ArticlePDF Available

Gut Microbiome Response to Sucralose and Its Potential Role in Inducing Liver Inflammation in Mice

Authors:

Abstract and Figures

Sucralose is the most widely used artificial sweetener, and its health effects have been highly debated over the years. In particular, previous studies have shown that sucralose consumption can alter the gut microbiota. The gut microbiome plays a key role in processes related to host health, such as food digestion and fermentation, immune cell development, and enteric nervous system regulation. Inflammation is one of the most common effects associated with gut microbiome dysbiosis, which has been linked to a series of human diseases, such as diabetes and obesity. The aim of this study was to investigate the structural and functional effects of sucralose on the gut microbiota and associated inflammation in the host. In this study, C57BL/6 male mice received sucralose in their drinking water for 6 months. The difference in gut microbiota composition and metabolites between control and sucralose-treated mice was determined using 16S rRNA gene sequencing, functional gene enrichment analysis and metabolomics. Inflammatory gene expression in tissues was analyzed by RT-PCR. Alterations in bacterial genera showed that sucralose affects the gut microbiota and its developmental dynamics. Enrichment of bacterial pro-inflammatory genes and disruption in fecal metabolites suggest that 6-month sucralose consumption at the human acceptable daily intake (ADI) may increase the risk of developing tissue inflammation by disrupting the gut microbiota, which is supported by elevated pro-inflammatory gene expression in the liver of sucralose-treated mice. Our results highlight the role of sucralose-gut microbiome interaction in regulating host health-related processes, particularly chronic inflammation.
Content may be subject to copyright.
ORIGINAL RESEARCH
published: 24 July 2017
doi: 10.3389/fphys.2017.00487
Frontiers in Physiology | www.frontiersin.org 1July 2017 | Volume 8 | Article 487
Edited by:
Zhaoping Li,
Ronald Reagan UCLA Medical Center,
United States
Reviewed by:
Andrew Patterson,
Pennsylvania State University,
United States
Benjamin Moeller,
University of California, Davis,
United States
*Correspondence:
Kun Lu
kunlu@unc.edu
Specialty section:
This article was submitted to
Gastrointestinal Sciences,
a section of the journal
Frontiers in Physiology
Received: 09 April 2017
Accepted: 26 June 2017
Published: 24 July 2017
Citation:
Bian X, Chi L, Gao B, Tu P, Ru H and
Lu K (2017) Gut Microbiome
Response to Sucralose and Its
Potential Role in Inducing Liver
Inflammation in Mice.
Front. Physiol. 8:487.
doi: 10.3389/fphys.2017.00487
Gut Microbiome Response to
Sucralose and Its Potential Role in
Inducing Liver Inflammation in Mice
Xiaoming Bian 1, Liang Chi 2, Bei Gao 1, Pengcheng Tu 2, Hongyu Ru 3and Kun Lu 2*
1Department of Environmental Health Science, University of Georgia, Athens, GA, United States, 2Department of
Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States,
3Department of Population Health and Pathobiology, North Carolina State University, Raleigh, NC, United States
Sucralose is the most widely used artificial sweetener, and its health effects have been
highly debated over the years. In particular, previous studies have shown that sucralose
consumption can alter the gut microbiota. The gut microbiome plays a key role in
processes related to host health, such as food digestion and fermentation, immune cell
development, and enteric nervous system regulation. Inflammation is one of the most
common effects associated with gut microbiome dysbiosis, which has been linked to a
series of human diseases, such as diabetes and obesity. The aim of this study was to
investigate the structural and functional effects of sucralose on the gut microbiota and
associated inflammation in the host. In this study, C57BL/6 male mice received sucralose
in their drinking water for 6 months. The difference in gut microbiota composition
and metabolites between control and sucralose-treated mice was determined using
16S rRNA gene sequencing, functional gene enrichment analysis and metabolomics.
Inflammatory gene expression in tissues was analyzed by RT-PCR. Alterations in
bacterial genera showed that sucralose affects the gut microbiota and its developmental
dynamics. Enrichment of bacterial pro-inflammatory genes and disruption in fecal
metabolites suggest that 6-month sucralose consumption at the human acceptable daily
intake (ADI) may increase the risk of developing tissue inflammation by disrupting the gut
microbiota, which is supported by elevated pro-inflammatory gene expression in the liver
of sucralose-treated mice. Our results highlight the role of sucralose-gut microbiome
interaction in regulating host health-related processes, particularly chronic inflammation.
Keywords: artificial sweetener, sucralose, gut microbiota, metabolomics, inflammation
INTRODUCTION
Artificial sweeteners are commonly used food additives that have much higher sweetness intensities
than table sugars (Gardner et al., 2012). Consumption of artificial sweeteners is increasing in
the United States, and in 2008, the prevalence of consumption of beverages containing artificial
sweeteners was 24.1% among adults. Sucralose, which is 600 times sweeter than sucrose, is one of
the most commonly used artificial sweeteners in the market due to its extremely sugar-like taste,
lack of a bitter aftertaste, stability at high temperatures, and long shelf-life (Grice and Goldsmith,
2000; Sylvetsky et al., 2012). The health effects of sucralose have been highly debated over the years.
A number of previous studies concluded that sucralose is safe for its intended use as an artificial
Bian et al. Sucralose Alters the Gut Microbiome
sweetener and that the body acquires no calories from sucralose
(Grotz and Munro, 2009; Sylvetsky et al., 2012). Most ingested
sucralose is not absorbed or metabolized and moves through
the gastrointestinal tract unchanged (Roberts et al., 2000).
However, this does not prove that sucralose has no effect on
the gut microbiota. One study showed that a product containing
sucralose altered the rat gut microbiota and induce inflammatory
lymphocyte infiltration (Abou-Donia et al., 2008), but the study
was considered to be deficient in several aspects (Brusick et al.,
2009), including the use of high doses and a sucralose mixture
instead of pure sucralose. Another study that focused on the
metabolic effects of sucralose on environmental bacteria showed
that sucralose can inhibit the growth of certain bacterial species
(Omran et al., 2013). Therefore, sucralose may inhibit intestinal
bacteria and alter the gut microbiota, and these alterations could
affect host health.
The mucosal surfaces of the human intestines are host to more
than 100 trillion microbes (including bacteria, fungi, viruses, and
parasites) from more than 1,000 species (Ley et al., 2006; Qin
et al., 2010). Gut microbes interact with the host mucosa directly
via the recognition of pathogen-associated molecular patterns
(PAMPs), such as lipopolysaccharide, flagellin, and bacterial
DNA and RNA, by mucosal pattern recognition receptors (PRRs;
Maloy and Powrie, 2011). Alternatively, the interaction can
occur indirectly through secreted metabolites (Nicholson et al.,
2012). These interactions are involved in maintaining symbiotic
homeostasis. Increasing evidence indicates that this homeostasis
is vital for human health (Holmes et al., 2011; Tremaroli
and Backhed, 2012). Gut microbes can help maintain good
host health by participating in digestion and fermentation of
food, development of immune cells, regulation of the enteric
nervous system, and prevention of colonization by pathogens
(Holmes et al., 2011). The host, in turn, provides a habitat
and nutrients and secretes antibodies to inhibit the aggressive
expansion of microbes (Kamada et al., 2013). Being highly
diverse, the gut microbiota can be shaped by various factors,
including aging, diet, drugs, antibiotics, diseases, stress, exercise,
and environmental pollutants (Ley et al., 2006; Nicholson et al.,
2012), and if homeostasis is disrupted as a result of this shaping,
many adverse outcomes may occur, such as cardiovascular
disease, obesity, diabetes, allergies, and cancer (Ley et al., 2006).
For example, an increased ratio of Firmicutes to Bacteroidetes
was found in obese mice compared with their lean littermates
(Turnbaugh et al., 2006), and obesity-related phenotypes were
found to be transmissible in a study in which fecal microbes
from obese and lean human twins were transferred to germ-free
mice (Ridaura et al., 2013). Likewise, a remarkable increase in
taurocholic acid and Bilophila wadsworthia induced by dietary
fat promotes colitis in IL10-deficient mice (Devkota et al., 2012).
Metabolites produced from dietary choline by gut microbes
have been shown to be modulated in obesity, diabetes, and
cardiovascular diseases (Kim et al., 2010; Wang et al., 2011).
Abbreviations: PAMPs, pathogen-associated molecular patterns; PRRs, pattern
recognition receptors; IBD, inflammatory bowel disease; LPS, lipopolysaccharide;
iNOS, inducible nitric-oxide synthase; MMP-2, matrix metalloproteinase 2; ROS,
reactive oxygen species; GI, gastrointestinal.
Inflammation is one of the most common physical conditions
associated with gut microbiota dysbiosis. For example, acute
or chronic inflammation is the primary characteristic of
inflammatory bowel diseases (IBDs; Xavier and Podolsky, 2007),
of which a disrupted gut microbiota is one of the major
triggers in addition to genetic factors and the host immune
system, although the precise etiology remains unclear (Hill and
Artis, 2010). Moreover, increasing evidence demonstrates that
low-grade chronic inflammation induced by gut microbiota
disruption is associated with metabolic diseases (Holmes et al.,
2011). Obesity and diabetes are associated with low-grade
inflammation not only in adipose tissues but also systemically.
A study of bariatric surgery, a method of reducing body weight
for obese individuals, showed that one gut microbe species,
Faecalibacterium prausnitzii, is directly related to the reduction
of low-grade inflammation in obesity and diabetes (Furet et al.,
2010). Dyslipidemia induced by a high-fat diet results in
increased levels of lipopolysaccharide (LPS), which is a pro-
inflammatory mediator (Holmes et al., 2011). Moreover, chronic
inflammation induced by gut microbes can drive the progression
of colorectal cancer from adenoma to invasive carcinoma (Uronis
et al., 2009). Thus, inflammation can be triggered and modulated
by an altered gut microbiota, and exposure to compounds that
can alter the gut microbiota may induce inflammation in the host.
