ArticlePDF AvailableLiterature Review

Influence of gut microbiota on the development and progression of nonalcoholic steatohepatitis

Authors:

Abstract and Figures

Introduction: Nonalcoholic steatohepatitis (NASH) is characterized by the presence of steatosis, inflammation, and ballooning degeneration of hepatocytes, with or without fibrosis. The prevalence of NASH has increased with the obesity epidemic, but its etiology is multifactorial. The current studies suggest the role of gut microbiota in the development and progression of NASH. The aim is to review the studies that investigate the relationship between gut microbiota and NASH. These review also discusses the pathophysiological mechanisms and the influence of diet on the gut-liver axis. Result: The available literature has proposed mechanisms for an association between gut microbiota and NASH, such as: modification energy homeostasis, lipopolysaccharides (LPS)-endotoxemia, increased endogenous production of ethanol, and alteration in the metabolism of bile acid and choline. There is evidence to suggest that NASH patients have a higher prevalence of bacterial overgrowth in the small intestine and changes in the composition of the gut microbiota. However, there is still a controversy regarding the microbiome profile in this population. The abundance of Bacteroidetes phylum may be increased, decreased, or unaltered in NASH patients. There is an increase in the Escherichia and Bacteroides genus. There is depletion of certain taxa, such as Prevotella and Faecalibacterium. Conclusion: Although few studies have evaluated the composition of the gut microbiota in patients with NASH, it is observed that these individuals have a distinct gut microbiota, compared to the control groups, which explains, at least in part, the genesis and progression of the disease through multiple mechanisms. Modulation of the gut microbiota through diet control offers new challenges for future studies.
This content is subject to copyright. Terms and conditions apply.
1 23
European Journal of Nutrition
ISSN 1436-6207
Eur J Nutr
DOI 10.1007/s00394-017-1524-x
Influence of gut microbiota on the
development and progression of
nonalcoholic steatohepatitis
Fabiana de Faria Ghetti, Daiane
Gonçalves Oliveira, Juliano Machado de
Oliveira, Lincoln Eduardo Villela Vieira
de Castro Ferreira, et al.
1 23
Your article is protected by copyright and
all rights are held exclusively by Springer-
Verlag GmbH Germany. This e-offprint is
for personal use only and shall not be self-
archived in electronic repositories. If you wish
to self-archive your article, please use the
accepted manuscript version for posting on
your own website. You may further deposit
the accepted manuscript version in any
repository, provided it is only made publicly
available 12 months after official publication
or later and provided acknowledgement is
given to the original source of publication
and a link is inserted to the published article
on Springer's website. The link must be
accompanied by the following text: "The final
publication is available at link.springer.com”.
Vol.:(0123456789)
1 3
Eur J Nutr
DOI 10.1007/s00394-017-1524-x
REVIEW
Influence ofgut microbiota onthedevelopment andprogression
ofnonalcoholic steatohepatitis
FabianadeFariaGhetti1,4 · DaianeGonçalvesOliveira1·
JulianoMachadodeOliveira1· LincolnEduardoVillelaVieiradeCastroFerreira1·
DionéiaEvangelistaCesar2· AnaPaulaBoroniMoreira3
Received: 31 January 2017 / Accepted: 6 August 2017
© Springer-Verlag GmbH Germany 2017
unaltered in NASH patients. There is an increase in the
Escherichia and Bacteroides genus. There is depletion of
certain taxa, such as Prevotella and Faecalibacterium.
Conclusion Although few studies have evaluated the com-
position of the gut microbiota in patients with NASH, it is
observed that these individuals have a distinct gut microbi-
ota, compared to the control groups, which explains, at least
in part, the genesis and progression of the disease through
multiple mechanisms. Modulation of the gut microbiota
through diet control offers new challenges for future studies.
Keywords Nonalcoholic fatty liver disease· Dysbiosis·
Steatohepatitis· Gut microbiota· Microbiome
Introduction
Nonalcoholic steatohepatitis (NASH) is the most severe his-
tological form of nonalcoholic fatty liver disease (NAFLD)
[1], characterized by the presence of hepatic steatosis and
inflammation, associated with ballooning degeneration, with
or without fibrosis [2]. Even as the prevalence of NASH in
the general population is approximately 2–5% [3, 4], around
70% of morbidly obese individuals are affected by this con-
dition [5]. Although in most cases, NASH carriers present
no symptoms, this condition may increase the risk of cirrho-
sis, liver failure, and hepatocellular carcinoma [69].
The exact cause of NASH is not yet clear, but studies have
suggested the role of the gut microbiota in the pathogenesis
of this disease [1012]. In fact, it has been shown in animal
models that gut microbiota increase intrahepatic fat through
mechanisms associated with increased dietary energy extrac-
tion or change in lipogenesis and β-oxidation [10, 11]. Fur-
thermore, hepatocellular inflammation may be secondary
to increased intestinal permeability and translocation of
Abstract
Introduction Nonalcoholic steatohepatitis (NASH) is char-
acterized by the presence of steatosis, inflammation, and bal-
looning degeneration of hepatocytes, with or without fibro-
sis. The prevalence of NASH has increased with the obesity
epidemic, but its etiology is multifactorial. The current stud-
ies suggest the role of gut microbiota in the development
and progression of NASH. The aim is to review the studies
that investigate the relationship between gut microbiota and
NASH. These review also discusses the pathophysiological
mechanisms and the influence of diet on the gut–liver axis.
Result The available literature has proposed mechanisms
for an association between gut microbiota and NASH, such
as: modification energy homeostasis, lipopolysaccharides
(LPS)–endotoxemia, increased endogenous production of
ethanol, and alteration in the metabolism of bile acid and
choline. There is evidence to suggest that NASH patients
have a higher prevalence of bacterial overgrowth in the
small intestine and changes in the composition of the gut
microbiota. However, there is still a controversy regarding
the microbiome profile in this population. The abundance
of Bacteroidetes phylum may be increased, decreased, or
* Fabiana de Faria Ghetti
bia.ghetti@hotmail.com
1 Universitary Hospital andSchool ofMedicine, Federal
University ofJuiz de Fora, JuizdeFora, MinasGerais,
Brazil
2 Department ofBiology, Federal University ofJuiz de Fora,
JuizdeFora, MinasGerais, Brazil
3 Department ofNutrition, Federal University ofJuiz de Fora,
JuizdeFora, MinasGerais, Brazil
4 Unidade de Nutrição Clínica, Hospital Universitário, Rua
Catulo Breviglieri, s/n, Bairro Santa Catarina, JuizdeFora,
MinasGeraisCEP36036-330, Brazil
Author's personal copy
Eur J Nutr
1 3
microbial cell components to the circulation [12]. Finally,
gut microbiota can contribute to fibrogenesis through activa-
tion of hepatic stellate cells [13].
Although animal experiments have associated gut micro-
biota with the histological components of NAFLD, there
are few clinical studies with emphasis on the composition
and functionality of the microbiome in NASH. There is evi-
dence that NASH patients have a higher prevalence of small
intestinal bacterial overgrowth (SIBO) [14, 15] and the low-
est percentage of Bacteroidetes in their fecal content when
compared to healthy subjects [16]. On the other hand, other
studies have observed a higher abundance of Bacteroidetes
in the gut microbiota of patients with NASH, compared to
healthy controls [17, 18]. Thus, the results of the studies are
still controversial regarding the microbiome profile in this
population.
A detailed study of the composition of the gut microbiota
and its metabolic functions can determine which microor-
ganisms contribute to gut health maintenance and what
changes can lead to the development of pathologies [19].
Therefore, the aim of this review is to highlight the studies
that investigate the relationship between gut microbiota and
NASH. Pathophysiological mechanisms and the influence of
diet on the gut–liver axis are also discussed.
Development andprogression ofnonalcoholic
steatohepatitis
Traditionally, the pathogenesis of NASH is explained by
the hypothesis of “two hits” proposed by Day and James
[20], in 1998. According to the authors, insulin resistance
would be the first stimulus (the first “hit”) that determines
the accumulation of fat in hepatocytes, resulting in steatosis.
The steatosis itself increases the sensitivity of the liver to the
second “hit”. The second “hit” would be the oxidative stress,
which promotes liver injury, characterized by tissue lesions,
inflammation, and fibrosis [20].
According to the hypothesis of the “two hits”, insulin
resistance promotes hepatic lipogenesis and lipolysis in the
adipose tissue, increasing the amount of fatty acids released
to the liver in the initial event. On a smaller scale, the avail-
ability of fatty acids in the liver may result from the trans-
port mediated by lipoproteins after intestinal absorption of
dietary fats [21]. Upon entering the hepatocytes, the free
fatty acids are oxidized by mitochondria to generate energy
or are esterified in triacylglycerols (TG), incorporated into
very-low density lipoprotein (VLDL) particles, and exported
from the liver to the peripheral tissues [22]. When free fatty
acids in the hepatocytes exceed their metabolization and
export capacity, they can cause hepatic steatosis [22]. The
accumulation of fatty acids in the liver results in excessive
increase in the production of reactive oxygen species (ROS)
from the mitochondria [23]. In addition, peroxisomes and
microsomal oxidation pathways are activated and generate
more ROS, culminating in hepatic oxidative stress [24]. Oxi-
dative stress appears to be responsible for initiating necroin-
flammation. The consequence of oxidative stress is hepatic
lipid peroxidation in cell membranes and mitochondria, pro-
ducing malondialdehyde and hydroxynonenal, resulting in
mitochondrial dysfunction [25]. Malondialdehyde activates
the regulatory transcription factor and the expression of pro-
inflammatory cytokines and adhesion molecules (NF-kB),
stimulating production of the tumor necrosis factor alpha
(TNF-α), interleukin 8 (IL-8), and selectin E [26]. Hydrox-
ynonenal activates hepatic stellate cells, promoting colla-
gen deposition, and hence, the development of fibrosis [27].
Thus, ROS, lipid peroxidation products, and cytokines are
involved in the second hit, which induces the progression of
simple steatosis to NASH.
At present, it is believed that the process of “two hits”
is insufficient to explain the pathogenesis of this heteroge-
neous disease, particularly in non-obese individuals [28].
Furthermore, simple hepatic steatosis, which is benign and
nonprogressive in a majority of patients, and NASH, may
reflect different pathogenesis [29]. In fact, the accumulation
of free fatty acids in the liver occurs mainly in the form of
TG [30]. There is evidence to indicate that TG by themselves
is not hepatotoxic, at least in mice (BKS.Cg-m/Leprdb/J)
with steatohepatitis [30]. Therefore, TG synthesis seems
to be an adaptive, beneficial response insituations, where
hepatocytes are exposed to potentially toxic TG metabolites
[30]. On the contrary, several other lipids, such as free fatty
acids, diacylglycerol, cholesterol, ceramide, and phospholip-
ids, also accumulate in the liver, and they are considered as
‘‘aggressive’’ lipids [31], that induce endoplasmic reticulum
(ER) stress, mitochondrial dysfunction, and oxidative stress,
resulting in hepatic inflammation and fibrogenesis [32, 33].
In this sense, a new theory was proposed by Tilg etal.
[31]: the hypothesis of “multiple parallel hits”. This hypoth-
esis proposes that several concurrent and not consecutive
stimuli (“multiple parallel hits”) induce oxidative stress,
which results in both hepatic steatosis and steatohepatitis.
Even as the two “hits” hypothesis suggests that steatosis
always precedes inflammation, the “multiple parallel hits”
hypothesis indicates that inflammation in NASH may pre-
cede steatosis in some cases [31]. In this new paradigm,
NASH results not only from oxidative stress, but also from
the interaction between different “hits”, including altered
lipid metabolism, mitochondrial dysfunction, ER stress,
genetic predisposition, and gut microbiota alterations [31].
It is increasingly recognized that the gut microbiota is impli-
cated in the pathogenesis and progression of NASH [34].
Thus, the gut microbiota has become a topic of interest in
recent investigations and a potential target of intervention
[35, 36].
Author's personal copy
Eur J Nutr
1 3
Gut microbiota
The human gut contains a large number of microorganisms,
mostly bacteria, collectively called gut microbiota [37]. The
latest estimate for the total number of bacteria cells in the
body is around 40 trillion (3.8×1013), the same order as the
number of human cells (3.0×1013), and their total mass is
about 0.2kg [38]. More than 1000 cultured gastrointestinal
species have been identified in the human microbiota [39].
Recent advances in molecular techniques, sequencing and
bioinformatic programs, have allowed the identification of
specific taxonomic groups, that is, phyla, classes, orders,
families, genera, and bacterial species [40]. Currently,
metagenomic techniques have been used to characterize the
composition, diversity, and potential physiological effects of
entire microbial communities, without cultivation and isola-
tion of the members of the community [41].
The Firmicutes and Bacteroidetes are the most prevalent
phyla in adults, followed by Actinobacteria, Proteobacteria,
Fusobacteria, Spirochaetae, and Verrucomicrobia [42]. The
stomach and small intestine are rich in Firmicutes (Lac-
tobacillaceae) and Proteobacteria (Enterobacteriaceae),
whereas the large intestine shows higher counts of Bacte-
roidetes (Bacteroidaceae, Prevotellaceae, and Rikenellaceae)
and Firmicutes (Lachnospiraceae and Ruminococcaceae)
[43]. The composition of adult microbiota remains relatively
stable, although the microbial diversity is acquired within
the first hours post birth, and is shaped over time as the diet
becomes more complex and the immune-system matures
[44]. A combination of multiple factors, including genetic
and environmental characteristics (type of delivery, antibi-
otic therapy, diet composition, lifestyle, social interactions,
and exposure to various xenobiotics) shapes the gut micro-
biota to make every individual microbially unique [45].
The gut microbiota can also be very dynamic and change
rapidly, for example, in response to dietary changes. One
study shows that an increase in caloric intake from 2400 to
3400kcal/day (with a similar nutrient profile that includes
24% protein, 16% fat, and 60% carbohydrates) over 3days
increases Firmicutes ratio and decreases the ratio of Bacte-
roidetes [10]. Diet is a major factor driving the composition
and metabolism of the gut microbiota [40] and the influence
of the diet on gut microbiota will be described in more detail
in the next sessions.
Nowadays, the gut microbiota is considered a metabolic
organ, which performs a wide range of functions including
an important role in the physiology of energy homeostasis
[46]. For example, some members of the Firmicutes phylum
are among the butyrate-producing bacteria that increase the
energy extraction from the diet [47]. In contrast, members
of the phylum Bacteroidetes participate in carbohydrate
metabolism and accomplish this by expressing enzymes
similar to glycosyl transferases, glycoside hydrolases, and
polysaccharide lyases [48]. In this context, the knowledge of
the composition and functions associated with the microbial
community is fundamental, as alterations in the composition
of the gut microbiota and/or its functions (called ‘dysbiosis’)
are associated with metabolic diseases, such as NASH [17,
18, 49]. Although human studies are scarce in the literature
(Table1), animal experiments support the link between gut
microbiota and the development of NASH.
