Pradhan Et Al. 2018

You might also like

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

Probiotics and Antimicrobial Proteins

https://doi.org/10.1007/s12602-018-9436-5

Probiotics L. acidophilus and B. clausii Modulate Gut Microbiota in Th1-


and Th2-Biased Mice to Ameliorate Salmonella Typhimurium-Induced
Diarrhea
Biswaranjan Pradhan 1,2 & Dipanjan Guha 1 & Aman Kumar Naik 1 & Arka Banerjee 1,3 & Subodh Tambat 4 &
Saurabh Chawla 1 & Shantibhusan Senapati 5 & Palok Aich 1

# Springer Science+Business Media, LLC, part of Springer Nature 2018

Abstract
Gut microbiota play important role in maintaining health. Probiotics are believed to augment it further. We aimed at comparing effects
of probiotics, Lactobacillus acidophilus (LA) and Bacillus clausii (BC) (a) on the gut microbiota abundance and diversity and (b) their
contributions to control intestinal dysbiosis and inflammation in Th1- and Th2-biased mice following Salmonella infection. We report
how could gut microbiota and the differential immune bias (Th1 or Th2) of the host regulate host responses when challenged with
Salmonella typhimurium in the presence and absence of either of the probiotics. LA was found to be effective in ameliorating the
microbial dysbiosis and inflammation caused by Salmonella infection, in Th1 (C57BL/6) and Th2 (BALB/c)-biased mouse. BC was
able to ameliorate Salmonella-induced dysbiosis and inflammation in Th2 but not in Th1-biased mouse. These results may support
probiotics LA as a treatment option in the case of Salmonella infection.

Keywords Probiotics . Microbiota . Dysbiosis . Inflammation . Lactobacillus . Salmonella

Introduction diversity of microbiota present in a host, gut microbiome is con-


sidered as the second genome, while gut is assumed as the sec-
It has been reported that microbiota and probiotics play important ond brain [2]. Gut microbiota, therefore, has a special place as-
role in improving health and protecting against bacterial over- sociated with the health of the host. Reports are available that has
growth and infection [1]. Because of high abundance and a large aimed at understanding interactions between microorganisms
and host to protect against pathogenic infection by (a) controlling
Electronic supplementary material The online version of this article the overgrowth of the pathogen and pacifying pathogen-
(https://doi.org/10.1007/s12602-018-9436-5) contains supplementary produced toxins [3, 4] and (b) by restoring epithelial integrity
material, which is available to authorized users.
to prevent invasion of the gut mucosa by pathogens [3, 4]. The
mutualistic partnership between microbiota and the host begins
* Palok Aich
palok.aich@niser.ac.in at birth and stabilizes in humans at around the age of 3 [5, 6].
However, change in the composition of gut microbiota
1
School of Biological Sciences, National Institute of Science (dysbiosis) was reported in subjects taking antibiotics [7], infect-
Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, ed with a pathogen [8], consuming high calorie diet [9], suffering
Jatni, Khurdha, Odisha 752050, India
from type II diabetes [9], gastrointestinal cancer [10], and inflam-
2
Present address: S. K. Dash Center of Excellence of Biosciences and matory bowel syndrome [11]. We wanted to understand (a) how
Engineering & Technology (SKBET), Indian Institute of Technology
does abundance and diversity of microbiome changes upon
Bhubaneswar, Bhubaneswar, Odisha, India
3
Salmonella challenge and (b) the ability of selected probiotics
Present address: Biozentrum der Universität Basel, 50-70
in clearing pathogen infection and improving health [12, 13].
Klingelbergstrasse, 4056 Basel, Switzerland
4
We used Salmonella enterica serovar typhimurium (ST) for chal-
Bionivid Technology Private Limited, 209, 4th Cross Rd, B
lenge model [14]. ST is the commonly infecting pathogens that
Channasandra, East of NGEF Layout, Kasturi Nagar,
Bengaluru, Karnataka 560043, India can efficiently induce destabilization of gut microbiota popula-
5 tions and cause diarrhea and populate liver. Despite knowing the
Institute of Life Sciences, Nalco Square, Chandrasekharpur,
Bhubaneswar, Odisha 751023, India etiology of Salmonella typhimurium-induced diarrhea, an
Probiotics & Antimicro. Prot.

effective prevention is yet to be developed. A few probiotic samples using species-specific primers for 3 days. Eating and
bacteria were reported to alleviate microbial dysbiosis and main- drinking pattern, normal cage activities, and body temperature
tain metabolic and immunological homeostasis in the gut of mice were recorded every day until any abnormality ob-
[15–17]. Current group screened four different bacterial strains served. Based on histopathological analysis bacterial dose was
that were either probiotic or potential probiotics and prescribed determined for the further experiments. All experiments were
widely by clinicians for supplementary therapy [18]. Out of four repeated twice to ensure consistency.
strains, we reported that probiotics, Lactobacillus acidophilus
MTCC10307 (LA) and Bacillus clausii MTCC8326 (BC) have
the ability to prime mouse and human macrophages [18]. Out of Mice Survivability
four potential probiotic strains LA, BC, Bifidobacterium bifidum
(BF), and Saccharomyces boulardii (SB) that we tried in vitro in Following a dose titration, the doses selected for
murine macrophages, only LA and BC could alleviate probiotics treatment were 2 × 10 9 CFU and 2 × 10 7
Salmonella typhimurium MTCC3232 (ST)-induced toxicity effi- CFU for LA and BC, respectively. Similarly, for ST,
ciently in vitro [18]. In this report, we evaluated (a) effects of ST the dose selected for infection was 5 × 108 CFU. Mice
challenge in gut microbiome perturbation in C57BL6 and were divided into six groups (n = 6 to 9) and orally
BALB/c mice, (b) role of LA and BC in (i) restoring gut micro- gavaged with the bacteria as described in the Table S1
biota of Th1-biased C57BL/6, and Th2-biased BALB/c mice in (Supplementary Material). The disease symptoms and
vivo, (ii) clearing ST infection from the streptomycin pretreated mortality were noted and plotted to get the survivability
and untreated mice. plot.

Materials and Methods Mice Treatment and Sample Collection

Bacterial Dose Titration in Mice On day 0 (D0), twelve (12) 7-week-old BALB/c or C57BL/6
mice for each treatment condition were treated with either LA
All the animals, used during the present study, were housed in or BC alone or along with ST. A booster dose of LA or BC
polysulfone cages with corncob bedding in a controlled envi- was given to respective LA or BC treated mice on day 1 (D1).
ronment with temperature and humidity ranging between 24 A positive control for infection with ST alone (n = 3) and no
± 3 °C and 40–70% under disease-free conditions. Mice of treatment mice (n = 3 per time point) for each time point was
two strains C57BL/6 (Th1-biased) and BALB/c (Th2- also studied. Mortality or morbidity as a symptom of infection
biased) were co-housed. Since, mice are coprophagous, co- was observed from day 3 (D3) post-ST challenge. Samples for
housing is needed to ensure that the differences observed, in various studies were collected on D3, D5, and D10 posttreat-
gut microbiota between C57BL/6 (Th-1 biased) and BALB/c ment or challenge following euthanasia of animals. Animals
(Th-2 biased) animals, was truly intrinsic of the mouse strains were euthanized by cervical dislocation as per standard oper-
used and not an artifact [19]. All animal experiments were ating procedures approved by Institutional Animal Ethics
done as per the guidelines of CPCSEA (Committee for the Committee. Based on the bacterial dose titration data, sample
Purpose of Control and Supervision of Experiments on collection days were planned. Duodenum, jejunum, ileum,
Animals, Govt. of India) and all protocols were approved by colon, spleen, liver, blood, and fecal samples were collected
the Institute Animal Ethics Committee constituted by from each of the mice after euthanization. Detailed treatment
CPCSEA. Animal House Facility of School of Biological plans and sample collection are depicted in Fig. S2. Another
Sciences, NISER, Bhubaneswar, India is governed and regu- batch of mice of 7 weeks of age are divided into different
lated by CPCSEA rules. Animals were exposed to 12-h light groups (n = 3) and treated with 7.5 mg/mouse of streptomycin
and 12-h dark cycle as a standard practice. Mice of 7 weeks of (Himedia, India) or treated with 20 mg/mouse of streptomy-
age were divided into five groups (n = 3) for each bacterial cin. A group (n = 6) of untreated mice kept as the time-
dose titration. LA, BC, and ST were collected at log phase matched absolute control. Another group (n = 6) of mice treat-
and orally gavaged with 250 μl of saline water at the doses ed with 20 mg/mouse of streptomycin and not treated/
of 2 × 106, 2 × 107, 2 × 108, and 2 × 109 number of bacteria to challenged with probiotics or ST is kept as streptomycin con-
their respective group of mice on day zero. One group of mice trol. After 24 h, the mice were treated with the probiotics or
was administered with saline water only as the absolute neg- challenged with Salmonella. The next day, a booster dose of
ative control. Fresh fecal samples were collected once per day probiotics was orally gavaged to the respective group of mice.
from each mouse and analyzed for the presence of adminis- The mice were then observed for the disease symptoms and
tered bacteria by culturing on nutrient agar plates as well as by sacrificed to collect samples for histology and fecal microbial
PCR amplification of the genomic DNA isolated from fecal analysis.
Probiotics & Antimicro. Prot.