In this study, we first used 16S rRNA gene sequencing to
examine the effects of sucralose on the gut microbiome of
C57BL6/J mice over a 6-month administration period. Next,
we used metabolomics to profile fecal metabolome changes
associated with a perturbed gut microbiome. Finally, we assessed
several markers of inflammation to define the effects of sucralose
consumption on host tissues. Our results show that sucralose
altered the gut microbiome and associated metabolic profiles,
which may contribute to inflammatory response in the mouse
liver.
MATERIALS AND METHODS
Animals and Sucralose Exposure
Male C57BL/6J mice (8 weeks old) purchased from the Jackson
Laboratory (Bar Harbor, ME) were used in this study. Twenty
male mice were housed in the University of Georgia animal
facility for a week before the study and then assigned to the
control or treatment group (ten mice in each group), which
received tap water or sucralose (Sigma-Aldrich, MO) in tap water,
respectively, for 6 months. The concentration of sucralose was 0.1
mg/ml, which was equivalent to the FDA-approved acceptable
daily intake (ADI) in humans (5 mg/kg/day). Fresh solutions
were made every week, and the consumption of water was
measured for both groups. Standard pelleted rodent diet and tap
water were provided to the mice ad libitum, and the mice were
housed in environmental conditions of 22C, 40–70% humidity,
and a 12:12 h light:dark cycle before and during the experiment.
Body weight was measured before and after the treatment. Fecal
pellets were collected at baseline and at three and 6 months
of treatment. Mice were euthanized with carbon dioxide and
necropsied after 6 months. All experiments were approved by
the University of Georgia Institutional Animal Care and Use
Frontiers in Physiology | www.frontiersin.org 2July 2017 | Volume 8 | Article 487
Bian et al. Sucralose Alters the Gut Microbiome
Committee. The animals were treated humanely and with regard
for alleviation of suffering.
16S rRNA Gene Sequencing of the Gut
Microbiota
The gut microbiota was investigated using 16S rRNA gene
sequencing in fecal samples at different time points. DNA was
isolated from the feces of individual mice using a PowerSoil
DNA Isolation Kit (MO BIO Laboratories) according to
the manufacturer’s instructions, and the resultant DNA was
quantified and stored at 80C until further analysis. The V4
region in the 16S rRNA gene was targeted using the universal
primers 515 (5-GTGCCAGCMGCCGCGGTAA) and 806 (5-
GGACTACHVGGGTWTCTAAT). For each sample, 1 ng of the
purified fecal DNA was used as a template for amplification
and then barcoded with specific indexes. The amplified products
were then normalized, pooled and sequenced by an Illumina
MiSeq at the Georgia Genomics Facility. Paired-end 250 ×250
(PE250, v2 kit) reads were generated at a depth of at least
25,000 reads per sample. Geneious 8.1.5 (Biomatters, Auckland,
New Zealand) was used to process the raw fastq files, and the
mate-paired files were trimmed to dispose of bases with an
error probability higher than 0.01 and then merged. The data
were then analyzed using quantitative insights into microbial
ecology (QIIME, version 1.9.1) (Caporaso et al., 2010), and
UCLUST was used to match operational taxonomic units (OTUs)
with 97% sequence similarity against Greengenes database 13.8.
The matched sequences were assigned at five different levels:
phylum, class, order, family and genus. The raw data of controls
and treated-mice have been uploaded into the MG-RAST
server (http://metagenomics.anl.gov/) with the following job
IDs: 317595, 317584, 317588, 317586, 317583, 317589, 317596,
317592, 317585, 317598, 317590, 317599, 317582, 317594,
317591, 317581, 317597, 317580, 317593, and 317587.
Functional Gene Enrichment Analysis
An open-source R package, Tax4Fun, was first used to analyze
the enrichment of functional genes of the microbiome of each
group (Asshauer et al., 2015). The output from QIIME with
a SILVA database extension (SILVA 119) was used for this
analysis. Tax4Fun can survey the functional genes of bacterial
communities based on the 16S rRNA sequencing data and
provide a good approximation to the gene profiles obtained
from metagenomic shotgun sequencing methods. The results
from Tax4Fun were further analyzed using Statistical Analysis
of Metagenomic Profiles (STAMP) (version 2.1.3) (Parks et al.,
2014).
Fecal Metabolomics Analysis
Metabolites in fecal samples collected at 6 months were extracted
using methanol and water as previously described (Lu et al.,
2014). In brief, 20 mg of feces was disrupted in 1 ml of a
methanol/water solution (1:1) with a TissueLyser at 50 Hz for 5
min, followed by centrifugation at 12,000 rpm for 10 min. The
resultant upper phase was collected and dried using a SpeedVac,
and the dried samples were re-suspended in 20% acetonitrile
for MS analysis. Metabolomic profiling was conducted using
a quadrupole-time-of-flight (Q-TOF) 6520 mass spectrometer
(Agilent Technologies, Santa Clara, CA) with an electrospray
ionization source interfaced with an Agilent 1200 HPLC system.
The detailed method for metabolomics was published previously
(Lu et al., 2012). The Q-TOF was calibrated with standard tuning
solution (Agilent Technologies) daily to ensure a mass accuracy
within 5 ppm. A YMC Hydrosphere C18 column was used to
separate the metabolites, and all detectable molecular features in
a mass range of 30–2,000 m/z were captured in the positive mode.
Metabolomic Data Processing and
Metabolite Identification
The data obtained from the HPLC-Q-TOF system were
processed and analyzed as previously described (Lu et al., 2012).
Briefly, the raw.d data were converted to .mzdata format using
MassHunter Workstation software (Agilent), and only signals
with an intensity higher than 1,000 counts were included in
the subsequent analysis. Peak alignment, intensity calculations,
and comparisons between the control and treatment group
were performed using the XCMS Online tools. Significantly
changed molecular features were profiled and searched against
the Human Metabolome Database (HMDB) (http://www.hmdb.
ca) and METLIN (http://metlin.scripps.edu) with a 10-ppm mass
accuracy threshold. The matched exact masses were stored and
used for the generation of MS/MS data to tentatively identify the
metabolites. The matched molecular features were fragmented
using MS/MS in the Q-TOF 6520 mass spectrometer to obtain
the product ions, and the spectra were compared with the HMDB
and METLIN MS/MS database to identify significantly altered
metabolites, with at least two matched fragments within a 200
ppm mass accuracy as the matching threshold.
Quantitative Real-Time Polymerase Chain
Reaction (qPCR)
Liver was first segmented into left lobe, media lobe, right lobe,
and caudate lobe during necropsy. Each liver segment was put
into a 2 ml tube, followed by immediate addition of 1 ml of
RNAlater R
solution (Thermo Fisher Scientific). The tubes were
stored at +4C overnight to allow the RNAlater R
solution to
inhibit the RNase before they were transferred to the 80C
freezer for storage. Liver samples (right lobe) treated with
RNAlater R
were used to isolate RNA with an RNeasy Mini
Kit (Qiagen, Valencia, CA) according to the manufacturer’s
instructions, and the resultant RNA was digested with a DNA-
freeTM DNA Removal Kit (Thermo Fisher Scientific) to remove
genomic DNA contamination. RNA integrity number (RIN) for
each RNA sample was measured with an Agilent Bioanalyzer.
The RIN typically was >9.0, indicating no RNA degradation
in the samples and processing. Then, cDNA was synthesized
from 1 µg of total RNA using iScriptTM Reverse Transcription
Supermix for RT-qPCR (Bio-Rad Laboratories, CA), and the
products were diluted to 1:5 before use in subsequent reactions.
Quantitative real-time PCR was performed on a Bio-Rad
CFX96 TouchTM Real-Time PCR Detection System using
SsoAdvancedTM Universal SYBR R
Green Supermix (Bio-Rad).
The sequences of the primers used for quantitative PCR were
Frontiers in Physiology | www.frontiersin.org 3July 2017 | Volume 8 | Article 487
Bian et al. Sucralose Alters the Gut Microbiome
as follows: TNF-α, 5-CCCTCACACTCAGATCATCTTCT and
5-GCTACGACGTGGGCTACAG; IL-6, 5-TAGTCCTTCCTA
CCCCAATTTCC and 5-TTGGTCCTTAGCCACTCCTTC;
IL-1β, 5-GCAACTGTTCCTGAACTCAACT and 5-ATCTTT
TGGGGTCCGTCAACT; iNOS, 5-GTTCTCAGCCCAACAATA
CAAGA and 5-GTGGACGGGTCGATGTCAC; MMP-2, 5-CAG
GGAATGAGTACTGGGTCTATT and 5-ACTCCAGTTAAA
GGCAGCATCTAC; MMP-9, 5-ATCTCTTCTAGAGACTGG
GAAGGAG and 5-AGCTGATTGACTAAAGTAGCTGGA;
MMP-13, 5-GTGTGGAGTTATGATGATGT and 5-TGCGAT
TACTCCAGATACTG; and β-actin, 5-CGTGCGTGACATCAA
AGAGAA and 5-TGGATGCCACAGGATTCCAT. All results
were normalized to the β-actin or GAPDH gene (endogenous
control). The fold change in expression over control samples was
calculated using the 11CT method by CFX manager software
(Bio-Rad). The qPCR conditions were 95C for 10 min, 40 cycles
of 15 s at 95C, 30 s at 60C, and 30 s at 72C, and a final melting
curve analysis performed by raising the temperature from 65
to 95C in 0.5C increments for 0.05 s each. Potential genomic
DNA contamination was controlled for by DNase digestion and
the inclusion of a No-RT control, and technical contamination
was controlled for by the inclusion of a No-template control.
