You are on page 1of 12

bs_bs_banner

Environmental Microbiology (2015) doi:10.1111/1462-2920.12934

Chronic cigarette smoke exposure induces microbial


and inflammatory shifts and mucin changes in the
murine gut

Liesbeth Allais,1* Frederiek-Maarten Kerckhof,2 decreased Tgf-β in the proximal colon. Tight junction
Stephanie Verschuere,3 Ken R. Bracke,4 gene expression remained unchanged. We infer that
Rebecca De Smet,1 Debby Laukens,5 the modulating role of chronic smoke exposure as a
Pieter Van den Abbeele,2 Martine De Vos,5 latently present risk factor in the gut may be driven by
Nico Boon,2 Guy G. Brusselle,4 Claude A. Cuvelier1 the altered epithelial mucus profiles and changes in
and Tom Van de Wiele2 microbiome composition and immune factors.
1
Department of Medical and Forensic Pathology,
2
Laboratory of Microbial Ecology and Technology Introduction
(LabMET), Faculty of Bioscience Engineering, Ghent
Inflammatory bowel diseases (IBD), comprising Crohn’s
University, Ghent, Belgium.
3
disease (CD) and ulcerative colitis (UC), result from a
Department of Pathology, AZ Sint-Jan, Brugge,
complex interplay between environmental factors, genet-
Belgium.
4
ics and intestinal microbiota, which combine to initiate and
Laboratory for Translational Research in Obstructive
perpetuate chronic inflammation in the gastrointestinal
Pulmonary Diseases, Department of Respiratory
tract. CD is associated with a transmural and discontinu-
Medicine, Ghent University Hospital, Ghent, Belgium.
5
ous inflammation that most frequently involves the ileum
Department of Gastroenterology, Ghent University,
and colon, but can affect the entire gastrointestinal tract,
Ghent, Belgium.
while in UC, a superficial inflammation occurs which is
limited to the colon (Khan and Collins, 2006; Jin et al.,
Summary 2012). The prevalence of IBD ranges from 174 to 210 for
CD and 79 to 122 for UC per 100 000 inhabitants in
Inflammatory bowel diseases (IBD) are complex mul-
Western countries and is still increasing (Loftus, 2004;
tifactorial diseases characterized by an inappropriate
Lakatos, 2006; Hovde and Moum, 2012). Disrupted intes-
host response to an altered commensal microbiome
tinal homeostasis and abnormal immune responses to
and dysfunctional mucus barrier. Cigarette smoking is
host intestinal resident microbiota play a major role in the
the best known environmental risk factor in IBD. Here,
pathogenesis, although the exact mechanisms have not
we studied the influence of chronic smoke exposure
yet been elucidated (Strober et al., 2007; Xavier and
on the gut microbiome, mucus layer composition and
Podolsky, 2007).
immune factors in conventional mice. We compared
Disturbance of the microbial equilibrium, termed
smoke-exposed with air-exposed mice (n = 12) after a
dysbiosis, is characterized by quantitative and qualitative
smoke exposure of 24 weeks. Both Illumina sequenc-
changes in the microbiota. This is marked by an increase of
ing (n = 6) and denaturing gradient gel electrophoresis
common bacterial inhabitants of the gut that become
(n = 12) showed that bacterial activity and community
pathogenic under permissive conditions (Stecher et al.,
structure were significantly altered in the colon due
2013). Dysbiosis provokes dysregulation of adaptive
to smoke exposure. Interestingly, an increase of
immune responses in the gut and is increasingly recog-
Lachnospiraceae sp. activity in the colon was
nized as a contributing factor in the pathogenesis of IBD
observed. Also, the mRNA expression of Muc2 and
(Carbonnel et al., 2009; Round and Mazmanian, 2009).
Muc3 increased in the ileum, whereas Muc4 increased
Previous studies showed that bacterial diversity is
in the distal colon of smoke-exposed mice (n = 6).
decreased in stool samples of IBD patients (Manichanh
Furthermore, we observed increased Cxcl2 and
et al., 2006; Erickson et al., 2012). Alterations in the
decreased Ifn-γ in the ileum, and increased Il-6 and
abundance of species of all prominent intestinal
phyla Firmicutes, Bacteroidetes, Verrucomicrobia, Actino-
Received 9 February, 2015; accepted 28 May, 2015. *For corre-
spondence. E-mail Liesbeth.allais@ugent.be or liesbeth.allais@ bacteria and Proteobacteria are associated with the devel-
gmail.com; Tel. +32 9 332 49 53; Fax +32-93324965. opment of IBD (Frank et al., 2007; Png et al., 2010b).
© 2015 Society for Applied Microbiology and John Wiley & Sons Ltd
2 L. Allais et al.

