Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

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

net/publication/368926457

Diets, Gut Microbiota and Metabolites

Article · March 2023


DOI: 10.1007/s43657-023-00095-0

CITATIONS READS

0 227

8 authors, including:

Yan Sun
Masonic Medical Research Laboratory
21 PUBLICATIONS   91 CITATIONS   

SEE PROFILE

All content following this page was uploaded by Yan Sun on 02 March 2023.

The user has requested enhancement of the downloaded file.


Phenomics
https://doi.org/10.1007/s43657-023-00095-0

REVIEW

Diets, Gut Microbiota and Metabolites


Yilian Liu1   · Wanglei Zhong1   · Xiao Li1   · Feng Shen2   · Xiaonan Ma1   · Qi Yang1   · Shangyu Hong1   ·
Yan Sun3 

Received: 26 July 2021 / Revised: 10 January 2023 / Accepted: 12 January 2023


© International Human Phenome Institutes (Shanghai) 2023

Abstract
The gut microbiota refers to the gross collection of microorganisms, estimated trillions of them, which reside within the gut
and play crucial roles in the absorption and digestion of dietary nutrients. In the past decades, the new generation ‘omics’
(metagenomics, transcriptomics, proteomics, and metabolomics) technologies made it possible to precisely identify micro-
biota and metabolites and describe their variability between individuals, populations and even different time points within
the same subjects. With massive efforts made, it is now generally accepted that the gut microbiota is a dynamically changing
population, whose composition is influenced by the hosts’ health conditions and lifestyles. Diet is one of the major contribu-
tors to shaping the gut microbiota. The components in the diets vary in different countries, religions, and populations. Some
special diets have been adopted by people for hundreds of years aiming for better health, while the underlying mechanisms
remain largely unknown. Recent studies based on volunteers or diet-treated animals demonstrated that diets can greatly and
rapidly change the gut microbiota. The unique pattern of the nutrients from the diets and their metabolites produced by the
gut microbiota has been linked with the occurrence of diseases, including obesity, diabetes, nonalcoholic fatty liver disease,
cardiovascular disease, neural diseases, and more. This review will summarize the recent progress and current understand-
ing of the effects of different dietary patterns on the composition of gut microbiota, bacterial metabolites, and their effects
on the host's metabolism.

Keywords  Gut microbiota · Metabolites · Diets · Nutrients · Metabolic diseases

Abbreviations TMA Trimethylamine


NAFLD Nonalcoholic fatty liver disease sBA Secondary bile acid
CVD Cardiovascular disease KD Ketogenic diet
LPS Lipopolysaccharide HFD High-fat diet
BCAA​ Branched-chain amino acid WD Western diet
SCFA Short-chain fatty acid HGD High-glucose diet
HFrD High-fructose diet
VD Vegan diet
Yilian Liu, Wanglei Zhong, Xiao Li, and Feng Shen have VeD Vegetarian diet
contributed equally to this paper.
MD Mediterranean diet
* Shangyu Hong GFD Gluten-free diet
shangyu_hong@fudan.edu.cn CD Chow diet
* Yan Sun RELMβ Resistin-like molecule β
ysun@mmri.edu NF-κB Nuclear factor-κB
1
MyD88 Myeloid differentiation factor 88
State Key Laboratory of Genetic Engineering and School
of Life Sciences, Human Phenome Institute, Fudan
IRAK Interleukin 1 receptor associated kinase
University, 2005 Songhu Road, Yangpu District, TRAF6 Tumor necrosis factor receptor associated factor
Shanghai 200433, China 6
2
Department of Hepatobiliary Surgery, Dongfeng Hospital, TAK1 Transforming growth factor β activated kinase 1
Hubei University of Medicine, Shiyan 442001, Hubei, China IRS1/2 Insulin receptor substrate 1/2
3
Masonic Medical Research Institute, 2150 Bleecker St, Utica, mTOR Mammalian target of rapamycin
NY 13501, USA

13
Vol.:(0123456789)
Y. Liu et al.

GABA Gamma-aminobutyric acid rapidly and frequently and shifts over one to two days when
OTUs Operational taxonomic units the hosts switch their diets (David et al. 2014; Walker et al.
GLP-1 Glucagon-like peptide-1 2011; Wu et al. 2011). Long-term consumption of these
diets plays a dominant role in shaping the hosts' gut micro-
biota and forming a stable and distinguished enterotype (Wu
Introduction et al. 2011). For example, HFD increases the abundance
of Firmicutes but decreases the abundance of Verrucomi-
There are more than 160 species of microorganisms in the crobia (Bisanz et al. 2019). On the contrary, KD decreases
human gastrointestinal tract (Laterza et al. 2016). Piles of the abundance of Firmicutes but increases the abundance
evidence had demonstrated that the disorder of gut micro- of Verrucomicrobia (Xie et al. 2017; Zhang et al. 2018b). In
biota might result in diseases, such as obesity (Ley et al. the last decade, tremendous efforts have been made to extend
2005), type 2 diabetes mellitus (T2DM) (Pascale et  al. our knowledge of how these diets impact the gut microbiota
2019), nonalcoholic fatty liver disease (NAFLD) (Ma et al. and the host's metabolism. In this review, we will summa-
2017), cardiovascular disease (CVD) (Tang et al. 2019), neu- rize the large body of data on how these diets affect gut
ral diseases (Olson et al. 2018), polycystic ovary syndrome microbiota and metabolites. We will also discuss how the
(Qi et al. 2019), and cancer (Raza et al. 2019). Accumulat- metabolic status of the hosts will be impacted by these diets
ing evidence, which was well reviewed by Wu et al. (2021), and the underlying mechanisms.
suggests that the metabolites produced by the gut microbiota
may be one of the major contributors. These metabolites,
including lipopolysaccharide (LPS), branched-chain amino High‑Fat Diet
acid (BCAA), short-chain fatty acid (SCFA), trimethylamine
(TMA), secondary bile acid (sBA), and so on, are often asso- HFD refers to the dietary pattern with the fat content gener-
ciated with the metabolic states, activation of inflammation, ally being 40–60%. Consumption of HFD profiles a pattern
and regulation of the hosts’ genes involved in energy stor- on gut microbiota and results in a different collection of
age and consumption (Blaut 2015; Gentile and Weir 2018; bacterial metabolites, which has been proven to contribute
Manco et al. 2010; Neis et al. 2015; Tsukumo et al. 2009; to the prevalence of overweight, and lead to obesity, T2DM,
Zhu et al. 2016). and CVD (Stocks et al. 2013; Wan et al. 2019; Woodie and
Diets have a great influence on the gut microbiota (Bibbo Blythe 2018).
et al. 2016; Requena et al. 2018). Due to the availability, Extensive evidence has demonstrated the influence of
tradition, religion, and special purposes related to diseases HFD consumption on the composition of gut microbiota.
or lifestyles, the nutrients in the diets vary in different coun- At the phylum level, HFD consumption decreases the
tries and populations. Among the popular diets studied, the abundance of Verrucomicrobia (Everard et al. 2014), but
different proportion of nutrients features these diets and was increases the abundance of Proteobacteria (Hildebrandt
believed to contribute to physiological consequence in the et al. 2009). At the genus level, the abundance of Biloph-
consumers of these diets, mainly the ketogenic diet (KD), ila, Butyrivibrio, Parabacteroides, and Roseburia  were
the high-fat diet (HFD), the western diet (WD), the high-glu- increased, but the abundance of Allobaculum, Coprococcus,
cose diet (HGD), the high-fructose diet (HFrD), the vegan Eubacterium, Lactobacillus, and Prevotella were decreased
diet (VD), the vegetarian diet (VeD), the Mediterranean by HFD consumption (Everard et al. 2014). At the species
diet (MD), and the gluten-free diet (GFD) (Rinninella et al. level, HFD consumption increases the abundance of Biloph-
2019). KD contains the highest proportion of fat (generally ila wadsworthia  (Devkota et  al. 2012),  and decreases
over 70%) and the lowest proportion of carbohydrates (less the abundance of  Bacteroides fragilis (Sun et  al. 2018)
than 10%) among the aforementioned diets (Veech 2004). and Akkermansia muciniphila (Schneeberger et al. 2015).
Both HFD and WD have higher levels of fat than other diets However, the reported effects of HFD on microbiota vary
except KD (Bortolin et al. 2018; Woodie and Blythe 2018), across studies. The reason might be the different sources of
while HGD and HFrD contain high levels of carbohydrates major nutrients in the HFD, different times on HFD, differ-
(Johnston et al. 2013). VD, VeD and MD are featured with ent animal handling policies, etc. To find out the robust and
a high level of dietary fiber but low in animal-derived fat reproducible changes in microbial composition induced by
and protein (Haddad et al. 1999; Kastorini et al. 2011). HFD feeding, Bisanz et al. performed a meta-analysis based
GFD is a diet lacking a specific class of proteins and gluten on 27 studies involving 1, 101 samples (including mouse and
(Biesiekierski 2017). The dietary fats, carbohydrates, pro- human samples) (Bisanz et al. 2019). Their analysis demon-
teins and dietary fibers were believed to impact gut microbi- strated that HFD consumption induces reproducible shifts in
ota (Candido et al. 2018; Deng et al. 2022; Kim et al. 2016). the gut microbiota and revealed an increased ratio of Firmi-
In general, the composition of the gut microbiota changes cutes: Bacteroidetes in all of those analyzed datasets from

13
Diets, Gut Microbiota and Metabolites

mice. This increased ratio of Firmicutes: Bacteroidetes was and isoleucine, mainly come from food and intestinal flora.
later reaffirmed by Munch et al. when they analyzed 16s BCAAs contribute to the development of obesity-associated
ribosomal RNA of the fecal samples from mice fed with a insulin resistance in humans and rodents (Goffredo et al.
chow diet (CD) or HFD for 30 weeks (Munch et al. 2019). 2017; Newgard et al. 2009), which are associated with the
The impact of HFD on human gut microbiota is more com- progression of obesity-related metabolic disorders, including
plicated and addresses more debates without a consensus T2DM (Asghari et al. 2018) and NAFLD (Li et al. 2013).
(Magne et al. 2020). Another concern about HFD-induced There are two proposed mechanisms by which BCAAs may
changes in microbiota is whether the changes in the micro- impair insulin sensitivity (Newgard et al. 2009). One is that
bial composition are directly caused by HFD consumption BCAAs activate mammalian target of rapamycin (mTOR)
or the obese state of the hosts resulting from HFD consump- signaling, and they lead to insulin resistance. Another pro-
tion. Hildebrandt et al. distinguished these two scenarios posed mechanism is that the increased BCAAs turnover
by using resistin-like molecule β (RELMβ) knockout mice, drives the production of toxic mitochondrial BCAAs cat-
which are resistant to diet-induced obesity, and they con- abolites (e.g., propionyl CoA, succinyl CoA, branched-chain
cluded that the high-fat diet itself, and not the obese state, keto acids), which are in turn proposed to impair mitochon-
was responsible for the altered microbiota (Hildebrandt et al. drial oxidative metabolism and thereby induce mitochondrial
2009). stress and lead to insulin resistance (Asghari et al. 2018).
Gut microbiota can impact the host through the metabo- HFD consumption decreases the circulating level of
lites they produced, including LPS, BCAAs, SCFAs, TMA, SCFAs by suppressing the abundance of SCFAs-producing
sBA, and so on (Gentile and Weir 2018; Manco et al. 2010; bacteria, Akkermansia Muciniphila (Alzaben et al. 2015;
Neis et al. 2015; Zhu et al. 2016). Cani et al. 2008; Derrien et al. 2004). The bacteria ferment
HFD consumption increases the circulating level of and degrade indigestible carbohydrates to produce SCFAs,
LPS and BCAAs by enhancing intestinal permeability and including acetate, propionate, and butyrate (Macfarlane and
decreases SCFA by suppressing the abundance of SCFA- Macfarlane 2011). It has been demonstrated that propion-
producing bacteria such as Akkermansia muciniphila (Alza- ate and butyrate concentrations correlate inversely with the
ben et al. 2015; Cani et al. 2008). LPS is mainly produced by risk of obesity, insulin resistance, and NAFLD (Ampong
Gram-negative bacteria (Kirkland and Ziegler 1984). Previ- et al. 2020). Mechanistically, the SCFAs bind to their recep-
ous studies have shown that high LPS levels would cause tors and increase the production of glucagon-like peptide-
subclinical inflammation, insulin resistance, and an increase 1(GLP-1) and peptide tyrosine tyrosine, which affect host's
in fat mass (Horton et al. 2014; Pedersen et al. 2016). In the energy balance and lipid metabolism (Hernandez et al. 2019;
liver, LPS activates nuclear factor-κB (NF-κB) and myeloid Koh et al. 2016; Liu et al. 2020; Sircana et al. 2018).
differentiation factor 88 (MyD88), and leads to NAFLD (Ma With the extensive research on gut microbiota in the con-
et al. 2017). In adipocytes, LPS will cause insulin resistance text of HFD consumption, it seems safe to say that HFD
by activating the MyD88- interleukin 1 receptor associated consumption can rapidly and reproducibly alter the mouse's
kinase (IRAK)-tumor necrosis factor receptor associated fac- gut microbiota and metabolites, which have a major impact
tor 6 (TRAF6)-transforming growth factor β activated kinase on both immunological and metabolic status of the hosts
1 (TAK1)-insulin receptor substrate 1/2 (IRS1/2) signaling (Table 1).
axis (Saad et al. 2016). BCAAs, including valine, leucine,