Data Analysis
The difference in individual gut bacterial components between
control and sucralose-treated mice at different time points was
assessed with the Mothur software. A two-tailed Welch’s t-test
(p<0.05) was used to compare the difference in metabolites
between the control and sucralose-treated mice. Additionally,
principle component analysis (PCA) was used to examine the
intrinsic clusters and outliers. Partial least squares discriminant
analysis (PLS-DA) and a hierarchical clustering heat map was
used to visualize metabolomic difference in different groups. A
two-tailed Student’s t-test was used to determine the statistical
significance of pro-inflammatory gene expression between the
controls and treated mice.
RESULTS
Sucralose Altered the Developmental
Dynamics of the Gut Microbiome
The gut microbiome is a dynamic system, and its bacterial
composition shifts over time. Maintaining normal developmental
trajectories of the gut microbiome is critical for its functions.
Feces collected from both groups of mice at baseline and
after three and 6 months of administration were employed
to investigate the effects of sucralose on the gut microbiome.
Using 16S rRNA gene sequencing, we found that 14 genera
exhibited different patterns over time in sucralose-treated mice
compared with control mice, as shown in Figure 1. These
bacterial genera exhibited no significant difference in abundance
at baseline but were significantly different after three and/or
6 months of treatment, indicating that sucralose disrupts the
developmental dynamics of gut bacteria. The genera included
Turicibacteraceae Turicibacter,Lachnospiraceae Ruminococcus,
Ruminococcaceae Ruminococcus,Verrucomicrobiaceae
Akkermansia,Staphylococcaceae Staphylococcus,Streptococcaceae
Streptococcus,Dehalobacteriaceae Dehalobacterium,
Lachnospiraceae Anaerostipes,Lachnospiraceae Roseburia,
and unclassified members in Family Clostridiaceae,
Christensenellaceae,Peptostreptococcaceae,Erysipelotrichaceae
and Order Bacillales..
Sucralose Increased the Abundance of
Bacterial Genes Related to
Pro-inflammatory Mediators
We next examined whether the changes in gut microbiome
composition were associated with functional perturbations of
the gut bacteria. Indeed, a number of bacterial functional genes
were enriched in sucralose-treated mice. For example, functional
gene enrichment analysis of the gut microbiome showed that
genes related to bacterial pro-inflammatory mediators were
highly elevated in sucralose-treated mice, as shown in Figure 2.
Specifically, genes related to LPS synthesis were significantly
increased after 6 months of treatment. In addition, multiple genes
related to flagella protein synthesis were increased in sucralose-
treated mice. Likewise, genes involved in fimbriae synthesis
increased in sucralose-treated mice. Numerous bacterial toxin
genes, such as toxic shock syndrome toxin-1 and shiga toxin
subunits, were also elevated in sucralose-treated mice.
Sucralose Changed the Fecal Metabolome
We next conducted metabolomic profiling to examine the
functional impact of sucralose on the fecal metabolome. The
combination in feces of a large quantity of gut bacteria and
their metabolic products creates an ideal biological sample to
assess functional changes in the gut microbiome. A total of
13,611 molecular features were detected in fecal samples, 1,764 of
which were significantly different (p<0.05 and fold change>1.5)
between the sucralose-treated and control mice (Figure 3A),
clearly indicating that sucralose perturbed the fecal metabolome.
A PLS-DA plot (Figure 3B) showed a separation in the molecular
patterns of the two groups, and the hierarchical clustering heat
map (Figure 3C) was consistent with this result. Molecular
features matched with the HMDB and METLIN database were
used for metabolite identification via MS/MS. We tentatively
identified 66 metabolites, including quorum sensing compounds,
amino acids and derivatives, lipids, fatty acids, bile acids, and
nucleic acids, among others (Supplementary Table 1).
Sucralose Altered Quorum Sensing Signals
Bacteria control multicellular behaviors, such as biofilm growth
and development, horizontal gene transfer, host-microbe cross-
talk, and microbe-microbe interactions, by the cell-cell signaling
process known as quorum sensing. Four acyl homoserine
lactones known to be quorum sensing signals (Bainton et al.,
1992; Winson et al., 1995; Passador et al., 1996; Stankowska
et al., 2008; Lade et al., 2014) were identified: N-butanoyl-
l-homoserine lactone, N-(3-oxo-hexanoyl)-homoserine lactone,
N-tetradecanoyl-L-homoserine lactone, and N-pentadecanoyl-L-
homoserine lactone. The reduced abundance of these quorum
sensing signals in sucralose-treated mice (Figure 4) indicates that
sucralose disrupts quorum sensing signaling.
Frontiers in Physiology | www.frontiersin.org 4July 2017 | Volume 8 | Article 487
Bian et al. Sucralose Alters the Gut Microbiome
FIGURE 1 | Sucralose altered the dynamics of gut microbiome development in C57BL6/J mice. Bacterial genera exhibited different patterns over time between the
control and sucralose-treated mice. *p<0.05.
Sucralose Altered Amino Acids and Derivatives
The gut microbiome is highly involved in the synthesis and
regulation of amino acids. Amino acids, such as L-tryptophan,
L-tyrosine, L-leucine, and L-isoleucine, as well as their
derivatives (Table S1) were affected by sucralose treatment.
Four compounds involved in tryptophan metabolism were
identified, including L-tryptophan (Trp), quinolinic acid,
kynurenic acid, and 2-aminomuconic acid, as shown in
Figure 5A. Compared with control mice, Trp, quinolinic acid,
and 2-aminomuconic acid were increased by 1.71-, 5.45-,
and 2.09-fold in sucralose-treated mice, while kynurenic
acid was reduced by 2.45-fold in sucralose-treated mice.
For tyrosine metabolism, though L-tyrosine increased (1.62-
fold), two of its metabolites, p-hydroxyphenylacetic acid and
cinnamic acid, decreased by 4.63- and 1.53-fold, respectively
(Figure 5B).
Sucralose Altered Bile Acids
The gut microbiome can transform primary hydrophilic bile
acids into secondary hydrophilic bile acids in the large intestine
through deconjugation, dehydroxylation, and dehydrogenation.
Bile acids not only facilitate fat and fat-soluble vitamin absorption
and maintain cholesterol homeostasis but are also viewed as
signaling molecules that bind to the nuclear receptor FXR and
the G-protein-coupled receptor TGR5. Several bile acids were
significantly different between the control and sucralose-treated
animals (Figure 6). 3-Oxo-4,6-choladienoic acid was increased
in sucralose-treated mice compared with control mice, while
other bile acids were reduced, including 3β,7α-dihydroxy-
5-cholestenoate, 3α,7β,12α-trihydroxyoxocholanyl-glycine
and lithocholic acid/isoallolithocholic acid/allolithocholic
acid/isolithocholic acid.
Sucralose Induced Elevated
Pro-inflammatory Gene Expression in Liver
As described above, sucralose could increase the production
of bacterial pro-inflammatory mediators, which may cause
inflammatory responses in host tissues after being translocated
into the host circulation. In fact, sucralose-treated mice exhibited
elevated gene expression of pro-inflammatory markers in the
liver (Figure 7), such as matrix metalloproteinase 2 (MMP-2) and
inducible nitric-oxide synthase (iNOS).
Frontiers in Physiology | www.frontiersin.org 5July 2017 | Volume 8 | Article 487
Bian et al. Sucralose Alters the Gut Microbiome
FIGURE 2 | Enrichment of bacterial pro-inflammatory genes after 6 months of sucralose treatment (*p<0.05, **p<0.01), including genes involved in LPS (A),
flagella (B), and fimbriae synthesis (C) as well as toxins (D) and multidrug resistance genes (E).
DISCUSSION
The gut microbiota is a dynamic system, and maintaining a
healthy balance is vital for the host (Nicholson et al., 2012).
Previous studies have demonstrated that changes to the gut
microbiota affect numerous host processes, such as immune
system development and energy metabolism and absorption,
and can also impact diseases in and beyond the GI tract
(Holmes et al., 2011). Xenobiotics in the food or environment
can affect the gut microbiome and host health (Lu et al.,
2014; Suez et al., 2014; Gao et al., 2017). One common
argument used to support sucralose safety is that the majority
of sucralose is not absorbed or metabolized in the body (Grice
and Goldsmith, 2000). However, we demonstrate here that
sucralose can affect the gut microbiome, its metabolic functions
and the host even though it passes through the GI tract
unchanged.
Specifically, we investigated the effect of sucralose
consumption on the gut microbiota and host in mice using
16S rRNA gene sequencing, functional gene enrichment
analysis, metabolomics and real-time PCR. Sucralose
consumption for 6 months altered the gut microbiome
composition, fecal metabolites, and pro-inflammatory gene
expression in the liver. The alterations induced by sucralose
consumption could affect the development of inflammation
and further influence other physiological functions in the
Frontiers in Physiology | www.frontiersin.org 6July 2017 | Volume 8 | Article 487
Bian et al. Sucralose Alters the Gut Microbiome
FIGURE 3 | Sucralose changed the fecal metabolome, as illustrated by the Cloud plot (A), PLS-DA plot (B), and heat map (C). A total of 1,764 molecular features
were significantly different (p<0.05 and fold change>1.5) between sucralose-treated mice and controls.
body. This study provides a new understanding of the
effect of artificial sweeteners on the gut microbiota and
host health.
