus rhamnosus ncdc 17 alone and in combination with herbal ingredients on diet induced adiposity
and related gene expression in c57bl/6j mice. Food Funct. 2015, 6, 3576–3584. [CrossRef] [PubMed]
87. Park, S.Y.; Seong, K.S.; Lim, S.D. Anti-obesity effect of yogurt fermented by Lactobacillus plantarum Q180 in
diet-induced obese rats. Korean J. Food Sci. Anim. Resour. 2016, 36, 77–83. [CrossRef]
88. Hong, S.M.; Chung, E.C.; Kim, C.H. Anti-obesity effect of fermented whey beverage using lactic acid bacteria
in diet-induced obese rats. Korean J. Food Sci. Anim. Resour. 2015, 35, 653–659. [CrossRef] [PubMed]
89. Park, J.E.; Oh, S.H.; Cha, Y.S. Lactobacillus plantarum LG42 isolated from gajami sik-hae decreases body and
fat pad weights in diet-induced obese mice. J. Appl. Microbiol. 2014, 116, 145–156. [CrossRef]
90. Ben Salah, R.; Trabelsi, I.; Hamden, K.; Chouayekh, H.; Bejar, S. Lactobacillus plantarum TN8 exhibits protective
effects on lipid, hepatic and renal profiles in obese rat. Anaerobe 2013, 23, 55–61. [CrossRef]
91. Lee, E.; Jung, S.R.; Lee, S.Y.; Lee, N.K.; Paik, H.D.; Lim, S.I. Lactobacillus plantarum strain Ln4 attenuates
diet-induced obesity, insulin resistance, and changes in hepatic mrna levels associated with glucose and
lipid metabolism. Nutrients 2018, 10, 643. [CrossRef] [PubMed]
92. Takemura, N.; Okubo, T.; Sonoyama, K. Lactobacillus plantarum strain no. 14 reduces adipocyte size in mice
fed high-fat diet. Exp. Biol. Med. (Maywood) 2010, 235, 849–856. [CrossRef] [PubMed]
Nutrients 2019, 11, 258 21 of 24
93. Pothuraju, R.; Sharma, R.K.; Kavadi, P.K.; Chagalamarri, J.; Jangra, S.; Bhakri, G.; De, S. Anti-obesity effect of
milk fermented by Lactobacillus plantarum ncdc 625 alone and in combination with herbs on high fat diet fed
c57bl/6j mice. Benef. Microbes 2016, 7, 375–385. [CrossRef] [PubMed]
94. Miyoshi, M.; Ogawa, A.; Higurashi, S.; Kadooka, Y. Anti-obesity effect of Lactobacillus gasseri SBT2055
accompanied by inhibition of pro-inflammatory gene expression in the visceral adipose tissue in diet-induced
obese mice. Eur. J. Nutr. 2014, 53, 599–606. [CrossRef] [PubMed]
95. Sato, M.; Uzu, K.; Yoshida, T.; Hamad, E.M.; Kawakami, H.; Matsuyama, H.; Abd El-Gawad, I.A.;
Imaizumi, K. Effects of milk fermented by Lactobacillus gasseri SBT2055 on adipocyte size in rats. Br. J. Nutr.
2008, 99, 1013–1017. [CrossRef] [PubMed]
96. 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. J. Microbiol. 2010, 48, 712–714. [CrossRef] [PubMed]
97. Kang, J.H.; Yun, S.I.; Park, M.H.; Park, J.H.; Jeong, S.Y.; Park, H.O. Anti-obesity effect of Lactobacillus gasseri
BNR17 in high-sucrose diet-induced obese mice. PLoS ONE 2013, 8, e54617. [CrossRef]
98. Tanida, M.; Shen, J.; Maeda, K.; Horii, Y.; Yamano, T.; Fukushima, Y.; Nagai, K. High-fat diet-induced obesity
is attenuated by probiotic strain Lactobacillus paracasei ST11 (NCC2461) in rats. Obes. Res. Clin. Pract. 2008, 2,
159–169. [CrossRef]
99. Thiennimitr, P.; Yasom, S.; Tunapong, W.; Chunchai, T.; Wanchai, K.; Pongchaidecha, A.; Lungkaphin, A.;
Sirilun, S.; Chaiyasut, C.; Chattipakorn, N.; et al. Lactobacillus paracasei hii01, xylooligosaccharides, and
synbiotics reduce gut disturbance in obese rats. Nutrition 2018, 54, 40–47. [CrossRef]
100. Ji, Y.S.; Kim, H.N.; Park, H.J.; Lee, J.E.; Yeo, S.Y.; Yang, J.S.; Park, S.Y.; Yoon, H.S.; Cho, G.S.; Franz, C.M.; et al.