RNA Isolation from Gut Wall Tissue (Distal Ileum normalized fold change values for all genes present on the
and Proximal Colon, n = 3 per Group) microarray slides using Arraypipe (v2.0) [21]. Differentially
regulated genes under various conditions were functionally
RNA isolation was done using the Rneasy mini kit (Qiagen, clustered using WEB-based GEne SeT AnaLysis Toolkit [22].
Germany) with minor modifications in the manufacturer’s
protocol. Briefly, 20 mg of tissue was gently homogenized
Genomic DNA Isolation from the Gut Content (n = 3,
with 600 μl of buffer RLT in a homogenizer (Himedia,
Technical Replicate 1–2)
India). The homogenate was centrifuged for 5 min at
5000 rpm in 4 °C to remove the debris. The supernatant was
Genomic DNA was isolated from the gut contents using the
passed through the Qiashredder (Qiagen, Germany) at
protocol standardized in our lab. In brief, 200 mg of gut con-
10000 rpm for 2 min. Six hundred microliters of 70% ethanol
tent from − 80 °C freezer was taken in a 2-ml sterile microfuge
was added to the clear lysate and mixed well by pipetting. The
tube and 1 ml of sterile PBS is added to it. The sample was
solution was again centrifuged at 5000 rpm for 5 min to re-
mixed properly by vortexing and pipetting. It is then centri-
move the pellets formed. The clear lysate-alcohol solution was
fuged at 4000 rpm for 5 min. To the pellet, 600 μl of lysis
passed through the Rneasy mini column (Qiagen, Germany)
buffer (Tris-HCl 0.1 M, EDTA 20 mM, NaCl 100 mM, 4%
for RNA binding to the silica column. The column was then
SDS) was added and mixed properly by pipetting. The mix-
washed with RW1 and RPE buffer to remove lipids, protein,
ture was kept in 70 °C for 30 min with intermittent mixing by
and DNA. RNA was then eluted from the matrix with 50 μl
inverting the microfuge tube several times. The lysate was
nuclease-free water. RNA quantity and quality was accessed
centrifuged at 13000 rpm in 4 °C for 15 min. The supernatant
with Nanodrop 2000 (Thermo Scientific, USA) and
was collected in a new 2-ml microfuge tube. To the clear
Bioanalyzer2100 (Agilent, USA).
lysate, 1 ml of phenol-chloroform-isoamyl alcohol (24:24:1)
was added and mixed properly by inverting several times. The
Reverse Transcription and qRT-PCR
mixture was centrifuged at 13000 rpm in 4 °C for 15 min. The
colorless upper aqueous layer was collected in a new
cDNA was synthesized from RNA as per the manufacturer’s
microfuge tube. Chilled absolute ethanol was added to the
instruction using AffinityScript One-Step RT-PCR Kit
clear lysate and gently mixed by inverting and rotating simul-
(Agilent, USA). qRT-PCR was done using the protocol de-
taneously to precipitate gDNA. The precipitate was then
scribed earlier [20]. The experiment was done with three tech-
pelleted by centrifugation at 13000 rpm in 4 °C for 15 min.
nical replicates along with no-template-control and no-primer-
The pellet was washed twice with 70% ethanol, dried at room
control.
temperature, and solubilized with 100 μl of nuclease-free wa-
ter. RNase treatment was given to the nucleic acid, and the
Microarray Experiment and Data Analysis (n = 3,
ethanol precipitation step was repeated to recover the micro-
Technical Replicate 1)
bial gDNA. The quality and quantity of the gDNA were
accessed with Nanodrop2000 and agarose gel electrophoresis.
RNA samples for gene expression analysis were labeled using
Agilent Quick-Amp labeling Kit (p/n5190-0444 Agilent,
USA). Time-matched control and treated samples of RNA Species-Specific Primer Designing
(500 ng each) containing RNA spikes were incubated with
reverse transcription mix primed by oligodT with a T7 poly- We designed species-specific primers against 16s rRNA gene
merase promoter site at 40 °C for 2 h to synthesize cDNA. to confirm the next generation sequencing data. Sequences of
cRNA was generated by in vitro transcription incorporating 16s rRNA of the species of interest were collected from NCBI
Cy3 labeled CTP in time-matched untreated control samples nucleotide database, aligned with the 16s rRNA database of
and Cy5 labeled CTP in test samples. Labeled cRNA samples bacteria and archaea using BLAST, top 25 hits are taken and
were cleaned and aliquots of the samples were assessed for aligned in multiple sequence alignment software MUSCLE.
yield and specific activity. Eight hundred nanograms of each In the HTML output format of MUSCLE, unique bases in the
of Cy3- and Cy5-labeled cRNA sample was mixed, sequence of interest were searched. We fixed the unique base
fragmented, and hybridized onto mouse GE 4x44k microarray G or C in the sequence of interest as the 3’-OH base and
slides. The hybridized slides were washed using Agilent Gene extended towards the 5′ end till the melting temperature
Expression wash buffers (Part Number 5188–5327, Agilent, (Tm) reaches 55 °C (Table S2). We did not fix the length of
USA). Slides were scanned to obtain images. Scanned images the primer constant, as done by many of the online primer
were uploaded in Feature Extraction software (Version 10.7, designing software. The primers are then optimized for their
Agilent, USA) to obtain normalized intensity for each spot efficiency and specificity through qRT-PCR taking five differ-
corresponding to a gene on the slide. We further obtained ent dilutions of templates.
Probiotics & Antimicro. Prot.

Histopathological Study bacterial diversity was identified by calculating alpha diversity


index using a EstimateS [30] tool.
Tissue samples were collected and preserved in 4% parafor-
maldehyde (PFA) buffer solution at room temperature for Statistical Analysis
minimum 72 h. For histopathological analysis, tissues were
processed for paraffin embedding, and multiple 5-μm sections Two-way ANOVA was used to calculate the level of signifi-
were prepared. For hematoxylin and eosin staining, slides cance of different treatment groups with respect to control
were deparaffinized and hydrated with deionized water groups as well as among various groups. A single asterisk
followed by hematoxylin (Sigma) staining for 3 min and eosin (*) corresponds to p ≤ 0.05, double aterisks (**) correspond
for 2 min. Slides were thoroughly washed in H2O and to p ≤ 0.01, triple asterisks (***) correspond to p ≤ 0.001, and
dehydrated through sequential alcohol grading, then cleared Bns^ corresponds to not significant. Error bars shown are stan-
in xylene and mounted with permanent mounting media dard deviations determined from average values of at least
(Vector Lab). Stained slides were observed under a Leica three replicates. All graphs were prepared and statistical anal-
DM500 light microscope, and representative images were tak- ysis was done using GraphPad Prism (V5.04, Prism, USA).
en at × 4, × 10, and × 40 magnifications. Images were random-
ly selected from several slides under microscope that represent Shannon-Weiner Index (H)
the area of interest clearly and accurately without much back-
ground or unwanted surrounding effects. Histological images Shannon’s index for diversity was calculated based on the
were scored as per the method explained in the Table S6. abundance value of gut microbial species in different taxo-
nomic categories [31]. Shannon-Weiner Index (H) is calculat-
ed by using the formula as follows:
16S rRNA Gene Profiling Through V3–V4 Sequencing
by NGS n
H ¼ ∑ pi lnpi ;
Stringent quality control of the Illumina reads was performed i¼1

using NGS QC toolkit to estimate the base call errors. Further, where pi is the proportion of S made up of the ith species. S is
high-quality reads from all the samples were merged and sub- total number of species in the community (aka richness).
jected to de-replication and de-noising. The de-replicated H stands for an index of diversity and EH denotes equita-
reads were processed for clustering of OTU’s followed by bility (or evenness) indicating the diversity of the microbial
chimera filtering. The non-redundant and representative community. EH is defined as
OTUs were annotated up to species level followed by individ-
ual sample quantitation. All the above analysis was performed

using UPARSE pipeline [23]. Operational taxonomic units EH ¼ H H max
;
(OTUs) were determined at a threshold of a phylum (80%),
class (90%), order (92%), genus (95%), and species (97%) where Hmax = lnS.
level. For our samples, we had performed the analysis with
different tools, such as QIIME [24] and UPARSE [23]. As
UPARSE is giving bacterial taxonomy up to genus level, we Results
subjected our samples further to get species-level classifica-
tion with MG-RAST [25] tool. OTUs along with their abun- Selection of Probiotics
dance in each sample were parametrically annotated with day
parameters, such as zero, three, five, and ten, and condition Previously, our group screened four potential probiotic strains
parameters, such as NT, ST, STLA, STBC, LA, and BC with (that are most commonly prescribed) LA, BC, BF, and SB in
each of the parameter being representing at least three samples vitro with respect to their cytotoxicity to the murine and hu-
as criteria for statistical analysis. OTUs abundance along with man macrophage/monocyte cells and their potential to induce
parameters was subjected to statistical and multivariate anal- innate immune gene expression [18]. BF was found to be
ysis using METAGENEassist [26]. Unsupervised hierarchical cytotoxic to the RAW 264.7 cells above the multiplicity of
clustering of OTUs based on parameters was done by apply- infection (MOI) of 10 and SB was unable to induce expression
ing Pearson un-centered algorithm with average linkage rule of select innate immune genes even at an MOI of 100 [18]. LA
using a cluster 3.0 software [27] and visualized using a Java and BC imparted insignificant cytotoxicity to the macrophage
Tree View software [28]. OTUs enrichment analysis was per- cells and were able to induce the expression of select innate
formed independently by plotting rarefaction curves for all the immune genes that helped to clear Salmonella infection in
samples using a MEGAN [29] tool. Parameter specific vitro [18]. LA and BC primed macrophages were significantly
Probiotics & Antimicro. Prot.