Dysbiosis andnonalcoholic steatohepatitis
Experimental data
Animal experiments have demonstrated direct roles for gut
microbiota in the development and progression of nonalco-
holic steatohepatitis (NASH). Using germ-free C57BL/6J
mice, Bäckhead etal. [50] have previously demonstrated that
mice devoid of gut microbiota are resistant to diet-induced
obesity, steatosis, and insulin resistance. Subsequently,
Le Roy etal. [51], using the transplantation experiment,
have shown that differences in microbiota composition can
determine responses to a high-fat diet (HFD) in mice and
contribute to the development of steatosis, independent of
obesity. In this study, the conventional C57BL/6J mice, fed
with an HFD, have generally displayed liver steatosis, hyper-
glycemia, and systemic inflammation (called the ‘respond-
ers’), but some mice are nonresponders, normoglycemic,
and have a lower level of systemic inflammation, with the
same diet. Germ-free mice have been colonized with gut
microbiota from either the responders or the nonrespond-
ers and then fed the same HFD. Despite a similar weight
gain, responder–receiver mice have been found to develop a
higher level of liver steatosis, glycemia, and insulin resist-
ance than nonresponder–receivers. Pyrosequencing of the
16S ribosomal RNA genes has revealed that responder and
nonresponder mice have distinct gut microbiota includ-
ing differences at the phylum, genera, and species levels.
Responder mice harbour a significantly increased number of
sequences belonging to the Firmicutes phylum, Barnesiella,
Roseburia genera, Lachnospiraceae bacterium 609, and
Barnesiella intestinihominis species [51].
Barnesiella intestinihominis, that is part of the Porphy-
romonadaceae family, in particular, showed an increase in
inflammasome-deficient mice (C57Bl/6) associated with
the progression of NASH. It was revealed that the nucle-
otide-binding domain, leucine-rich repeat protein (NLRP)
6, NLRP3 inflammasomes, and the effector protein IL-18
negatively regulated the exacerbated hepatic steatosis and
inflammation via modulation of the gut microbiota. Antibi-
otic treatment with ciprofloxacin and metronidazole reduced
the severity of NASH in inflammasome-deficient mice
and abolished transmission of the phenotype to wild-type
Author's personal copy
Eur J Nutr
1 3
animals, showing that gut microbiota drove NASH progres-
sion in this model [52].
Progression of NASH was also strictly related to reduced
microbial diversity and an increased ratio of Firmicutes to
Bacteroidetes in model C57BL/6J mice [53]. The abundance
of Bacteroides spp., Bacteroides vulgatus, Desulfovibrio
spp., Atopobium spp., Clostridium cocleatum, and Clostrid
ium xylanolyticumin was increased in these animals and
positively correlated with the increased levels of lipopoly-
saccharides (LPS) [53], an endotoxin present on the cell sur-
face of Gram-negative bacteria, which induced inflammation
[54]. The authors also observed a reduction in the abundance
of gut barrier-protecting bacteria, such as the Lactobacillus
spp. [53]. Another study with rats also showed that during
the progression of NASH, the levels of LPS were highly
increased. In addition, an increase was found in Escherichia
coli and Enterococcus as well as a decrease was seen in
Lactobacillus, Bifidobacteria, and Bacteroide [36]. Based on
the connection between the intestine and liver, also termed
‘gut–liver axis’, the gut microbiota and their metabolic by-
products may influence liver pathology [36].
Dysbiosis could also promote liver fibrogenesis. Indeed,
C57BL/6 mice fed an HFD developed more severe liver
fibrosis than control mice that were fed a standard chow
diet, by changes in gut microbiota, activating an inflamma-
some cascade [55]. HFD-related increases in liver fibrosis
were associated with an increase in the percentage of Gram-
negative (mainly Proteobacteria) versus Gram-positive bac-
teria (mainly reduction in Erysipelotrichaceae and a com-
plete disappearance of Bifidobacteriaceae) and a reduced
ratio between Bacteroidetes and Firmicutes [55]. Bifido-
bacteriaceae (Firmicutes) was known to exert a protective
role during liver injury [56, 57], whereas Proteobacteria was
considered the main pathogen bacteria, expressing endotox-
ins [58]. Thus, the outcome suggested that dietary habits,
by increasing the percentage of intestinal Gram-negative
Table 1 Human studies that have evaluated the gut microbiota in nonalcoholic steatohepatitis
, increase; , decrease; 16S rRNA, 16S ribosomal RNA sequencing; NASH, nonalcoholic steatohepatitis; qRT-PCR, quantitative real-time
polymerase chain reaction; SS, simple steatosis
References Study Sample Method Results and p value
Boursier etal. [49] Cross-sectional Adult NASH (n=35), obese no-
NASH (n=22)
16S rRNA Bacteroides and Prevotella
in NASH vs. obese no-NASH
(p=0.013 e p=0.053, respec-
tively)
Del Chierico etal. [69] Cross-sectional Children NASH (n=26), healthy
controls (n=54)
16S rRNA Ruminococcus, Blautia, Dorea
Oscillospira in NASH vs. healthy
controls (p<0.05)
Zhu etal. [18] Cross-sectional Children NASH (n=22), obesity
(n=25) and healthy controls
(n=16)
16S rRNA Bacteroidetes and Firmicutes
in NASH vs. healthy controls
(p=0.009 e p=0.002, respec-
tively)
Proteobacteria, Enterobacteriaceae
and Escherichia in NASH vs.
healthy controls and obesity
(p=0.0007)
Wong etal. [17] Longitudinal (6months) Adult NASH (n=16) and healthy
control (n=22)
16S rRNA Faecalibacterium and Anaero‑
sporobacter in NASH vs. healthy
controls (p<0.05)
Parabacteroides and Allisonella
in NASH vs. healthy control
(p<0.05)
Firmicutes in NASH vs. healthy
controls (p<0.05)
Mouzaki etal. [16] Cross-sectional Adult NASH (n=22), SS (n=11)
and healthy controls (n=17)
qRT-PCR Bacteroidetes in NASH vs. SS and
healthy controls (p=0.006)
Shanab etal. [60] Cross-sectional Adult NASH (n=18) and healthy
controls (n=16)
Lactulose
breath
hydrogen
test
SIBO in NASH (77.78%) vs. SIBO
in healthy controls (31.25%)
(p<0.0001)
Wigg etal. [14] Cross-sectional Adult NASH (n=22) and healthy
controls (n=23)
Combined
xylose and
lactulose
breath test
SIBO in NASH (50%) vs. SIBO in
healthy controls (22%) (p=0.048)
Author's personal copy
Eur J Nutr
1 3
endotoxin producers, might accelerate liver fibrogenesis,
introducing dysbiosis as a cofactor that contributed to
chronic liver injury in NASH [55].
Human data
The first report, on humans, of the relationship between
gut microbiota and pathogenesis of NASH was published
by Drenick etal. [59]. In this study, patients undergoing
intestinal bypass developed parallel NASH and SIBO. After
being treated with antibiotics, the patients showed regres-
sion of hepatic steatosis, suggesting that microbiota were the
possible cause of NASH [59]. Subsequently, other studies
investigated gut microbiota in patients with NASH (Table1).
Wigg etal. [14] observed small intestinal bacterial over-
growth (SIBO) in 50% of the patients with NASH and in
22% of the control subjects (p=0.048). Shanab etal. [60]
also observed a higher prevalence of SIBO in the NASH
group, compared to the control group (77 vs. 31%).
SIBO may be characterized by an increase in the num-
ber of bacteria in the proximal small intestine (≥105 col-
ony-forming units/mL of intestinal content) or a change in
microbial composition, with a profile that is typical of the
microorganism species that colonize the large intestine [61].
Although the “gold standard” for the diagnosis of SIBO is
still thought to be jejunal aspiration and culture, this tech-
nique requires intestinal intubation that may not be well tol-
erated and may not detect the un-culturable species [62]. To
investigate the possible presence of SIBO, all the studies in
patients with NASH used the breath test (Table1), because
it provides the simplest noninvasive and widely available
diagnostic modality for suspected SIBO, by determination of
hydrogen and/or methane concentration produced by intes-
tinal bacterial metabolism in the exhaled air [63]. However,
there is a lack of consensus on the breath test interpretation
[64]. Studies seeking to validate breath testing have calcu-
lated sensitivities and specificities ranging from 31 to 77
and 44 to 100%, respectively [65, 66], leading to high false-
positive rates [67]. In addition, a variety of test methods
and diagnostic criteria are used in studies and they are not
standardized to define a positive test for SIBO. These factors
have led to a controversy regarding the diagnostic utility of
breath testing in SIBO [64].
Many microbial studies have focused on the fecal micro-
biota. It is important to highlight the major drawback of the
use of stool analyses. It is the fact that a fecal sample does
not reflect the microbiota composition from the small intes-
tine, because it represents mainly fecal samples from the end
of the colon [68]. Therefore, studies with NASH patients
show changes in the composition of the fecal microbiota, but
there is controversy regarding the profile of resident bacte-
ria in the gut. For example, Mouzaki etal. [16] show a low
percentage of Bacteroidetes (Bacteroidetes to total bacteria
counts) and no differences in Firmicutes, in the stool sam-
ples of NASH patients. Instead, two studies [17, 18] have
observed an increase in Bacteroidetes and decrease in Fir-
micutes in NASH patients, compared to the healthy controls.
There is recent evidence that shows that the abundance of
Bacteroidetes and Firmicutes is similar between NASH and
no-NASH patients [49].
Other changes in the gut microbiota are related to NASH
(Table1). Recent evidence shows that the percentage of Bac
teroides genus, one of the most important genera within the
Bacteroidetes phylum, is significantly increased in NASH,
whereas the percentage of the Prevotella genus is decreased
[49]. In addition, pediatric NASH patients have a lower fecal
abundance of Faecalibacterium and Anaerosporobacter, but
higher abundance of Parabacteroides and Allisonella. A sig-
nificant difference is observed at the phylum, family, and
genera level in the fecal samples of children with NASH.
Proteobacteria/Enterobacteriaceae/Escherichia are higher in
NASH compared to healthy controls and obese patients [18].
Another study with pediatric patients shows a decrease of
Oscillospira and increases of Ruminococcus, Blautia, and
Dorea in NASH compared to the controls [69].
The variability of methods (qPCR vs. pyrosequencing),
exclusion of all taxa with an abundance below 1% and pro-
file of subjects (adults vs. children) may explain, in part, why
there is still no consensus in the literature about which bac-
terial groups are increased or reduced in the gut of NASH
patients, compared to no-NASH patients. In these studies, all
NASH patients have a high body mass index (BMI) (>29kg/
m2), and the BMI of NASH patients is significantly higher
than that of healthy controls in two studies [16, 18]. As obe-
sity itself is linked to gut microbiota composition changes
[47, 70], BMI can be a major confounder [28]. Thus far, no
study has directly assessed the gut microbiota composition
in non-obese patients with NASH, but recent evidence has
shown that non-obese patients with nonalcoholic fatty liver
disease (NAFLD) have 20% more Bacteroidetes phylum and
24% fewer Firmicutes phylum, compared to healthy con-
trols [28]. Future studies should include non-obese NASH
patients in their analyses, to exclude the impact of obesity.
Although few studies have evaluated the composition of
the gut microbiota in NASH patients, it was observed that
these individuals have a distinct gut microbiota, compared to
the healthy control groups, which explains, at least in part,
the genesis and progression of the disease through multiple
mechanisms.
Mechanistic pathways inthedevelopment
andprogression ofNASH
The mechanisms involved in the relationship between gut
microbiota and NASH are not yet fully known, but the
Author's personal copy
Eur J Nutr
1 3
proposed mechanisms in the literature are described below
(Fig.1). The proposed mechanisms may potentiate each
other through shared molecular pathways of fat accumula-
tion, activation of inflammation, and fibrogenesis in the liver
[71].
Modification ofenergy homeostasis
Energy harvest fromthediet
The gut microbiota has the ability to extract energy from
food via glycoside hydrolases and polysaccharide lyases,
which are not encoded by the human genome (Fig.1). Such
enzymes in the colon metabolize undigested polysaccharides
into monosaccharides and short chain fatty acids (SCFA)
[50]. The monosaccharides produced by microbial fer-
mentation are absorbed and transferred to the liver through
portal circulation, where they activate factors like the car-
bohydrate-responsive element-binding protein (ChREBP),
which increases the transcription of proteins involved in
hepatic lipogenesis [72]. The SCFA (acetate, propionate,
and butyrate) can be used for lipid or gluconeogenesis [73].
Thus, bacterial SCFA provide an additional source of energy
for the body, promoting fatty liver accumulation [74].
The first investigation in this line of evidence has been
conducted by Bäckhed etal. [11]. Colonization of C57BL/6
mice, germ-free (raised in absence of microorganisms), with
cecal content from mice that were colonized with a normal
microbiota at birth (termed ‘conventionally raised’) resulted
in 60% of increased total body fat and consequently hepatic
TG accumulation, without any increase in food consumption
or energy expenditure [11]. Subsequently, Turnbaugh etal.
[47] showed that the C57BL/6J obese mice had a higher con-
centration of SCFA and fewer calories in their stool, suggest-
ing that in these animals, the microbiota contribute to the
extraction of additional calories from their diet. These ani-
mals showed higher levels of Firmicutes than Bacteroidetes
compared to their lean counterparts. The changes observed
in obese mice microbiota could increase energy delivery to
the liver and reduce fecal energy loss [47].
Although the experimental data indicate that the gut
microbiota influence the energy balance, it remains uncer-
tain as to what extent gut microbiota are an important regu-
lator of nutrient absorption in humans. One clinical study
showed that the total amount of SCFA and propionate were
higher in the obese group than in the lean group [73]. How-
ever, another study found no difference in energy excre-
tion in the stools and no difference in bacterial abundance
between the obese and lean groups [10].
There is evidence that SCFA produced in the colon con-
tribute to approximately 5–10% of the energy requirements
[75]. It may be possible that the additional calories provided
to the host by the microbiota, due to the fermentation of
undigested dietary molecules, are not sufficient to induce
significant changes in weight [76]. One of the arguments to
support this hypothesis is that consumption of a high-fiber
diet could increase SCFA production, which usually helps
to reduce weight and adipose tissue [77, 78]. Studies with
Fig. 1 Mechanisms proposed
in the relationship between gut
microbiota and nonalcoholic
steatohepatitis. LPL lipoprotein
lipase, LPS lipopolysaccharide,
NASH nonalcoholic steato-
hepatitis, ROS reactive oxygen
species, TMAO trimethylamine-
N-oxide, TLR toll-like recep-
tor, VLDL very-low density
lipoprotein
Author's personal copy
Eur J Nutr
1 3
prebiotics also have indicated that a higher intestinal pro-
duction of SCFA is associated with an increase in satiety
and a consequent reduction in dietary intake. These effects
are in part related to the increase of glucagon-like peptides
(GLP-1 and GLP-2) and peptide YY (PYY), which lead to
hypothalamic effects related to the reward mechanism [79,
80]. Thus, more mechanistic studies are required to under-
stand the role of each of the SCFA on NASH.
