You are on page 1of 14

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/315303554

Nonalcoholic Fatty Liver Disease, the Gut Microbiome, and Diet

Article  in  Advances in Nutrition · March 2017


DOI: 10.3945/an.116.013151

CITATIONS READS

63 182

3 authors, including:

Zeinab Mokhtari Azita Hekmatdoost


Shahid Beheshti University of Medical Sciences University of British Columbia - Vancouver
14 PUBLICATIONS   165 CITATIONS    224 PUBLICATIONS   2,892 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Nutrition and Non Alcoholic fatty liver disease (NAFLD) View project

synbiotic supplements in patients with irritable bowel syndrome View project

All content following this page was uploaded by Zeinab Mokhtari on 05 April 2018.

The user has requested enhancement of the downloaded file.


REVIEW

Nonalcoholic Fatty Liver Disease, the Gut


Microbiome, and Diet1,2
Zeinab Mokhtari,3 Deanna L Gibson,4 and Azita Hekmatdoost3,5*
3
Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology
Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran; 4Department of Biology, University of British Columbia, Okanagan
Campus, Kelowna, British Columbia, Canada; and 5Department of Gastroenterology, Hepatology, and Nutrition, University of British Columbia,
Vancouver, British Columbia, Canada

Downloaded from advances.nutrition.org at Fudan University on March 15, 2017


ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common liver disorder in the world, yet the pathogenesis of the disease is not well
elucidated. Due to the close anatomic and functional association between the intestinal lumen and the liver through the portal system, it is
speculated that the gut microbiome may play a pivotal role in the pathogenesis of NAFLD. Furthermore, diet, which can modulate the gut
microbiome and several metabolic pathways involved in NAFLD development, shows a potential tripartite relation between the gut, diet, and
the liver. In this review, we summarize the current evidence that supports the association between NAFLD, the gut microbiome, and the role of
diet. Adv Nutr 2017;8:240–52.

Keywords: nonalcoholic fatty liver disease, gut microbiota, diet, NAFLD, NASH, probiotics

Introduction health, and disease of the host (10), which suggests that
Nonalcoholic fatty liver disease (NAFLD)6 is the most com- the gut microbiome is a metabolic organ in the host. Con-
mon cause of chronic liver diseases in the world (1). NAFLD sidering that the anatomy of the liver facilitates a close inter-
is defined as a liver pathologic spectrum, which is initiated action between the liver and gut microbiome, it is possible
by steatosis that may progress to nonalcoholic steatohepati- that gut microbial metabolites and circulating byproducts
tis (NASH). NASH manifestations beyond steatosis include influence liver function. Indeed, it is known that slow blood
necro-inflammation and fibrosis (2). The disease can prog- flow and fenestrated endothelium in the sinusoids lead to
ress to cirrhosis and eventual hepatocellular carcinoma the interaction between substances derived from the gut
(3). The reported prevalence of NAFLD is 30% in Western and hepatocytes, parenchymal cells, and hepatic immune
countries (1, 4) and 12–24% in Asia (5), which shows the cells (11). Similarly, dietary antigens are known to affect
global importance of understanding the disease pathology. liver function, and because dietary components are among
In addition to genetic background, environmental factors the strongest factors that predict the ecosystem that makes
such as diet and the intestinal microbiome should be consid- up the gut microbiome (12, 13), both diet and the micro-
ered critical factors in the development of NAFLD (6, 7). biome need to be considered in the pathogenesis of NAFLD.
The gut microbiome, the trillions of microbes living in In the present review, we summarize the current evidence
the gut, may contribute to the pathogenesis of liver diseases that supports the association between NAFLD, the gut
(8, 9). Increasing evidence in recent decades shows that the microbiome, and dietary factors.
gut microbiome plays a significant role in metabolism,
Current Status of Knowledge
1
The authors reported no funding received for this study. NAFLD and the gut microbiome. The gut harbors
;1013–1014 bacteria, which is predicted to encode genes
2
Author disclosures: Z Mokhtari, DL Gibson, and A Hekmatdoost, no conflicts of interest.
*To whom correspondence should be addressed. E-mail: a_hekmat2000@yahoo.com.
6
Abbreviations used: ACC, acetyl-CoA carboxylase; AMPK, AMP-activated protein kinase; ASC, >100 times as in the human genome (14). Despite the
apoptosis-associated speck-like protein; FAS, fatty acid synthase; FIAF, fasting-induced ad- wide microbial diversity in humans, only 4 main phyla dom-
ipocyte factor; FOS, fructo-oligosaccharide; GLP1, glucagon-like peptide 1; GPR, G-protein– inate: Firmicutes, Bacteroidetes, Actinobacteria, and Proteo-
coupled receptor; HFD, high-fat diet; LPL, lipoprotein lipase; NAFLD, nonalcoholic fatty liver
disease; NASH, nonalcoholic steatohepatitis; PYY, peptide YY; RS, resistant starch; SREBP-1, bacteria. Approximately 90% of the microbes come from the
sterol regulatory element binding protein 1; TLR, toll-like receptor. Firmicutes and Bacteroidetes phyla (14) and each has unique

240 ã2017 American Society for Nutrition. Adv Nutr 2017;8:240–52; doi:10.3945/an.116.013151.
functions. Major contributors of Firmicutes are as follows: 38). Inflammasomes are cytoplasmic multiprotein complexes
Lachnospiraceae, which are composed mostly of microbes that sense endogenous or exogenous stress- or damage-
from the genera Blautia, Clostridium, Coprococcus, Eubac- associated molecular patterns (39, 40). Upon sensing the rel-
terium, Roseburia, and Ruminococcaceae, which are com- evant signal, they assemble, typically together with the adaptor
posed mostly of the genera Faecalibacterium, Oscillospira, protein, apoptosis-associated speck-like protein (ASC), into a
and Ruminococcus. Major contributors of the Bacteroidetes multiprotein complex that governs caspase-1 activation and
phylum are Bacteroides, Parabacteroides, Porphyromonas, subsequent cleavage of effector proinflammatory cytokines,
Prevotellaceae (Prevotella), and Rikenellaceae (Alistipes). including pro–IL-1b and pro–IL-18, which ultimately leads
The most prominent Actinobacteria are Bifidobacteriaceae to apoptosis (38). In inflammasome deficiency, large amounts
(Bifidobacterium), and the most abundant Proteobacteria of bacterial endotoxins enter the portal vein, and the liver then
are the Enterobacteriaceae (Escherichia) (15, 16). Although has to process high amounts of endotoxins, which leads to ab-
the functions of most Firmicutes are not yet clear, there is errant and damaging inflammatory responses that promote
some evidence that indicates that some members of this progression to NASH (37). Another mechanism by which
phylum are among the butyrate-producing bacteria that the gut microbiota contributes to the development of NAFLD
increase energy extraction from the diet (17–19). On the may be through increasing the number of ethanol-producing
other hand, members of the phylum Bacteroidetes bacteria (e.g., Escherichia coli) (15). These bacteria produce al-
participate in carbohydrate metabolism and accomplish cohol, which could participate in the disruption of gut per-

Downloaded from advances.nutrition.org at Fudan University on March 15, 2017


this by expressing enzymes similar to glycosyl transferases, meability, the generation of ROS, and consequently, liver
glycoside hydrolases, and polysaccharide lyases (20). inflammation (41, 42). However, the specific mechanisms
Understanding the functions associated with the microbial of this hypothesis require further investigation.
community is important, because alterations in the The characterization of the gut microbial composition
intestinal microbiota have been associated with host and comparing it between patients and healthy individuals
diseases, including obesity, type 2 diabetes, and NAFLD is regarded as one way of identifying a healthy microbial ec-
(8, 21, 22). In patients with NAFLD, the microbiome’s osystem (43). Indeed, a few studies have examined dysbiosis
abundance (23) and community structure is altered (also in NAFLD (Table 1). Patients with NASH harbor a lower abun-
called dysbiosis) (15, 24). The importance of the gut dance of Faecalibacterium and Firmicutes (Clostridiales
microbiome in the prevention and treatment of NAFLD is family, Anaerosporobacter) but a higher abundance of Para-
highlighted by the fact that fecal transplantation from bacteroides and Allisonella in their fecal microbiome (45).
mice with NAFLD into wild-type mice caused NAFLD in Importantly, it has been reported that improved intrahepatic
mice that received the microbiota (25). In addition, the TG content is related to a lower abundance of Firmicutes
alteration of microbiota can change the disease severity and a higher abundance of Bacteroidetes (45). In support
(26–30). Overall, these results suggest that the microbiome of this, obese individuals with NAFLD have increased mem-
is important in NAFLD. bers of the phylum Firmicutes [Lachnospiraceae (Dorea,
Several lines of evidence have shown that NAFLD can be Robinsoniella, and Roseburia)] (46). Yet, children and adoles-
affected by the gut microbiome through various proposed cents with NASH do not show similar patterns and instead
mechanisms. The contribution of increased intestinal per- have increased Bacteriodetes [Prevotellaceae (Prevotella,
meability (20), overgrowth of bacteria (15), and elevated se- Porphyromonas)] and Proteobacteria [Enterobacteriaceae
rum LPS (31, 32) has been shown in patients with NAFLD. (Escherichia), Alcaligenaceae] and a decrease in Firmicutes
Overgrowth of bacteria, in particular gram-negative bacte- [Lachnospiraceae (Blautia, Coproccus, Eubacterium, Roseburia),
ria, increases the production of hepatotoxic products such Ruminococcaceae (Faecalibacterium, Oscillospira, Ruminococcus)]
as LPS (23). The disruption of the intestinal integrity in- and Actinobacteria [Bifidobacteriaceae (Bifidobacterium)]
creases intestinal permeability, which leads to bacterial in their fecal microbiomes (15). In support of these stud-
translocation, and bacterial endotoxins penetrate into the ies, pediatric patients with NAFLD showed an increase in
portal vein, which increases the risk of NAFLD development Bradyrhizobium, Anaerococcus, Peptoniphilus, Propionibacterium
through the activation of hepatic inflammatory cells (23, acnes, Dorea, and Ruminococcus and a decrease in Oscillospira
33). Bacterial endotoxins are recognized by toll-like recep- and Rikenellaceae (44). Although it is clear that there may
tors (TLRs) on hepatocytes, which recognize several compo- be some discrepancies in what defines dysbiosis in liver dis-
nents of microbes and initiate immunologic responses (32). ease, the frequency of disease also occurs in association with
When bacterial LPS signals through TLR4, signaling ulti- obesity and is considered a manifestation of metabolic syn-
mately activates NF-kB and proinflammatory cytokines drome. Thus, the dysbiosis may be related to these metabolic
(32, 34). Moreover, dysbiosis is associated with reduced syn- disturbances, considering that several studies suggested
thesis and secretion of fasting-induced adipocyte factor that increased Firmicutes and reduced Bacteroidetes
[(FIAF) also known as angiopoietin-like 4] in enterocytes, may be a cause of obesity (17, 47). However, the reduction
which results in increased activity of lipoprotein lipase in Bacteroidetes is not simply a cause of obesity in patients
(LPL) and the accumulation of TGs in the liver (35, 36). with NASH, considering that Bacteroidetes abundance is re-
Dysbiosis and inflammasome deficiency may be important duced in these patients even after adjusting for BMI and fat
in the development of hepatic steatosis and inflammation (37, intake (24).