(p < 0.001) protected against the ST-induced cytotoxicity up lachnospiracea, 8.7% flavonifractor, 6.3% anaerostipes,
to 12-h post-challenge. Therefore, based on previous reports 3.2% alistipes, 2.8% desulfovibrio, and several other low
by our group, LA and BC were selected for the present study abundant genera (Fig. 1a). C57BL/6 microbiota is less di-
[18]. In this report, we studied the efficacy of LA and BC verse, consisting of 54.9% prevotella, 16.7% oscillospira,
against Salmonella challenge in vivo in mouse model. The 12.2% bacteroides, 3.9% ruminococcus, 3.4% odoribacter,
differential immune biasness of BALB/c and C57BL/6 mouse and several other genera in small percentages (Fig. 1b). The
may influence the population structure of the gut microbiota. major phyla that constitute BALB/c gut microbiota are 83.7%
Therefore, we checked the cecal microbial composition of Firmicutes, 11% Bacteroides, and 3.3% Proteobacteria (inset
both types of mouse through 16S rRNA sequencing. to Fig. 1a). Similarly, C57BL/6 microbiota consists of 49.5%
Firmicutes, 46.2% Bacteroides, and 3.4% Spirochaetes (inset
Gut Microbial Composition of BALB/C and C57BL/6 to Fig. 1b). Detailed information on the abundance of gut
Mice Are Different microbiota in BALB/c and C57BL/6 at the level of class,
family, and order is shown in Fig. 1c–e. Clostridia is the major
Genetic makeup and the immunological bias of C57BL/6 and class in the gut microbiota of both the mice models.
BALB/c mice are different. We also observed significant dif- Deferribacteres is the second major class of bacteria present
ferences in gut microbial abundance even after co-housing the in both mice models. However, its abundance is more in the
Th1- and Th2-biased mice. It is expected that differential mi- C57BL/6 mice compared to BALB/c mice. Clostridiales is the
crobiota of these two mice strains may lead to different re- major order present in the gut microbiota of BALB/c. In case
sponses following treatment with LA or BC as well as for of C57BL/6 mice, Clostridiales and Deferribacterales consti-
challenge with ST. Results from 16S rRNA sequencing for tute the majority of the order present in the gut microbiota.
both types of mice of 7 weeks of age are shown in Fig. 1. The major family constituting the gut microbiota of BALB/c
Our results show, at the genus level, the BALB/c microbiota is mice is Lachnospiraceae, whereas in the case of C57BL/6
more diverse, consisting of 39.7% moryella, 14.4% mice the major family is Prevotellaceae. The differential gut

Fig. 1 Gut microbial compositions at genus and phylum levels (inset) for are shown as error bars. Gut microbial compositions of BALB/c and
BALB/c (a) and C57BL/6 (b) mouse are shown. The gut microbial di- C57BL/6 mouse are also represented at the level of class (c), order (d),
versity of BALB/c mouse is higher (Shannon index = 6.75) compared to and family (e)
C57BL/6 mouse (Shannon index = 5.82). Standard deviations of the data
Probiotics & Antimicro. Prot.

microbiota in Th1- and Th2-biased co-housed mouse may be 0.02–2 × 109 CFU of bacteria per mouse. LA is able to colo-
attributed to their immune biasness. The differential gut mi- nize in the BALB/c (Fig. 2a) and C57BL/6 (Fig. 2c) mouse
crobiota may utilize the abundant food sources differently, gut, orally gavaged up to a dose of 2 × 109 CFU/mouse with-
imparting differential colonization of the introduced out any significant adverse effects in the animal. Bacterial
probiotics and pathogen. Our next aim is to see the site and presence in the fecal samples of mice as well as health and
duration of colonization of the treated probiotic strains as well behavior of the mice was tracked for several days to find the
as the challenged pathogenic bacteria. optimal dose of LA and BC. Both LA and BC were able to
colonize in the distal ileum and proximal colon (Fig. 2a–d).
ST, LA, and BC Were Able to Colonize in Distal Ileum However, when LA was being tolerated by the mice up to the
and Proximal Colon of Mice Gut dose of 2 × 109 CFU/mouse, BC-induced bloating (Fig. 2g),
and villi atrophy (Fig. 2h) in mice when administered beyond
The strains of probiotics (LA and BC), as well as pathogen ST, the dose of 2 × 107 CFU/mouse. BC could also colonize in the
were tested in vitro using murine macrophage RAW 264.7 for BALB/c (Fig. 2b) and C57BL/6 (Fig. 2d) mouse gut when
their immune-modulatory and cytotoxic ability. It was report- orally gavaged up to a dose of 2 × 107 CFU of BC. However,
ed that the two strains of LA and BC, that were used, could LA-treated mouse was found to have normal gut morphology
clear ST infection in vitro [18]. Results from the current in (Fig. 2e) and histological structure (Fig. 2f), even at a dose of
vivo ST challenge studies with streptomycin treated and un- 2 × 109 CFU/mouse. We have chosen lower dosage (2 × 107
treated BALB/c and C57BL/6 mice following treatment with CFU/mouse) of BC to work with for further studies, while
or without these strains of LA and BC are reported and kept the higher dosage (2 × 109 CFU/mouse) for LA, since
discussed. Figure 2a–d showed colonization ability of LA equivalent dose (2 × 107 CFU/mouse) of LA did not have
and BC in the gut of BALB/c and C57BL/6 mouse. In both any significant protection against salmonella challenge as de-
mice strains, colonization of LA and BC is significantly higher scribed in the next section. We tracked the health of mice with
in colon and ileum compared to colonization in duodenum respect to their feeding, drinking, grooming, and playful ac-
and jejunum. BALB/c and C57BL/6 mice were treated with tivities. Histology of the gut, spleen, and liver of the mice
LA and BC through oral gavage with doses ranging from about to die was performed to quantify the level of

Fig. 2 Colonization of LA (a, c) and BC (b, d) in mouse gut is shown. like bloating rather intestine look normal (e) and no inflammation in the
LA is able to colonize in the BALB/c (a) and C57BL/6 (c) mouse gut gut as shown by histopathology (f) when treated with 2 × 109 CFU of LA
when orally gavaged at a dose of 2 × 109 CFU/mouse. BC is also able to (e, f). The higher dose of BC (2 × 108) is detrimental to the mouse and
colonize in the BALB/c (b) and C57BL/6 (d) mouse gut when orally induced bloating (g) and inflammation (h) in the mouse gut
gavaged at a dose of 2 × 107 CFU of BC. There was no adverse effect
Probiotics & Antimicro. Prot.

infection-mediated inflammation and tissue damage. Results day 3 and on day 5, posttreatment to find out the kinetics of the
also revealed that ST was able to colonize the mice gut for at infection and curing processes. We also followed the STLA-
least 3-day post-challenge (Fig. S1). ST infection load was treated group of mice until day 10 to find out the degree of
found to be higher in the C57BL/6 mice compared to the clearance of ST from the mouse by the probiotic LA. The
BALB/c mice. The optimal ST infective dose per mouse was detailed study plan is depicted graphically in Fig. S2 and the
determined to be 5 × 108 CFU of bacteria based on mice sur- doses of different bacteria per mice as well as sample collec-
vivability (Fig. 3) and histopathology. In addition, we evalu- tion plan are listed in Table S1. The survivability of the mouse
ated the efficiency of LA at a low (2 × 107 CFU per mouse) was determined following ST challenge and probiotic
and a high dose (2 × 109 CFU per mouse) in ameliorating the treatment.
effects of ST infection in mouse. The data revealed that LA at
the dose of 2 × 107 CFU per mouse is ineffective in protecting LA and BC Protects BALB/c Mice from ST Infection,
the ST infected mouse. But LA at a dose of 2 × 109 CFU per but Only LA (Not BC) Was Able to Protect C57BL/6
mouse is effective in protecting the mouse against ST infec- Mice from ST Infection
tion (Fig. 3b). Therefore, the optimized doses selected for the
further studies were 2 × 109 CFU/mouse for LA and 2 × 107 The rate of mortality of the mouse increased with increasing
CFU/mouse for BC. The symptoms of diarrhea and mortality CFU of ST infection (Fig. 3a). BALB/c mice infected with ST
in the mice have been observed post 4 days of ST infection. are significantly (p < 0.001) protected following treatment
Therefore, the study is designed to sacrifice a group of mice on with LA at CFU of 2 × 10 9, but not at CFU of 2 × 107

Fig. 3 The rate of mortality of the


mouse increased with increasing
CFU of ST infection (a). BALB/c
mice infected with ST is signifi-
cantly (p < 0.001, n = 6) protected
following treatment with LA at a
CFU of 2 × 109, but not at a CFU
of 2 × 107 (b). Similarly, C57BL/
6 mice infected with ST is signif-
icantly (p < 0.001, n = 6)
protected after treatment with LA
at a CFU of 2 × 109, but not at a
CFU of 2 × 107(c). BC could
protect BALB/c mice (b) but not
C57BL/6 mice (c) significantly
(p < 0.001, n = 6) from ST infec-
tion. BALB/c-mice pretreated
with streptomycin at a dose of
7.5 mg/mouse (d) and 20 mg/
mouse (e) is also significantly (p
< 0.001, n = 3) protected from ST
infection after treatment with LA
but not with BC. Similarly,
C57BL/6-mice pretreated with
streptomycin at a dose of 7.5 mg/
mouse (f) and 20 mg/mouse (g) is
also significantly (p < 0.01, n = 3)
protected from ST infection after
treatment with LA but not with
BC
Probiotics & Antimicro. Prot.

(Fig. 3b). Similarly, C57BL/6 mice infected with ST is signif- compared to non-streptomycin model of mouse (Fig. S3).
icantly (p < 0.001) protected against Salmonella challenge fol- This phenomenon is similar in both the Th1 and Th2
lowing treatment with LA at a CFU of 2 × 109, but not at a immune-biased mice strains (Fig. S3).
CFU of 2 × 107 (Fig. 3c). BC can protect BALB/c mice
(Fig. 3b) but not C57BL/6 mice (Fig. 3c) significantly (p < Microbial Dysbiosis in Mice Following Treatment
0.001) against ST challenge. BALB/c mice pretreated with with ST and STBC but Not with STLA
streptomycin at a dose of 7.5 mg/mouse (Fig. 3d) and
20 mg/mouse (Fig. 3e) was also significantly (p < 0.001) Changes in abundance (%) of gut microbiota and diversity on
protected against ST challenge following treatment with LA day 3 and day 5 following various treatment conditions in
but not with BC. Similarly, C57BL/6 mice pretreated with BALB/c and C57BL/6 mice are shown in Fig. 4. Figure 4a,
streptomycin at a dose of 7.5 mg/mouse (Fig. 3f) and b shows changes in BALB/c and changes for C57BL/6 are
20 mg/mouse (Fig. 3g) was also significantly (p < 0.01) shown in Fig. 4c, d. Figure 4e shows kinetics of changes in the
protected against ST challenge following treatment with LA diversity of gut microbiome. It is observed that abundance of
but not with BC. In addition, we compared the pathological bacterial genus Moryella decreased and abundance of
scores of the group of ST infected mice with or without pre- Lachnospirace increased in BALB/c on day 3 and day 5 fol-
treatment with streptomycin. The rate of mortality of the ST- lowing ST challenge (Fig. 4a–b). The abundance of bile me-
infected mice pretreated with streptomycin is higher compared tabolizing Flavonifractor was increased significantly (p ≤
to the non-streptomycin model of ST-infected mice (Fig. 3). 0.01) on day 5 in the BALB/c mice treated with ST and
The pathology score of the ST-infected mouse model with STBC (Fig. 4b). Reduced abundance of Oscillospira and
streptomycin pretreatment is significantly (p ≤ 0.05) higher Prevotella was observed in the microbiota of C57BL/6 mice