Activation ofG protein‑coupled receptors
The SCFA act on the G protein-coupled receptors, such as
Gpr41 and Gpr43, expressed in intestine and adipose tissues
[81]. GPR41 and GPR43 have been renamed free fatty acid
receptors FFA3 and FFA2, respectively [82], based on their
responsiveness to SCFA. There is a power order of SCFA in
activating human FFA2 and FFA3 receptors, where FFA2 is
activated more potently by acetate=propionate>butyrate,
whereas for FFA3, it is propionate=butyrate>acetate [83].
The FFA3 activation stimulates enteroendocrine cells
to increase production of the PYY, a hormone that reduces
intestinal motility and provides greater absorption of nutri-
ents, particularly of SCFA [84]. The FFA2 activation con-
tributes to inhibition of lipolysis and adipocyte differen-
tiation, leading to increased adipose tissue [85]. FFA2 is
also present on intestinal neutrophils and might, therefore,
contribute to NASH pathogenesis by increasing intestinal
inflammation and permeability [86, 87].
Effects ofadenosine 5‑monophosphate protein kinase
andfasting‑induced adipose factor
Experimental studies suggest that the presence of microbiota
inhibits the enzyme adenosine 5-monophosphate protein
kinase (AMPK) pathway and suppresses intestinal expres-
sion of the protein fasting-induced adipose factor (FIAF)
[11, 50]. AMPK is a key enzyme that controls the cellular
energy status, which in turn activates the key enzymes of
mitochondrial fatty acid oxidation, including acetyl-CoA
carboxylase (ACC) and carnitine-palmitoyltransferase I
(CTP1). When inhibited, the AMPK suppresses muscle
oxidation of fatty acids, favoring adiposity [50]. FIAF is
a circulating lipoprotein lipase (Lpl) inhibitor produced by
the intestine, liver, and adipose tissue [88]. Inhibition of
FIAF increases the activity of the lipoprotein lipase, lead-
ing to fat accumulation in the adipose tissue and increases
hepatic uptake of free fatty acids [11]. Inhibition of FIAF
further decreases expression of the peroxisome prolifera-
tor-activated receptor gamma coactivator 1 alpha (PGC1-α)
and enzymes involved in mitochondrial fatty acid oxidation
[50]. Together, these effects may increase insulin resistance,
resulting in obesity and hepatic steatosis (Fig.1) [11, 50].
Lipopolysaccharide–endotoxemia
It has been proposed that changes in the gut microbiota
favor an increase in circulatory lipopolysaccharides (LPS),
particularly when the diets are rich in fat and energy [89,
90]. Increased circulatory LPS may contribute to meta-
bolic endotoxemia (low-grade inflammation) [91], which
plays a pivotal role in the development and progression of
NASH (Fig.1) [90]. LPS is recognized by pattern recogni-
tion receptors. These receptors include membranous toll-
like receptors (TLRs) (especially TLR-4) and intracellular
NOD-like receptors (NLRP3 and NLRP6 inflammasomes).
Stimulation of TLR-4 results in the activation of several dif-
ferent intracellular signaling cascades, inducing the synthe-
sis of a variety of inflammatory cytokines (especially TNF-
α), which induce inflammation, oxidative stress, and insulin
resistance [92]. Kupffer cells, which express the highest lev-
els of TLR-4 liver, are cells that respond to LPS to produce
cytokines and ROS [93]. The interaction between LPS and
TLR-4 also activates receptors on stellate cells, resulting
in hepatic fibrogenesis [15]. Increase in the levels of LPS
also leads to liver injury through a mechanism mediated by
the inflammasome, which includes NLRPs, a group of cyto-
plasmatic and multiprotein complexes [94]. NLRPs manip-
ulate the cleavage of proinflammatory interleukins (ILs),
such as pro-IL-1β and pro-IL-18 [94]. Henao-Mejia etal.
[52] experimentally revealed that the NLRP6 and NLRP3
inflammasome alterations or IL-18 deficiency cause intesti-
nal microbial changes by enhancing portal influx of TLR-4
and TLR-9 ligands, which in turn increase hepatic TNF-α
production in C57Bl/6 mice. Apart from LPS, TNF-α, ILs,
and plasminogen activator inhibitor-1 (PAI-1) may represent
a good marker of NASH. The increased portal endotoxemia
could induce the expression of PAI-1, a fibrinolysis inhibi-
tor [95]. Elevated PAI-1 has been correlated with enhanced
LPS-induced liver damage and induction of liver inflamma-
tory response [96, 97].
Several studies conducted on animals and humans support
the link between LPS–endotoxemia and the development of
NASH [54, 98100]. An animal study demonstrated that
chronic infusion of LPS caused endotoxemia, hepatic stea-
tosis, and changed gut microbiota in HFD C57bl6/J mice
[54]. Genetically obese mice exhibit increased sensitivity
to endotoxin hepatotoxicity, quickly developing steatohepa-
titis after exposure to low doses of LPS [99]. In addition,
intraperitoneal administration of LPS-augmented hepatic
inflammation, apoptosis, and reactive substances in the
methionine choline-deficient nutritional model of NASH/
C57/BL6 mice [100]. In human studies, increased levels of
endotoxin were found in NASH patients, as compared to
healthy individuals [101, 102]. Similarly, it was reported that
morbidly obese subjects have increased levels of LPS and
Author's personal copy
Eur J Nutr
1 3
LPS-binding proteins (LBP), which correlate with a major
liver expression of TNF-α and the presence of NASH [98].
Possible mechanisms for endotoxemia in patients with
NASH include SIBO and disruption of the intestinal mucosa
barrier integrity, which may lead to an increased intestinal
permeability and excessive absorption of LPS, resulting
in low-grade inflammation and hepatic fibrosis [15, 103].
This association is supported by a growing body of experi-
mental and human data. Genetically obese C57BL/6J mice
display enhanced intestinal permeability, leading to portal
endotoxemia. Moreover, murine hepatic stellate cells iso-
lated from the livers of the animals were more sensitive to
LPS, developing a stronger inflammatory and fibrogenic
phenotype [103]. In human studies, Miele etal. [15] have
shown that patients with NAFLD have increased intestinal
permeability, and this abnormality is related to the increased
prevalence of SIBO and disrupt tight junctions compared
to healthy adults. Another study has associated SIBO with
the expression of TLR-4 and IL-8 in NASH patients [60].
Wigg etal. [14] have found increased prevalence of SIBO
and elevated TNF-α levels in patients with NASH, but have
found no difference in the intestinal permeability or serum
endotoxin levels. Despite the negative result of this study,
the authors have suggested that endotoxin may still be an
important factor in the pathogenesis of NASH. Some pos-
sible explanations for the paradox in this study are under-
estimated endotoxin levels, due to a retrospective collection
of endotoxins. Endotoxins bound to plasma proteins are not
measured and systemic levels may not reflect portal endo-
toxins [14]. In addition, NASH patients may have significant
susceptibility to gut leakiness, and gut leakiness may still be
an important pathogenic factor in patients with NASH and
‘normal’ intestinal permeability [104].
Increased endogenous ethanol production
The hypothesis that endogenous ethanol contributes to the
pathogenesis of NASH dates back from the Cope etal. study
[105]. The authors have reported elevated alcohol concentra-
tion in the breath of obese mice and have demonstrated that
breath alcohol concentration can be reduced by gut micro-
bial intervention with antibiotics [105]. Human studies have
reported increased endogenous ethanol in NASH. Nair etal.
[106] have demonstrated that obese women with NASH have
higher breath ethanol concentrations than healthy controls
detected by gas chromatography. Another study has shown
that pediatric NASH patients have higher plasma concentra-
tions of ethanol when compared to healthy or obese indi-
viduals [18]. In addition, an increased expression of etha-
nol-metabolizing enzymes, alcohol dehydrogenase, catalase,
and aldehyde dehydrogenase is seen in NASH liver [107].
In summary, these outcomes suggest that the microbiota of
patients with NASH produce more ethanol, which induces
the expression of ethanol-metabolizing enzymes in the liver
[107].
The normal microbiota in the human large intestine is
capable of producing and metabolizing ethanol [108]. It has
been shown that under anaerobic conditions, the bacterial
metabolism of pyruvate, produced during the breakdown of
carbohydrates, generates acetaldehyde, which can then be
further reduced to form ethanol [109]. This metabolic fate of
carbohydrates is favored when there is intestinal overgrowth
of bacteria or yeast [110] or if carbohydrates, particularly
sugar (e.g., glucose, sucrose, and fructose), are consumed
excessively [111].
Despite the lack of consistent NASH-related gut micro-
biota changes, the possible overgrowth of ethanol-producing
bacteria may underlie an increase in the circulation of etha-
nol levels in NASH. Zhu etal. [18] have shown a higher E.
coli rate in the NASH group, compared to groups without
NASH. E. coli is a member of the Family of Enterobacte-
riaceae, which typically aerobically degrade carbohydrates
by mixed acid fermentation [112]. Ethanol is one of the
common-end products of this pathway [112]. Furthermore,
it is possible to suggest that the intestinal overgrowth of
other bacteria-producing ethanol or yeast (e.g., Lactobacil
lus fermentum, Weissella confusa, and Saccharomyces cer
evisiae) explain the higher plasma concentration of ethanol
in some NASH patients [113], mainly in those with rich-in-
carbohydrate diets [114]. L. fermentum and W. confusa are
both heterolactic organisms [115, 116]. Ethanol is one of the
dominating metabolites of heterolactic intestinal microbes
by mixed acid fermentation [115]. Finally, S. cerevisiae,
typically for yeasts, metabolizes hexoses via ethanol fer-
mentation, yielding just ethanol and carbon dioxide [117].
The intestinally derived ethanol may contribute to the
pathogenesis of NASH (Fig.1), because gut-derived ethanol
can induce hepatic steatosis [118]. In addition, increases in
the production of ethanol by gut microbiota may injure the
intestinal barrier and promote increasing permeability and
endotoxemia [105, 119, 120]. Consequently, tissues, includ-
ing the liver, that are exposed to this blood flow are stimu-
lated to produce cytokines, such as TNF-α [105] and ROS,
causing liver injury [119].
Altered bile acid metabolism
Primary bile acid (BA) species (cholic and chenode-
oxycholic acids) are synthesized and conjugated with
glycine or taurine in the liver, stored in the gallbladder,
and released into the duodenum until ingestion of a fat
meal (Fig.2) [121]. In the intestine, BA are metabolized
by bacteria to more hydrophobic BA species, through
7α-dehydroxylation and/or deconjugation of hydrophilic
groups, resulting in secondary BA species (deoxycholic
Author's personal copy
Eur J Nutr
1 3
and lithocholic acid) [122]. Over 95% of BA are reab-
sorbed in the distal ileum and then recycled via the portal
vein into the liver [123].
In addition to promoting the absorption of fat, cholesterol,
and fat-soluble vitamins in the intestinal tract, BA also act
as signaling molecules that modulate a variety of physio-
logical processes [124]. Regulatory actions of BA are medi-
ated through specific BA-activated receptors, including the
farnesoid X receptor (FXR), and members of the G protein-
coupled receptor, mainly the TGR5 [125]. FXR, which is
highly expressed in hepatocytes and enterocytes, is activated
by free and conjugated primary BA [124]. FXR induces the
expression of a short heterodimer partner (SHP), which
inhibits CYP7A1 activation, the first enzyme of BA syn-
thesis [121]. In the small intestine, FXR induces the fibro-
blast growth factor (human FGF19 and mouse FGF15), an
intestinal hormone, to repress hepatic BA synthesis through
FGF receptor 4 (FGFR4) expressed in the liver [125]. Acti-
vation of the FXR pathways not only regulates the synthesis
and enterohepatic cycle of BA, but also acts on the control
of hepatic de novo lipogenesis in the liver, exportation of
TG by VLDL, and gluconeogenesis [126]. TGR5 is widely
distributed and expressed in various tissues, including the
intestines, enteroendocrine cells, and liver [121]. Activation
of TGR5 by secondary BA induces intestinal glucagon-like
peptide-1 (GLP-1) release from the intestinal enteroendo-
crine L cells and GLP-1-associated improvements in glucose
tolerance and liver function [127]. Therefore, BA plays a
crucial role in lipid and glucose homeostasis [126].
By altering BA metabolism and its regulated signaling
pathways, gut microbiota could contribute to the patho-
genesis of NASH (Figs.1, 2) [128]. Although the precise
mechanism is unknown, altered BA concentrations have
been reported in patients with NASH [34, 129, 130]. Patients
with NASH have higher fasting and postprandial total serum
BA concentrations, including secondary BA, which tend to
be a more hydrophobic and cytotoxic species [129]. In a
similar study [130], total and secondary BA were increased
in the liver tissues of NASH patients. This increase in BA
concentration could be the consequence of a higher rate
of BA synthesis or possibly be an adaptive response to
the accumulation of TG in the liver [131]. A healthy liver
is very efficient in removing BAs from the enterohepatic
cycle. When the liver function is compromised, more BA
appears in the circulation, because the liver is not adequately
removing them [131]. Higher levels of serum 7α-hydroxy-
4-cholesten-3-one (C4), a BA synthesis intermediate and a
reliable marker of de novo BA synthesis, were also observed
in NASH patients. C4 may represent the hepatic response
to the increased fecal BA losses [35]. Indeed, higher fecal
BA levels have been demonstrated in patients with NASH.
In this study, higher levels of unconjugated primary BA in
the stool correlated with dysbiosis [35].
Dysbiosis could substantially alter BA homeostasis
[35], especially in the colon, where some bacteria, includ-
ing Bacteroides, Clostridium, and Escherichia, are able to
deconjugate and/or dehydroxylate BA, which may lead to an
increase in the circulation of unconjugated secondary BA
Fig. 2 Influence of gut micro-
biota in the development of
nonalcoholic steatohepatitis by
altering bile acid metabolism
and its regulated signaling path-
ways. BA bile acid, CYP7A1 the
first enzyme of bile acid syn-
thesis, FGF15 fibroblast growth
factor 15, FGFR4 FGF receptor
4, FXR farsenoide X receptor;
GLP‑1 glucagon-like peptide-1,
LPS lipopolysaccharide, NASH
nonalcoholic steatohepatitis,
ROS reactive oxygen species,
TNF‑α tumor necrosis factor
alpha, TRG‑5 G protein-coupled
receptor, TLR‑4 Toll-like recep-
tor 4, SHP short heterodimer
partner, VLDL very-low density
lipoprotein
Author's personal copy
Eur J Nutr
1 3
species [129]. At high levels, BA are able to activate inflam-
matory and oxidative stress, resulting in apoptosis or necro-
sis, and eventually fibrosis and cirrhosis [132]. In contrast,
the relative abundance of Clostridium leptum (C. leptum to
total bacteria counts) is decreased in patients with NASH
compared to the controls and correlates with higher cholic
and chenodeoxycholic acids in the stool [35]. Higher levels
of unconjugated primary BA in the stool are positively cor-
related with steatosis, ballooning, and fibrosis. These find-
ings may represent the hepatotoxic impact of hydrophobic
BA. BA can also contribute to the development of NASH
through its effects on intestinal permeability [35]. BA has
bactericidal activity and reduces the intestinal permeability
to endotoxin [133]. However, increased deconjugation of
BA reduces the bactericidal properties of the bile, causing
growth of bacteria that promotes more deconjugation of BA,
and ultimately, translocation and endotoxemia in homeo-
stasis conditions [134]. The interplay between BA and gut
microbiota in human NASH needs to be investigated further.
