ArticlePDF Available

Anti‐inflammatory potential of Lactobacillus reuteri LM 1071 in LPS‐stimulated RAW264.7 cells

Authors:

Abstract

Aims: To investigate anti-inflammatory effects of Lactobacillus reuteri LM1071 in lipopolysaccharides (LPS)-induced inflammation RAW264.7 cells. Methods and results: To evaluate anti-inflammatory activities of L. reuteri LM1071, LPS-stimulated RAW264.7 cells were used. Gene expression levels of eight immune-associated genes including IL-1β, IL-6, and TNF-α and protein production levels of COX-1 and COX-2 were analyzed. Moreover, the production of eicosanoids as important biomarkers for anti-inflammation was determined. Conclusions: The current study demonstrates that L. reuteri LM1071 has anti-inflammatory potential by inhibiting the production of inflammation mediators such as NO, eicosanoids such as PGE1 & PGE2, pro-inflammatory cytokines, and COX proteins. It can also enhance the production of inflammatory associated genes such as IL-11, BMP4, LEFTY2, and EET metabolite. Significance and impact of the study: Lactobacillus reuteri is one of crucial bacteria for food fermentation. It can be found in the gastrointestinal system of human and animals. Several studies have shown that L. reuteri has valuable effects on host health. The current study firstly demonstrated that L. reuteri has beneficial effect on the inflammation containing the variation of eicosanoids which are the one of the most important biomarkers and moreover eicosanoid-associated genes as well as proteins.
J Appl Microbiol. 2021;00:1–9. wileyonlinelibrary.com/journal/jam
|
1
© 2021 The Society for Applied Microbiology
INTRODUCTION
Inflammation is a primary host defence mechanism
against many stimuli such as external infections, chem-
ical damage and physical damage that can lead to tissue
injury (Yu et al., 2019). Macrophages are among import-
ant innate immune cells that respond to these stimuli by
releasing of pro- inflammatory mediators and phagocy-
tosis for injured tissue reparation (Ran & Montgomery,
2012; Watanabe et al., 2019). During inflammation, mac-
rophages regulate the production of pro- inflammatory
cytokines such as interleukin (IL)- 1β, IL- 6 and tumour
necrosis factor- α (TNF- α) (Dinarello, 2006) as well as the
Received: 10 June 2021
|
Revised: 13 October 2021
|
Accepted: 18 October 2021
DOI: 10.1111/jam.15331
ORIGINAL ARTICLE
Anti- inflammatory potential of Lactobacillus reuteri
LM1071 via eicosanoid regulation in LPS- stimulated
RAW264.7 cells
A- yeongJang1
|
WeerawanRod- in1
|
ChaiwatMonmai1
|
MinnSohn2
|
Tae- rahkKim2
|
Min- GyuJeon2
|
Woo JungPark1
A- yeong Jang and Weerawan Rod- in are contributed equally to this
research.
1Department of Marine Food Science
and Technology, Gangneung- Wonju
National University, Gangneung,
Gangwon, Korea
2Center for Research and Development,
LACTOMASON, Jinju, Korea
Correspondence
Woo Jung Park, Department of
Marine Food Science and Technology,
Gangneung- Wonju National University,
Gangneung, Gangwon 25457, Korea.
Email: pwj0505@gwnu.ac.kr
Funding information
LACTOMASON
Abstract
Aims: To investigate anti- inflammatory effects of Lactobacillus reuteri LM1071 in
lipopolysaccharides (LPS)- induced inflammation RAW264.7 cells.
Methods and Results: To evaluate anti- inflammatory activities of L. reuteri
LM1071, LPS- stimulated RAW264.7 cells were used. Gene expression levels
of eight immune- associated genes including IL- 1β, IL- 6 and TNF- α and
protein production levels of COX- 1 and COX- 2 were analysed. Moreover, the
production of eicosanoids as important biomarkers for anti- inflammation was
determined.
Conclusions: The current study demonstrates that L. reuteri LM1071 has anti-
inflammatory potential by inhibiting the production of inflammation mediators such
as NO, eicosanoids such as PGE1 & PGE2, pro- inflammatory cytokines and COX
proteins. It can also enhance the production of inflammatory associated genes such
as IL- 11, BMP4, LEFTY2 and EET metabolite.
Significance and Impact of the Study: Lactobacillus reuteri is one of the crucial
bacteria for food fermentation. It can be found in the gastrointestinal system of
human and animals. Several studies have shown that L. reuteri has valuable effects
on host health. The current study firstly demonstrated that L. reuteri has a benefi-
cial effect on the inflammation containing the variation of eicosanoids (PGE1 and
PGE2) which are one of the most important biomarkers and moreover eicosanoid-
associated genes as well as proteins (COX- 2).
KEYWORDS
anti- inflammation, COX, eicosanoid, Lactobacillus reuteri LM1071, RAW264.7 cells
2
|
JANG  .
production of pro- inflammatory mediators such as cyclo-
oxygenase- 2 (COX- 2) and inducible nitric oxide synthase
(iNOS) (Choi et al., 2019).
Lactobacillus spp. exist in various food products in
every part of the world (Greifová et al., 2017). They
are probiotic bacteria that can provide good effects
on several physiological processes including immune
regulation (Ding et al., 2017). Several studies have
reported the potential of Lactobacillus spp. to regula-
tion immunity by minimizing inflammatory responses
through T- lymphocytes (Jang et al., 2012; Shah et al.,
2012; Smits et al., 2005), B- lymphocytes (Hosoya et al.,
2014; Lee et al., 2013), nature killer (NK) cells (Makino
et al., 2016; Matsusaki et al., 2016; Shida et al., 2017)
and macrophages (Sohn et al., 2015; Ukibe et al.,
2015). In addition, Lactobacillus rhamnosus and L.
plantarum can remarkably induce the production of
pro- inflammatory cytokines such as IL- 6 and TNF- α
(Jorjão et al., 2015; Lee et al., 2016a). However, L. gas-
seri and L. rhamnosus can decrease the production
of IL- 6 and TNF- α (Mortaz et al., 2015; Ukibe et al.,
2015).
Lactobacillus reuteri is one of crucial bacteria for
food fermentation. It can be found in the gastrointes-
tinal system of human and animals (Duar et al., 2017).
Several studies have shown that L. reuteri has valuable
effects on host health. It can protect host against gas-
trointestinal infections by bacteria such as Clostridium
difficile (Cherian et al., 2015), Salmonella (Abhisingha
et al., 2018) and Escherichia coli (Genís et al., 2017).
Many strains of L. reuteri have anti- inflammatory po-
tential effects. For example it has been reported that
L. reuteri GMNL- 263 can decrease serum levels of
TNF- α, IL- 6 and monocyte chemoattractant protein- 1
(MCP- 1) in high- fat diet administrated rats (Hsieh
et al., 2016). The production of TNF- α in lipopoly-
saccharides (LPS)- induced THP- 1 cells is suppressed
by L. reuteri BM36301 (Lee et al., 2016b). L. reuteri
DSM12246 can suppress the production of IL- 12, IL- 6
and TNF- α in dendritic cells of mice (Christensen
et al., 2002). Recently, it has been reported that L. re-
uteri LM1071 isolated from human breast milk exhib-
its anti- inflammatory effects on mRNA expression of
IL- 6, TNF- α and IL- 4 in IL- 1β- stimulated HT- 29 cells
and that it can be used as an effective and safe pro-
biotic for humans (Kim et al., 2020). However, anti-
inflammatory effects of L. reuteri based on the evidence
of eicosanoid production have not been reported yet.
Thus, the objective of the current study was to firstly
investigate the beneficial effects of L. reuteri LM1071
on inflammation including the production of eicosa-
noids known to be important biomarkers. Effects of
L. reuteri LM1071 on eicosanoid- associated genes and
proteins in LPS- induced inflammation RAW264.7 cells
were also determined.
MATERIALS AND METHODS
Lactic acid bacteria strain
L. reuteri LM1071 was obtained from LACTOMASON at
a concentration of 4.8×109 cells g1 (Kim et al., 2020).
