ArticlePDF Available

Bacillus coagulans MZY531 alleviates intestinal mucosal injury in immunosuppressive mice via modulating intestinal barrier, inflammatory response, and gut microbiota

Springer Nature
Scientific Reports
Authors:

Abstract and Figures

Bacillus coagulans has a potential role in improving intestinal injury. However, the specific mechanism is still unclear. In this study, the protective effect of B. coagulans MZY531 on intestinal mucosa injury in cyclophosphamide (CYP)-induced immunosuppressed mice were investigated. The results indicated that the immune organ (thymus and spleen) indices of B. coagulans MZY531 treatment groups were significantly increased compared to the CYP group. B. coagulans MZY531 administration promotes the expression of immune proteins (IgA, IgE, IgG, and IgM). B. coagulans MZY531 could upregulate the ileum levels of IFN-γ, IL-2, IL-4, and IL-10 in immunosuppressed mice. Moreover, B. coagulans MZY531 restores the villus height and crypt depth of the jejunum and alleviates injury of intestinal endothelial cells caused by CYP. Furthermore, the western blotting results showed that B. coagulans MZY531 ameliorated CYP-induced intestinal mucosal injury and inflammatory via up-regulates the ZO-1 pathway and down-regulates the expression of the TLR4/MyD88/NF-κB pathway. After treatment with B. coagulans MZY531, the relative abundance of Firmicutes phylum was dramatically increased, as well as the genera of Prevotella and Bifidobacterium, and reducing harmful bacteria. These findings suggested that B. coagulans MZY531 has a potential immunomodulatory activity on chemotherapy-induced immunosuppression.
This content is subject to copyright. Terms and conditions apply.
1
Vol.:(0123456789)
Scientic Reports | (2023) 13:11181 | https://doi.org/10.1038/s41598-023-38379-0
www.nature.com/scientificreports
Bacillus coagulans MZY531
alleviates intestinal mucosal
injury in immunosuppressive mice
via modulating intestinal barrier,
inammatory response, and gut
microbiota
Zhongwei Zhao
1,2, Manqing Sun
1, Xinmu Cui
1, Jiaxin Chen
1, Chunhong Liu
2 &
Xuewu Zhang
1*
Bacillus coagulans has a potential role in improving intestinal injury. However, the specic mechanism
is still unclear. In this study, the protective eect of B. coagulans MZY531 on intestinal mucosa injury
in cyclophosphamide (CYP)-induced immunosuppressed mice were investigated. The results indicated
that the immune organ (thymus and spleen) indices of B. coagulans MZY531 treatment groups were
signicantly increased compared to the CYP group. B. coagulans MZY531 administration promotes
the expression of immune proteins (IgA, IgE, IgG, and IgM). B. coagulans MZY531 could upregulate
the ileum levels of IFN-γ, IL-2, IL-4, and IL-10 in immunosuppressed mice. Moreover, B. coagulans
MZY531 restores the villus height and crypt depth of the jejunum and alleviates injury of intestinal
endothelial cells caused by CYP. Furthermore, the western blotting results showed that B. coagulans
MZY531 ameliorated CYP-induced intestinal mucosal injury and inammatory via up-regulates
the ZO-1 pathway and down-regulates the expression of the TLR4/MyD88/NF-κB pathway. After
treatment with B. coagulans MZY531, the relative abundance of Firmicutes phylum was dramatically
increased, as well as the genera of Prevotella and Bidobacterium, and reducing harmful bacteria.
These ndings suggested that B. coagulans MZY531 has a potential immunomodulatory activity on
chemotherapy-induced immunosuppression.
Normal gastrointestinal function is essential for the absorption of nutrients, while gastrointestinal changes can
lead to serious defects in the intestinal barrier and gastrointestinal diseases1,2. Tight junction proteins, includ-
ing ZO-1, occludin, and claudin-1, constitute intestinal epithelial cells as a physical barrier, and their complex
interactions maintain the integrity of the intestinal barrier and reduce intestinal permeability3. Intestinal leakage
can activate immune cells to secrete inammatory cytokines, which in turn increases intestinal permeability and
causes systemic inammation4. In addition, microorganisms in the gastrointestinal tract can also damage the
intestinal barrier function by promoting mucosal reaction, reducing the expression of tight junction proteins.
Aer reaching the blood, bacteria can trigger inammation by binding TLR4 receptors expressed on the liver
through the portal vein5,6.
Cyclophosphamide (CYP) was an alkylated anticancer agent which was widely used in the treatment of vari-
ous cancers. However, long-term treatment of CYP may could result various side eects such as acute cytotoxicity,
immunosuppression, and gastrointestinal mucosal barrier damage. To improve this situation, some protective
