You are on page 1of 10

Best Practice & Research Clinical Gastroenterology 31 (2017) 579e588

Contents lists available at ScienceDirect

Best Practice & Research Clinical Gastroenterology


journal homepage: https://ees.elsevier.com/ybega/default.asp

11

The interaction between smoking, alcohol and the gut microbiome


Gabriele Capurso*, Edith Lahner
Digestive and Liver Disease Unit, Sant’Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Italy

a r t i c l e i n f o a b s t r a c t

Article history: The gastrointestinal microbiome is a complex echosystem that establishes a symbiotic, mutually bene-
Received 20 September 2017 ficial relation with the host, being rather stable in health, but affected by age, drugs, diet, alcohol, and
Received in revised form smoking. Alcohol and smoking contribute to changes in the stomach and affect H pylori-related disorders
17 October 2017
including the risk of gastric cancer. In the small intestine and in the colon alcohol causes depletion of
Accepted 20 October 2017
bacteria with anti-inflammatory activity, eventually resulting in intestinal damage with “leaky gut”.
These changes contribute to hepatic damage in both alcoholic and non-alcoholic liver disease and have
Keywords:
been associated with other disorders. Lactobacillus GG and A. muciniphila exert a protective effect in this
Gastric microbiota
Gastric atrophy
setting. Smoking leads to modifications of the gut microbiome linked with a protective effect toward
Gastric cancer ulcerative colitis and deleterious for Crohn's disease. The exact cause-effect relation between alcohol and
Helicobacter pylori smoking and changes of the gastrointestinal microbiome needs further exploration with high throughput
Alcohol methodologies, and controlled studies are necessary to define the role of microbiome modulation on the
Smoking immune response and systemic activation of pro-inflammatory pathways.
Intestinal microbiota © 2017 Elsevier Ltd. All rights reserved.
Dysbiosis
Liver
Inflammatory bowel disease

1. Introduction diet, alcohol, and smoking [4].


The most common of the bacterial groups are the gram-negative
As defined by Quigley EMM, the microbiota refers to the Bacteroidetes and the gram-positive Firmicutes, the largest phylum,
assemblage of microorganisms (including also others than bacteria) that includes several Lactobacillus strains used as probiotics.
present in a defined environment, while the microbiome comprises In the past decades, it has become clear that the microbiome is
the full complement of microorganisms, such as bacteria, viruses, not a passive victim in many pathological processes, but its modi-
fungi, and protozoa, together with their genes and genomes in a fication often play a contributive or causative role in pathophysio-
given locus [1]. The gastrointestinal microbiome is a complex logical processes [5].
echosystem of 10e100 trillion microorganisms, of which the ma- The present review will present the available evidence
jority are bacteria in the gut, but also including virus and fungal regarding the interaction between smoking, alcohol and the gut
species, that develops immediately after birth depending on mul- microbiome. The effects of alcohol and smoking on the microbiome
tiple factors among whom diet and the host genotype seem the of either the upper or the lower gastrointestinal tract will be pre-
most important ones [2]. sented separately, highlighting key points and area for further
The interaction between the microbiome and the host is a research.
symbiotic, mutually beneficial one.
Data from the Human Microbiome Project Consortium [3] hel-
ped characterizing the microbiome composition and function in 1.1. What do we know about the gastric microbiome?
terms of genomics, trascriptomics and metabolomics. In adults the
microbiome tends to become rather stable in health, but is affected From a physiological point of view, the stomach may be
by a number of factors such as age, drugs (especially antibiotics), considered a defensive barrier against orally ingested microor-
ganisms, as it functions to inactivate ingested microorganisms to
prevent them from reaching the intestine [6]. The observation that
* Corresponding author. Digestive and Liver Disease Unit, S. Andrea Hospital,
reduced gastric acid secretion increases the susceptibility toward
Sapienza University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy. infection [6,7] supports this concept. The gastric environment is
E-mail address: gabriele.capurso@gmail.com (G. Capurso). hostile and difficult to colonize, mainly due to its acidic pH and

https://doi.org/10.1016/j.bpg.2017.10.006
1521-6918/© 2017 Elsevier Ltd. All rights reserved.
580 G. Capurso, E. Lahner / Best Practice & Research Clinical Gastroenterology 31 (2017) 579e588

proteolytic enzymes, and the stomach has therefore been consid- molecular approaches, but with the limit that they are unable to
ered to be a nearly sterile organ. The stomach displays a much differentiate living and dead microorganisms [20]. The acquisition
lower microbial load (102-104 colony-forming units [CFU]/ml of of functional microbiota data requires the sequencing of the com-
bacteria) than the intestine (104e1012 CFU/ml) [8,9]. Some bacteria plete microbial community DNA by shotgun metagenomics
adapt to these unfriendly conditions, i.e. obligate or facultative methods and subsequent matching of the obtained sequences to
acidophiles able to live at pH < 4.0 [10]. the known functional genes available on datasets [9]. Due to the
In 1983, the discovery of Helicobacter pylori (Hp), a Gram- low bacterial load in the stomach, the application of these methods
negative bacterium responsible of gastric inflammation eventu- may result technically difficult. The techniques used to analyze the
ally leading to peptic ulcer, MALToma or adenocarcinoma, drasti- human microbiota are thoroughly reviewed amongst others by
cally changed this view. Meanwhile, Hp is certainly the most Schulz C et al. [21].
thoroughly investigated component of the gastric microbiota [11].
Compared to other gastrointestinal districts, knowledge on 1.2. Who are the bugs in the stomach? what is the composition of
gastric microbiota and its role in human health and disease is the gastric microbiota?
limited. The development of new high-throughput sequencing
methods and technologies as microbiomics and metagenomics The major component of the gastric microbiota in more than
made feasible the investigation of non-cultured microorganisms half of the world population is Hp, a spiral-shaped member of the
[12]. Parallel to this, the wide diffusion of gastrointestinal endos- Gram-negative Epsilonproteobacteria [22]. Hp has been classified as
copy made easier the accessibility of the stomach to obtain bioptic a definitive (class I) carcinogen by the World Health Organization
mucosal or gastric juice samples for molecular analysis. (WHO). The mucus layer of the stomach is the natural habitat of Hp;
How to investigate the gastric microbiota: from culture to the majority of Hp organisms swim within the mucus layer pro-
culture-independent molecular methods. pelled by a bundle of rotating flagella [23]. Hp infection profoundly
The presence of gastric microorganisms other than Hp was at affects gastric physiologic functions, which in turn may affect the
first investigated by using conventional methods, such as histology properties of gastric mucosa to be colonized by other bacteria [24].
of gastric mucosal biopsies and the culture of gastric juice or gastric Whether Hp infection in humans is able to influence the compo-
mucosa [13]. In contrast to animal studies usually using the entire, sition of the gastric microbiota in not fully understood.
aseptically removed stomach, the sample collection in humans is Hp infection leads to changes of the intragastric microenviron-
more complex. Gastric mucosal samples are generally obtained by ment possibly creating novel niches for bacterial species. Rodents'
biopsies during gastroscopy, but the contamination of the biopsy gastric microbiota may be influenced by Hp infection [25], while
channel with oral or throat microorganisms cannot be excluded, the gastric microbiota in mice seems not to be affected [26]. Simi-
potentially leading to distortion of microbial composition results larly, in humans no significant differences between phylotype dis-
given the low gastric bacterial load [11]. Moreover, data are lacking tribution of gastric microbiota or gastric pH levels by using 16S
about microbial differences between different mucosal layers in rDNA sequencing analysis were observed between Hp-positive and
humans; microbiota analyses from gastric biopsies represent a enegative subjects [27]. A 2008 European study [28] reported a
bacteria mix in the luminal fluid, upper mucus layer (non-Hp), and higher gastric microbiota diversity in Hpenegative compared to
juxtaepithelial mucus (mainly Hp) [11]. Instead, gastric juice may Hp-positive subjects. Maldonado-Contreras et al. [29] observed a
be sampled in a sterile manner by using a bull-nosed capsule, positive correlation between Hp and Spirochaetae and a negative
double or triple nastrogastric tube systems, or flexibile silicone correlation between Hp and Bacteroidetes, Chloroflexi, Cianobacteria,
sampling probes [14,15]. Fusobacteria, Plactomycetes, Beta- and Gamaproteobacteria, and
By culture, the most represented bacteria in the stomach of Verrucomicrobia, further showing that 28% of the microbiota
healthy subjects were found to be Clostridium sp, Lactobacillus sp, composition variation was explained by Hp status, but 44% was
and Veillonella sp [14]. The main advantage of the culture-based explained by patients' origin. Geographical differences may play a
approach toward culture-independent methods is the selective role in the diversity of the human gastric microbiota and in in-
detection of viable bacteria. Unfortunately, the vast majority (~80%) teractions variations between Hp and other gastric microbiota
of microbial species are not cultivable [16]. components [13]. In a very recent study Hp eradication increased
The more recent culture-independent, molecular approaches bacterial diversity and restored the relative abundance of other
allowed to give new insights in the gastric microbiota composition bacteria to levels similar to Hp-negative subjects suggesting that Hp
and depend on DNA-based approaches, either relying on whole- colonization results in alterations of gastric microbiota, reversible
genome information or focusing on the 16S rRNA gene as stan- by antibiotic treatment [30]. Thus, to date, whether Hp acts as key
dard phylogenetic marker [11,17]. First, the bacterial diversity of the player or house keeper for other gastric microbes by modulating
gastric microbial ecosystem was evidenced by more traditional the gastric acidity or by other mechanisms stills remains to be
DNA fingerprinting techniques, such as denaturating gradient gel clarified [21].
electrophoresis (DGGE), temporal gradient gel electrophoresis In the recent years, culture-independent studies provided evi-
(TTGE) or terminal restriction fragment length polymorphism dence on the microbial diversity in the human stomach
(TRFLP) of polymerase chain reaction (PCR)-amplified 16S rDNA [8,27,28,31e33]. Some authors reported the gastric microbiota in
fragments [18]; more recently, methods like Sanger-sequencing 16S healthy, Hp-free gastric mucosa: Zilberstein et al. [14] isolated 18
rRNA libraries, microarrays targeting the 16S rRNA gene and high- different bacterial taxa by culture of gastric mucus aspirate; the
throughput sequencing methods (454, Illumina, and Ion Torrent) most frequent species were Lactobacillus sp., Veillonella sp., Clos-
have been developed [11,12]. These latter methods allow a tridium sp., and Corynebacterium sp. A study based on 454-
considerably higher sequencing coverage than Sanger sequencing pyrosequencing technology (short rRNA gene sequence reads)
of 16S clone libraries and a much better phylogenetic resolution [28] was performed on three Hp-negatives and three Hp-positives):
than fingerprinting methods, permitting the identification of un- 33 phylotypes were found in all three Hp-negative samples, and
expected or previously unknown bacteria, not detectable by most of the prominent phylotypes (e.g. Streptococcus, Actinomyces,
microarray approaches [11,19]. Prevotella and Gemella), were also abundant in the throat suggest-
A complete view of the overall microbial composition of a def- ing that they were swallowed microorganisms from upstream
inite ecosystem may thus be provided by these innovative microbiota; the majority of the 177 phylotypes found in the
G. Capurso, E. Lahner / Best Practice & Research Clinical Gastroenterology 31 (2017) 579e588 581