Modulation of the murine microbiome with a concomitant anti-obesity effect by Lactobacillus rhamnosus gg
and lactobacillus sakei nr28. Benef. Microbes 2012, 3, 13–22. [CrossRef]
101. Pothuraju, R.; Sharma, R.K. Interplay of gut microbiota, probiotics in obesity: A review. Endocr. Metab.
Immune Disord. Drug Targets 2018, 18, 212–220. [CrossRef] [PubMed]
102. Everard, A.; Matamoros, S.; Geurts, L.; Delzenne, N.M.; Cani, P.D. Saccharomyces boulardii administration
changes gut microbiota and reduces hepatic steatosis, low-grade inflammation, and fat mass in obese and
type 2 diabetic db/db mice. MBio 2014, 5, e01011-14. [CrossRef] [PubMed]
103. Murphy, E.F.; Cotter, P.D.; Hogan, A.; O’Sullivan, O.; Joyce, A.; Fouhy, F.; Clarke, S.F.; Marques, T.M.;
O’Toole, P.W.; Stanton, C.; et al. Divergent metabolic outcomes arising from targeted manipulation of the gut
microbiota in diet-induced obesity. Gut 2013, 62, 220–226. [CrossRef] [PubMed]
104. Arora, T.; Anastasovska, J.; Gibson, G.; Tuohy, K.; Sharma, R.K.; Bell, J.; Frost, G. Effect of lactobacillus
acidophilus ncdc 13 supplementation on the progression of obesity in diet-induced obese mice. Br. J. Nutr.
2012, 108, 1382–1389. [CrossRef] [PubMed]
105. An, H.M.; Park, S.Y.; Lee, D.K.; Kim, J.R.; Cha, M.K.; Lee, S.W.; Lim, H.T.; Kim, K.J.; Ha, N.J. Antiobesity and
lipid-lowering effects of Bifidobacterium spp. In high fat diet-induced obese rats. Lipids Health Dis. 2011, 10,
116. [CrossRef]
106. Yin, Y.N.; Yu, Q.F.; Fu, N.; Liu, X.W.; Lu, F.G. Effects of four bifidobacteria on obesity in high-fat diet induced
rats. World J. Gastroenterol. 2010, 16, 3394–3401. [CrossRef] [PubMed]
107. Chen, J.; Wang, R.; Li, X.F.; Wang, R.L. Bifidobacterium adolescentis supplementation ameliorates visceral
fat accumulation and insulin sensitivity in an experimental model of the metabolic syndrome. Br. J. Nutr.
2012, 107, 1429–1434. [CrossRef] [PubMed]
108. Reichold, A.; Brenner, S.A.; Spruss, A.; Forster-Fromme, K.; Bergheim, I.; Bischoff, S.C. Bifidobacterium
adolescentis protects from the development of nonalcoholic steatohepatitis in a mouse model.
J. Nutr. Biochem. 2014, 25, 118–125. [CrossRef] [PubMed]
109. Wang, J.; Tang, H.; Zhang, C.; Zhao, Y.; Derrien, M.; Rocher, E.; van-Hylckama Vlieg, J.E.; Strissel, K.; Zhao, L.;
Obin, M.; et al. Modulation of gut microbiota during probiotic-mediated attenuation of metabolic syndrome
in high fat diet-fed mice. ISME J. 2015, 9, 1–15. [CrossRef] [PubMed]
110. Zhao, X.; Higashikawa, F.; Noda, M.; Kawamura, Y.; Matoba, Y.; Kumagai, T.; Sugiyama, M. The obesity
and fatty liver are reduced by plant-derived Pediococcus pentosaceus lp28 in high fat diet-induced obese mice.