Sucralose has been shown to be safe using different
endpoints in previous studies, but very few studies have
reported its effects on the gut microbiome and, particularly,
its functions (Grotz and Munro, 2009). In this study, we
examined sucralose-induced gut microbiome functional
perturbation, which may contribute to the development of
systemic inflammation in the host. Altering the gut bacterial
composition may confer an increased risk of developing
inflammation in sucralose-treated mice. For example, among
the 14 changed genera, several were found to be associated
with host inflammation. Ruminococcaceae Ruminococcus,
which were more abundant in sucralose-treated mice in
this study, were shown to be more abundant in colonic
Crohn’s disease samples than in healthy samples in a previous
study (Willing et al., 2010); Streptococcaceae Streptococcus,
Dehalobacteriaceae Dehalobacterium,Lachnospiraceae
Anaerostipes, and Lachnospiraceae Ruminococcus, which
were reduced in sucralose-treated mice, were found to be
negatively associated with inflammation in previous studies
(Willing et al., 2010; Collins et al., 2014; Fernández et al.,
2016; Munyaka et al., 2016). The functional impact of these
altered gut bacteria remains to be further elucidated in the
future. Nevertheless, alterations in gut microbiome composition
may lead to differential functional bacterial metagenomes and
metabolic capacities of the gut microbiome.
Frontiers in Physiology | www.frontiersin.org 7July 2017 | Volume 8 | Article 487
Bian et al. Sucralose Alters the Gut Microbiome
FIGURE 4 | Quorum sensing signals were altered by sucralose consumption. *p<0.05, ***p<0.001.
FIGURE 5 | Amino acids and derivatives altered by sucralose. *p<0.05, **p<0.01, ***p<0.001) (A) tryptophan metabolites; (B) tyrosine metabolites.
Frontiers in Physiology | www.frontiersin.org 8July 2017 | Volume 8 | Article 487
Bian et al. Sucralose Alters the Gut Microbiome
FIGURE 6 | Sucralose altered bile acids in the fecal samples of mice treated with sucralose for 6 months. *p<0.05, **p<0.01.
FIGURE 7 | Sucralose consumption increased the gene expression of inflammatory markers in the liver, as examined by qRT-PCR. The mRNA expression of matrix
metalloproteinase 2 (MMP-2) and inducible nitric-oxide synthase (iNOS) was elevated in sucralose-treated mice. *p<0.05, N.S., no statistical significance.
Frontiers in Physiology | www.frontiersin.org 9July 2017 | Volume 8 | Article 487
Bian et al. Sucralose Alters the Gut Microbiome
Previous studies have demonstrated that functional genes of
the bacterial community are related to 16S rRNA marker genes,
allowing the functional capacities of the gut microbiome to be
surveyed using 16S rRNA gene sequencing (Asshauer et al.,
2015). Using functional gene enrichment analysis, a number
of genes related to bacterial pro-inflammatory mediators were
shown to be significantly increased in the sucralose-treated gut
microbiome, including genes involved in LPS synthesis, flagella
protein synthesis, and fimbriae synthesis as well as bacterial
toxins and drug resistance genes. LPS, flagella, and fimbriae are
known PAMPs that can trigger pathological inflammation in
the host, and various toxins produced by bacteria can induce
toxicity in the host. LPS, a known endotoxin from the outer
membrane of gram-negative bacteria, can initiate inflammatory
events, such as the secretion of pro-inflammatory cytokines like
interleukin-6 or tumor necrosis factor (TNF)-α(de La Serre et al.,
2010). Flagella protein levels are low in a healthy gut, and high
levels of flagella proteins have been shown to be associated with
gut mucosal barrier breakdown and inflammation in previous
studies (Cullender et al., 2013). Fimbriae play an important
role in bacterial adhesion to and invasion of epithelial cells
and are known virulence factors (Nakagawa, 2002). Additionally,
multidrug resistance genes were increased in the sucralose-
treated gut microbiome, and the increase in multidrug resistance
genes and/or multidrug-resistant bacteria may lead to a more
hostile gut environment (Marshall et al., 2009). These data
indicate that 6 months of sucralose consumption increased the
pro-inflammatory products of the gut microbiome and its ability
to potentially induce systemic inflammation.
Likewise, the metabolites identified in fecal samples may be
involved in regulating inflammation. For example, several amino
acids were perturbed in sucralose-treated fecal metabolites in this
study. In particular, we found that tryptophan metabolism was
disrupted by sucralose, and this disruption was related to changes
in the expression of functional genes of the gut microbiome.
As shown in Supplementary Figure 1, several genes related to
tryptophan metabolism were elevated, while the abundance of
tryptophan and its metabolites were altered in the fecal samples.
The four metabolites identified are involved in the kynurenine
pathway, which is the most important tryptophan metabolism
pathway, consuming 95% of the tryptophan in the body
(Keszthelyi et al., 2009). The balance between two metabolites
in this pathway, quinolinic acid, and kynurenic acid, plays an
important role in mediating inflammation and the excitability
of cells such as enteric neurons. Quinolinic acid is pro-
inflammatory and excitotoxic, whereas kynurenic acid is anti-
inflammatory and neuroprotective (Keszthelyi et al., 2009). Here,
we found an elevated level of quinolinic acid and a decreased
level of kynurenic acid. This indicates that sucralose may shift
cells to a pro-inflammatory state. Likewise, tyrosine and two of its
metabolites, p-hydroxyphenylacetic acid and cinnamic acid, have
previously been shown to decrease the production of reactive
oxygen species (ROS) in neutrophils (Beloborodova et al.,
2012), and the reduced level of these compounds in our study
indicated that ROS levels may be increased in sucralose-treated
mice. Bacterial antioxidative enzyme genes, such as catalase
and catalase-peroxidase, which respond to ROS, were also
significantly enriched in sucralose-treated mice (Supplementary
Figure 2). ROS can stimulate the release of pro-inflammatory
cytokines (Chapple, 1997); therefore, the decrease in the two
tyrosine metabolites may contribute to the development of a pro-
inflammatory state. Additionally, secondary bile acids that have
FIGURE 8 | Proposed functional link between sucralose-induced gut microbiota alterations and host inflammation. Sucralose perturbs the gut microbiome and its
metabolic functions, inducing the enrichment of bacterial pro-inflammatory mediators, and disrupting metabolites involved in inflammation regulation. Together, these
consequences may contribute to the induction of liver inflammation in the host.
Frontiers in Physiology | www.frontiersin.org 10 July 2017 | Volume 8 | Article 487
Bian et al. Sucralose Alters the Gut Microbiome
antimicrobial effects were decreased, which may allow the growth
of pathogens (Begley et al., 2005).
Pro-inflammatory mediators, such as LPS, and metabolites
can translocate into host circulation and tissues, leading to
systemic inflammatory response (de La Serre et al., 2010).
In accordance with this expectation, real-time PCR results
showed that MMP-2 and iNOS expression was elevated in the
liver of sucralose-treated mice. MMP-2 is strongly associated
with inflammatory responses, because it can cleave and
activate TNF-αand IL-1β, which are both pro-inflammatory
cytokines that contribute to the induction of inflammation
(Medina and Radomski, 2006; Wang et al., 2006). Likewise,
iNOS-derived NO regulates pro-inflammatory genes and
significantly contributes to inflammatory liver injury. iNOS
exerts numerous effects associated with the progression of
inflammatory conditions in multiple liver diseases, such as
increasing the liver inflammatory response, promoting the
induction of liver tumors and contributing to liver fibrosis
caused by a chronic viral infection (Sass et al., 2001; La
Mura et al., 2014). The expression of both MMP-2 and iNOS
was found to be increased in the liver of sucralose-treated
mice compared with control mice, indicating that sucralose
exposure increases the risk of developing inflammation in
the liver. Notably, most of the sucralose consumed passes
through the GI tract unabsorbed and unchanged (Roberts
et al., 2000); therefore, the inflammatory response observed
in the liver was unlikely to be stimulated by sucralose
directly.
Taken together, as illustrated in Figure 8, our data show
that 6-month sucralose consumption at human ADI alters the
gut microbiome and its functions in mice. In particular, the
enrichment of gut microbial pro-inflammatory genes and fecal
metabolites suggests that sucralose alters the gut environment to
release more pro-inflammatory mediators and alter functional
metabolites, which may contribute to the increased expression
of pro-inflammatory markers in the liver, such as iNOS and
MMP-2. Notably, while the majority of ingested sucralose
passes through the GI tract unabsorbed, it does disrupt
the gut microbiota and its functions; therefore, our results
highlight the role of sucralose-gut microbiome interaction
in regulating host health-related processes, such as chronic
inflammation.
There are a few limitations associated with this study. First,
we only assessed inflammatory response in the liver of sucralose-
treated mice by RT-PCR. Examination of host response using
other endpoints and methods, such as circulating LPS and
histological assessment, in related samples and tissues would
be needed to better characterize the effects of sucralose in
the body. Second, we conducted experiments using a single
dose of sucralose at the human ADI, while human intake of
sucralose is typically lower than this concentration. Our ongoing
study using multiple human-relevant doses aims at better
understanding time- and dose-dependent effects of sucralose on
the gut microbiome and host. Third, the enrichment analysis
of functional bacterial genes was performed based on the
16S rRNA sequencing data. Metagenomic shotgun sequencing
and/or metatranscriptomics will further shed light on sucralose-
induced functional perturbation of the gut microbiome. Finally,
the identification of altered metabolites was based on the
matching with the metabolite database. Future validation of key
metabolites of interest with authentic compounds is warranted.
Likewise, a more accurate quantitative analysis of altered
metabolites using stable isotope labeled standards should be
conducted.
AUTHOR CONTRIBUTIONS
XB, HR, and KL designed the study. XB, BG, LC, and PT
acquired, analyzed and interpreted the data. XB, PT, HR, and
KL drafted and critically revised the manuscript, approved the
version to be published, and are accountable for all aspects of the
work.
FUNDING
This research was partially supported by the University of
Georgia, University of North Carolina at Chapel Hill and
NIH/NIEHS (R01ES024950).
SUPPLEMENTARY MATERIAL
The Supplementary Material for this article can be found
online at: http://journal.frontiersin.org/article/10.3389/fphys.