stomach but not in throat belonged to Proteobacteria; in contrast, to the pre-infection microbiota [37], suggesting that gastric
the three Hp-positive samples were totally dominated by this microbiota variations may have an effect of Hp-associated disease.
bacterium (93e97% of reads), corroborating the finding that the An impaired gastric microbiota (dysbiosis) has been identified as a
stomach displays a diverse microbiota when Hp is absent [27]. possible trigger of different gastric conditions as atrophic gastritis
Li et al. [31] investigated the gastric microbiota composition of (AG), intestinal metaplasia, and even gastric cancer (GC)
Hp negative NSAIDs-off individuals by 16S rRNA gene-libraries from [33,38e40].
antral and corporal biopsies and identified as dominant genera The gastric microbiota has long been considered an essential
Streptococcus, Prevotella, Neisseria, Haemophilus, and Porphyr- factor contributing to GC development [41]: The impaired gastric
omonas, accounting together for 70.5% of all sequences, Strepto- acid secretion in gastric atrophy results in gastric bacterial over-
coccus and Prevotella alone accounted for 41% of all sequences. With growth, increased production of N-nitroso-compounds leading to
regard to gastric topography, the genus Prevotella was more DNA damage and epithelial cells methylation, potentially promot-
abundant in the antrum compared to the corpus. ing gastric carcinogenesis [42]. This is supported by data showing
Similarly to previous studies with different methodological ap- increased gastric bacterial counts and enhanced nitrites production
proaches [27,28] Stearns and colleagues [32] by using Illumina in the stomach during antisecretory treatment [43].
sequencing reported Firmicutes, Actinobacteria, Bacteroidetes, Pro-
teobacteria, and Fusobacteria as the most abundant phyla of the
1.3.1. Gastric microbiota modulation and gastric cancer
human stomach. Albeit the microbial research in the human
Yu G et al. [38] showed that lower microbial richness was
stomach has been defined to be still in the early stages, current data
significantly associated with lower pepsinogen I/II ratio, a sero-
show that the stomach microbiota displays a microbial diversity
logical marker of gastric atrophy predisposing to GC, supporting the
beyond Hp and seems to be distinguishable from oral, throat, nose,
potential role of the gastric microbiota in GC risk. In GC patients, the
and distal gut microbiota [8,13], supporting the idea that in healthy
gastric microbiota was dominated by different species of the genera
subjects harbor a “core gastric microbiota”. Table 1 gives an over-
Streptococcus, Lactobacillus, Veillonella, Prevotella, and Haemophilus
view on the reported composition of the gastric microbiota other
[44]. Veillonella and Haemophilus parainfluenzae had a higher ca-
than Hp.
pacity in nitrate than nitrite reduction, thus increasing nitrite
accumulation in the gastric juice [45].
1.3. Potential modulators of the gastric microbiota: alcohol and A recent Korean study analyzed the gastric microbiota of Hp-
smoking positive and -negative GC patients compared to controls by bar-
coded 454 pyrosequencing of the 16S rRNA gene and showed a
Microbiota diversity or composition may be shaped by the host higher composition of Streptococcus pseudopneumoniae,
physiology as well as by environmental factors. Many factors have S. parasanguinis, and S. oralis in the Hp-negative cancer group [46],
been postulated to potentially exert an influence on the gut suggesting a possible role of non-Hp bacteria in the gastric carci-
microbiota by causing significant shifts of its composition, such as nogenesis. However, a very recent Swedish study investigated the
diet, antibiotics, other drugs, Hp-related inflammation, intra-gastric composition of the transcriptionally active microbial community
pH, as well as smoking and alcohol [33]. For example, the mucus and the Hp gene expression by using metatranscriptomic RNA
layer in the stomach is a protective barrier preventing microbial sequencing of stomach biopsies from individuals with different Hp
epithelial colonization and diffusion of gastric acid to the epithelial infection status and pre-malignant tissue changes observing that
surface. The capability to get across the mucus layer highly depends Hp completely dominates the microbiota not only in infected in-
on bacterial factors (motility, shape and mucus-degrading-ability) dividuals, but also in most individuals classified as Hp-uninfected
[34], but also on host factors: an increased pH results in lower using conventional methods, and that Hp abundance is positively
viscosity making the mucus layer easier to penetrate for microor- correlated with presence of Campylobacter, Deinococcus, and Sul-
ganisms [35], thus potentially modulating gastric microbiota. Hp furospirillum [47]. This study clearly strengthens how functional
infection is linked to chronic inflammation, oxidative stress, and analysis of the gastric microbiota may provide new insights into
DNA damage [36], potentially influencing the gastric microbiota how bacteria respond in vivo to variations in the stomach micro-
composition. An elegant study in wild-type C57BL/6 mice reported environment and how much we have still to learn about its in-
that the entity of Hp-associated inflammation was directly related teractions and modulators.

Table 1
Composition of the gastric microbiota beyond Helicobacter pylori.

Most frequent phyla/genera Detection technique Subjects Reference

Corynebacterium Culture of gastric mucus aspirate Healty volunteers [14]


Clostridia
Lactobacillus
Veillonella
Actinomyces 454-pyrosequencing technology (short rRNA gene sequence reads) from gastric biopsies Hp-negative subjects [27]
Gemella
Prevotella
Proteobacteriaa
Streptococcus
Haemophilus 16S rRNA gene-libraries from antral and corporal biopsies Hp-negative NSAIDs-off subjects [31]
Neisseria
Porphyromonas Prevotella
Streptococcus
Actinobacteria Illumina 16S rRNA sequencing of gastric biopsies Healthy volunteers [32]
Bacteroidetes, Firmicutes
Fusobacteria Proteobacteria
a
Proteobacteria were detected the stomach only, the other also in the throat; Hp ¼ Helicobacter pylori.
582 G. Capurso, E. Lahner / Best Practice & Research Clinical Gastroenterology 31 (2017) 579e588