PLoS ONE 2012, 7, e30696. [CrossRef]
111. Gauffin Cano, P.; Santacruz, A.; Moya, A.; Sanz, Y. Bacteroides uniformis cect 7771 ameliorates metabolic and
immunological dysfunction in mice with high-fat-diet induced obesity. PLoS ONE 2012, 7, e41079. [CrossRef]
[PubMed]
Nutrients 2019, 11, 258 22 of 24
112. Poutahidis, T.; Kleinewietfeld, M.; Smillie, C.; Levkovich, T.; Perrotta, A.; Bhela, S.; Varian, B.J.; Ibrahim, Y.M.;
Lakritz, J.R.; Kearney, S.M.; et al. Microbial reprogramming inhibits western diet-associated obesity.
PLoS ONE 2013, 8, e68596. [CrossRef] [PubMed]
113. Santacruz, A.; Collado, M.C.; Garcia-Valdes, L.; Segura, M.T.; Martin-Lagos, J.A.; Anjos, T.; Marti-Romero, M.;
Lopez, R.M.; Florido, J.; Campoy, C.; et al. Gut microbiota composition is associated with body weight,
weight gain and biochemical parameters in pregnant women. Br. J. Nutr. 2010, 104, 83–92. [CrossRef]
[PubMed]
114. Derrien, M.; Vaughan, E.E.; Plugge, C.M.; de Vos, W.M. Akkermansia muciniphila gen. Nov., sp. Nov., a
human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol. 2004, 54, 1469–1476. [CrossRef]
115. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.;
Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between akkermansia muciniphila and intestinal epithelium
controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [CrossRef] [PubMed]
116. Yoo, S.R.; Kim, Y.J.; Park, D.Y.; Jung, U.J.; Jeon, S.M.; Ahn, Y.T.; Huh, C.S.; McGregor, R.; Choi, M.S. Probiotics
L. plantarum and L. curvatus in combination alter hepatic lipid metabolism and suppress diet-induced obesity.
Obesity (Silver Spring) 2013, 21, 2571–2578. [CrossRef] [PubMed]
117. Savcheniuk, O.; Kobyliak, N.; Kondro, M.; Virchenko, O.; Falalyeyeva, T.; Beregova, T. Short-term periodic
consumption of multiprobiotic from childhood improves insulin sensitivity, prevents development of
non-alcoholic fatty liver disease and adiposity in adult rats with glutamate-induced obesity. BMC Complement.
Altern. Med. 2014, 14, 247. [CrossRef]
118. Kobyliak, N.; Falalyeyeva, T.; Beregova, T.; Spivak, M. Probiotics for experimental obesity prevention: Focus
on strain dependence and viability of composition. Endokrynol. Pol. 2017, 68, 659–667. [CrossRef]
119. Alard, J.; Lehrter, V.; Rhimi, M.; Mangin, I.; Peucelle, V.; Abraham, A.L.; Mariadassou, M.; Maguin, E.;
Waligora-Dupriet, A.J.; Pot, B.; et al. Beneficial metabolic effects of selected probiotics on diet-induced obesity
and insulin resistance in mice are associated with improvement of dysbiotic gut microbiota. Environ. Microbiol.
2016, 18, 1484–1497. [CrossRef]
120. Karimi, G.; Jamaluddin, R.; Mohtarrudin, N.; Ahmad, Z.; Khazaai, H.; Parvaneh, M. Single-species versus
dual-species probiotic supplementation as an emerging therapeutic strategy for obesity. Nutr. Metab.