2017.00487/full#supplementary-material
REFERENCES
Abou-Donia, M. B., El-Masry, E. M., Abdel-Rahman, A. A., McLendon, R. E., and
Schiffman, S. S. (2008). Splenda alters gut microflora and increases intestinal
p-glycoprotein and cytochrome p-450 in male rats. J. Toxicol. Environ. Health
A71, 1415–1429. doi: 10.1080/15287390802328630
Asshauer, K. P., Wemheuer, B., Daniel, R., and Meinicke, P. (2015).
Tax4Fun: predicting functional profiles from metagenomic 16S rRNA
data. Bioinformatics 31, 2882–2884. doi: 10.1093/bioinformatics/btv287
Bainton, N. J., Stead, P., Chhabra, S. R., Bycroft, B. W., Salmond, G. P.,
Stewart, G. S., et al. (1992). N-(3-oxohexanoyl)-L-homoserine lactone regulates
carbapenem antibiotic production in Erwinia carotovora. Biochem. J. 288,
997–1004. doi: 10.1042/bj2880997
Begley, M., Gahan, C. G., and Hill, C. (2005). The interaction between bacteria and
bile. FEMS Microbiol. Rev. 29, 625–651. doi: 10.1016/j.femsre.2004.09.003
Beloborodova, N., Bairamov, I., Olenin, A., Shubina, V., Teplova, V., and
Fedotcheva, N. (2012). Effect of phenolic acids of microbial origin on
production of reactive oxygen species in mitochondria and neutrophils. J.
Biomed. Sci. 19:89. doi: 10.1186/1423-0127-19-89
Brusick, D., Borzelleca, J. F., Gallo, M., Williams, G., Kille, J., Wallace Hayes, A.,
et al. (2009). Expert panel report on a study of splenda in male rats. Regul.
Toxicol. Pharmacol. 55, 6–12. doi: 10.1016/j.yrtph.2009.06.013
Caporaso, J. G., Kuczynski, J., Stombaugh, J., Bittinger, K., Bushman,
F. D., Costello, E. K., et al. (2010). QIIME allows analysis of high-
throughput community sequencing data. Nat. Methods 7, 335–336.
doi: 10.1038/nmeth.f.303
Frontiers in Physiology | www.frontiersin.org 11 July 2017 | Volume 8 | Article 487
Bian et al. Sucralose Alters the Gut Microbiome
Chapple, I. L. C. (1997). Reactive oxygen species and antioxidants in inflammatory
diseases. J. Clin. Periodontol. 24, 287–296.
Collins, J. W., Chervaux, C., Raymond, B., Derrien, M., Brazeilles, R.,
Kosta, A., et al. (2014). Fermented dairy products modulate citrobacter
rodentium-induced colonic hyperplasia. J. Infect. Dis. 210, 1029–1041.
doi: 10.1093/infdis/jiu205
Cullender, T. C., Chassaing, B., Janzon, A., Kumar, K., Muller, C. E., Werner,
J. J., et al. (2013). Innate and adaptive immunity interact to quench
microbiome flagellar motility in the gut. Cell Host Microbe 14, 571–581.
doi: 10.1016/j.chom.2013.10.009
de La Serre, C. B., Ellis, C. L., Lee, J., Hartman, A. L., Rutledge, J. C., and Raybould,
H. E. (2010). Propensity to high-fat diet-induced obesity in rats is associated
with changes in the gut microbiota and gut inflammation. Am. J. Physiol.
Gastrointest. Liver Physiol. 299, G440–G448. doi: 10.1152/ajpgi.00098.2010
Devkota, S., Wang, Y., Musch, M. W., Leone, V., Fehlner-Peach, H.,
Nadimpalli, A., et al. (2012). Dietary-fat-induced taurocholic acid promotes
pathobiont expansion and colitis in II10/mice. Nature 487, 104–108.
doi: 10.1038/nature11225
Fernández, J., Redondo-Blanco, S., Gutiérrez-del-Río, I., Miguélez, E. M.,
Villar, C. J., and Lombó, F. (2016). Colon microbiota fermentation of
dietary prebiotics towards short-chain fatty acids and their roles as anti-
inflammatory and antitumour agents: a review. J. Funct. Foods 25, 511–522.
doi: 10.1016/j.jff.2016.06.032
Furet, J. P., Kong, L. C., Tap, J., Poitou, C., Basdevant, A., Bouillot, J. L., et al.
(2010). Differential adaptation of human gut microbiota to bariatric surgery-
induced weight loss: links with metabolic and low-grade inflammation markers.
Diabetes 59, 3049–3057. doi: 10.2337/db10-0253
Gao, B., Bian, X., Mahbub, R., and Lu, K. (2017). Sex-specific effects of
organophosphate diazinon on the gut microbiome and its metabolic functions.
Environ. Health Perspect. 125, 198–206. doi: 10.1289/EHP202
Gardner, C., Wylie-Rosett, J., Gidding, S. S., Steffen, L. M., Johnson, R. K., Reader,
D., et al. (2012). Nonnutritive sweeteners: current use and health perspectives:
a scientific statement from the American heart association and the american
diabetes association. Diabetes Care 35, 1798–1808. doi: 10.2337/dc12-9002
Grice, H. C., and Goldsmith, L. A. (2000). Sucralose—an overview of the toxicity
data. Food Chem. Toxicol. 38, S1–S6. doi: 10.1016/S0278-6915(00)00023-5
Grotz, V. L., and Munro, I. C. (2009). An overview of the safety of sucralose. Regul.
Toxicol. Pharmacol. 55, 1–5. doi: 10.1016/j.yrtph.2009.05.011
Hill, D. A., and Artis, D. (2010). Intestinal bacteria and the regulation
of immune cell homeostasis. Annu. Rev. Immunol. 28, 623–667.
doi: 10.1146/annurev-immunol-030409-101330
Holmes, E., Li, J. V., Athanasiou, T., Ashrafian, H., and Nicholson, J.
K. (2011). Understanding the role of gut microbiome-host metabolic
signal disruption in health and disease. Trends Microbiol. 19, 349–359.
doi: 10.1016/j.tim.2011.05.006
Kamada, N., Seo, S. U., Chen, G. Y., and Nunez, G. (2013). Role of the gut
microbiota in immunity and inflammatory disease. Nat. Rev. Immunol. 13,
321–335. doi: 10.1038/nri3430
Keszthelyi, D., Troost, F. J., and Masclee, A. A. (2009).
Understanding the role of tryptophan and serotonin metabolism in
gastrointestinal function. Neurogastroenterol. Motil. 2, 1239–1249.
doi: 10.1111/j.1365-2982.2009.01370.x
Kim, I. Y., Jung, J., Jang, M., Ahn, Y. G., Shin, J. H., Choi, J. W., et al. (2010).
1H NMR-based metabolomic study on resistance to diet-induced obesity in
AHNAK knock-out mice. Biochem. Biophys. Res. Commun. 403, 428–434.
doi: 10.1016/j.bbrc.2010.11.048
Lade, H., Paul, D., and Kweon, J. H. (2014). N-acyl homoserine lactone-
mediated quorum sensing with special reference to use of quorum quenching
bacteria in membrane biofouling control. Biomed. Res. Int. 2014:162584.
doi: 10.1155/2014/162584
La Mura, V., Pasarin, M., Rodriguez-Vilarrupla, A., Garcia-Pagan, J. C., Bosch,
J., and Abraldes, J. G. (2014). Liver sinusoidal endothelial dysfunction after
LPS administration: a role for inducible-nitric oxide synthase. J. Hepatol. 61,
1321–1327. doi: 10.1016/j.jhep.2014.07.014
Ley, R. E., Peterson, D. A., and Gordon, J. I. (2006). Ecological and evolutionary
forces shaping microbial diversity in the human intestine. Cell 124, 837–848.
doi: 10.1016/j.cell.2006.02.017
Lu, K., Abo, R. P., Schlieper, K. A., Graffam, M. E., Levine, S., Wishnok, J. S., et al.
(2014). Arsenic exposure perturbs the gut microbiome and its metabolic profile
in mice: an integrated metagenomics and metabolomics analysis. Environ.
Health Perspect. 122, 284–291. doi: 10.1289/ehp.1307429
Lu, K., Knutson, C. G., Wishnok, J. S., Fox, J. G., and Tannenbaum, S. R.
(2012). Serum metabolomics in a Helicobacter hepaticus mouse model of
inflammatory bowel disease reveal important changes in the microbiome,
serum peptides, and intermediary metabolism. J. Proteome Res. 11, 4916–4926.
doi: 10.1021/pr300429x
Maloy, K. J., and Powrie, F. (2011). Intestinal homeostasis and its breakdown in
inflammatory bowel disease. Nature 474, 298–306. doi: 10.1038/nature10208
Marshall, B. M., and Ochieng, D. J., Levy, S. B. (2009). Commensals:
underappreciated reservoir of antibiotic resistance. Microbe 4, 231–238.
doi: 10.1128/microbe.4.231.1
Medina, C., and Radomski, M. W. (2006). Role of matrix metalloproteinases
in intestinal inflammation. J. Pharmacol. Exp. Ther. 318, 933–938.
doi: 10.1124/jpet.106.103465
Munyaka, P. M., Rabbi, M. F., Khafipour, E., and Ghia, J. E. (2016). Acute dextran
sulfate sodium (DSS)-induced colitis promotes gut microbial dysbiosis in mice.
J. Basic Microbiol. 56, 986–998. doi: 10.1002/jobm.201500726
Nakagawa, I. (2002). Functional differences among fima variants of
porphyromonas gingivalis and their effects on adhesion to and
invasion of human epithelial cells. Infect. Immun. 70, 277–285.
doi: 10.1128/IAI.70.1.277-285.2002
Nicholson, J. K., Holmes, E., Kinross, J., Burcelin, R., Gibson, G., Jia, W., et al.