1.3.2. Gastric microbiota modulation and alcohol genotoxic or tumor promoting effects. In AG, a predisposing con-
Alcohol may be considered a further modulator of gastric dition for GC characterized by impaired gastric acid secretion and
microenvironment. Attention has been paid to the harmful conse- alterations of the gastric microbiota [40,67], ethanol metabolism by
quences of alcohol in the stomach. Both acute and chronic alcohol the gastric microbiota may lead to high intragastric acetaldehyde
consumption produce harmful effects in the stomach by multiple, production [68]: the acetaldehyde production from gastric juice
complex mechanisms related to direct contact of ethanol or its samples from AG patients incubated with 1% ethanol has been re-
metabolite acetaldehyde as well as by non-alcoholic components of ported to be 7.6-times higher compared to control samples [69].
alcoholic beverages: (i) gastric motility impairment; (ii) alteration Thus, alcohol consumption may exert effects on the gastric
of gastric acid output, and (iii) direct mucosal damaging [48,49]. microenvironment and this may, in turn, lead to potential alter-
Ethanol solutions exert an inhibitory, not dose-dependent effect on ations of the gastric microbiota composition, including Hp. The
gastric emptying, and alcoholic beverages produced by fermenta- magnitude and the modalities with which alcohol intake may
tion (beer and wine) are emptied even more slowly than their modulate Hp infection and the gastric microbiota remain a subject
corresponding ethanol concentrations, presumably due to addi- open for investigation, as specific studies on the gastric microbiota
tional inhibitory factors in the non-alcoholic ingredients [50]. Fer- beyond Hp are lacking.
mented alcoholic drinks are potent stimulators of gastric acid
output, and succinic acid and maleic acid have been reported as the 1.4. Gastric microbiota modulation and smoking
responsible substances [51]. In contrast, highly concentrated alco-
holic beverages produced by distillation (whiskey and cognac) do N-nitroso compounds, consisting of N-nitroamines and N-
not stimulate gastric acid secretion; interestingly, alcoholic bever- nitroamides, are potent carcinogens to which humans are exposed
ages produced by fermentation followed by distillation (i.e. aperi- from diet and smoking. Increased exposure to these exogenous N-
tifs and hard liquors) do not stimulate acid secretion as well [52]. nitroso compounds has been linked to an increased incidence of GC,
Alcohol has been associated with a breakdown of gastric barrier albeit an important role has been ascribed to the endogenous N-
thus contributing to the toxic damage of alcohol to gastric mucosa nitroso compounds [70]. A 2008 systematic review provided
and alterations in mucosal defense [53]. Ethanol may directly and summary RR estimates of 1.62 in males and 1.20 in females for GC in
dose-dependently impair the gastric mucosal barrier; the acidifi- current smokers compared to never smokers, classifying smoking
cation of the mucosal cells and ethanol itself induce release of in- as one the most important behavioral risk factor for GC [71]. More
flammatory and vasoactive substances, leading to ischemia and recently, data from the Stomach Cancer Pooling (StoP) Project
mucosal damage [52]. Therefore, analogously to tobacco smoking, confirmed the detrimental effect of cigarette smoking on the risk of
acute or chronic use/abuse of alcohol may be postulated to signif- GC reporting a RR 1.25 for current cigarette smokers compared to
icantly alter the gastric microenvironment in turn affecting the never smokers [72].
gastric microbiota composition. Another proposed mechanism by which smoking may lead to
With regard to Hp, an alcohol intake lower than 75 g weekly has negative effects on health, as cancer and periodontitis, may be
been reported to protect against active infection [54]. Another through microbiome alterations [73]. Vogtman E et al. showed by a
study reported a lower risk of Hp infection in subjects consuming 16S rRNA microarray that current smokers had an increased
between 3 and 6 units of wine or beer weekly compared to those within-participant-diversity for a 10 species increase, the OR for
not drinking (OR 0.89, 95% 0.90e0.99; OR 0.83, 95%CI 0.75 to 0.91) current smoking was 1.29; Dialister invisus and Megasphaera
[55]. A recent dose-response meta-analysis showed an inverse micronuciformis were more commonly detected in current smokers
relationship between alcohol intake and Hp infection (OR 0.78,95% compared to never smokers [74].
CI 0.69 to 0.89), regardless of gender, age, geographic areas, Among host-related factors, smoking has been studied as a
detection methods or beverage [56], thus confirming the results possible risk factor for Hp eradication failure: a meta-analysis re-
from former studies. ported an OR of 1.95 for eradication failure in smokers compared to
Alcohol consumption has been reported to be positively asso- non-smokers [75]. A very recent Israeli study on >48,000 subjects
ciated with successful eradication treatment: treatment failure was confirmed that eradication failure (first-line treatment for Hp
significantly more frequent in non-consumers (29.9%) than in infection) was positively associated with current smoking (OR 1.15),
consumers (12.2%) (OR 3.24) [57]. To date it is not understood together with female gender (OR 1.20) and low socioeconomic
whether alcohol-related alterations of the gastric microenviron- status (OR 1.24) [76]. Whether the significant association between
ment and possible subsequent changes of the gastric microbiota the likelihood of unsuccessful first-line treatment for Hp infection
may be linked to this observation. and smoking status may be linked to an altered gastric microbiome
In contrast to the observed protective effects of alcohol on Hp remains to be investigated.
infection, alcohol intake was reported being a risk factor for GC Table 2summarizes the supposed mechanisms and the
[58e60]: Low alcohol intake was negatively associated with GC risk described modulating effects of alcohol and smoking on the gastric
(HR 0.74) [61]. This association was stronger only in heavy alcohol microbiota.
consumers (4 drinks daily) (RR 1.20) [62]. A more recent huge
Japanese study reported a significant association with GC risk and 1.5. Effects of alcohol on the microbiome of the lower
different levels of alcohol intake (HR 1.39) [63]. Similarly to gastrointestinal tract
smoking, alcohol intake has been reported as being associated with
Hp eradication failure [64]. The association between alcohol consumption and changes in
Ethanol is metabolized by alcohol dehydrogenases (ADH), intestinal microbiome has been investigated extensively in the past
catalase or cytochrome P450 2E1 to acetaldehyde which is then decade and there is sufficient evidence suggesting that both acute
further oxidized to acetate by aldehyde dehydrogenase (ALDH) and chronic exposure to alcohol determine specific modifications of
[65]. Acetaldehyde induces DNA damage in the gastrointestinal the microbiome composition, bacterial overgrowth, and disruption
mucosa [65], classified as class 1 carcinogen to humans by the WHO of the mucosal barrier function [77] (see Table 3). Many of these
[66]. Several mechanisms have been suggested to delineate alcohol studies were conducted in animal models with liver disease, as it
consumption and cancer development. Ethanol itself is not carci- has become clear that alcohol-induced intestinal dysbiosis alters
nogenic, but data suggest that acetaldehyde and ROS have the homeostasis between the gut and the liver and plays a central
G. Capurso, E. Lahner / Best Practice & Research Clinical Gastroenterology 31 (2017) 579e588 583

Table 2
Alcohol and smoking as potential modulators of the gastric microbiota.

Gastric microbiota Proposed mechanisms Alteration of the gastric microbiota


modulators

Alcohol Impairment of gastric motility Inverse relationship between alcohol intake and Hp infection
Alteration of gastric acid output Alcohol consumption is positively associated with successful Hp
Breakdown of gastric mucosal barrier eradication treatment
Production of acetaldehyde Alcohol intake is associated with higher gastric cancer risk; in atrophic
gastritis, higher acetaldehyde levels and gastric microbiota alterations
Other gastric microbes: data are lacking
Smoking Alteration of gastric microenvironment: increased exposure to N- Smoking is associated with a higher risk of Hp eradication failure
nitroso compounds which are potent carcinogens inducing DNA Other gastric microbes: data are scanty
damage and epithelial cells methylation, possibly enhanced by impaired Smoking is associated with higher gastric microbial diversity Smoking is
gastric acid secretion leading in turn to increased nitroso-compounds associated with a higher risk of gastric cancer
production due to gastric bacterial overgrowth In GC patients, gastric microbiota dominated by Streptococcus,
Lactobacillus, Veillonella, Prevotella

Hp ¼ Helicobacter.

Table 3
Effects of alcohol on intestinal microbiome composition and associated disorders.

Animal Models Human Studies Intestinal damage Associated Disorder(s)

Phyla (Genera) Firmicutes (Lactococci, Bacteroidetes [88,91,92], Bacterial overgrowth, leaky gut, Intestinal inflammation, ASH,
Depleted by Alcohol Pediococci, Lactobacilli) [78,80], Proteobacteria changes of mucus composition, Klebsiella pneumoniae
A muciniphila [84] (Enterobacteriaceae) [91,92] bacterial translocation. pulmonary infection.
Firmicutes (Lachnospiraceae,
Ruminococcaceae) [89,91],
Verrucomicrobia (A muciniphila)
Phyla/genera increased Verrucomicrobia, Bacteroidetes Proteobacteria
by alcohol Actinobaceria [78,80,89,91] (Enterobacteriaceae)
Bacteroides, Clostridium
[88,89,91]
Genera associated with LGG [79,80], A muciniphila [85] LGG [92] e
recovery of damage

LGG ¼ Lactobacillus rhamnosus GG, ASH ¼ alcoholic steatohepatitis.