L. reuteri LM1071 was prepared in stock solution at
1 mg ml1 (w/v) that dissolved in dimethyl sulfoxide
(DMSO), and diluted various concentrations using cul-
ture medium for experiment. Treatment groups included
10, 20, 30 and 40 μg ml1 corresponding to 4.8 × 104,
9.6×104, 1.44×105 and 1.92×105 cells ml1 respectively.
Animal cell culture
RAW264.7 cells were purchased from Korean Cell Line
Bank (KCLB, South Korea). Cells were maintained in RPMI-
1640medium (Gibco™, USA) supplemented with 10% foetal
bovine serum (FBS) and 1% penicillin/streptomycin (Welgene,
South Korea) at 37°C in a humidified 5% CO2 incubator.
Nitric oxide (NO) production and cell
proliferation assay
RAW264.7 cells were seeding at a density of 1×105 cells
well1 in a 96- well plate. After 24h incubation, the culture
medium was removed. Cells were pretreated with vari-
ous concentrations (10, 20, 30 and 40 μg ml1) of L. reu-
teri LM1071 for 1h. Cells were stimulated with or without
1μgml1 of LPS (from Escherichia coli, Sigma- Aldrich, USA)
and incubated at 37°C for 24h. NO production in culture su-
pernatant was determined using Griess reagent (Promega,
USA). The culture supernatant was mixed with the same
volume of the Griess reagent following the manufacture's
instruction. The absorbance at 540nm was measured using
an EL800 Absorbance Microplate Reader (BioTek, USA). A
standard curve generated using sodium nitrite (provided in
the kit) was used for nitrite concentration determination.
Stimulated cells were used to evaluate the cytotoxicity
of L. reuteri LM1071 to RAW264.7 cells using an EZ- cytox
cell viability assay kit (Daeil Labservice, Korea) accord-
ing to the manufacturer's instructions. Cell proliferation
ratio was calculated according to the following formula:
Cell proliferation ratio (%)=
The absorbance at 450 nm of test group
The absorbance at 450 nm of control group
×
100
|
3
ANTI- INFLAMMATORY EFFECT OF L. REUTERI
RNA extraction and cDNA synthesis
After treatment with L. reuteri LM1071 and LPS, total
RNA was extracted from RAW264.7 cells using Tri rea-
gent® (Molecular Research Center, Inc., USA). RNA was
precipitated using absolute isopropanol alcohol at 4°C for
30min. The RNA pellet was collected by centrifugation
and washed with 70% ethanol thrice. Total RNA was dis-
solved in nuclease- free water. Its quantity was measured
using a nanophotometer (Implen, Germany). Then, 1g
of RNA was transcribed into cDNA using a high- capacity
cDNA reverse transcription kit (Applied biosystems,
USA) according to the manufacturer's recommended
instructions.
Quantification of immune- associated
gene expression
To evaluate inflammatory gene expression, 5 ng
of cDNA from each treatment group was used as a
template. The qPCR was performed with TB Green®
Premix Ex Taq™ II (Takara Bio Inc., Japan) in a
QuantStudio™ 3 FlexReal- Time PCR System (Applied
Biosystem, USA). The PCR reaction contained
0.4μmoll1 of specific primer set (Table 1). Relative
mRNA expression was normalized against β- actin
as a reference gene and analysed using QuantStudio
3software.
Eicosanoid level determination
Levels of prostaglandin E1 (PGE1: Enzo Life Science,
USA), prostaglandin E2 (PGE2: Enzo Life Science, USA),
leukotriene B4 (LTB4: Enzo Life Science, USA) and epox-
yeicosatrienoic acids (EET: MyBioSource, USA) were
measured using ELISA kits following to the manufactur-
er's instructions.
Western blot
RAW264.7 cells were placed in a six- well plate and treated
with different concentrations of L. reuteri LM1071 and LPS
as described in the NO production section. Cells were har-
vested and lysed with RIPA buffer (Tech & Innovation,
China) supplemented with 0.1% of protease inhibitor
(Thermo Scientific, USA). Protein concentration was
evaluated using Pierce™ BCA Protein Assay Kit (Thermo
Scientific, USA). The same amount of protein from each
treatment group (30 μg) was separated on 10% SDS-
polyacrylamide gel electrophoresis (SDS- PAGE). These
separated proteins were transferred onto a polyvinylidene
fluoride (PVDF) membrane. Western blot was carried out
as described by Narayanan et al. (2003). Briefly, the mem-
brane was incubated with antibodies specific to COX- 1
(Cell Signaling Technology, USA), COX- 2 (Cell Signaling
Technology, USA) and α- tubulin (Abcam, United Kingdom)
at 4°C overnight. The membrane was then incubated with
TABLE List of primers used in quantitative real- time PCR experiment
Gene Accession No. Primer sequence
iNOS BC062378.1 Forward: 5’- TTCCAGAATCCCTGGACAAG3’
Reverse: 5’- TGGTCAAACTCTTGGGGTTC3’
COX−2 NM_011198.4 Forward: 5’- AGAAGGAAATGGCTGCAGAA3’
Reverse: 5’- GCTCGGCTTCCAGTATTGAG3’
IL−1βNM_008361.4 Forward: 5’- GGGCCTCAAAGGAAAGAATC3’
Reverse: 5’- TACCAGTTGGGGAACTCTGC3’
IL−6 NM_031168.2 Forward: 5’- AGTTGCCTTCTTGGGACTGA3’
Reverse: 5’- CAGAATTGCCATTGCACAAC3’
TNF- αD84199.2 Forward: 5’- ATGAGCACAGAAAGCATGATC3’
Reverse: 5’- TACAGGCTTGTCACTCGAATT3’
IL−11 BC134354.1 Forward: 5’- TCCCCTCGAGTCTCTTCAGA3’
Reverse: 5’- TCTCCGTCAGCTGGGAATTT3’
BMP4 D14814.1 Forward: 5’- CTTCAACCTCAGCAGCATCC3’
Reverse: 5’- GATGAGGTGTCCAGGAACCA3’
LEFTY2 NM_177099.4 Forward: 5’- CAGCTGCAGCTCAGCCAGGCCC3’
Reverse: 5’- AGCGGTCAGCGTGACTTCCC3’
β- actin NM_007393.5 Forward: 5’- CCACAGCTGAGAGGGAAATC3’
Reverse: 5’- AAGGAAGGCTGGAAAAGAGC3’
4
|
JANG  .
goat anti- rabbit IgG- HRP secondary antibody at room tem-
perature for 1h. Expression levels of proteins were detected
using Pierce® ECL Plus Western Blotting Substrate (Thermo
Scientific, USA). ChemiDoc XRS + imaging system (Bio-
Rad, USA) was used for blot imaging and ImageLab soft-
ware (version 4.1; Bio- Rad, USA) was used to determine
the quantity of protein expression.
Statistical analysis
All statistical analyses were performed using the Statistix
8.1software. Data are presented as means ± standard de-
viation. One- way analysis of variance (ANOVA) with a
Tukey's honest significance test was then performed to
determine differences among treatment groups. Statistical
significance was considered at p <0.05. All experiments
were performed three times independently.
RESULTS
Effect of L. reuteri LM1071 on NO
production and cell proliferation
Anti- inflammatory activities of L. reuteri LM1071
were evaluated using LPS- stimulated RAW264.7 cells.
Different concentrations of L. reuteri LM1071 cells were
used to treat cells to determine the production of NO and
cell proliferation. As shown in Figure 1a, RAW264.7 (- )
cells (cells cultured with RPMI medium without stimula-
tion by LPS) showed very low NO production level, while
FIGURE Effect of L. reuteri
LM1071 in LPS- stimulated RAW264.7
cells. (a) effect on NO production and
(b) effect on cell proliferation. Data
are presented as mean ± SD of three
independent experiments (n=3).