Cigarette smoke (CS) is the most prominent environ- exposed and 12 air-exposed mice was analysed by dena-
mental risk factor for developing CD; however, it turing gradient gel electrophoresis (DGGE) as a profiling
exerts a protective role in UC (Persson et al., 1990; technique. Dendrograms applying the abundance-based
Ananthakrishnan, 2013). In addition, smoke exposure Jaccard index (Fig. 1) and Yue and Clayton’s theta index
may have an important impact on the composition and (Fig. S1) were used to visualize the clustering of the
dynamics of the gut microbiome. For instance, the samples. Considering the distinct intestinal regions, the
Bifidobacterium population increased in the caecum of dendrograms showed clear clusters for air-exposed mice
rats after 4 weeks of CS exposure (Tomoda et al., 2011). (49 ± 11% and 58 ± 9% within-group similarity, respec-
In mouse, side-stream smoking increased Clostridium tively), separated from smoke-exposed mice in caecum
sp., but decreased the Firmicutes phylum (Lactococcus and distal colon, suggesting a bacterial shift (P = 0.001 for
and Ruminococcus sp.), the Enterobacteriaceae family both using multivariate analysis of variance (MANOVA)).
and the segmented filamentous bacteria in the caecum This kind of separated clusters was not observed in the
(Wang et al., 2012). In human studies, smoking has dendrogram of the ileal samples, with the air-exposed
been associated with a higher rate of Clostridium difficile samples only showing 42 ± 11% within-group similarity.
infection, in which current smokers (actively smoking
during the study) have the highest risk compared with
The activity of specific bacterial species is influenced by
former (quitted smoking before the start of the study)
smoke exposure
and never smokers (never smoked in their whole life)
(Rogers et al., 2012). Moreover, CS is a known risk A shift in the microbial composition may potentially
determinant for both bacterial and viral infections imply a shift in microbial activity. Using cDNA samples,
through alterations in cell- and humoral-mediated we evaluated the activity of the microbiome with Illumina
immune responses in the respiratory tract (Arcavi and sequencing as a more high-resolution method. We ran-
Benowitz, 2004). We recently demonstrated that CS trig- domly selected 12 mice (six smoke- and six air-exposed
gers the gut immune system through the recruitment of mice, originating from four distinct cages) of which
immune cells to the Peyer’s patches (PP), the main lym- ileal, caecal and distal colonic samples were included in
phoid organs in the gut, and through the induction the analysis. Changes in 16S rRNA levels were evalu-
of autophagy and apoptosis in the follicle-associated ated by the sparse partial least square discriminant
epithelium (FAE) overlying the PP (Verschuere et al., analysis (sPLS-DA). This clearly showed a distinct ordi-
2011; 2012). nation for proximal and distal colon between the smoke-
Many studies have investigated the impact of specific and air-exposed murine microbiome activity (Fig. 2),
intestinal bacteria on host gene and protein expression in but not for ileum, which parallels the shift in microbial
the intestine and their role in IBD development (Rolli et al., composition as shown by the DGGE-based clustering
2010). Nevertheless, the role of smoking in the emergence (Fig. 1).
of dysbiosis, which might modulate the risk for the To further investigate the effect of smoke exposure on
development of IBD, still needs further investigation. CS bacterial activity in the murine gut, we used Illumina
alters host–microorganism interaction dynamics in the sequencing to identify the operating taxonomic units
airways, causing respiratory tract infections and contribut- (OTUs) showing changes in activity in the different gut
ing to chronic obstructive pulmonary disease (COPD) regions of six smoke- and six air-exposed mice. We
(Garmendia et al., 2012). This study addresses the effect applied the sPLS-DA method to identify the specific OTUs
of chronic CS exposure on the gut mucosa and its micro- in each separate intestinal region. All relevant OTUs were
bial environment. We examined the diversity of the bacte- clustered using the unweighted pair group method with
rial community in the ileum and colon of mice exposed to arithmetic mean (UPGMA) algorithm and displayed in a
CS or air for 24 weeks. Additionally, we explored which heatmap (Fig. 3), which demonstrated that the expressed
species were sensitive to CS exposure and to what extent 16S rRNA level and therefore the activity of OTU010,
the composition of the mucus layer and inflammatory gene representing Lachnospiraceae sp., strongly increases in
expression is affected. proximal and distal colon (571.74% and 300.76% change,
respectively) in response to smoke exposure.
Results
The gut bacterial community shifts in response to Cigarette smoke exposure affects the specific mucin
cigarette smoke expression pattern, but not the main mucin classes in
ileum and distal colon
To determine the response of the host microbiome to CS,
the taxonomical community structure of the microbiome in The detected bacterial changes prompted us to investigate
ileal, caecal and distal colonic samples of 12 smoke- whether the mucin composition of the mucus layer is

© 2015 Society for Applied Microbiology and John Wiley & Sons Ltd, Environmental Microbiology
Cigarette smoke induces shifts in the murine gut 3

altered upon CS exposure. Mucins in the gut can be


subdivided in two main classes: secretable mucins and cell
surface mucins (Hoebler et al., 2006). We examined the
amount of goblet cells, which secrete mucins into the gut
lumen, in Alcian Blue (AB)/periodic acid Schiff (PAS)-staine
ileum and distal colon sections of 10 smoke-exposed of 10
air-exposed mice. For each group, ileum and distal colon
sections were scored. No significant differences were
detected. Additionally, tissue sections of the same gut
samples were stained with high iron diamine (HID) and AB
to differentiate between sulfo- and sialomucins. Again, no
significant differences were detected.
Furthermore, we examined the mRNA expression
pattern of MUC1, MUC2, MUC3 and MUC4. MUC2 is
stored in goblet cells and typically secreted, whereas
MUC1, MUC3 and MUC4 are membrane-bound cell
surface mucins, which are anchored in the glycocalyx,
located at the apical surface of the enterocyte (Hoebler
et al., 2006). Expression analysis in both the smoke-
and air-exposed group (n = 6 in each group) showed a
shift in the mucin expression pattern in response to CS
in ileum and distal colon (Fig. 4A and B). In the ileum,
Muc2 and Muc3 expression was upregulated in
response to CS, while Muc1 and Muc4 expression was
not significantly altered. Muc2 and Muc3 expression
in air-exposed ileum was much lower than in air-
exposed distal colon, which is not surprising as bacterial
load and therefore bacteria-induced mucus in the ileum
is much lower (Johansson, 2014). Interestingly, being
induced by CS exposure, Muc2 and Muc3 expression in
the ileum resembles a more colon-like expression sig-
nature. In the distal colon, only Muc4 expression is
increased in smoke-exposed compared with air-exposed
animals.

Cigarette smoke exposure affects inflammatory gene


expression, but not the tight junction gene expression
The gut microbiota play an important role in the immuno-
logical response of the gut (Round et al., 2010). Interest-
ingly, we have previously shown that 24 weeks of CS
exposure affects the ileal immunological response by trig-
gering the attraction of immune cells to the PP, the
immune inductive sites of the gut (Verschuere et al.,
2011). Therefore, we investigated the effect of CS expo-
sure on inflammatory gene expression in the immune
effector regions of the gut (n = 6 in each group). We
observed a significant increase of Cxcl2, a decrease of
Ifn-γ and a nominal decrease of Il-6 in the ileum (Fig. 4C),
Fig. 1. Agglomerative nesting clustering dendrograms using the
abundance-based Jaccard distance measure based on the DGGE
whereas Tnf-α, Il-10, Nfκb and Tgf-β remained unaltered
data with average linkage (UPGMA), normalized in BIONUMERICS (Fig. S2A). In the proximal colon, Il-6 was increased and
5.10 software. (A) Clustering dendrogram for ileum, (B) clustering Tgf-β was decreased (Fig. 4D), while Il-10, Nfκb, Ifn-γ,
dendrogram for caecum and (C) clustering dendrogram for distal
colon. SM, smoking; NSM, non-smoking; DC, distal colon; IL,
Cxcl2 and Il-1β did not change (Fig. S2B). However, in the
ileum; CC, caecum. distal colon, inflammatory gene expression remained

© 2015 Society for Applied Microbiology and John Wiley & Sons Ltd, Environmental Microbiology
4 L. Allais et al.