Table 1  The impacts of HFD on gut microbiota, secondary metabolites and metabolic status
Microbiota Secondary metabolites Metabolic status
Phylum Genus Species

HFD Firmicutes↑ Bilophila↑ B. wadsworthia↑ LPS↑ Subclinical inflammation↑


Proteobacteria↑ Butyrivibrio↑ B. fragilis↓ BCAA↑ Insulin resistance↑
Bacteroidetes↓ Parabacteroides↑ A. Muciniphila↓ SCFA↓ Adipose mass↑
Verrucomicrobia↓ Roseburia↑ Metabolic disorders↑
Allobaculum↓ T2DM↑
Coprococcus↓ NAFLD↑
Eubacterium↓
Lactobacillus↓
Prevotella↓

HFD high fat diet, LPS lipopolysaccharide, BCAA​ branched chain amino acid, SCFA short chain fatty acid, T2DM type 2 diabetes mellitus,
NAFLD Nonalcoholic fatty liver disease

13
Y. Liu et al.

Western Diet Ketogenic Diet

WD is a typical diet pattern in western countries. It is KD is a normal caloric diet that is high in fat (over 70% in
high in both saturated fats (> 40%) and refined carbohy- most cases), low in carbohydrates, and moderate in pro-
drates (> 40%) and has higher efficiency to induce obesity, tein. KD consumption results in a very low carbohydrate
increase visceral adipose tissue, hepatic steatosis, inflam- uptake (less than 10% of total caloric intake), and forces
mation, fasting hyperglycemia, and dyslipidemia in rats ketone production (Veech 2004). KD is initially introduced
compared to HFD mentioned above (Bortolin et al. 2018; as an effective treatment for drug-resistant epilepsy (Hohn
Tiniakos et al. 2010). et al. 2019). Later, KD is also applied as a treatment for
WD consumption has adverse effects on gut micro- other neuronal diseases (Rusek et al. 2019). But nowadays,
biota, which include disorder of intestinal flora, damage KD is prevalently used by people aiming to lose weight,
of intestinal mucosal, increased intestinal permeability, and the mechanism is under investigation (Badman et al.
and consequent inflammation (Chassaing et  al. 2015; 2009; Garbow et al. 2011; Kosinski and Jornayvaz 2017).
Schroeder et  al. 2018; Zinocker and Lindseth 2018). Meanwhile, some studies raised the concern that the con-
In 2021, Low et al. comprehensively compared the gut sumption of KD might cause some adverse effects such as
microbiota between the mice fed with WD and normal NAFLD and insulin resistance (Dashti et al. 2003; Ellen-
diets and found that these changes are largely reversible broek et al. 2014; Jornayvaz et al. 2010).
and repeatable (Low et al. 2021). At the phylum level, KD dramatically influences the composition of gut
Firmicutes and Verrucomicrobiota showed significantly microbiota (Table 3). At the phylum level, KD decreases
higher abundances, while Bacteroidota and Cyanobac- the abundance of Firmicutes but increases the abundance
teria have lower abundances in WD-fed mice than in of  Bacteroidetes  and  Proteobacteria (Xie et  al. 2017;
control mice (Low et al. 2021). At the family level, eight Zhang et al. 2018b). At the family level, KD decreases the
families have significantly higher abundances while 16 abundance of  Enterobacteriaceae,  Sinobacteraceae,
families were significantly less abundant in WD-fed and Comamonadaceae, but increases the abundance of
mice than in normal diets-fed mice (Low et al. 2021). Ruminococcaceae and Mogibacteriaceae (Gutierrez-Rep-
Nine families were similarly abundant among the groups iso et al. 2019). At the genus level, KD decreases the abun-
(Low et al. 2021). In 2022, Romualdo et al. analyzed the dance of Serratia, Erwinia, and Citrobacter, and increases
impacts of WD on mouse's gut microbiota and linked the abundance of Oscillospira and Butyricimonas (Gutier-
the changes in gut bacteria with liver steatosis. Their rez-Repiso et al. 2019). KD was also reported to decrease
data confirmed the WD-induced alteration in mouse's the abundance of Dialister and Cronobacter and increases
gut microbiota with differences in phyla and families the abundance of Desulfovibrio, Prevotella, Lactobacil-
(Romualdo et al. 2022). The combined information from lus, and Parabacteroides (Lindefeldt et al. 2019; Ma et al.
the abovementioned studies were listed in Table 2. 2018; Olson et al. 2018; Tagliabue et al. 2017; Xie et al.
Similar to HFD, WD increases the plasma level of 2017). At the species level, KD decreases the abundance
LPS and decreases the level of SCFAs (Kaye et al. 2020; of Bifidobacteria, but increases the abundance of Akker-
Simkin 2019), which contribute to the increased insulin mansia muciniphila (Ma et al. 2018; Olson et al. 2018),
resistance, body weight, plasma and liver triglycerides, which has been reported to be associated with weight
cholesterol, and risk of CVD (Baena et al. 2017). The loss, improved insulin sensitivity, alleviated insulinemia,
direct comparison of the gut microbiota between WD- decreased plasma total cholesterol, and the development
and HFD-fed rats showed minor differences between the of T2DM and hypertension (Brahe et al. 2015; Dao et al.
two groups (Bortolin et al. 2018). Overall, WD induced 2016; Depommier et al. 2019; Kong et al. 2021; Li et al.
similar alterations in gut microbiota and metabolites 2017; Liu et al. 2017; Yassour et al. 2016; Zhang et al.
when compared with HFD (Table  2). Nevertheless, a 2013). However, to what extent the regulated abundance
closer look at the features of gut microbiota and metabo- of Akkermansia muciniphila contributes to the physiologi-
lites in WD-fed mammals will be helpful for us to under- cal impact of KD remains unclear.
stand the mechanisms of this popular obesogenic diet. The consumption of KD leads to the change of metab-
olites of gut microbiota. Akkermansia muciniphila  is
regarded as a key propionate-producing bacterium and
butyrate production is dominated by  Ruminococcus

13
Diets, Gut Microbiota and Metabolites

Table 2  The impacts of WD on gut microbiota, secondary metabolites and metabolic status


Microbiota Secondary Metabolic status
metabolites
Phylum Class Order Family Genus

WD Firmicutes↑ Clostridia↑ Clostridiales↑ Erysipel- Coprococcus↑ LPS↑ vWAT↑


Proteobacteria↑ Deltaproteobac- Desulfovibrion- otrichaceae↑ Ruminococcus↑ SCFA↓ Body weight↑
Verrucomicro- teria↑ ales↑ Streptococ- Desulfovibrio↑ Plasma triglycer-
bia↑ Bacili↑ Lactobacillales↑ caceae↑ Faecalibacte- ides↑
Actinobacteria↓ Bacterioidia↓ Bacterioidales↓ Clostridiaceae↑ rium↓ Liver triglycer-
Bacteroidetes↓ Fusobacteria↓ Fusobacteriales↓ Eubacterium Fusobacterium↓ ides↑
Cyanobacteria↓ coprostanoli- Bacteroides↓ Plasma choles-
Fusobacteria↓ genes group↑ Prevotella↓ terol↑
Enterococcaceae↑ Alloprevotella↓ Liver cholesterol↑
Christensenel- Alistipes↓ Insulin resistance↑
laceae↑ RC9 gut group↓ Adipose mass↑
Staphylococ- Parabacteroides↓
caceae↑ Odoribacter↓
Akkermansi-
aceae↑
Ruminococ-
caceae↑
Desulfovibrion-
aceae↑
Lachnospiraceae↓
Lactobacil-
laceae↓
Clostridia UCG-
014↓
Butyricicoc-
caceae↓
Clostridia
vadinBB60
group↓
Acholeplasmata-
ceae↓
Monoglobaceae↓
RF39↓
Peptococcaceae↓
Bacteroidaceae↓
Rikenellaceae↓
Muribaculaceae↓
Bifidobacte-
riaceae↓
Coriobacteriales
Incertae Sedis↓
Eggerthellaceae↓
Gastranaer-
ophilales↓
Prevotellaceae↓
Porphyromona-
daceae↓
Fusobacte-
riaceae↓

WD western diet, LPS lipopolysaccharide, SCFA short chain fatty acid, vWAT​visceral adipose tissue

bromii (Morrison and Preston 2016). However, a recent et al. 2021). Further studies will be needed to clarify the
study reported a reduction of SCFAs in KD-fed mice when impacts of KD on the metabolism of SCFAs, sBAs and
compared to normal chow diet-fed mice (Li et al. 2021). tryptophan, and the consequences on the hosts' metabolic
In their report, Li et al. also revealed that KD feeding status. In 2018, Olson et al. reported that the consump-
decreased four main tryptophan metabolites and changed tion of KD decreased systemic gamma-Aminobutyric
the levels of 10 sBAs, among the 40 sBAs monitored (Li acid (GABA) and elevated hippocampal GABA/glutamate

13
Y. Liu et al.

Table 3  The impacts of KD on gut microbiota, secondary metabolites and metabolic status


Microbiota Secondary metabolites Metabolic status
Phylum Family Genus Species

KD Bacteroidetes↑ Ruminococcaceae↑ Oscillospira↑ A. muciniphila↑ SCFA↑ Insulin resistance↓


Proteobacteria↑ Mogibacteriaceae ↑ Butyricimonas↑ sBA↓
Firmicutes↓ Enterobacteriaceae↓ Desulfovibrio↑
Sinobacteraceae↓ Prevotella↑
Comamonadaceae↓ Lactobacillus↑
Parabacteroides↑
Bifidobacteria↓
Serratia↓
Erwinia↓
Citrobacter↓
Dialister↓
Cronobacter↓

KD ketogenic diet, SCFA short chain fatty acid, sBA secondary Bile Acids

levels, which play critical roles in the anti-seizure effect 2018; Mastrocola et al. 2018; Vasques-Monteiro et al. 2021).
induced by KD (Olson et al. 2018). However, this idea was challenged by Bier et al. (2020).
Although more and more attention was put on the impact They claimed that antibiotic treatment significantly reduced
of KD on the gut microbiota and their metabolites, there microbial diversity and altered the microbial composition,
are still several critical questions and concerns remained but had minimal or no effect on the metabolic phenotypes of
unanswered. First, as partially addressed by Li et al. in their the HFrD-treated rats (Bier et al. 2020). Further investiga-
report, both the sources and proportions of fat in the KD are tion will be needed to find out the cause of this inconsist-
critical for the KD-induced alteration in gut microbiota and ency. Nevertheless, there's no doubt that HFD or HFrD has a
their metabolites (Li et al. 2021). The differential alteration great impact on gut microbiota. At the phylum level, HGD-
that would impact the hosts differently will be an intriguing or HFrD-fed mice had a significantly lower abundance
topic for future investigation. Second, it has been shown that of Bacteroidetes and a higher abundance of Proteobacte-
there is a biphasic effect of KD in microbiota: a quick but ria compared to normal diet-fed mice after 12 weeks of feed-
dramatic decrease in the richness and diversity of the bacte- ing (Do et al. 2018). At the family levels, the levels of nine
ria, followed by a slow recovery of bacterial concentration different families were significantly changed in the feces of
(Swidsinski et al. 2017). Thus, the microbiota analyzed at C57BL/6J mice fed with HFrD for 12 weeks (Table 4) (Ahn
different time points may show different results. Third, the et al. 2020). While at the genus level, they found HFrD feed-
proportion of fat, carbohydrates, and proteins varies a lot in ing increased the abundance of Akkemansia but decreased
different versions of KDs. The KDs used in animal studies the abundance of Dehalobacterium and an unknown genus
are generally more extreme than the KD used in patients, and in Mogibacteriaeae (Ahn et al. 2020). At the species level,
the KDs used by patients with epilepsy are generally more HFrD-fed mice had higher levels of Akkermansia muciniph-
extreme than the KDs used by weight losers, which may ila, Clostridium sp, Lachnoclostridium clostridium alden-
cause inconsistency in the conclusion from animal studies ense, Roseburia sp, Lachnoclostridium clostridium scindens,
and clinical trials (Li et al. 2021; Zhu et al. 2022). and lower levels of Clostridium disporicum, Lactobacillus
johnsonii, Bacteroides acidifaciens, Allobaculum sp in their
feces after one-week feeding. Different mice strains may also
High‑Glucose or Fructose Diet respond differently to fructose (Montrose et al. 2021).
The intestinal permeability of HGD or HFrD group mice
Excessive sugar intake contributes to the development of was enhanced due to the lower expression of tight junction
NAFLD, CVD, and T2DM in both direct and indirect man- proteins, such as zonula occludens-1 and occluding, which
ners (Stanhope 2016). Consumption of two commonly used can increase the intestinal permeability and contribute to
high-sugar diets: HGD or HFrD results in similar fatty liver the translocation of LPS (Cani et al. 2008; Do et al. 2018).
parameters, including elevated levels of hepatic triacylglyc- Given the increased abundance of Proteobacteria (Do et al.
erol and insulin resistance (Johnston et al. 2013). Data from 2018), one of the major producers of LPS (Rizzatti et al.
multiple groups suggested that fructose consumption is spe- 2017), it is not a surprise that HFrD increases plasma LPS
cifically linked to gut microbial dysbiosis, which mediated (Rivero-Gutierrez et al. 2017), and the increased plasma
the hosts' metabolic changes (Di Luccia et al. 2015; Do et al. levels of LPS leads to insulin resistance in murine fed with