Cardiovasc. Dis. 2017, 27, 910–918. [CrossRef]
121. Roselli, M.; Finamore, A.; Brasili, E.; Rami, R.; Nobili, F.; Orsi, C.; Zambrini, A.V.; Mengheri, E. Beneficial
effects of a selected probiotic mixture administered to high fat-fed mice before and after the development of
obesity. J. Funct. Foods 2018, 45, 321–329. [CrossRef]
122. Luoto, R.; Kalliomäki, 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. Int. J. Obes. 2010, 34,
1531–1537. [CrossRef]
123. Kadooka, Y.; Sato, M.; Imaizumi, K.; Ogawa, A.; Ikuyama, K.; Akai, Y.; Okano, M.; Kagoshima, M.; Tsuchida, T.
Regulation of abdominal adiposity by probiotics (Lactobacillus gasseri sbt2055) in adults with obese tendencies
in a randomized controlled trial. Eur. J. Clin. Nutr. 2010, 64, 636–643. [CrossRef]
124. Jung, S.P.; Lee, K.M.; Kang, J.H.; Yun, S.I.; Park, H.O.; Moon, Y.; Kim, J.Y. Effect of Lactobacillus gasseri bnr17
on overweight and obese adults: A randomized, double-blind clinical trial. Korean J. Fam. Med. 2013, 34,
80–89. [CrossRef]
125. Stadlbauer, V.; Leber, B.; Lemesch, S.; Trajanoski, S.; Bashir, M.; Horvath, A.; Tawdrous, M.; Stojakovic, T.;
Fauler, G.; Fickert, P.; et al. Lactobacillus casei shirota supplementation does not restore gut microbiota
composition and gut barrier in metabolic syndrome: A randomized pilot study. PLoS ONE 2015, 10, e0141399.
[CrossRef]
126. Brahe, L.K.; Le Chatelier, E.; Prifti, E.; Pons, N.; Kennedy, S.; Blaedel, T.; Hakansson, J.; Dalsgaard, T.K.;
Hansen, T.; Pedersen, O.; et al. Dietary modulation of the gut microbiota—A randomised controlled trial in
obese postmenopausal women. Br. J. Nutr. 2015, 114, 406–417. [CrossRef]
127. Jones, M.L.; Martoni, C.J.; Di Pietro, E.; Simon, R.R.; Prakash, S. Evaluation of clinical safety and tolerance of
a Lactobacillus reuteri ncimb 30242 supplement capsule: A randomized control trial. Regul. Toxicol. Pharmacol.
2012, 63, 313–320. [CrossRef]
128. Chung, H.J.; Yu, J.G.; Lee, I.A.; Liu, M.J.; Shen, Y.F.; Sharma, S.P.; Jamal, M.A.; Yoo, J.H.; Kim, H.J.; Hong, S.T.
Intestinal removal of free fatty acids from hosts by lactobacilli for the treatment of obesity. FEBS Open Bio
2016, 6, 64–76. [CrossRef]
Nutrients 2019, 11, 258 23 of 24
129. Hariri, M.; Salehi, R.; Feizi, A.; Mirlohi, M.; Kamali, S.; Ghiasvand, R. The effect of probiotic soy milk
and soy milk on anthropometric measures and blood pressure in patients with type II diabetes mellitus:
A randomized double-blind clinical trial. ARYA Atheroscler. 2015, 11, 74–80.
130. Sharafedtinov, K.K.; Plotnikova, O.A.; Alexeeva, R.I.; Sentsova, T.B.; Songisepp, E.; Stsepetova, J.; Smidt, I.;
Mikelsaar, M. Hypocaloric diet supplemented with probiotic cheese improves body mass index and blood
pressure indices of obese hypertensive patients—A randomized double-blind placebo-controlled pilot study.