(2012). Host-gut microbiota metabolic interactions. Science 3366, 1262–1267.
doi: 10.1126/science.1223813
Omran, A., Ahearn, G., Bowers, D., Swenson, J., and Coughlin, C. (2013).
Metabolic effects of sucralose on environmental bacteria. J. Toxicol.
2013:372986. doi: 10.1155/2013/372986
Parks, D. H., Tyson, G. W., Hugenholtz, P., and Beiko, R. G. (2014). STAMP:
statistical analysis of taxonomic and functional profiles. Bioinformatics 30,
3123–3124. doi: 10.1093/bioinformatics/btu494
Passador, L., Tucker, K. D., Guertin, K. R., Journet, M. P., Kende,
A. S., and Iglewski, B. H. (1996). Functional analysis of the
Pseudomonas aeruginosa autoinducer PAI. J. Bacteriol. 178, 5995–6000.
doi: 10.1128/jb.178.20.5995-6000.1996
Qin, J., Li, R., Raes, J., Arumugam, M., Burgdorf, K. S., Manichanh, C., et al.
(2010). A human gut microbial gene catalogue established by metagenomic
sequencing. Nature 464, 59–65. doi: 10.1038/nature08821
Ridaura, V. K., Faith, J. J., Rey, F. E., Cheng, J., Duncan, A. E., Kau, A. L., et al.
(2013). Gut microbiota from twins discordant for obesity modulate metabolism
in mice. Science 341:1241214. doi: 10.1126/science.1241214
Roberts, A., Renwick, A. G., Sims, J., and Snodin, D. J. (2000). Sucralose
metabolism and pharmacokinetics in man. Food Chem. Toxicol. 38, 31–41.
doi: 10.1016/S0278-6915(00)00026-0
Sass, G., Koerber, K., Bang, R., Guehring, H., and Tiegs, G. (2001). Inducible nitric
oxide synthase is critical for immune-mediated liver injury in mice. J. Clin.
Invest. 107, 439–447. doi: 10.1172/JCI10613
Stankowska, D., Kwinkowski, M., and Kaca, W. (2008). Quantification of proteus
mirabilis virulence factors and modulation by acylated homoserine lactones.
J. Microbiol. Immunol. Infect. 41, 243–253. Avilable online at: http://www.ejmii.
com/issue_abstract.php?code=PDT4b0d2267aca1b
Suez, J., Korem, T., Zeevi, D., Zilberman-Schapira, G., Thaiss, C. A., Maza, O.,
et al. (2014). Artificial sweeteners induce glucose intolerance by altering the gut
microbiota. Nature 514, 181–186. doi: 10.1038/nature13793
Sylvetsky, A. C., Welsh, J. A., Brown, R. J., and Vos, M. B. (2012). Low-calorie
sweetener consumption is increasing in the United States. Am. J. Clin. Nutr. 96,
640–646. doi: 10.3945/ajcn.112.034751
Tremaroli, V., and Backhed, F. (2012). Functional interactions between
the gut microbiota and host metabolism. Nature 489, 242–249.
doi: 10.1038/nature11552
Turnbaugh, P. J., Ley, R. E., Mahowald, M. A., Magrini, V., Mardis, E. R., and
Gordon, J. I. (2006). An obesity-associated gut microbiome with increased
capacity for energy harvest. Nature 444, 1027–1031. doi: 10.1038/nature05414
Uronis, J. M., Muhlbauer, M., Herfarth, H. H., Rubinas, T. C., Jones,
G. S., and Jobin, C. (2009). Modulation of the intestinal microbiota
Frontiers in Physiology | www.frontiersin.org 12 July 2017 | Volume 8 | Article 487
Bian et al. Sucralose Alters the Gut Microbiome
alters colitis-associated colorectal cancer susceptibility. PLoS ONE 4:e6026.
doi: 10.1371/journal.pone.0006026
Wang, L., Zhang, Z. G., Zhang, R. L., Gregg, S. R., Hozeska-Solgot, A., LeTourneau,
Y., et al. (2006). Matrix metalloproteinase 2 (MMP2) and MMP9 secreted
by erythropoietin-activated endothelial cells promote neural progenitor cell
migration. J. Neurosci. 26, 5996–6003. doi: 10.1523/JNEUROSCI.5380-05.2006
Wang, Z., Klipfell, E., Bennett, B. J., Koeth, R., Levison, B. S., Dugar, B., et al. (2011).
Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease.
Nature 472, 57–63. doi: 10.1038/nature09922
Willing, B. P., Dicksved, J., Halfvarson, J., Andersson, A. F., Lucio, M., Zheng,
Z., et al. (2010). A pyrosequencing study in twins shows that gastrointestinal
microbial profiles vary with inflammatory bowel disease phenotypes.
Gastroenterology 139, 1844–1854. doi: 10.1053/j.gastro.2010.08.049
Winson, M. K., Camara, M., Latifi, A., Foglino, M., Chhabra, S. R., Daykin,
M., et al. (1995). Multiple N-acyl-L-homoserine lactone signal molecules
regulate production of virulence determinants and secondary metabolites
in Pseudomonas aeruginosa. Proc. Natl. Acad. Sci. U.S.A. 92, 9427–9431.
doi: 10.1073/pnas.92.20.9427
Xavier, R. J., and Podolsky, D. K. (2007). Unravelling the pathogenesis
of inflammatory bowel disease. Nature 448, 427–434. doi: 10.1038/nature
06005
Conflict of Interest Statement: The authors declare that the research was
conducted in the absence of any commercial or financial relationships that could
be construed as a potential conflict of interest.
Copyright © 2017 Bian, Chi, Gao, Tu, Ru and Lu. This is an open-access article
distributed under the terms of the Creative Commons Attribution License (CC BY).
The use, distribution or reproduction in other forums is permitted, provided the
original author(s) or licensor are credited and that the original publication in this
journal is cited, in accordance with accepted academic practice. No use, distribution
or reproduction is permitted which does not comply with these terms.
Frontiers in Physiology | www.frontiersin.org 13 July 2017 | Volume 8 | Article 487

Supplementary resource (1)

... There are several scientific studies and systematic reviews reporting an association between significant alterations in the gut microbiome and LNCS consumption (Schiffman & Nagle, 2019). One example is the artificial sweetener sucralose, which has been reported to produce dysbiosis even at safe levels approved by regulatory agencies (Bian et al., 2017). Several studies (Bian et al., 2017;Frankenfeld et al., 2015;Ruiz-Ojeda et al., 2019;Schiffman & Rother, 2013;Suez et al., 2014;Uebanso et al., 2017) based on the animal model (with rats) as used by the FDA (Abou-Donia et al., 2008) showed an association between the consumption of sucralose with a generalized reduction of gut bacteria, affecting to a greater extent to the beneficial bacteria such as Lactobacillus and Bifidobacterium, compared to pathogenic bacteria such as enterobacteria. ...
... One example is the artificial sweetener sucralose, which has been reported to produce dysbiosis even at safe levels approved by regulatory agencies (Bian et al., 2017). Several studies (Bian et al., 2017;Frankenfeld et al., 2015;Ruiz-Ojeda et al., 2019;Schiffman & Rother, 2013;Suez et al., 2014;Uebanso et al., 2017) based on the animal model (with rats) as used by the FDA (Abou-Donia et al., 2008) showed an association between the consumption of sucralose with a generalized reduction of gut bacteria, affecting to a greater extent to the beneficial bacteria such as Lactobacillus and Bifidobacterium, compared to pathogenic bacteria such as enterobacteria. These alterations caused histological damage at the level of the intestinal epithelium. ...
... In addition, the recovery period of the microbiota bacteria levels at the pre-study level was 12 weeks (Abou-Donia et al., 2008). Other studies obtained similar results on the sucralose effect and alteration in stool bacterial counts (Bian et al., 2017). In addition to altering bacterial counts, proinflammatory bacterial genes were also increased (Uebanso et al., 2017). ...
... In mice, the regular intake of sweeteners for 12 weeks led to a lower abundance and diversity of small intestinal microbiota and significantly increased lymphocytes in Peyer's patches and the lamina propria (Martínez-Carrillo et al. 2019). Moreover, proinflammatory gene expression in the liver associated with GM dysbiosis was observed after a 6-month period of regular sucralose consumption, even at the ADI for humans (Bian et al. 2017;Ruiz-Ojeda et al. 2019). Bian et al. (2017) also reported that sucralose caused liver inflammation when it was consumed regularly. ...
... Moreover, proinflammatory gene expression in the liver associated with GM dysbiosis was observed after a 6-month period of regular sucralose consumption, even at the ADI for humans (Bian et al. 2017;Ruiz-Ojeda et al. 2019). Bian et al. (2017) also reported that sucralose caused liver inflammation when it was consumed regularly. Such liver inflammation may have been related to the overgrowth of GM proinflammatory bacteria (e.g., Turicibacter) (Dhurandhar et al. 2018). ...
Article
Full-text available
To improve performance and recovery faster, athletes are advised to eat more often than usual and consume higher doses of simple carbohydrates, during and after exercise. Sports energetic supplements contain food additives, such as artificial sweeteners, emulsifiers, acidity regulators, preservatives, and salts, which could be harmful to the gut microbiota and impair the intestinal barrier function. The intestinal barrier plays a critical function in bidirectionally regulation of the selective transfer of nutrients, water, and electrolytes, while preventing at the same time, the entrance of harmful substances (selective permeability). The gut microbiota helps to the host to regulate intestinal homeostasis through metabolic, protective, and immune functions. Globally, the gut health is essential to maintain systemic homeostasis in athletes, and to ensure proper digestion, metabolization, and substrate absorption. Gastrointestinal complaints are an important cause of underperformance and dropout during endurance events. These complications are directly related to the loss of gut equilibrium, mainly linked to microbiota dysbiosis and leaky gut. In summary, athletes must be cautious with the elevated intake of ultra-processed foods and specifically those contained on sports nutrition supplements. This review points out the specific nutritional interventions that should be implemented and/or discontinued depending on individual gut functionality.