13
Diets, Gut Microbiota and Metabolites

Table 4  The impacts of HGD or HFrD on gut microbiota, secondary metabolites and metabolic status
Microbiota Secondary Metabolic status
metabolites
Phylum Family Genus Species

HGD or HFrD Proteobacteria↑ S24-7↑ Akkermansia↑ A. muciniphila↑ LPS↑ Insulin resistance↑


Bacteroidetes↓ Pseudomona- Dehalobacterium↓ C. sp↑ Hepatic inflamma-
daceae↑ Unknown genus L. clostridium tion↑
Verrucomicrobi- (Mogibacte- aldenense↑ Hepatic lipid accu-
aceae↑ riaceae)↓ R. sp↑ mulation↑
Rikenellaceae↓ L. clostridium Enlargement of
Dehalobacte- scindens↑ adipocytes↑
riaceae↓ C. disporicum↓
Lachnospiraceae↓ L. johnsonii↓
Mogibacteriaceae↓ B. acidifaciens↓
Ruminococcaceae↓ A. sp↓
Turicibacteraceae↓

HGD or HFrD high glucose diet or high fructose diet, LPS lipopolysaccharide

HGD or HFrD (Pedersen et al. 2016; Rivero-Gutierrez et al. diet are advocated as a dietary strategy for maintaining good
2017). Silva et al. also compared the fecal metabolites from health and treating metabolic disorders, including obesity,
mice fed with ND, HGD or HFrD, and found that high glu- diabetes, and CVD (Craig 2009; Jenkins et al. 2014; Le and
cose or fructose feeding caused different alterations in fecal Sabate 2014; Tonstad et al. 2013).
SCFAs (Silva et al. 2018). Their results indicated that both The influence of vegan and vegetarian diets on gut micro-
high glucose- and high fructose-feeding caused higher levels biota has been studied in many laboratories, and the results
of fecal acetate. But only high fructose-, not high glucose- are somewhat contradictory. The overall differential micro-
fed mice had lower levels of fecal butyrate. On the other biota composition in vegans or vegetarians, when compared
side, high glucose- but not high fructose-fed mice had signif- to omnivores, and inconsistency were well summarized in
icantly higher levels of fecal propionate compared to normal a systematic review (Trefflich et al. 2020). The reasons for
chow diet-fed mice (Silva et al. 2018). Ahn et al. also raised the inconsistency could be the different ways the vegan or
the concern of substantial variability in both fecal metabo- vegetarian were defined, the different regions the subjects
lites and microbial profiles between animals within the same were recruited, the time the subjects stayed on the diets, and
feeding group, as well as at different time points for one how the samples were collected and analyzed. One single
animal across the feeding interval (Ahn et al. 2020). Thus, study with the biggest number of differences in microbiota
it is recommended to have multiple feces sampling collected composition is from Zhang et al. (2018a). They recruited
during a feeding trial to obtain representative levels of fecal 29 healthy volunteers and divided them into three groups (a
metabolites and microbial populations. study group with 15 members normally consume an omnivo-
In recent decades, the introduction of high-fructose corn rous but were put on a three-months lacto-ovo-vegetarian
syrup in the food industry dramatically increased the con- diet, an omnivorous group with seven members stayed on an
sumption of fructose. How the HFrD would change our lives omnivorous diet and a vegetarian group with seven members
and the safe limit of daily fructose consumption remains stayed on a vegetarian diet). They collected samples on day
largely unknown. Further investigation on fructose-induced 0 and day 91 from each group and surveyed the effect on
alteration of microbiota and their metabolites is needed for a the diversity of gut microbiota by metagenomic sequencing
better understanding of the impact of fructose consumption. (Zhang et al. 2018a). A combined short-term effect (Day
91 vs Day 0 in the study group) and long-term effects (the
omnivorous group vs the vegetarian group at Day 91 and
Vegan or Vegetarian Diet Day 0) on gut microbiota were listed in Table 5.
Bacterial metabolites were more abundant while lipid and
There are two kinds of vegetarianism: one is vegetarian, amino acid metabolites were less abundant in the plasma
and the population are on the traditional vegetarian diet; metabolome of vegans when compared to omnivores (Wu
the other one is vegan, also named a strict vegetarian, and et  al. 2016). A vegetarian diet increases the abundance
a vegan avoids all animal and animal-derived products. of Prevotella, which produces SCFAs (de Moraes et al.
Accordingly, these two plant foods-based diets are featured 2017; Rios-Covian et al. 2016). The elevation of SCFAs may
a high level of fiber and carbohydrates but low in fat and contribute to diet-induced weight loss and increased insu-
protein (Haddad et al. 1999). Both vegan diet and vegetarian lin sensitivity in vegans (De Filippo et al. 2010; Pilis et al.

13
Y. Liu et al.

Table 5  The impacts of VD or VeD on gut microbiota, secondary metabolites and metabolic status
Microbiota Secondary metabolites Metabolic status
Phylum Genus Species

VD or VeD Firmicutes↑ Prevotella↑ P. copri↑ SCFA↑ Insulin sensitivity↑


Proteobacteria↑ Capnocytophaga↑ B. Hydrogenotrophica↑ TMA↓
Bacteroidetes↓ Porphyromonas↑ C. Ramosum↑ sBA↓
Verrucomicrobia↓ Roseburia↑ C. Symbiosum↑
Faecalibacterium↑ P. duerdenii↑
Lachnospira↑ S. peroris↑
Veillonella↑ V. dispar↑
Actinobacillus↑ V. parvula↑
Atopobium↑ V. atypica↑
Actinoplanes↑ M. luteus↑
Cryptobacterium↑ A. Segnis↑
Micrococcus↑ A. actinomycetemcomitans↑
Aggregatibacter↑ C. concisus↑
Aeromonas/Pseudomona↑ H. haemolyticus↑
Campylobacter↑ H. influenzae↑
Haemophilus↑ H. parainfluenzae↑
Klebsiella↑ K. pneumonia↑
Neisseria↑ N. mucosa↑
Alistipes↓ B. Vulgatus↓
Parabacteroides↓ B. fragilis↓
Peptostreptococcus↓ B. Dorei↓
Desulfitobacterium↓ B. Thetaiotaomicron ↓
Ruminococcus↓ B. Uniformis↓
Dialister↓ B. Finegoldii↓
Phascolarctobacterium↓ B. Stercoris↓
Acetobacterium↓ P. gingivalis↓
Bulleidia↓ P. Distasonis↓
Holdemania↓ P. buccalis↓
Dorea↓ P. oris↓
Caldanaerobacter↓ P. tannerae↓
Lachnoclostridium↓ L. amylovorus↓
Lactobacillus↓ C. Clostridioforme↓
Lactococcus↓ C. Kluyveri↓
Oscillospira↓ C. coccoides-E. rectale↓
Bifidobacterium↓ C. Innocuum↓
Eggerthella↓ C. Paraputrificum↓
Corynebacterium↓ R. torques↓
Oxalobacter↓ D. invisus↓
Ruegeria↓ P. Succinatutens↓
Taylorella↓ E. Coli↓
Syntrophobacter↓ E. faecium↓
Ralstonia↓ R. Eubacterium rectale↓
Campylobacter↓ D. longicatena↓
Desulfovibrio↓ B. hansenii↓
Bilophila↓ A. caccae↓
Succinivibrio↓ F. magna↓
Halomas↓ T. pseudo ethanolicus↓
Methanosphaera↓ P. denticolens↓
Dehalogenimona↓ M. curtisii↓
Streptobacillus ↓ E. hermannii↓
Akkermansia ↓ S. fumaroxidans↓
A. baumannii↓
D. piger↓
D. alaskensis↓
D. aespoeensis↓
B. wadsworthia↓
E. ictaluri/ tarda↓
M. stadtmanae↓
D. lykanthroporepellens↓
F. ulcerans↓

VD or VeD vegan diet or vegetarian diet, SCFA short chain fatty acid, sBA secondary Bile Acids, TMA trimethylamine

13
Diets, Gut Microbiota and Metabolites

2014; Tindall et al. 2018; Xie et al. 2017). The vegetarian 2011; Ryan et al. 2013; Salas-Salvado et al. 2014; Wang
or vegan diets are low in choline, phosphatidylcholine, and et al. 2021).
L-carnitine, which cause the reduction of TMA in the hosts. The high level of dietary fibers and low content of
TMA can be further oxidized to trimethylamine N-oxide, red meat in an MD orchestrate a special pattern of gut
which could trigger insulin resistance and increase the risk microbiota (Table 6), which is believed to mediate, at least
of T2DM and NAFLD in humans and rodents (Barrea et al. partially, the beneficial effects of MD (De Filippis et al.
2018; Fu et al. 2020; Heianza et al. 2019; Schugar et al. 2016; Garcia-Mantrana et al. 2018; Ghosh et al. 2020;
2017; Shan et al. 2017). Primary bile acids are produced by Mitsou et al. 2017; Rinninella et al. 2019; Tindall et al.
the liver and secreted into the intestine, and then converted 2018; Wang et al. 2021). De Filippis et al. first convinc-
to sBAs by gut microbiota. The increased level of sBAs has ingly demonstrated the connection between MD and gut
been reported to positively correlate with the risk of insulin microbiota (De Filippis et al. 2016). In 2020, based on
resistance and T2DM (Sircana et al. 2018). sBAs activate 16S rRNA sequencing data, Ghosh et al. reported that, at
farnesoid X receptor (FXR) and G protein-coupled bile acid the species level, a 12-month MD intervention increased
receptor-1 (also named TGR5), which are both expressed in the abundance of  Faecalibacterium prausnitzii, Rose-
endocrine L cells. The activation of FXR and TGR5 stimu- buria hominis, Eubacterium rectale, Eubacterium eligens,
lates the secretion of GLP-1, which improves insulin sensi- Eubacterium xylanophilum, Bacteroides thetaiotaomicron,
tivity (Pathak et al. 2018). Vegans had lower levels of total Prevotella copri, and Anaerostipes hadrus in 612 subjects
BA and sBA in their feces than omnivores (Trefflich et al. across five European countries (Ghosh et al. 2020). These
2019), which might be because of the high intake of fibers species positively associate with the production of SCFAs
and low intake of fat (O'Keefe et al. 2015). Whether a plant- and negatively associate with the development of T2DM
derived diet improves insulin sensitivity by impacting the (Machiels et  al. 2014; Qin et  al. 2012). Consistently,
levels of sBA remains unknown. better adherence to the MD was associated with signifi-
cantly higher levels of total SCFA (Garcia-Mantrana et al.
2018). On the other side, MD decreased the abundance
Mediterranean Diet of Ruminococcus torques, Collinsella aerofaciens, Copro-
coccus comes, Dorea formicigenerans, Clostridium ram-
MD is a diet with a high level of vegetables, fruits, fish, osum, Veillonella dispar, Flavonifractor plautii, and Actin-
whole grains, beans, wine and olive oil, popular in south- omyces lingnae (Ghosh et al. 2020). In 2021, Wang et al.
ern European countries around the olive-tree-bearing based on metagenomics sequencing analysis, confirmed
regions of the Mediterranean basin. It has been reported the impacts of MD on these species, which were featured
that higher adherence to the MD is associated with a lower as fiber metabolizers and SCFA producers (Wang et al.
risk of CVD and metabolic syndrome (Kastorini et al. 2021). Based on their analysis, the abundances of whole
grams, vegetables, fruits and reduced red meat in MD were
Table 6  The impacts of MD on gut microbiota, secondary metabolites and metabolic status
Microbiota Secondary metabolites Metabolic status
Phylum Family Genus Species

MD Bacteroidetes↑ Lachnospiraceae↑ Lactobacillus ↑ C. albicans↑ SCFA↑ T2DM↓


Bifidobacteria↑ F. prausnitzii↑ TMA↓ Obesity↓
Clostridium ↓ R. hominis↑ sBA↓ Insulin sensitivity↑
E. rectale↑
E. eligens↑
E. xylanophilum↑
B. thetaiotaomicron↑
P. copri↑
A. hadrus↑
R. torques↓
C. aerofaciens↓
C. comes↓
D. formicigenerans↓
C. ramosum↓
V. dispar↓
F. plautii↓
A. lingnae↓

MD Mediterranean diet, SCFA short chain fatty acid, sBA secondary Bile Acids, TMA trimethylamine, T2DM type 2 diabetes mellitus