Gut microbiome and NAFLD: role of nutrition 241


TABLE 1 Intestinal microbiota composition in patients with NAFLD1
Study, year Change in
(ref) Subjects Method microbiota
Del Chierico et al., 2016 Pediatric NAFLD, NASH, or obesity (n = 61); 16S rRNA pyrosequencing NAFLD vs. healthy controls:
(44) healthy subjects (n = 54) [Bradyrhizobium, Anaerococcus, Peptoniphilus,
Propionibacterium acnes, Dorea, Ruminococcus
YOscillospira and Rikenellaceae
Mouzaki et al., 2013 (24) NASH (n = 22); SS (n = 11); healthy qPCR NASH vs. both SS and healthy controls:
controls (n = 17) YPercentage of Bacteroidetes (Bacteroidetes to
total bacteria counts)
NASH vs. SS:
[Clostridium coccoides
Wong et al., 2013 (45) NASH (n = 16); healthy controls (n = 22) 16S rRNA pyrosequencing NASH vs. healthy controls:
YFaecalibacterium, YAnaerosporobacter
[Parabacteroides, [Allisonella
Improvement in intrahepatic TG content was
associated with a reduction in the abundance of
Firmicutes (R2 = 0.4820, P = 0.0028) and an
increase in Bacteroidetes (R2 = 0.4366,
P = 0.0053)

Downloaded from advances.nutrition.org at Fudan University on March 15, 2017


Raman et al., 2013 (46) Obese NAFLD patients (n = 30); healthy 16S rRNA pyrosequencing Obese NAFLD vs. healthy controls:
controls (n = 30) [Lactobacillus species
[Firmicutes (Lachnospiraceae; genera: Dorea,
Robinsoniella, and Roseburia)
YOne member of phylum Firmicutes
(Ruminococcaceae; genus, Oscillibacter)
YA member of phylum Bacteroidetes
(Porphyromonadaceae)
Zhu et al., 2013 (15) NASH children (n = 22); obese children 16S rRNA pyrosequencing NASH vs. healthy controls:
(n = 25); healthy children (n = 16) [Proteobacteria [Enterobacteriaceae (Escherichia),
Alcaligenaceae]
[Bacteroidetes [Prevotellaceae (Prevotella),
Porphyromonas], YRikenellaceae (Alistipes)
YActinobacteria [Bifidobacteriaceae
(Bifidobacterium)]
YFirmicutes [Lachnospiraceae (Blautia, Coproccus,
Eubacterium, Roseburia), Ruminococcaceae
(Faecalibacterium, Oscillospira, Ruminococcus)],
[Peptoniphilus
Wigg et al., 2001 (23) Healthy controls (n = 23) C-D-xylose and lactulose Small intestinal bacterial overgrowth was present in
breath test 50% of patients with nonalcoholic steatosis and
in 22% of control subjects (P = 0.048)
1
NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; ref, reference; rRNA, ribosomal RNA; SS, simple steatosis; [, increased; Y, decreased.

Overall, the evidence suggests that the gut microbiome Diet and the gut microbiome. Dietary factors are strong
may have an important role in NAFLD pathology, but predictors of the gut microbiota composition (49–51). In
the studies have not identified a particular microbe in- fact, it has been projected that dietary factors play a more im-
volved due to the heterogeneous results. Similar to other portant role in shaping the gut microbiota composition than
microbiome studies, discrepancies can be due to variations do genetic factors (52). To understand the role of nutrition,
in the study designs. Some of the studies, such as the study the gut microbiome, and NAFLD, we summarized the exper-
conducted by Zhu et al. (15), used patients with no history imental studies that evaluated this potential relation (Table 2).
of antibiotics, probiotics, proton pump inhibitors, and his-
tamine receptor antagonists within 3 mo before examining Energy homeostasis and the gut microbiota. An imbal-
the fecal microbiota; however, others did not consider all of ance between energy intake and expenditure is regarded as
these precautions. In addition, the collection and process- the main cause of increased hepatic lipogenesis. Increased
ing of fecal samples have been shown to produce large var- lipogenesis and reduced FA oxidation result in increased sus-
iances and inaccuracies in the interpretation of the taxa ceptibility to hepatic steatosis and the subsequent conse-
present in the microbiota (48), which may be a contribut- quences (63). Several studies indicated that an increase in
ing factor to the conflicting data found in NAFLD micro- the relative abundance of the members of Firmicutes, and
biome studies. Even so, all of these studies have evaluated especially of some Clostridium clusters, is involved in har-
the association, and well-designed studies are needed to vesting energy from the diet (17, 18, 64), and this contrib-
unravel any causal relation between the gut microbes and utes to the development of excessive weight gain and
NAFLD. hepatic lipogenesis.

242 Mokhtari et al.


TABLE 2 Studies evaluating the effects of dietary factors on the gut microbiota in animal models of NAFLD1
Study, year Animal Main
(ref) model Treatment Method(s) results
Saha and Reimer, Wistar rats HF or HP diet from 3 to 15 wk of qPCR HF vs. control group:
2014 (53) age; a high-fat, high-sucrose diet [Total bacteria, [Bifidobacteria,
from 15 to 21 wk; the respective and [Bacteroides:Prevotella
HF or HP diets from 21 to 25 wk HF vs. HP:
YFirmicutes, YFirmicutes:
Bacteroidetes, Yhepatic
cholesterol content
Negative correlation between
liver weight and Bacteroides:
Prevotella (r = 20.415, P = 0.044)
Bomhof et al., 2014 (54) Sprague-Dawley rats Initiate with a high-fat, high-sucrose qPCR Prebiotic oligofructose vs control:
diet for 8 wk and then prebiotic YEnergy intake, Y weight gain,
OFSs vs. the probiotic BB-12 Yfat mass, [PYY, [Bifidobacteria,
for 8 wk [Lactobacilli
Improved glycemia and
Yinsulin concentrations,
Yliver TGs in OFSs and BB-12

Downloaded from advances.nutrition.org at Fudan University on March 15, 2017


[GLP-1 in OFSs
[GLP-2 in probiotic BB-12
No differences in plasma LPS,
TNF-a, IL-6, IL-1β
Ritze et al., 2014 (55) C57BL/6 mice High-fructose diet with LGG vs. qPCR High-fructose diet with LGG vs.
high-fructose diet over 8 wk high-fructose diet:
YALT, Yfat, Yaccumulation in liver,
YChREBP, YACC1, YFAS,
YTNF-a, YIL-1β, [occludin,
YLPS, [total bacterial numbers
Zeng et al., 2013 (56) C57BL/6 mice HFD vs. LFD for 10 wk Sequencing 16S rRNA HFD vs. LFD:
[Hepatic lipid accumulation,
[inflammatory cell infiltration,
[leptin, [TNF-a
[Lactobacillus gasseri and/or
[Lactobacillus taiwanensis
Positive correlation of L. gasseri
and/or L. taiwanensis DNA and
lipid droplets in liver
Park et al., 2013 (57) C57BL/6J mice HFD + probiotic (Lactobacillus Sequencing 16S rRNA HFD + probiotic vs. HFD + placebo:
curvatus HY7601 and Lactobacillus YALT, [FA oxidation–related
plantarum KY1032) vs. genes, Yproinflammatory
HFD + placebo for 10 wk. genes (TNFa, IL6, IL1b), Ygut
microbiota diversity,
[Bifidobacterium Pseudolongum
Pachikian et al., C57BL/6J mice n–3 PUFA–depleted diet for 3 mo DGGE, qPCR n-3 PUFA-depleted diet
2013 (58) supplemented with FOSs during the supplemented with FOSs
last 10 d of treatment vs. n-3 PUFA-depleted diet:
[Bifidobacterium spp.
YRoseburia spp., YSREBP2,
[PPAR-a, YLDL, YHDL, [GLP-1
Cano et al., 2013 (59) C57BL/6 mice HFD supplemented with Bifidobacterium qPCR HFD supplemented with
pseudocatenulatum CECT 7765 vs. B. pseudocatenulatum CECT
HFD for 7 wk 7765 vs. HFD:
YSerum cholesterol, Yserum TGs,
Yserum glucose, Yinsulin
resistance, Yhepatocytes with
grade 3 steatosis,
Yfat absorption, Yleptin,
YIL-6, YMCP-1, [IL-4, [IL-10,
[Bifidobacteria,
YEnterobacteriaceae,
Ybody weight gain,
Yfood intake

(Continued)

Gut microbiome and NAFLD: role of nutrition 243


TABLE 2 (Continued )

Study, year Animal Main


(ref) model Treatment Method(s) results
Axling et al., 2012 (60) C57BL/6J mice HFD with or without a supplement GT Sequencing 16S Lp + GT vs. control:
and supplemented with Lp or the rRNA, qPCR [Lactobacillus, [diversity
combination of both (Lp + GT) of bacteria, Yliver weights,
for 22 wk Yliver size, YTGs, YALT
Akkermansia correlated negatively
with body fat content
(r = 20.43, P = 0.04), plasma
insulin (r = 20.47, P = 0.03),
and liver TGs (r = 20.44,
P = 0.03).
Neyrinck et al., 2011 (61) C57BL/6J mice HFD supplemented with arabinoxylans DGGE, qPCR HFD supplemented with
from wheat vs. HFD for 4 wk arabinoxylans from
wheat vs. HFD:
[Bifidobacteria, Yadipocyte
size, Yserum and hepatic
cholesterol accumulation