role in the development of alcoholic liver disease. In this view, the with faeces of control animals fed with alcohol, intestinal and he-
microbiome plays the role of an important mediator of a number of patic inflammation increases, suggesting that the pro-
negative effects of alcohol. However, it has also been demonstrated inflammatory effect of alcohol is mediated by the intestinal
that bacterial phyla are able to produce alcohol themselves, such as microbiome [81].
Proteobacteria, are increased in subjects with metabolic alterations Chronic exposure to alcohol also modifies the composition of
and non-alcoholic steatohepatitis (NASH), possibly supporting the intestinal mucus. Interestingly, MUC2 knock out animals are less
hypothesis of a direct role of the microbiome in determining NASH. prone to develop bacterial overgrowth, bacterial translocation and
At any rate, manipulation of the microbiome is an intriguing ther- intestinal inflammation in response to exposure to alcohol, sug-
apeutic possibility for alcohol-related disorders. gesting that the interaction between alcohol, the mucosal layer and
microbiome are complex [82]. An interesting bacterial species in
this setting is Akkermansia muciniphila, a member of the phyla
1.5.1. Studies in animal models
Verrucomicrobia, with growing data supporting potential anti-
There are a number of studies in animal models in which
inflammatory properties [83]. A muciniphila exerts an anti-
exposition to alcohol causes ‘leaky gut’ with decreased levels of
inflammatory action, and its depletion is an early event in
bacteria with “anti-inflammatory activity” such as Firmicutes (Lac-
alcohol-fed animals [84]. In mice treated with alcohol, when A
tococcus, Pediococcus, Lactobacillus and the Leuconostoc genera) and
muciniphila is administered in a preventive setting, all markers of
increased levels of Verrucomicrobia and Bacteroidetes [78] in the
intestinal (gut barrier function and mucus thickness) and liver
cecum. These changes seem to occur after 3 weeks of exposure to
damage are reduced [85].
alcohol. In another study, rats fed with alcohol for 10 weeks showed
Taken together, these studies strongly suggest that alcohol
modifications of the microbiome in the colonic but not in the ileal
causes significant changes in the composition of the colonic
mucosa, and pre-treatment with either a probiotic (Lactobacillus
microbiome in animal models, which in turn causes pro-
GG, LGG) or a prebiotic (oats) prevented these changes [79].
inflammatory changes, leaky gut and bacterial translocation asso-
Similar results have been confirmed by others, with data sug-
ciated with liver disease [86].
gesting that alcohol causes an increase of Proteobacteria and Acti-
The liver is not the only organ distant from the gut that has been
nobaceria phyla, and a decrease of Firmicutes with these changes
associated with deleterious effects of intestinal dysbiosis due to
being restored by treatment with LGG [80].
alcohol. As alcohol is a risk factor for pulmonary infections,
Germ-free animals are an interesting model to investigate the
Samuelson and colleagues investigated the effect of alcohol-
role of alcohol on intestinal microbiota. When germ-free rats are
induced dysbiosis on the susceptibility to Klebsiella pneumoniae
fed with alcohol for seven days the induction of pro-inflammatory
infection in an animal model [87]. When alcohol-naïve rats were
cytokines and the liver inflammation are less pronounced
recolonized with microbiota from alcohol-fed animals, there was
compared to those seen in control animals with normal micro-
an increased risk of K. pneumoniae infection and related damage,
biome. Furthermore, when germ-free animals are transplanted
584 G. Capurso, E. Lahner / Best Practice & Research Clinical Gastroenterology 31 (2017) 579e588

associated with impaired immunological defense. These findings received the microbiome of patients with severe alcoholic hepatitis
point out toward a wider role of alcohol-related intestinal dysbiosis developed a worse intestinal, damage with increased intestinal
with potential effects on the immunological response and conse- permeability and bacterial translocation, and more prominent in-
quences that are not limited to the digestive system. flammatory changes in the liver. This result strongly supports the
case of an active role of the microbiome, independently from
alcohol and the host.
1.5.2. Studies in human
Another intriguing aspect regards the existence of a gut-brain
There is also a growing body of evidence form studies in humans
axis that could be modulated by microbiome imbalance due to
supporting the association between alcohol consumption, dysbio-
alcohol. In an interesting study, Leclercq et al. [96] investigated the
sis and eventually intestinal and hepatic inflammation. Chronic
composition of gut microbiome and intestinal permeability and
alcohol consumption leads to an increase in Proteobacteria and a
their association with psychological symptoms of alcohol depen-
decrease in Bacteroidetes and increased intestinal permeability [88]
dence in 75 subjects with alcohol dependence. They found that
resulting in traslocation of LPS and endotoxins in the bloodstream
subjects with increased intestinal permeability had higher scores of
which eventually contributes to hepatic damage [89].
depression and anxiety and alcohol craving after 3 weeks of
Indeed, the microbiome of cirrhotic patients differs from that of
abstinence, possibly suggesting that the microbiome might be a
controls, and a “cirrhosis dysbiosis ratio” (CDR) has been proposed
potential target to reduce the risk of alcohol use relapse in
[90].
alcoholics.
This index, defined by the ratio between Lachnospiraceae and
An interesting issue regards the cause-effect relation between
Ruminococcaceae which can exert a protective effect and Entero-
alcohol and microbiome. Indeed, while the above mentioned
bacteriaceae and Bacteroidaceae that are potentially pathogens is
studies suggest that alcohol assumption modifies the microbiome
typically lower in cirrhotics, especially in those with an alcoholic
and that their interaction determines a number of pathophysio-
etiology [89,91].
logical changes related with intestinal and liver damage, dysbiosis
The causal effect of these alteration is unclear. However,
itself can lead to ethanol production even in the absence of alcohol
intriguingly, administration of Lactobacillus rhamnosus GG to
assumption, thus possibly resulting in secondary liver damage.
cirrhotic patients in a randomized-controlled trial, determines an
It has been demonstrated that some bacteria, such as Escherichia
increase of Lachnospiraceae and a decrease of Enterobacteriaceae
coli can produce up to 0.8 g of ethanol per hour in anaerobic con-
with consequent reduction of pro-inflammatory cytokines [92].
ditions [97]. Interestingly, the presence of Enterobacteriaceae which
Although all these data clearly show that alcohol favours dys-
under certain circumstances can produce alcohol, is increased in
biosis, some alcoholic beverages might exert a positive effect on the
patients with NASH [98]. In another study ethanol was measured by
microbiome. This is the case of red wine. In a cross-over study
gas chromatography in 21 with NASH and in 10 controls [99]. While
performed in healthy subjects, gin caused an increase in the Bac-
NASH patients did not produce more ethanol compared to controls,
teroides and Clostridium frequencies while both red wine and de-
its levels were higher in obese women. One might therefore spec-
alcoholized red wine consumption led to an increase of the
ulate that a certain type of dysbiosis, typical of obesity and meta-
amount of a number of bacterial group including Enterococcus,
bolic syndrome, favours increased ethanol production in the gut,
Bacteroides and Bifidobacterium [93]. Red wine induced changes
contributing to liver injury in a vicious circle in which increased
were associated with a parallel reduction of systolic and diastolic
ethanol production would in turn cause “leaky gut” and bacterial
blood pressures, triglyceride, cholesterol, and C-reactive protein.
translocation eventually causing increased liver damage.
The results suggest that the effect is mediated by polyphenols
contained in red wine.
A decrease of faecal A. muciniphila when compared with healthy 1.6. Smoking and the microbiome in the lower GI tract
controls has also been reported in patients with alcoholic steato-
hepatitis (ASH) which seems associated the severity of the liver Cigarettes produce as much as 4000 individual chemical com-
damage [85]. A. muciniphila is a key health-promoting bacteria pounds such as hydrogen, hydrogen cyanide, methane, aniline,
related with active and healthy lifestyle, as regular physical activity aldehyde, phenols, nicotine and nar containing in turn simple
increases its abundance together with that of Bifidobacterium spp, phenols, cresols, naphthols, pyrene, benzopyrene and hydrocar-
R. hominis, and F. prausnitzii [94]. bons and heavy metals such as lead, cadmium, nickel, polonius,
Taken collectively these data demonstrate that specific modifi- strontium [100]. All these compounds have some kind of delete-
cations of the microbiome occur in association with alcohol intake rious effects on different human organs and some of them exert an
in humans, causing intestinal damage and increase in inflammatory effect on the intestinal health and on the microbiome (see Table 4).
changes, eventually linked with increased hepatic damage. With
this in mind, Llopis et al. designed a study in which germ-free and 1.6.1. Studies in animal models
conventional mice received microbiome transplants from alcoholic Animal models offer a good opportunity to analyze the affects of
patients with or without alcoholic hepatitis and investigated different compounds derived from cigarette smoking on the in-
possible different effects of feeding the animals with alcohol related testine [101]. However, there is a relative paucity of these kind of
with this “humanized” microbiome [95]. They found that mice who data in the literature. Thomas et al. investigated the effect on

Table 4
Effects of smoking on intestinal microbiome composition and associated disorder(s).