13
Y. Liu et al.

the determining components shaping the specific pattern samples at both six and 12 months on GFD compared to
of gut microbiota in MD-consuming individuals (Wang baseline samples.
et al. 2021). The effects of GFD on healthy people were
In addition to the aforementioned SCFAs, the other also investigated. The abundance of  Bifidobacte-
metabolites were also found to be affected by MD and play rium,  Clostridium lituseburense, and  Faecalibacte-
roles in hosts' pathophysiological conditions. Tindall et al. rium prausnitzii was decreased but the abundance of Enter-
demonstrated that MD can reduce the plasma level of TMA, obacteriaceae  and  Escherichia coli  was increased in
leading to a lower risk of insulin resistance (Tindall et al. healthy people after one month of GFD consumption (De
2018). Ghosh et al. demonstrated that MD can decrease the Palma et al. 2009). The results from a similar clinical trial
level of sBA (Ghosh et al. 2020), which is positively associ- on healthy adults on GFD for one month demonstrated that
ated with insulin resistance (Sircana et al. 2018). GFD can decrease the abundance of some beneficial bacte-
ria, such as Bifidobacterium longum and Lactobacillus, but
also increase the abundance of Enterobacteriaceae, which
Gluten‑Free Diet is regarded as harmful bacteria (Sanz 2010). Bonder et al.
(2016) investigated the change in intestinal microbiota of
Gluten is a mixture of hundreds of distinct proteins but is 21 healthy participants who were kept on GFD for four
primarily made up of two different classes of proteins: glia- weeks, and their results showed that the abundance of Veil-
din and glutenin (Biesiekierski 2017). GFD is a diet that lonellaceae changed most obviously in GFD and its abun-
excludes all gluten-containing foods, such as pasta, pizza, dance was markedly decreased. Besides, the abundance
beer, oatmeal, toast, sandwiches, cakes, bread, cookies, etc. of Ruminicoccus bromii and Roseburia was also decreased
GFD is the only efficient treatment available for Celiac dis- by GFD (Bonder et al. 2016). On the contrary, the abun-
ease, a chronic inflammatory pathology of the small intestine dance of Victivallaceae, Clostridiaceae, and Coriobacte-
triggered by dietary gluten (Newnham 2017), which affects riaceae was increased (Bonder et al. 2016).
about 1% of the general population worldwide (Catassi et al. GFD will lead to the change of metabolites of intestinal
2015; Lionetti et al. 2015). GFD is also taken by healthy microbiota (Table 7). As mentioned above, GFD decreases
people aiming for better health, but the potential usage of the abundance of Bifidobacterium and Lactobacillus, both
GFD in preventing obesity and diabetes is controversial were the source of SCFAs. Therefore, it is not a surprise to
(Haupt-Jorgensen et al. 2018). see that GFD consumers have a lower circulating level of
Zafeiropoulou et al. (2020) compared the gut micro- SCFAs (Di Cagno et al. 2011; Garcia-Mazcorro et al. 2018).
biota of celiac disease children on GFD (45 patients) and Given the strong impacts of SCFAs on insulin sensitivity
gluten-containing diets (20 patients). And then, among (Hernandez et al. 2019), logically, the decreased circulat-
these 20 children on gluten-containing diets, 13 patients ing level of SCFAs by GFD will increase the risk of meta-
participated in a 12 months prospective study and started bolic diseases. Indeed, GFD consumption increased the risk
to consume GFD (Zafeiropoulou et al. 2020). Their fecal of NAFLD in Celiac disease patients (Tovoli et al. 2018;
samples were collected at six months and 12 months on Imperatore et al. 2018). Although some studies suggest that
GFD and analyzed by 16s ribosomal RNA sequencing. In GFD consumption reduced the risk of obesity and diabetes
their cross-sectional study, Zafeiropoulou et al. identified (Haupt-Jorgensen et al. 2018), others argued that GFD might
51 Operational taxonomic units (OTUs) with significantly aggravate obesity, serum lipid levels, insulin resistance, and
higher abundance, and three OTUs with significantly atherosclerosis (Alzaben et al. 2015; Anania et al. 2017;
lower abundance in GFD diet children (Zafeiropoulou Kemppainen et al. 1995; Mariani et al. 1998). More studies
et al. 2020). In the prospective study, they found seven are needed to increase our understanding of how a GFD is
significantly decreased and three increased OTUs in the
Table 7  The impacts of GFD on gut microbiota, secondary metabolites and metabolic status
Microbiota Secondary metabolites Metabolic status
Family Genus Species

GFD Enterobacteriaceae↑ Roseburia↓ R. bromii↓ SCFA↓ NAFLD↑


Bacteroidaceae↑ Bifidobacterium↓ C. lituseburen↓ Hepatic steatosis↑
Clostridiaceae↑ Lactobacillus↓ F. prausnitzii↓ Serum Lipid levels↑
Victivallaceae↑ E. coli↑ Insulin resistance↑
Lachnospiraceas↓ B. longum↓ Atherosclerosis↑
Veillonellaceae↓

GFD gluten-free diet, SCFA short chain fatty acid, NAFLD nonalcoholic fatty liver disease

13
Diets, Gut Microbiota and Metabolites

associated with metabolic disease and to apply GFD effec- other and influence the hosts' pathophysiology? What are
tively and safely. the key sensors or message receivers in the hosts that help
the hosts quickly and harmoniously adapt to the dietary
changes? Nowadays, more and more people acknowledge
Conclusion the featured diets as medical treatments or nutritional strat-
egies aiming for better health. But, the potential adverse
Leveraging advanced technology, researchers have made effects of some diets are underestimated. How the gut
huge progress in the identification and systematization microbiota gets involved in these beneficial and/or harm-
of microbiota in the past decades. However, researchers ful effects is largely unknown. Without any doubt, further
realized a big obstacle when generalizing results from research is warranted and will keep extending our knowl-
the previous literature: the inconsistency among differ- edge of the gut microbiota.
ent  laboratories (Bier et  al. 2020; Bisanz et  al. 2019;
Acknowledgements  This work was supported by grants from the
Trefflich et al. 2020), or even within the same laboratory National Key R&D Program of China (2019YFA0802300 to S. H.),
(Ahn et al. 2014). Many factors contribute to these incon- Training Program of the Major Research Plan of the National Natu-
sistencies. First, the techniques used for identifying the ral Science Foundation of China (91957117), National Natural Sci-
microbes are different, from real-time PCR, 16s ribosomal ence Foundation of China (31971082), Shanghai Pujiang Program
(18PJ1400500), and Open Research Fund of the National Key Labo-
RNA sequencing to metagenomic sequencing. Second, the ratory of Genetic Engineering (SKLGE1805) to S. H., Y.S. was sup-
diets provided for the patients are different. For instance, ported by the Halfond-Weil Postdoctoral Fellowship.
the fat content and ratio in HFD vary from study to study,
although they are all called HFD. Third, even though the Authors' Contributions  SH concepted  the idea for the article, all
authors contributed to the literature search and data analysis, and YL,
diets fed to animals are better controlled, the diet storage WZ, XL, FS, SH, YS drafted and/or critically revised the work. All
condition also makes difference (Yi et al. 2022). Fourth, authors read and approved the final manuscript.
the environment in different animal facilities affects the
microbiota in the animals. Fifth, the genetic background Data availability  All data generated or analyzed during this study are
included in this published article (and its supplementary information
of the research objects also affects the microbiota (Ahn files).
et al. 2014). A better description of the detailed materials
and methods will be very helpful and necessary in future Declarations 
microbiota-related publications.
The differences in major components in the diets not Conflict of interest  On behalf of all authors, the corresponding author
states that there is no conflict of interest.
only discriminate them from others but also shape specific
patterns of gut microbiota in the hosts. The proportion of
macronutrients, namely fats, carbohydrates and proteins
in the diets varies in KD, HFD, WD, HGD/HFrD, and References
so does the gut microbiota. However, the composition of
the microbiota does not necessarily match the contents of Ahn Y, Narous M, Tobias R, Rho JM, Mychasiuk R (2014) The
the macronutrients. For example, fat-rich HFD and WD ketogenic diet modifies social and metabolic alterations identified
in the prenatal valproic acid model of autism spectrum disorder.
both cause a significant increase in the abundance of Fir-
Dev Neurosci 36(5):371–380. https://d​ oi.o​ rg/1​ 0.1​ 159/0​ 00362​ 645
micutes in the hosts' gut (Bisanz et al. 2019; Low et al. Ahn IS, Lang JM, Olson CA, Diamante G, Zhang G, Ying Z, Byun HR,
2021), but KD, with an even higher level of fat, induces a Cely I, Ding J, Cohn P, Kurtz I, Gomez-Pinilla F, Lusis AJ, Hsiao
dramatic reduction of Firmicutes (Xie et al. 2017; Zhang EY, Yang X (2020) Host genetic background and gut microbiota
contribute to differential metabolic responses to fructose con-
et al. 2018b), and V/VeD, with an extremely low level of
sumption in mice. J Nutr 150(10):2716–2728. https://​doi.​org/​
fat, surprisingly increased the abundance of Firmicutes 10.​1093/​jn/​nxaa2​39
in the gut (Zhang et al. 2018a). The precise connection Alzaben AS, Turner J, Shirton L, Samuel TM, Persad R, Mager D
between the components in the diets and the gut micro- (2015) Assessing nutritional quality and adherence to the glu-
ten-free diet in children and adolescents with celiac disease.
biota still needs a lot of work, with the consideration of
Can J Diet Pract Res 76(2):56–63. https://​doi.​org/​10.​3148/​
the complicated ecological system in the gut. cjdpr-​2014-​040
Here, we review the large body of data that is struc- Ampong I, Watkins A, Gutierrez-Merino J, Ikwuobe J, Griffiths HR
turing our understanding of how the diets alter the gut (2020) Dietary protein insufficiency: an important consideration
in fatty liver disease? Br J Nutr 123(6):601–609. https://​doi.​org/​
microbiota, bacterial metabolites, and the hosts' metabolic
10.​1017/​S0007​11451​90030​64
status. Given the large population and fast dynamic of the Anania C, Pacifico L, Olivero F, Perla FM, Chiesa C (2017) Cardio-
gut microbiota, what we have known about gut microbiota metabolic risk factors in children with celiac disease on a gluten-
is just the tip of the iceberg. Many questions remain unan- free diet. World J Clin Pediatr 6(3):143–148. https://​doi.​org/​10.​
5409/​wjcp.​v6.​i3.​143
swered: How do these altered microbes crosstalk with each