Different letters (a, b, c and d) indicate
statistical differences between treatment
groups at p<0.05. (- ), RPMI medium as
the negative control
50
(a)
40
30
20
10
0
Nitric oxide production (µmol l-1)
(-) DMSO 10 20
L. reuteri concentration (µg ml-1)
30 40LPS
(-) DMSO 10 20
L. reuteri concentration (µg ml
-1
)
30 40LPS
a
b
c
dddd
140
(b)
120
100
80
60
40
20
0
Cell proliferation ratio (%)
cc
b
aa
cc
|
5
ANTI- INFLAMMATORY EFFECT OF L. REUTERI
FIGURE The immune- associated gene expression effect of L. reuteri LM1071 in LPS- stimulated RAW264.7 cells. (a) iNOS, (b) COX-
2, (c) IL- 1β, (d) IL- 6, (e) TNF- α, (f) IL- 11, (g) BMP4 and (h) LEFTY2. Data are presented as mean ± SD of three independent experiments
(n=3). Different letters (a, b, c, d, e and f) indicate statistical differences between treatment groups at p<0.05. (- ), RPMI medium as the
negative control
25
(a)
20
15
10
iNOS expression (fold)
5
0
e
(-) DMSO LPS 10 20 30 40
L. reuteri concentration (µg ml-1)
e
b
a
c
d
e
140
120
100
80
60
40
20
0
(b)
COX-2 expression (fold)
(-)DMSO LPS 10 20 30 40
L. reuteri concentration (µg ml-1)
e
d
c
ff
b
a
(c)
IL-1
β
expression (fold)
0
10
20
30
40
50
60
(-) DMSO LPS 10 20 30 40
L. reuteri concentration (µg ml-1)
e
d
ff
c
b
a
20
15
10
5
0
(d)
IL-6 expression (fold)
(-)DMSO LPS 10 20 30 40
L. reuteri concentration (µg ml-1)
(-) DMSO LPS 10 20 30 40
L. reuteri concentration (µg ml-1)
(-) DMSO LPS 10 20 30 40 (-)DMSO LPS 10 20 30 40
L. reuteri concentration (µg ml
-1
)
L. reuteri concentration (µg ml-1)
(-)DMSO LPS 10 20 30 40
L. reuteri concentration (µg ml-1)
a
b
c
ff
d
e
TNF-
α
expression (fold)
40
(e)
30
20
10
0
a
b
c
d
ff
e
IL-11 expression (fold)
2·5
2·0
1·5
1·0
0·5
0·0
(f)
(g) (h)
e
f
c
b
a
dd
BMP4 expression (fold)
3·0
2·5
2·0
1·5
1·0
0·5
0·0
f
dc
b
a
ee
LEFTY2 expression (fold)
3·0
2·5
2·0
1·5
1·0
0·5
0·0
d
c
b
a
dd
e
6
|
JANG  .
LPS- stimulated cells generated 42.10±0.45μmoll1NO.
Treatments with various concentrations of L. reuteri
LM1071 significantly decreased the production of NO
in LPS- stimulated RAW264.7 cells. However, L. reuteri
LM1071 was not cytotoxic to LPS- stimulated RAW264.7
cells at any concentration up to 40μgml1 (Figure 1b).
Effect of L. reuteri LM1071 on expression of
immune- associated genes
LPS obviously increased gene expression levels of inflam-
matory markers such as iNOS (Figure 2a) and COX- 2 in
RAW264.7 cells (Figure 2b). It also induced the expression
of pro- inflammatory cytokine genes such as IL- 1β, IL- 6 and
TNF- α (Figure 2c– e). However, L. reuteri LM1071 effectively
suppressed the expression of pro- inflammatory cytokines
known to be inflammatory markers in a concentration-
dependent manner in RAW264.7 cells (Figure 2a– e).
Conversely, LPS significantly reduced expression levels
of IL- 11, bone morphogenic protein 4 (BMP4) and left-
right determination factor 2 (LEFTY2) (Figure 2f– h). Gene
expression of IL- 11, BMP4 and LEFTY2 significantly in-
creased depending on the concentration of L. reuteri.
Effect of L. reuteri LM1071 on
eicosanoid level
To investigate the effect of L. reuteri LM1071 on LPS-
induced eicosanoid production, RAW264.7 cells were pre-
treated with various concentrations of L. reuteri LM1071
along with 1μgml1 of LPS. As shown in Figure 3, LPS
significantly increased the production of PGE1, PGE2
and LTB4, but significantly reduced the production of
EET. Different concentrations of L. reuteri LM1071 dose-
dependently decreased the production of PGE2 in LPS-
stimulated RAW264.7 cells. Production levels of PGE1
and LTB4 were also significantly declined when LPS-
stimulated cells were pretreated with L. reuteri LM1071
at high concentrations (30– 40μgml1 for PGE1 and 20–
40μgml1 for LTB4). Contrarily, the production of EET
remarkably was raised according to the concentration of
L. reuteri LM1071 in RAW264.7 cells.
FIGURE Effect of L. reuteri LM1071 on eicosanoid productions. (a) PGE1, (b) PGE2, (c) LTB4 and (d) EET. Data are presented as mean
± SD of three independent experiments (n=3). Different letters (a, b, c, d, e and f) indicate statistical differences between treatment groups
at p<0.05. (- ), RPMI medium as the negative control
4000
aaa
bb
c
(-) DMSO LPS 2010 30 40
L. reuteri concentration (µg ml-1)
(-) DMSO LPS 2010 30 40
(-) DMSO LPS 2010 30 40
(-) DMSO LPS 2010 30 40
L. reuteri concentration (µg ml-1)
L. reuteri concentration (µg ml
-1
)
L. reuteri concentration (µg ml
-1
)
c
(a)
(c) (d)
(b)
3000
2000
PEG1 production (pg ml-1)
1000
0
PEG2 production (pg ml-1)
EET production (µg ml-1)
350
300
250
200
150
100
50
0
a
b
c
d
ff
e
300
250
200
150
100
50
0
ee
f
d
c
b
a
LTB4 production (pg ml-1)
250
200
150
100
50
0
a
a
b
c
ccc
|
7
ANTI- INFLAMMATORY EFFECT OF L. REUTERI
Effect of L. reuteri LM1071 on COX- 1 and
COX- 2 production
Protein production levels of COX- 1 and COX- 2 in LPS-
treated RAW264.7 cells were greatly up- regulated than
those in the untreated group (Figure 4). However, L.
reuteri LM1071 dose- dependently suppressed such in-
creases of production levels of COX- 1 and COX- 2 in
LPS- stimulated cells. Moreover, treatment with high con-
centrations of L. reuteri LM1071 (30 and 40μgml1) dra-
matically depressed the production of COX- 1 and COX- 2.
DISCUSSION
Inflammation response is a process of preventing host
cells against external intrusion (Mariathasan & Monack,
2007). Macrophages are native immune cells associ-
ated with innate immunity. They play a key role in in-
flammatory responses (Fujiwara & Kobayashi, 2005).
However, the imbalance of immune system caused
by excessive production of inflammatory mediators
and long period of inflammation may lead to chronic
inflammatory disorders (Shanura Fernando et al., 2018).
The present study was performed to investigate the anti-
inflammatory potential of L. reuteri LM1071 in LPS-
stimulated RAW264.7 cells.