Fig. 2. Individuals plot, based on the Illumina sequencing data, showing a clearly distinct ordination between the proximal and distal colonic
samples of air- and smoke-exposed mice. No distinct ordination could be detected in the ileum. This confirms the DGGE data. SM, smoking;
NSM, non-smoking; DC, distal colon; IL, ileum; PC, proximal colon.

unchanged (Fig. S2C and D). In addition, we analysed the increase in the activity of Lachnospiraceae sp. in the
effect of chronic CS exposure on tight junction gene colon. Furthermore, in the ileum, mRNA expression of
expression. It is known that loss of gut epithelial integrity Muc2 and Muc3 significantly increased after cigarette
and barrier function are predisposing factors in IBD, for smoke exposure, whereas in the colon, an increased
which CS is a major risk factor, and local inflammation expression of Muc4 was observed. In addition, inflamma-
impairs the barrier function of gut epithelium (Wang et al., tory gene expression is affected in the ileum and proximal
2012). We did not observe any changes in the tight junc- colon, with an increase of Cxcl2 and a decrease of Ifn-γ in
tion genes β-catenin, occludin, claudin-4 and E-cadherin the ileum, and an increase in Il-6 and a decrease in Tgf-β
(Fig. S3). in the proximal colon. However, no changes in inflamma-
tory gene expression occurred in the distal colon. Also, no
changes in tight junction gene expression were detected
Discussion
in response to CS exposure.
In this study, we demonstrate that chronic exposure to First, we studied the influence of CS exposure on the
cigarette smoke significantly affects the mucosa- bacterial community structure and activity in the mucosa
associated bacterial community, mucins and inflammatory of different parts of the gut (ileum, caecum and distal
gene expression in the gut of conventional mice. We colon). Therefore, we applied two complementary
revealed a shift of the community structure and an methods based on overlapping regions of the 16S rDNA

© 2015 Society for Applied Microbiology and John Wiley & Sons Ltd, Environmental Microbiology
Cigarette smoke induces shifts in the murine gut 5

Fig. 3. Heatmap with UPGMA clustering using the sparse patrimonial least squares discriminant analysis (sPLS-DA). The 40 most active
OTUs are displayed (ranked by sPLS-DA Xw score [X within], Liquet et al., 2012). The activity of OTU0010 (=Lachnospiraceae sp. according
to RDP9) is increased in the proximal and distal colon (571.74% and 300.76% change, respectively) of CS-exposed mice.

of bacteria: DGGE and Illumina sequencing. DGGE segmented filamentous bacteria (Wang et al., 2012).
analysis is a quick profiling technique and showed a shift There is a constant turnover of the microbiota in the gut,
in the bacterial community structure in response to CS in rendering its composition vulnerable to short-term expo-
the caecum and distal colon, likewise paralleling a shift in sure to environmental hazards. In our study, we assume
bacterial activity shown with Illumina sequencing. To date, that 24 weeks of CS exposure induces a stable microbial
the effect of a subacute CS exposure of 4 weeks has been community, thereby modelling long-term cigarette smoke-
investigated in a rat model showing a decrease of induced changes in the gut microbiota.
Bifidobacterium sp. in the caecum in response to CS Second, we explored which bacterial species might be
(Tomoda et al., 2011). In a mouse model, 8 weeks of CS susceptible to CS exposure. Analysis of our Illumina
exposure induced an increase in Clostridium sp. and a sequencing data using the sPLS-DA allows detecting
decrease in the Firmicutes, Enterobacteriaceae sp. and changes in the activity of species groups through analysis

© 2015 Society for Applied Microbiology and John Wiley & Sons Ltd, Environmental Microbiology
6 L. Allais et al.

Fig. 4. (A) mRNA expression of mucins in the ileum after air and smoke exposure, relative to the expression of two reference genes
[glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and hydroxymethylbilane synthase (HMBS)]. Expression of Muc2 and Muc3
significantly increases after smoke exposure (P = 0.039 and P = 0.0321 respectively). (B) Expression Muc4 significantly increased after smoke
exposure (P = 0.0274). mRNA expression of Muc4 increases in the distal colon after smoke exposure (P = 0.0274) relative to the expression
of two reference genes [glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and hydroxymethylbilane synthase (HMBS)]. (C) Expression of
CXCL2 significantly increases and IFN-γ significantly decreases after smoke exposure (P = 0.0186 and P = 0.0463, respectively), while IL-6
shows a nominal decrease (P = 0.068) and IL-1β is not significantly altered (P = 0.0983). (D) mRNA expression of inflammatory genes in the
proximal colon after air and smoke exposure. Expression of IL-6 significantly increases and TGF-β significantly decreases after smoke
exposure (P = 0.0209 and P = 0.0388 respectively). P-values lower than 0.05 were considered significant. Data are represented as
mean ± SEM. n = 6 in each group. *P < 0.05.

of cDNA. This analysis showed an increase in the activity Clostridium XIVa cluster, which includes Lachnospiraceae
of Lachnospiraceae sp. in the proximal and distal colon of sp., strengthens the tight junctions and thereby the epi-
smoke-exposed mice, but not in the ileum. It has been thelial barrier through the production of butyrate (Ma
shown previously that Lachnospiraceae sp. induces et al., 2012). Another study reported an increase in tight
monocyte/macrophage recruitment into the inflamed junction proteins claudin-3 and ZO-2 after 8 weeks of CS
colon, and thus triggers colitis upon disruption of the exposure (Wang et al., 2012). According to our own
colonic epithelial cell barrier function (Nakanishi et al., data, a long-term cigarette smoke exposure of 24
2014). This is in agreement with our findings that chronic weeks no longer affects tight junction gene expression.
smoke exposure affects the gut immune system of In the faeces of healthy human subjects, an increase
healthy mice by inducing the recruitment of immune cells in Firmicutes and Actinobacteria and a decrease in
to the PPs and apoptosis and autophagy in the FAE Bacteroidetes and Proteobacteria have been reported
(Verschuere et al., 2011; 2012). In this way, CS exposure after smoking cessation (Biedermann et al., 2013). A
may contribute to an increased risk for the creation of an study by Wang and colleagues has shown that 8 weeks
inflammatory environment, and combined with other of CS exposure increased Clostridium sp., and decreased
factors eventually may initiate IBD. Surprisingly, we could the Firmicutes (Lactococcus sp. and Ruminococcus sp.),
not detect changes in tight junction gene expression, Enterobacteriaceae sp. and segmented filamentous bac-
although it has previously been reported that the teria in the caecum (Wang et al., 2012). Although this