13
Y. Liu et al.

Asghari G, Farhadnejad H, Teymoori F, Mirmiran P, Tohidi M, Azizi Catassi C, Gatti S, Lionetti E (2015) World perspective and celiac
F (2018) High dietary intake of branched-chain amino acids is disease epidemiology. Dig Dis 33(2):141–146. https://​doi.​org/​
associated with an increased risk of insulin resistance in adults. 10.​1159/​00036​9518
J Diabetes 10(5):357–364. https://​doi.​org/​10.​1111/​1753-​0407.​ Chassaing B, Koren O, Goodrich JK, Poole AC, Srinivasan S, Ley RE,
12639 Gewirtz AT (2015) Dietary emulsifiers impact the mouse gut
Badman MK, Kennedy AR, Adams AC, Pissios P, Maratos-Flier microbiota promoting colitis and metabolic syndrome. Nature
E (2009) A very low carbohydrate ketogenic diet improves 519(7541):92–96. https://​doi.​org/​10.​1038/​natur​e14232
glucose tolerance in ob/ob mice independently of weight loss. Craig WJ (2009) Health effects of vegan diets. Am J Clin Nutr
Am J Physiol Endocrinol Metab 297(5):E1197–1204. https://​ 89(5):1627S-1633S. https://​doi.​org/​10.​3945/​ajcn.​2009.​26736N
doi.​org/​10.​1152/​ajpen​do.​00357.​2009 Dao MC, Everard A, Aron-Wisnewsky J, Sokolovska N, Prifti E, Ver-
Baena M, Sanguesa G, Hutter N, Beltran JM, Sanchez RM, Rog- ger EO, Kayser BD, Levenez F, Chilloux J, Hoyles L, Consor-
lans N, Alegret M, Laguna JC (2017) Liquid fructose in West- tium MI-O, Dumas ME, Rizkalla SW, Dore J, Cani PD, Clem-
ern-diet-fed mice impairs liver insulin signaling and causes ent K (2016) Akkermansia muciniphila and improved metabolic
cholesterol and triglyceride loading without changing calorie health during a dietary intervention in obesity: relationship
intake and body weight. J Nutr Biochem 40:105–115. https://​ with gut microbiome richness and ecology. Gut 65(3):426–436.
doi.​org/​10.​1016/j.​jnutb​io.​2016.​10.​015 https://​doi.​org/​10.​1136/​gutjnl-​2014-​308778
Barrea L, Annunziata G, Muscogiuri G, Di Somma C, Laudisio D, Dashti HM, Bo-Abbas YY, Asfar SK, Mathew TC, Hussein T, Behba-
Maisto M, de Alteriis G, Tenore GC, Colao A, Savastano S hani A, Khoursheed MA, Al-Sayer HM, Al-Zaid NS (2003)
(2018) Trimethylamine-N-oxide (TMAO) as novel potential Ketogenic diet modifies the risk factors of heart disease in obese
biomarker of early predictors of metabolic syndrome. Nutrients patients. Nutrition 19(10):901–902. https://​doi.​org/​10.​1016/​
10(12):1971. https://​doi.​org/​10.​3390/​nu101​21971 s0899-​9007(03)​00161-8
Bibbo S, Ianiro G, Giorgio V, Scaldaferri F, Masucci L, Gasbarrini David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE,
A, Cammarota G (2016) The role of diet on gut microbiota Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA,
composition. Eur Rev Med Pharmacol Sci 20(22):4742–4749 Biddinger SB, Dutton RJ, Turnbaugh PJ (2014) Diet rapidly
Bier A, Khasbab R, Haberman Y, Braun T, Hadar R, Sosnovski K, and reproducibly alters the human gut microbiome. Nature
Amir A, Leibowitz A, Grossman E (2020) Antibiotic treat- 505(7484):559–563. https://​doi.​org/​10.​1038/​natur​e12820
ment does not ameliorate the metabolic changes in rats present- De Filippis F, Pellegrini N, Vannini L, Jeffery IB, La Storia A, Laghi
ing dysbiosis after consuming a high fructose diet. Nutrients L, Serrazanetti DI, Di Cagno R, Ferrocino I, Lazzi C, Turroni
12(1):203. https://​doi.​org/​10.​3390/​nu120​10203 S, Cocolin L, Brigidi P, Neviani E, Gobbetti M, O’Toole PW,
Biesiekierski JR (2017) What is gluten? J Gastroenterol Hepatol Ercolini D (2016) High-level adherence to a Mediterranean
32(Suppl 1):78–81. https://​doi.​org/​10.​1111/​jgh.​13703 diet beneficially impacts the gut microbiota and associated
Bisanz JE, Upadhyay V, Turnbaugh JA, Ly K, Turnbaugh PJ (2019) metabolome. Gut 65(11):1812–1821. https://​doi.​org/​10.​1136/​
Meta-analysis reveals reproducible gut microbiome alterations gutjnl-​2015-​309957
in response to a high-fat diet. Cell Host Microbe 26(2):265– De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB,
272. https://​doi.​org/​10.​1016/j.​chom.​2019.​06.​013 Massart S, Collini S, Pieraccini G, Lionetti P (2010) Impact of
Blaut M (2015) Gut microbiota and energy balance: role in obesity. diet in shaping gut microbiota revealed by a comparative study in
Proc Nutr Soc 74(3):227–234. https://​doi.​org/​10.​1017/​S0029​ children from Europe and rural Africa. Proc Natl Acad Sci USA
66511​40017​00 107(33):14691–14696. https://d​ oi.o​ rg/1​ 0.1​ 073/p​ nas.1​ 00596​ 3107
Bonder MJ, Tigchelaar EF, Cai X, Trynka G, Cenit MC, Hrdlickova De Palma G, Nadal I, Collado MC, Sanz Y (2009) Effects of a gluten-
B, Zhong H, Vatanen T, Gevers D, Wijmenga C, Wang Y, free diet on gut microbiota and immune function in healthy adult
Zhernakova A (2016) The influence of a short-term gluten- human subjects. Br J Nutr 102(8):1154–1160. https://​doi.​org/​10.​
free diet on the human gut microbiome. Genome Med 8(1):45. 1017/​S0007​11450​93717​67
https://​doi.​org/​10.​1186/​s13073-​016-​0295-y de Moraes AC, Fernandes GR, da Silva IT, Almeida-Pititto B, Gomes
Bortolin RC, Vargas AR, Gasparotto J, Chaves PR, Schnorr CE, EP, Pereira AD, Ferreira SR (2017) Enterotype may drive the
Martinello KB, Silveira AK, Rabelo TK, Gelain DP, Moreira dietary-associated cardiometabolic risk factors. Front Cell Infect
JCF (2018) A new animal diet based on human Western diet Microbiol 7:47. https://​doi.​org/​10.​3389/​fcimb.​2017.​00047
is a robust diet-induced obesity model: comparison to high-fat Deng P, Durham J, Liu J, Zhang X, Wang C, Li D, Gwag T, Ma M,
and cafeteria diets in term of metabolic and gut microbiota Hennig B (2022) Metabolomic, lipidomic, transcriptomic,
disruption. Int J Obes (lond) 42(3):525–534. https://​doi.​org/​ and metagenomic analyses in mice exposed to pfos and fed
10.​1038/​ijo.​2017.​225 soluble and insoluble dietary fibers. Environ Health Perspect
Brahe LK, Le Chatelier E, Prifti E, Pons N, Kennedy S, Hansen T, 130(11):117003. https://​doi.​org/​10.​1289/​EHP11​360
Pedersen O, Astrup A, Ehrlich SD, Larsen LH (2015) Specific Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-
gut microbiota features and metabolic markers in postmeno- Silva S, Falony G, Raes J, Maiter D, Delzenne NM, de Barsy M,
pausal women with obesity. Nutr Diab 5:159. https://​doi.​org/​ Loumaye A, Hermans MP, Thissen JP, de Vos WM, Cani PD
10.​1038/​nutd.​2015.9 (2019) Supplementation with Akkermansia muciniphila in over-
Candido FG, Valente FX, Grzeskowiak LM, Moreira APB, Rocha D, weight and obese human volunteers: a proof-of-concept explora-
Alfenas RCG (2018) Impact of dietary fat on gut microbiota tory study. Nat Med 25(7):1096–1103. https://​doi.​org/​10.​1038/​
and low-grade systemic inflammation: mechanisms and clinical s41591-​019-​0495-2
implications on obesity. Int J Food Sci Nutr 69(2):125–143. Derrien M, Vaughan EE, Plugge CM, de Vos WM (2004) Akkermansia
https://​doi.​org/​10.​1080/​09637​486.​2017.​13432​86 muciniphila gen. nov., sp. nov., a human intestinal mucin-degrad-
Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne ing bacterium. Int J Syst Evol Microbiol 54(Pt 5):1469–1476.
NM, Burcelin R (2008) Changes in gut microbiota control https://​doi.​org/​10.​1099/​ijs.0.​02873-0
metabolic endotoxemia-induced inflammation in high-fat diet- Devkota S, Wang Y, Musch MW, Leone V, Fehlner-Peach H, Nadim-
induced obesity and diabetes in mice. Diabetes 57(6):1470– palli A, Antonopoulos DA, Jabri B, Chang EB (2012) Dietary-
1481. https://​doi.​org/​10.​2337/​db07-​1403 fat-induced taurocholic acid promotes pathobiont expansion and

13
Diets, Gut Microbiota and Metabolites

colitis in Il10–/– mice. Nature 487(7405):104–108. https://​doi.​ characterizes obese adolescents with non-alcoholic fatty liver
org/​10.​1038/​natur​e11225 disease. Nutrients 9(7):642. https://​doi.​org/​10.​3390/​nu907​0642
Di Cagno R, De Angelis M, De Pasquale I, Ndagijimana M, Vernocchi Gutierrez-Repiso C, Hernandez-Garcia C, Garcia-Almeida JM, Bellido
P, Ricciuti P, Gagliardi F, Laghi L, Crecchio C, Guerzoni ME, D, Martin-Nunez GM, Sanchez-Alcoholado L, Alcaide-Torres J,
Gobbetti M, Francavilla R (2011) Duodenal and faecal micro- Sajoux I, Tinahones FJ, Moreno-Indias I (2019) Effect of syn-
biota of celiac children: molecular, phenotype and metabolome biotic supplementation in a very-low-calorie ketogenic diet on
characterization. BMC Microbiol 11:219. https://​doi.​org/​10.​ weight loss achievement and gut microbiota: a randomized con-
1186/​1471-​2180-​11-​219 trolled pilot study. Mol Nutr Food Res 63(19):e1900167. https://​
Di Luccia B, Crescenzo R, Mazzoli A, Cigliano L, Venditti P, Wal- doi.​org/​10.​1002/​mnfr.​20190​0167
ser JC, Widmer A, Baccigalupi L, Ricca E, Iossa S (2015) Haddad EH, Berk LS, Kettering JD, Hubbard RW, Peters WR (1999)
Rescue of fructose-induced metabolic syndrome by antibiotics Dietary intake and biochemical, hematologic, and immune status
or faecal transplantation in a rat model of obesity. PLoS ONE of vegans compared with nonvegetarians. Am J Clin Nutr 70(3
10(8):e0134893. https://​doi.​org/​10.​1371/​journ​al.​pone.​01348​93 Suppl):586S-593S. https://​doi.​org/​10.​1093/​ajcn/​70.3.​586s
Do MH, Lee E, Oh MJ, Kim Y, Park HY (2018) High-glucose or -fruc- Haupt-Jorgensen M, Holm LJ, Josefsen K, Buschard K (2018) Pos-
tose diet cause changes of the gut microbiota and metabolic dis- sible prevention of diabetes with a gluten-free diet. Nutrients
orders in mice without body weight change. Nutrients 10(6):761. 10(11):1746. https://​doi.​org/​10.​3390/​nu101​11746
https://​doi.​org/​10.​3390/​nu100​60761 Heianza Y, Sun D, Li X, DiDonato JA, Bray GA, Sacks FM, Qi L
Ellenbroek JH, van Dijck L, Tons HA, Rabelink TJ, Carlotti F, Bal- (2019) Gut microbiota metabolites, amino acid metabolites and
lieux BE, de Koning EJ (2014) Long-term ketogenic diet causes improvements in insulin sensitivity and glucose metabolism: the
glucose intolerance and reduced beta- and alpha-cell mass POUNDS Lost trial. Gut 68(2):263–270. https://d​ oi.o​ rg/1​ 0.1​ 136/​
but no weight loss in mice. Am J Physiol Endocrinol Metab gutjnl-​2018-​316155
306(5):E552-558. https://​doi.​org/​10.​1152/​ajpen​do.​00453.​2013 Hernandez MAG, Canfora EE, Jocken JWE, Blaak EE (2019) The
Everard A, Lazarevic V, Gaia N, Johansson M, Stahlman M, Backhed short-chain fatty acid acetate in body weight control and insulin
F, Delzenne NM, Schrenzel J, Francois P, Cani PD (2014) Micro- sensitivity. Nutrients 11(8):1943. https://​doi.​org/​10.​3390/​nu110​
biome of prebiotic-treated mice reveals novel targets involved in 81943
host response during obesity. ISME J 8(10):2116–2130. https://​ Hildebrandt MA, Hoffmann C, Sherrill-Mix SA, Keilbaugh SA, Ham-
doi.​org/​10.​1038/​ismej.​2014.​45 ady M, Chen YY, Knight R, Ahima RS, Bushman F, Wu GD
Fu BC, Hullar MAJ, Randolph TW, Franke AA, Monroe KR, Cheng (2009) High-fat diet determines the composition of the murine
I, Wilkens LR, Shepherd JA, Madeleine MM, Le Marchand L, gut microbiome independently of obesity. Gastroenterology
Lim U, Lampe JW (2020) Associations of plasma trimethylamine 137(5):1716–1724 e1711–1712. https://​doi.​org/​10.​1053/j.​gas-
N-oxide, choline, carnitine, and betaine with inflammatory and tro.​2009.​08.​042
cardiometabolic risk biomarkers and the fecal microbiome in the Hohn S, Dozieres-Puyravel B, Auvin S (2019) History of dietary treat-
Multiethnic Cohort Adiposity Phenotype Study. Am J Clin Nutr ment from Wilder's hypothesis to the first open studies in the
111(6):1226–1234. https://​doi.​org/​10.​1093/​ajcn/​nqaa0​15 1920s. Epilepsy Behav 101(Pt A):106588. https://​doi.​org/​10.​
Garbow JR, Doherty JM, Schugar RC, Travers S, Weber ML, Wentz 1016/j.​yebeh.​2019.​106588
AE, Ezenwajiaku N, Cotter DG, Brunt EM, Crawford PA (2011) Horton F, Wright J, Smith L, Hinton PJ, Robertson MD (2014)
Hepatic steatosis, inflammation, and ER stress in mice main- Increased intestinal permeability to oral chromium (51 Cr)
tained long term on a very low-carbohydrate ketogenic diet. Am -EDTA in human Type 2 diabetes. Diabet Med 31(5):559–563.
J Physiol Gastrointest Liver Physiol 300(6):G956-967. https://​ https://​doi.​org/​10.​1111/​dme.​12360
doi.​org/​10.​1152/​ajpgi.​00539.​2010 Imperatore N, Tortora R, Testa A, Gerbino N, Caporaso N, Rispo A
Garcia-Mantrana I, Selma-Royo M, Alcantara C, Collado MC (2018) (2018) Proton pump inhibitors as risk factor for metabolic syn-
Shifts on gut microbiota associated to Mediterranean diet adher- drome and hepatic steatosis in coeliac disease patients on gluten-
ence and specific dietary intakes on general adult population. free diet. J Gastroenterol 53(4):507–516. https://d​ oi.o​ rg/1​ 0.1​ 007/​
Front Microbiol 9:890. https://​doi.​org/​10.​3389/​fmicb.​2018.​ s00535-​017-​1381-7
00890 Jenkins DJ, Wong JM, Kendall CW, Esfahani A, Ng VW, Leong TC,
Garcia-Mazcorro JF, Noratto G, Remes-Troche JM (2018) The Faulkner DA, Vidgen E, Paul G, Mukherjea R, Krul ES, Singer
effect of gluten-free diet on health and the gut microbiota can- W (2014) Effect of a 6-month vegan low-carbohydrate ('Eco-
not be extrapolated from one population to others. Nutrients Atkins') diet on cardiovascular risk factors and body weight in
10(10):1421. https://​doi.​org/​10.​3390/​nu101​01421 hyperlipidaemic adults: a randomised controlled trial. BMJ Open
Gentile CL, Weir TL (2018) The gut microbiota at the intersection of 4(2):e003505. https://​doi.​org/​10.​1136/​bmjop​en-​2013-​003505
diet and human health. Science 362(6416):776–780. https://​doi.​ Johnston RD, Stephenson MC, Crossland H, Cordon SM, Palcidi E,
org/​10.​1126/​scien​ce.​aau58​12 Cox EF, Taylor MA, Aithal GP, Macdonald IA (2013) No dif-
Ghosh TS, Rampelli S, Jeffery IB, Santoro A, Neto M, Capri M, ference between high-fructose and high-glucose diets on liver
Giampieri E, Jennings A, Candela M, Turroni S, Zoetendal EG, triacylglycerol or biochemistry in healthy overweight men.
Hermes GDA, Elodie C, Meunier N, Brugere CM, Pujos-Guillot Gastroenterology 145(5):1016–1025. https://​doi.​org/​10.​1053/j.​
E, Berendsen AM, De Groot L, Feskins EJM, Kaluza J, Pie- gastro.​2013.​07.​012
truszka B, Bielak MJ, Comte B, Maijo-Ferre M, Nicoletti C, De Jornayvaz FR, Jurczak MJ, Lee HY, Birkenfeld AL, Frederick DW,
Vos WM, Fairweather-Tait S, Cassidy A, Brigidi P, Franceschi Zhang D, Zhang XM, Samuel VT, Shulman GI (2010) A high-
C, O’Toole PW (2020) Mediterranean diet intervention alters the fat, ketogenic diet causes hepatic insulin resistance in mice,
gut microbiome in older people reducing frailty and improving despite increasing energy expenditure and preventing weight
health status: the NU-AGE 1-year dietary intervention across gain. Am J Physiol Endocrinol Metab 299(5):E808–815. https://​
five European countries. Gut 69(7):1218–1228. https://​doi.​org/​ doi.​org/​10.​1152/​ajpen​do.​00361.​2010
10.​1136/​gutjnl-​2019-​319654 Kastorini CM, Milionis HJ, Esposito K, Giugliano D, Goudevenos JA,
Goffredo M, Santoro N, Trico D, Giannini C, D’Adamo E, Zhao Panagiotakos DB (2011) The effect of Mediterranean diet on
H, Peng G, Yu X, Lam TT, Pierpont B, Caprio S, Herzog RI metabolic syndrome and its components: a meta-analysis of 50
(2017) A branched-chain amino acid-related metabolic signature