Altered choline metabolism
Choline is an essential nutrient and phospholipid component
of cell membranes required for the formation of VLDL and
exportation of liver lipids [135]. The metabolism of dietary
choline by microbiota reduces the bioavailability of choline
free to the secretion of VLDL, favoring the accumulation
of fat in the liver (Fig.1) [135, 136]. It is also known that
enzymes produced by the gut microbiota catalyze the con-
version of dietary choline into toxic methylamines, such as
trimethylamine (TMA) [1]. TMA is subsequently oxidized in
the liver, forming trimethylamine-N-oxide (TMAO), which
induces liver inflammation. Moreover, TMAO may affect
glucose and lipid metabolism, promoting the development
of fatty liver [136]. TMAO increases fasting insulin levels
and homeostasis model assessment-estimated insulin resist-
ance (HOMA-IR) and also exacerbates the impaired glucose
tolerance in HFD mice (C57BL/6). These effects are associ-
ated with the expression of genes related to the insulin signal
pathway, glycogen synthesis, gluconeogenesis, and glucose
transport in the liver [137]. The effects of TMAO on lipid
metabolism involve the overexpression of flavin contain-
ing monooxygenase (FMO3) in the human hepatoma cell
line, resulting in increased lipogenesis. These effects may
be mediated through the peroxisome proliferator-activated
receptor alpha (PPARα) and Kruppel-like factor 15 (KLF15)
pathways [138].
A few studies have examined the association of choline
and its metabolites with fatty liver disease. A choline-defi-
cient diet greatly exacerbates a fatty liver induced by HFD
consumption in C57Bl/6 mice [139]. Moreover, Pfp/Rag2
mice, submitted to a choline-deficient diet, develop a fatty
liver featuring fibrosis and elevation of the proinflammatory
markers serum amyloid A (SAA) and TNFα. Hepatic TG is
significantly increased as well as alanine aminotransferase,
demonstrating inflammation-linked hepatocyte damage
[140]. Human studies have shown that the consumption of a
low-choline diet promotes accumulation of TG in the liver
and worsens fibrosis [141]. In a recent study, Chen etal.
[136] have shown adverse associations between the circulat-
ing TMAO level and the presence and severity of NAFLD in
Chinese adults, but no significant choline–NAFLD associa-
tion has been observed. The composition of the gut micro-
biota can change with a low-choline diet. Healthy women
during choline depletion show rate variations of Erysipel
otrichi (Firmicutes) and Gammaproteobacteria (Proteobac-
teria) in their fecal contents, which are directly associated
with changes in the liver fat [142]. The Gammaproteobac
teria genera in particular, identified in their study, includ-
ing Klebsiella spp., Enterobacter spp., and Escherichia spp.,
are gram-negative bacteria with LPS-containing membranes
[142]. Therefore, the increase of circulating LPS can be one
of the possible mechanisms involved in NASH development
in choline-deficient patients.
On the basis of all these mechanistic pathways, it is
possible to suggest that modulation of the gut microbiota,
through strategies that can include diet, probiotics, antibiot-
ics, or fecal microbiota transplantation, is a possibility for
the treatment of NASH [143]. However, diet appears to be
the simplest, most physiological, and most effective method
to improve intestinal health [144].
Diet andgut microbiota
The diet is among the most easily controlled factors that
can potentially manipulate the gut microbiota [145]. As dis-
cussed above, a high-energy diet, HFD, high-carbohydrate
diet (mainly high-fructose diet), and decreased choline
intake can alter the gut microbiota, which has been shown
to be associated with NASH. Correcting dietary habits is
typically part of the standard recommendations for NASH
treatment [146]. However, we have not found studies in the
literature that have investigated the effects of dietary habit
modifications on the gut microbiota of NASH patients, with-
out the use of probiotics and/or prebiotics. There is some
evidence in animal models and no-NASH subjects that sug-
gests that the amount of dietary calories and the balance
between the three dietary macronutrients (fats, carbohy-
drates, and proteins) have the potential to improve the gut
microbiota [147].
The impact of calorie restriction on gut microbiota has
been demonstrated in C57BL/6J mice [148]. Calorie restric-
tion enriches phylotypes correlated with probiotic effects,
such as Lactobacillus and Bifidobacterium, and reduces
phylotypes correlated with inflammation and obesity, such
Author's personal copy
Eur J Nutr
1 3
as Streptococcaceae and Desulfovibrionaceae. These calo-
rie restriction-induced changes in the gut microbiota are
concomitant with significantly reduced serum levels of the
LPS-binding protein, suggesting that animals under calorie
restriction can establish a structurally balanced architecture
of the gut microbiota that may exert a health benefit to the
host via reduction of antigen load from the gut. In obese sub-
jects, the reduction in food energy content decreases the phy-
lum Firmicutes and increases Bacteroidetes [10, 149, 150].
Moreover, these calorie-restricted diets increase microbial
gene richness in subjects with obesity [151] and normalize
the circulating LPS levels [152].
The effects of dietary macronutrients on the human
microbiota are still poorly understood. A number of studies
focus on the impact of a “Western” diet (high in animal fat
and protein and low in fiber), compared to a “non-Western”
diet (low in animal fat and protein and high in fiber) [43,
153, 154]. Western and non-Western human diets are con-
sistently associated with distinct gut microbial communities
[43, 153, 154]. Amato etal. [154] observed elevated micro-
bial richness and a relatively higher abundance of Prevotella
in non-Western humans and a relatively elevated abundance
of Bacteroides in Western humans. These results are con-
cordant with a study of the human gut microbiota that asso-
ciates diets high in protein and animal fat with high levels
of Bacteroides and diets high in plant carbohydrates with
high levels of Prevotella [155, 156]. Similarly, all published
studies of Western and non-Western humans to date report
higher microbial richness and a higher abundance of Prevo
tella in non-Western populations [43, 153, 154]. One study
also shows a higher abundance of Xylanibacter [156]. The
genera Prevotella and Xylanibacter are known to contain
a set of bacterial genes for cellulose and xylan hydrolysis,
completely lacking in the non-Western population. In addi-
tion, Enterobacteriaceae (Shigella and Escherichia) were
significantly under-represented in non-Western populations
compared to Western populations [156]. Moreover, a clini-
cal trial has shown that healthy subjects with a prudent-style
diet (20% of fat) for 1month reduced plasma LPS levels by
38%, whereas a Western-style diet induced a 71% increase
in plasma levels of endotoxin [157].
The composition of gut microbiota could also be influ-
enced by the quality of dietary lipids. One experiment
evaluated the effect of a fat-type diet, varying in polyun-
saturated-to-saturated fatty acid ratios in the gut microbiota
composition and hepatic TG accumulation [158]. C57Bl/6J
mice were fed purified HFDs (45E% fat) containing palm
oil (saturated lipids), olive oil (monounsaturated lipids), or
safflower oil (polyunsaturated lipids) for 8weeks. According
to the authors, HFD containing palm oil induced a higher
liver TG content, reduced microbial diversity, and increased
the Firmicutes:Bacteroidetes ratio, whereas HFDs contain-
ing olive oil or safflower did not change the gut microbiota
[157]. Similarly, Caesar etal. [159] fed mice isocaloric
diets that differed only in fat composition (either lard or fish
oil, which are rich in saturated and polyunsaturated lipids,
respectively) to assess how the dietary fat sources affected
the microbiota. This study showed that the genera Bacte
roides, Turicibacter, and Bilophila had increased in lard-fed
mice, while Actinobacteria (Bifidobacterium and Adlercreut
zia), lactic acid bacteria (Lactobacillus and Streptococcus),
Verrucomicrobia (Akkermansia muciniphila), Alphaproteo
bacteria, and Deltaproteobacteria had increased in fish-oil-
fed mice. Moreover, TLR-4 was activated by serum from
mice fed with lard, suggesting that a lard diet promotes an
increase in the influx of microbial factors into the systemic
circulation [159].
Regarding carbohydrate intake, there is evidence that
low-carbohydrate diets can impair the gut microbiota, since
they are also restricted in source foods, such as fruits, veg-
etables, and grains, which are rich in fiber (non-digestible
carbohydrates) [160, 161]. In general, the consumption of
fiber-rich diets is associated with greater richness and diver-
sity of the gut microbiota, being positively associated with
the presence of Bacteroidetes and Actinobacteria and with
reduction in the Firmicutes:Bacteroides ratio [150]. Fiber-
rich diets also promote a higher concentration of SCFA in
the fecal contents, especially of butyrate, which has a benefi-
cial effect on inflammation [162]. Thus, the replacement of
fructose, and other simple carbohydrates, with non-digesti-
ble carbohydrates can avoid the consequences of dysbiosis
in the gut–liver axis, especially in NASH.
Finally, the relationship between protein intake and gut
microbiota, specifically in NASH patients, is lacking. The
excess protein has been linked with potentially damaging
effects on the gut microbiota and health. Hoodia etal. [163]
have verified that a high-protein diet reducts Faecalibac
terium prausnitzii and increases colon permeability and
secretion of cytokines. Other evidence has reported high
levels of Clostridium spp. and Bacteroides spp., with con-
current reductions in Bifidobacterium spp., Roseburia spp.,
and Eubacterium spp., in subjects who consumed a high-
protein diet [161, 164, 165]. Reductions in Bifidobacterium
spp., Roseburia spp., and Eubacterium may increase the risk
of NASH, as these bacterial species are usually associated
with butyrate production and control of endotoxemia [166].
It is important to mention that, generally, the highest pro-
portion of dietary protein is accompanied by a reduction in
the amount of carbohydrates. Therefore, it is possible that
the impact of the consumption of high-protein diets on the
gut microbiota is related not only to the production of toxic
substances derived from protein fermentation, but also to the
reduction of dietary carbohydrate consumption, especially of
non-digestible carbohydrates [162]. Besides that, the source
of protein varies between studies. For example, populations
that consume meat-rich diets have higher fecal Bacteroides,
Author's personal copy
Eur J Nutr
1 3
Bifidobacterium, Peptococcus, and anaerobic Lactobacillus
species [167]. A vegetarian diet is associated with higher
rates of Bacteroides thetaiotaomicron, Clostridium clostridi
oforme, and F. prausnitzii compared to an omnivorous diet
[168].
Although the studies use different methods and varying
manipulations of diets, which make comparability and gen-
eralization of the outcomes difficult, it seems clear that the
various diets are important environmental factors that regu-
late and modify the gut microbiota.
Conclusions
The development and progression of NASH is a complex
and multifactorial process, which cannot be completely
explained by the “two hits” hypothesis. It is increasingly
recognized that the gut microbiota is implicated in the
pathogenesis and progression of NASH. There are evi-
dences suggesting that NASH patients have a higher prev-
alence of bacterial overgrowth of the small intestine and
changes in the composition of gut microbiota, but there
is controversy regarding the profile of resident bacteria in
the gut. An abundance of the Bacteroidetes phylum may
be increased, decreased, or unaltered in NASH patients.
There is an increase in the Enterobacteriaceae family (phy-
lum Proteobacteria), especially Escherichia. Moreover, the
Bacteroides genus (phylum Bacteroidetes) is also increased.
There is depletion of certain taxa, such as Prevotella, Fecali
bacterium, Anaerosporobacter, Oscillospira, Ruminococcus,
Blautia, and Dorea. Although few studies have evaluated the
gut microbiota in NASH patients, it was observed that these
subjects have a distinct gut microbiota compared to the con-
trol groups, which explains, at least in part, the genesis and
progression of the disease through multiple mechanisms.
Changes in the gut microbiota have consequences on
energy homeostasis, resulting in hepatic steatosis. Dysbiosis
is also responsible for increased intestinal permeability and
metabolic endotoxemia, which correlate with inflammation
and liver fibrosis. In addition, it is observed that the metabo-
lism of other related pathways is affected by the gut micro-
biome in NASH, such as choline and bile acid metabolism
and the endogenous production of ethanol. The role of LPS
and bile acids in NASH pathogenesis has been discussed
in several studies and they appear to be the most relevant
factors in humans.
It is essential to identify strategies to modulate the gut
microbiota and probably minimize the development and pro-
gression of NASH. Modulation of gut microbiota by diet
control offers new challenges for future studies.
Acknowledgements The authors gratefully acknowledge the com-
bined support of the Brazilian government organizations (Comissão de
Aperfeiçoamento de Pessoal do Nível Superior-CAPES and Fundação
de Amparo à Pesquisa do Estado de Minas Gerais-FAPEMIG).
Author contributions The author contributions were as follows:
FFG and DGO designed the concept of the study, and all authors were
involved in the literature search and review. FFG and DGO wrote the
manuscript. APBM, JMO, DEC and LEVVCF were involved with
editing the manuscript, and all authors read and approved the final
manuscript.
Compliance with ethical standards
Conflict of interest The authors declare that they have no conflict
of interest.
References
1. Liu W, Baker RD, Bhatia T etal (2016) Pathogenesis of non-
alcoholic steatohepatitis. Cell Mol Life Sci 73(10):1969–1987.
doi:10.1007/s00018-016-2161-x
2. Tijera FH, Servín-Caamaño AI (2015) Pathophysiological
mechanisms involved in nonalcoholic steatohepatitis and novel
potential therapeutic targets. World J Hepatol 7(10):1297–1301.
doi:10.4254/wjh.v7.i10.1297
3. Browning JD, Szczepaniak LS, Dobbins R etal (2004) Preva-
lence of hepatic steatosis in an urban population in the United
States: impact of ethnicity. Hepatology 40(6):1387–1395
4. Wanless IR, Lentz JS (1990) Fatty liver hepatitis (steatohepati-
tis) and obesity: an autopsy study with analysis of risk factors.
Hepatology 12(5):1106–1110
5. Losekann A, Weston AC, Mattos AA etal (2015) Nonalcoholic
steatohepatitis (NASH): risk factors in morbidly obese patients.
Int J Mol Sci 16(10):25552–25559
6. Ekstedt M, Franzén LE, Mathiesen UL etal (2006) Long-term
follow-up of patients with NAFLD and elevated liver enzymes.