Downloaded from advances.nutrition.org at Fudan University on March 15, 2017


and insulin resistance,
[tight junction proteins
(ZO-1 and occludin),
Yinflammatory markers
(IL-6 and MCP-1)
Cani et al., 2009 (62) C57BL/6mice Prebiotic (oligofructose) vs. nonprebiotic DGGE, qPCR analysis Prebiotic vs. nonprebiotic:
carbohydrates (microcrystalline [Total bacteria count,
cellulose) for 4 wk [Lactobacillus spp.,
[Bifidobacterium spp.,
[Clostridium coccoides–
Eubacterium rectale cluster,
YLPS, [ZO-1 and occludin,
YPAI-1, YNADPHox, YiNOS,
YTLR4, YTNF-a, [GLP-2,
Yhepatic expression of
inflammatory and oxidative
stress markers
Cani et al., 2007 (33) C57BL/6J mice High-fat and carbohydrate-free diet vs. FISH High-fat and carbohydrate-free
normal-diet group for 4 wk diet vs. normal-diet group:
[Liver weight, [Liver TGs,
[TNF-a, [IL-1, [IL-6, [LPS,
YBifidobacterium spp.,
E. rectale/C. coccoides
1
ACC1, acetyl-CoA carboxylase 1; ALT, alanin transferase; BB-12, Bifidobacterium animalis subsp. lactis BB-12; ChREBP, transcription factor carbohydrate-responsive element-
binding protein; DGGE, denaturing gradient gel electrophoresis; FAS, fatty acid synthase; FISH, Fuorescent in situ hybridization; FOS, fructo-oligosaccharide; GLP,
glucagon-like peptide; GT, green tea powder; HF, high prebiotic fiber; HFD, high-fat diet; HP, high protein; iNOS, inducible nitric oxide synthase; LFD, low-fat diet; LGG,
Lactobacillus rhamnosus GG; Lp, Lactobacillus plantarum DSM 15313; MCP-1, monocyte chemoattractant protein-1; NADPHox, NAD(P)H oxidase; NAFLD, nonalcoholic fatty
liver disease; OFS, oligofructosaccharide; PAI-1, plasminogen activator inhibitor 1; PYY, peptide YY; ref, reference; rRNA, ribosomal RNA; SREBP2, sterol regulatory element
binding protein 2; TLR4, Toll-like receptor 4; TNFa, tumor necrosis factor a (gene); ZO-1, zonula occludens-1; [, increased; Y, decreased.

With the use of germ-free animal models, it has been obesity. In normal mice with normal gut microbiota, inac-
shown that the gut microbiota increases energy harvested tive AMPK is associated with decreased FA oxidation in
from dietary polysaccharides and augments key enzymes in skeletal muscle (36). In addition, gut satiety hormones are
hepatic de novo FA biosynthesis, including acetyl-CoA car- affected by the gut microbiome through the gut-brain axis
boxylase (ACC) and FA synthase (FAS), and consequently, (65). Therefore, the gut microbiome influences energy in-
hepatic TG accumulation. On the other hand, the gut take in addition to energy expenditure. Any changes in the
microbiota drives the host to increase LPL activity and hepatic gut microbial taxa can increase gastrointestinal satiety hor-
production of TGs by the inhibition of FIAF (an LPL inhib- mones, hence influencing energy hemostasis (66–68).
itor) in intestinal epithelium cells (35). In addition, germ- Another key function of the intestinal bacteria is to hy-
free mice showed increased AMP-activated protein kinase drolyze polysaccharides and oligosaccharides that are not
(AMPK) activity, which is an intracellular energy sensor in completely catabolized by the host enzymes. Although
the liver and skeletal muscle, leading to increased FA oxida- much is still to be learned about which microbes synthesize
tion and insulin sensitivity and reduced glycogen concentra- and/or metabolize the SCFAs and the roles of these potent
tions. This would protect germ-free mice against diet-induced metabolites, saccharolytic fermentations are major players

244 Mokhtari et al.


in metabolic disorders (69). A wide range of bacterial groups reduced VLDL secretion (83). Maintenance of mice fed an
are able to produce acetate, whereas the production of pro- HFD for 4 wk resulted in a significant increase in
pionate and butyrate seems to be more specific. Akkermansia Bifidobacterium spp. and the E. rectale/Clostridium
municiphila has been known as a major propionate-producing coccoides group, while decreasing the C. coccoides group
bacterium (70) and Eubacterium hallii, Eubacterium rectale, and Bifidobacteria E. rectale. Moreover, fat consumption
Roseburia inulinivorans, Faecalibacterium prausnitzii, Clostridium can produce a wide amount of chylomicrons, which
lavalense, Bacteroides uniformis, and Ruminococcus bromii translocate LPS toward other organs, especially the liver
appear to be responsible for most of butyrate production (84), which can bind to TLR4 on hepatic immune cells
(70–72). Butyrate and propionate at low amounts exert mul- and initiate the inflammatory process. It has been shown
tiple advantageous effects on the host, including the preven- that plasma inflammatory cytokines are positively
tion of colonic carcinogenesis, inflammation, and oxidative correlated with plasma LPS concentrations and negatively
stress; improvement in intestinal barrier function; and stim- correlated with intestinal Bifidobacteria count (85). An
ulation of satiety and lipid oxidation in hepatocytes (73, 74). HFD not only changes the gut microbiota but also
Acetate may be more important for modulating insulin sen- plays a role in the development of steatohepatitis in mice
sitivity and metabolic disease (75) and the development of (56). It has been shown that Lactobacillus gasseri and
diabetes (76). However, more studies are required to under- Lactobacillus taiwanensis increase after consumption of an
stand which microbes produce each SCFA and the relation HFD. These bacteria are bile acid–resistant and probably

Downloaded from advances.nutrition.org at Fudan University on March 15, 2017


this has on liver health and disease. contribute to bile acid deconjugation and reduction in fat
SCFAs are ligands for G-protein–coupled receptor (GPR) absorption. There was a positive significant association
43 expressed on intestinal epithelial cells. The stimulation of between L. gasseri and/or L. taiwanensis DNA and lipid
GPR43 by SCFAs is necessary during immune responses droplets in the liver (R2 = 0.3, P = 0.02). Increased fat
(77). GPR41 is another SCFA receptor. Binding to GPR41 intake simultaneously increases bile acid secretion and
is associated with upregulation of peptide YY (PYY) and bile acid–resistant bacteria. Increased Lactobacillus in HFD
glucagon-like peptide 1 [(GLP1), anorectic hormones] that may affect lipid metabolism through bile acids (86, 87).
have effects on appetite control; therefore, SCFAs are linked A high-protein diet also affects the gut microbiome,
to food intake by activating these receptors (78, 79). Current favoring a potentially pathogenic and proinflammatory mi-
evidence indicates that SCFAs have paradoxical effects on crobiota profile with increased ammonia, phenols, and hy-
hepatic health. SCFAs have been identified as rich sources drogen sulfide concentrations, which induce mucosal
of energy for the host and act as intestinotrophic agents to inflammation and alter the enteric nervous system and in-
promote intestinal absorption of nutrients. On the other testinal motility (88). In addition, the dietary source of pro-
hand, through their receptor GPR43 and other pathways, teins plays a critical role in shaping the gut microbiota and
they reduce gut permeability and invasion of bacterial pro- their ability to produce secondary metabolites, so that
ducts through the portal vein and liver, and thus protect meat protein–fed rats showed a higher relative abundance
the liver from the ensuing damaging inflammation, which of the beneficial genus Lactobacillus but lower amounts of
then results in downregulation of insulin signaling in adi- SCFAs and SCFA-producing bacteria, including Fusobacte-
pose tissue, thereby decreasing fat accumulation (80). rium, Bacteroides, and Prevotella spp. compared with the
Thus, it seems that SCFAs are beneficial for hepatic health soy protein–fed group (89). Overall, studies evaluating the
if the body is receiving a balanced diet, whereas they exag- effects of different dietary patterns on the human gut micro-
gerate steatosis when extra calories are consumed. However, biome are limited; further studies are needed to elucidate
more mechanistic studies are required to understand the more details in this area.
role of each of the SCFAs on NAFLD.
Probiotics and the gut microbiome in NAFLD. Current
Macronutrient intakes and the gut microbiome in studies hypothesize the use of probiotics for modulating
NAFLD. Several studies have shown that high-fat diets dysbiosis in many diseases, including NAFLD. Probiotics
(HFDs) participate in the development of dysbiosis through are beneficial microorganisms when ingested in specified
diet-induced reductions in Bacteroidetes, Lactobacilli, and dosages. Several studies showed that gut microbiota manip-
Bifidobacteria and increases in Firmicutes (81, 82). Thus, ulation by probiotic supplements is associated with reduced
an HFD may contribute to the development of NAFLD at least liver damage, decreased concentrations of LPS, as well as im-
partially through dysbiosis, which may increase intestinal proved aminotransferase concentrations (90–93). A recent
permeability. Furthermore, HFD exposure impairs choline review summarized the studies that show that probiotic sup-
metabolism via dysbiosis. Choline is converted to methylamines plementation in animal models and human studies improve
(dimethylamine, trimethylamine, and trimethylamine-N-oxide) inflammatory status and clinical manifestations in NAFLD
by intestinal microbes with consumption of an HFD. It is (94). Manipulating the gut microbiota with probiotics
associated with a reduction in plasma phosphatidylcholine may improve dysbiosis associated with an HFD. In support
and increases in urinary methylamines. Indeed, changes in of this, consumption of an HFD upregulates the expression
the gut microbiota mimic the effect of a choline-deficient of genes involved in hepatic lipid biosynthesis, and supple-
diet, which leads to accumulation of TGs in hepatocytes and mentation with Lactobacillus curvatus HY7601 and