13
Y. Liu et al.

studies and 534,906 individuals. J Am Coll Cardiol 57(11):1299– Liu R, Hong J, Xu X, Feng Q, Zhang D, Gu Y, Shi J, Zhao S, Liu
1313. https://​doi.​org/​10.​1016/j.​jacc.​2010.​09.​073 W, Wang X, Xia H, Liu Z, Cui B, Liang P, Xi L, Jin J, Ying
Kaye DM, Shihata WA, Jama HA, Tsyganov K, Ziemann M, Kiriazis X, Wang X, Zhao X, Li W, Jia H, Lan Z, Li F, Wang R, Sun
H, Horlock D, Vijay A, Giam B, Vinh A, Johnson C, Fiedler A, Y, Yang M, Shen Y, Jie Z, Li J, Chen X, Zhong H, Xie H,
Donner D, Snelson M, Coughlan MT, Phillips S, Du XJ, El-Osta Zhang Y, Gu W, Deng X, Shen B, Xu X, Yang H, Xu G, Bi Y,
A, Drummond G, Lambert GW, Spector TD, Valdes AM, Mac- Lai S, Wang J, Qi L, Madsen L, Wang J, Ning G, Kristiansen
kay CR, Marques FZ (2020) Deficiency of prebiotic fiber and K, Wang W (2017) Gut microbiome and serum metabolome
insufficient signaling through gut metabolite-sensing receptors alterations in obesity and after weight-loss intervention. Nat
leads to cardiovascular disease. Circulation 141(17):1393–1403. Med 23(7):859–868. https://​doi.​org/​10.​1038/​nm.​4358
https://​doi.​org/​10.​1161/​CIRCU​LATIO​NAHA.​119.​043081 Liu JL, Segovia I, Yuan XL, Gao ZH (2020) Controversial roles
Kemppainen T, Uusitupa M, Janatuinen E, Jarvinen R, Julkunen R, of gut microbiota-derived short-chain fatty acids (SCFAs) on
Pikkarainen P (1995) Intakes of nutrients and nutritional status pancreatic beta-cell growth and insulin secretion. Int J Mol Sci
in coeliac patients. Scand J Gastroenterol 30(6):575–579. https://​ 21(3):910. https://​doi.​org/​10.​3390/​ijms2​10309​10
doi.​org/​10.​3109/​00365​52950​90897​92 Low A, Soh M, Miyake S, Aw VZJ, Feng J, Wong A, Seedorf H
Kim E, Kim DB, Park JY (2016) Changes of mouse gut microbiota (2021) Longitudinal changes in diet cause repeatable and
diversity and composition by modulating dietary protein and car- largely reversible shifts in gut microbial communities of labo-
bohydrate contents: a pilot study. Prev Nutr Food Sci 21(1):57– ratory mice and are observed across segments of the entire
61. https://​doi.​org/​10.​3746/​pnf.​2016.​21.1.​57 intestinal tract. Int J Mol Sci 22(11):5981. https://​doi.​org/​10.​
Kirkland TN, Ziegler EJ (1984) An immunoprotective monoclonal 3390/​ijms2​21159​81
antibody to lipopolysaccharide. J Immunol 132(5):2590–2592. Ma J, Zhou Q, Li H (2017) Gut microbiota and nonalcoholic fatty
https://​doi.​org/​10.​4049/​jimmu​nol.​132.5.​2590 liver disease: insights on mechanisms and therapy. Nutrients
Koh A, De Vadder F, Kovatcheva-Datchary P, Backhed F (2016) From 9(10):1124. https://​doi.​org/​10.​3390/​nu910​1124
dietary fiber to host physiology: short-chain fatty acids as key Ma D, Wang AC, Parikh I, Green SJ, Hoffman JD, Chlipala G, Murphy
bacterial metabolites. Cell 165(6):1332–1345. https://d​ oi.o​ rg/1​ 0.​ MP, Sokola BS, Bauer B, Hartz AMS, Lin AL (2018) Ketogenic
1016/j.​cell.​2016.​05.​041 diet enhances neurovascular function with altered gut microbi-
Kong C, Yan X, Liu Y, Huang L, Zhu Y, He J, Gao R, Kalady MF, ome in young healthy mice. Sci Rep 8(1):6670. https://​doi.​org/​
Goel A, Qin H, Ma Y (2021) Ketogenic diet alleviates colitis 10.​1038/​s41598-​018-​25190-5
by reduction of colonic group 3 innate lymphoid cells through Macfarlane GT, Macfarlane S (2011) Fermentation in the human large
altering gut microbiome. Signal Transduct Target Ther 6(1):154. intestine: its physiologic consequences and the potential contri-
https://​doi.​org/​10.​1038/​s41392-​021-​00549-9 bution of prebiotics. J Clin Gastroenterol 45(Suppl):S120–127.
Kosinski C, Jornayvaz FR (2017) Effects of ketogenic diets on cardio- https://​doi.​org/​10.​1097/​MCG.​0b013​e3182​2fecfe
vascular risk factors: evidence from animal and human studies. Machiels K, Joossens M, Sabino J, De Preter V, Arijs I, Eeckhaut V,
Nutrients 9(5):517. https://​doi.​org/​10.​3390/​nu905​0517 Ballet V, Claes K, Van Immerseel F, Verbeke K, Ferrante M,
Laterza L, Rizzatti G, Gaetani E, Chiusolo P, Gasbarrini A (2016) Verhaegen J, Rutgeerts P, Vermeire S (2014) A decrease of the
The gut microbiota and immune system relationship in human butyrate-producing species Roseburia hominis and Faecali-
graft-versus-host disease. Mediterr J Hematol Infect Dis bacterium prausnitzii defines dysbiosis in patients with ulcera-
8(1):e2016025. https://​doi.​org/​10.​4084/​MJHID.​2016.​025 tive colitis. Gut 63(8):1275–1283. https://​doi.​org/​10.​1136/​
Le LT, Sabate J (2014) Beyond meatless, the health effects of vegan gutjnl-​2013-​304833
diets: findings from the Adventist cohorts. Nutrients 6(6):2131– Magne F, Gotteland M, Gauthier L, Zazueta A, Pesoa S, Navarrete
2147. https://​doi.​org/​10.​3390/​nu606​2131 P, Balamurugan R (2020) The firmicutes/bacteroidetes ratio:
Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon a relevant marker of gut dysbiosis in obese patients? Nutrients
JI (2005) Obesity alters gut microbial ecology. Proc Natl Acad 12(5):1474. https://​doi.​org/​10.​3390/​nu120​51474
Sci USA 102(31):11070–11075. https://​doi.​org/​10.​1073/​pnas.​ Manco M, Putignani L, Bottazzo GF (2010) Gut microbiota, lipopoly-
05049​78102 saccharides, and innate immunity in the pathogenesis of obesity
Li T, Geng L, Chen X, Miskowiec M, Li X, Dong B (2013) Branched- and cardiovascular risk. Endocr Rev 31(6):817–844. https://​doi.​
chain amino acids alleviate nonalcoholic steatohepatitis in rats. org/​10.​1210/​er.​2009-​0030
Appl Physiol Nutr Metab 38(8):836–843. https://​doi.​org/​10.​ Mariani P, Viti MG, Montuori M, La Vecchia A, Cipolletta E, Calvani
1139/​apnm-​2012-​0496 L, Bonamico M (1998) The gluten-free diet: a nutritional risk
Li J, Zhao F, Wang Y, Chen J, Tao J, Tian G, Wu S, Liu W, Cui Q, factor for adolescents with celiac disease? J Pediatr Gastroenterol
Geng B, Zhang W, Weldon R, Auguste K, Yang L, Liu X, Chen Nutr 27(5):519–523. https://​doi.​org/​10.​1097/​00005​176-​19981​
L, Yang X, Zhu B, Cai J (2017) Gut microbiota dysbiosis contrib- 1000-​00004
utes to the development of hypertension. Microbiome 5(1):14. Mastrocola R, Ferrocino I, Liberto E, Chiazza F, Cento AS, Collotta D,
https://​doi.​org/​10.​1186/​s40168-​016-​0222-x Querio G, Nigro D, Bitonto V, Cutrin JC, Rantsiou K, Durante
Li Y, Yang X, Zhang J, Jiang T, Zhang Z, Wang Z, Gong M, Zhao L, M, Masini E, Aragno M, Cordero C, Cocolin L, Collino M
Zhang C (2021) Ketogenic diets induced glucose intolerance and (2018) Fructose liquid and solid formulations differently affect
lipid accumulation in mice with alterations in gut microbiota and gut integrity, microbiota composition and related liver toxicity: a
metabolites. mBio 12(2). https://d​ oi.o​ rg/1​ 0.1​ 128/m
​ Bio.0​ 3601-2​ 0 comparative in vivo study. J Nutr Biochem 55:185–199. https://​
Lindefeldt M, Eng A, Darban H, Bjerkner A, Zetterstrom CK, Allander doi.​org/​10.​1016/j.​jnutb​io.​2018.​02.​003
T, Andersson B, Borenstein E, Dahlin M, Prast-Nielsen S (2019) Mitsou EK, Kakali A, Antonopoulou S, Mountzouris KC, Yannak-
The ketogenic diet influences taxonomic and functional com- oulia M, Panagiotakos DB, Kyriacou A (2017) Adherence to the
position of the gut microbiota in children with severe epilepsy. Mediterranean diet is associated with the gut microbiota pattern
NPJ Biofilms Microbiomes 5(1):5. https://​doi.​org/​10.​1038/​ and gastrointestinal characteristics in an adult population. Br J
s41522-​018-​0073-2 Nutr 117(12):1645–1655. https://​doi.​org/​10.​1017/​S0007​11451​
Lionetti E, Gatti S, Pulvirenti A, Catassi C (2015) Celiac disease from a 70015​93
global perspective. Best Pract Res Clin Gastroenterol 29(3):365– Montrose DC, Nishiguchi R, Basu S, Staab HA, Zhou XK, Wang H,
379. https://​doi.​org/​10.​1016/j.​bpg.​2015.​05.​004 Meng L, Johncilla M, Cubillos-Ruiz JR, Morales DK, Wells MT,