Fig. 4 The gut microbial


composition at genus level of
BALB/c mice treated with either
LA or BC at day 3 (a) and at day 5
(b) following challenge with ST.
The gut microbial composition in
genus level of C57BL/6 mice
treated with either LA or BC at
day 3 (c) and at day 5 (d) follow-
ing challenge with ST. Heatmap
for BALB/c and C57BL/6 (e) be-
tween treatment groups is being
made based on weighted unifrac
approach. STLA is grouped along
with untreated (NT) for both mice
models. This implies that LA is
aiding in normalizing the gut
microbiome of the mice disrupted
with ST infection
Probiotics & Antimicro. Prot.

on day 3 posttreatment with ST and STBC in comparison to presence of abovementioned bacteria in the mice gut is listed
time-matched untreated mice (Fig. 4c d). From the species- in Table S2. Together, these data implicated that LA treatment
level analysis of the C57BL/6 gut microbiota, we found that of the ST-infected mice relieved inflammation by stimulating
the short-chain-fatty-acids-producing bacterial species in- SCFA producers in gut microbiota, whereas BC treatment to
creased in the STLA-treated mice group. The bacteria from the ST-infected mice have no such effect. In addition, BC
the genus Moryella and Lachnospirace metabolize complex treatment to ST-infected C57BL/6 mice aids in destabilizing
carbohydrates, and produce short chain fatty acids (SCFAs) the microbiota by stimulating the growth of a few
that maintains gut homeostasis via signaling through anti- inflammation-inducing sulfate reducers. The effect of LA or
inflammatory pathways. Reduced SCFA producers in ST BC administration on infected mice is well defined, to study
and STBC-treated mice indicated the inflammatory condition the effect of LA or BC on host response; we studied genome-
in their gut. This was further validated by gut microbial dis- wide gene expression in the gut wall tissue taken from the
tribution calculated by Shannon diversity index (Fig. 4e). For distal ileum and proximal colon of the mice to access response
both C57BL/6 and BALB/c, STLA treatment increased the to the changed microbiota in the mouse gut. It is important to
gut microbial diversity. This implies that LA is aiding in nor- note that BALB/c and C57BL/6 mice behaved differently in
malizing the gut microbiome of the mice disrupted with ST terms of microbiota rearrangement following treatment with
infection but BC failed to do that. Species-level information LA and BC. For BALB/c, on D3 post LA treatment gut mi-
on day 5 for BALB/c following treatment with STLA revealed crobial composition was similar to that of untreated mice,
significant fold increases (values shown in bracket by the bac- whereas on D5 following LA treatment Lachnospiracea,
teria) with respect to (WRT) untreated time-matched (day 5) Moryella, and Flavonifractor group of bacteria changed sig-
mice in abundance of Butyricicoccus pullicaecorum (6.3- nificantly. BC treatment on the contrary significantly affected
fold), Candidatus arthromitus (41.6-fold), Clostridium abundance of Lachnospiracea and Moryella on both days 3
scindens (41.6-fold), and Faecalibacterium prausnitzii and 5 posttreatment compared to untreated mice. In C57BL/6
(143.2-fold) (Table 1). These significantly increased bacteria on the other genus that were significantly altered compared to
are known to ferment complex carbohydrates, helps in an untreated mice were Oscillospira and Prevotella. Interestingly,
expansion of T-reg cells and produce anti-inflammatory on D3, following treatment with BC composition of microbi-
SCFAs, such as butyrate, propionate, acetate, isobutyrate, ota was more similar to D3 following challenge with ST. Our
isovalerate, and valerate. The presence of all these species in results laid the foundation for BC as less effective in treating
the fecal sample was confirmed through qRT-PCR using dysbiosis following challenge with ST.
species-specific primers designed against 16 s rRNA. The
bacteria that became highly abundant (numbers increased) Gene Expression in the Gut Wall Tissue in the Distal
on day 5 in C57BL/6 mice following STLA treatment are Ileum and Proximal Colon of Treated Mice
Clostridium lepttum (13.7-fold), Ruminococcus gnavus (8.8-
fold), and Acetatifactor muris (6846 folds), and similarly on Gut wall tissue comprised of a mixed population of cells,
day 3, posttreatment Herbinix hemicellulosilytica (17.8) and including intestinal epithelial cells, goblet cells, M cells, en-
Helicobacter typhlonius (6.3) (Table 2). C57BL/6 mice treat- terochromaffin cells, and immune cells, such as macrophages,
ed with STBC have increased numbers of inflammatory- dendritic cells, B cells, T cells, and NKT cells. Gut microbiota
inducing sulfate-reducing bacteria, such as Desulfovibrio and the gut tissues are in continuous crosstalk through the
intestinalis (2.6-fold), and Helicobacter typhlonius (17.4-fold) interaction of various secretory molecules with gut tissue re-
(Table 2). The species-specific primers used to validate the ceptors, and the respective activation of various metabolic and

Table 1 BALB/c microbiota changes in species level

OTU_ID Species name Fold changes wrt time-matched NT mice

D3STLA D3STBC D5STLA D5STBC Impt function

gi_ Butyricicoccus pullicaecorum 2.6 − 3.0 6.3 3.4 Anti-inflammatory


343205971
gi_ Candidatus arthromitus − 2.0 − 1.2 41.6 1.2 Innate immune response
444304121
gi_ Clostridium scindens 1.7 − 1.4 16.7 − 1.8 Expansion of T-reg cells
265678482
gi_ Faecalibacterium prausnitzii − 4.6 − 3.4 143.2 5.8 SCFA, butyrate
265678656
Probiotics & Antimicro. Prot.

Table 2 C57BL/6 microbiota changes in species level

OTU_ID Species name Fold changes wrt time-matched NT mice

D3STLA D3STBC D5STLA D5STBC

denovo9209 Acetatifactor muris 1 1 6846 6 Bioremediation


denovo1769 Ruminococcus gnavus − 2.8 − 1.6 8.8 36.6 SCFA, acetate, butyrate
denovo8429 Herbinix hemicellulosilytica 17.8 1.5 1 2.2 Cellolose metabolism
denovo3701 Mucispirillum schaedleri 1 6 − 2.1 18 Maintain ENS and anti-diabetic
denovo2541 Helicobacter typhlonius 6.3 17.4 −3 3.3 Antiinflammatory
denovo465 Clostridium leptum − 1.6 30.3 13.7 − 44.5 Expansion of T-reg cells

immunological signaling pathways required for gut homeosta- enhanced renal glucose reabsorption or synthesis of complex
sis. Therefore, it is necessary to monitor the gene expression in lipids or long chain fatty acids that might have been compro-
gut wall tissue following its response to microbial perturba- mised as a result of ST. STLA-treated mice also expressed the
tion, rather than analyzing intestinal epithelial cells alone. We toxin-clearing cytochrome p450 gene Cyp26b1 to clear the
extracted total RNA from mice gut wall tissue from the distal toxins that might have been produced as a result of ST chal-
ileum and proximal colon to analyze host gene expression. lenge. Overexpression of Tnf and Il1b in STBC treated mice,
The genes were considered to be differentially expressed if
their fold changes are more than 1.5 with respect to time-
matched untreated controls and if the p value is less than
0.05. A detailed number of differentially expressed genes that
are either conserved or unique following various treatment
and challenge conditions are shown in Venn diagram (Fig.
S4). Following analysis of the microarray data, we have found
that lipopolysaccharide mediated TLR pathway is enriched in
BALB/c mice after 3 days of ST challenge and for C57BL/6
mice after 5 days of ST challenge, which is also validated
through qRT-PCR (Fig. 5). We have observed several proin-
flammatory cytokines like Il1b, Il6, Nfkb1, Tnf gets upregu-
lated in ST challenged mice and downregulated in STLA-
treated mice (Fig. 5a, b). STLA treatment can reduce proin-
flammatory response in C57BL/6 mice compared to ST-
treated mice group. Primer sequences of the genes used for
validation in qRT-PCR are listed in Table S3.
Genes were clustered together based on their function and
the pathways they represent. Several pathways came up fol-
lowing our analysis, and we focused mainly on pathways
associated with immunological, metabolic, and gut barrier
functions. Some of the functionally important genes and their
expression values in terms of fold changes on day 3, treated
groups with respect to time-matched untreated controls of
BALB/c mice are listed in Table 3. Results, from day 3 sug-
gested higher expression of mucin genes (Muc13, Muc16, and
Muc20) and glucose transporters (Slc2a2 and Slc2a7). Results
also revealed a higher expression of Slc27a5 gene that sug-
gested significantly more synthesis of long chain fatty acid Fig. 5 Genes of TLR pathway differentially expressed in mice after
following LA-treated ST-challenged mice compared to LA- 3 days of treatment with ST, STBC, and STLA in BALB/c and C57BL/
untreated ST-challenged mice (Table 3). Upregulation of these 6 mice (c) and after 5 days of treatment with ST, STBC, and STLA in
BALB/c and C57BL/6 mice (b). Note that the higher expression of in-
genes in STLA-treated mice but downregulation in ST chal- flammatory cytokines il1b and tnf in ST infected C57BL/6 mice, which is
lenged mice suggest repair or maintenance of the (a) gut mu- decreased significantly (p < 0.01, n = 3) following treatment with LA
cosa and (b) epithelial tissue barrier function as well as
Probiotics & Antimicro. Prot.