Cytokines are critical mediators coordinating in-
flammatory processes. The production of TNF- α, IL-
1β and IL- 6 cytokines may increase inflammation and
tissue injury (Oishi & Manabe, 2018). Conversely,
BMP4, IL- 11 and LEFTY2have been indicated as anti-
inflammatory molecules (Baraban et al., 2016; Ma et al.,
2013). Therefore, reducing the production of IL- 1β,
IL- 6 and TNF- α and preserving the activity of BMP4,
IL- 11 and LEFTY2might be an effective way to bal-
ance inflammatory disease defence. Figure 2showed
that mRNA expression levels of pro- inflammatory cy-
tokines (TNF- α, IL- 1β and IL- 6) were down- regulated
by L. reuteri in LPS- stimulated RAW264.7 cells in a
concentration- dependent manner. Similarly, it has
been reported that L. reuteri can decrease mRNA
expression levels of IL- 6, TNF- α and IL- 4 in IL- 1β-
induced HT- 29 cells (Kim et al., 2020). Conversely,
treatment with various concentrations of L. reuteri
LM1071 significantly increased mRNA expression
FIGURE The effect of L. reuteri
LM1071 on proteins associated COXs
production in LPS- stimulated RAW264.7
cells. (a) Western blot and (b) relative
band intensity. Data are presented
as mean ± SD of three independent
experiments (n=3). Different letters (a,
b, c and d) indicate statistical differences
between treatment groups at p<0.05. (- ),
RPMI medium as the negative control
-Tubulin
COX-2
COX-1
(-) DMSO LPS 10
L. reuteri concentration (µg ml-1)
20 30 40
Relative protein expression (fold)
400
300
200
100
0d
d
10 µg ml
-1
L. reuteri
20 µg ml-1 L. reuteri
30 µg ml-1 L. reuteri
40 µg ml-1 L. reuteri
1 µg ml-1 LPS
1% DMSO
(-) RPMI
a
b
ccd cd
cc
c
c
b
b
a
COX-1 COX-2
(a)
(b)
8
|
JANG  .
levels of BMP4, IL- 11 and LEFTY2 dose dependently.
These results indicate that L. reuteri LM1071 possesses
biological activities to regulate inflammation- related
cytokines and other genes.
Prostaglandins are known to be important mediators of
inflammation (Bamba et al., 2003). During inflammation,
production levels of PGE2 and LTB4 as pro- inflammatory
metabolites derived from arachidonic acid are up-
regulated, whereas EET as an anti- inflammatory metab-
olite derived from arachidonic acid is down- regulated
(Zhang et al., 2018). Like previous references, our results
also showed that treatment with L. reuteri LM1071signifi-
cantly decreased the production of PGE2 and LTB4 but
increased the production of EET.
Excessive production of NO and pro- inflammatory cy-
tokines might cause a wide range of severe cell injuries and
induce inflammation (Tak & Firestein, 2001). LPS is rec-
ognized by macrophage TLR- 4, leading to the activation of
immune- associated pathways such as MAPK and NF- κB
pathways (Shanura Fernando et al., 2018). Such activation
can lead to increased production of NO, PGE2 and pro-
inflammatory cytokines (Yu et al., 2019). LPS can activate
inflammatory responses, resulting in up- regulated pro-
duction of inflammatory mediators such as NO, COX- 1,
COX- 2 and pro- inflammatory cytokines (Hu et al., 2008).
Our results showed that L. reuteri LM1071 exhibited an
anti- inflammatory activity through down- regulating the
production of NO in LPS- stimulated RAW264.7 cells. The
expression of iNOS, a gene for the generation of NO, was
also significantly decreased depending on the concen-
tration of L. reuteri. In addition, L. reuteri LM1071 sup-
pressed the production of COX- 2 at both gene and protein
levels (Figure 4). COX- 2 is one of the most important bio-
markers to confirm the variation of inflammation in LPS-
stimulated RAW264.7 cells (Lee et al., 2008; Wang et al.,
2018).
In conclusion, the current study demonstrates that L.
reuteri LM1071has an anti- inflammatory potential by in-
hibiting the production of inflammation mediators such as
NO, eicosanoids such as PGE1 & PGE2, pro- inflammatory
cytokines and COXs protein. It can also enhance the pro-
duction of inflammation- associated genes such as IL- 11,
BMP4, LEFTY2 and EET metabolite.
ACKNOWLEDGEMENT
This study was supported by a Research Program funded
by LACTOMASON in Korea.
CONFLICT OF INTEREST
The authors declare no conflict of interest.
ORCID
Woo Jung Park https://orcid.org/0000-0001-9804-3444
REFERENCES
Abhisingha, M., Dumnil, J. & Pitaksutheepong, C. (2018) Selection
of potential probiotic Lactobacillus with inhibitory activity
against Salmonella and fecal coliform bacteria. Probiotics and
Antimicrobial Proteins, 10, 218– 227.
Bamba, H., Ota, S., Kato, A., Miyatani, H., Kawamoto, C., Yoshida,
Y. et al. (2003) Effect of rebamipide on prostaglandin receptors-
mediated increase of inflammatory cytokine production by mac-
rophages. Alimentary Pharmacology & Therapeutics, 18, 113– 118.
Baraban, E., Chavakis, T., Hamilton, B.S., Sales, S., Wabitsch,
M., Bornstein, S.R. et al. (2016) Anti- inflammatory proper-
ties of bone morphogenetic protein 4 in human adipocytes.
International Journal of Obesity, 40, 319– 327.
Cherian, P.T., Wu, X., Yang, L., Scarborough, J.S., Singh, A.P., Alam,
Z.A. et al. (2015) Gastrointestinal localization of metronidazole
by a lactobacilli- inspired tetramic acid motif improves treat-
ment outcomes in the hamster model of Clostridium difficile in-
fection. Journal of Antimicrobial Chemotherapy, 70, 3061– 3069.
Choi, S.H., Lee, S.H., Kim, M.G., Lee, H.J. & Kim, G.B. (2019)
Lactobacillus plantarum CAU1055 ameliorates inflammation
in lipopolysaccharide- induced RAW264.7 cells and a dextran
sulfate sodium- induced colitis animal model. Journal of Dairy
Science, 102, 6718– 6725.
Christensen, H.R., Frøkiaer, H. & Pestka, J.J. (2002) Lactobacilli
differentially modulate expression of cytokines and matura-
tion surface markers in murine dendritic cells. The Journal of
Immunology, 168, 171– 178.
Dinarello, C.A. (2006) The paradox of pro- inflammatory cytokines
in cancer. Cancer and Metastasis Reviews, 25, 307– 313.
Ding, Y.H., Qian, L.Y., Pang, J., Lin, J.Y., Xu, Q., Wang, L.H.
et al. (2017) The regulation of immune cells by Lactobacilli:
a potential therapeutic target for anti- atherosclerosis therapy.
Oncotarget, 8, 59915– 59928.
Duar, R.M., Lin, X.B., Zheng, J., Martino, M.E., Grenier, T., Pérez-
Muñoz, M.E. et al. (2017) Lifestyles in transition: evolution and
natural history of the genus Lactobacillus. FEMS Microbiology
Reviews, 41, S27– S48.
Fujiwara, N. & Kobayashi, K. (2005) Macrophages in inflammation.
Current Drug Targets: Inflammation & Allergy, 4, 281– 286.
Genís, S., Sánchez- Chardi, A., Bach, À., Fàbregas, F. & Arís, A. (2017)
A combination of lactic acid bacteria regulates Escherichia
coli infection and inflammation of the bovine endometrium.
Journal of Dairy Science, 100, 479– 492.
Greifová, G., Májeková, H., Greif, G., Body, P., Greifová, M. &
Dubničková, M. (2017) Analysis of antimicrobial and immu-
nomodulatory substances produced by heterofermentative
Lactobacillus reuteri. Folia Microbiol (Praha), 62, 515– 524.
Hosoya, T., Sakai, F., Yamashita, M., Shiozaki, T., Endo, T., Ukibe, K.
et al. (2014) Lactobacillus helveticus SBT2171 inhibits lympho-
cyte proliferation by regulation of the JNK signaling pathway.
PLoS One, 9, e108360.
Hsieh, F.C., Lan, C.C., Huang, T.Y., Chen, K.W., Chai, C.Y., Chen, W.T.
et al. (2016) Heat- killed and live Lactobacillus reuteri GMNL- 263
exhibit similar effects on improving metabolic functions in high-
fat diet- induced obese rats. Food Funct, 7, 2374– 2388.