Animal Models Human Studies Intestinal damage Associated Disorder(s)

Phyla (Genera) Depleted by Bifidobacteria [111], Firmicutes, Actinobacteria [115] Anti-inflammatory effect in the Crohn's disease and Ulcerative
Smoking Firmicutes colon mucosa and Colitis.
(Lactococcus,Ruminococcus), proinflammatory in the ileum;
Enterobacteriaceae [112] changes in mucins expression.
Phyla/genera increased Clostridium [112], Clostridium [114] Bacteroidetes,
smoking Lachnospiraceae [113] Proteobacteria [115]
G. Capurso, E. Lahner / Best Practice & Research Clinical Gastroenterology 31 (2017) 579e588 585

colorectal mucosa of subcutaneous nicotine at different doses as Bacteroidetes and Proteobacteria. Of course, the observed results
compared to saline, in ferrets. They did not report significant might be due to other uninvestigated factors related with smoking
changes but for the nicotine and cotinine serum levels [102]. Van habit or with its cessation, such as modification of the diet and
Dijk et al. reported the potential anti-inflammatory effect of nico- increase in body weight. Interestingly, similar changes have been
tine, as upon its administration the synthesis of pro-inflammatory reported in obese men with increased levels of Firmicutes and
cytokines such as interleukin-1 beta and tumor necrosis factor reduced levels of Bacteroidetes [116].
alpha was completely abolished [103]. As stated above, the heterogeneous effects of smoking on the
The effects of smoking on the large and small bowel mucosa human microbiome at different sites might partially explain the
seem to diverge. It has been demonstrated that both nicotine and reduced risk of ulcerative colitis [117] and the increased risk of
carbone-monoxide do not cause macroscopic or microscopic Crohn's disease (CD) [118] associated with this habit.
damage and decrease inflammation in the colon, while nicotine Indeed, in Crohn's disease patients who smoke, the microbial
worsens jejunitis in experimental models, decreasing PGE2 gen- population and diversity is decreased with reduction in particular
eration and increasing NOS activity [104e106]. These data support of Collinsella, Enterorhabdus and Gordonibacter [119]. In another
the clinical observation of opposite effects of smoking on ulcerative study the decreased levels of Faecalibacterium and smoking are
colitis and Crohn's disease. However, there are data in opposition associated independently with CD recurrence [120].
with this view, as others found that cigarette smoke (CS) exposure
in a rat model of trinitrobenzene sulfonic acid (TNBS)-induced 2. Conclusion remarks
colitis worsened the inflammatory damage [107].
Zijlstra and colleagues investigated the effect of different con- In conclusion, microbiome research has enormously developed
centration of subcutaneous nicotine on the colonic mucosa of in the last years tempting to move its first steps from the laboratory
rabbits [108]. The thickness of adherent mucus on rectal mucosa into clinical practice. New nucleotide sequencing techniques and
was significantly reduced by low dose nicotine, but increased by new biocomputational tools techniques have allowed to better
high doses. Prostaglandins (PG) were reduced with an inverse dose characterize the microbiome of the gastrointestinal tract. Among
dependence, as lower doses caused a more significant reduction. many factors that contribute to the modulation of the gastroin-
This heterogeneity might be due to the different models, the testinal microbiome, the intake of alcohol and smoking seem
different substances (either nicotine or cigarette smoking) and relevant. The interactions between alcohol and smoking, the gastric
their dosage and way of administration employed in the above microbiome and the host remain enigmatic. This holds true also for
mentioned studies. the role of H. pylori which might act as a key player or house keeper
In this context, it is worth reminding that the intestinal micro- for other GI microbes by modulating gastric acidity. As stated by
biota is an important scavenger of potentially toxic compounds, Quigley EMM [1], for the real clinical applicability of gut micro-
with a number of enzymatic families (azoreductases, nitro- biome research at least three essential conditions need to be ful-
reductases, b-glucuronidases, sulfatases and b-lyases) affecting the filled: (i) we need to know what the normal microbiota is like, (ii)
metabolism of over 30 environmental pollutants. On the other we need to accurately and reliably define an altered/abnormal
hand, environmental contaminants alter the composition of the microbiota, and (iii) we need to establish a biologically plausible
microbiome [109,110]. and clinically meaningful relationship between a certain micro-
It is therefore not surprising that exposure to smoking causes biome profile and a certain disease [1]. This holds true especially for
modifications of the gut microbiome. A decrease of Bifidobacteria the gastric microbiota, as to date the composition of the normal
and of proprionic and butirric acid in the cecum has been reported “core” gastric microbiota is still not completely defined. The precise
by Tomoda et al. in rats exposed to smoking [111]. role of potential modulators of gastric microbiota, such as smoking
In a similar animal model, even passive smoking increases and alcohol, still needs to be clarified, but current available data
Clostridium sp and reduces Firmicutes phylum (Lactococcus and suggest a key role in gastric diseases, in particular as risk modulator
Ruminococcus sp.) and Enterobacteriaceae [112] for gastric cancer. As far as regards the interaction between alcohol
In a more recent study, the effect of chronic exposure to smoking and smoking and the intestinal microbiome, there is sufficient ev-
compared to normal air was evaluated on the mucosa and on the idence suggesting that alcohol causes depletion of certain microbes
microbiome composition of rats at different levels of the GI tract and increase of others, and that the resulting dysbiosis, coupled
(ileum, cecum, colon) employing the Illumina high throughput with leaky gut causes increased inflammatory changes in the in-
sequencing [113]. testine and liver. These alterations seem particularly relevant for
Microbial clusters in the cecum and colon differed significantly the pathogenesis of alcoholic liver disease. Specific bacteria such as
in animals exposed to smoking with an important increase of Lactobacilli and A. muciniphila that are depleted by alcohol seem
Lachnospiraceae sp. The mRNA expression of mucins was also able to exert a significant anti-inflammatory effect in this setting.
modified by smoking, as Muc2 and Muc3 increased in the ileum, Interestingly, dysbiosis itself in non-alcoholic steatohepatitis in-
whereas Muc4 increased in the distal colon of smoke-exposed crease alcohol production in a potential vicious circle. Alcohol-
mice, while the expression of tight junction genes remained induced modifications of the gut microbiome also seem associ-
unchanged. ated with other non-gastrointestinal disorders, including pneu-
monia and behavioral patterns. The role of smoking in determining
1.6.2. Studies in humans modifications of the gut microbiome has been investigated spe-
The reported increase of Clostridia induced by smoking in rats cifically in the context of inflammatory bowel disease, and links
seems also indirectly confirmed in humans by an epidemiological with the long-standing clinical observation of smoking being pro-
study on 16781 elderly subjects. The authors reported an increased tective toward ulcerative colitis and deleterious for Crohn's disease.
rate of C. difficile infection in smokers, with odds 33% greater in In the next few years the puzzle of knowledge on this topic will
former smokers and 80% greater in current smokers when certainly become more complete.
compared to never smokers [114]. However, in this complex research field, many confounders
Smoking withdrawal in humans determines profound changes come into play. Not least because the microbiota-gut-brain axis is
in the microbiome with increased microbial diversity [115], an in- bidirectional, as shown by the effects of stress on gut physiology,
crease of Firmicutes and Actinobacteria and a decrease of immune function and gut microbiota composition [121]. This may
586 G. Capurso, E. Lahner / Best Practice & Research Clinical Gastroenterology 31 (2017) 579e588