© 2015 Society for Applied Microbiology and John Wiley & Sons Ltd, Environmental Microbiology
Cigarette smoke induces shifts in the murine gut 7

study suggests that Firmicutes, to which the immune responses. This kind of immune inductive sites
Lachnospiraceae sp. belongs, decreases due to smoking, are much less frequent in the colon, which makes it mainly
many other species groups determine changes in the an effector site (lamina propria and surface epithelia) of
phylum besides the Lachnospiraceae sp. Also, we ileal-generated immune responses (Brandtzaeg and
exposed the mice to 24 weeks of CS, which is a long-term Pabst, 2004). Wang et al. showed an inhibition of the
exposure and sheds light on long-term changes in the NFκB pathway, but no changes in the expression of
microbiota. In future experiments, it might be of interest to colonic Tnf-α and Il-6 after 8 weeks of CS exposure
monitor microbiota changes over time in response to CS (Wang et al., 2012). In our study, we did not observe any
exposure. changes in Nfκb expression; however, we showed that
A third important finding in this study was that smoking colonic Il-6 was increased. Again, this might be due to the
is able to induce changes in mucin expression. Previous long-term (24 weeks) effect of CS exposure in our own
studies have shown that smoking causes mucin experimental set-up.
hypersecretion in the lung of patients suffering from Our data show that chronic CS exposure has a pro-
COPD (Di et al., 2012; Kim, 2012; Yu et al., 2012). The found effect on the gut microbiota, the associated mucin
major mucins in the intestine are MUC2, a secretable composition and cytokine/chemokine production in mice,
mucin mainly produced and delivered to the lumen by which is likely to occur in human as well. Although the gut
goblet cells, and MUC3, a cell surface mucin (Shirazi microbial similarity between mice and humans at the
et al., 2000; Hoebler et al., 2006; Linden et al., 2008). phylum level is remarkable, many differences exist at the
MUC4 is a cell surface mucin that acts purely as an species level (Dethlefsen et al., 2007). Both in human and
anti-adhesive (Hattrup and Gendler, 2008). Here, we C57BL6/J mice, the two most abundant phyla are the
showed that ileal expression of Muc2 and Muc3 increased Firmicutes and the Bacteroidetes; however, 85% of the
significantly upon CS exposure. In the distal colon, CS murine sequences represent species that have not been
caused an increase of Muc4 mRNA. Interestingly, quan- detected in humans (Ley et al., 2005). Therefore, extrapo-
titative changes in mucin secretion also occur in human lation from mouse to man might be challenging due to the
IBD (Boltin et al., 2013). The composition of the protective heterogeneity of the human population. Nevertheless, the
mucin layer plays an important role in inhibiting direct use of a murine model offers the advantage of housing
contact between the host and potentially offending bacte- in standardized conditions, minimizing environmental
ria, and reducing the exposure time by increasing bacte- influences.
rial transit. The changes in mucin expression that we Interestingly, our findings show that chronic CS expo-
observed might either be a direct effect of the exposure to sure affects the immune system in the ileum while it exerts
the chemical components of CS or induced as a protec- a more pronounced effect on the microbiota in the colon.
tion mechanism to counteract local physiological changes This may be partly explained by the much lower bacterial
in the gut. In addition, a shifting composition of mucolytic load in the ileum compared with the colon. It is known that
organisms from A. muciniphila to other mucolytic species, smoking worsens CD, while UC is rather a disease of
such as Ruminococcus gnavus, occurs in pathological ex-smokers (Ananthakrishnan, 2015). Inflammation in CD
conditions, such as IBD, and may therefore be a suitable mainly originates in the terminal ileum, whereas UC is
biomarker for mucosal integrity (Png et al., 2010a; Berry initiated in the colon. Taking all these facts together, it
and Reinisch, 2013). However, to date, the mucin degra- might be that CS affects the ileum and colon via
dation specificity of different Akkermansia species distinct mechanisms, resulting in an even more distinct
remains unclear. outcome for inflammatory diseases as CD and UC. Given
In addition, we found that inflammatory gene expres- our data, we speculate that cigarette smoke exposure
sion is altered in the ileum and proximal colon in response tends to affect the ileal immune system, which may give
to CS exposure, but not in the distal colon. We demon- rise to CD-related inflammation. In contrast, cigarette
strated that ileal Cxcl2, a critical effector for neutrophil smoke exposure targets the colonic microbiome rather
trafficking, is increased by chronic CS exposure. In con- than its immune system. Moreover, the activity of
trast, ileal Ifn-ɣ, involved in macrophage activation, is Lachnospiraceae sp., a butyrate producer, is increased in
decreased. Furthermore, we showed that proximal response to CS exposure, which may contribute to the
colonic Il-6 was increased. Proximal colonic Tgf-β, being smoke-induced protective effect on UC-related colonic
involved in tempering the immune response, was inflammation.
decreased. Surprisingly, expression of inflammatory Today, the link between host (patho)physiology and
factors was not affected in the distal colon, although the gut microbiome is being increasingly recognized
smoke-induced changes in the microbiota occur in the (Arumugam et al., 2011; Elinav et al., 2011). In-depth
colon. The ileum contains important immune inductive knowledge of the human bacterial ecosystem in relation to
sites, such as the PPs, taking up antigens and generating disease might pave the way for the generation of

© 2015 Society for Applied Microbiology and John Wiley & Sons Ltd, Environmental Microbiology
8 L. Allais et al.