Hepatology 44(4):865–873
7. Bacon BR, Farahvash MJ, Janney CG etal (1994) Nonalcoholic
steatohepatitis: an expanded clinical entity. Gastroenterology
107(4):1103–1109
8. White DL, Kanwal F, El-Serag HB (2012) Association between
nonalcoholic fatty liver disease and risk for hepatocellular can-
cer, based on systematic review. Clin Gastroenterol Hepatol
10(12):1342–1359
9. Orman ES, Barritt AS, Wheeler SB etal (2013) Declining liver
utilization for transplantation in the United States and the impact
of donation after cardiac death. Liver Transpl 19(1):59–68
10. Jumpertz R, Le DS, Turnbaugh PJ etal (2011) Energy-balance
studies reveal associations between gut microbes, caloric load,
and nutrient absorption in humans. Am J Clin Nutr 94(1):58–65
11. Backhed F, Ding H, Wang T etal (2004) The gut microbiota as
an environmental factor that regulates fat storage. Proc Natl Acad
Sci 101(4):15718–15723
12. Csak T, Ganz M, Pespisa J etal (2011) Fatty acid and endotoxin
activate inflammasomes in mouse hepatocytes that release danger
signals to stimulate immune cells. Hepatology 54(1):133–144
13. Miura K, Kodama Y, Inokuchi S etal (2010) Toll-like receptor
9 promotes steatohepatitis by induction of interleukin-1beta in
mice. Gastroenterology 139(1):323–334
14. Wigg AJ, Roberts-Thomson IC, Dymock RB etal (2001) The role
of small intestinal bacterial overgrowth, intestinal permeability,
endotoxaemia, and tumour necrosis factor á in the pathogenesis
of nonalcoholic steatohepatitis. Gut 48(2):206–211
Author's personal copy
Eur J Nutr
1 3
15. Miele L, Valenza V, La Torre G etal (2009) Increased intestinal
permeability and tight junction alterations in nonalcoholic fatty
liver disease. Hepatology 49(6):1877–1887
16. Mouzaki M, Comelli EM, Arendt BM (2013) Intestinal micro-
biota in patients with nonalcoholic fatty liver disease. Hepatol-
ogy 58(1):120–127
17. Wong VW-S, Tse C-H, Lam TT-Y etal (2013) Molecular char-
acterization of the fecal microbiota in patients with nonalcoholic
steatohepatitis—a longitudinal study. PLoS One 8(4):e62885.
doi:10.1371/journal.pone.0062885
18. Zhu L, Baker SS, Gill C etal (2013) Characterization of gut
microbiomes in nonalcoholic steatohepatitis (NASH) patients: a
connection between endogenous alcohol and NASH. Hepatology
57(2):601–609
19. Preidis GA, Versalovic J (2009) Targeting the human micro-
biome with antibiotics, probiotics, and prebiotics: gastro-
enterology enters the metagenomics era. Gastroenterology
136(6):2015–2031
20. Day CP, James OF (1998) Steatohepatitis: a tale of two “hits”?
Gastroenterology 114(4):842–845
21. Donnelly KL, Smith CI, Schwarzenberg SJ etal (2005) Sources
of fatty acids stored in liver and secreted via lipoproteins in
patients with nonalcoholic fatty liver disease. J Clin Invest
115(5):1343–1351
22. Mittendorfer B, Yoshino M, Patterson BW etal (2016) VLDL
triglyceride kinetics in lean, overweight, and obese men and
women. J Clin Endocrinol Metab 101:4151–4160
23. Pessayre D, Berson A, Fromenty B etal (2001) Mitochondria in
steatohepatitis. Semin Liver Dis 21:57–69
24. Leclerq IA, Farrell GC, Fiels J etal (2000) CYP2EI and CYP4A
as microsomal catalysts of lipid peroxides in murine nonalco-
holic steatohepatitis. J Clin Investig 105:1067–1075
25. Parola M, Pinzani M, Casini A etal (1993) Stimulation of lipid
peroxidation or 4-hydroxynonenal treatment increases procol-
lagen alpha 1(I) gene expression in human liver fat-storing cells.
Biochim Biophys Res Commun 194:1044–1050
26. Jaeschke H, Wang Y, Essani NA (1996) Reactive oxygen species
activate the transcription factor NF-kB in the liver by induction
of lipid peroxidation (abstr). Hepatology 24:238A
27. Lee KS, Buck M, Houglum K, Chojkier M (1995) Activation of
hepatic stellate cells by TGF alpha and collagen type I is medi-
ated by oxidative stress through c-myb expression. J Clin Investig
96:2461–2468
28. Wang B, Jiang X, Cao M etal (2016) Altered fecal microbiota
correlates with liver biochemistry in nonobese patients with
nonalcoholic fatty liver disease. Sci Rep 6:32002. doi:10.1038/
srep32002
29. Takaki A, Kawai D, Yamamoto K (2013) Multiple hits, including
oxidative stress, as pathogenesis and treatment target in nonal-
coholic steatohepatitis (NASH). Int J Mol Sci 14:20704–20728.
doi:10.3390/ijms141020704
30. Yamaguchi K, Yang L, McCall S etal (2007) Inhibiting triglyc-
eride synthesis improves hepatic steatosis but exacerbates liver
damage and fibrosis in obese mice with nonalcoholic steatohepa-
titis. Hepatology 45(6):1366–1374
31. Tilg H, Moschen AR (2010) Evolution of Inflammation in non-
alcoholic fatty liver disease: the multiple parallel hits hypothesis.
Hepatology 52(5):1836–1846
32. Feldstein AE, Werneburg NW, Canbay A etal (2004) Free fatty
acids promote hepatic lipotoxicity by stimulating TNF-alpha
expression via a lysosomal pathway. Hepatology 40:185–194
33. Mari M, Caballero F, Colell A etal (2006) Mitochondrial free
cholesterol loading sensitizes to TNF- and Fas-mediated steato-
hepatitis. Cell Metab 4:185–198
34. Buzzetti E, Pinzani M, Tsochatzis EA (2016) The mul-
tiple-hit pathogenesis of nonalcoholic fatty liver disease
(NAFLD). Metabolism 65(8):1038–1048. doi:10.1016/j.
metabol.2015.12.012
35. Mouzaki M, Wang AY, Bandsma R etal (2016) Bile acids
and dysbiosis in nonalcoholic fatty liver disease. PLoS One
11(5):e0151829. doi:10.1371/journal.pone.0151829
36. Xue L, He J, Gao N etal (2017) Probiotics may delay the pro-
gression of nonalcoholic fatty liver disease by restoring the gut
microbiota structure and improving intestinal endotoxemia. Sci
Rep 7:45176. doi:10.1038/srep45176
37. Erejuwa OO, Sulaiman SA, Wahab MAS (2014) Modulation
of gut microbiota in the management of metabolic disorders:
the prospects and challenges. Int J Mol Sci 15(3):4158–4188
38. Sender R, Fuchs S, Milo R (2016) Are we really vastly outnum-
bered? Revisiting the ratio of bacterial to host cells in humans.
Cell 164:337–340
39. Rajilic-Stojanovic M (2013) Function of the microbiota. Best
Pract Res Clin Gastroenterol 27:5–16
40. Zhernakova A, Kurilshikov A, Bonder MJ etal (2016)
Population-based metagenomics analysis reveals mark-
ers for gut microbiome composition and diversity. Science
352(6285):565–569. doi:10.1126/science.aad3369
41. Lepage P, Leclerc MC, Joossens M etal (2013) A metagenomic
insight into our gut’s microbiome. Gut 62:146–158
42. Costello EK, Lauber CL, Hamady M etal (2009) Bacterial
community variation in human body habitats across space
and time. Science 326(5960):1694–1697. doi:10.1126/
science.1177486
43. Scheithauer TPM, Dallinga-Thie GM, De Vos WM etal (2016)
Causality of small and large intestinal microbiota in weight
regulation and insulin resistance. Mol Metab 5:759–770
44. Yatsunenko T, Rey FE, Manary MJ etal (2012) Human
gut microbiome viewed across age and geography. Nature
486(7402):222–227. doi:10.1038/nature11053
45. Turnbaugh PJ, Hamady M, Yatsunenko T et al (2009)
A core gut microbiome in obese and lean twins. Nature
457(7228):480–484. doi:10.1038/nature07540
46. Nadal I, Santacruz A, Marcos A etal (2009) Shifts in clostridia,
bacteroides and immunoglobulin-coating fecal bacteria associ-
ated with weight loss in obese adolescents. Int J Obes (Lond)
33:758–767
47. Turnbaugh PJ, Ley RE, Mahowald MA etal (2006) An obesity-
associated gut microbiome with increased capacity for energy
harvest. Nature 444(7122):1027–1031
48. Cantarel BL, Lombard V, Henrissat B (2012) Complex carbo-
hydrate utilization by the healthy human microbiome. PLoS
One 7:e2874. doi:10.1371/journal.pone.0028742
49. Boursier J, Mueller O, Barret M etal (2016) The severity of
nonalcoholic fatty liver disease is associated with gut dysbio-
sis and shift in the metabolic function of the gut microbiota.
Hepatology 63(3):764–775
50. Bäckhed F, Manchester JK, Semenkovich CF etal (2007)
Mechanisms underlying the resistance to diet-induced obesity
in germ-free mice. PNAS 104(3):979–984
51. Le Roy T, Llopis M, Lepage P etal (2013) Intestinal microbi-
ota determines development of nonalcoholic fatty liver disease
in mice. Gut 62:1787–1794
52. Henao-Mejia J, Elinav E, Jin C etal (2012) Inflammasome-
mediated dysbiosis regulates progression of NAFLD and obe-
sity. Nature 482(7384):179–185
53. Xie G, Wang X, Liu P etal (2016) Distinctly altered gut
microbiota in the progression of liver disease. Oncotarget
7(15):19355–19366. doi:10.18632/oncotarget.8466
54. Cani PD, Amar J, Iglesias MA etal (2007) Metabolic endo-
toxemia initiates obesity and insulin resistance. Diabetes
56(7):1761–1772
Author's personal copy
Eur J Nutr
1 3
55. De Minicis S, Rychlicki C, Agostinelli L etal (2014) Dysbiosis
contributes to fibrogenesis in the course of chronic liver injury
in mice. Hepatology 59:1738–1749
56. Osman N, Adawi D, Ahrne S etal (2007) Endotoxin- and
d-galactosamine-induced liver injury improved by the admin-
istration of Lactobacillus, Bifidobacterium and blueberry. Dig
Liver Dis 39:849–856
57. Xing HC, Li LJ, Xu KJ etal (2006) Protective role of supple-
ment with foreign Bifidobacterium and Lactobacillus in experi-
mental hepatic ischemia–reperfusion injury. J Gastroenterol
Hepatol 21:647–656
58. Ueyama J, Nadai M, Kanazawa H etal (2005) Endotoxin from
various gram-negative bacteria has differential effects on func-
tion of hepatic cytochrome P450 and drug transporters. Eur J
Pharmacol 510:127–134
59. Drenick EJ, Fisler J, Johnson D (1982) Hepatic steatosis after
intestinal by-pass. Prevention and reversal by metronidazole
irrespective of protein-calorie malnutricion. Gastroenterology
82(3):535–548
60. Shanab AA, Scully P, Crosbie O etal (2011) Small intestinal
bacterial overgrowth in nonalcoholic steatohepatitis: associa-
tion with toll-like receptor 4 expression and plasma levels of
interleukin 8. Dig Dis Sci 56(5):1524–1534
61. Bures J, Cyrany J, Kohoutova D etal (2010) Small intesti-
nal bacterial overgrowth syndrome. World J Gastroenterol
16:2978–2990
62. Corazza GR, Menozzi MG, Strocchi A etal (1990) The diagno-
sis of small bowel bacterial overgrowth. Reliability of jejunal
culture and inadequacy of breath hydrogen testing. Gastroen-
terology 98(2):302–309
63. Saad RJ, Chey WD (2014) Breath testing for small intestinal
bacterial overgrowth: maximizing test accuracy. Clin Gastro-
enterol Hepatol 12:1964–1972
64. Krajicek EJ, Hansel SL (2016) Small intestinal bacte-
rial overgrowth: a primary care review. Mayo Clin Proc
91(12):1828–1833
65. Khoshini R, Dai SC, Lezcano S (2008) A systematic review of
diagnostic tests for small intestinal bacterial overgrowth. Dig
Dis Sci 53(6):1443–1454
66. Erdogan A, Lee YY, Badger C etal (2014) What is the opti-
mal threshold for an increase in hydrogen and methane levels
with glucose breath test (GBT) for detection of small intestinal
bacterial overgrowth (SIBO)? Gastroenterology 146(5):S532
67. Lin EC, Massey BT (2016) Scintigraphy demonstrates high
rate of false-positive results from glucose breath tests for
small bowel bacterial overgrowth. Clin Gastroenterol Hepatol
14(2):203–208
68. Zoetendal EG, Raes J, Van Den Bogert B etal (2012) The
human small intestinal microbiota is driven by rapid uptake
and conversion of simple carbohydrates. ISME J 6:1415–1426.
doi:10.1038/ismej.2011.212
69. Del Chierico F, Nobili V, Vernocchi P etal (2016) Gut micro-
biota profiling of pediatric nonalcoholic fatty liver disease and
obese patients unveiled by na integrated meta-omics-based
approach. Hepatology. doi:10.1002/hep.28572
70. Ley RE, Turnbaugh PJ, Klein S, Gordon JI etal (2006) Micro-
bial ecology: human gut microbes associated with obesity.
Nature 444(7122):1022–1023
71. Wieland A, Frank DN, Harnke B etal (2015) Systematic
review: microbial dysbiosis and nonalcoholic fatty liver dis-
ease. Aliment Pharmacol Ther 42:1051–1063
72. Benhamed F, Denechaud FD, Lemoine M etal (2012) The lipo-
genic transcription factor ChREBP dissociates hepatic steatosis
from insulin resistance in mice and humans. J Clin Investig
122(6):2176–2194. doi:10.1172/JCI41636
73. Wolever TM, Brighenti F, Royall D etal (1989) Effect of rectal
infusion of short chain fatty acids in human subjects. Am J Gas-
troenterol 84:1027–1033
74. Schwiertz A, Taras D, Schäfer K etal (2009) Microbiota and
SCFA in lean and overweight healthy subjects. Obesity 18:190–
195. doi:10.1038/oby.2009.167
75. McNeil NI (1984) The contribution of the large intestine to
energy supplies in man. Am J Clin Nutr 39:338–342
76. Moreira APB, Teixeira TFS, Ferreira AB etal (2012) Influence
of a high-fat diet on gut microbiota, intestinal permeability and
metabolic endotoxaemia. Br J Nutr 108(5):801–809
77. Lu Y, Fan C, Li P etal (2016) Short chain fatty acids prevent
high-fat-diet-induced obesity in mice by regulating G pro-
tein coupled receptors and gut microbiota. Sci Rep 6:37589.