Gut microbiome and NAFLD: role of nutrition 245


Lactobacillus plantarum KY1032 upregulates the expression could induce special species of bacteria that produce meta-
of genes involved in hepatic FA oxidation (57). Another study bolic products such as butyrate. Butyrate could act as a sig-
showed that increasing tissue n–3 PUFAs in the gut alters the naling molecule in the host that participates in various
gut bacterial composition, which is associated with a decrease signaling pathways (110); thus, products of the gut micro-
in LPS and gut permeability and eventually results in meta- biota could explain part of the role of nutritional factors
bolic endotoxemia and inflammation (95). Overall, although in the gut liver axis and their relation with NAFLD.
there are some exciting possibilities with regard to the use of It has been reported that prebiotic supplementation can
probiotics as a treatment for NAFLD, experimental studies are modulate the effects of HFDs. Prebiotic consumption leads
still required to test the mechanisms of action and clinical to increased Bifidobacteria and total bacteria Bacteroides:
trials are still required to test their efficacy. Prevotella spp. and decreased Firmicutes (53). Cano et al.
(59) reported that Bifidobacterium pseudocatenulatum con-
Prebiotics and intestinal microbiota in NAFLD. In addi- sumption improved glucose tolerance and inflammatory
tion to probiotics being potentially protective in NAFLD, ac- status while decreasing serum LPS and hepatic steatosis in
cumulating data also suggest that there are beneficial effects HFD-fed mice. Another study showed that prebiotic oligo-
of prebiotics in NAFLD. A prebiotic is defined as “a selec- fructose in combination with probiotic Bifidobacterium
tively fermented ingredient that results in specific changes, animalis subsp. lactis BB-12 decreased the ratio of Firmicutes
in the composition and/or activity of the gastrointestinal mi- to Bacteroidetes and improved glycemia. This decrease in

Downloaded from advances.nutrition.org at Fudan University on March 15, 2017


crobiota, thus conferring benefit(s) upon host health” (96). the Firmicutes-to-Bacteroidetes ratio is hypothesized to de-
Prebiotics are naturally found in foods such as onions, as- crease obesity-inducing effects of the microbiome (54). In
paragus, garlic, and leeks and resistant starch (RS) sources addition, oligofructose decreases energy intake, weight
such as legumes, potatoes, and cereals. Prebiotics could gain, and fat mass (54). In this study, prebiotics and pro-
modulate the intestinal microbiota, because they would in- biotics had no effect on increased LPS and inflammatory
crease beneficial bacteria for the host, in particular the factors induced by high-fat and high-sucrose diets (54). A
indigenous Bifidobacteria and Lactobacilli, and inhibit significant increase in the abundance of Bifidobacterium
bacterial activities harmful to host health (97). Furthermore, spp. and Lactobacillus spp. by the administration of inulin-
in the large intestine, inulin-type fructans and RS are type fructo-oligosaccharides (FOSs) has been documented
completely catabolized by the microbiota into SCFAs, (112). Increasing total bacteria counts of Lactobacillus spp.
bacterial biomass, and organic acids such as lactic acid and and Bifidobacterium spp. by prebiotics improved intestinal
gases (carbon dioxide, hydrogen, methane). barrier function and hepatic inflammation and oxidation.
Prebiotics have beneficial effects on the gut microbiota, Furthermore, stimulating the proliferation of these bacteria
serum lipids, and the prevention of hepatic steatosis (61, was associated with a reduction in intestinal endotoxin and
62, 98, 99). In fact, the addition of RS to rodent feed pellets modulated gut mucosal function (113, 114). Lactobacillus
increases Bacteriodetes, Actinobacteria (Bifidobacteria spp.), spp. inhibit proinflammatory responses that result from
and Verrucomicrobia (A. muciniphilia) while decreasing endotoxins by suppressing NF-kB (115).
Firmicutes (100–102). Also, in humans, the administration Inulin-type fructans or FOSs are also able to modify
of RS has been shown to induce phylum-level changes, selec- groups of bacteria other than Bifidobacteria or Lactobacilli.
tively increasing numbers of Actinobacteria and Bacteroidetes Inulin-containing diets result in an increase in bacteria be-
and reducing Firmicutes; at the species level, an increase longing to C. coccoides and E. rectale cluster in animal models
in Bifidobacterium adolescentis, E. rectale, Roseburia spp., (116). In addition, inulin can stimulate the metabolic activity
and R. bromii have been reported (103–107). These var- of Roseburia spp. and E. rectale genera of the Firmicutes phy-
ious bacteria differ significantly in the production of lum (117, 118). Roseburia spp. and E. rectale belong to the
metabolic byproducts and potential interactions with the butyrate-producing bacteria; therefore, they may mediate
host. It has been well established that Bifidobacteria spp., a some of the advantageous properties of prebiotics. Further-
source of lactate and acetate and low amounts of ethanol more, the consumption of inulin, galacto-oligosaccharides,
and formate, are broadly used as probiotics and have and raffinose stimulates F. prausnitzii (119–121). F. prausnitzii,
health-promoting properties because of their potential which belong to the butyrate-producing bacteria, could be
modulating effects on the immune system, metabolism, crucial to gut barrier function and could modulate systemic
and elimination of pathogenic bacteria of the host (108). inflammation (common to NAFLD) (79, 122). All of these
Members of E. rectale, Roseburia, and R. bromii are main proposed actions may play a role in the beneficial effects of
producers of butyrate in the colon (71, 107, 109). It has prebiotics in NAFLD prevention and/or treatment. How-
been shown that increasing butyrate-producing bacteria ever, well-designed clinical trials are required to support
and butyrate production is closely linked with prevention the efficacy of prebiotics during NAFLD.
of the progression from steatosis to hepatocarcinogenesis
and improving hepatic inflammatory and oxidative stress in- Choline and other nutrients and the gut microbiome in
dexes and gut integrity (74, 110, 111). This would appear to NAFLD. Choline deficiency is related to NAFLD pathogen-
indicate that there is a mutualistic relation between the gut esis. Although choline is synthesized in the body, it is con-
microbiota and the host; dietary ingredients such as RS sidered to be an essential nutrient and dietary intake is

246 Mokhtari et al.


required to fulfill its roles in the body (123). Choline plays a use of Lactobacillus rhamnosus GG protects against NAFLD
role in brain function, acetylcholine and neurotransmitter in animals fed a high-fructose diet through increasing the
synthesis, cell signaling, lipid transport, and lipid metabo- proteins involved in gut integrity and decreasing inflamma-
lism (124, 125). Several studies have shown that choline is tory factors and endotoxins. The authors suggested that
critical for the development of hepatic steatosis due to its L. rhamnosus GG may increase occludin and claudin-1 (ef-
contribution to lipid metabolism (125–127). When hepatic fective proteins in gut integrity), which lead to reductions
de novo lipogenesis or TG synthesis from diacylglycerol is in LPS and other toxins translocated to the liver.
greater than the lipid secretion from the liver and/or fat ox- Several recent studies reported that polyphenol com-
idation, hepatic steatosis occurs. Choline is one of the methyl pounds attenuate inflammatory markers and hepatic steato-
group donors in the body that contributes to hepatic synthe- sis grade in patients with NAFLD (142–145). Some of these
sis of phosphatidylcholine (125). Phosphatidylcholine is act through the amelioration of insulin resistance, whereas
required for VLDL synthesis and secretion. Reduced some act through the modulation of the gut microbiome
phosphatidylcholine contributes to TG accumulation in (146, 147). These compounds can reduce pathogen bacteria
the liver and NAFLD (128). Choline-deficient diets disturb and increase lactic acid bacteria, such as some strains of Bi-
the mitochondrial function, resulting in increased hepatic fidobacteria (148). The manipulation of the gut microbiota
lipid accumulation that contributes to NAFLD pathogenesis may be 1 of the mechanisms of action involved in the ben-
(129). Gut bacteria can convert choline to trimethylamine eficial effects of polyphenols on NAFLD. Axling et al. (60)

Downloaded from advances.nutrition.org at Fudan University on March 15, 2017


and reduce choline bioavailability, mimicking choline de- reported that green tea in combination with Lactobacillus
ficiency side effects (130). A reduction in choline bio- plantarum (which metabolizes phenolic acids) in HFD-fed
availability is associated with increased reactive oxygen mice was able to promote growth of Lactobacillus in the in-
species resulting from FFA oxidation, more hepatic lipid testine and to attenuate HFD-induced inflammation and he-
accumulation, and reduced hepatic VLDL excretion, all of patic TG and cholesterol accumulation. Furthermore, there
which contribute to the development of hepatic steatosis was a significant inverse association between the amount of
(125, 126). Choline metabolism is mainly disrupted by 3 Akkermansia and hepatic TG accumulation. A. muciniphila is a
main bacterial phyla in the gut microbiota: Proteobacteria, mucus-degrading bacterium that contributes to the regulation
Firmicutes, and Actinobacteria (130). Spencer et al. (131), of immune responses through upregulation of the expression
in a longitudinal experimental study in healthy women who of some genes involved in intestinal mucus integrity (149).
consumed a rigorously controlled diet, showed that choline
deficiency led to hepatic lipid accumulation. In addition, n–3 FAs and the intestinal microbiota in NAFLD. A low
decreased amounts of Gammaproteobacteria and increased intake of n–3 PUFAs is associated with hepatic steatosis
Erysipelotrichi were directly associated with elevated (150). n–3 PUFAs play an important role in lipid metabo-
accumulation of hepatic fat. This study was the only study, to lism because they can induce FA oxidation by activating
our knowledge, that examined gut microbiota composition PPAR-a and inhibition of lipogenesis by suppressing sterol
in relation to diet and development of NAFLD in humans. regulatory element binding protein 1 (SREBP-1) (151). It
In addition to the evidence of gut dysbiosis in NAFLD, has been shown that n–3 PUFA supplementation, and not
patients with NASH also have dysbiosis, including small in- total calories from fat, alters the gut microbial composition
testinal bacterial overgrowth (23). Bacterial overgrowth en- by enhancing numbers of Lactobacillus and Bifidobacteria
hances the number of bacteria metabolizing choline and spp. in adult mice (152) as well as in aged mice, which
influences hepatic lipid accumulation (125). Pyruvate pro- modulated both gut immunity and oxidative stress (153). In
duced by the gut microbiota from carbohydrate increases support of these data, other studies have shown that changes
metabolites such as acetaldehyde and ethanol. These metab- in gut microbiota composition by n–3 PUFA deficiency can
olites are toxic and associated with gut permeability (132, be modulated by prebiotics such as FOS. FOS consumption
133). Overall, although better designed studies are needed, was associated with increased cecal Bifidobacterium spp. and
there are sufficient data to support that choline and the reduced Roseburia spp. in n–3 PUFA–depleted mice (58).
gut microbiome together play a role in liver disease. Moreover, it has been shown that an increase in lactic acid
High-fructose dietary intake is also considered to be a sig- bacteria, such as Bifidobacterium spp., is related to a reduc-
nificant risk factor in the development of NAFLD (134, tion in cholesterol absorption (154) and an increase in
135). Some studies have indicated that antibiotics, prebi- bile acid deconjugation, which leads to more excretion of
otics, and probiotics are protective factors in fructose- bile acids and elimination of hepatic cholesterol (155).
induced liver damage (136–139). These studies suggested
that gut microbe manipulation may protect from the side ef- Conclusions
fects of high-fructose diets on the liver. The chronic intake In summary, intestinal pathogenic bacteria contribute to
of fructose is associated with bacterial overgrowth, which NAFLD pathogenesis through the following ways: 1) by in-
causes increases in endotoxins in the portal vein, develop- creasing LPS, which binds to TLR4 in hepatic immune cells,
ment of inflammatory responses in the liver, and insulin re- resulting in the activation of the NF-kB pathway and its re-
sistance in hepatocytes, resulting in increased hepatic lipid lated inflammatory pathways, leading to hepatic injury, ne-
accumulation (140, 141). Ritze et al. (55) reported that the crosis, and finally fibrosis; 2) inhibition of FIAF, which