Nutr. J. 2013, 12, 138. [CrossRef]
131. Schwiertz, A.; Taras, D.; Schafer, K.; Beijer, S.; Bos, N.A.; Donus, C.; Hardt, P.D. Microbiota and scfa in lean
and overweight healthy subjects. Obesity (Silver Spring) 2010, 18, 190–195. [CrossRef] [PubMed]
132. Gøbel, R.J.; Larsen, N.; Jakobsen, M.; Mølgaard, C.; Michaelsen, K.F. Probiotics to adolescents with obesity:
Effects on inflammation and metabolic syndrome. J. Pediatr. Gastroenterol. Nutr. 2012, 55, 673–678. [CrossRef]
[PubMed]
133. Larsen, N.; Vogensen, F.K.; Gobel, R.J.; Michaelsen, K.F.; Forssten, S.D.; Lahtinen, S.J.; Jakobsen, M. Effect
of lactobacillus salivarius ls-33 on fecal microbiota in obese adolescents. Clin. Nutr. 2013, 32, 935–940.
[CrossRef] [PubMed]
134. Jones, R.B.; Alderete, T.L.; Martin, A.A.; Geary, B.A.; Hwang, D.H.; Palmer, S.L.; Goran, M.I. Probiotic
supplementation increases obesity with no detectable effects on liver fat or gut microbiota in obese hispanic
adolescents: A 16-week, randomized, placebo-controlled trial. Pediatr. Obes. 2018, 13, 705–714. [CrossRef]
[PubMed]
135. Higashikawa, F.; Noda, M.; Awaya, T.; Danshiitsoodol, N.; Matoba, Y.; Kumagai, T.; Sugiyama, M. Antiobesity
effect of Pediococcus pentosaceus lp28 on overweight subjects: A randomized, double-blind, placebo-controlled
clinical trial. Eur. J. Clin. Nutr. 2016, 70, 582–587. [CrossRef] [PubMed]
136. Bernini, L.J.; Simao, A.N.; Alfieri, D.F.; Lozovoy, M.A.; Mari, N.L.; de Souza, C.H.; Dichi, I.; Costa, G.N.
Beneficial effects of bifidobacterium lactis on lipid profile and cytokines in patients with metabolic syndrome:
A randomized trial. Effects of probiotics on metabolic syndrome. Nutrition 2016, 32, 716–719. [CrossRef]
137. Chang, B.J.; Park, S.U.; Jang, Y.S.; Ko, S.H.; Joo, N.M.; Kim, S.I.; Kim, C.H.; Chang, D.K. Effect of functional
yogurt NY-YP901 in improving the trait of metabolic syndrome. Eur. J. Clin. Nutr. 2011, 65, 1250–1255.
[CrossRef]
138. Spaiser, S.J.; Culpepper, T.; Nieves, C., Jr.; Ukhanova, M.; Mai, V.; Percival, S.S.; Christman, M.C.;
Langkamp-Henken, B. Lactobacillus gasseri ks-13, bifidobacterium bifidum g9-1, and bifidobacterium
longum mm-2 ingestion induces a less inflammatory cytokine profile and a potentially beneficial shift in
gut microbiota in older adults: A randomized, double-blind, placebo-controlled, crossover study. J. Am.
Coll. Nutr. 2015, 34, 459–469.
139. Nabavi, S.; Rafraf, M.; Somi, M.-H.; Homayouni-Rad, A.; Asghari-Jafarabadi, M. Probiotic yogurt improves
body mass index and fasting insulin levels without affecting serum leptin and adiponectin levels in
non-alcoholic fatty liver disease (NAFLD). J. Funct. Foods 2015, 18, 684–691. [CrossRef]
140. Madjd, A.; Taylor, M.A.; Mousavi, N.; Delavari, A.; Malekzadeh, R.; Macdonald, I.A.; Farshchi, H.R.
Comparison of the effect of daily consumption of probiotic compared with low-fat conventional yogurt on
weight loss in healthy obese women following an energy-restricted diet: A randomized controlled trial1.
Am. J. Clin. Nutr. 2016, 103, 323–329. [CrossRef]
141. Gomes, A.C.; de Sousa, R.G.; Botelho, P.B.; Gomes, T.L.; Prada, P.O.; Mota, J.F. The additional effects
of a probiotic mix on abdominal adiposity and antioxidant status: A double-blind, randomized trial.