customized therapeutics and probiotics. We hypothesize incubation in 1% aqueous periodic acid for 5 min. Slides
that the induction of dysbiosis and the alteration of epi- were then washed in distilled water and immersed in
thelial mucus and cytokine/chemokine profiles by chronic Schiff’s reagent for 15 min. In case of the HID/AB
staining method, slides were treated with diamine solu-
cigarette smoke exposure modulate the risk for the devel-
tion (N,N-dimethyl-meta-phenylenediamine-dihydrochloride;
opment of inflammation-related disorders, such as IBD. N,N-dimethyl-para-phenylene-diamine-dihydrochloride; ferric
Further work in this field may contribute to the adaptation chloride 60% solution and distilled water) for 24 h at room
of treatment strategies for IBD depending on smoking temperature. After incubation, slides were washed and
behaviour. Future research exposing colitic mice to counterstained in 1% AB solution for 5 min. Finally, sections
chronic smoke exposure will be necessary to be able to from both staining methods were dehydrated in absolute
attribute a specific role of dysbiosis and mucus changes in alcohol, cleared in three changes of xylene and mounted.
the development of disease.
Scoring of microscopic analysis
Experimental procedures Histological and goblet cell assessment was performed using
light microscopy. For 10 AB/PAS-stained sections per group,
Animals
positively stained goblet cells were counted in two regions of
Male C57BL/6 wild-type mice were purchased from Charles 10 aligning longitudinal crypts. For 10 HID/AB-stained sec-
River Laboratories and were co-housed in the same animal tions per group, sulfomucin-positive stained goblet cells were
facility before the start of the experiment to homogenize gut counted in two regions of 10 aligning longitudinal crypts.
microbiota between groups. All mice were 8–9 weeks old at Histological assessments were carried out in a blinded
the start of the smoke exposure. Mice were housed in a fashion.
temperature-controlled room with a 12 h light and 12 h dark-
ness cycle and were given standardized food (Carfil,
Quantitative real-time polymerase chain reaction
Turnhout, Belgium) and water ad libitum. The mice were
divided in two cages per experimental group (n = 12) with six (qRT-PCR)
mice per cage. The local ethics committee for animal experi- RNA from ileum and distal colon (both were taken from 10
mentation of the Faculty of Medicine and Health Sciences smoke- and 10 air-exposed mice) was extracted using the
(Ghent, Belgium) approved all experiments (ECD 27/07). Qiagen miRNeasy Mini Kit (Qiagen, Hilden, Germany). Sub-
sequently, cDNA was synthesized by reverse transcription
Cigarette smoke exposure using the iScript™ cDNA synthesis kit (Bio-Rad Laboratories,
Nazareth, Belgium) following the manufacturer’s instructions.
Mice were exposed to mainstream cigarette smoke, as Expression of target genes Muc1, Muc2, Muc3, Muc4, Cxcl2,
described previously (D’Hulst et al., 2005). Briefly, groups of I1-1β, I1-6, Ifn-γ, Tnf-α, Il-10, Nfκb, Tgfb1, Ctnb1, Cldb-4,
12 mice were exposed to the tobacco smoke of 20 cigarettes Ocln and Cdh1, and reference genes glyceraldehyde-3-
(Reference Cigarette 3R4F without filter; University of Ken- phosphate dehydrogenase (GAPDH) and hydroxymethyl-
tucky, Lexington, KY, USA). The exposure to five cigarettes bilane synthase (HMBS) (Table 1), was analysed by
was applied for 7 min, with 30 min smoke-free intervals, qRT-PCR using the SensiMix™ SYBR No-ROX Kit (Bioline,
which was repeated four times a day, 5 days per week for 24 London, UK). The qRT-PCR was performed on a
weeks (chronic smoke exposure). An optimal smoke : air LightCycler480 detection system (Roche Diagnostics) with
ratio of 1:6 was obtained. The control groups were exposed to the following cycling conditions: 10 min incubation at 95°C,
air. Carboxyhaemoglobin in serum of smoke-exposed mice 45 cycles of 95°C for 10 s and 60°C for 1 min. Melting curve
reached a non-toxic level of 8.7 ± 0.31% (compared with analysis confirmed primer specificity. The PCR efficiency of
0.65 ± 0.25% in air-exposed mice), which is similar to each primer pair was calculated using a standard curve from
carboxyhaemoglobin blood concentrations of human reference cDNA. The amplification efficiency was determined
smokers (Macdonald et al., 2004). using the formula 10−1/SLOPE − 1.

Staining methods DGGE


Paraffin-embedded tissue sections of 4 μm taken from The ileal and colonic samples of 12 smoke- and 12 air-
ileum and distal colon were de-waxed and re-hydrated. The exposed mice were obtained snap-frozen and stored at
sections were stained with either AB/PAS, in which AB −80°C. The 16S rRNA genes for all bacteria were amplified by
stains blue for secretable mucins and PAS stains purple- PCR adding a GC-clamp of 40 bp. The DGGE was performed
pink for cell surface mucins, or with HID/AB, in which using the INGENYPHORU System (Ingeny International BV,
sulfated mucins are stained dark brown by HID and The Netherlands), after which PCR fragments were loaded
sialylated mucins are stained blue by AB. In case of the onto 8% (w/v) polyacrylamide gels containing 40–60% dena-
AB/PAS staining method, slides were stained using the PAS turing gradients (Muyzer et al., 1993; Ovreas et al., 1997). To
Staining Kit and AB for PAS Staining Kit on the automated process and compare the different gels, an in-house devel-
Ventana system (Roche Diagnostics, Vilvoorde, Belgium). oped marker of different PCR fragments was loaded on each
Slides were immersed in the AB solution for 5 min before gel (Boon et al., 2002).

© 2015 Society for Applied Microbiology and John Wiley & Sons Ltd, Environmental Microbiology
Cigarette smoke induces shifts in the murine gut 9
Table 1. Mouse primer sequences qRT-PCR.

Gene symbol Accession number Forward primer (5′-3′) Reverse primer (3′-5′) Effic R2

Hmbs NM_001110251 AAGGGCTTTTCTGAGGCACC AGTTGCCCATCTTTCATCACTG 99 0.99


Gapdh NM_008084 CATGGCCTTCCGTGTTCCTA GCGGCACGTCAGATCCA 106 0.9986
Muc1* NM_013605 GCAGTCCTCAGTGGCACCTC CACCGTGGGCTACTGGAGAG 105 0.99
Muc2 NM_023566 CAAGGGCTCGGAACTCCAG CCAGGGAATCGGTAGACATCG 97 0.92
Muc3* XM_355711 CGTGGTCAACTGCGAGAATGG CGGCTCTATCTCTACGCTCTCC 104 0.94
Muc4* AF520422 CAGCAGCCAGTGGGGACAG CTCAGACACAGCCAGGGAACTC 110 0.96
Cxcl2 NM_009140 GCGCCCAGACAGAAGTCATAG AGCCTTGCCTTTGTTCAGTATC 89,2 0.99
Il-1β NM_000576 CACGATGCACCTGTACGATCA GTTGCTCCATATCCTGTCCCT 97 0.9987
Tnf-α NM_013693 ATGAGCACTGAAAGCATGATCC GAGGGCTGATTAGAGAGAGGTC 92 0.9761
Il-10 NM_010548 GGTGTCCTTTCAATTGCTCTCAT TCACAACTCTCTTAGGAGCTCTGAACT 90 0.9974
Nfκb NM_023526 GAAGGGCGTGTTTGACAAGGA GCATCCCGAACAAGAGACAGAAT 93 0.9957
Tgfb1 NM_011577 CTCCCGTGGCTTCTAGTGC GCCTTAGTTTGGACAGGATCTG 87 0.9828
Ctnb1 NM_001165902.1 TCACATTTGAGAAGCGATCCTAC TCCAGCTCGGATTCCATGAAC 89 0.9971
Cldn-4 NM_009903.2 AGCCTTCCAGGTCCTCAACT AGCAGCGAGTAGAAG 195 0.9975
Ocln NM_008756.2 ACAGACTACACAACTGGCGG TCATCAGCAGCAGCCATGTA 101 0.9991
Cdh1 NM_009864.2 TTACTGCCCCCAGAGGATGA TGCAACGTCGTTACGAGTCA 195 0,9957
Ifng NM_008337 GCCAAGCGGCTGACTGA TCAGTGAAGTAAAGGTACAAGCTACAATCT 197 0,9635

*Primer sequences were adapted from Hoebler and colleagues (2006).