Gut microbiome and NAFLD: role of nutrition 247


increases the activity of LPL and lipogenesis; 3) enhance- 13. Sima H, Hekmatdoost A, Ghaziani T, Alavian SM, Mashayekh A,
ment in polysaccharide digestion and absorption, which in- Zali MR. The prevalence of celiac autoantibodies in hepatitis patients.
Iran J Allergy Asthma Immunol 2010;9:157–62.
creases the production of SCFAs and hepatic lipogenesis;
14. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L,
and 4) conversion of choline into methylamines, which re- Sargent M, Gill SR, Nelson KE, Relman DA. Diversity of the human
duces choline availability and induces fat accumulation intestinal microbial flora. Science 2005;308:1635–8.
and reactive oxygen species production in the liver. 15. Zhu L, Baker SS, Gill C, Liu W, Alkhouri R, Baker RD, Gill SR.
Nutritional factors and the gut microbiota have bilat- Characterization of gut microbiomes in nonalcoholic steatohepatitis
(NASH) patients: a connection between endogenous alcohol and
eral interactions that contribute to the development and/or
NASH. Hepatology 2013;57:601–9.
protection from NAFLD. High-fat, high-protein, and high- 16. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR,
fructose diets and diets that are low in n–3 FAs (Western di- Fernandes GR, Tap J, Bruls T, Batto JM, et al. Enterotypes of the
etary pattern) may contribute to the development of NAFLD human gut microbiome. Nature 2011;473(7346):174–80.
by dysbiosis in the intestinal microbiota. On the other hand, 17. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER,
gut microbiota manipulation by nutritional factors such as Gordon JI. An obesity-associated gut microbiome with increased ca-
pacity for energy harvest. Nature 2006;444:1027–31.
prebiotics, probiotics, and polyphenols can improve charac- 18. Santacruz A, Collado MC, Garcia-Valdes L, Segura MT, Martin-
teristics of NAFLD. Few studies have investigated the effects Lagos JA, Anjos T, Martí-Romero M, Lopez RM, Florido J,
of nutritional factors on the gut microbiota in patients with Campoy C, et al. Gut microbiota composition is associated with body
NAFLD, and in particular, human studies are scarce. Further weight, weight gain and biochemical parameters in pregnant women.

Downloaded from advances.nutrition.org at Fudan University on March 15, 2017


studies are needed to elucidate the exact mechanism(s) of Br J Nutr 2010;104:83–92.
19. Zoetendal EG, Rajilic-Stojanovic M, De Vos WM. High-throughput
action and probable therapeutic options. diversity and functionality analysis of the gastrointestinal tract micro-
biota. Gut 2008;57:1605–15.
Acknowledgments 20. Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M,
ZM and AH contributed to the conception of the article, the Reddy DN. Role of the normal gut microbiota. World J Gastroenterol
2015;21:8787–803.
literature search and interpretation, writing the article, and
21. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, Liang S, Zhang W, Guan Y,
the critical revisions related to important intellectual con- Shen D, et al. A metagenome-wide association study of gut microbiota
tent of the manuscript and approved the final manuscript; in type 2 diabetes. Nature 2012;490:55–60.
and DLG contributed to the final revisions of the manu- 22. Shen J, Obin MS, Zhao L. The gut microbiota, obesity and insulin
script. All authors read and approved the final manuscript. resistance. Mol Aspects Med 2013;34:39–58.
23. Wigg AJ, Roberts-Thomson IC, Dymock RB, McCarthy PJ, Grose RH,
Cummins AG. The role of small intestinal bacterial overgrowth, intes-
References tinal permeability, endotoxaemia, and tumour necrosis factor alpha in
1. Vernon G, Baranova A, Younossi ZM. Systematic review: the epide- the pathogenesis of non-alcoholic steatohepatitis. Gut 2001;48:206–11.
miology and natural history of non-alcoholic fatty liver disease and
24. Mouzaki M, Comelli EM, Arendt BM, Bonengel J, Fung SK,
non-alcoholic steatohepatitis in adults. Aliment Pharmacol Ther
Fischer SE, McGilvray ID, Allard JP. Intestinal microbiota in patients
2011;34:274–85.
with nonalcoholic fatty liver disease. Hepatology 2013;58:120–7.
2. Tiniakos DG, Vos MB, Brunt EM. Nonalcoholic fatty liver disease:
25. Le Roy T, Llopis M, Lepage P, Bruneau A, Rabot S, Bevilacqua C,
pathology and pathogenesis. Annu Rev Pathol 2010;5:145–71.
Martin P, Philippe C, Walker F, Bado A, et al. Intestinal microbiota
3. Angulo P. Nonalcoholic fatty liver disease. N Engl J Med 2002;346: determines development of non-alcoholic fatty liver disease in mice.
1221–31. Gut 2013;62:1787–94.
4. Bedogni G, Miglioli L, Masutti F, Tiribelli C, Marchesini G, Bellentani S. 26. Eslamparast T, Poustchi H, Zamani F, Sharafkhah M, Malekzadeh R,
Prevalence of and risk factors for nonalcoholic fatty liver disease: the Hekmatdoost A. Synbiotic supplementation in nonalcoholic fatty liver
Dionysos Nutrition and Liver Study. Hepatology 2005;42:44–52. disease: a randomized, double-blind, placebo-controlled pilot study.
5. Fan JG, Saibara T, Chitturi S, Kim BI, Sung JJ, Chutaputti A. What are Am J Clin Nutr 2014;99:535–42.
the risk factors and settings for non-alcoholic fatty liver disease in 27. Yari Z, Rahimlou M, Eslamparast T, Ebrahimi-Daryani N, Poustchi H,
Asia-Pacific? J Gastroenterol Hepatol 2007;22:794–800. Hekmatdoost A. Flaxseed supplementation in non-alcoholic fatty liver
6. Wilfred de Alwis NM, Day CP. Genetics of alcoholic liver disease and disease: a pilot randomized, open labeled, controlled study. Int J Food
nonalcoholic fatty liver disease. Semin Liver Dis 2007;27:44–54. Sci Nutr 2016;67:461–9.
7. Ghaemi A, Taleban FA, Hekmatdoost A, Rafiei A, Hosseini V, Amiri Z, 28. Eslamparast T, Eghtesad S, Poustchi H, Hekmatdoost A. Recent ad-
Homayounfar R, Fakheri H. How much weight loss is effective on vances in dietary supplementation, in treating non-alcoholic fatty liver
nonalcoholic fatty liver disease? Hepat Mon 2013;13:e15227. disease. World J Hepatol 2015;7:204–12.
8. Schnabl B, Brenner DA. Interactions between the intestinal micro- 29. Shavakhi A, Minakari M, Firouzian H, Assali R, Hekmatdoost A,
biome and liver diseases. Gastroenterology 2014;146:1513–24. Ferns G. Effect of a probiotic and metformin on liver aminotrans-
9. Balmer ML, Slack E, de Gottardi A, Lawson MA, Hapfelmeier S, ferases in non-alcoholic steatohepatitis: a double blind randomized
Miele L, Grieco A, Van Vlierberghe H, Fahrner R, Patuto N, et al. The clinical trial. Int J Prev Med 2013;4:531–7.
liver may act as a firewall mediating mutualism between the host and its 30. Rahimlou M, Ahmadnia H, Hekmatdoost A. Dietary supplements and
gut commensal microbiota. Sci Transl Med 2014;6:237ra66. pediatric non-alcoholic fatty liver disease: present and the future.
10. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune World J Hepatol 2015;7:2597–602.
responses during health and disease. Nat Rev Immunol 2009;9:313–23. 31. Brun P, Castagliuolo I, Di Leo V, Buda A, Pinzani M, Palu G,
11. Racanelli V, Rehermann B. The liver as an immunological organ. Martines D. Increased intestinal permeability in obese mice: new
Hepatology 2006;43(2, Suppl 1):S54–62. evidence in the pathogenesis of nonalcoholic steatohepatitis. Am J
12. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Physiol Gastrointest Liver Physiol 2007;292:G518–25.
Bewtra M, Knights D, Walters WA, Knight R, et al. Linking long-term 32. Soares JB, Pimentel-Nunes P, Roncon-Albuquerque R, Leite-
dietary patterns with gut microbial enterotypes. Science 2011;334: Moreira A. The role of lipopolysaccharide/Toll-like receptor 4 signal-
105–8. ing in chronic liver diseases. Hepatol Int 2010;4:659–72.