doi:10.1038/srep37589
78. Anastasovska J, Arora T, Canon GJs etal (2012) Fermentable
carbohydrate alters hypothalamic neuronal activity and protects
against the obesogenic environment. Obesity (Silver Spring)
20(5):1016–1023
79. Cani PD, Possemiers S, Van de Wiele T etal (2009) Changes
in gut microbiota control inflammation in obese mice through a
mechanism involving GLP-2-driven improvement of gut perme-
ability. Gut 58(8):1091–1103
80. Delzenne NM, Cani PD, Neyrinck AM (2007) Modula-
tion of glucagonlike peptide 1 and energy metabolism by
inulin and oligofructose:experimental data. J Nutr 137(11
Suppl):2547S–2551S
81. Le Poul E, Loison C, Struyf S etal (2003) Functional charac-
terization of human receptors for short chain fatty acids and
their role in polymorphonuclear cell activation. J Biol Chem
278:25481–25489
82. Stoddart LA, Smith NJ, Milligan G (2008) International union
of pharmacology. LXXI. Free fatty acid receptors FFA1, -2, and
-3: pharmacology and pathophysiological functions. Pharmacol
Rev 60:405–417
83. Milligan G, Stoddart LA, Smith NJ (2009) Agonism and alloster-
ism: the pharmacology of the free fatty acid receptors FFA2 and
FFA3. Br J Pharmacol 158:146–153
84. Samuel BS, Shaito A, Motoike T etal (2008) Effects of the gut
microbiota on host adiposity are modulated by the short-chain
fatty-acid binding G protein-coupled receptor, Gpr41. PNAS
5(430):16767–16772
85. Ge H, Li X, Weiszmann J etal (2008) Activation of G protein-
coupled receptor 43 in adipocytes leads to inhibition of lipoly-
sis and suppression of plasma free fatty acids. Endocrinology
149(9):4519–4526
86. Dewulf EM, Ge Q, Bindels LB etal (2013) Evaluation of the
relationship between GPR43 and adiposity in human. Nutr Metab
10(1):11. doi:10.1186/1743-7075-10-11
87. Ulven T (2012) Short-chain free fatty acid receptors FFA2/
GPR43 and FFA3/GPR41 as new potential therapeutic tar-
gets. Front Endocrinol (Lausanne) 3:111. doi:10.3389/
fendo.2012.00111
88. Yoon JC, Chickering TW, Rosen ED etal (2000) Peroxisome
proliferator-activated receptor gamma target gene encoding
a novel angiopoietin-related protein associated with adipose
differentiation. Mol Cell Biol 20:5343–5349. doi:10.1128/
MCB.20.14.5343-5349.2000
89. Erridge C, Attina T, Spickett CM etal (2007) A high-fat meal
induces low-grade endotoxemia: evidence of a novel mechanism
of postprandial inflammation. Am J Clin Nutr 86(5):1286–1292
90. Cani PD, Bibiloni R, Knauf C etal (2008) Changes in gut micro-
biota control metabolic endotoxemia-induced inflammation in
high-fat diet-induced obesity and diabetes in mice. Diabetes
57:1470–1481
Author's personal copy
Eur J Nutr
1 3
91. Manco M, Putignani L, Bottazzo GF (2010) Gut microbiota,
lipopolysaccharides, and innate immunity in the pathogenesis
of obesity and cardiovascular risk. Endocr Rev 31(6):817–844
92. Ye D, Li FY, Lam KS etal (2012) Toll-like receptor-4 mediates
obesity-induced nonalcoholic steatohepatitis through activation
of X-box binding protein-1 in mice. Gut 61(7):1058–1067
93. Su GL (2002) Lipopolysaccharides in liver injury: molecular
mechanisms of Kupffer cell activation. Am J Physiol Gastro-
intest Liver Physiol 283:G256–G265
94. Szabo G, Csak T (2012) Inflammasomes in liver diseases. J
Hepatol 57(3):642–654. doi:10.1016/j.jhep.2012.03.035
95. Alisi A, Manco M, Devito R etal (2010) Endotoxin and plas-
minogen activator inhibitor-1 serum levels associated with
nonalcoholic steatohepatitis in children. JPGN 50:645–649
96. Luyendyk JP, Maddox JF, Green CD etal (2004) Role of
hepatic fibrin in idiosyncrasy-like liver injury from lipopol-
ysaccharide-ranitidine coexposure in rats. Hepatology
40:1342–1351
97. Targher G, Bertolini L, Scala L etal (2007) Plasma PAI-1 levels
are increased in patients with nonalcoholic steatohepatitis. Dia-
betes Care 30:e31–e32. doi:10.2337/dc07-0109
98. Ruiz AG, Casafont F, Crespo J etal (2007) Lipopolysaccharide-
binding protein plasma levels in liver TNF-alpha gene expression
in obese patients: evidence for the potential role of endotoxin
in the pathogenesis of nonalcoholic steatohepatitis. Obes Surg
17(10):1374–1380
99. Yang SQ, Lin HZ, Lane MD etal (1997) Obesity increases sensi-
tivity to endotoxin liver injury: implications for the pathogenesis
of steatohepatitis. Proc Natl Acad Sci USA 94:2557–2562
100. Kirsch R, Clarkson V, Verdonk RC etal (2006) Rodent nutri-
tional model of steatohepatitis: effects of endotoxin (lipopolysac-
charide) and tumor necrosis factor alpha deficiency. J Gastroen-
terol Hepatol 21(1 Pt 1):174–182
101. Thuy S, Ladurner R, Volynets V etal (2008) Nonalcoholic fatty
liver disease in humans is associated with increased plasma endo-
toxin and plasminogen activator inhibitor 1 concentrations and
with fructose intake. J Nutr 138(8):1452–1455
102. Harte AL, da Silva NF, Creely SJ etal (2010) Elevated endo-
toxin levels in nonalcoholic fatty liver disease. J Inflamm 7:15.
doi:10.1186/1476-9255-7-15
103. Brun P, Castagliuolo I, Di Leo V etal (2007) Increased intestinal
permeability in obese mice: new evidence in the pathogenesis
of nonalcoholic steatohepatitis. Am J Physiol Gastrointest Liver
Physiol 292:G518–G525
104. Farhadi A, Gundlapalli S, Shaikh M etal (2008) Susceptibil-
ity to gut leakiness: a possible mechanism for endotoxaemia in
nonalcoholic steatohepatitis. Liver Int 28:1026–1033
105. Cope K, Risby T, Diehl AM (2000) Increased gastrointestinal
ethanol production in obese mice: implications for fatty liver
disease pathogenesis. Gastroenterology 119:1340–1347
106. Nair S, Cope K, Terence RH etal (2001) Obesity and female
gender increase breath ethanol concentration: potential implica-
tions for the pathogenesis of nonalcoholic steatohepatitis. Am J
Gastroenterol 96(4):1200–1204
107. Baker SS, Baker RD, Liu W etal (2010) Role of alcohol metab-
olism in nonalcoholic steatohepatitis. PLoS One 5(3):e9570.
doi:10.1371/journal.pone.0009570
108. Nosova T, Jokelainen K, Kaihovaara P etal (1996) Aldehyde
dehydrogenase activity and acetate production by aerobic bacte-
ria representing the normal flora of human large intestine. Alco-
hol Alcohol 31(6):555–564
109. McManus IR, Contag AO, Olson RE (1960) Characterization of
endogenous ethanol in the mammal. Science 131:102–103
110. Baraona E, Julkunen R, Tannenbaum L etal (1986) Role of intes-
tinal bacterial overgrowth in ethanol production and metabolism
in rats. Gastroenterology 90:103–110
111. Hunnisett A, Howard J, Davies S (1990) Gut fermentation (or
autobrewery) syndrome. A new clinical test with initial obser-
vations and discussion of clinical and biological implications. J
Nutr Med 1:33–38. doi:10.3109/13590849009003132
112. Böck A, Sawers G (1996) Fermentation. In: Neidhardt FC,
Curtiss R III, Ingraham JL, Lin ECC, Low KB, Magasanik B,
Reznikoff B, Riley M, Schaechter M, Umbarger HE (eds) Escher
ichia coli and Salmonella typhimurium: cellular and molecular
biology, 2nd edn. ASM Press, Washington, DC, pp 262–282
113. Elshaghabee FMF, Bockelmann W, Meske D etal (2016) Ethanol
production by selected intestinal microorganisms and lactic acid
bacteria growing under different nutritional conditions. Front
Microbiol 7:47. doi:10.3389/fmicb.2016.00047
114. Volynets V, Küper MA, Strahl S etal (2012) Nutrition, intesti-
nal permeability, and blood ethanol levels are altered in patients
with nonalcoholic fatty liver disease (NAFLD). Dig Dis Sci
57:1932–1941
115. Collins MD, Samelis J, Metaxopoulos J etal (1993) Taxonomic
studies on some leuconostoc-like organisms from fermented sau-
sages: description of a new genus Weissella for the Leuconostoc
paramesenteroides group of species. J Appl Bacteriol 75:595–
603. doi:10.1111/j.1365-2672.1993.tb01600.x
116. Fusco V, Quero GM, Cho G-S etal (2015) The genus Weissella:
taxonomy, ecology and biotechnological potential. Front Micro-
biol 6:155. doi:10.3389/fmicb.2015.00155
117. Buckel W (1999) Anaerobic energy metabolism. In: Lengeler JW,
Drews G, Schlegel HG (eds) Biology of the prokaryotes. Georg
Thieme Verlag, Stuttgart, pp 278–326
118. Lieber C (1991) Hepatic, metabolic and toxic effect of ethanol.
Alcohol Clin Exp Res 15:573–592
119. Gustot T, Lemmers A, Moreno C etal (2006) Differential liver
sensitization to toll-like receptor pathways in mice with alcoholic
fatty liver. Hepatology 43:989–1000
120. Forsyth CB, Tang Y, Shaikh M etal (2011) Role of snail
activation in alcohol-induced iNOS-mediated disruption of
intestinal epithelial cell permeability. Alcohol Clin Exp Res
35(9):1635–1643
121. Yuan L, Bambha K (2015) Bile acid receptors and nonalcoholic
fatty liver disease. World J Hepatol 7(28):2811–2818
122. Ridlon JM, Kang DJ, Hylemon PB (2006) Bile salt biotransfor-
mations by human intestinal bacteria. J Lipid Res 47:241–259
123. Dawson PA, Karpen SJ (2015) Intestinal transport and metabo-
lism of bile acids. J Lipid Res 56:1085–1099
124. Copple BL, Li T (2016) Pharmacology of bile acid receptors:
evolution of bile acids from simple detergents to complex
signaling molecules. Pharmacol Res 104:9–21. doi:10.1016/j.
phrs.2015.12.007
125. Schaap FG, Trauner M, Jansen PLM (2013) Bile acid receptors
as targets for drug development. Nat Rev Gastroenterol Hepatol.
doi:10.1038/nrgastro.2013.151 (Advance online publication)
126. Claudel T, Staels B, Kuipers F (2005) The farnesoid X receptor:
a molecular link between bile acid and lipid and glucose metabo-
lism. Arterioscler Thromb Vasc Biol 25:2020–2031
127. Thomas C, Gioiello A, Noriega L etal (2009) TGR5-mediated
bile acid sensing controls glucose homeostasis. Cell Metab
10(3):167–177
128. Sayin SI, Wahlström A, Felin J etal (2013) Gut microbiota
regulates bile acid metabolism by reducing the levels of tauro-
beta-muricholic acid, a naturally occurring FXR antagonist. Cell
Metab 17:225–235
129. Ferslew BC, Xie G, Johnston CK etal (2015) Altered bile acid
metabolome in patients with nonalcoholicv steatohepatitis. Dig
Dis Sci 60(11):3318–3328. doi:10.1007/s10620-015-3776-8
130. Aranha MM, Cortez-Pinto H, Costa A etal (2008) Bile acid
levels are increased in the liver of patients with steatohepatitis.
Eur J Gastroenterol Hepatol 20(6):519–525
Author's personal copy
Eur J Nutr
1 3
131. Kalhan S, Guo L, Edmison J etal (2011) Plasma metabolomic pro-
file in nonalcoholic fatty liver disease. Metabolism 60(3):404–413
132. Faubion WA, Guicciardi ME, Miyoshi H etal (1999) Toxic bile
salts induce rodent hepatocyte apoptosis via direct activation of
Faz. J Clin Invest 103:137–145
133. Parlesak A, Schaeckeler S, Moser L etal (2007) Conjugated pri-
mary bile salts reduce permeability of endotoxin through intes-
tinal epithelial cells and synergize with phosphatidylcholine in
suppression of inflammatory cytokine production. Crit Care Med
35:2367–2374
134. Lorenzo-Zúñiga V, Bartolí R, Planas R etal (2003) Oral bile acids
reduce bacterial overgrowth, bacterial translocation, and endotox-
emia in cirrhotic rats. Hepatology 37(3):551–557. doi:10.1053/
jhep.2003.50116
135. Sherriff JL, O’Sullivan TA, Properzi C etal (2016) Choline, its
potential role in nonalcoholic fatty liver disease, and the case for
human and bacterial genes. Adv Nutr 7(1):5–13
136. Y-m Chen, Liu Y, R-f Zhou etal (2016) Associations of gut-flora-
dependent metabolite trimethylamine-N-oxide, betaine and choline
with nonalcoholic fatty liver disease in adults. Sci Rep 6:19076.
doi:10.1038/srep19076
137. Gao X, Liu X, Xu J etal (2014) Dietary trimethylamine N-oxide
exacerbates impaired glucose tolerance in mice fed a high fat diet.
J Biosci Bioeng 118:476–481
138. Shih DM, Wang Z, Lee R etal (2015) Flavin containing monooxy-
genase 3 exerts broad effects on glucose and lipid metabolism and
atherosclerosis. J Lipid Res 56:22–37
139. Raubenheimer PJ, Nyirenda MJ, Walker BR (2006) A choline-
deficient diet exacerbates fatty liver but attenuates insulin resist-
ance and glucose intolerance in mice fed a high-fat diet. Diabetes
55:2015–2020
140. Pelz S, Stock P, Brückner S etal (2012) A methionine-choline-
deficient diet elicits NASH in the immunodeficient mouse featuring
a model for hepatic cell transplantation. Exp Cell Res 318:276–287.
doi:10.1016/j.yexcr.2011.11.005
141. Guerrerio AL, Colvin RM, Schwartz AK etal (2012) Choline
intake in a large cohort of patients with nonalcoholic fatty liver
disease. Am J Clin Nutr 95:892–900
142. Spencer MD, Hamp TJ, Reid RW etal (2011) Association between
composition of the human gastrointestinal microbiome and devel-
opment of fatty liver with choline deficiency. Gastroenterology
140(3):976–986
143. Walsh CJ, Guinane CM, O’Toole PW etal (2014) Beneficial
modulation of the gut microbiota. FEBS Lett. doi:10.1016/j.
febslet.2014.03.035
144. Fava F, Gitau R, Griffin BA etal (2013) The type and quantity of
dietary fat and carbohydrate alter fecal microbiome and short-chain
fatty acid excretion in a metabolic syndrome ‘at-risk’ population.
Int J Obes 37:216–223
145. Xu Z, Knight R (2015) Dietary effects on human gut microbiome
diversity. Br J Nutr 113:S1–S5
146. National Institute for Health and Care Excellence (2016) Nonal-
coholic fatty liver disease: assessment and management. NICE
Guideline NG49, Methods, evidence and recommendations. ISBN:
978-1-4731-1955-0. http://nice.org.uk/guidance/ng49
147. Schwenger KJP, Allard JP (2014) Clinical approaches to nonalco-
holic fatty liver disease. World J Gastroenterol 20(7):1712–1723
148. Zhang C, Li S, Yang L etal (2013) Structural modulation of gut
microbiota in life-long calorie-restricted mice. Nat Commun
4:2163. doi:10.1038/ncomms3163
149. Santacruz A, Marcos A, Wärnberg J etal (2009) Interplay between
weight loss and gut microbiota composition in overweight adoles-
cents. Obesity 17:1906–1915
150. Kim MS, Hwang SS, Park EJ etal (2013) Strict vegetarian diet
improves the risk factors associated with metabolic diseases by
modulating gut microbiota and reducing intestinal inflammation.