The normalization and analysis of DGGE gel patterns were with subsequent classification using the RDP release 9 into
done with the BIONUMERICS software 5.10 (Applied Maths, 904 classified OTUs at the 85% bootstrap cut-off.
Sint-Martens-Latem, Belgium). During this processing, the
different lanes were defined, background was subtracted, Statistical analysis
differences in the intensity of the lanes were compensated
during normalization, and bands and band classes were Reported gene expression values are expressed as
detected. mean ± standard error of the mean (SEM) and error bars
depict the SEM. Statistical analysis was performed by SPSS
21 Software (SPSS 21, Chicago, IL, USA) using Student’s
Illumina sequencing t-test for normally distributed populations, and Mann–
Whitney U-test for populations where normal distribution was
Illumina sequencing was performed on amplicons from cDNA not accomplished. A P-value of less than 0.05 was consid-
extracted from snap-frozen gut tissue samples of six smoke- ered significant.
and six air-exposed mice. Sequencing was performed on Clustering of DGGE data was done based on the
cDNA, which was synthesized from extracted RNA. Using abundance-based Jaccard index (with fuzzy logic) or the Yue
random primers for the cDNA synthesis, 16S rRNA was also and Claytons theta index, and the UPGMA. Similarities and
translated into cDNA, which made the samples suitable for abundances were extracted from the software, and statistical
16S sequencing. The MicroRneasy Mini Kit (Qiagen) was analysis was performed using R version 2.15.1. P-values
used for RNA extraction, for which the manufacturer ensures were calculated using permutation-based MANOVA
high-purity RNA. When measuring samples using a (vegan::Adonis).
NanoDrop after RNA extraction, high concentrations (500– For β-diversity statistics on the Illumina sequencing data,
1000 ng μl−1) were obtained and 260/280 nm ratios were all we applied the sPLS-DA method with two-factor design and
close to 2, which indicates good RNA quality. Quality and ad hoc optimization from the MixOmics package, considering
concentrations were similar for samples of different gut parts. ileum, caecum and distal colon originating from the same
The sequencing was performed by LGC Genomics (Berlin, mouse as paired samples. The sPLS-DA is a combined mul-
Germany). The raw flowgrams were processed and analysed tilevel and multivariate method and distinguishes between
in an in-house MOTHUR (Schloss et al., 2009) (http:// within-sample and between-sample variation, after which the
www.mothur.org, version 1.26.0) and R (http://www.r discriminant analysis is executed on the between-sample
-project.org/ version 2.15.1)/Sweave pipeline. Sequencing variation. We applied this method to identify the abundance of
error was reduced using the MOTHUR implementation of the specific OTUs in each separate intestinal region. An ad hoc
SeqNoise algorithm (Quince et al., 2011). Alignment with the estimation procedure is used to include only the statistically
SILVA 16S reference (Pruesse et al., 2007) was performed relevant OTUs. Applying tuning parameter two, the relevant
and sequences were trimmed to overlap in the same align- OTUs were selected in order to model the data into three
ment space. Chimeric sequences were removed using sPLS-DA components (Liquet et al., 2012).
Uchime (Edgar et al., 2011). A Bayesian classifier was used
with the ribosomal database project (RDP) training set (Cole Acknowledgements
et al., 2009) of RDP release 9 (http://rdp.cme.msu.edu/) to
classify the sequences. Unique sequences were then clus- We thank the FLAMES statistical centre for advice in our
tered into 904 OTUs with a 97% sequence identity threshold, statistical analyses. We are grateful to Dorothea van