248 Mokhtari et al.


33. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, 50. Hekmatdoost A, Mirshafiey A, Feizabadi MM, Djazayeri A. Polyunsat-
Neyrinck AM, Fava F, Tuohy KM, Chabo C, et al. Metabolic endo- urated fatty acids, microflora and colitis. Ann Nutr Metab 2009;55:325.
toxemia initiates obesity and insulin resistance. Diabetes 2007;56: 51. Hekmatdoost A, Wu X, Morampudi V, Innis SM, Jacobson K. Dietary
1761–72. oils modify the host immune response and colonic tissue damage
34. Su GL. Lipopolysaccharides in liver injury: molecular mechanisms of following Citrobacter rodentium infection in mice. Am J Physiol
Kupffer cell activation. Am J Physiol Gastrointest Liver Physiol 2002; Gastrointest Liver Physiol 2013;304:G917–28.
283:G256–65. 52. Carmody RN, Gerber GK, Luevano JM, Gatti DM, Somes L,
35. Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Svenson KL, Turnbaugh PJ. Diet dominates host genotype in shaping
Semenkovich CF, Gordon JI. The gut microbiota as an environmental the murine gut microbiota. Cell Host Microbe 2015;17:72–84.
factor that regulates fat storage. Proc Natl Acad Sci USA 2004;101: 53. Saha DC, Reimer RA. Long-term intake of a high prebiotic fiber diet
15718–23. but not high protein reduces metabolic risk after a high fat challenge
36. Backhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms and uniquely alters gut microbiota and hepatic gene expression. Nutr
underlying the resistance to diet-induced obesity in germ-free mice. Res 2014;34:789–96.
Proc Natl Acad Sci USA 2007;104:979–84. 54. Bomhof MR, Saha DC, Reid DT, Paul HA, Reimer RA. Combined ef-
37. Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, fects of oligofructose and Bifidobacterium animalis on gut microbiota
Thaiss CA, Kau AL, Eisenbarth SC, Jurczak MJ, et al. Inflammasome- and glycemia in obese rats. Obesity (Silver Spring) 2014;22:763–71.
mediated dysbiosis regulates progression of NAFLD and obesity. Na- 55. Ritze Y, Bárdos G, Claus A, Ehrmann V, Bergheim I, Schwiertz A,
ture 2012;482:179–85. Bischoff SC. Lactobacillus rhamnosus GG protects against non-
38. Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ, alcoholic fatty liver disease in mice. PLoS One 2014;9:e80169.
Peaper DR, Bertin J, Eisenbarth SC, Gordon JI, et al. NLRP6 in- 56. Zeng H, Liu J, Jackson MI, Zhao FQ, Yan L, Combs GF Jr. Fatty liver
accompanies an increase in Lactobacillus species in the hind gut of

Downloaded from advances.nutrition.org at Fudan University on March 15, 2017


flammasome regulates colonic microbial ecology and risk for colitis.
Cell 2011;145:745–57. C57BL/6 mice fed a high-fat diet. J Nutr 2013;143:627–31.
39. Lee HJ, Yeon JE, Ko EJ, Yoon EL, Suh SJ, Kang K, Kim HR, Kang SH, 57. Park DY, Ahn YT, Park SH, Huh CS, Yoo SR, Yu R, Sung MK,
Yoo YJ, Je J, et al. Peroxisome proliferator-activated receptor-delta ag- McGregor RA, Choi MS. Supplementation of Lactobacillus curvatus
onist ameliorated inflammasome activation in nonalcoholic fatty liver HY7601 and Lactobacillus plantarum KY1032 in diet-induced obese
disease. World J Gastroenterol 2015;21:12787–99. mice is associated with gut microbial changes and reduction in obe-
40. Sui YH, Luo WJ, Xu QY, Hua J. Dietary saturated fatty acid and sity. PLoS One 2013;8:e59470.
58. Pachikian BD, Essaghir A, Demoulin JB, Catry E, Neyrinck AM,
polyunsaturated fatty acid oppositely affect hepatic NOD-like receptor
Dewulf EM, Sohet FM, Portois L, Clerbaux LA, Carpentier YA, et al.
protein 3 inflammasome through regulating nuclear factor-kappa B
Prebiotic approach alleviates hepatic steatosis: implication of fatty acid
activation. World J Gastroenterol 2016;22:2533–44.
oxidative and cholesterol synthesis pathways. Mol Nutr Food Res
41. Volynets V, Kuper MA, Strahl S, Maier IB, Spruss A, Wagnerberger S,
2013;57:347–59.
Königsrainer A, Bischoff SC, Bergheim I. Nutrition, intestinal per-
59. Cano PG, Santacruz A, Trejo FM, Sanz Y. Bifidobacterium CECT 7765
meability, and blood ethanol levels are altered in patients with non-
improves metabolic and immunological alterations associated with obe-
alcoholic fatty liver disease (NAFLD). Dig Dis Sci 2012;57:1932–41.
sity in high-fat diet-fed mice. Obesity (Silver Spring) 2013;21:2310–21.
42. Kirpich IA, Feng W, Wang Y, Liu Y, Barker DF, Barve SS, McClain CJ.
60. Axling U, Olsson C, Xu J, Fernandez C, Larsson S, Strom K, Ahrné S,
The type of dietary fat modulates intestinal tight junction integrity,
Holm C, Molin G, Berger K. Green tea powder and Lactobacillus
gut permeability, and hepatic Toll-like receptor expression in a mouse
plantarum affect gut microbiota, lipid metabolism and inflammation
model of alcoholic liver disease. Alcohol Clin Exp Res 2012;36:835–46.
in high-fat fed C57BL/6J mice. Nutr Metab (Lond) 2012;9:105.
43. Harris K, Kassis A, Major G, Chou CJ. Is the gut microbiota a new
61. Neyrinck AM, Possemiers S, Druart C, Van de Wiele T, De Backer F,
factor contributing to obesity and its metabolic disorders? J Obes
Cani PD, Larondelle Y, Delzenne NM. Prebiotic effects of wheat
2012;2012:879151. arabinoxylan related to the increase in Bifidobacteria, Roseburia and
44. Del Chierico F, Nobili V, Vernocchi P, Russo A, De Stefanis C,
Bacteroides/Prevotella in diet-induced obese mice. PLoS One 2011;
Gnani D, Furlanello C, Zandonà A, Paci P, Capuani G, et al. Gut 6:e20944.
microbiota profiling of pediatric NAFLD and obese patients unveiled 62. Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O,
by an integrated meta-omics based approach. Hepatology 2016 Mar Geurts L, Naslain D, Neyrinck A, Lambert DM, et al. Changes in gut
29 (Epub ahead of print; DOI: 10.1002/hep.28572). microbiota control inflammation in obese mice through a mechanism
45. Wong VW, Tse CH, Lam TT, Wong GL, Chim AM, Chu WC, involving GLP-2-driven improvement of gut permeability. Gut 2009;
Yeung DK, Law PT, Kwan HS, Yu J, et al. Molecular characterization of 58:1091–103.
the fecal microbiota in patients with nonalcoholic steatohepatitis—a 63. Fabbrini E, Sullivan S, Klein S. Obesity and nonalcoholic fatty liver
longitudinal study. PLoS One 2013;8:e62885. disease: biochemical, metabolic, and clinical implications. Hepatology
46. Raman M, Ahmed I, Gillevet PM, Probert CS, Ratcliffe NM, Smith S, 2010;51:679–89.
Greenwood R, Sikaroodi M, Lam V, Crotty P, et al. Fecal microbiome 64. Jumpertz R, Le DS, Turnbaugh PJ, Trinidad C, Bogardus C, Gordon JI,
and volatile organic compound metabolome in obese humans with Krakoff J. Energy-balance studies reveal associations between gut mi-
nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2013;11 crobes, caloric load, and nutrient absorption in humans. Am J Clin
(7):868–75, e1–3. Nutr 2011;94:58–65.
47. Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, 65. Perry RJ, Peng L, Barry NA, Cline GW, Zhang D, Cardone RL,
Griffin NW, Lombard V, Henrissat B, Bain JR, et al. Gut microbiota Petersen KF, Kibbey RG, Goodman AL, Shulman GI. Acetate
from twins discordant for obesity modulate metabolism in mice. mediates a microbiome-brain-beta-cell axis to promote metabolic
Science 2013;341:1241214. syndrome. Nature 2016;534:213–7.
48. Gorzelak MA, Gill SK, Tasnim N, Ahmadi-Vand Z, Jay M, Gibson DL. 66. Musso G, Gambino R, Cassader M. Gut microbiota as a regulator of
Methods for improving human gut microbiome data by reducing energy homeostasis and ectopic fat deposition: mechanisms and im-
variability through sample processing and storage of stool. PLoS One plications for metabolic disorders. Curr Opin Lipidol 2010;21:76–83.
2015;10:e0134802. 67. Cani PD, Lecourt E, Dewulf EM, Sohet FM, Pachikian BD, Naslain D,
49. Hekmatdoost A, Feizabadi MM, Djazayery A, Mirshafiey A, De Backer F, Neyrinck AM, Delzenne NM. Gut microbiota fermen-
Eshraghian MR, Yeganeh SM, Sedaghat R, Jacobson K. The effect of tation of prebiotics increases satietogenic and incretin gut peptide
dietary oils on cecal microflora in experimental colitis in mice. Indian production with consequences for appetite sensation and glucose re-
J Gastroenterol 2008;27:186–9. sponse after a meal. Am J Clin Nutr 2009;90:1236–43.