Table 3 Important genes expressed in BALB/c after 3 days of treatment

GeneSymbol Gene Name D3BC D3LA D3ST D3STBC D3STLA


Adh4 Alcohol Dehydrogenase 4 11.6 1.5 4.2 4.7 7.0
Maob Monoamine Oxidase B 2.0 -1.1 1.9 3.1 2.9
UDP Glucuronosyltransferase Family 2
Ugt2a3 Member A3 9.6 1.2 5.4 7.9 6.3
UDP Glucuronosyltransferase Family 1
Ugt1a6a Member A6 -2.8 -1.2 -1.2 -3.0 3.4
Gzmg Granzyme G 1.3 1.7 1.9 1.1 140.8
Slc27a5 Solute Carrier Family 27 Member 5 12.5 4.1 1.8 5.6 5.6
Nme6 NDP Kinase 6 1.2 1.2 1.3 -1.0 124.7
Oas1h 2'-5'-Oligoadenylate Synthetase 1 -1.1 1.0 1.2 1.0 31.0
Protein Phosphatase 1 Regulatory
Ppp1r12b Subunit 12B -5.4 -1.3 -1.3 1.0 29.1
Ptger4 Prostaglandin E Receptor 4 -2.6 1.4 1.4 -1.5 22.6
Slc2a2 Solute Carrier Family 2 Member 2 202.2 -1.2 1.8 40.2 22.1
Slc2a7 Solute Carrier Family 2 Member 7 7.0 6.4 2.4 3.3 15.9
Mpo Myeloperoxidase 1.2 1.2 9.7 -1.1 1.3
Lect2 Leukocyte Cell Derived Chemotaxin 2 5.0 -1.5 7.4 25.6 5.6
C-Type Lectin Domain Family 4
Cd209c Member M -1.0 -1.0 1.4 -1.3 1.2
Hdc Histidine Decarboxylase 2.6 1.9 1.1 7.4 3.1
Trim6 Tripartite Motif Containing 6 5.7 7.6 4.3 3.2 5.6
Cytochrome P450 Family 26 Subfamily
Cyp26b1 B Member 1 4.3 1.2 7.0 8.4 1.8
Muc 5ac Mucin 5Ac -1.4 3.4 -1.1 1.6 2.4
Muc 5B Mucin 5B -1.4 1 -1.3 -1.7 2.7
Muc 13 Mucin 13 -1.8 1.3 -3.4 -1.1 1.8
Muc16 Mucin 16 3.3 1.5 -1.2 3.0 1.8
Muc20 Mucin 20 1.3 2.3 -2.0 1.4 1.9
Tjp1 Tight Junction Protein 1 2.6 2.2 -1.7 -3.0 2.4
Hdac1 Histone deacetylase 1 1.3 1.1 1.5 -1.1 -1.2
Hdac3 Histone deacetylase 3 1.5 -1.0 1.2 1.4 1.5
Hdac6 Histone deacetylase 6 1.9 1.3 1.4 1.5 1.2
Hdac7 Histone deacetylase 7 1.3 -1.3 -1.0 1.2 1.1
Hdac8 Histone deacetylase 8 7.7 1.2 1.2 3.3 -1.0
Hdac9 Histone deacetylase 9 2.6 -2.6 -3.4 1.4 -2.5

but in STLA-treated mice, suggesting induction of a proin- compared with respect to time-matched untreated control
flammatory condition in STBC-treated mice but not in group of C57BL/6 mice is shown in Table 4. The high
STLA (Fig. 5a and Table S4). expression level of the defensins, DEFA4 (42.4 folds) and
Similarly, expression of a few more relevant important DEFA1 (25.1 folds), in STBC, treated mice suggests that
genes and their expression values in terms of fold changes tissue damage occurred in the gut [32]. Il12b and Il17a
on day 3 in mice with various treatment conditions were expression in STLA-treated mice suggest the upregulation
Probiotics & Antimicro. Prot.

Table 4 Important genes expressed in C57BL/6 after 3 days of treatment

GeneSymbol Gene Name D3BC D3LA D3ST D3STBC D3STLA


Defa4 Defensin Alpha 4 -2.1 -1.9 -1.1 42.4 7.7
Defa1 Defensin Alpha 1 2.3 1.0 1.3 25.1 7.6
Tumor Necrosis Factor
Tnfsf4 Superfamily Member 4 -1.8 4.5 7.7 3.1 7.2
Itga5 Integrin Subunit Alpha 5 -5.5 -1.3 3.7 7.0 6.4
Il17f Interleukin 17F 2.4 4.0 1.2 3.1 4.5
Il19 Interleukin 19 2.2 -1.3 1.2 -1.6 4.1
Tlr6 Toll Like Receptor 6 13.2 5.1 2.5 8.9 3.6
Il17a Interleukin 17A 2.3 1.5 -1.3 1.9 3.5
Transforming Growth Factor
Tgfb3 Beta 3 -1.0 1.3 1.3 1.4 2.4
C-C Motif Chemokine Receptor
Ccr4 4 3.3 4.1 4.5 2.0 2.1
C-X-C Motif Chemokine Ligand
Cxcl1 1 1.2 2.2 1.8 -1.8 2.0
Il12b Interleukin 12B -1.1 1.2 -1.1 1.4 1.9
Ctse Cathepsin E 1.7 10.8 5.3 11.6 1.9
Cytotoxic T-Lymphocyte
Ctla4 Associated Protein 4 2.3 1.9 -1.1 2.5 1.7
Csf3 Colony Stimulating Factor 3 3.9 2.5 3.5 1.7 1.7
C3 Complement Component 3 1.3 1.4 1.0 1.7 1.4
Il6 Interleukin 6 1.7 1.6 9.2 1.8 -1.3
Ccl2 C-C Motif Chemokine Ligand 2 -1.1 1.9 -1.1 1.1 1.1
Fcer1g Fc Fragment Of IgE Receptor Ig -1.3 -1.4 -1.2 1.5 -1.1
Cathelicidin Antimicrobial
Camp Peptide 1.4 2.2 -1.1 1.8 -1.1
Il1b Interleukin 1 Beta 1.9 2.0 14.8 2 1.9
Tnf Tumor Necrosis Factor 1.4 1.4 2.2 1.1 1.5
Fpr2 Formyl Peptide Receptor 2 1.0 -2.1 1.2 2.1 -1.2
Ccl7 C-C Motif Chemokine Ligand 7 1.4 2.6 1.4 4.2 -1.3
Muc13 Mucin 13 4.0 5.9 1.2 1.8 2.5
Muc16 Mucin 16 8.0 3.8 -1.9 4.7 6.9
Tjp1 Tight junction protein 1 1.9 2.4 1.2 1.5 3.2
Hdac6 Histone deacetylase 6 1.6 1.6 1.3 1.5 1.0

of proinflammatory signals for pathogen clearance. The S5) was minimal. Together, these data suggested that LA-
requirement of Il17a was reported before to suppress induced anti-inflammation and BC-induced inflammation
Salmonella invasion [33, 34]. Downregulation of Il1b in ST infected mice, findings which are in accordance
and Tnf in STLA-treated mice, but upregulation in with our data on the microbiota. However, the inflamma-
STBC-treated mice suggested the presence of severe in- tory damage to the gut by ST and STBC needs to be
flammation in the STBC group, whereas inflammation further explored for confirmation and establishing the
observed in the STLA group of mice (Fig. 5b and Table mechanism.
Probiotics & Antimicro. Prot.

BALB/C and C57BL/6 Mice Treated with ST and STBC damage, immune infiltration into the tissue, tissue abscises,
Had Intensive Damage to Ileum, Colon, Spleen, and immune infiltrations into the red pulp area of the spleen
and Liver are observed in the BALB/c mice treated with ST and STBC
(Fig. S5 and S6). Few of the representative colon images of
Histopathological scoring of the eosin and hematoxylin the mouse across treatment conditions are presented in Fig. 6a.
(H&E) stained transverse slices of BALB/c and C57BL/6 This was further supported by the higher pathological score of
mice ileum, colon, spleen, and liver were performed using 3.4 ± 0.2, 1.9 ± 0.2, 2.6 ± 0.2, 1.9 ± 0.3 for the ileum, colon,
the method explained in the materials and methods section. spleen, and liver respectively in BALB/c mice treated with ST
The histological images were scored for the level of damage compared to the NT (Fig. 6b). Similarly, a pathological score
based on the scoring method listed in Table S6. Extensive of 3.6 ± 0.1, 2.6 ± 0.2, 2.4 ± 0.4, and 2.1 ± 0.5 for the ileum,

Fig. 6 Representative images of the histology of colon from BALB/c and mentioned in Fig. S2. The pathological scores of the histology images
C57BL/6 mice treated with ST, LA, BC, and their combinations (a). The of different treatment groups were plotted and tested for statistical signif-
histological images (n = 3) were scored for the pathological assessment icance (two-way ANOVA) among the treatment groups. Note that ST and
based on the criteria explained in Table S6 and normalized to the scale STBC treated groups of mice have significantly (p < 0.001) higher pa-
ranging from zero to four. The histology scores of various treatment thology score compared to the NT- and STLA-treated groups of mice
groups of mice are represented graphically (b). The treatment plan is
Probiotics & Antimicro. Prot.

colon, spleen, and liver respectively in BALB/c mice treated via production of SCFA or other routes. Bacterial species,
with STBC suggested an inflammatory state in this group of include Butyricoccus pullicaecorum, Clostridium scindens,
mice (Fig. 6b). Tissues of the abovementioned organs collect- Candidatus arthromitus, and Faecalibacterium prausnitzii,
ed from mice treated with LA, BC, and STLA have lower in STLA-treated BALB/c mice and Clostridium leptum,
pathological scores compared to the ST-treated mice in the Herbinix hemicellulosilytica, Ruminococcus gnavus,
groups (Fig. 6b). Acetatifactor muris, Mucisspirillum schaedleri, and
Extensive damage, immune infiltration into the tissue, tis- Helicobactor typholnis. SCFAs usually induce anti-
sue abscises, and immune infiltrations into the red pulp area inflammatory responses in the intestinal epithelial cells of
are also observed in the C57BL/6 mice treated with ST and mice by inhibiting HDACs, followed by NF-kB suppression
STBC. This was further supported by a higher pathological and downregulation of TNF and IL1B [41]. The rise in SCFA
score of 3.2 ± 0.2, 2.3 ± 0.3, 2.7 ± 0.2, and 3.2 ± 0.3 for the producing bacteria in both mice species following LA treat-
ileum, colon, spleen, and liver respectively in C57BL/6 mice ment might be suggestive of a plausible mechanism by which
treated with ST compared to the NT (Fig. 6b). Similarly, a treatment of mice with LA could alleviate ST-induced inflam-
pathological score of 3.6 ± 0.2, 3.6 ± 0.1, 2.7 ± 0.4, and 2.6 mation in the gut. Such pronounced increase following BC
± 0.6 for the ileum, colon, spleen, and liver respectively in treatment was not evident from the current results. Dysbiosis
C57BL/6 mice treated with STBC suggests an inflammatory was evident in ST and STBC-treated mice with reduced mi-
state in this group of mice (Fig. 6b). Tissues of the crobial diversity compared to untreated mice; however, little
abovementioned organs collected from mice treated with perturbation in STLA-, LA-, and BC-treated mice was ob-
LA, BC, and STLA have lower pathological scores compared served. To validate, we evaluated inflammatory and physio-
to the ST treated mice in the groups (Fig. 6). Microbial logical status in the gut by gene expression analysis using
dysbiosis and inflammatory gene expression in ST- and expression microarray. The inflammatory status of the gut
STBC-treated BALB/c and C57BL/6 mice were further sup- was also visualized through H & E stained transverse sections
ported with significantly higher pathological scores in these of the gut, as well as spleen and liver.
groups of mice with respect to time-matched untreated control
groups of mice. ST and STBC Treated Mice Exhibit Severe
Inflammation in Gut Compared to STLA-Treated Mice