... Similarly, Ace-K (E950) is one of the most used synthetic sweeteners due to its: low calorie, high sweetness, and excellent thermal stability (up to 250 °C) [57]. However, studies in vitro found that its consumption could affect cognitive functions by altering neuro-metabolic functions [63] and the perturbation of the gut microbiota [64,65]. Saccharin (E954) is a sweetener widely manufactured around the world in four commercial forms: acid saccharin, sodium saccharin, potassium saccharin, and calcium saccharin. ...
... Sucralose (E955) is industrially prepared to utilize sucrose as the starting material. Although it is widely used in foods and beverages all over the world, its safety is being put in doubt by several studies revealing the gut damage and inflammation observed in animal models [64,66,67]. Also, sucralose together with Ace-K was associated with increased coronary heart disease risk by recent cohort findings [58,59]. ...
... Additionally, sucralose administration for six months influenced the abundance of 14 different taxonomic levels, as well as the regulation of amino acids and chronic inflammation, in C57BL/6 mice [60]. This shows the urgent need for further research to investigate the observed effects on humans. of Clostridium cluster XIVa was affected in mice given 15 mg of sucralose/kg [9]. ...
... Additionally, sucralose administration for six months influenced the abundance of 14 different taxonomic levels, as well as the regulation of amino acids and chronic inflammation, in C57BL/6 mice [60]. This shows the urgent need for further research to investigate the observed effects on humans. ...
Article
Full-text available
Worldwide, the demand for natural and synthetic sweeteners in the food industry as an alternative to refined sugar is increasing. This has prompted more research to be conducted to estimate its safety and effects on health. The gut microbiome is critical in metabolizing selected sweeteners which might affect overall health. Recently, more studies have evaluated the relationship between sweeteners and the gut microbiome. This review summarizes the current knowledge regarding the role played by the gut microbiome in metabolizing selected sweeteners. It also addresses the influence of the five selected sweeteners and their metabolites on GI cancer-related pathways. Overall, the observed positive effects of sweetener consumption on GI cancer pathways, such as apoptosis and cell cycle arrest, require further investigation in order to understand the underlying mechanism.
... Furthermore, sucralose is only minimally absorbed, and thus, most of the sucralose that is ingested remains intact in the colon (Magnuson et al. 2016). One biologically plausible hypothesis supported by accumulating rodent (Abou-Donia et al. 2008;Suez et al. 2014;Bian et al. 2017aBian et al. , 2017bBian et al. , 2017cZhang et al. 2021) and human (Suez et al. 2014;Mendez-Garcia et al. 2022;Suez et al. 2022) data is that LCS may alter the composition and function of gut microbiota, leading to systemic inflammation and metabolic dysregulation (Pepino 2015;Suez et al. 2015;Harrington et al. 2022). For example, consumption of several different LCSs was shown to induce glucose intolerance via effects on gut microbiota in rodents (Suez et al. 2014), and sucralose was recently shown to disturb gut microbiota and impair glycemia in humans (Mendez-Garcia et al. 2022;Suez et al. 2022). ...
Article
Sucralose and acesulfame-potassium consumption alters gut microbiota in rodents, with unclear effects in humans. We examined effects of three-times daily sucralose- and acesulfame-potassium-containing diet soda consumption for 1 ( n = 17) or 8 ( n = 8) weeks on gut microbiota composition in young adults. After 8 weeks of diet soda consumption, the relative abundance of Proteobacteria, specifically Enterobacteriaceae, increased; and, increased abundance of two Proteobacteria taxa was also observed after 1 week of diet soda consumption compared with sparkling water. In addition, three taxa in the Bacteroides genus increased following 1 week of diet soda consumption compared with sparkling water. The clinical relevance of these findings and effects of sucralose and acesulfame-potassium consumption on human gut microbiota warrant further investigation in larger studies. Clinical trial registration: NCT02877186 and NCT03125356.
... While data from long-term randomized controlled trials (RCTs) in humans are lacking [9], findings of mechanistic studies [10] and small, relatively short-term RCTs [3] have also demonstrated that chronic LCS consumption may unfavorably impact cardiometabolic risk factors. LCS are metabolically active and are proposed to act through a variety of potential mechanisms [11] including the disruption of glucose-insulin homeostasis [12][13][14][15], disturbance of gut microbiota [16][17][18], and dysregulation of inflammatory pathways [19][20][21]. However, despite their widespread use among children [22,23], LCS consumption in children with T1D is severely understudied. ...
Article
Full-text available
Low-calorie sweeteners (LCS) are commonly consumed by children with type 1 diabetes (T1D), yet their role in cardiometabolic health is unclear. This study examined the feasibility, acceptability, and preliminary effects of 12 weeks of LCS restriction among children with T1D. Children (n = 31) with T1D completed a two-week run-in (n = 28) and were randomly assigned to avoid LCS (LCS restriction, n = 15) or continue their usual LCS intake (n = 13). Feasibility was assessed using recruitment, retention, and adherence rates percentages. Acceptability was assessed through parents completing a qualitative interview (subset, n = 15) and a satisfaction survey at follow-up. Preliminary outcomes were between-group differences in change in average daily time-in-range (TIR) over 12 weeks (primary), and other measures of glycemic variability, lipids, inflammatory biomarkers, visceral adiposity, and dietary intake (secondary). Linear regression, unadjusted and adjusted for age, sex, race, and change in BMI, was used to compare mean changes in all outcomes between groups. LCS restriction was feasible and acceptable. No between-group differences in change in TIR or other measures of glycemic variability were observed. However, significant decreases in TNF-alpha (−0.23 ± 0.08 pg/mL) and improvements in cholesterol (−0.31 ± 0.18 mmol/L) and LDL (−0.60 ± 0.39 mmol/L) were observed with usual LCS intake, compared with LCS restriction. Those randomized to LCS restriction did not report increases in total or added sugar intake, and lower energy intake was reported in both groups (−190.8 ± 106.40 kcal LCS restriction, −245.3 ± 112.90 kcal usual LCS intake group). Decreases in percent energy from carbohydrates (−8.5 ± 2.61) and increases in percent energy from protein (3.2 ± 1.16) and fat (5.2 ± 2.02) were reported with usual LCS intake compared with LCS restriction. Twelve weeks of LCS restriction did not compromise glycemic variability or cardiometabolic outcomes in this small sample of youth with T1D. Further examination of LCS restriction among children with T1D is warranted.
... Additionally, sucralose administration for six months influenced the abundance of 14 different taxonomic levels, regulation of amino acids, and chronic inflammation in C57BL/6 mice [60]. This shows the urgent need for further research to investigate the observed effects on humans. ...
Preprint
Full-text available
Worldwide, the demand for natural and synthetic sweeteners in the food industry as an alternative to refined sugar is increasing. This prompted more research to estimate its safety and effect on health. The gut microbiome is critical in metabolizing selected sweeteners, which might affect overall health. Recently, more studies evaluated the relationship between sweeteners and the gut microbiome. This review summarizes the current knowledge addressing the role played by the gut microbiome in metabolizing selected sweeteners. It also addresses the influence of the five selected sweeteners or their metabolites on GI cancer-related pathways. Overall, the observed positive effect of sweetener consumption on GI cancer pathways, such as apoptosis and cell cycle arrest, requires further investigation to understand the underlying mechanism. Furthermore, more research is needed to address the main challenges of studying the field, thus advancing it further.
Article
Full-text available
NAFLD is the most common chronic liver disease worldwide, characterized by lipid accumulation in the liver, and usually evolves from steatohepatitis to fibrosis, cirrhosis, or even HCC. Its incidence is rapidly rising in parallel with the increasing prevalence of obesity and metabolic syndrome. Current therapies are limited to lifestyle changes including dietary intervention and exercise, in which dietary modification exerts an important part in losing weight and preventing NAFLD. In this review, we briefly discuss the roles and mechanisms of dietary components including fructose, non-nutritive sweeteners, fat, proteins, and vitamins in the progression or prevention of NAFLD. We also summarize several popular dietary patterns such as calorie-restricted diets, intermittent fasting, ketogenic diets, Mediterranean diets, and dietary approach to stop hypertension diets and compare the effects of low-fat and low-carbohydrate diets in preventing the development of NAFLD. Moreover, we summarize the potential drugs targeting metabolic-related targets in NAFLD.
Article
Brazil is experiencing an increased prevalence of overweight and obesity. To overcome these health problems, several strategies have been implemented, including incentives to reduce sugar intake and new packaging labeling. This has promoted an increase in the use of low or non-caloric sweeteners (LNCS). In this study, the use of LNCS in six Brazilian food groups (non-alcoholic beverages, dairy products, baked goods, confectionery, cereals, and condiments) was investigated through label declarations. Three supermarkets were visited in Belo Horizonte, MG, Brazil. Ten out of the 16 LNCS allowed by the Brazilian legislation were declared. Altogether, among the 441 products included, 17.7% contained at least one LNCS, with an average of 2.21 LNCS per sweetened product. Non-alcoholic beverages (37.2%) and dairy products (29.5%) had the highest LNCS occurrence. Combinations of two, three, four, and seven LNCS were found. Artificial sweeteners represented 87.8% of the declared LNCS, with a higher prevalence of acesulfame-K, followed by cyclamate, and sucralose, respectively. Meanwhile, 53.9% of 78 products containing LNCS also had added sugars, and 70.5% used nutritional claims about reduced sugars and/or calories. This scenario highlights the importance of continuous monitoring of LNCS in foods and beverages as front-of-package labeling is not fully implemented yet.