Gut microbiome and NAFLD: role of nutrition 249


68. Parnell JA, Reimer RA. Weight loss during oligofructose supplemen- 86. de la Serre CB, Ellis CL, Lee J, Hartman AL, Rutledge JC,
tation is associated with decreased ghrelin and increased peptide YY in Raybould HE. Propensity to high-fat diet-induced obesity in rats is as-
overweight and obese adults. Am J Clin Nutr 2009;89:1751–9. sociated with changes in the gut microbiota and gut inflammation.
69. Puertollano E, Kolida S, Yaqoob P. Biological significance of short- Am J Physiol Gastrointest Liver Physiol 2010;299:G440–8.
chain fatty acid metabolism by the intestinal microbiome. Curr 87. Tremaroli V, Backhed F. Functional interactions between the gut
Opin Clin Nutr Metab Care 2014;17:139–44. microbiota and host metabolism. Nature 2012;489:242–9.
70. Morrison DJ, Preston T. Formation of short chain fatty acids by the 88. Yao CK, Muir JG, Gibson PR. Review article: insights into colonic
gut microbiota and their impact on human metabolism. Gut Mi- protein fermentation, its modulation and potential health implica-
crobes 2016;7:189–200. tions. Aliment Pharmacol Ther 2016;43:181–96.
71. Louis P, Young P, Holtrop G, Flint HJ. Diversity of human colonic 89. Zhu Y, Lin X, Li H, Li Y, Shi X, Zhao F, Xu X, Li C, Zhou G. Intake of
butyrate-producing bacteria revealed by analysis of the butyryl-CoA: meat proteins substantially increased the relative abundance of genus
acetate CoA-transferase gene. Environ Microbiol 2010;12:304–14. lactobacillus in rat feces. PLoS One 2016;11:e0152678.
72. Takahashi K, Nishida A, Fujimoto T, Fujii M, Shioya M, Imaeda H, 90. Wong VW, Won GL, Chim AM, Chu WC, Yeung DK, Li KC,
Inatomi O, Bamba S, Sugimoto M, Andoh A. Reduced abundance of Chan HL. Treatment of nonalcoholic steatohepatitis with probiotics: a
butyrate-producing bacteria species in the fecal microbial community proof-of-concept study. Ann Hepatol 2013;12:256–62.
in Crohn’s disease. Digestion 2016;93:59–65. 91. Everard A, Matamoros S, Geurts L, Delzenne NM, Cani PD. Sac-
73. Hosseini E, Grootaert C, Verstraete W, Van de Wiele T. Propionate charomyces boulardii administration changes gut microbiota and
as a health-promoting microbial metabolite in the human gut. Nutr reduces hepatic steatosis, low-grade inflammation, and fat mass in
Rev 2011;69:245–58. obese and type 2 diabetic db/db mice. MBio 2014;5:e01011–4.
74. Hamer HM, Jonkers D, Venema K, Vanhoutvin S, Troost F, 92. Nardone G, Compare D, Liguori E, Di Mauro V, Rocco A, Barone M,
Napoli A, Lapi D, Iovene MR, Colantuoni A. Protective effects of

Downloaded from advances.nutrition.org at Fudan University on March 15, 2017


Brummer RJ. Review article: the role of butyrate on colonic function.
Aliment Pharmacol Ther 2008;27:104–19. Lactobacillus paracasei F19 in a rat model of oxidative and metabolic
75. van der Beek CM, Canfora EE, Lenaerts K, Troost FJ, Olde hepatic injury. Am J Physiol Gastrointest Liver Physiol 2010;299:
Damink SW, Holst JJ, Masclee AA, Dejong CH, Blaak EE. Distal, G669–76.
not proximal, colonic acetate infusions promote fat oxidation and 93. Compare D, Coccoli P, Rocco A, Nardone OM, De Maria S, Carteni M,
improve metabolic markers in overweight/obese men. Clin Sci (Lond) Nardone G. Gut–liver axis: the impact of gut microbiota on nonalco-
2016;130:2073–82. holic fatty liver disease. Nutr Metab Cardiovasc Dis 2012;22:471–6.
76. Brown K, Godovannyi A, Ma C, Zhang Y, Ahmadi-Vand Z, Dai C, 94. Eslamparast T, Eghtesad S, Hekmatdoost A, Poustchi H. Probiotics and
Gorzelak MA, Chan Y, Chan JM, Lochner A, et al. Prolonged anti- nonalcoholic fatty liver disease. Middle East J Dig Dis 2013;5:129–36.
95. Kaliannan K, Wang B, Li XY, Kim KJ, Kang JX. A host-microbiome
biotic treatment induces a diabetogenic intestinal microbiome that
interaction mediates the opposing effects of omega-6 and omega-3
accelerates diabetes in NOD mice. ISME J 2016;10:321–32.
fatty acids on metabolic endotoxemia. Sci Rep 2015;5:11276.
77. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D,
96. Roberfroid M, Gibson GR, Hoyles L, McCartney AL, Rastall R,
Schilter HC, Rolph MS, Mackay F, Artis D, et al. Regulation of in-
Rowland I, Wolvers D, Watzl B, Szajewska H, Stahl B, et al. Prebiotic
flammatory responses by gut microbiota and chemoattractant re-
effects: metabolic and health benefits. Br J Nutr 2010;104(Suppl 2):
ceptor GPR43. Nature 2009;461:1282–6.
S1–63.
78. Samuel BS, Shaito A, Motoike T, Rey FE, Backhed F, Manchester JK,
97. Blaut M. Relationship of prebiotics and food to intestinal microflora.
Hammer RE, Williams SC, Crowley J, Yanagisawa M, et al. Effects of
Eur J Nutr 2002;41:I11–6.
the gut microbiota on host adiposity are modulated by the short-chain
98. Tuohy KM, Probert HM, Smejkal CW, Gibson GR. Using probiotics
fatty-acid binding G protein-coupled receptor, Gpr41. Proc Natl Acad
and prebiotics to improve gut health. Drug Discov Today 2003;8:692–
Sci USA 2008;105:16767–72.
700.
79. Sleeth ML, Thompson EL, Ford HE, Zac-Varghese SE, Frost G. Free
99. Daubioul CA, Taper HS, De Wispelaere LD, Delzenne NM. Dietary
fatty acid receptor 2 and nutrient sensing: a proposed role for fibre, oligofructose lessens hepatic steatosis, but does not prevent hyper-
fermentable carbohydrates and short-chain fatty acids in appetite reg- triglyceridemia in obese zucker rats. J Nutr 2000;130:1314–9.
ulation. Nutr Res Rev 2010;23:135–45. 100. Birt DF, Boylston T, Hendrich S, Jane JL, Hollis J, Li L, McClelland J,
80. Zhu L, Baker RD, Baker SS. Gut microbiome and nonalcoholic fatty Moore S, Phillips GJ, Rowling M, et al. Resistant starch: promise for
liver diseases. Pediatr Res 2015;77:245–51. improving human health. Adv Nutr 2013;4(6):587–601.
81. Hildebrandt MA, Hoffmann C, Sherrill-Mix SA, Keilbaugh SA, 101. Keenan MJ, Zhou J, Hegsted M, Pelkman C, Durham HA, Coulon DB,
Hamady M, Chen YY, Knight R, Ahima RS, Bushman F, Wu GD. Martin RJ. Role of resistant starch in improving gut health, adiposity,
High-fat diet determines the composition of the murine gut micro- and insulin resistance. Adv Nutr 2015;6(2):198–205.
biome independently of obesity. Gastroenterology 2009;137(5): 102. Zaman SA, Sarbini SR. The potential of resistant starch as a prebiotic.
1716–24, e1–2. Crit Rev Biotechnol 2016;36:578–84.
82. Turnbaugh PJ, Backhed F, Fulton L, Gordon JI. Diet-induced obesity 103. Flint HJ, Scott KP, Louis P, Duncan SH. The role of the gut microbiota
is linked to marked but reversible alterations in the mouse distal gut in nutrition and health. Nat Rev Gastroenterol Hepatol 2012;9:577–89.
microbiome. Cell Host Microbe 2008;3:213–23. 104. Martínez I, Kim J, Duffy PR, Schlegel VL, Walter J. Resistant starches
83. Dumas ME, Barton RH, Toye A, Cloarec O, Blancher C, Rothwell A, types 2 and 4 have differential effects on the composition of the fecal
Fearnside J, Tatoud R, Blanc V, Lindon JC, et al. Metabolic profiling microbiota in human subjects. PLoS One 2010;5:e15046.
reveals a contribution of gut microbiota to fatty liver phenotype in 105. Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, Ze X,
insulin-resistant mice. Proc Natl Acad Sci USA 2006;103:12511–6. Brown D, Stares MD, Scott P, Bergerat A, et al. Dominant and diet-
84. Vreugdenhil AC, Rousseau CH, Hartung T, Greve JW, van’t Veer C, responsive groups of bacteria within the human colonic microbiota.
Buurman WA. Lipopolysaccharide (LPS)-binding protein mediates ISME J 2011;5:220–30.
LPS detoxification by chylomicrons. J Immunol 2003;170(3):1399– 106. Abell GC, Cooke CM, Bennett CN, Conlon MA, McOrist AL. Phy-
405. lotypes related to Ruminococcus bromii are abundant in the large
85. Cani PD, Neyrinck AM, Fava F, Knauf C, Burcelin RG, Tuohy KM, bowel of humans and increase in response to a diet high in resistant
Gibson GR, Delzenne NM. Selective increases of bifidobacteria in gut starch. FEMS Microbiol Ecol 2008;66:505–15.
microflora improve high-fat-diet-induced diabetes in mice through a 107. Ze X, Duncan SH, Louis P, Flint HJ. Ruminococcus bromii is a key-
mechanism associated with endotoxaemia. Diabetologia 2007;50: stone species for the degradation of resistant starch in the human
2374–83. colon. ISME J 2012;6:1535–43.

250 Mokhtari et al.


108. O’Callaghan A, Van Sinderen D. Bifidobacteria and their role as 129. Teodoro JS, Rolo AP, Duarte FV, Simoes AM, Palmeira CM. Differential
members of the human gut microbiota. Front Microbiol 2016;7:925. alterations in mitochondrial function induced by a choline-deficient
109. Flint HJ, Duncan SH, Scott KP, Louis P. Interactions and competition diet: understanding fatty liver disease progression. Mitochondrion
within the microbial community of the human colon: links between 2008;8:367–76.
diet and health. Environ Microbiol 2007;9:1101–11. 130. Martínez-del Campo A, Bodea S, Hamer HA, Marks JA, Haiser HJ,
110. Endo H, Niioka M, Kobayashi N, Tanaka M, Watanabe T. Butyrate- Turnbaugh PJ, Balskus EP. Characterization and detection of a widely
producing probiotics reduce nonalcoholic fatty liver disease progres- distributed gene cluster that predicts anaerobic choline utilization by
sion in rats: new insight into the probiotics for the gut-liver axis. human gut bacteria. MBio 2015;6.
PLoS One 2013;8:e63388. 131. Spencer MD, Hamp TJ, Reid RW, Fischer LM, Zeisel SH, Fodor AA.
111. Jin CJ, Sellmann C, Engstler AJ, Ziegenhardt D, Bergheim I. Supple- Association between composition of the human gastrointestinal mi-
mentation of sodium butyrate protects mice from the development of crobiome and development of fatty liver with choline deficiency.
non-alcoholic steatohepatitis (NASH). Br J Nutr 2015;114:1745–55. Gastroenterology 2011;140:976–86.
112. Macfarlane S, Macfarlane GT, Cummings JH. Review article: prebiotics 132. Nair S, Cope K, Risby TH, Diehl AM. Obesity and female gender
in the gastrointestinal tract. Aliment Pharmacol Ther 2006;24:701–14. increase breath ethanol concentration: potential implications for the
113. Griffiths EA, Duffy LC, Schanbacher FL, Qiao H, Dryja D, Leavens A, pathogenesis of nonalcoholic steatohepatitis. Am J Gastroenterol
Rossman J, Rich G, Dirienzo D, Ogra PL. In vivo effects of bifido- 2001;96:1200–4. Erratum in: Am J Gastroenterol 2001;23:2809.
bacteria and lactoferrin on gut endotoxin concentration and mucosal 133. Cope K, Risby T, Diehl AM. Increased gastrointestinal ethanol pro-
immunity in Balb/c mice. Dig Dis Sci 2004;49:579–89. duction in obese mice: implications for fatty liver disease pathogen-
114. Hevia A, Delgado S, Sanchez B, Margolles A. Molecular players in- esis. Gastroenterology 2000;119:1340–7.
volved in the interaction between beneficial bacteria and the immune 134. Tetri LH, Basaranoglu M, Brunt EM, Yerian LM, Neuschwander-
system. Front Microbiol 2015;6:1285. Tetri BA. Severe NAFLD with hepatic necroinflammatory changes in