Discussion SCFAs are inhibitors of histone deacetylases (HDACs) and


ligands for G-protein coupled receptors (GPCRs), and thereby
Microbial Diversity and its Resilience to Perturbation act as signaling molecules that influence the expansion and
function of hematopoietic and non-hematopoietic cell line-
It has been reported that microbial diversity is reduced in the ages. SCFA-driven inhibition of − 1 s tends to promote a
gut from the patients suffering from Crohn’s disease [35], tolerogenic, antiinflammatory cell phenotype that is crucial
ulcerative colitis [36], irritable bowel syndrome [37], for maintaining immune homeostasis [42]. Exposure of pe-
Clostridium difficile associated diarrhea [38], and antibiotic- ripheral blood mononuclear cells and neutrophils to SCFAs
associated diarrhea [39]. Diversity in the microbial composi- inhibits HDAC and inactivated nuclear factor–kB (NF-kB),
tion is, therefore, a major factor in maintaining health. Fransen and downregulated production of the proinflammatory cyto-
et al. reported that BALB/c gut microbiota was more diverse kine, tumor necrosis factor (TNF) [42, 43]. We observed acti-
than that of C57BL/6 mice [40], which corroborated with the vation of Hdac1, Hdac3, Hdac6, Hdac7, Hdac8, and Hdac9
current report. We, therefore, propose that because of greater expression by BC and STBC in BALB/c mice followed by
microbial diversity, BALB/c mice might be less vulnerable to activation of Tnfsf10, Tnfsf13b, and Tnfsf14 through NF-kB
perturbation by ST than C57BL/6 mice. Our 16s rRNA pro- pathway leading to inflammation and immune cell infiltration
filing data from the mice gut microbiota suggested that ST in STBC-treated mice. On the contrary, LA-treated BALB/c
perturbed the microbiota of C57BL/6 mice more than that of mice downregulated HDACs and TNFs to induce low inflam-
the BALB/c mice. Surprisingly, the microbial perturbation mation and immune cell infiltration in STLA-treated mice.
was more pronounced in STBC-treated mice than in ST- Upregulated HDACs and TNFs by BC and ST might explain
treated mice. There was little perturbation of microbiota in the synergistic activity of ST and BC with respect to inflam-
STLA-, LA-, and BC-treated mice implying that LA and BC mation in BALB/c mice, whereas HDAC downregulation by
by itself did not perturb or alter the microbiota composition LA antagonized the ST-induced inflammation [44]. However,
significantly and LA was restoring ST-perturbed microbiota to moderate activation (low-fold changes) of chemokines like
a normal level (Fig. 4). Our species-level information revealed Ccl17, Ccl24, Ccrl2, CXCL12, and interleukins, such as Il2,
an increase in the number of bacteria that enhances anti- Il6, Il22, Il22ra1, and Il24 in STLA-treated BALB/c mice
inflammatory responses in BALB/c and C57BL/6 mice either indicated the immune activation for ST clearance without
Probiotics & Antimicro. Prot.

inducing major inflammation. Higher activation (fold changes corroborated the inflammatory status of ST-treated mice after
greater than five) of the aforementioned chemokines and cy- 5 days. Expression mucin genes (Muc13, Muc16, and Muc20)
tokines, along with Tnf and Il1b in STBC-treated C57BL/6 in LA and STLA-treated mice indicated an intact IEC barrier,
mice, indicated immune activation for ST clearance, high in- while downregulation of those same genes in ST- and STBC-
flammation might have caused the severe immune infiltration treated mice indicated compromised IEC barrier. Tjp1 was
and damage to the gut mucosa. Overexpression of formyl moderately expressed in LA- and STLA-treated mice group
peptide receptor 1 (Fpr1) in STBC-treated BALB/c mice, but was downregulated in ST- and STBC-treated mice to com-
which are known to be aberrantly expressed during inflamma- promise of IEC barrier integrity in ST- and STBC-treated
tion [45], corroborated with the inflammatory condition ob- mice. E-cadherin (Cdh2, Cdh7, Cdh12, Cdh20, Cdhr2, and
served in these groups of mice. SCFAs are also essential for Cdhr4) were downregulated in ST- and STBC-treated mice,
the maintenance of mucosal immunity by fortifying the intes- indicating compromised gut barrier integrity in these groups
tinal epithelial cell barrier function. It was reported that in of mice. It is also important to note that there are reports that
response to SCFAs, intestinal epithelial goblet cells increased suggested efficacy of treatment of probiotics, in general and
transcription of mucin genes [45, 46]. Results from the current LA and BC, in particular, against pathogen challenge or infec-
transcriptome study suggested that, while ST downregulates tion [49–56]. It was reported that secreted compounds of BC
mucins, including Muc5Ac, Muc5B, Muc13, Muc16, and could alleviate infection because of C. difficile and BC could
Muc20, LA restored expression of the Mucin genes in have human use as well as act against viral infection by acti-
STLA-treated BALB/c mice. Apart from mucins other pro- vating TLR-2 dependent mechanism [49, 52, 54, 57].
teins contributing to IEC barrier function, are intercellular Similarly, LA could be effective against breast cancer, allevi-
junction proteins, such as occludins, zonula occludens 1 ate inflammation following Salmonella infection by Tgf-β
(known as Tjp1), and E-cadherins. Downregulation of tight signaling pathway and promote gut immunity as well can treat
junction protein 2 (Tjp2) in STBC and ST-treated mice im- respiratory infections [50, 51, 53, 55, 56]. The current report
plied compromised intestinal barrier in these groups of mice. revealed differential efficacy of LA and BC in treating
However, Tjp2 is overexpressed in STLA and LA-treated Salmonella infection in BALB/c and C57BL/6 mice.
mice, indicating the high integrity of the intestinal barrier in Together, the microbiota, host transcriptome, and host
these groups of mice. Downregulation of E-cadherins, such as histopathological data suggested that BALB/c mice mi-
Cdh10, Cdh17, Cdh22, Cdhr1, Cdhr2, and Cdhr5 in ST- and crobiota were less perturbed than that of C57BL/6 mice
STBC-treated mice, indicates a compromised gut barrier in with ST infection. LA antagonizes the ST infection
these groups of mice. completely by inducing SCFA producing microbial spe-
Activation of Hdac6 by BC and STBC in C57BL/6 mice cies like B. pullicaecorum, C. scindens, C. arthromitus, C.
followed by activation of Tnf, Tnfsf4, Il1b, and Tnfrsf4 tertium, C. xylanolyticum, and F. prausnitzii in BALB/c
through NF-kB pathway corroborated with high inflammation mice. However, BC could not induce these SCFA produc-
and immune cell infiltration observed in STBC-treated groups ing bacteria significantly; therefore, STBC-treated BALB/
of mice. However, LA-treated C57BL/6 mice had downregu- c mice survived, but with severe inflammation in the gut.
lated HDACs and TNFs, which was corroborated by low in- Downregulated Tnf and Il1b in STLA-treated mice but
flammation and low immune cell infiltration in STLA-treated upregulated Tnf and Il1b in STBC-treated mice, in addi-
mice. Upregulated HDACs and TNFs by treatment with BC tion to evidence from histological data, was in accordance
and ST might explain the synergistic activity of ST and BC with the previously mentioned observations. Further ex-
with respect to inflammation in C57BL/6 mice, whereas pression of several mucins, zonula occludens 1, and sev-
HDAC downregulation by LA could alleviate the ST- eral E-cadherins in STLA-treated mice indicated the intact
induced inflammation. Moderate activation (low-fold IEC barrier function, which was supported by gut histo-
changes) of chemokines, such as Ccl2, Ccl7, Ccr4, Cxcl1, logical data (Fig. S7, the graphical summary).
Cxcl13, and interleukins, like Il12b, Il6, Il17a, Il17f, Il19, The microbiota of C57BL/6 mice was readily perturbed by
and Il22 in STLA-treated C57BL/6 mice, indicated the im- the ST-induced growth of sulfate-reducing proteobacteria D.
mune activation for ST clearance without inducing major in- intestinalis and inflammation-inducing proteobacteria H.
flammation [47, 48]. Higher activation of the aforementioned typhlonius. However, LA-treated mice have increased
chemokines and cytokines along with Tnf and Il1b in STBC- antiinflammatory SCFA producing bacteria, such as C. lepum,
treated C57BL/6 mice indicated immune activation for ST C . p o l y s a c c h a r o l y t i c u m , P. d e n t a l i s , a n d H .
clearance, but over inflammation might have compromised hemicellulosilytica through which consequently neutralized
survival. Aberrant expression of formyl peptide receptors the ST-induced inflammation in mice gut. The overactivation
(FPRs) has been well established in states of inflammation, of HDACs by BC and ST in C57BL/6 mice induces NF-kB
autoimmune diseases, neurodegenerative disorders, and can- mediated production of the inflammation-inducing Tnf and
cer [44]. Overexpression of Fpr1 and Fpr2 in ST-treated mice Il1b. The inflammation is again accompanied by
Probiotics & Antimicro. Prot.