Environ Microbiol Rep 5:765–775
151. Cotillard A, Kennedy SP, Kong LC etal (2013) Dietary intervention
impact on gut microbial gene richness. Nature 29(500):585–588
152. Breusing N, Lagerpusch M, Engstler AJ etal (2017) Influence of
energy balance and glycemic index on metabolic endotoxemia in
healthy men. J Am Coll Nutr 0(0):1–8. doi:10.1080/07315724.20
16.1156036
153. Obregon-Tito AJ, Tito RY, Metcalf JL etal (2015) Subsistence
strategies in traditional societies distinguish gut microbiomes. Nat
Commun 6:6505
154. Amato KR, Yeoman CJ, Cerda G etal (2015) Variable responses
of human and non-human primate gut microbiomes to a Western
diet. Microbiome 3:53. doi:10.1186/s40168-015-0120-7
155. Wu GD, Chen J, Hoffmann C etal (2011) Linking long-term die-
tary patterns with gut microbial enterotypes. Science 334:105–108
156. De Filippo C, Cavalieri D, Di Paola M etal (2010) Impact of diet in
shaping gut microbiota revealed by a comparative study in children
from Europe and rural Africa. PNAS 107(33):14691–14696
157. Pendyala S, Walker JM, Holt PR (2012) A high-fat diet is associ-
ated with endotoxemia that originates from the gut. Gastroenterol-
ogy 142(5):1100–1101.e2. doi:10.1053/j.gastro.2012.01.034
158. De Wit N, Derrien M, Bosch-Vermeulen H etal (2012) Saturated
fat stimulates obesity and hepatic steatosis and affects gut microbi-
ota composition by an enhanced overflow of dietary fat to the distal
intestine. Am J Physiol Gastrointest Liver Physiol 303:589–599
159. Caesar R, Tremaroli V, Kovatcheva-Datchary P etal (2015) Cross-
talk between gut microbiota and dietary lipids aggravates WAT
Inflammation through TLR signaling. Cell Metab 22(4):658–668
160. Brinkworth GD, Noakes M, Clifton PM etal (2009) Comparative
effects of very low-carbohydrate, high-fat and highcarbohydrate,
low-fat weight-loss diets on bowel habit and fecal short-chain fatty
acids and bacterial populations. Br J Nutr 101:1493–1502
161. Duncan SH, Belenguer A, Holtrop G etal (2007) Reduced dietary
intake of carbohydrate, by obese subjects, results in decreased con-
centrations of butyrate and butyrate-producing bacteria in feces.
Appl Environ Microbiol 73:1073–1078
162. Lopez-Legarrea P, Fuller NR, Zulet MA etal (2014) The influ-
ence of Mediterranean, carbohydrate and high protein diets on gut
microbiota composition in the treatment of obesity and associated
inflammatory state. Asia Pac J Clin Nutr 23(3):360–368
163. Hooda S, Boler BMV, Kerr KR etal (2013) The gut microbiome of
kittens is affected by dietary protein: carbohydrate ratio and associ-
ated with blood metabolite and hormone concentrations. Br J Nutr
109(9):1637–1646
164. Smith EA, Macfarlane GT (1996) Studies on amine production in
the human colon: enumeration of amine forming bacteria and phys-
iological effects of carbohydrate and pH. Anaerobe 2(5):285–297
165. Russell WR, Gratz SW, Duncan SH et al (2011) Highprotein,
reduced-carbohydrate weight-loss diets promote metabolite pro-
files likely to be detrimental to colonic health. Am J Clin Nutr
93(5):1062–1072
166. Cani PD, Neyrinck AM, Fava F etal (2007) Selective increases of
bifidobacteria in gut microflora improve high-fat-diet-induced dia-
betes in mice through a mechanism associated with endotoxaemia.
Diabetologia 50:2374–2383
167. Reddy BS, Weisburger JH, Wynder EL (1995) Effects of high risk
and low risk diets for colon carcinogenesis on fecal microflora and
steroids in man. J Nutr 105(7):878–884
168. Matijasic BB, Obermajer T, Lipoglavsek L etal (2014) Asso-
ciation of dietary type with fecal microbiota in vegetarians and
omnivores in Slovenia. Eur J Nutr 53(4):1051–1064
Author's personal copy
... Помимо липидной нагрузки, активизации цитокинов дополнительный параллельный удар может оказывать микробиота кишечника. Так, согласно последним данным, «ось кишечник -печень» -тесная двунаправленная связь между микробиотой кишечника и печенью, которая играет важную роль в развитии и прогрессировании НАЖБП и сопутствующих заболеваний, включая СД2 типа [23,24]. Желудочнокишечный тракт содержит наибольшее количество бактерий в организме человека и выполняет множество физиологических функций, в том числе принимает активное участие в метаболизме глюкозы и липидов. ...
... В кишечнике человека преобладают четыре основных вида комменсальных бактерий: Firmicutes, Bacteroidetes, Actinobacteria и Proteobacteria. При НАЖБП возникают специфические дисбиотические изменения: преобладают грамотрицательные бактерии, в основном из семейства Enterobacteriaceae, Escherichia (Proteobacteria spp.) и Bacteroidetes (Bacteroidetes spp.), продуцирующие эндотоксины [23,24]. Хроническая энтеротоксемия приводит к избыточному образованию и накоплению свободных радикалов в гепатоцитах, способствуя развитию воспалительной реакции в печени [25]. ...
Article
Today, despite the introduction of primary and secondary prevention methods, chronic non-communicable diseases remain the main cause of high morbidity and mortality throughout the world. According to some studies, hepatic component metabolic syndrome and one of the main factors contributing to the development of cardiometabolic diseases is non-alcoholic severe liver disease. According to the results of studies, NAFLD is currently the leading cause of chronic liver diseases worldwide and is closely associated with the development of cardiometabolic pathology, in particular type 2 diabetes and CVD. This review is devoted to the epidemiology, pathophysiology of the close relationship between NAFLD and type 2 diabetes, as well as therapy affecting insulin resistance, a key link connecting these two nosologies.
... Нарушения кишечного микробиоценоза тесно связаны с патогенезом НАЖБП, посредством изменений бактериального состава и кишечных метаболитов [75,76]. ...
Article
Among dental pathologies, periodontal diseases occupy the 2nd place in prevalence after caries. The prevalence of non-alcoholic fatty liver disease (NAFLD) in the general population in the world reaches 33.0% and is detected in all age categories. The presence of common risk factors suggests an indirect interaction between periodontitis and NAFLD, but the question of the possibility of direct interaction without the participation of “intermediaries” is relevant. The review examines the bidirectional relationship between periodontal pathology and NAFLD. The results of experimental and clinical studies indicate that periodontal bacteria, especially Porphyromonas gingivalis, correlate with the development of NAFLD. P. gingivalis has been detected in the liver, and LPS from this bacterium has been shown to be involved in the progression of NAFLD, suggesting a possible direct role of P. gingivalis in NAFLD. In addition, P. gingivalis causes disruption of the intestinal microbiocenosis, which contributes to the progression of NAFLD. There are two possible routes connecting the oral cavity and the liver - hematogenous and enteral diffusion of hepatotoxic components. The emerging concept of changes in the oral microbiota associated with periodontitis is closely related to the role of the mouth-gut-liver axis in the pathogenesis of NAFLD, based on the close relationship between the gut and liver connected by the enterohepatic circulation.
... Among them, Faecalibacterium prausnitzii is an important bacterium in the human gut that produces butyric acid, protecting the digestive system from intestinal pathogens (Miquel et al., 2015;Zhou et al., 2018Zhou et al., , 2021). An increased abundance of Bacteroides may trigger inflammatory effects and contribute to the development of diseases such as insulin resistance and obesity (Grasset et al., 2017;De Faria Ghetti et al., 2018;Lieber et al., 2019). Among them, Bacteroides fragilis, which is thought to have anti-inflammatory effects, is the more widely reported species. ...
Article
Full-text available
The research on the correlation or causality between gut microbiota and the occurrence, development, and treatment of colorectal cancer (CRC) is receiving increasing emphasis. At the same time, the incidence and mortality of colorectal cancer vary among individuals and regions, as does the gut microbiota. In order to gain a better understanding of the characteristics of the gut microbiota in CRC patients and the differences between different regions, we initially compared the gut microbiota of 25 CRC patients and 26 healthy controls in the central region of China (Hubei Province) using 16S rRNA high-throughput sequencing technology. The results showed that Corynebacterium, Enterococcus, Lactobacillus , and Escherichia-Shigella were significantly enriched in CRC patients. In addition, we also compared the potential differences in functional pathways between the CRC group and the healthy control group using PICRUSt’s functional prediction analysis. We then analyzed and compared it with five cohort studies from various regions of China, including Central, East, and Northeast China. We found that geographical factors may affect the composition of intestinal microbiota in CRC patients. The composition of intestinal microbiota is crucial information that influences colorectal cancer screening, early detection, and the prediction of CRC treatment outcomes. This emphasizes the importance of conducting research on CRC-related gut microbiota in various regions of China.
... The exact role of the gut microbiota in NASH pathogenesis is still unknown. Gut endotoxins, mainly bacterial lipopolysaccharides (LPS), have been shown to promote hepatic inflammation [18]. Further possible hypotheses involve induction of a microbiota-induced gut barrier dysfunction by increasing epithelial permeability, shifts in bile acid composition influencing microbiota community structure, and bacterial metabolites, like ethanol, influencing hepatic metabolism [19]. ...
Article
Full-text available
Liver steatosis is the most frequent liver disorder and its advanced stage, non-alcoholic steatohepatitis (NASH), will soon become the main reason for liver fibrosis and cirrhosis. The “multiple hits hypothesis” suggests that progression from simple steatosis to NASH is triggered by multiple factors including the gut microbiota composition. The Epstein Barr virus induced gene 2 (EBI2) is a receptor for the oxysterol 7a, 25-dihydroxycholesterol synthesized by the enzymes CH25H and CYP7B1. EBI2 and its ligand control activation of immune cells in secondary lymphoid organs and the gut. Here we show a concurrent study of the microbial dysregulation and perturbation of the EBI2 axis in a mice model of NASH. We used mice with wildtype, or littermates with CH25H−/−, EBI2−/−, or CYP7B1−/− genotypes fed with a high-fat diet (HFD) containing high amounts of fat, cholesterol, and fructose for 20 weeks to induce liver steatosis and NASH. Fecal and small intestinal microbiota samples were collected, and microbiota signatures were compared according to genotype and NASH disease state. We found pronounced differences in microbiota composition of mice with HFD developing NASH compared to mice did not developing NASH. In mice with NASH, we identified significantly increased 33 taxa mainly belonging to the Clostridiales order and/ or the family, and significantly decreased 17 taxa. Using an Elastic Net algorithm, we suggest a microbiota signature that predicts NASH in animals with a HFD from the microbiota composition with moderate accuracy (area under the receiver operator characteristics curve = 0.64). In contrast, no microbiota differences regarding the studied genotypes (wildtype vs knock-out CH25H−/−, EBI2−/−, or CYP7B1−/−) were observed. In conclusion, our data confirm previous studies identifying the intestinal microbiota composition as a relevant marker for NASH pathogenesis. Further, no link of the EBI2 – oxysterol axis to the intestinal microbiota was detectable in the current study.
... Gut dysbiosis increases intestinal permeability, favoring toxic bacterial product translocation, higher free fatty acid (FFA) absorption, increased proinflammatory cytokines, lipopolysaccharide and ammonia, also intestinal inflammation and dysmotility, etc. [131,133]. Patients with NAFLD have an altered composition of gut microbiota and exhibit a distinct microbial signature, although the microbiome profile is not entirely deciphered [134]. It seems though that there is an increase in the Bacteroidetes and a decrease in the Firmicutes phyla associated with significant liver fibrosis, although other changes have been observed (increase in Proteobacteria phylum and several species such as Bacteroides, Escherichia coli, and possibly Ruminococcus; decrease in Prevotella, etc.) [135][136][137]. ...
Article
Full-text available
The bidirectional relationship between type 2 diabetes and (non-alcoholic fatty liver disease) NAFLD is indicated by the higher prevalence and worse disease course of one condition in the presence of the other, but also by apparent beneficial effects observed in one, when the other is improved. This is partly explained by their belonging to a multisystemic disease that includes components of the metabolic syndrome and shared pathogenetic mechanisms. Throughout the progression of NAFLD to more advanced stages, complex systemic and local metabolic derangements are involved. During fibrogenesis, a significant metabolic reprogramming occurs in the hepatic stellate cells, hepatocytes, and immune cells, engaging carbohydrate and lipid pathways to support the high-energy-requiring processes. The natural history of NAFLD evolves in a variable and dynamic manner, probably due to the interaction of a variable number of modifiable (diet, physical exercise, microbiota composition, etc.) and non-modifiable (genetics, age, ethnicity, etc.) risk factors that may intervene concomitantly, or subsequently/intermittently in time. This may influence the risk (and rate) of fibrosis progression/regression. The recognition and control of the factors that determine a rapid progression of fibrosis (or its regression) are critical, as the fibrosis stages are associated with the risk of liver-related and all-cause mortality.
Preprint
Full-text available
Context Polycystic ovary syndrome (PCOS), a common endocrine disorder in women of reproductive age, is closely associated with chronic low-grade inflammation and metabolic disturbances. In PCOS mice, dietary inulin has been demonstrated to regulate intestinal flora and inflammation. However, the efficacy of dietary inulin in clinical PCOS remains unclear. Objective The intestinal flora and related metabolic indexes of obese patients with polycystic ovary syndrome (PCOS) after 3 months of inulin treatment were analyzed. Setting and design To analyze the intestinal flora and related metabolic indexes in healthy controls and obese patients with polycystic ovary syndrome after 3 months of inulin treatment. Results The results showed that dietary inulin improved sex hormone disorders, reduced BMI and WHR levels in obese women with PCOS. In addition, the inulin intervention reduced plasma TNF-α, IL-1β, IL-6, and MCP-1levels. Inulin intervention increased the abundance of Actinobacteria, Fusobacteria, Lachnospira, and Bifidobacterium, as well as decreased the ratio of F/B and the abundance of proteobacteria, Sutterella, and Enterobacter.Correlation analyses showed a strong relationship among plasma inflammatory factors, sex steroid hormones, and the intestinal flora of patients. Conclusions Dietary inulin may improve obese PCOS women disease through the gut flora-inflammation-steroid hormone pathway.
Article
Full-text available
Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are closely related liver conditions that have become more prevalent globally. This review examines the intricate interplay between microbiome dysbiosis and mitochondrial dysfunction in the development of NAFLD and NASH. The combination of these two factors creates a synergistic situation referred to as “double trouble”, which promotes the accumulation of lipids in the liver and the subsequent progression from simple steatosis (NAFLD) to inflammation (NASH). Microbiome dysbiosis, characterized by changes in the composition of gut microbes and increased intestinal permeability, contributes to the movement of bacterial products into the liver. It triggers metabolic disturbances and has anti-inflammatory effects. Understanding the complex relationship between microbiome dysbiosis and mitochondrial dysfunction in the development of NAFLD and NASH is crucial for advancing innovative therapeutic approaches that target these underlying mechanisms.