13
Diets, Gut Microbiota and Metabolites

Simpson KW, Zhang S, Dogan B, Jiao C, Fei Z, Oka A, Herzog Qi X, Yun C, Sun L, Xia J, Wu Q, Wang Y, Wang L, Zhang Y, Liang X,
JW, Sartor RB, Dannenberg AJ (2021) Dietary fructose alters the Wang L, Gonzalez FJ, Patterson AD, Liu H, Mu L, Zhou Z, Zhao
composition, localization, and metabolism of gut microbiota in Y, Li R, Liu P, Zhong C, Pang Y, Jiang C, Qiao J (2019) Gut
association with worsening colitis. Cell Mol Gastroenterol Hepa- microbiota-bile acid-interleukin-22 axis orchestrates polycystic
tol 11(2):525–550. https://​doi.​org/​10.​1016/j.​jcmgh.​2020.​09.​008 ovary syndrome. Nat Med 25(8):1225–1233. https://​doi.​org/​10.​
Morrison DJ, Preston T (2016) Formation of short chain fatty acids 1038/​s41591-​019-​0509-0
by the gut microbiota and their impact on human metabolism. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, Liang S, Zhang W, Guan Y,
Gut Microbes 7(3):189–200. https://​doi.​org/​10.​1080/​19490​976.​ Shen D, Peng Y, Zhang D, Jie Z, Wu W, Qin Y, Xue W, Li J, Han
2015.​11340​82 L, Lu D, Wu P, Dai Y, Sun X, Li Z, Tang A, Zhong S, Li X, Chen
Munch NS, Fang HY, Ingermann J, Maurer HC, Anand A, Kellner W, Xu R, Wang M, Feng Q, Gong M, Yu J, Zhang Y, Zhang M,
V, Sahm V, Wiethaler M, Baumeister T, Wein F, Einwachter Hansen T, Sanchez G, Raes J, Falony G, Okuda S, Almeida M,
H, Bolze F, Klingenspor M, Haller D, Kavanagh M, Lysaght J, LeChatelier E, Renault P, Pons N, Batto JM, Zhang Z, Chen H,
Friedman R, Dannenberg AJ, Pollak M, Holt PR, Muthupalani Yang R, Zheng W, Li S, Yang H, Wang J, Ehrlich SD, Nielsen R,
S, Fox JG, Whary MT, Lee Y, Ren TY, Elliot R, Fitzgerald R, Pedersen O, Kristiansen K, Wang J (2012) A metagenome-wide
Steiger K, Schmid RM, Wang TC, Quante M (2019) High-fat diet association study of gut microbiota in type 2 diabetes. Nature
accelerates carcinogenesis in a mouse model of barrett's esopha- 490(7418):55–60. https://​doi.​org/​10.​1038/​natur​e11450
gus via interleukin 8 and alterations to the gut microbiome. Gas- Raza MH, Gul K, Arshad A, Riaz N, Waheed U, Rauf A, Aldakheel
troenterology 157(2):492–506. https://​doi.​org/​10.​1053/j.​gastro.​ F, Alduraywish S, Rehman MU, Abdullah M, Arshad M
2019.​04.​013 (2019) Microbiota in cancer development and treatment. J
Neis EP, Dejong CH, Rensen SS (2015) The role of microbial amino Cancer Res Clin Oncol 145(1):49–63. https://​doi.​org/​10.​1007/​
acid metabolism in host metabolism. Nutrients 7(4):2930–2946. s00432-​018-​2816-0
https://​doi.​org/​10.​3390/​nu704​2930 Requena T, Martinez-Cuesta MC, Pelaez C (2018) Diet and microbiota
Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF, linked in health and disease. Food Funct 9(2):688–704. https://​
Haqq AM, Shah SH, Arlotto M, Slentz CA, Rochon J, Gallup doi.​org/​10.​1039/​c7fo0​1820g
D, Ilkayeva O, Wenner BR, Yancy WS Jr, Eisenson H, Musante Rinninella E, Cintoni M, Raoul P, Lopetuso LR, Scaldaferri F, Pulcini
G, Surwit RS, Millington DS, Butler MD, Svetkey LP (2009) G, Miggiano GAD, Gasbarrini A, Mele MC (2019) Food com-
A branched-chain amino acid-related metabolic signature that ponents and dietary habits: keys for a healthy gut microbiota
differentiates obese and lean humans and contributes to insulin composition. Nutrients 11(10):2393. https://​doi.​org/​10.​3390/​
resistance. Cell Metab 9(4):311–326. https://​doi.​org/​10.​1016/j.​ nu111​02393
cmet.​2009.​02.​002 Rios-Covian D, Ruas-Madiedo P, Margolles A, Gueimonde M, de Los
Newnham ED (2017) Coeliac disease in the 21st century: paradigm Reyes-Gavilan CG, Salazar N (2016) Intestinal short chain fatty
shifts in the modern age. J Gastroenterol Hepatol 32(Suppl acids and their link with diet and human health. Front Microbiol
1):82–85. https://​doi.​org/​10.​1111/​jgh.​13704 7:185. https://​doi.​org/​10.​3389/​fmicb.​2016.​00185
O’Keefe SJ, Li JV, Lahti L, Ou J, Carbonero F, Mohammed K, Posma Rivero-Gutierrez B, Gamez-Belmonte R, Suarez MD, Lavin JL,
JM, Kinross J, Wahl E, Ruder E, Vipperla K, Naidoo V, Mtshali Aransay AM, Olivares M, Martinez-Augustin O, Sanchez de
L, Tims S, Puylaert PG, DeLany J, Krasinskas A, Benefiel AC, Medina F, Zarzuelo A (2017) A synbiotic composed of Lacto-
Kaseb HO, Newton K, Nicholson JK, de Vos WM, Gaskins HR, bacillus fermentum CECT5716 and FOS prevents the develop-
Zoetendal EG (2015) Fat, fibre and cancer risk in African Ameri- ment of fatty acid liver and glycemic alterations in rats fed a
cans and rural Africans. Nat Commun 6:6342. https://d​ oi.o​ rg/1​ 0.​ high fructose diet associated with changes in the microbiota.
1038/​ncomm​s7342 Mol Nutr Food Res 61(8):1600622. https://d​ oi.o​ rg/1​ 0.1​ 002/m
​ nfr.​
Olson CA, Vuong HE, Yano JM, Liang QY, Nusbaum DJ, Hsiao EY 20160​0622
(2018) The gut microbiota mediates the anti-seizure effects of Rizzatti G, Lopetuso LR, Gibiino G, Binda C, Gasbarrini A (2017)
the ketogenic diet. Cell 173(7):1728–1741. https://​doi.​org/​10.​ Proteobacteria: a common factor in human diseases. Biomed Res
1016/j.​cell.​2018.​04.​027 Int 2017:9351507. https://​doi.​org/​10.​1155/​2017/​93515​07
Pascale A, Marchesi N, Govoni S, Coppola A, Gazzaruso C (2019) The Romualdo GR, Valente LC, Sprocatti AC, Bacil GP, de Souza IP, Rod-
role of gut microbiota in obesity, diabetes mellitus, and effect of rigues J, Rodrigues MAM, Vinken M, Cogliati B, Barbisan LF
metformin: new insights into old diseases. Curr Opin Pharmacol (2022) Western diet-induced mouse model of non-alcoholic fatty
49:1–5. https://​doi.​org/​10.​1016/j.​coph.​2019.​03.​011 liver disease associated with metabolic outcomes: Features of gut
Pathak P, Xie C, Nichols RG, Ferrell JM, Boehme S, Krausz KW, Pat- microbiome-liver-adipose tissue axis. Nutrition 103–104:111836.
terson AD, Gonzalez FJ, Chiang JYL (2018) Intestine farnesoid https://​doi.​org/​10.​1016/j.​nut.​2022.​111836
X receptor agonist and the gut microbiota activate G-protein bile Rusek M, Pluta R, Ulamek-Koziol M, Czuczwar SJ (2019) Ketogenic
acid receptor-1 signaling to improve metabolism. Hepatology diet in Alzheimer’s disease. Int J Mol Sci 20(16):3892. https://​
68(4):1574–1588. https://​doi.​org/​10.​1002/​hep.​29857 doi.​org/​10.​3390/​ijms2​01638​92
Pedersen HK, Gudmundsdottir V, Nielsen HB, Hyotylainen T, Nielsen Ryan MC, Itsiopoulos C, Thodis T, Ward G, Trost N, Hofferberth S,
T, Jensen BA, Forslund K, Hildebrand F, Prifti E, Falony G, Le O’Dea K, Desmond PV, Johnson NA, Wilson AM (2013) The
Chatelier E, Levenez F, Dore J, Mattila I, Plichta DR, Poho P, Mediterranean diet improves hepatic steatosis and insulin sen-
Hellgren LI, Arumugam M, Sunagawa S, Vieira-Silva S, Jor- sitivity in individuals with non-alcoholic fatty liver disease. J
gensen T, Holm JB, Trost K, Meta HITC, Kristiansen K, Brix S, Hepatol 59(1):138–143. https://​doi.​org/​10.​1016/j.​jhep.​2013.​02.​
Raes J, Wang J, Hansen T, Bork P, Brunak S, Oresic M, Ehrlich 012
SD, Pedersen O (2016) Human gut microbes impact host serum Saad MJ, Santos A, Prada PO (2016) Linking gut microbiota and
metabolome and insulin sensitivity. Nature 535(7612):376–381. inflammation to obesity and insulin resistance. Physiology
https://​doi.​org/​10.​1038/​natur​e18646 (bethesda) 31(4):283–293. https://​d oi.​o rg/​1 0.​1 152/​p hysi​o l.​
Pilis W, Stec K, Zych M, Pilis A (2014) Health benefits and risk asso- 00041.​2015
ciated with adopting a vegetarian diet. Rocz Panstw Zakl Hig Salas-Salvado J, Bullo M, Estruch R, Ros E, Covas MI, Ibarrola-
65(1):9–14 Jurado N, Corella D, Aros F, Gomez-Gracia E, Ruiz-Gutierrez V,
Romaguera D, Lapetra J, Lamuela-Raventos RM, Serra-Majem