be particular true for life style habits as smoking and alcohol con- 2017;34:1070e86.
[8] Delgado S, Cabrera-Rubio R, Mira A, Sua rez A, Mayo B. Microbiological survey
sumption related with psychological and social factors, which
of the human gastric ecosystem using culturing and pyrosequencing
might in turn influence the gastric microbiota. methods. Microb Ecol 2013;65:763e72.
[9] Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, sta-
bility and resilience of the human gut microbiota. Nature 2012;489:220e30.
[10] Merrell DS, Camilli A. The cadA gene of Vibrio cholerae is induced during
Practice Points: infection and plays a role in acid tolerance. Mol Microbiol 1999;34:836e49.
[11] Yang I, Nell S, Suerbaum S. Survival in hostile territory: the microbiota of the
stomach. FEMS Microbiol Rev 2013;37:736e61.
[12] Loman NJ, Constantinidou C, Chan JZ, Halachev M, Sergeant M, Penn CW,
et al. High-throughput bacterial genome sequencing: an embarrassment of
choice, a world of opportunity. Nat Rev Microbiol 2012;10:599e606.
 Alcohol and smoking deeply modify the microbiome of [13] Ianiro G, Molina-Infante J, Gasbarrini A. Gastric microbiota. Helicobacter
both the upper and the lower gastrointestinal tract 2015;1:68e71.
 The resulting dysbiosis is related with immune response, [14] Zilberstein B, Quintanilha AG, Santos MA, Pajecki D, Moura EG, Alves PR,
et al. Digestive tract microbiota in healthy volunteers. Clinics 2007;62:
“leaky gut” and systemic consequences contributing to 47e54.
gastric, intestinal, liver and distant organs damage [15] Sharma BK, Santana IA, Wood EC, Walt RP, Pereira M, Noone P, et al. Intra-
 Modulation of the gastrointestinal microbiome with spe- gastric bacterial activity and nitrosation before, during, and after treatment
with omeprazole. Br Med J 1984;289:717e9.
cific bacteria, such as LGG or A muciniphila has the po-
[16] Vartoukian SR, Palmer RM, Wade WG. Strategies for culture of 'unculturable'
tential to modify disease course of alcoholic and non- bacteria. FEMS Microbiol Lett 2010;309:1e7.
alcoholic liver disease, inflammatory bowel disease and [17] Human Microbiome Project Consortium. A framework for human micro-
biome research. Nature 2012;486:215e21.
other disorders
[18] Sekirov I, Russell SL, Antunes LC, Finlay BB. Gut microbiota in health and
disease. Physiol Rev 2010;90:859e904.
[19] Liu Z, Lozupone C, Hamady M, Bushman FD, Knight R. Short pyrosequencing
reads suffice for accurate microbial community analysis. Nucleic Acids Res
Research agenda: 2007;35:e120.
[20] Wu WM, Yang YS, Peng LH. Microbiota in the stomach: new insights. J Dig
Dis 2014;15:54e61.
[21] Schulz C, Koch N, Schütte K, Pieper DH, Malfertheiner P. H. pylori and its
modulation of gastrointestinal microbiota. J Dig Dis 2015;16:109e17.
[22] Frenck Jr RW, Clemens J. Helicobacter in the developing world. Microbes
Infect 2003;5:705e13.
[23] Josenhans C, Suerbaum S. The role of motility as a virulence factor in bac-
 The exact cause-effect relation between alcohol and teria. Int J Med Microbiol 2002;291:605e14.
smoking and changes of the gastrointestinal microbiome [24] McColl KE, el-Omar E, Gillen D. Helicobacter pylori gastritis and gastric
needs further exploration with high throughput method- physiology. Gastroenterol Clin North Am 2000;29:687e703.
[25] Osaki T, Matsuki T, Asahara T, Zaman C, Hanawa T, Yonezawa H, et al.
ologies, especially in the upper GI tract. Comparative analysis of gastric bacterial microbiota in Mongolian gerbils
 Detailed controlled studies are necessary to define the after long-term infection with Helicobacter pylori. Microb Pathog 2012;53:
role of microbiome modulation on the immune response 12e8.
[26] Tan MP, Kaparakis M, Galic M, Pedersen J, Pearse M, Wijburg OL, et al.
and systemic activation of pro-inflammatory pathways Chronic Helicobacter pylori infection does not significantly alter the micro-
 The role of the gut-brain axis is also a potential interesting biota of the murine stomach. Appl Environ Microbiol 2007;73:1010e3.
area for research as dysbiosis can influence the central [27] Bik EM, Eckburg PB, Gill SR, Nelson KE, Purdom EA, Francois F, et al. Mo-
lecular analysis of the bacterial microbiota in the human stomach. Proc Natl
nervous system response Acad Sci U. S. A 2006;103:732e7.
[28] Andersson AF, Lindberg M, Jakobsson H, B€ ackhed F, Nyren P, Engstrand L.
Comparative analysis of human gut microbiota by barcoded pyrosequencing.
PLoS One 2008;3:e2836.
[29] Maldonado-Contreras A, Goldfarb KC, Godoy-Vitorino F, Karaoz U,
Conflict of interest
Contreras M, Blaser MJ, et al. Structure of the human gastric bacterial com-
munity in relation to Helicobacter pylori status. ISME J 2011;5:574e9.
None. [30] Li TH, Qin Y, Sham PC, Lau KS, Chu KM, Leung WK. Alterations in gastric
microbiota after H. Pylori eradication and in different histological stages of
gastric carcinogenesis. Sci Rep 2017;7:44935.
Sources of funding and conflicts [31] Li XX, Wong GL, To KF, Wong VW, Lai LH, Chow DK, et al. Bacterial micro-
biota profiling in gastritis without Helicobacter pylori infection or non-
steroidal anti-inflammatory drug use. PLoS One 2009;4:e7985.
The authors declare no supporting funds and no conflicts of [32] Stearns JC, Lynch MD, Senadheera DB, Tenenbaum HC, Goldberg MB,
interest. Cvitkovitch DG, et al. Bacterial biogeography of the human digestive tract. Sci
Rep 2011;1:170.
[33] Engstrand L, Lindberg M. Helicobacter pylori and the gastric microbiota. Best
References Pract Res Clin Gastroenterol 2013;27:39e45.
[34] Swidsinski A, Sydora BC, Doerffel Y, Loening-Baucke V, Vaneechoutte M,
[1] Quigley EMM. Gut microbiome as a clinical tool in gastrointestinal disease Lupicki M, et al. Viscosity gradient within the mucus layer determines the
management: are we there yet? Nat Rev Gastroenterol Hepatol 2017;14: mucosal barrier function and the spatial organization of the intestinal
315e20. microbiota. Inflamm Bowel Dis 2007;13:963e70.
[2] Gill SR, Pop M, Deboy RT, Eckburg PB, Turnbaugh PJ, Samuel BS, et al. Met- [35] Goddard AF, Spiller RC. The effect of omeprazole on gastric juice viscosity, pH
agenomic analysis of the human distal gut microbiome. Science 2006;312: and bacterial counts. Alimentary Pharmacol Ther 1996;10:105e9.
1355e9. [36] Hardbower DM, de Sablet T, Chaturvedi R, Wilson KT. Chronic inflammation
[3] Human Microbiome Project Consortium: structure, function and diversity of and oxidative stress: the smoking gun for Helicobacter pylori-induced
the healthy human microbiome. Nature 2012;486:207e14. gastric cancer? Gut Microbes 2013;4:475e81.
[4] Faith JJ, Guruge JL, Charbonneau M, Subramanian S, Seedorf H, Goodman AL, [37] Rolig AS, Cech C, Ahler E, Carter JE, Ottemann KM. The degree of Helicobacter
et al. The long-term stability of the human gut microbiota. Science 2013;5: pylori-triggered inflammation is manipulated by preinfection host micro-
84e7. biota. Infect Immun 2013;81:1382e9.
[5] Owyang C, Wu GD. The gut microbiome in health and disease. Gastroen- [38] Yu G, Gail MH, Shi J, Klepac-Ceraj V, Paster BJ, Dye BA, et al. Association
terology 2014;146:1433e6. between upper digestive tract microbiota and cancer-predisposing states in
[6] Martinsen TC, Bergh K, Waldum HL. Gastric juice: a barrier against infectious the esophagus and stomach. Cancer Epidemiol Biomarkers Prev 2014;23:
diseases. Basic Clin Pharmacol Toxicol 2005;96:94e102. 735e41.
[7] Yu LY, Sun LN, Zhang XH, Li YQ, Yu L, Yuan ZQ, et al. A review of the novel [39] Abreu MT, Peek Jr RM. Gastrointestinal malignancy and the microbiome.
application and potential adverse effects of proton pump inhibitors. Adv Ther Gastroenterology 2014;146:1534e46. e3.
G. Capurso, E. Lahner / Best Practice & Research Clinical Gastroenterology 31 (2017) 579e588 587