Hu, S.S., Bradshaw, H.B., Chen, J.S., Tan, B. & Walker, J.M. (2008)
Prostaglandin E2 glycerol ester, an endogenous COX- 2 metabolite
of 2- arachidonoylglycerol, induces hyperalgesia and modulates
NF- κB activity. British Journal of Pharmacology, 153, 1538– 1549.
|
9
ANTI- INFLAMMATORY EFFECT OF L. REUTERI
Jang, S.O., Kim, H.J., Kim, Y.J., Kang, M.J., Kwon, J.W., Seo, J.H.
et al. (2012) Asthma prevention by Lactobacillus Rhamnosus
in a mouse model is associated with CD4(+)CD25(+)Foxp3(+)
T cells. Allergy, Asthma & Immunology Research, 4, 150– 156.
Jorjão, A.L., de Oliveira, F.E., Leão, M.V., Carvalho, C.A., Jorge, A.O.
& de Oliveira, L.D. (2015) Live and heat- killed Lactobacillus
rhamnosus ATCC 7469 may induce modulatory cytokines pro-
files on macrophages RAW264.7. The Scientific World Journal,
2015, 716749.
Kim, T.R., Choi, K.S., Ji, Y., Holzapfel, W.H. & Jeon, M.G. (2020)
Anti- inflammatory effects of Lactobacillus reuteri LM1071 via
MAP kinase pathway in IL- 1β- induced HT- 29 cells. Journal of
Animal Science and Technology, 62, 864– 874.
Lee, H.A., Kim, H., Lee, K.W. & Park, K.Y. (2016a) Dead Lactobacillus
plantarum stimulates and skews immune responses toward T
helper 1 and 17 polarizations in RAW264.7 cells and mouse sple-
nocytes. Journal of Microbiology and Biotechnology, 26, 469– 476.
Lee, J., Bang, J. & Woo, H.J. (2013) Effect of orally administered
Lactobacillus brevis HY7401 in a food allergy mouse model.
Journal of Microbiology and Biotechnology, 23, 1636– 1640.
Lee, J.M., Hwang, K.T., Jun, W.J., Park, C.S. & Lee, M.Y. (2008)
Antiinflammatory effect of lactic acid bacteria: inhibition
of cyclooxygenase- 2 by suppressing nuclear factor- kappaB
in RAW264.7 macrophage cells. Journal of Microbiology and
Biotechnology, 18, 1683– 1688.
Lee, J., Yang, W., Hostetler, A., Schultz, N., Suckow, M.A., Stewart,
K.L. et al. (2016b) Characterization of the anti- inflammatory
Lactobacillus reuteri BM36301 and its probiotic benefits on
aged mice. BMC Microbiology, 16, 69– 81.
Ma, H., Hong, M., Duan, J., Liu, P., Fan, X., Shang, E. et al. (2013)
Altered cytokine gene expression in peripheral blood mono-
cytes across the menstrual cycle in primary dysmenorrhea: a
case- control study. PLoS One, 8, e55200.
Makino, S., Sato, A., Goto, A., Nakamura, M., Ogawa, M., Chiba, Y.
et al. (2016) Enhanced natural killer cell activation by exopoly-
saccharides derived from yogurt fermented with Lactobacillus
delbrueckii ssp. bulgaricus OLL1073R- 1. Journal of Dairy
Science, 99, 915– 923.
Mariathasan, S. & Monack, D.M. (2007) Inflammasome adaptors
and sensors: intracellular regulators of infection and inflam-
mation. Nature Reviews Immunology, 7, 31– 40.
Matsusaki, T., Takeda, S., Takeshita, M., Arima, Y., Tsend- Ayush,
C., Oyunsuren, T. et al. (2016) Augmentation of T helper
type 1 immune response through intestinal immunity in mu-
rine cutaneous herpes simplex virus type 1 infection by pro-
biotic Lactobacillus plantarum strain 06CC2. International
Immunopharmacology, 39, 320– 327.
Mortaz, E., Adcock, I.M., Ricciardolo, F.L., Varahram, M., Jamaati,
H., Velayati, A.A. et al. (2015) Anti- inflammatory effects of
Lactobacillus rahmnosus and Bifidobacterium breve on cigarette
smoke activated human macrophages. PLoS One, 10, e0136455.
Narayanan, B.A., Narayanan, N.K., Simi, B. & Reddy, B.S. (2003)
Modulation of inducible nitric oxide synthase and related proin-
flammatory genes by the omega- 3 fatty acid docosahexaenoic
acid in human colon cancer cells. Cancer Research, 63, 972– 979.
Oishi, Y. & Manabe, I. (2018) Macrophages in inflammation, repair
and regeneration. International Immunology, 30, 511– 528.
Ran, S. & Montgomery, K.E. (2012) Macrophage- mediated lymph-
angiogenesis: the emerging role of macrophages as lymphatic
endothelial progenitors. Cancers (Basel), 4, 618– 657.
Shah, M.M., Saio, M., Yamashita, H., Tanaka, H., Takami, T., Ezaki,
T. et al. (2012) Lactobacillus acidophilus strain L- 92 induces
CD4(+)CD25(+)Foxp3(+) regulatory T cells and suppresses
allergic contact dermatitis. Biological &/and Pharmaceutical
Bulletin, 35, 612– 616.
Shanura Fernando, I.P., Asanka Sanjeewa, K.K., Samarakoon, K.W.,
Lee, W.W., Kim, H.S., Ranasinghe, P. et al. (2018) Antioxidant
and anti- inflammatory functionality of ten Sri Lankan seaweed
extracts obtained by carbohydrase assisted extraction. Food
Science and Biotechnology, 27, 1761– 1769.
Shida, K., Sato, T., Iizuka, R., Hoshi, R., Watanabe, O., Igarashi, T.
et al. (2017) Daily intake of fermented milk with Lactobacillus
casei strain Shirota reduces the incidence and duration of upper
respiratory tract infections in healthy middle- aged office work-
ers. European Journal of Nutrition, 56, 45– 53.
Smits, H.H., Engering, A., van der Kleij, D., de Jong, E.C., Schipper,
K., van Capel, T.M. et al. (2005) Selective probiotic bacteria in-
duce IL- 10- producing regulatory T cells in vitro by modulating
dendritic cell function through dendritic cell- specific intercel-
lular adhesion molecule 3- grabbing nonintegrin. The Journal of
Allergy and Clinical Immunology, 115, 1260– 1267.
Sohn, W., Jun, D.W., Lee, K.N., Lee, H.L., Lee, O.Y., Choi, H.S. et al.
(2015) Lactobacillus paracasei induces M2- dominant kupffer
cell polarization in a mouse model of nonalcoholic steatohepa-
titis. Digestive Diseases and Sciences, 60, 3340– 3350.
Tak, P.P. & Firestein, G.S. (2001) NF- kappaB: a key role in inflam-
matory diseases. The Journal of Clinical Investigation, 107,
7– 11.
Ukibe, K., Miyoshi, M. & Kadooka, Y. (2015) Administration of
Lactobacillus gasseri SBT2055 suppresses macrophage infil-
tration into adipose tissue in diet- induced obese mice. British
Journal of Nutrition, 114, 1180– 1187.
Wang, H., Zhang, L., Xu, S., Pan, J., Zhang, Q. & Lu, R. (2018)
Surface- layer protein from Lactobacillus acidophilus NCFM
inhibits lipopolysaccharide- induced inflammation through
MAPK and NF- κB signaling pathways in RAW264.7 cells.
Journal of Agriculture and Food Chemistry, 66, 7655– 7662.
Watanabe, S., Alexander, M., Misharin, A.V. & Budinger, G.R.S.
(2019) The role of macrophages in the resolution of inflamma-
tion. Journal of Clinical Investigation, 129, 2619– 2628.
Yu, H.S., Lee, N.K., Choi, A.J., Choe, J.S., Bae, C.H. & Paik, H.D.
(2019) Anti- inflammatory potential of probiotic strain Weissella
cibaria JW15 isolated from Kimchi through regulation of NF-
κB and MAPKs pathways in LPS- induced RAW264.7 cells.
Journal of Microbiology and Biotechnology, 29, 1022– 1032.
Zhang, Q., Wang, X., Yan, G., Lei, J., Zhou, Y., Wu, L. et al. (2018)
Anti- versus pro- inflammatory metabololipidome upon cup-
ping treatment. Cellular Physiology and Biochemistry, 45,
1377– 1389.