13
Y. Liu et al.

L, Pinto X, Basora J, Munoz MA, Sorli JV, Martinez-Gonzalez ketogenic diet. Front Microbiol 8:1141. https://​doi.​org/​10.​3389/​
MA (2014) Prevention of diabetes with Mediterranean diets: fmicb.​2017.​01141
a subgroup analysis of a randomized trial. Ann Intern Med Tagliabue A, Ferraris C, Uggeri F, Trentani C, Bertoli S, de Giorgis
160(1):1–10. https://​doi.​org/​10.​7326/​M13-​1725 V, Veggiotti P, Elli M (2017) Short-term impact of a classical
Sanz Y (2010) Effects of a gluten-free diet on gut microbiota and ketogenic diet on gut microbiota in GLUT1 deficiency syndrome:
immune function in healthy adult humans. Gut Microbes a 3-month prospective observational study. Clin Nutr ESPEN
1(3):135–137. https://​doi.​org/​10.​4161/​gmic.1.​3.​11868 17:33–37. https://​doi.​org/​10.​1016/j.​clnesp.​2016.​11.​003
Schneeberger M, Everard A, Gomez-Valades AG, Matamoros S, Tang WHW, Backhed F, Landmesser U, Hazen SL (2019) Intestinal
Ramirez S, Delzenne NM, Gomis R, Claret M, Cani PD (2015) microbiota in cardiovascular health and disease: JACC state-of-
Akkermansia muciniphila inversely correlates with the onset of the-art review. J Am Coll Cardiol 73(16):2089–2105. https://d​ oi.​
inflammation, altered adipose tissue metabolism and metabolic org/​10.​1016/j.​jacc.​2019.​03.​024
disorders during obesity in mice. Sci Rep 5:16643. https://​doi.​ Tindall AM, Petersen KS, Kris-Etherton PM (2018) Dietary patterns
org/​10.​1038/​srep1​6643 affect the gut microbiome—the link to risk of cardiometabolic
Schroeder BO, Birchenough GMH, Stahlman M, Arike L, Johansson diseases. J Nutr 148(9):1402–1407. https://​doi.​org/​10.​1093/​jn/​
MEV, Hansson GC, Backhed F (2018) Bifidobacteria or fiber nxy141
protects against diet-induced microbiota-mediated colonic mucus Tiniakos DG, Vos MB, Brunt EM (2010) Nonalcoholic fatty liver dis-
deterioration. Cell Host Microbe 23(1):27–40. https://d​ oi.o​ rg/1​ 0.​ ease: pathology and pathogenesis. Annu Rev Pathol 5:145–171.
1016/j.​chom.​2017.​11.​004 https://​doi.​org/​10.​1146/​annur​ev-​pathol-​121808-​102132
Schugar RC, Shih DM, Warrier M, Helsley RN, Burrows A, Ferguson Tonstad S, Stewart K, Oda K, Batech M, Herring RP, Fraser GE
D, Brown AL, Gromovsky AD, Heine M, Chatterjee A, Li L, Li (2013) Vegetarian diets and incidence of diabetes in the Advent-
XS, Wang Z, Willard B, Meng Y, Kim H, Che N, Pan C, Lee RG, ist Health Study-2. Nutr Metab Cardiovasc Dis 23(4):292–299.
Crooke RM, Graham MJ, Morton RE, Langefeld CD, Das SK, https://​doi.​org/​10.​1016/j.​numecd.​2011.​07.​004
Rudel LL, Zein N, McCullough AJ, Dasarathy S, Tang WHW, Tovoli F, Negrini G, Fari R, Guidetti E, Faggiano C, Napoli L, Bolondi
Erokwu BO, Flask CA, Laakso M, Civelek M, Naga Prasad SV, L, Granito A (2018) Increased risk of nonalcoholic fatty liver
Heeren J, Lusis AJ, Hazen SL, Brown JM (2017) The TMAO- disease in patients with coeliac disease on a gluten-free diet:
producing enzyme flavin-containing monooxygenase 3 regu- beyond traditional metabolic factors. Aliment Pharmacol Ther
lates obesity and the Beiging of white adipose tissue. Cell Rep 48(5):538–546. https://​doi.​org/​10.​1111/​apt.​14910
20(1):279. https://​doi.​org/​10.​1016/j.​celrep.​2017.​06.​053 Trefflich I, Marschall HU, Giuseppe RD, Stahlman M, Michalsen A,
Shan Z, Sun T, Huang H, Chen S, Chen L, Luo C, Yang W, Yang Lampen A, Abraham K, Weikert C (2019) Associations between
X, Yao P, Cheng J, Hu FB, Liu L (2017) Association between dietary patterns and bile acids-results from a cross-sectional
microbiota-dependent metabolite trimethylamine-N-oxide and study in vegans and omnivores. Nutrients 12(1):47. https://​doi.​
type 2 diabetes. Am J Clin Nutr 106(3):888–894. https://​doi.​org/​ org/​10.​3390/​nu120​10047
10.​3945/​ajcn.​117.​157107 Trefflich I, Jabakhanji A, Menzel J, Blaut M, Michalsen A, Lampen
Silva JCP, Mota M, Martins FO, Nogueira C, Goncalves T, Carneiro T, A, Abraham K, Weikert C (2020) Is a vegan or a vegetarian diet
Pinto J, Duarte D, Barros AS, Jones JG, Gil AM (2018) Intestinal associated with the microbiota composition in the gut? Results
microbial and metabolic profiling of mice fed with high-glucose of a new cross-sectional study and systematic review. Crit Rev
and high-fructose diets. J Proteome Res 17(8):2880–2891. Food Sci Nutr 60(17):2990–3004. https://d​ oi.o​ rg/1​ 0.1​ 080/1​ 0408​
https://​doi.​org/​10.​1021/​acs.​jprot​eome.​8b003​54 398.​2019.​16766​97
Simkin DR (2019) Microbiome and mental health, specifically as it Tsukumo DM, Carvalho BM, Carvalho-Filho MA, Saad MJ (2009)
relates to adolescents. Curr Psychiatry Rep 21(9):93. https://​doi.​ Translational research into gut microbiota: new horizons in obe-
org/​10.​1007/​s11920-​019-​1075-3 sity treatment. Arq Bras Endocrinol Metabol 53(2):139–144.
Sircana A, Framarin L, Leone N, Berrutti M, Castellino F, Parente R, https://​doi.​org/​10.​1590/​s0004-​27302​00900​02000​04
De Michieli F, Paschetta E, Musso G (2018) Altered gut micro- Vasques-Monteiro IML, Silva-Veiga FM, Miranda CS, de Andrade
biota in type 2 diabetes: just a coincidence? Curr Diab Rep Goncalves ECB, Daleprane JB, Souza-Mello V (2021) A rise in
18(10):98. https://​doi.​org/​10.​1007/​s11892-​018-​1057-6 Proteobacteria is an indicator of gut-liver axis-mediated nonalco-
Stanhope KL (2016) Sugar consumption, metabolic disease and obe- holic fatty liver disease in high-fructose-fed adult mice. Nutr Res
sity: the state of the controversy. Crit Rev Clin Lab Sci 53(1):52– 91:26–35. https://​doi.​org/​10.​1016/j.​nutres.​2021.​04.​008
67. https://​doi.​org/​10.​3109/​10408​363.​2015.​10849​90 Veech RL (2004) The therapeutic implications of ketone bodies: the
Stocks T, Taylor MA, Angquist L, Macdonald IA, Arner P, Holst C, effects of ketone bodies in pathological conditions: ketosis,
Oppert JM, Martinez JA, Rossner S, Polak J, Langin D, Saris ketogenic diet, redox states, insulin resistance, and mitochon-
WH, Astrup A, Sorensen TI (2013) Change in proportional pro- drial metabolism. Prostaglandins Leukot Essent Fatty Acids
tein intake in a 10-week energy-restricted low- or high-fat diet, 70(3):309–319. https://​doi.​org/​10.​1016/j.​plefa.​2003.​09.​007
in relation to changes in body size and metabolic factors. Obes Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, Ze X, Brown
Facts 6(3):217–227. https://​doi.​org/​10.​1159/​00035​1726 D, Stares MD, Scott P, Bergerat A, Louis P, McIntosh F, John-
Sun L, Xie C, Wang G, Wu Y, Wu Q, Wang X, Liu J, Deng Y, Xia J, stone AM, Lobley GE, Parkhill J, Flint HJ (2011) Dominant and
Chen B, Zhang S, Yun C, Lian G, Zhang X, Zhang H, Bisson diet-responsive groups of bacteria within the human colonic
WH, Shi J, Gao X, Ge P, Liu C, Krausz KW, Nichols RG, Cai J, microbiota. ISME J 5(2):220–230. https://d​ oi.o​ rg/1​ 0.1​ 038/i​ smej.​
Rimal B, Patterson AD, Wang X, Gonzalez FJ, Jiang C (2018) 2010.​118
Gut microbiota and intestinal FXR mediate the clinical benefits Wan Y, Wang F, Yuan J, Li J, Jiang D, Zhang J, Li H, Wang R, Tang
of metformin. Nat Med 24(12):1919–1929. https://​doi.​org/​10.​ J, Huang T, Zheng J, Sinclair AJ, Mann J, Li D (2019) Effects
1038/​s41591-​018-​0222-4 of dietary fat on gut microbiota and faecal metabolites, and their
Swidsinski A, Dorffel Y, Loening-Baucke V, Gille C, Goktas O, relationship with cardiometabolic risk factors: a 6-month ran-
Reisshauer A, Neuhaus J, Weylandt KH, Guschin A, Bock M domised controlled-feeding trial. Gut 68(8):1417–1429. https://​
(2017) Reduced mass and diversity of the colonic microbiome doi.​org/​10.​1136/​gutjnl-​2018-​317609
in patients with multiple sclerosis and their improvement with Wang DD, Nguyen LH, Li Y, Yan Y, Ma W, Rinott E, Ivey KL, Shai
I, Willett WC, Hu FB, Rimm EB, Stampfer MJ, Chan AT,

13
Diets, Gut Microbiota and Metabolites

Huttenhower C (2021) The gut microbiome modulates the pro- Zafeiropoulou K, Nichols B, Mackinder M, Biskou O, Rizou E, Kara-
tective association between a Mediterranean diet and cardiometa- nikolou A, Clark C, Buchanan E, Cardigan T, Duncan H, Wands
bolic disease risk. Nat Med 27(2):333–343. https://​doi.​org/​10.​ D, Russell J, Hansen R, Russell RK, McGrogan P, Edwards CA,
1038/​s41591-​020-​01223-3 Ijaz UZ, Gerasimidis K (2020) Alterations in intestinal micro-
Woodie L, Blythe S (2018) The differential effects of high-fat and biota of children with celiac disease at the time of diagnosis and
high-fructose diets on physiology and behavior in male rats. on a gluten-free diet. Gastroenterology 159(6):2039–2051 e2020.
Nutr Neurosci 21(5):328–336. https://​doi.​org/​10.​1080/​10284​ https://​doi.​org/​10.​1053/j.​gastro.​2020.​08.​007
15X.​2017.​12878​34 Zhang C, Bjorkman A, Cai K, Liu G, Wang C, Li Y, Xia H, Sun L,
Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Kristiansen K, Wang J, Han J, Hammarstrom L, Pan-Ham-
Bewtra M, Knights D, Walters WA, Knight R, Sinha R, Gilroy marstrom Q (2018a) Impact of a 3-months vegetarian diet on
E, Gupta K, Baldassano R, Nessel L, Li H, Bushman FD, Lewis the gut microbiota and immune repertoire. Front Immunol 9:908.
JD (2011) Linking long-term dietary patterns with gut micro- https://​doi.​org/​10.​3389/​fimmu.​2018.​00908
bial enterotypes. Science 334(6052):105–108. https://d​ oi.o​ rg/1​ 0.​ Zhang Y, Zhou S, Zhou Y, Yu L, Zhang L, Wang Y (2018b) Altered
1126/​scien​ce.​12083​44 gut microbiome composition in children with refractory epilepsy
Wu GD, Compher C, Chen EZ, Smith SA, Shah RD, Bittinger K, after ketogenic diet. Epilepsy Res 145:163–168. https://​doi.​org/​
Chehoud C, Albenberg LG, Nessel L, Gilroy E, Star J, Weljie 10.​1016/j.​eplep​syres.​2018.​06.​015
AM, Flint HJ, Metz DC, Bennett MJ, Li H, Bushman FD, Zhang X, Shen D, Fang Z, Jie Z, Qiu X, Zhang C, Chen Y, Ji L (2013)
Lewis JD (2016) Comparative metabolomics in vegans and Human gut microbiota changes reveal the progression of glu-
omnivores reveal constraints on diet-dependent gut microbiota cose intolerance. PLoS One 8(8):e71108. https://d​ oi.o​ rg/1​ 0.1​ 371/​
metabolite production. Gut 65(1):63–72. https://d​ oi.o​ rg/1​ 0.1​ 136/​ journ​al.​pone.​00711​08
gutjnl-​2014-​308209 Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, Li L, Fu
Wu J, Wang K, Wang X, Pang Y, Jiang C (2021) The role of X, Wu Y, Mehrabian M, Sartor RB, McIntyre TM, Silverstein
the gut microbiome and its metabolites in metabolic dis- RL, Tang WHW, DiDonato JA, Brown JM, Lusis AJ, Hazen SL
eases. Protein Cell 12(5):360–373. https://​doi.​org/​10.​1007/​ (2016) Gut microbial metabolite TMAO enhances platelet hyper-
s13238-​020-​00814-7 reactivity and thrombosis risk. Cell 165(1):111–124. https://​doi.​
Xie G, Zhou Q, Qiu CZ, Dai WK, Wang HP, Li YH, Liao JX, Lu XG, org/​10.​1016/j.​cell.​2016.​02.​011
Lin SF, Ye JH, Ma ZY, Wang WJ (2017) Ketogenic diet poses a Zhu H, Bi D, Zhang Y, Kong C, Du J, Wu X, Wei Q, Qin H (2022)
significant effect on imbalanced gut microbiota in infants with Ketogenic diet for human diseases: the underlying mechanisms
refractory epilepsy. World J Gastroenterol 23(33):6164–6171. and potential for clinical implementations. Signal Transduct Tar-
https://​doi.​org/​10.​3748/​wjg.​v23.​i33.​6164 get Ther 7(1):11. https://​doi.​org/​10.​1038/​s41392-​021-​00831-w
Yassour M, Lim MY, Yun HS, Tickle TL, Sung J, Song YM, Lee Zinocker MK, Lindseth IA (2018) The western diet-microbiome-host
K, Franzosa EA, Morgan XC, Gevers D, Lander ES, Xavier interaction and its role in metabolic disease. Nutrients 10(3):365.
RJ, Birren BW, Ko G, Huttenhower C (2016) Sub-clinical https://​doi.​org/​10.​3390/​nu100​30365
detection of gut microbial biomarkers of obesity and type
2 diabetes. Genome Med 8(1):17. https://​d oi.​o rg/​1 0.​1 186/​ Springer Nature or its licensor (e.g. a society or other partner) holds
s13073-​016-​0271-6 exclusive rights to this article under a publishing agreement with the
Yi C, Li D, Guo X, Wang J, Liu C, Lu G, Sun Y, Huang H, Hong author(s) or other rightsholder(s); author self-archiving of the accepted
S, Li J (2022) The storage conditions of high-fat diet are the manuscript version of this article is solely governed by the terms of
key factors for diet-induced obesity and liver damage. Nutrients such publishing agreement and applicable law.
14(11):2222. https://​doi.​org/​10.​3390/​nu141​12222

13

View publication stats

You might also like