Downloaded from advances.nutrition.org at Fudan University on March 15, 2017


115. Petrof EO, Kojima K, Ropeleski MJ, Musch MW, Tao Y, De Simone C, mice fed trans fats and a high-fructose corn syrup equivalent. Am J
Chang EB. Probiotics inhibit nuclear factor-kappaB and induce heat Physiol Gastrointest Liver Physiol 2008;295:G987–95.
shock proteins in colonic epithelial cells through proteasome inhibi- 135. Ouyang X, Cirillo P, Sautin Y, McCall S, Bruchette JL, Diehl AM,
tion. Gastroenterology 2004;127:1474–87. Johnson RJ, Abdelmalek MF. Fructose consumption as a risk factor for
116. Kleessen B, Hartmann L, Blaut M. Oligofructose and long-chain inu- non-alcoholic fatty liver disease. J Hepatol 2008;48:993–9.
lin: influence on the gut microbial ecology of rats associated with a 136. Bergheim I, Weber S, Vos M, Kramer S, Volynets V, Kaserouni S,
human faecal flora. Br J Nutr 2001;86:291–300. McClain CJ, Bischoff SC. Antibiotics protect against fructose-induced
117. Duncan SH, Belenguer A, Holtrop G, Johnstone AM, Flint HJ, hepatic lipid accumulation in mice: role of endotoxin. J Hepatol 2008;
Lobley GE. Reduced dietary intake of carbohydrates by obese subjects 48:983–92.
results in decreased concentrations of butyrate and butyrate- 137. Busserolles J, Gueux E, Rock E, Demigne C, Mazur A, Rayssiguier Y.
producing bacteria in feces. Appl Environ Microbiol 2007;73:1073–8. Oligofructose protects against the hypertriglyceridemic and pro-
118. Rivière A, Selak M, Lantin D, Leroy F, De Vuyst L. Bifidobacteria and oxidative effects of a high fructose diet in rats. J Nutr 2003;133:1903–8.
butyrate-producing colon bacteria: importance and strategies for their 138. Suganthi R, Rajamani S, Ravichandran MK, Anuradha CV. Effect of
stimulation in the human gut. Front Microbiol 2016;7:979. food seasoning spices mixture on biomarkers of oxidative stress in tis-
119. Ramirez-Farias C, Slezak K, Fuller Z, Duncan A, Holtrop G, Louis P. Effect sues of fructose-fed insulin-resistant rats. J Med Food 2007;10:149–53.
of inulin on the human gut microbiota: stimulation of Bifidobacterium 139. Li YC, Hsieh CC. Lactoferrin dampens high-fructose corn syrup-
adolescentis and Faecalibacterium prausnitzii. Br J Nutr 2009;101:541–50. induced hepatic manifestations of the metabolic syndrome in a mu-
120. Davis LM, Martinez I, Walter J, Goin C, Hutkins RW. Barcoded py- rine model. PLoS One 2014;9:e97341.
rosequencing reveals that consumption of galactooligosaccharides 140. Sellmann C, Priebs J, Landmann M, Degen C, Engstler AJ, Jin CJ,
results in a highly specific bifidogenic response in humans. PLoS One Gärttner S, Spruss A, Huber O, Bergheim I. Diets rich in fructose, fat
2011;6:e25200. or fructose and fat alter intestinal barrier function and lead to the
121. Fernando WM, Hill JE, Zello GA, Tyler RT, Dahl WJ, Van Kessel AG. development of nonalcoholic fatty liver disease over time. J Nutr
Diets supplemented with chickpea or its main oligosaccharide com- Biochem 2015;26:1183–92.
ponent raffinose modify faecal microbial composition in healthy 141. Spruss A, Bergheim I. Dietary fructose and intestinal barrier: potential
adults. Benef Microbes 2010;1:197–207. risk factor in the pathogenesis of nonalcoholic fatty liver disease. J
122. Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermúdez- Nutr Biochem 2009;20:657–62.
Humarán LG, Gratadoux J-J, Blugeon S, Bridonneau C, Furet JP, 142. Askari F, Rashidkhani B, Hekmatdoost A. Cinnamon may have
Corthier G, et al. Faecalibacterium prausnitzii is an anti-inflammatory therapeutic benefits on lipid profile, liver enzymes, insulin resistance,
commensal bacterium identified by gut microbiota analysis of Crohn and high-sensitivity C-reactive protein in nonalcoholic fatty liver
disease patients. Proc Natl Acad Sci USA 2008;105:16731–6. disease patients. Nutr Res 2014;34:143–8.
123. Zeisel SH, da Costa KA. Choline: an essential nutrient for public 143. Faghihzadeh F, Adibi P, Hekmatdoost A. The effects of resveratrol
health. Nutr Rev 2009;67:615–23. supplementation on cardiovascular risk factors in patients with non-
124. Zeisel SH. Choline: an essential nutrient for humans. Nutrition 2000; alcoholic fatty liver disease: a randomised, double-blind, placebo-
16:669–71. controlled study. Br J Nutr 2015;114:796–803.
125. Sherriff JL, O’Sullivan TA, Properzi C, Oddo JL, Adams LA. Choline, 144. Faghihzadeh F, Adibi P, Rafiei R, Hekmatdoost A. Resveratrol sup-
its potential role in nonalcoholic fatty liver disease, and the case for plementation improves inflammatory biomarkers in patients with
human and bacterial genes. Adv Nutr 2016;7(1):5–13. nonalcoholic fatty liver disease. Nutr Res 2014;34:837–43.
126. Corbin KD, Zeisel SH. Choline metabolism provides novel insights 145. Faghihzadeh F, Hekmatdoost A, Adibi P. Resveratrol and liver: a
into nonalcoholic fatty liver disease and its progression. Curr Opin systematic review. J Res Med Sci 2015;20(8):797–810.
Gastroenterol 2012;28:159–65. 146. Eslamparast T, Zamani F, Hekmatdoost A, Sharafkhah M, Eghtesad S,
127. Buchman AL, Dubin MD, Moukarzel AA, Jenden DJ, Roch M, Malekzadeh R, Poustchi H. Effects of synbiotic supplementation
Rice KM, Gornbein J, Ament ME. Choline deficiency: a cause of on insulin resistance in subjects with the metabolic syndrome: a
hepatic steatosis during parenteral nutrition that can be reversed with randomised, double-blind, placebo-controlled pilot study. Br J Nutr
intravenous choline supplementation. Hepatology 1995;22:1399–403. 2014;112:438–45.
128. Noga AA, Zhao Y, Vance DE. An unexpected requirement for phos- 147. Ghorbani Z, Hekmatdoost A, Mirmiran P. Anti-hyperglycemic and in-
phatidylethanolamine N-methyltransferase in the secretion of very sulin sensitizer effects of turmeric and its principal constituent cur-
low density lipoproteins. J Biol Chem 2002;277:42358–65. cumin. Int J Endocrinol Metab 2014;12:e18081.

Gut microbiome and NAFLD: role of nutrition 251


148. Lee HC, Jenner AM, Low CS, Lee YK. Effect of tea phenolics and their 152. Ghosh S, DeCoffe D, Brown K, Rajendiran E, Estaki M, Dai C,
aromatic fecal bacterial metabolites on intestinal microbiota. Res Yip A, Gibson DL. Fish oil attenuates omega-6 polyunsaturated
Microbiol 2006;157:876–84. fatty acid-induced dysbiosis and infectious colitis but impairs
149. Derrien M, Van Baarlen P, Hooiveld G, Norin E, Muller M, de LPS dephosphorylation activity causing sepsis. PLoS One 2013;8:
Vos WM. Modulation of mucosal immune response, tolerance, and e55468.
proliferation in mice colonized by the mucin-degrader Akkermansia 153. Huang EY, Leone VA, Devkota S, Wang Y, Brady MJ, Chang EB.
muciniphila. Front Microbiol 2011;2:166. Composition of dietary fat source shapes gut microbiota architecture
150. Zelber-Sagi S, Nitzan-Kaluski D, Goldsmith R, Webb M, Blendis L, and alters host inflammatory mediators in mouse adipose tissue.
Halpern Z, Oren R. Long term nutritional intake and the risk for non- JPEN J Parenter Enteral Nutr 2013;37:746–54.
alcoholic fatty liver disease (NAFLD): a population based study. J 154. Pereira DI, Gibson GR. Cholesterol assimilation by lactic acid bacteria
Hepatol 2007;47:711–7. and bifidobacteria isolated from the human gut. Appl Environ Mi-
151. Araya J, Rodrigo R, Videla LA, Thielemann L, Orellana M, Pettinelli P, crobiol 2002;68:4689–93.
Poniachik J. Increase in long-chain polyunsaturated fatty acid n-6/n-3 155. St-Onge MP, Farnworth ER, Jones PJ. Consumption of fermented and
ratio in relation to hepatic steatosis in patients with non-alcoholic nonfermented dairy products: effects on cholesterol concentrations
fatty liver disease. Clin Sci (Lond) 2004;106:635–43. and metabolism. Am J Clin Nutr 2000;71:674–81.

Downloaded from advances.nutrition.org at Fudan University on March 15, 2017

252 Mokhtari et al.

View publication stats

You might also like