Article
Full-text available
Oral microbiome dysbiosis mediates chronic periodontal disease, gut microbial dysbiosis, and mucosal barrier disfunction that leads to steatohepatitis via the enterohepatic circulation. Improving this dysbiosis towards health may improve liver disease. Treatment with antibiotics and probiotics have been used to modulate the microbial, immunological, and clinical landscape of periodontal disease with some success. The aim of the present investigation was to evaluate the potential for nisin, an antimicrobial peptide produced by Lactococcus lactis , to counteract the periodontitis-associated gut dysbiosis and to modulate the glycolipid-metabolism and inflammation in the liver. Periodontal pathogens, namely Porphyromonas gingivalis , Treponema denticola , Tannerella forsythia and Fusobacterium nucleatum , were administrated topically onto the oral cavity to establish polymicrobial periodontal disease in mice. In the context of disease, nisin treatment significantly shifted the microbiome towards a new composition, commensurate with health while preventing the harmful inflammation in the small intestine concomitant with decreased villi structural integrity, and heightened hepatic exposure to bacteria and lipid and malondialdehyde accumulation in the liver. Validation with RNA Seq analyses, confirmed the significant infection-related alteration of several genes involved in mitochondrial dysregulation, oxidative phosphorylation, and metal/iron binding and their restitution following nisin treatment. In support of these in vivo findings indicating that periodontopathogens induce gastrointestinal and liver distant organ lesions, human autopsy specimens demonstrated a correlation between tooth loss and severity of liver disease. Nisin’s ability to shift the gut and liver microbiome towards a new state commensurate with health while mitigating enteritis, represents a novel approach to treating NAFLD-steatohepatitis-associated periodontal disease.
Article
Full-text available
Gut-derived bacterial lipopolysaccharide (LPS) and subsequent hepatic toll-like receptor 4 (TLR4) activation have been recognized to be involved in the onset of diet-induced nonalcoholic fatty liver disease (NAFLD), but little is known about the variation of LPS and TLR4 during the progression of NAFLD. Probiotics were able to inhibit proliferation of harmful bacteria and improve gastrointestinal barrier function. However, it?s unclear whether LPS/TLR4 is involved in the protection effect of probiotics on NAFLD. In this study, we described characteristic of gut microbiota structure in the progression of NAFLD, and we also analyzed the relationship between gut microbiota and LPS/TLR4 in this process. Furthermore, we applied probiotics intervention to investigate the effect of probiotics on gut flora structure, intestinal integrity, serum LPS, liver TLR4 and liver pathology. Our results showed that serum LPS and liver TLR4 were highly increased during progression of NAFLD, with gut flora diversity and gut mircobiological colonization resistance (B/E) declining. Furthermore, probiotics could improve gut microbiota structure and liver pathology. Probiotics could also downregulate serum LPS and liver TLR4. Our results suggested that both gut flora alteration and endotoxemia may be involved in the progression of NAFLD. Probiotics may delay the progression of NAFLD via LPS/TLR4 signaling.
Article
Full-text available
Elucidating the mechanisms by which short chain fatty acids (SCFA) reduce body weight may assist in the development of an effective weight control strategy. Dietary supplementation of acetate, propionate, butyrate or their admixture was shown to significantly inhibit the body weight gain induced by high-fat diet feeding. Supplementation of SCFAs caused significant changes in the expressions of G-protein coupled receptor 43 (GPR43) and GPR41 characterized by increases in the adipose tissue and reductions in the colon. Additionally, they influenced the bacterial community structure in feces, with a reduction in the proportion of Firmicutes and an increase in the proportion of Bacteroidetes. The effects of dietary SCFAs on the GPR expression and gut microbiota composition may further result in body weight reduction by enhancing triglyceride hydrolysis and FFA oxidation in the adipose tissue, promoting beige adipogenesis and mitochondrial biogenesis, and inhibiting chronic inflammation.
Article
Full-text available
Increasing evidence suggests a role of intestinal dysbiosis in obesity and non-alcoholic fatty liver disease (NAFLD). But it remains unknown in nonobese NAFLD. This prospective, cross-sectional study sought to characterize differences in fecal microbiota between nonobese adult individuals with and without NAFLD and their potential association with metabolic markers of disease progression. A total of 126 nonobese subjects were enrolled: 43 NAFLD and 83 healthy controls (HC). The microbial community was profiled by denaturing gradient gel electrophoresis and examined by 454 pyrosequencing of the 16S ribosomal RNA V3 region. Lower diversity and a phylum-level change in the fecal microbiome were found in NAFLD. Compared with HC, patients had 20% more phylum Bacteroidetes (p = 0.005) and 24% less Firmicutes (p = 0.002). Within Firmicutes, four families and their 8 genera, which were short-chain fatty acids-producing and 7α-dehydroxylating bacteria, were significantly decreased. Moreover, Gram-negative (G−) bacteria were prevalent in NAFLD (p = 0.008). Furthermore, a significant correlation with metabolic markers was revealed for disturbed microbiota in NAFLD. This novel study indicated that intestinal dysbiosis was associated with nonobese NAFLD and might increase the risk of NAFLD progression.
Article
Full-text available
Objective: The twin pandemics of obesity and Type 2 diabetes (T2D) are a global challenge for health care systems. Changes in the environment, behavior, diet, and lifestyle during the last decades are considered the major causes. A Western diet, which is rich in saturated fat and simple sugars, may lead to changes in gut microbial composition and physiology, which have recently been linked to the development of metabolic diseases. Methods: We will discuss evidence that demonstrates the influence of the small and large intestinal microbiota on weight regulation and the development of insulin resistance, based on literature search. Results: Altered large intestinal microbial composition may promote obesity by increasing energy harvest through specialized gut microbes. In both large and small intestine, microbial alterations may increase gut permeability that facilitates the translocation of whole bacteria or endotoxic bacterial components into metabolic active tissues. Moreover, changed microbial communities may affect the production of satiety-inducing signals. Finally, bacterial metabolic products, such as short chain fatty acids (SCFAs) and their relative ratios, may be causal in disturbed immune and metabolic signaling, notably in the small intestine where the surface is large. The function of these organs (adipose tissue, brain, liver, muscle, pancreas) may be disturbed by the induction of low-grade inflammation, contributing to insulin resistance. Conclusions: Interventions aimed to restoring gut microbial homeostasis, such as ingestion of specific fibers or therapeutic microbes, are promising strategies to reduce insulin resistance and the related metabolic abnormalities in obesity, metabolic syndrome, and type 2 diabetes. This article is part of a special issue on microbiota.
Article
There is evidence that nonalcoholic fatty liver disease (NAFLD) is affected by gut microbiota. Therefore, we investigated its modifications in pediatric NAFLD patients using targeted metagenomics and metabolomics. Stools were collected from 61 consecutive patients diagnosed with nonalcoholic fatty liver (NAFL), nonalcoholic steatohepatitis (NASH), or obesity and 54 healthy controls (CTRLs), matched in a case-control fashion. Operational taxonomic units were pyrosequenced targeting 16S ribosomal RNA and volatile organic compounds determined by solid-phase microextraction gas chromatography-mass spectrometry. The alpha-diversity was highest in CTRLs, followed by obese, NASH, and NAFL patients; and beta-diversity distinguished between patients and CTRLs but not NAFL and NASH. Compared to CTRLs, in NAFLD patients Actinobacteria were significantly increased and Bacteroidetes reduced. There were no significant differences among the NAFL, NASH, and obese groups. Overall NAFLD patients had increased levels of Bradyrhizobium, Anaerococcus, Peptoniphilus, Propionibacterium acnes, Dorea, and Ruminococcus and reduced proportions of Oscillospira and Rikenellaceae compared to CTRLs. After reducing metagenomics and metabolomics data dimensionality, multivariate analyses indicated a decrease of Oscillospira in NAFL and NASH groups and increases of Ruminococcus, Blautia, and Dorea in NASH patients compared to CTRLs. Of the 292 volatile organic compounds, 26 were up-regulated and 2 down-regulated in NAFLD patients. Multivariate analyses found that combination of Oscillospira, Rickenellaceae, Parabacteroides, Bacteroides fragilis, Sutterella, Lachnospiraceae, 4methyl-2-pentanone, 1-butanol, and 2-butanone could discriminate NAFLD patients from CTRLs. Univariate analyses found significantly lower levels of Oscillospira and higher levels of 1-pentanol and 2-butanone in NAFL patients compared to CTRLs. In NASH, lower levels of Oscillospira were associated with higher abundance of Dorea and Ruminococcus and higher levels of 2-butanone and 4-methyl-2-pentanone compared to CTRLs. Conclusion: An Oscillospira decrease coupled to a 2-butanone up-regulation and increases in Ruminococcus and Dorea were identified as gut microbiota signatures of NAFL onset and NAFL-NASH progression, respectively.
Article
Objective: Overfeeding with a high-fat and/or high-carbohydrate (CHO) diet is known to increase plasma concentrations of endotoxin (lipopolysaccharide [LPS]) that may lead to metabolic disturbances like insulin resistance. The impact of CHO quality (i.e., the glycemic index [GI]) independent of fat intake on metabolic endotoxemia remains unclear. In the present study, the effects of changes in energy balance and GI on plasma endotoxin were studied. Methods: Fifteen healthy young men overconsumed diets containing 65% CHO and 20% fat for 1 week (OF; +50% of energy requirement) followed by 3 weeks of caloric restriction (CR; -50% of energy requirement) and were then randomized to 2 weeks hypercaloric refeeding (RF, +50% of energy requirement) with either a low- or high-GI (40 vs 74) diet. Results: During OF, subjects gained 1.9 ± 0.7 kg body weight (+0.6 ± 0.8% fat mass) followed by a weight loss of 6.1 ± 0.8 kg (-2.0 ± 0.6% fat mass) and weight regain of 4.0 ± 0.6 kg (0.9 ± 0.8% fat mass). Fasting insulin and homeostasis model assessment-insulin resistance (HOMAIR) increased with OF and RF and decreased with CR, MatsudaISI decreased by 37% after RF (all p < 0.05). Endotoxin significantly increased by 30.8% with OF and by 24.7% with RF (both p < 0.05), whereas CR normalized endotoxin levels. No difference in endotoxin levels was observed between refeeding a hypercaloric high- or low-GI diet. Changes in endotoxin levels with RF were not related to changes in insulin sensitivity. Conclusion: A hypercaloric diet (OF and RF) increased plasma endotoxin irrespective of GI, whereas a negative energy balance did not reduce endotoxemia. Impaired insulin sensitivity with hypercaloric refeeding on a high-GI diet was not explained by metabolic endotoxemia.
Article
Gastrointestinal symptoms are commonly seen in the primary care setting. 1 These patient presentations can be nonspecific, leading to a broad differential diagnosis. Small intestinal bacterial overgrowth is a clinical entity that can present with many of these nonspecific gastrointestinal symptoms. The recent interest in the microbiome by those in the medical and lay communities has made this syndrome all the more relevant. This review gives the primary care provider an up-to-date understanding of the etiology, risk factors and predisposing factors, presentation, diagnostic testing, and management of small intestinal bacterial overgrowth.
Article
Context: High plasma VLDL-triglyceride (TG) concentration and alterations in VLDL-TG metabolism are associated with cardiometabolic disease. Objective: To evaluate the inter-relationships among factors purported to regulate VLDL-TG metabolism in a large cohort of men and women with a wide range in body adiposity and fat distribution but without diabetes. Subjects and design: We assessed body composition and fat distribution, plasma insulin concentration, free fatty acid (FFA) availability, and basal VLDL-TG and VLDL-apoB-100 (VLDL particle number) kinetics in 233 lean, overweight and obese men and women. Results: We found that: 1) plasma VLDL-TG concentration is determined primarily by VLDL-TG secretion rate (SR) in men and by VLDL-TG clearance rate in women; 2) there is a dissociation between VLDL-TG and VLDL-apoB-100 SRs, and VLDL-apoB-100 SR only explains ∼30% of the variance in VLDL-TG SR; 3) ∼50% of people with obesity have high plasma VLDL-TG concentration due to both an increased VLDL-TG SR and a decreased rate of VLDL-TG plasma clearance, and they have lower plasma HDL-cholesterol concentration and more intra-abdominal and liver fat than those with normal VLDL-TG concentration; 4) fat-free mass, liver fat content and the rate of FFA release into plasma are independent predictors (with a sex × race interaction) of VLDL-TG SR. Conclusions: The regulation of plasma VLDL-TG concentration is complex and influenced by multiple metabolic factors. Many people with obesity have normal plasma VLDL-TG concentrations and kinetics, whereas those with high plasma VLDL-TG concentrations have increased VLDL-TG SR and other markers of cardiometabolic disease risk.
Article
The reliability of a single jejunal culture in the diagnosis of small bowel bacterial overgrowth has recently been questioned. Seventy-seven patients thought to have bacterial overgrowth, defined as a jejunal culture yielding at least 106 organisms per milliliter of aspirate, took part in the study. Bacterial overgrowth was found in 74% of the patients with predisposing conditions and in 32% of those with no clear causes of bacterial colonization. The intestinal juice of some patients was taken at two different levels of the proximal jejunum, using both the closed-and open-tube systems. Highly significant correlations (r8 = 0.90, p < 0.001) were found between the numbers of bacteria per milliliter at the 2 jejunal levels and between the numbers of bacteria per milliliter of jejunal aspirate obtained from the closed and open tubes (r8 = 0.84, p < 0.001). Compared with the jejunal culture, the gas chromatography of volatile fatty acids in jejunal aspirate and the glucose-and lactulose-hydrogen breath tests showed sensitivities of 56%, 62%, and 68% and specificities of 100%, 83%, and 44%, respectively. This work demonstrates the reliability of jejunal cultures and the inadequacy of breath hydrogen testing in the prediction of positive jejunal cultures. When results of testing for volatile fatty acids in jejunal aspirates are positive, this always indicates the presence of bacterial overgrowth; thus, this procedure would avoid the more complicated, time-consuming, and costly bacteriological analysis of jejunal samples.
Article
Background/Aims: In the past, nonalcoholic steatohepatitis has been described mostly in obese women with diabetes. The aim of this study was to describe a series of patients with nonalcoholic steatohepatitis with a different clinical profile. Methods: The clinical, biochemical, and histological features of 33 patients with nonalcoholic steatohepatitis seen from July 1990 to June 1993 were analyzed. Results: The mean age was 47 years. All patients were antibody to hepatitis C virusnegative. Nineteen of 33 (58%) were men, 20 of 33 (61%) were nonobese, 26 of 33 (79%) had normal glucose levels, and 26 of 33 (79%) had normal lipid levels. Fourteen of 33 (42%) had normal glucose and lipid levels and were not obese. Thirteen of 33 (39%) had pathological increases in fibrosis, 5 of whom had micronodular cirrhosis. Of these 13 with severe, progressive disease, 8 (62%) were women, 8 (62%) were obese, 4 (31%) were diabetic or had an elevated glucose level, and 3 (23%) had hyperlipidemia. Although serum iron studies (transferrin saturation and ferritin) were abnormal in 18 of 31 (58%), no patient had hemochromatosis. Conclusions: Nonalcoholic Steatohepatitis can be a severe, progressive liver disease leading to the development of cirrhosis. It should no longer be considered a disease predominantly seen in obese women with diabetes.