Obesity (Silver Spring) 2017, 25, 30–38. [CrossRef] [PubMed]
142. Szulinska, M.; Loniewski, I.; van Hemert, S.; Sobieska, M.; Bogdanski, P. Dose-dependent effects of
multispecies probiotic supplementation on the lipopolysaccharide (LPS) level and cardiometabolic profile
in obese postmenopausal women: A 12-week randomized clinical trial. Nutrients 2018, 10, 773. [CrossRef]
[PubMed]
143. Hamad, E.M.; Sato, M.; Uzu, K.; Yoshida, T.; Higashi, S.; Kawakami, H.; Kadooka, Y.; Matsuyama, H.; Abd
El-Gawad, I.A.; Imaizumi, K. Milk fermented by Lactobacillus gasseri sbt2055 influences adipocyte size via
inhibition of dietary fat absorption in zucker rats. Br. J. Nutr. 2009, 101, 716–724. [CrossRef]
144. Renga, B.; Mencarelli, A.; Vavassori, P.; Brancaleone, V.; Fiorucci, S. The bile acid sensor fxr regulates insulin
transcription and secretion. Biochim. Biophys. Acta 2010, 1802, 363–372. [CrossRef]
Nutrients 2019, 11, 258 24 of 24
145. Shyangdan, D.S.; Royle, P.; Clar, C.; Sharma, P.; Waugh, N.; Snaith, A. Glucagon-like peptide analogues for
type 2 diabetes mellitus. Cochrane Database Syst. Rev. 2011, 10, CD006423. [CrossRef] [PubMed]
146. Mei, L.; Tang, Y.; Li, M.; Yang, P.; Liu, Z.; Yuan, J.; Zheng, P. Co-administration of cholesterol-lowering
probiotics and anthraquinone from cassia obtusifolia l. Ameliorate non-alcoholic fatty liver. PLoS ONE 2015,
10, e0138078. [CrossRef] [PubMed]
147. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.;
Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772.
[CrossRef]
148. Puddu, A.; Sanguineti, R.; Montecucco, F.; Viviani, G.L. Evidence for the gut microbiota short-chain fatty
acids as key pathophysiological molecules improving diabetes. Mediators Inflamm. 2014, 2014, 162021.
[CrossRef] [PubMed]
149. Cani, P.D.; Van Hul, M. Novel opportunities for next-generation probiotics targeting metabolic syndrome.
Curr. Opin. Biotechnol. 2015, 32, 21–27. [CrossRef] [PubMed]
150. Hiippala, K.; Jouhten, H.; Ronkainen, A.; Hartikainen, A.; Kainulainen, V.; Jalanka, J.; Satokari, R.
The potential of gut commensals in reinforcing intestinal barrier function and alleviating inflammation.
Nutrients 2018, 10, 988. [CrossRef]
151. Rutten, N.B.; Gorissen, D.M.; Eck, A.; Niers, L.E.; Vlieger, A.M.; Besseling-van der Vaart, I.; Budding, A.E.;
Savelkoul, P.H.; van der Ent, C.K.; Rijkers, G.T. Long term development of gut microbiota composition in
atopic children: Impact of probiotics. PLoS ONE 2015, 10, e0137681. [CrossRef] [PubMed]
152. Lundelin, K.; Poussa, T.; Salminen, S.; Isolauri, E. Long-term safety and efficacy of perinatal probiotic
intervention: Evidence from a follow-up study of four randomized, double-blind, placebo-controlled trials.
Pediatr. Allergy Immunol. 2017, 28, 170–175. [CrossRef] [PubMed]
153. Karimi, G.; Sabran, M.R.; Jamaluddin, R.; Parvaneh, K.; Mohtarrudin, N.; Ahmad, Z.; Khazaai, H.;
Khodavandi, A. The anti-obesity effects of Lactobacillus casei strain Shirota versus orlistat on high fat
diet-induced obese rats. Food Nutr. Res. 2015, 59, 29273. [CrossRef] [PubMed]
154. Nagata, S.; Chiba, Y.; Wang, C.; Yamashiro, Y. The effects of the Lactobacillus casei strain on obesity in children:
A pilot study. Benef. Microbes 2017, 8, 535–543. [CrossRef] [PubMed]
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).