compromised IEC barrier function through downregulation of Funding Information This study is provided by the Department of
Biotechnology (DBT), Government of India for partially funding this
mucins, Tjp1, and several E-cadherins in STBC-treated
project through extramural support. The funders had no role in study
C57BL/6 mice, which is evident from the histology of the design, data collection, and interpretation, or the decision to submit the
mice gut (Fig. S8, the graphical summary). work for publication
Microbial diversity has been reported to be depleted in
various inflammatory intestinal diseases [40, 58, 59]. It has Compliance with Ethical Standards
been observed that strain difference, strain dependent differ-
ence in secretoryIgA (sIgA) production as well as conserved Conflict of Interest The authors declare that they have no conflicts of
interest with the contents of this article.
signatures of salmonellosis in mouse and human hinted at
important strategies of Salmonella infection [40, 58, 59].
However, a common treatment or prevention was yet to be
established. The present study reported that LA could be a
References
strain independent solution for Salmonella infection. Here,
also we reported that gut microbiota is depleted by ST-
1. Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM,
induced diarrhea accompanied by severe gut inflammation. Hold G, Quraishi MN, Kinross J, Smidt H, Tuohy KM, Thomas LV,
Therefore, depletion of microbial diversity can be regarded Zoetendal EG, Hart A (2016) The gut microbiota and host health: a
as a marker for several gut-related diseases. While probiotics new clinical frontier. Gut 65(2):330–339. https://doi.org/10.1136/
gutjnl-2015-309990
were supplemented successfully offset the dysbiosis in gut
2. Moelling K (2016) Nutrition and the microbiome. Ann N Y Acad
microbiota caused by antibiotics, it should be kept in mind Sci 1372(1):3–8. https://doi.org/10.1111/nyas.13039
that a few probiotics species exist, such as BC, which may 3. Smoot DT, Mobley HLT, Chippendale GR, Lewison JF, Resau JH
exacerbate inflammation during ST infection. We conclude (1990) Helicobacter pylori urease activity is toxic to human gastric
that each species and each strain of probiotics should be pre- epithelial-cells. Infect Immun 58(6):1992–1994
screened for their compatibility with a particular type of infec- 4. Winter SE, Thiennimitr P, Winter MG, Butler BP, Huseby DL,
Crawford RW, Russell JM, Bevins CL, Adams LG, Tsolis RM,
tion or disease; further study in this area is highly needed and Roth JR, Baumler AJ (2010) Gut inflammation provides a respira-
future insights may serve as promising prospects for probiotic tory electron acceptor for Salmonella. Nature 467(7314):426–429.
use in clinical settings. Since current study is done in mice, we https://doi.org/10.1038/nature09415
cannot directly corroborate this with humans as well as the 5. Huurre A, Kalliomäki M, Rautava S, Rinne M, Salminen S, Isolauri
E (2007) Mode of delivery—effects on gut microbiota and humoral
SCFA associated microbiome identified in Th1 and Th2 mice immunity. Neonatology 93(4):236–240
need to be validated in humans under appropriate immune 6. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello
bias. MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin
AP, Heath AC, Warner B, Reeder J, Kuczynski J, Caporaso JG,
Acknowledgments We acknowledge and we are thankful to Mr. Madan Lozupone CA, Lauber C, Clemente JC, Knights D, Knight R,
Mohan Mallick and Mr. Susanta Kumar Swain, the technicians of Dr. Gordon JI (2012) Human gut microbiome viewed across age and
Shantibhusan Senapati’s laboratory for making the histological slides. geography. Nature 486(7402):222–227. https://doi.org/10.1038/
We deeply appreciate Bionivid, Xcelris Lab and SciGenome for timely nature11053
execution of 16S rRNA sequencing. We acknowledge Mr. Sibabrata 7. Modi SR, Collins JJ, Relman DA (2014) Antibiotics and the gut
Sarangi, attendant, NISER animal house for taking care of the animals microbiota. J Clin Invest 124(10):4212–4218. https://doi.org/10.
during the experiments. We heartily appreciate Mr. David Alexander 1172/JCI72333
Datzkiw for proofreading this manuscript. 8. Schultz BM, Paduro CA, Salazar GA, Salazar-Echegarai FJ,
Sebastian VP, Riedel CA, Kalergis AM, Alvarez-Lobos M,
Data Submission The transcriptomics data discussed in this publication Bueno SM (2017) A potential role of Salmonella infection in the
have been deposited in NCBI’s Gene Expression Omnibus and are acces- onset of inflammatory bowel diseases. Front Immunol 8:191.
sible through GEO series accession number GSE98353 (https://www. https://doi.org/10.3389/fimmu.2017.00191
ncbi.nlm.nih.gov/geo/query/acc.cgi?token=excdimugvlubnkj&acc= 9. Everard A, Cani PD (2013) Diabetes, obesity and gut microbiota.
GSE98353). Best Pract Res Clin Gastroenterol 27(1):73–83. https://doi.org/10.
The 16S rDNA sequencing data discussed in this publication have 1016/j.bpg.2013.03.007
been deposited in NCBI’s Sequence Read Archive and are accessible 10. Louis P, Hold GL, Flint HJ (2014) The gut microbiota, bacterial
through Bio project number PRJNA388784 (http://www.ncbi.nlm.nih. metabolites and colorectal cancer. Nat Rev Microbiol 12(10):661–
gov/bioproject/388784) and PRJNA392028 (https://www.ncbi.nlm.nih. 672. https://doi.org/10.1038/nrmicro3344
gov/bioproject/PRJNA392028). 11. Hold GL, Smith M, Grange C, Watt ER, El-Omar EM,
Mukhopadhya I (2014) Role of the gut microbiota in inflammatory
Author Contributions P.A. and B.P. planned the study and designed the bowel disease pathogenesis: what have we learnt in the past 10
experiments. B.P. executed most of the experiments and D.G., A.K.N, years? World J Gastroentero 20(5):1192–1210. https://doi.org/10.
A.B. assisted in the experiments with supervision from P.A., S.S., S.C., 3748/wjg.v20.i5.1192
B.P., and D.G. analyzed the histopathological data. P.A., B.P., and D.G. 12. Alzahrani S, Lina TT, Gonzalez J, Pinchuk IV, Beswick EJ, Reyes
analyzed the microarray data. S.T., B.P., P.A., and A.B. analyzed the VE (2014) Effect of Helicobacter pylori on gastric epithelial cells.
microbiome data. BP prepared the first draft of the manuscript. P.A. World J Gastroenterol 20(36):12767–12780. https://doi.org/10.
overall supervised the work and finalized the manuscript. 3748/wjg.v20.i36.12767
Probiotics & Antimicro. Prot.