[40] Eun CS, Kim BK, Han DS, Kim SY, Kim KM, Choi BY, et al. Differences in gastric this disease? Curr Gastroenterol Rep 2016;18:34.
mucosal microbiota profiling in patients with chronic gastritis, intestinal [68] Vakevainen S, Mentula S, Nuutinen H, Salmela KS, Jousimies-Somer H,
metaplasia, and gastric cancer using pyrosequencing methods. Helicobacter Farkkila M, et al. Ethanol-derived microbial production of carcinogenic
2014;19:407e16. acetaldehyde in achlorhydric atrophic gastritis. Scand J Gastroenterol
[41] Correa P. Human gastric carcinogenesis: a multistep and multifactorial 2002;37:648e55.
processefirst american cancer society award lecture on cancer epidemiology [69] Yokoyama A, Yokoyama T, Omori T, Matsushita S, Mizukami T, Takahashi H,
and prevention. Cancer Res 1992;52:6735e40. et al. Helicobacter pylori, chronic atrophic gastritis, inactive aldehyde
[42] Wang LL, Yu XJ, Zhan SH, Jia SJ, Tian ZB, Dong QJ. Participation of microbiota dehydrogenase-2, macrocytosis and multiple upper aerodigestive tract
in the development of gastric cancer. World J Gastroenterol 2014;20: cancers and the risk for gastric cancer in alcoholic Japanese men.
4948e52. J Gastroenterol Hepatol 2007;22:210e7.
[43] Sanduleanu S, Jonkers D, De Bruine A, Hameeteman W, Stockbrügger RW. [70] Jakszyn P, Bingham S, Pera G, Agudo A, Luben R, Welch A, et al. Endogenous
Non-Helicobacter pylori bacterial flora during acid-suppressive therapy: versus exogenous exposure to N-nitroso compounds and gastric cancer risk
differential findings in gastric juice and gastric mucosa. Aliment Pharmacol in the European Prospective Investigation into Cancer and Nutrition (EPIC-
Ther 2001;15:379e88. EURGAST) study. Carcinogenesis 2006;27:1497e501.
[44] Dicksved J, Lindberg M, Rosenquist M, Enroth H, Jansson JK, Engstrand L. [71] Ladeiras-Lopes R, Pereira AK, Nogueira A, Pinheiro-Torres T, Pinto I, Santos-
Molecular characterization of the stomach microbiota in patients with Pereira R, et al. Smoking and gastric cancer: systematic review and meta-
gastric cancer and in controls. J Med Microbiol 2009;58:509e16. analysis of cohort studies. Cancer Causes Control 2008;19:689e701.
[45] Forsythe SJ, Cole JA. Nitrite accumulation during anaerobic nitrate reduction [72] Praud D, Rota M, Pelucchi C, Bertuccio P, Rosso T, Galeone C, et al. Cigarette
by binary suspensions of bacteria isolated from the achlorhydric stomach. smoking and gastric cancer in the stomach cancer pooling (StoP) project. Eur
J Gen Microbiol 1987;133:1845e9. J Cancer Prev 2016. https://doi.org/10.1097/CEJ.0000000000000290.
[46] Sohn SH, Kim N, Jo HJ, Kim J, Park JH, Nam RH, et al. Analysis of gastric body [73] Yao QW, Zhou DS, Peng HJ, Ji P, Liu DS. Association of periodontal disease
microbiota by pyrosequencing: possible role of bacteria other than Heli- with oral cancer: a meta-analysis. Tumour Biol 2014;35:7073e7.
cobacter pylori in the gastric carcinogenesis. J Cancer Prev 2017;22:115e25. [74] Vogtmann E, Flores R, Yu G, Freedman ND, Shi J, Gail MH, et al. Association
[47] Thorell K, Bengtsson-Palme J, Liu OH, Gonzales RVP, Nookaew I, Rabeneck L, between tobacco use and the upper gastrointestinal microbiome among
et al. In vivo analysis of the viable microbiota and Helicobacter pylori tran- Chinese men. Cancer Causes Control 2015;26:581e8.
scriptome in gastric infection and early stages of carcinogenesis. Infect [75] Suzuki T, Matsuo K, Ito H, Sawaki A, Hirose K, Wakai K, et al. Smoking in-
Immun 2017. https://doi.org/10.1128/IAI.00031-17. creases the treatment failure for Helicobacter pylori eradication. Am J Med
[48] Rocco A, Compare D, Angrisani D, Sanduzzi Zamparelli M, Nardone G. 2006;119:217e24.
Alcoholic disease: liver and beyond. World J Gastroenterol 2014;20: [76] Itskoviz D, Boltin D, Leibovitzh H, Tsadok Perets T, Comaneshter D, Cohen A,
14652e9. et al. Smoking increases the likelihood of Helicobacter pylori treatment
[49] Teyssen S, Singer MV. Alcohol-related diseases of the oesophagus and failure. Dig Liver Dis 2017;49:764e8.
stomach. Best Pract Res Clin Gastroenterol 2003;17:557e73. [77] Leung C, Rivera L, Furness JB, Angus P. The role of the gut microbiota in
[50] Franke A, Teyssen S, Harder H, Singer MV. Effect of ethanol and some alco- NAFLD. Nat Rev Gastroenterol Hepatol 2016;13:412e25.
holic beverages on gastric emptying in humans. Scand J Gastroenterol [78] Aw Yan, De Fouts, Brandl J, Starkel P, Torralba M, Schott E, et al. Enteric
2004;39:638e44. dysbiosis associated with a mouse model of alcoholic liver disease. Hep-
[51] Teyssen S, Gonza lez-Calero G, Schimiczek M, Singer MV. Maleic acid and atology 2011;53:96e105.
succinic acid in fermented alcoholic beverages are the stimulants of gastric [79] Mutlu EA, Keshavarzian A, Engen P, Forsyth CB, Sikaroodi M, Gillevet P. In-
acid secretion. J Clin Invest 1999;103:707e13. testinal dysbiosis a possible mechanism of alcohol-induced endotoxemia and
[52] Franke A, Teyssen S, Singer MV. Alcohol-related diseases of the esophagus alcoholic steatohepatitis in rats Alcohol. Clin Exp Res 2009;33:1836e46.
and stomach. Dig Dis 2005;23:204e13. [80] Bull-Otterson L, Feng W, Kirpich I, Wang Y, Qin X, Liu Y, et al. Metagenomic
[53] Boltin D, Niv Y. Pharmacological and alimentary alteration of the gastric analyses of alcohol induced pathogenic alterations in the intestinal micro-
barrier. Best Pract Res Clin Gastroenterol 2014;28:981e94. biome and the effect of Lactobacillus rhamnosus GG treatment. PloS one
[54] Brenner H, Rothenbacher D, Bode G, Adler G. Relation of smoking and alcohol 2013;8:e53028.
and coffee consumption to active Helicobacter pylori infection: cross [81] Campos Canesso M, Lacerda NL, Ferreira CM, Gonçalves JL, Almeida D,
sectional study. BMJ 1997;315:1489e92. Gamba C, et al. Comparing the effects of acute alcohol consumption in germ-
[55] Murray LJ, Lane AJ, Harvey IM, Donovan JL, Nair P, Harvey RF. Inverse rela- free and conventional mice: the role of the gut microbiota BMC Microbi-
tionship between alcohol consumption and active Helicobacter pylori ology, vol. 14; 2014. p. 240.
infection: the Bristol Helicobacter project. Am J Gastroenterol 2002;97: [82] Hartmann P, Chen P, Hj Wang, Wang L, Df Mccole, Brandl K, et al. Deficiency
2750e5. of intestinal mucin-2 ameliorates experimental alcoholic liver disease in
[56] Liu SY, Han XC, Sun J, Chen GX, Zhou XY, Zhang GX. Alcohol intake and mice. Hepatology 2013;58:108e19.
Helicobacter pylori infection: a dose-response meta-analysis of observa- [83] Derrien M, Belzer C, de Vos WM. Akkermansia muciniphila and its role in
tional studies. Infect Dis (Lond) 2016;48:303e9. regulating host functions. Microb Pathog 2017;106:171e81.
[57] Baena Jose  M, Lo
pez C, Hidalgo A, Rams F, Jimenez S, et al. Relation between [84] Lowe PP, Gyongyosi B, Satishchandran A, Iracheta-Vellve A, Ambade A,
alcohol consumption and the success of Helicobacter pylori eradication Kodys K, et al. Alcohol-related changes in the intestinal microbiome influ-
therapy using omeprazole, clarithromycin and amoxicillin for 1 week. Eur J ence neutrophil infiltration, inflammation and steatosis in early alcoholic
Gastroenterol Hepatol 2002;14:291e6. hepatitis in mice. PLoS One 2017. https://doi.org/10.1371/
[58] Ma SH, Jung W, Weiderpass E, Jang J, Hwang Y, Ahn C, et al. Impact of alcohol journal.pone.0174544.
drinking on gastric cancer development according to Helicobacter pylori [85] Grander C, Adolph TE, Wieser V, Lowe P, Wrzosek L, Gyongyosi B, et al.
infection status. Br J Cancer 2015;113:1381e8. Recovery of ethanol-induced Akkermansia muciniphila depletion ameliorates
[59] Ma K, Baloch Z, He TT, Xia X. Alcohol consumption and gastric cancer risk: a alcoholic liver disease. Gut 2017. https://doi.org/10.1136/gutjnl-2016-
meta-analysis. Med Sci Monit 2017;23:238e46. 313432.
[60] Ferronha I, Bastos A, Lunet N. Prediagnosis lifestyle exposures and survival of [86] Betrapally NS, Gillevet PM, Bajaj JS. Changes in the intestinal microbiome and
patients with gastric cancer: systematic review and meta-analysis. Eur J alcoholic- and non-alcoholic liver diseasesdcauses or effects? Gastroenter-
Cancer Prev 2012;21:449e52. ology 2016;150:1745e55.
[61] Buckland G, Travier N, Huerta JM, Bueno-de-Mesquita HB, Siersema PD, [87] Samuelson DR, Shellito JE, Maffei VJ, Tague ED, Campagna SR, Blanchard EE,
Skeie G, et al. Healthy lifestyle index and risk of gastric adenocarcinoma in et al. Alcohol-associated intestinal dysbiosis impairs pulmonary host defense
the EPIC cohort study. Int J Cancer 2015;137:598e606. against Klebsiella pneumoniae. PLoS Pathog 2017;13:e1006426. https://doi.
[62] Tramacere I, Negri E, Pelucchi C, Bagnardi V, Rota M, Scotti L, et al. A meta- org/10.1371/journal.ppat.1006426.
analysis on alcohol drinking and gastric cancer risk. Ann Oncol 2012;23: [88] Rao RK, Seth A, Sheth P. Recent advances in alcoholic liver disease I: role of
28e36. intestinal permeability and endotoxemia in alcoholic liver disease. American
[63] Tamura T, Wada K, Tsuji M, Konishi K, Kawachi T, Hori A, et al. Association of Journal of Physiology. Gastrointest Liver Physiol 2004;286:G881e4.
alcohol consumption with the risk of stomach cancer in a Japanese popu- [89] Kakiyama G, Pandak WM, Gillevet PM, Hylemon PB, Heuman DM, Daita K,
lation: a prospective cohort study. Eur J Cancer Prev 2016. https://doi.org/ et al. Modulation of the fecal bile acid profile by gut microbiota in cirrhosis.
10.1097/CEJ.0000000000000278. J hepatol 2013;58:949e55.
[64] Pan KF, Zhang L, Gerhard M, Ma JL, Liu WD, Ulm K, et al. A large randomised [90] Bajaj JS, Heuman DM, Hylemon PB, Sanyal AJ, White MB, Monteith P, et al.
controlled intervention trial to prevent gastric cancer by eradication of Altered profile of human gut microbiome is associated with cirrhosis and its
Helicobacter pylori in Linqu County, China: baseline results and factors complications. J hepatol 2014c;60:940e7.
affecting the eradication. Gut 2016;65:9e18. [91] Chen Y, Yang F, Lu H, Wang B, Lei D, Wang Y, et al. Characterization of fecal
[65] Na HK, Lee JY. Molecular basis of alcohol-related gastric and colon cancer. Int microbial communities in patients with liver cirrhosis. Hepatology 2011;54:
J Mol Sci 2017. https://doi.org/10.3390/ijms18061116. 562e72.
[66] IARC working group on the evaluation of carcinogenic risks to humans [92] Bajaj JS, Heuman DM, Hylemon PB, Sanyal AJ, Puri P, Sterling RK, et al.
personal habits and indoor combustions. The role of acetaldehyde in alcohol- Randomised clinical trial: Lactobacillus GG modulates gut microbiome,
induced carcinogenesis. IARC monogr. Eval Carcinog Risks Hum 2012;100E: metabolome and endotoxemia in patients with cirrhosis. Alimentary Phar-
471. macol Ther 2014;39:1113e25.
[67] den Hoed CM, Kuipers EJ. Gastric cancer: how can we reduce the incidence of [93] Queipo-Ortuno MI, Boto-Ordonez M, Murri M, et al. Influence of red wine
588 G. Capurso, E. Lahner / Best Practice & Research Clinical Gastroenterology 31 (2017) 579e588