How to cite this article: Jang, A.- Y., Rod- in, W.,
Monmai, C., Sohn, M., Kim, T.- R., Jeon, M.- G. &
et al. (2021) Anti- inflammatory potential of
Lactobacillus reuteri LM1071 via eicosanoid
regulation in LPS- stimulated RAW264.7 cells.
Journal of Applied Microbiology, 00, 1– 9. https://
doi.org/10.1111/jam.15331
... rhamnosus GG, Lactobacillus reuteri LM1071, B. breve CECT7263 и Bifidobacterium animalis subsp. lactis , мультиштаммовых пробиотиков и синбиотических продуктов [2,5,6,10,12,13]. В ряде рандомизированных плацебо-контролированных исследованиях продемонстрировано уменьшение времени плача на фоне приема смеси L. rhamnosus, L. reuteri, B. infantis и фруктоолигосахарида [14], смеси L. rhamnosus 19070- 2, L. reuteri 12246, фруктоолигосахарида и 200 МЕ витамина D3 [15] [16]. ...
Article
Introduction. Probiotics have been shown to be effective in the treatment of infant colic for Lactobacillus reuteri. In recent years, multi-strain probiotics have been used for the same purpose. Aim. To evaluate the clinical course of infant colic during treatment with mono- and multiprobiotics and in follow-up. М aterial and methods. 110 children aged from 1 to 5 months (average age 3.2 ± 0.3 months) suffering from colic were examined. Patients were randomized into three groups: group 1 (n = 30), received the BioGaia Probiotic drops for baby, group 2 (n = 40) – Bac- Set Bab multiprobiotic, group 3 (n = 40) – symptomatic therapy. Results. The average duration of crying of children who received multiprobiotic (group 2) was significantly less than that of children of the first and third groups (group 1 – 1.62 ± 0.5 hours; group 2 – 1.1 ± 0.3 hours; group 3 – 2.5 ± 0.6 hours; p < 0.001). The duration of crying during colic was in correlation with the level of zonulin in the stool (r = 0.58, p < 0.05). Treatment with the multiprobiotic was accompanied by a decrease in the frequency of bowel movements from 4.15 times a day to 3.27 times and was not accompanied by an increase in constipation, while monostrain probiotic and symptomatic therapy did not change the frequency of daily bowel movements and did not affect pre-existing constipation. Conclusions. The multiprobiotic turned out to be more effective than the single-strain monostrain probiotic and symptomatic therapy in the speed of relieving infant colic; it completely stops them in 87.5% of children and reduces them in 12.5%. The effect of multiprobiotic is persistent, relapse was noted in only 13.3% of children, whereas after taking monostrain probiotic the relapse rate was 33.33%. The effect of multiprobiotic is associated with its ability to normalize intestinal microbiota and reduce the permeability of the intestinal wall.
... and is instrumental in regulating antimicrobial, antiviral, and immune responses [35]. Pathogenic bacteria can be reduced by the substantial presence of Lactobacillus spp. in the gastrointestinal tract of animals [36]. There is a significant enrichment of the Prevotella genus in the LADG, indicating that Prevotella significantly decreases the daily weight gain in pigs. ...
Article
Full-text available
The average daily weight gain (ADG) is considered a crucial indicator for assessing growth rates in the swine industry. Therefore, investigating the gastrointestinal microbiota and serum metabolites influencing the ADG in pigs is pivotal for swine breed selection. This study involved the inclusion of 350 purebred Yorkshire pigs (age: 90 ± 2 days; body weight: 41.20 ± 4.60 kg). Concurrently, serum and fecal samples were collected during initial measurements of blood and serum indices. The pigs were categorized based on their ADG, with 27 male pigs divided into high-ADG (HADG) and low-ADG (LADG) groups based on their phenotype values. There were 12 pigs in LADG and 15 pigs in HADG. Feces and serum samples were collected on the 90th day. Microbiome and non-targeted metabolomics analyses were conducted using 16S rRNA sequencing and liquid chromatography-mass spectrometry (LC-MS). Pearson correlation, with Benjamini–Hochberg (BH) adjustment, was employed to assess the associations between these variables. The abundance of Lactobacillus and Prevotella in LADG was significantly higher than in HADG, while Erysipelothrix, Streptomyces, Dubosiella, Parolsenella, and Adlercreutzia in LADG were significantly lower than in HADG. The concentration of glutamine, etiocholanolone glucuronide, and retinoyl beta-glucuronide in LADG was significantly higher than in HADG, while arachidonic acid, allocholic acid, oleic acid, phenylalanine, and methyltestosterone in LADG were significantly lower than in HADG. The Lactobacillus–Streptomyces networks (Lactobacillus, Streptomyces, methyltestosterone, phenylalanine, oleic acid, arachidonic acid, glutamine, 3-ketosphingosine, L-octanoylcarnitine, camylofin, 4-guanidinobutyrate 3-methylcyclopentadecanone) were identified as the most influential at regulating swine weight gain. These findings suggest that the gastrointestinal tract regulates the daily weight gain of pigs through the network of Lactobacillus and Streptomyces. However, this study was limited to fecal and serum samples from growing and fattening boars. A comprehensive consideration of factors affecting the daily weight gain in pig production, including gender, parity, season, and breed, is warranted.
... Самый высокий уровень доказательства эффективности среди вышеописанных методик имеет терапия пробиотиками [5,14,15,18]. У детей, находящихся на естественном вскармливании, продемонстрирована эффективность разных штаммов Lactobacillus reuteri [5,[18][19][20]. В недавнем обзоре представлены результаты 10 рандомизированных плацебо-контролируемых двой ных слепых 1 Materials and methods. ...
Article
Introduction. The highest level of evidence for the treatment of colic in breastfed infants is with L. reuteri monoprobiotic therapy. Recently multistrain probiotics have been used to treat colic. А im. Is to evaluate the effectiveness of various strategies for the treatment of infantile colic, including monoand multistrain probiotics. Materials and methods. A total of 110 infants aged 1–5 months with colic and 20 healthy infants were examined. The effectiveness of treatment was assessed in three groups of infants with colic: Group 1 consisting of 30 children who received BioGaia Probiotic drops for baby ( L. reuteri ); Group 2 consisting of 40 children who received Bac-Set Baby multistrain probiotic, and Group 3 consisting of 40 children who received symptomatic therapy. The estimated parameters included duration of crying, fecal levels of zonulin measured by ELISA assays, and gut microbiota composition. Results. Children with colic had longer crying duration compared to healthy children, higher levels of zonulin in the stool and differences in the microbiota. In group 2, colic relief occurred on average within 5 days, which is significantly faster than in groups 1 and 3 (10 and 12 days, respectively). In group 2, colic relief occurred in the majority of children (87.5%), the differences with groups 1 and 3 were statistically significant (53.3% and 10% of children, respectively). Also in group 2 there was a decrease in the level of zonulin by 41.4% from the initial level, in group 1 – by 40.1%, in group 3 – by 10.8%. Group 2 has also showed positive changes in the microbiota. Discussion. The results confirm the effectiveness of multistrain probiotics in the treatment of colic. Conclusions. Bac-Set Baby multistrain probiotic proved to be more effective than BioGaia Probiotic drops for baby monostrain probiotic and symptomatic therapy in the relief of infantile colic. The effect of Bac-Set Baby is associated with its ability to normalize the intestinal microbiota and reduce the permeability of the intestinal wall.
... Crosstalk between the gut and the lungs is strongly correlated with lung disease severity, and the main direction of this crosstalk is from the gut to the lungs, although it cannot be ruled out that it occurs in the opposite direction [6,[27][28][29]. Previous studies have shown that L. reuteri has significant potential for supporting intestinal epithelial regeneration, repairing the intestinal barrier, remodeling the intestinal microbiota, and inhibiting the production of proinflammatory factors, among many other activities [14,30,31]. However, the molecular mechanisms by which L. reuteri ameliorates ALI symptoms remain unclear. ...