13. Patel S, McCormick BA (2014) Mucosal inflammatory response to 29. Huson DH, Weber N (2013) Microbial community analysis using
Salmonella typhimurium infection. Front Immunol 5:311. https:// MEGAN. Methods Enzymol 531:465–485. https://doi.org/10.
doi.org/10.3389/fimmu.2014.00311 1016/B978-0-12-407863-5.00021-6
14. Wotzka SY, Nguyen BD, Hardt WD (2017) Salmonella typhimurium 30. Colwell RK, Elsensohn JE (2014) EstimateS turns 20: statistical
diarrhea reveals basic principles of enteropathogen infection and estimation of species richness and shared species from samples,
disease-promoted DNA exchange. Cell Host Microbe 21(4):443– with non-parametric extrapolation. Ecography 37(6):609–613.
454. https://doi.org/10.1016/j.chom.2017.03.009 https://doi.org/10.1111/ecog.00814
15. Cario E (2008) Innate immune signalling at intestinal mucosal sur- 31. Menke S, Gillingham MA, Wilhelm K, Sommer S (2017) Home-
faces: a fine line between host protection and destruction. Curr Opin made cost effective preservation buffer is a better alternative to
Gastroenterol 24(6):725–732. https://doi.org/10.1097/MOG. commercial preservation methods for microbiome research. Front
0b013e32830c4341 Microbiol 8:102. https://doi.org/10.3389/fmicb.2017.00102
16. Servin AL, Coconnier MH (2003) Adhesion of probiotic strains to 32. Roth S, Franken P, Sacchetti A, Kremer A, Anderson K, Sansom O,
the intestinal mucosa and interaction with pathogens. Best Pract Res Fodde R (2012) Paneth cells in intestinal homeostasis and tissue
Clin Gastroenterol 17(5):741–754 injury. PLoS One 7(6):e38965. https://doi.org/10.1371/journal.
17. Wells JM, Rossi O, Meijerink M, van Baarlen P (2010) Epithelial pone.0038965
crosstalk at the microbiota-mucosal interface. Proc Natl Acad Sci 33. Jin W, Dong C (2013) IL-17 cytokines in immunity and inflamma-
108(Supplement 1):4607–4614. https://doi.org/10.1073/pnas. tion. Emerg Microbes Infect 2(9):e60. https://doi.org/10.1038/emi.
1000092107 2013.58
18. Pradhan B, Guha D, Ray P, Das D, Aich P (2016) Comparative 34. Mayuzumi H, Inagaki-Ohara K, Uyttenhove C, Okamoto Y,
analysis of the effects of two probiotic bacterial strains on metabo- Matsuzaki G (2010) Interleukin-17A is required to suppress inva-
lism and innate immunity in the RAW 264.7 murine macrophage sion of Salmonella enterica serovar Typhimurium to enteric muco-
cell line. Probiotics Antimicrob Proteins 8(2):73–84. https://doi. sa. Immunology 131(3):377–385. https://doi.org/10.1111/j.1365-
org/10.1007/s12602-016-9211-4 2567.2010.03310.x
19. Moore RJ, Stanley D (2016) Experimental design considerations in 35. Dicksved J, Halfvarson J, Rosenquist M, Jarnerot G, Tysk C,
microbiota/inflammation studies. Clin Transl Immunology 5(7): Apajalahti J, Engstrand L, Jansson JK (2008) Molecular analysis
e92. https://doi.org/10.1038/cti.2016.41 of the gut microbiota of identical twins with Crohn’s disease. ISME
J 2(7):716–727. https://doi.org/10.1038/ismej.2008.37
20. Sur A, Pradhan B, Banerjee A, Aich P (2015) Immune activation
36. Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N,
efficacy of indolicidin is enhanced upon conjugation with carbon
Pace NR (2007) Molecular-phylogenetic characterization of micro-
nanotubes and gold nanoparticles. PLoS One 10(4):e0123905
bial community imbalances in human inflammatory bowel dis-
21. Hokamp K, Roche FM, Acab M, Rousseau ME, Kuo B, Goode D,
eases. Proc Natl Acad Sci U S A 104(34):13780–13785. https://
Aeschliman D, Bryan J, Babiuk LA, Hancock RE, Brinkman FS
doi.org/10.1073/pnas.0706625104
(2004) ArrayPipe: a flexible processing pipeline for microarray
37. Carroll IM, Ringel-Kulka T, Keku TO, Chang YH, Packey CD,
data. Nucleic Acids Res 32(Web Server):W457–W459. https://
Sartor RB, Ringel Y (2011) Molecular analysis of the luminal-
doi.org/10.1093/nar/gkh446
and mucosal-associated intestinal microbiota in diarrhea-
22. Zhang B, Kirov S, Snoddy J (2005) WebGestalt: an integrated predominant irritable bowel syndrome. Am J Physiol Gastrointest
system for exploring gene sets in various biological contexts. Liver Physiol 301(5):G799–G807. https://doi.org/10.1152/ajpgi.
Nucleic Acids Res 33(suppl 2):W741–W748. https://doi.org/10. 00154.2011
1093/nar/gki475 38. Chang JY, Antonopoulos DA, Kalra A, Tonelli A, Khalife WT,
23. Edgar RC (2013) UPARSE: highly accurate OTU sequences from Schmidt TM, Young VB (2008) Decreased diversity of the fecal
microbial amplicon reads. Nat Methods 10(10):996–998. https:// microbiome in recurrent Clostridium difficile-associated diarrhea. J
doi.org/10.1038/nmeth.2604 Infect Dis 197(3):435–438. https://doi.org/10.1086/525047
24. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman 39. Young VB, Schmidt TM (2004) Antibiotic-associated diarrhea ac-
FD, Costello EK, Fierer N, Pena AG, Goodrich JK, Gordon JI, companied by large-scale alterations in the composition of the fecal
Huttley GA, Kelley ST, Knights D, Koenig JE, Ley RE, microbiota. J Clin Microbiol 42(3):1203–1206
Lozupone CA, McDonald D, Muegge BD, Pirrung M, Reeder J, 40. Fransen F, Zagato E, Mazzini E, Fosso B, Manzari C, El Aidy S,
Sevinsky JR, Turnbaugh PJ, Walters WA, Widmann J, Yatsunenko Chiavelli A, D'Erchia AM, Sethi MK, Pabst O, Marzano M, Moretti
T, Zaneveld J, Knight R (2010) QIIME allows analysis of high- S, Romani L, Penna G, Pesole G, Rescigno M (2015) BALB/c and
throughput community sequencing data. Nat Methods 7(5):335– C57BL/6 mice differ in polyreactive IgA abundance, which impacts
336. https://doi.org/10.1038/nmeth.f.303 the generation of antigen-specific IgA and microbiota diversity.
25. Meyer F, Paarmann D, D'Souza M, Olson R, Glass EM, Kubal M, Immunity 43(3):527–540. https://doi.org/10.1016/j.immuni.2015.
Paczian T, Rodriguez A, Stevens R, Wilke A, Wilkening J, Edwards 08.011
RA (2008) The metagenomics RAST server—a public resource for 41. Rooks MG, Garrett WS (2016) Gut microbiota, metabolites and
the automatic phylogenetic and functional analysis of host immunity. Nat Rev Immunol 16(6):341–352. https://doi.org/
metagenomes. Bmc Bioinformatics 9(1):1 10.1038/nri.2016.42
26. Arndt D, Xia J, Liu Y, Zhou Y, Guo AC, Cruz JA, Sinelnikov I, 42. Usami M, Kishimoto K, Ohata A, Miyoshi M, Aoyama M, Fueda
Budwill K, Nesbo CL, Wishart DS (2012) METAGENassist: a Y, Kotani J (2008) Butyrate and trichostatin A attenuate nuclear
comprehensive web server for comparative metagenomics. factor kappaB activation and tumor necrosis factor alpha secretion
Nucleic Acids Res 40(Web Server issue):W88–W95. https://doi. and increase prostaglandin E2 secretion in human peripheral blood
org/10.1093/nar/gks497 mononuclear cells. Nutr Res 28(5):321–328. https://doi.org/10.
27. de Hoon MJ, Imoto S, Nolan J, Miyano S (2004) Open source 1016/j.nutres.2008.02.012
clustering software. Bioinformatics 20(9):1453–1454. https://doi. 43. Vinolo MA, Rodrigues HG, Hatanaka E, Sato FT, Sampaio SC,
org/10.1093/bioinformatics/bth078 Curi R (2011) Suppressive effect of short-chain fatty acids on pro-
28. Saldanha AJ (2004) Java Treeview—extensible visualization of duction of proinflammatory mediators by neutrophils. J Nutr
microarray data. Bioinformatics 20(17):3246–3248. https://doi. Biochem 22(9):849–855. https://doi.org/10.1016/j.jnutbio.2010.
org/10.1093/bioinformatics/bth349 07.009
Probiotics & Antimicro. Prot.

44. Bloes DA, Kretschmer D, Peschel A (2015) Enemy attraction: bac- spores in the prevention of recurrent respiratory infections in chil-
terial agonists for leukocyte chemotaxis receptors. Nat Rev dren: a pilot study. Ther Clin Risk Manag 3(1):13–17
Microbiol 13(2):95–104. https://doi.org/10.1038/nrmicro3390 53. Perdigon G, Maldonado Galdeano C, Valdez JC, Medici M (2002)
45. Willemsen LEM, Koetsier MA, Van Deventer SJH, Van Tol EAF Interaction of lactic acid bacteria with the gut immune system. Eur J
(2003) Short chain fatty acids stimulate epithelial mucin 2 expres- Clin Nutr 56(Suppl 4):S21–S26. https://doi.org/10.1038/sj.ejcn.
sion through differential effects on prostaglandin E1 and E2 pro- 1601658
duction by intestinal myofibroblasts. Gut 52(10):1442–1447 54. Ripert G, Racedo SM, Elie AM, Jacquot C, Bressollier P, Urdaci
46. Gaudier E, Jarry A, Blottiere HM, de Coppet P, Buisine MP, Aubert MC (2016) Secreted compounds of the probiotic Bacillus clausii
JP, Laboisse C, Cherbut C, Hoebler C (2004) Butyrate specifically strain O/C inhibit the cytotoxic effects induced by Clostridium dif-
modulates MUC gene expression in intestinal epithelial goblet cells ficile and Bacillus cereus toxins. Antimicrob Agents Chemother
deprived of glucose. Am J Physiol Gastrointest Liver Physiol 60(6):3445–3454. https://doi.org/10.1128/AAC.02815-15
287(6):G1168–G1174. https://doi.org/10.1152/ajpgi.00219.2004 55. Weiss G, Rasmussen S, Zeuthen LH, Nielsen BN, Jarmer H,
47. Chen J, Rao JN, Zou T, Liu L, Marasa BS, Xiao L, Zeng X, Turner Jespersen L, Frokiaer H (2010) Lactobacillus acidophilus induces
DJ, Wang JY (2007) Polyamines are required for expression of virus immune defence genes in murine dendritic cells by a Toll-like
Toll-like receptor 2 modulating intestinal epithelial barrier integrity. receptor-2-dependent mechanism. Immunology 131(2):268–281.
Am J Physiol Gastrointest Liver Physiol 293(3):G568–G576. https://doi.org/10.1111/j.1365-2567.2010.03301.x
https://doi.org/10.1152/ajpgi.00201.2007 56. Yan F, Polk DB (2011) Probiotics and immune health. Curr Opin
48. Liu L, Guo X, Rao JN, Zou TT, Xiao L, Yu TX, Timmons JA, Gastroenterol 27(6):496–501. https://doi.org/10.1097/MOG.
Turner DJ, Wang JY (2009) Polyamines regulate E-cadherin tran- 0b013e32834baa4d
scription through c-Myc modulating intestinal epithelial barrier
57. Menconi A, Morgan MJ, Pumford NR, Hargis BM, Tellez G (2013)
function. Am J Phys Cell Phys 296(4):C801–C810. https://doi.
Physiological properties and Salmonella growth inhibition of pro-
org/10.1152/ajpcell.00620.2008
biotic Bacillus strains isolated from environmental and poultry
49. Duc le H, Hong HA, Barbosa TM, Henriques AO, Cutting SM
sources. Int J Bacteriol 2013:958408. doi:https://doi.org/10.1155/
(2004) Characterization of Bacillus probiotics available for human
2013/958408, 1, 8
use. Appl Environ Microbiol 70(4):2161–2171
50. Huang IF, Lin IC, Liu PF, Cheng MF, Liu YC, Hsieh YD, Chen JJ, 58. Lee SJ, Liang L, Juarez S, Nanton MR, Gondwe EN, Msefula CL,
Chen CL, Chang HW, Shu CW (2015) Lactobacillus acidophilus Kayala MA, Necchi F, Heath JN, Hart P, Tsolis RM, Heyderman
attenuates Salmonella-induced intestinal inflammation via TGF- RS, MacLennan CA, Felgner PL, Davies DH, McSorley SJ (2012)
beta signaling. BMC Microbiol 15:203. https://doi.org/10.1186/ Identification of a common immune signature in murine and human
s12866-015-0546-x systemic Salmonellosis. Proc Natl Acad Sci U S A 109(13):4998–
51. Maroof H, Hassan ZM, Mobarez AM, Mohamadabadi MA (2012) 5003. https://doi.org/10.1073/pnas.1111413109
Lactobacillus acidophilus could modulate the immune response 59. Wickham ME, Brown NF, Provias J, Finlay BB, Coombes BK
against breast cancer in murine model. J Clin Immunol 32(6): (2007) Oral infection of mice with Salmonella enterica serovar
1353–1359. https://doi.org/10.1007/s10875-012-9708-x Typhimurium causes meningitis and infection of the brain. BMC
52. Marseglia GL, Tosca M, Cirillo I, Licari A, Leone M, Marseglia A, Infect Dis 7:65. https://doi.org/10.1186/1471-2334-7-65
Castellazzi AM, Ciprandi G (2007) Efficacy of Bacillus clausii

You might also like