polyphenols and ethanol on the gut microbiota ecology and biochemical [108] Zijlstra FJ, Srivastava ED, Rhodes M, van Dijk AP, Fogg F, Samson HJ, et al.
biomarkers. Am J Clin Nutr 2012;95:1323e34. Effect of nicotine on rectal mucus and mucosal eicosanoids. Gut 1994;35:
[94] Bressa C, Bailen-Andrino M, Pe rez-Santiago J, Gonzalez-Soltero R, Pe
rez M, 247e51.
Montalvo-Lominchar MG, et al. Differences in gut microbiota profile be- [109] Claus SP, Guillou H, Ellero-Simatos S. The gut microbiota: a major player in
tween women with active lifestyle and sedentary women. PLoS One the toxicity of environmental pollutants? npj Biofilms Microbiomes 2016;2:
2017;12:e0171352. https://doi.org/10.1371/journal.pone.0171352. 16003.
[95] Llopis M, Cassard AM, Wrzosek L, Boschat L, Bruneau A, Ferrere G, et al. [110] Van de Wiele T, Vanhaecke L, Boeckaert C, Peru K, Headley J, Verstraete W,
Intestinal microbiota contributes to individual susceptibility to alcoholic et al. Human colon microbiota transform polycyclic aromatic hydrocarbons
liver disease. Gut 2016;65:830e9. to estrogenic metabolites environmental health perspectives, 113; 2005.
[96] Leclercq S, Matamoros S, Cani PD, Neyrinck AM, Jamar F, Sta €rkel P, et al. p. 6e10.
Intestinal permeability, gut-bacterial dysbiosis, and behavioral markers of [111] Tomoda K, Kubo K, Asahara T, Andoh A, Nomoto K, Nishii Y, et al. Cigarette
alcoholdependence severity. Proc Natl Acad Sci U. S. A 2014;111:E4485e93. smoke decreases organic acids levels and population of bifidobacterium in
[97] Clark DP. The fermentation pathways of Escherichia coli. FEMS Microbiol Rev the caecum of rats. J Toxicol Sci 2011;36:261e6.
1989;5:223e34. [112] Wang H, Zhao JX, Hu N, Ren J, Du M, Zhu MJ. Side-stream smoking reduces
[98] Zhu L, et al. Characterization of gut microbiomes in nonalcoholic steatohe- intestinal inflammation and increases expression of tight junction proteins.
patitis (NASH) patients: a connection between endogenous alcohol and World J Gastroenterol 2012;18:2180e7.
NASH. Hepatology 2013;57:601e9. [113] Allais L, Kerckhof FM, Verschuere S, Bracke KR, De Smet R, Laukens D, et al.
[99] Nair S, Cope K, Risby TH, Diehl AM. Obesity and female gender increase Chronic cigarette smoke exposure induces microbial and inflammatory shifts
breath-ethanol concentration: potential implications for the pathogenesis of and mucin changes in the murine gut. Environ Microbiol 2016;18:1352e63.
nonalcoholic steatohepatitis. Am J Gastroenterol 2001;96:1200e4. [114] Rogers MAM, Greene MT, Saint S, Chenoweth CE, Malani PN, Trivedi I, et al.
[100] Hecht SS. Tobacco carcinogens, their biomarkers and tobacco-induced can- Higher rates of Clostridium difficile infection among smokers. PLoS One
cer. Nat Rev Cancer 2003;3:733e44. 2012;7:e4209.
[101] Verschuere S, Rebecca De Smet R, Liesbeth Allais L, Cuvelier CA. The effect of [115] Biedermann L, Zeitz J, Mwinyi J, Sutter-Minder E, Rehman ASJ, et al. Smoking
smoking on intestinal inflammation: what can be learned from animal cessation induces profound changes in the composition of the intestinal
models? J Crohn's Colitis 2012;6:1e12. microbiota in humans. PlosOne 2013;8:e59260.
[102] Thomas GA, Pullan RD, Zijlstra FJ, Rankin BJ, Russell MA, Feyerabend C, et al. [116] Koliada A, Syzenko G, Moseiko V, Budovska L, Puchkov K, Perederiy V. As-
Effect of nicotine on large bowel mucus thickness, eicosanoids and faecal sociation between body mass index and Firmicutes/Bacteroidetes ratio in an
proteinase in ferrets. Eur J Gastroenterol Hepatol 1997;9:179e82. adult Ukrainian population. BMC Microbiol 2017;17:120.
[103] Van Dijk JP, Madretsma GS, Keuskamp ZJ, Zijlstra FJ. Nicotine inhibits cyto- [117] Harries AD, Baird A, Rhodes J. Non-smoking: a feature of ulcerative colitis.
kine synthesis by mouse colonic mucosa. Eur J Pharmacol 1995;278:R11e2. BMJ 1982;284:706.
[104] Verschuere S, Bracke KR, Demoor T, Plantinga M, Verbrugghe P, [118] Birrenbach T, Bocker U. Inflammatory bowel disease and smoking: a review
Ferdinande L, et al. Cigarette smoking alters epithelial apoptosis and im- of epidemiology, pathophysiology, and therapeutic implications. Inflamm
mune composition in murine GALT. Lab Invest 2011;91:1056e67. Bowel Dis 2004;10:848e59.
[105] Eliakim R, Fan QX, Babyatsky MW. Chronic nicotine administration differ- [119] Opstelten JL, Plassais J, van Mil SWC, Achouri E. Gut microbial diversity is
entially alters jejunal and colonic inflammation in interleukin-10 deficient reduced in smokers with Crohn's disease. Inflamm Bowel Dis 2016;22:
mice. Eur J Gastroenterol Hepatol 2002;14:607e14. 2070e7.
[106] Eliakim R, Karmeli F, Cohen P, Heyman SN, Rachmilewitz D. Dual effect of [120] Wright E, Kamm M, Wagner J, Teo SM, De Cruz P, Hamilton A, et al. What
chronic nicotine administration: augmentation of jejunitis and amelioration causes recurrence of Crohn's Disease after intestinal resection? A prospective
of colitis induced by iodoacetamide in rats. Int J Colorectal Dis 2001;16: evaluation of microbiota, smoking and anti-TNF therapy. Results POCER
14e21. study J Crohn's Colitis 2015:S436.
[107] Guo X, Ko JK, Mei QB, Cho CH. Aggravating effect of cigarette smoke exposure [121] De Palma G, Collins SM, Bercik P, Verdu EF. The microbiota-gut-brain axis in
on experimental colitis is associated with leukotriene B4 and reactive oxy- gastrointestinal disorders: stressed bugs, stressed brain or both? J Physiol
gen metabolites. Digestion 2001;63:180e7. 2014;592:2989e97.

You might also like