© 2015 Society for Applied Microbiology and John Wiley & Sons Ltd, Environmental Microbiology
10 L. Allais et al.
Limbergen, Ran Rumes and Lynn Supply for the support with Dethlefsen, L., McFall-Ngai, M., and Relman, D.A. (2007) An
the animal experiments and the processing of the samples, ecological and evolutionary perspective on human-microbe
and Eliane Castrique, Christelle Snauwaert, Marie-Rose mutualism and disease. Nature 449: 811–818.
Mouton, Katleen de Saedeleer, Anouk Goethals, Ann D’Hulst, A.I., Vermaelen, K.Y., Brusselle, G.G., Joos, G.F.,
Neesen, Indra de Borle, Evelyn Spruyt and Greet Barbier for and Pauwels, R.A. (2005) Time course of cigarette smoke-
the excellent technical support with the animal experiments. induced pulmonary inflammation in mice. Eur Respir J 26:
We thank Tim Lacoere, Siska Maertens and Lois Maignien 204–213.
from LabMET for the support with the DGGE and Illumina Di, Y.P., Zhao, J., and Harper, R. (2012) Cigarette smoke
sequencing. This work was supported by the Special induces MUC5AC protein expression through the activa-
Research Fund of Ghent University (01D41012), the Con- tion of Sp1. J Biol Chem 287: 27948–27958.
certed Research Actions of Ghent University (BOF09/GOA/ Edgar, R.C., Haas, B.J., Clemente, J.C., Quince, C., and
005, BOF10/GOA/021 and BOF12/GOA/008) and the Knight, R. (2011) UCHIME improves sensitivity and speed
Interuniversity Attraction Poles Program (IUAP, P7/30). of chimera detection. Bioinformatics 27: 2194–2200.
Liesbeth Allais is supported by a doctoral grant from the Elinav, E., Strowig, T., Kau, A.L., Henao-Mejia, J., Thaiss,
Special Research Fund of Ghent University (01D41012). C.A., Booth, C.J., et al. (2011) NLRP6 inflammasome regu-
Frederiek-Maarten Kerckhof is supported by a doctoral grant lates colonic microbial ecology and risk for colitis. Cell 145:
from the Concerted Research Actions of Ghent University 745–757.
(BOF09/GOA/005). Ken R. Bracke is a postdoctoral Erickson, A.R., Cantarel, B.L., Lamendella, R., Darzi, Y.,
researcher of the Fund for Scientific Research Flanders Mongodin, E.F., Pan, C., et al. (2012) Integrated
(FWO Vlaanderen). No author has an ethical or financial metagenomics/metaproteomics reveals human host-
conflict of interest. microbiota signatures of Crohn’s disease. PLoS ONE 7:
e49138.
References Frank, D.N., St Amand, A.L., Feldman, R.A., Boedeker, E.C.,
Harpaz, N., and Pace, N.R. (2007) Molecular-phylogenetic
Ananthakrishnan, A.N. (2013) Environmental risk factors for characterization of microbial community imbalances in
inflammatory bowel disease. Gastroenterol Hepatol (N Y) human inflammatory bowel diseases. Proc Natl Acad Sci
9: 367–374. USA 104: 13780–13785.
Ananthakrishnan, A.N. (2015) Environmental risk factors for Garmendia, J., Morey, P., and Bengoechea, J.A. (2012)
inflammatory bowel diseases: a review. Dig Dis Sci 60: Impact of cigarette smoke exposure on host-bacterial
290–298. pathogen interactions. Eur Respir J 39: 467–477.
Arcavi, L., and Benowitz, N.L. (2004) Cigarette smoking and Hattrup, C.L., and Gendler, S.J. (2008) Structure and function
infection. Arch Intern Med 164: 2206–2216. of the cell surface (tethered) mucins. Annu Rev Physiol 70:
Arumugam, M., Raes, J., Pelletier, E., Le Paslier, D., 431–457.
Yamada, T., Mende, D.R., et al. (2011) Enterotypes of the Hoebler, C., Gaudier, E., De Coppet, P., Rival, M., and
human gut microbiome. Nature 473: 174–180. Cherbut, C. (2006) MUC genes are differently expressed
Berry, D., and Reinisch, W. (2013) Intestinal microbiota: a during onset and maintenance of inflammation in dextran
source of novel biomarkers in inflammatory bowel dis- sodium sulfate-treated mice. Dig Dis Sci 51: 381–389.
eases? Best Pract Res Clin Gastroenterol 27: 47–58. Hovde, O., and Moum, B.A. (2012) Epidemiology and clinical
Biedermann, L., Zeitz, J., Mwinyi, J., Sutter-Minder, E., course of Crohn’s disease: results from observational
Rehman, A., Ott, S.J., et al. (2013) Smoking cessation studies. World J Gastroenterol 18: 1723–1731.
induces profound changes in the composition of the intes- Jin, Y., Lin, Y., Lin, L., and Zheng, C. (2012) IL-17/IFN-gamma
tinal microbiota in humans. PLoS ONE 8: e59260. interactions regulate intestinal inflammation in TNBS-
Boltin, D., Perets, T.T., Vilkin, A., and Niv, Y. (2013) Mucin induced acute colitis. J Interferon Cytokine Res 32: 548–
function in inflammatory bowel disease: an update. J Clin 556.
Gastroenterol 47: 106–111. Johansson, M.E. (2014) Mucus layers in inflammatory bowel
Boon, N., De Windt, W., Verstraete, W., and Top, E.M. (2002) disease. Inflamm Bowel Dis 20: 2124–2131.
Evaluation of nested PCR-DGGE (denaturing gradient gel Khan, W.I., and Collins, S.M. (2006) Gut motor function:
electrophoresis) with group-specific 16S rRNA primers for immunological control in enteric infection and inflamma-
the analysis of bacterial communities from different waste- tion. Clin Exp Immunol 143: 389–397.
water treatment plants. FEMS Microbiol Ecol 39: 101–112. Kim, K.C. (2012) Role of epithelial mucins during airway
Brandtzaeg, P., and Pabst, R. (2004) Let’s go mucosal: com- infection. Pulm Pharmacol Ther 25: 415–419.
munication on slippery ground. Trends Immunol 25: 570– Lakatos, P.L. (2006) Recent trends in the epidemiology of
577. inflammatory bowel diseases: up or down? World J
Carbonnel, F., Jantchou, P., Monnet, E., and Cosnes, J. Gastroenterol 12: 6102–6108.
(2009) Environmental risk factors in Crohn’s disease and Ley, R.E., Backhed, F., Turnbaugh, P., Lozupone, C.A.,
ulcerative colitis: an update. Gastroenterol Clin Biol 33 Knight, R.D., and Gordon, J.I. (2005) Obesity alters gut
(Suppl. 3): S145–S157. microbial ecology. Proc Natl Acad Sci USA 102: 11070–
Cole, J.R., Wang, Q., Cardenas, E., Fish, J., Chai, B., Farris, 11075.
R.J., et al. (2009) The Ribosomal Database Project: Linden, S.K., Florin, T.H., and McGuckin, M.A. (2008) Mucin
improved alignments and new tools for rRNA analysis. dynamics in intestinal bacterial infection. PLoS ONE 3:
Nucleic Acids Res 37: D141–D145. e3952.