Article
Full-text available
Acute lung injury (ALI) causes lung inflammation and edema as well as resulting in gut microbiota disorder. Probiotics, however, can improve the gut microbiota composition and modulate its immune response, playing an important role in ALI pathogenesis. Therefore, our study aims to investigate the effect of Lactobacillus reuteri on Lipopolysaccharide (LPS)-induced ALI in mice and to probe the mechanism of its synergistic modulatory effect on the lungs and intestines. We assessed the therapeutic effects of L. reuteri in the ALI mouse model by histopathology, alveolar lavage fluid and serum inflammatory factor analysis and explored microbiome and transcriptome alterations. L. reuteri intervention effectively attenuated lung tissue injury and significantly reduced the LPS-induced inflammatory response and macrophage and neutrophil infiltration. Additionally, L. reuteri improved the intestinal barrier function and remodeled the disordered microbiota. In conclusion, our study showed that L. reuteri attenuated the inflammatory response, ameliorated the pulmonary edema, repaired the intestinal barrier, and remodeled the gut microbiota in ALI mice. This study provides new perspectives on the clinical treatment of ALI.
Article
Full-text available
The gut microbiome has emerged as a potential target for the treatment of cardiovascular disease. Ischemia/reperfusion (I/R) after myocardial infarction is a serious complication and whether certain gut bacteria can serve as a treatment option remains unclear. Lactobacillus reuteri (L. reuteri) is a well‐studied probiotic that can colonize mammals including humans with known cholesterol‐lowering properties and anti‐inflammatory effects. Here, the prophylactic cardioprotective effects of L. reuteri or its metabolite γ‐aminobutyric acid (GABA) against acute ischemic cardiac injury caused by I/R surgery are demonstrated. The prophylactic gavage of L. reuteri or GABA confers cardioprotection mainly by suppressing cardiac inflammation upon I/R. Mechanistically, GABA gavage results in a decreased number of proinflammatory macrophages in I/R hearts and GABA gavage no longer confers any cardioprotection in I/R hearts upon the clearance of macrophages. In vitro studies with LPS‐stimulated bone marrow‐derived macrophages (BMDM) further reveal that GABA inhibits the polarization of macrophages toward the proinflammatory M1 phenotype by inhibiting lysosomal leakage and NLRP3 inflammasome activation. Together, this study demonstrates that the prophylactic oral administration of L. reuteri or its metabolite GABA attenuates macrophage‐mediated cardiac inflammation and therefore alleviates cardiac dysfunction after I/R, thus providing a new prophylactic strategy to mitigate acute ischemic cardiac injury.
Article
Full-text available
The imbalance of microbial composition and diversity in favor of pathogenic microorganisms combined with a loss of beneficial gut microbiota taxa results from factors such as age, diet, antimicrobial administration for different infections, other underlying medical conditions, etc. Probiotics are known for their capacity to improve health by stimulating the indigenous gut microbiota, enhancing host immunity resistance to infection, helping digestion, and carrying out various other functions. Concurrently, the metabolites produced by these microorganisms, termed postbiotics, which include compounds like bacteriocins, lactic acid, and hydrogen peroxide, contribute to inhibiting a wide range of pathogenic bacteria. This review presents an update on using probiotics in managing and treating various human diseases, including complications that may emerge during or after a COVID-19 infection.
Article
A radiation-induced brain injury (RIBI) is a major adverse event following radiotherapy of malignant tumors. RIBI would affect cognitive function, leading to a series of complications and even death. However, the pathogenesis of RIBI is still unclear, and it still lacks specific therapeutic drugs. The gut-brain bidirectional communication may be mediated by various microbiota and metabolites in the gastrointestinal tract. Probiotics are closely related to physiological health. The theory of the gut-brain axis provides us with a new idea to improve the gut microenvironment by supplementing probiotics against RIBI. Here, Lactobacillus reuteri microcapsules (LMCs) were prepared, which were predominantly irregular spheres with a rough surface under a scanning electron microscope and a narrow size distribution ranging from 20 to 700 μm. The transmission electron microscopy images showed that the structure of microcapsules containing Lactobacillus reuteri (L. reuteri) was a core and shell structure. The survival of L. reuteri in microcapsules was significantly more than that of free L. reuteri in the simulated stomach environment of pH 1.2. 16S rDNA sequencing showed that LMCs observably increased the relative abundance of Lactobacillus in RIBI mice. More importantly, compared with the RIBI model mice, the behavior of RIBI mice treated with LMCs was significantly improved. In addition, LMCs greatly alleviated the pathological damage of the hippocampus and intestines in the mice after irradiation and reduced the level of TNF-α and IL-6 in vivo. Generally, LMCs are a promising oral preparation, which provide new ideas and methods for the treatment of RIBI.
Article
Full-text available
Lactic acid bacteria are well-known probiotics, conferring several health benefits. In this study, we isolated lactobacilli from human breast milk and identified Lactobacillus reuteri LM1071 (RR-LM1071) using 16S rDNA sequencing. We tested the hemolytic activity, biogenic amine production, and antibiotic susceptibility of this strain to assess its safety. RR-LM1071 was found to be negative for hemolytic activity and biogenic amine production, as well as was measured in susceptible level for antibiotics in the minimal inhibitory concentration (MIC) test. The adhesive properties of RR-LM1071 were higher than those of LGG in HT-29 cells, and showed a greater hydrophobicity than LGG in hexadecane solvent. Under inflammatory conditions, RR-LM1071 suppressed the mRNA expression of IL-6, TNF-α, and IL-4 produced in IL-1β-induced HT-29 cells. Our results suggest that RR-LM1071 is a safe and valuable probiotic that can be used for the treatment of inflammatory bowel disease.
Article
Full-text available
Enzyme-assisted extraction is a cost-effective, safe, and efficient method to obtain bioactives from plant materials. During this study, 10 different marine algae from Sri Lanka were individually extracted by using five commercial food-grade carbohydrases. The enzymatic and water extracts of the seaweeds were analyzed for their antioxidant and anti-inflammatory activities. The highest DPPH, hydrogen peroxide (H2O2) and intracellular H2O2 scavenging abilities were observed from the Celluclast extract of Sargassum polycystum (CSp). CSp exerted protective effects against oxidative stress-induced cell death in hydrogen peroxide-induced Chang cells and in model zebrafish. The Celluclast extract of Chnoospora minima (CCm) showed the strongest anti-inflammatory activity against lipopolysaccharide (LPS)-induced NO production in RAW 264.7 macrophages (IC50 = 44.47 µg/mL) and in model zebrafish. CCm inhibited the levels of iNOS, COX-2, PGE2, and TNF-α in LPS stimulated RAW 264.7 macrophages. Hence, CSp and CCm could be utilized in developing functional ingredients for foods, and cosmeceuticals.
Article
Full-text available
Background/aims: This study aimed to explore the metabololipidome in mice upon cupping treatment. Methods: A nude mouse model mimicking the cupping treatment in humans was established by administrating four cupping sets on the back skin for 15 minutes. UPLC-MS/ MS was performed to determine the PUFA metabolome in mice skin and blood before and after cupping treatment. The significantly changed lipids were administered in macrophages to assess the production of pro-inflammatory cytokines IL-6 and TNF-α by ELISA. Results: The anti-inflammatory lipids, e.g. PGE1, 5,6-EET, 14,15-EET, 10S,17S-DiHDoHE, 17R-RvD1, RvD5 and 14S-HDoHE were significantly increased while pro-inflammatory lipids, e.g. 12-HETE and TXB2 were deceased in the skin or plasma post cupping treatment. Cupping treatment reversed the LPS-stimulated IL-6 and TNF-α expression in mouse peritoneal exudates. Moreover, 5,6-EET, PGE1 decreased the level of TNF-α, while 5,6-EET, 5,6-DHET downregulated IL-6 production in macrophages. Importantly, 14,15-EET and 14S-HDoHE inhibited both IL-6 and TNF-α induced by lipopolysaccharide (LPS). 17-RvD1, RvD5 and PGE1 significantly reduced the LPS-initiated TNF-α, while TXB2 and 12-HETE further upregulated the LPS-enhanced IL-6 and TNF-α expression in macrophages. Conclusion: Our results reveal the identities of anti-inflammatory versus pro-inflammatory metabolipidome and suggest the potential therapeutic mechanism of cupping treatment.