Article
Full-text available
Background: There is growing recognition of the significance of the gut microbiome to human health, and the association between a perturbed gut microbiome with human diseases has been established. Previous studies also show the role of environmental toxicants in perturbing the gut microbiome and its metabolic functions. The wide agricultural use of diazinon, an organophosphate insecticide, has raised serious environmental health concerns since it is a potent neurotoxicant. With studies demonstrating the presence of a microbiome-gut-brain axis, it is possible that gut microbiome perturbation may also contribute to diazinon toxicity. Objectives: We investigated the impact of diazinon exposure on the gut microbiome composition and its metabolic functions in C57BL/6 mice. Methods: We used a combination of 16S rRNA gene sequencing, metagenomics sequencing, and mass spectrometry-based metabolomics profiling in a mouse model to examine the functional impact of diazinon on the gut microbiome. Results: 16S rRNA gene sequencing revealed that diazinon exposure significantly perturbed the gut microbiome, and metagenomic sequencing found that diazinon exposure altered the functional metagenome. Moreover, metabolomics profiling revealed an altered metabolic profile arising from exposure. Of particular significance, these changes were more pronounced for male mice than females. Conclusions: Diazinon exposure perturbed the gut microbiome community structure, functional metagenome, and associated metabolic profiles in a gender-specific manner. These findings may provide novel insights regarding perturbations of the gut microbiome and its functions as a potential new mechanism contributing to diazinon neurotoxicity and, in particular, its sex-selective effects.
Article
Full-text available
Unlabelled: The most widely used and characterized experimental model of ulcerative colitis (UC) is the epithelial erosion, dextran sulfate sodium (DSS)-induced colitis, which is developed by administration of DSS in drinking water. We investigated fecal and colonic mucosa microbial composition and functional changes in mice treated with DSS. C57Bl/6 mice received 5% DSS in drinking water for 5 days. Inflammation was evaluated clinically and by analysis of colonic tissue cytokine levels and C-reactive protein (CRP) in the serum. Colonic mucosa and fecal samples were used for DNA extraction and the V4 region of bacterial 16S rRNA gene was subjected to MiSeq Illumina sequencing. Alpha- and beta-diversities, and compositional differences at phylum and genus levels were determined, and bacterial functional pathways were predicted. DSS increased disease severity, serum CRP and cytokines IL-1β and IL-6, but decreased bacterial species richness, and shifted bacterial community composition. Bacteroides, Turicibacter, Escherichia, Clostridium, Enterobacteriaceae, Clostridiaceae, Bacteroidaceae, Bacteroidales, among other taxa were associated with DSS treatment in fecal and colonic samples. Also, DSS altered microbial functional pathways in both colonic mucosa and fecal samples. Conclusions: The development of colitis in DSS model was accompanied with reduced microbial diversity and dysbiosis of gut microbiota at lower taxonomical levels.
Article
Full-text available
The characterization of phylogenetic and functional diversity are key elements in the analysis of microbial communities. Amplicon-based sequencing of marker genes, such as 16S rRNA, is a powerful tool for assessing and comparing the structure of microbial communities at a high phylogenetic resolution. Because 16S rRNA sequencing is more cost-effective than whole metagenome shotgun sequencing, marker gene analysis is frequently used for broad studies that involve a large number of different samples. However, in comparison to shotgun sequencing approaches, insights into the functional capabilities of the community get lost when restricting the analysis to taxonomic assignment of 16S rRNA data. Tax4Fun is a software package that predicts the functional capabilities of microbial communities based on 16S rRNA datasets. We evaluated Tax4Fun on a range of paired metagenome/16S rRNA datasets to assess its performance. Our results indicate that Tax4Fun provides a good approximation to functional profiles obtained from metagenomic shotgun sequencing approaches. Tax4Fun is an open-source R package and applicable to output as obtained from the SILVAngs web server or the application of QIIME with a SILVA database extension. Tax4Fun is freely available for download at http://tax4fun.gobics.de/. kasshau@gwdg.de. © The Author(s) 2015. Published by Oxford University Press.
Article
Full-text available
Non-caloric artificial sweeteners (NAS) are among the most widely used food additives worldwide, regularly consumed by lean and obese individuals alike. NAS consumption is considered safe and beneficial owing to their low caloric content, yet supporting scientific data remain sparse and controversial. Here we demonstrate that consumption of commonly used NAS formulations drives the development of glucose intolerance through induction of compositional and functional alterations to the intestinal microbiota. These NAS-mediated deleterious metabolic effects are abrogated by antibiotic treatment, and are fully transferrable to germ-free mice upon faecal transplantation of microbiota configurations from NAS-consuming mice, or of microbiota anaerobically incubated in the presence of NAS. We identify NAS-altered microbial metabolic pathways that are linked to host susceptibility to metabolic disease, and demonstrate similar NAS-induced dysbiosis and glucose intolerance in healthy human subjects. Collectively, our results link NAS consumption, dysbiosis and metabolic abnormalities, thereby calling for a reassessment of massive NAS usage.
Article
Full-text available
Membrane biofouling remains a severe problem to be addressed in wastewater treatment systems affecting reactor performance and economy. The finding that many wastewater bacteria rely on N -acyl homoserine lactone-mediated quorum sensing to synchronize their activities essential for biofilm formations; the quenching bacterial quorum sensing suggests a promising approach for control of membrane biofouling. A variety of quorum quenching compounds of both synthetic and natural origin have been identified and found effective in inhibition of membrane biofouling with much less environmental impact than traditional antimicrobials. Work over the past few years has demonstrated that enzymatic quorum quenching mechanisms are widely conserved in several prokaryotic organisms and can be utilized as a potent tool for inhibition of membrane biofouling. Such naturally occurring bacterial quorum quenching mechanisms also play important roles in microbe-microbe interactions and have been used to develop sustainable nonantibiotic antifouling strategies. Advances in membrane fabrication and bacteria entrapment techniques have allowed the implication of such quorum quenching bacteria for better design of membrane bioreactor with improved antibiofouling efficacies. In view of this, the present paper is designed to review and discuss the recent developments in control of membrane biofouling with special emphasis on quorum quenching bacteria that are applied in membrane bioreactors.
Article
Full-text available
We evaluated the protective effects of fermented dairy products (FDPs) in an infection model, using the mouse pathogen Citrobacter rodentium (CR). Treatment of mice with FDP formulas A, B, and C or a control product did not affect CR colonization, organ specificity, or attaching and effacing lesion formation. Fermented dairy product A (FDP-A), but neither the supernatant from FDP-A nor β-irradiated (IR) FDP-A, caused a significant reduction in colonic crypt hyperplasia and CR-associated pathology. Profiling the gut microbiota revealed that IR-FDP-A promoted higher levels of phylotypes belonging to Alcaligenaceae and a decrease in Lachnospiraceae (Ruminococcus) during CR infection. Conversely, FDP-A prevented a decrease in Ruminococcus and increased Turicibacteraceae (Turicibacter). Importantly, loss of Ruminococcus and Turicibacter has been associated with susceptibility to dextran sodium sulfate–induced colitis. Our results demonstrate that viable bacteria in FDP-A reduced CR-induced colonic crypt hyperplasia and prevented the loss of key bacterial genera that may contribute to disease pathology.
Article
Full-text available
The human intestine is host to an enormously complex, diverse, and vast microbial community, the gut microbiota. The gut microbiome plays a profound role in metabolic processing, energy production, immune and cognitive development, epithelial homeostasis, and so forth. However, the composition and diversity of the gut microbiome can be readily affected by external factors, which raises the possibility that exposure to toxic environmental chemicals leads to gut microbiome alteration or dysbiosis. Arsenic exposure affects large human populations worldwide and has been linked to a number of diseases, including cancer, diabetes and cardiovascular disorders. We investigated the impact of arsenic exposure on the gut microbiome composition and its metabolic profiles. We used an integrated approach combining 16S rRNA gene sequencing and mass spectrometry-based metabolomics profiling to examine the functional impact of arsenic exposure on the gut microbiome. 16S rRNA gene sequencing discovered that arsenic significantly perturbed the gut microbiome composition in C57BL/6 mice following exposure to 10 ppm arsenic for 4 weeks in drinking water. Moreover, metabolomics profiling revealed a concurrent effect, with a number of gut-flora-related metabolites being perturbed in multiple biological matrices. These data clearly illustrated that arsenic exposure not only alters the gut microbiome community at the abundance level, but also substantially disturbs its metabolic profiles at the function level. These findings may provide novel insights regarding perturbations of the gut microbiome and its functions as a potential new mechanism by which arsenic exposure leads to or exaggerates human diseases.
Article
Antibiotic resistance, reported for sulfonamides in the mid-1930s and for penicillins in the 1940s, remains a stubborn quandary. What was once confined mainly to hospitals increasingly involves multidrug resistance that encompasses communities and encircles the globe. Virtually all types of bacterial infections are becoming resistant to antibiotic treatments, according to officials at the Centers for Disease Control and Prevention in Atlanta, Ga. Yet, despite decades of grappling with these issues, we still do not understand fully how genes carrying resistance traits spread, what makes certain species highly promiscuous in transferring those traits, whether there are effective barriers to their spread, and the frequency with which resistance genes move independently or in tandem with other migrating genes.
Article
STAMP is a graphical software package that provides statistical hypothesis tests and exploratory plots for analysing taxonomic and functional profiles. It supports tests for comparing pairs of samples or samples organized into two or more treatment groups. Effect sizes and confidence intervals are provided to allow critical assessment of the biological relevancy of test results. A user-friendly graphical interface permits easy exploration of statistical results and generation of publication-quality plots. Availability and implementation: STAMP is licensed under the GNU GPL. Python source code and binaries are available from our website at: http://kiwi.cs.dal.ca/Software/STAMP Contact: donovan.parks{at}gmail.com Supplementary information: Supplementary data are available at Bioinformatics online.