© 2015 Society for Applied Microbiology and John Wiley & Sons Ltd, Environmental Microbiology
Cigarette smoke induces shifts in the murine gut 11
Liquet, B., Le Cao, K.A., Hocini, H., and Thiebaut, R. (2012) flagellin elicits widespread innate immune defense mecha-
A novel approach for biomarker selection and the integra- nisms, apoptotic signaling, and a sepsis-like systemic
tion of repeated measures experiments from two assays. inflammatory response in mice. Crit Care 14: R160.
BMC Bioinformatics 13: 325. Round, J.L., and Mazmanian, S.K. (2009) The gut microbiota
Loftus, E.V. (2004) Clinical epidemiology of inflammatory shapes intestinal immune responses during health and
bowel disease: incidence, prevalence, and environmental disease. Nat Rev Immunol 9: 313–323.
influences. Gastroenterology 126: 1504–1517. Round, J.L., O’Connell, R.M., and Mazmanian, S.K. (2010)
Ma, X., Fan, P.X., Li, L.S., Qiao, S.Y., Zhang, G.L., and Li, Coordination of tolerogenic immune responses by the com-
D.F. (2012) Butyrate promotes the recovering of intestinal mensal microbiota. J Autoimmun 34: J220–J225.
wound healing through its positive effect on the tight junc- Schloss, P.D., Westcott, S.L., Ryabin, T., Hall, J.R.,
tions. J Anim Sci 90 (Suppl. 4): 266–268. Hartmann, M., Hollister, E.B., et al. (2009) Introducing
Macdonald, G., Kondor, N., Yousefi, V., Green, A., Wong, F., Mothur: open-source, platform-independent, community-
and Aquino-Parsons, C. (2004) Reduction of supported software for describing and comparing microbial
carboxyhaemoglobin levels in the venous blood of ciga- communities. Appl Environ Microbiol 75: 7537–7541.
rette smokers following the administration of carbogen. Shirazi, T., Longman, R.J., Corfield, A.P., and Probert, C.S.
Radiother Oncol 73: 367–371. (2000) Mucins and inflammatory bowel disease. Postgrad
Manichanh, C., Rigottier-Gois, L., Bonnaud, E., Gloux, K., Med J 76: 473–478.
Pelletier, E., Frangeul, L., et al. (2006) Reduced diversity of Stecher, B., Maier, L., and Hardt, W.D. (2013) ‘Blooming’ in
faecal microbiota in Crohn’s disease revealed by a the gut: how dysbiosis might contribute to pathogen evo-
metagenomic approach. Gut 55: 205–211. lution. Nat Rev Microbiol 11: 277–284.
Muyzer, G., Dewaal, E.C., and Uitterlinden, A.G. (1993) Pro- Strober, W., Fuss, I., and Mannon, P. (2007) The fundamental
filing of complex microbial-populations by denaturing gra- basis of inflammatory bowel disease. J Clin Invest 117:
dient gel-electrophoresis analysis of polymerase chain 514–521.
reaction-amplified genes-coding for 16S ribosomal-RNA. Tomoda, K., Kubo, K., Asahara, T., Andoh, A., Nomoto, K.,
Appl Environ Microbiol 59: 695–700. Nishii, Y., et al. (2011) Cigarette smoke decreases organic
Nakanishi, Y., Sato, T., and Ohteki, T. (2014) Commensal acids levels and population of bifidobacterium in the
Gram-positive bacteria initiates colitis by inducing caecum of rats. J Toxicol Sci 36: 261–266.
monocyte/macrophage mobilization. Mucosal Immunol 8: Verschuere, S., Bracke, K.R., Demoor, T., Plantinga, M.,
152–160. Verbrugghe, P., Ferdinande, L., et al. (2011) Cigarette
Ovreas, L., Forney, L., Daae, F.L., and Torsvik, V. (1997) smoking alters epithelial apoptosis and immune composi-
Distribution of bacterioplankton in meromictic Lake tion in murine GALT. Lab Invest 91: 1056–1067.
Saelenvannet, as determined by denaturing gradient gel Verschuere, S., Allais, L., Bracke, K.R., Lippens, S.,
electrophoresis of PCR-amplified gene fragments coding De Smet, R., Vandenabeele, P., et al. (2012) Cigarette
for 16S rRNA. Appl Environ Microbiol 63: 3367–3373. smoke and the terminal ileum: increased autophagy in
Persson, P.G., Ahlbom, A., and Hellers, G. (1990) Inflamma- murine follicle-associated epithelium and Peyer’s patches.
tory bowel disease and tobacco smoke–a case-control Histochem Cell Biol 137: 293–301.
study. Gut 31: 1377–1381. Wang, H., Zhao, J.X., Hu, N., Ren, J., Du, M., and Zhu, M.J.
Png, C.W., Linden, S.K., Gilshenan, K.S., Zoetendal, E.G., (2012) Side-stream smoking reduces intestinal inflamma-
McSweeney, C.S., Sly, L.I., et al. (2010a) Mucolytic bacte- tion and increases expression of tight junction proteins.
ria with increased prevalence in IBD mucosa augment in World J Gastroenterol 18: 2180–2187.
vitro utilization of mucin by other bacteria. Am J Xavier, R.J., and Podolsky, D.K. (2007) Unravelling the
Gastroenterol 105: 2420–2428. pathogenesis of inflammatory bowel disease. Nature 448:
Png, C.W., Linden, S.K., Gilshenan, K.S., Zoetendal, E.G., 427–434.
McSweeney, C.S., Sly, L.I., et al. (2010b) Mucolytic bacte- Yu, H., Li, Q., Kolosov, V.P., Perelman, J.M., and Zhou, X.
ria with increased prevalence in IBD mucosa augment in (2012) Regulation of cigarette smoke-mediated mucin
vitro utilization of mucin by other bacteria. Am J expression by hypoxia-inducible factor-1alpha via epider-
Gastroenterol 105: 2420–2428. mal growth factor receptor-mediated signaling pathways.
Pruesse, E., Quast, C., Knittel, K., Fuchs, B.M., Ludwig, J Appl Toxicol 32: 282–292.
W.G., Peplies, J., and Glockner, F.O. (2007) SILVA: a com-
prehensive online resource for quality checked and aligned
ribosomal RNA sequence data compatible with ARB. Supporting information
Nucleic Acids Res 35: 7188–7196.
Additional Supporting Information may be found in the online
Quince, C., Lanzen, A., Davenport, R.J., and Turnbaugh, P.J.
version of this article at the publisher’s web-site:
(2011) Removing noise from pyrosequenced amplicons.
BMC Bioinformatics 12: 38. Fig. S1. Agglomerative nesting clustering dendrograms
Rogers, M.A., Greene, M.T., Saint, S., Chenoweth, C.E., using the abundance-based Yue and Clayton’s θ based on
Malani, P.N., Trivedi, I., and Aronoff, D.M. (2012) Higher the DGGE data with average linkage (UPGMA), normalized
rates of Clostridium difficile infection among smokers. in BIONUMERICS 5.10 software. (A) clustering dendrogram for
PLoS ONE 7: e42091. ileum, (B) clustering dendrogram for caecum and (C) cluster-
Rolli, J., Loukili, N., Levrand, S., Rosenblatt-Velin, N., ing dendrograms for distal colon. SM, smoking; NSM, non-
Rignault-Clerc, S., Waeber, B., et al. (2010) Bacterial smoking; DC, distal colon; IL, ileum; CC, caecum.

© 2015 Society for Applied Microbiology and John Wiley & Sons Ltd, Environmental Microbiology
12 L. Allais et al.
Fig. S2. (A) mRNA expression of TNF-α, IL-10, NFκB and exposure. Data are represented as mean ± SEM; n = 6 in
TGF-β in the ileum after air and smoke exposure, relative to each group.
the expression of two reference genes [glyceraldehyde-3- Fig. S3. mRNA expression of tight junction genes in the
phosphate dehydrogenase (GAPDH) and hydroxymethyl- ileum, proximal and distal colon after air and smoke expo-
bilane synthase (HMBS)]. (B) mRNA expression of IL-10, sure, relative to the expression of two reference genes
NFκB, IFN-γ, CXCL2, IL-1β and TNF-α in the proximal colon [glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and
after air and smoke exposure. (C) mRNA expression of hydroxymethylbilane synthase (HMBS)]. Data are repre-
CXCL2, IL-1β, IL-6 and IFN-γ in the distal colon after air and sented as mean ± SEM; n = 6 in each group.
smoke exposure. (D) mRNA expression of TNF-α, IL-10, Appendix. Raw Illumina sequencing data, containing OTU
NFκB and IFN-γ in the distal colon after air and smoke table and LefSe data.

© 2015 Society for Applied Microbiology and John Wiley & Sons Ltd, Environmental Microbiology

You might also like