Article
Full-text available
Three hundred and sixty presumptive lactic acid bacteria (LAB) isolated from pregnant sows, newborn, suckling, and weaned piglets were preliminarily screened for anti-Salmonella activity. Fifty-eight isolates consisting of Lactobacillus reuteri (n = 32), Lactobacillus salivarius (n = 10), Lactobacillus mucosae (n = 8), Lactobacillus johnsonii (n = 5), and Lactobacillus crispatus (n = 3) were selected and further characterized for probiotic properties including production of antimicrobial substances, acid and bile tolerance, and cell adherence to Caco-2 cells. Eight isolates including Lact. johnsonii LJ202 and Lact. reuteri LR108 were identified as potential probiotics. LJ202 was selected for further use in co-culture studies of two-bacterial and multiple-bacterial species to examine its inhibitory activity against Salmonella enterica serovar Enteritidis DMST7106 (SE7106). Co-culture of LJ202 and SE7106 showed that LJ202 could completely inhibit the growth of SE7106 in 10 h of co-culture. In co-culture of multiple-bacterial species, culturable fecal bacteria from pig feces were used as representative of multiple-bacterial species. The study was performed to examine whether interactions among multiple-bacterial species would influence antagonistic activity of LJ202 against SE7106 and fecal coliform bacteria. Co-culture of SE7106 with different combinations of fecal bacteria and probiotic (LJ202 and LR108) or non-probiotic (Lact. mucosae LM303) strains revealed that the growth of SE7106 was completely inhibited either in the presence or in the absence of probiotic strains. Intriguingly, LJ202 exhibited notable inhibitory activity against fecal coliform bacteria while LR108 did not. Taken together, the results of co-culture studies suggested that LJ202 is a good probiotic candidate for further study its inhibitory effects against pathogen infections in pigs.
Article
Probiotics are known to provide the host with immune-modulatory effects and are therefore of remarkable interest for therapeutic and prophylactic applications against various disorders, including inflammatory diseases. Weissella cibaria JW15 (JW15) has been reported to possess probiotic and antioxidant properties. However, the effect of JW15 on inflammatory responses has not yet been reported. Therefore, the objective of current study was to evaluate the anti-inflammatory potential of JW15 against lipopolysaccharide (LPS) stimulation. The production of pro-inflammatory factors and the cellular signaling pathways following treatment with heat-killed JW15 was examined in LPS-induced RAW 264.7 cells. Treatment with heat-killed JW15 decreased nitric oxide and prostaglandin E2 production via down-regulation of the inducible nitric oxide synthase and cyclooxygenase-2. In addition, treatment with heat-killed JW15 suppressed the expression of pro-inflammatory cytokines, interleukin (IL)-1β, IL-6, and tumor necrosis factor-α. The anti-inflammatory properties treating with heat-killed JW15 were associated with mitogen-activated protein kinases signaling pathway-mediated suppression of nuclear factor-κB. These results indicated that JW15 possesses anti-inflammatory potential and provide a molecular basis regarding the development of functional probiotic products.
Article
This study aimed to screen lactic acid bacteria (LAB) for their anti-inflammatory activity by using RAW264.7 cells and dextran sulfate sodium (DSS)-induced colitis. In all, 192 LAB strains were isolated from healthy human feces, of which 8 strains showed excellent nitric oxide (NO) inhibitory activity. Peptidoglycan extracts of these 8 LAB strains were subjected to NO assay, Western blot, and ELISA. Among the 8 tested strains, extracts of 4 strains significantly inhibited the production of NO, related enzyme activities such as inducible nitric oxide synthase and cyclooxygenase 2, and key cytokines such as tumor necrosis factor-α and IL-6 in RAW264.7 cells. The 4 strains belonged to Lactobacillus (CAU1054, CAU1055, CAU1064, and CAU1301). Oral administration of the 4 strains inhibited DSS-induced body weight loss, colon shortening, and colon damage in ICR mice. The colon tissue of the mice treated with Lactobacillus plantarum strain CAU1055 had significantly reduced levels of inducible nitric oxide synthase, cyclooxygenase 2, tumor necrosis factor-α, and IL-6. We found that strain CAU1055 could be used as a candidate probiotic strain for the prevention and treatment of inflammatory bowel disease. Further studies are warranted to confirm the mechanisms of interaction between peptidoglycan of L. plantarum strain CAU1055 and upstream cellular signaling mediators.
Article
Macrophages are tissue-resident or infiltrated immune cells critical for innate immunity, normal tissue development, homeostasis, and repair of damaged tissue. Macrophage function is a sum of their ontogeny, the local environment in which they reside, and the type of injuries or pathogen to which they are exposed. In this Review, we discuss the role of macrophages in the restoration of tissue function after injury, highlighting important questions about how they respond to and modify the local microenvironment to restore homeostasis.
Article
Tissue injury triggers a complex series of cellular responses, starting from inflammation activated by tissue and cell damage and proceeding to healing. By clearing cell debris, activating and resolving inflammation, and promoting fibrosis, macrophages play key roles in most, if not all, phases of the response to injury. Recent studies of the mechanisms underlying the initial inflammation and later tissue regeneration and repair revealed that macrophages bridge these processes in part by supporting and activating stem/progenitor cells, clearing damaged tissue, remodeling extracellular matrix to prepare scaffolding for regeneration, and promoting angiogenesis. However, macrophages also have a central role in the development of pathology induced by failed resolution (e.g., chronic inflammation) and excessive scarring. In this review we summarize the activities of macrophages in inflammation and healing in response to acute injury in tissues with differing regenerative capacities. While macrophages lead similar processes in response to tissue injury in these tissues, their priorities and the consequences of their activities differ among tissues. Moreover, the magnitude, nature, and duration of injury also greatly affect cellular responses and healing processes. In particular, continuous injury and/or failed resolution of inflammation leads to chronic ailments in which macrophage activities may become detrimental.
Article
The objective of our research was to evaluate the molecular mechanism of the anti-inflammatory effects of surface layer protein (Slp) derived from Lactobacillus acidophilus NCFM in lipopolysaccharide-induced RAW264.7 cells. Our results presented that Slp, with an apparent size of 46 kDa, attenuated the production of TNF-α, IL-1β, and reactive oxygen species (ROS), by inhibiting the MAPK and NF-κB signaling pathways. In addition, 10 μg mL-1 of Slp significantly inhibited NO and PGE2 production (P < 0.001) through down-regulating the expression levels of iNOS and COX-2 protein. Furthermore, Slp was found to inhibit NF-κB p65 translocation into the nucleus to activate inflammatory gene transcription. These findings suggest that Slp is a potential immune-modulating bioactive protein derived from probiotics and holds promise for use as an additive in functional foods.
Article
Lactobacillus species are found in nutrient-rich habitats associated with food, feed, plants, animals and humans. Due to their economic importance, the metabolism, genetics and phylogeny of lactobacilli have been extensively studied. However, past research primarily examined lactobacilli in experimental settings abstracted from any natural history, and the ecological context in which these bacteria exist and evolve has received less attention. In this review, we synthesize phylogenetic, genomic and metabolic metadata of the Lactobacillus genus with findings from fine-scale phylogenetic and functional analyses of representative species to elucidate the evolution and natural history of its members. The available evidence indicates a high level of niche conservatism within the well-supported phylogenetic groups within the genus, with lifestyles ranging from free-living to strictly symbiotic. The findings are consistent with a model in which host-adapted Lactobacillus lineages evolved from free-living ancestors, with present-day species displaying substantial variations in terms of the reliance on environmental niches and the degree of host specificity. This model can provide a framework for the elucidation of the natural and evolutionary history of Lactobacillus species and valuable information to improve the use of this important genus in industrial and therapeutic applications.