agents are increasingly being used to alleviate adverse side eects in chemotherapy patients. Certain anti-para-
sitics have also been reported to have an immunomodulatory activity such as levamisole. Levamisole is a drug
widely used to enhance the immunity of various human diseases, including leprosy, rheumatoid arthritis, and
in adjuvanted therapy of colorectal cancer. In recent years, more and more attention has been paid to alleviating
CYP induced immunosuppression based on gut microbiota regulation. In 2013, Viaud etal. demonstrated that
OPEN
1Medical College, Yanbian University, Yanji 133002, Jilin, People’s Republic of China. 2College of Special Education,
Changchun University, Changchun 130022, People’s Republic of China. *email: zhangxuewu@ybu.edu.cn
Content courtesy of Springer Nature, terms of use apply. Rights reserved
2
Vol:.(1234567890)
Scientic Reports | (2023) 13:11181 | https://doi.org/10.1038/s41598-023-38379-0
www.nature.com/scientificreports/
CYP alters microbiota composition in the small intestine and induces the translocation of selected species of
Gram-positive bacteria into secondary lymphoid organs7. A few years later, Xie etal. discovered that Lactobacil-
lus plantarum intervention could protect the intestinal mucosal injury and intestinal barrier function in mice
induced by CYP by regulating intestinal ora imbalance8. erefore, regulating intestinal ora, inammation,
and intestinal barrier may be a novel potential therapeutic strategy for the treatment of the intestinal injury.
Probiotics are considered a novel choice to reduce the side eects of chemotherapy on patients. Probiotics
can maintain the balance of intestinal microorganisms and protect the intestinal integrity of epithelial cells by
increasing the mucus layer and the expression of TJ9. Bacillus coagulans, as a probiotic, can stay in the gut for a
longer time; it also has a strong adhesive ability and signicant immunomodulatory eect10. It has been reported
that some active components in the fermentation supernatant of B. coagulans can form a biological protective
barrier in the human intestinal tract, promote the immune response of the digestive tract mucosa, and thus
improve intestinal immunity11. In addition, B. coagulans 13002 can stimulate the growth of Bidobacterium and
Lactobacillus and reduce the intestinal side eects caused by cyclophosphamide12. Furthermore, B. coagulans
TL3 can protect rats from inammation caused by endotoxin and inhibit the reproduction of harmful bacteria by
blocking the expression of the TLR4 pathway so as to enhance intestinal immunity13. Moreover, previous stud-
ies have shown that B. coagulans 13002 and fructooligosaccharides signicantly reduce CYP-induced intestinal
mucosal damage and improve immune function by regulating intestinal microora12. In this study, we exam-
ined the eects of B. coagulans MZY531 (MZY531) on intestinal mucosal injury, inammation, and intestinal
microora induced by CYP in mice and explored its protective mechanism. ese data provide a theoretical
basis for the development and utilization of B. coagulans and support its addition to functional foods to improve
intestinal health.
Materials and methods
Preparation of bacterial strain. B. coagulans MZY531 is a probiotic strain isolated from naturally fer-
mented kimchi and stored in China Center for Type Culture Collection (CCTCC, accession M2021622, Wuhan,
China). B. coagulans MZY531 was inoculated in LB liquid medium and cultured in a constant temperature
vibration incubator for 48h (180 r/min, 50°C). en the culture medium was centrifuged (2000×g, 10min) and
washed three times with aseptic phosphate-buered saline (PBS, pH 7.4) to remove the residual medium and
collect bacteria. Next, the bacteria were resuspended in saline solution, and the concentration was adjusted to
1.0 × 109CFU/mL, which was stored at 4°C for subsequent intragastric administration of mice.
Animals and experimental design. Immunosuppressive model was induced by CYP according to pre-
vious study14. A total of 40 7-week-old female BALB/C mice were purchased from Changchun Yisi Experi-
mental Animal Technology Co., Ltd. (Changchun, China). All mice were kept in a suitable environment with
a temperature of 22 ± 1°C, relative humidity of 50 ± 1%, and a light/dark cycle of 12h, and had free access to
water and food. All animal studies (including the mice euthanasia procedure) were done in compliance with the
regulations and guidelines of the Jilin Academy of Agricultural Sciences institutional animal care and conducted
according to the AAALAC and the IACUC guidelines.
e experimental animal protocol is shown in Fig.1A. Aer 1week of adaptation, the mice were randomly
divided into four groups (n = 10 in each group): Control group, CYP group, CYP + LH group, and CYP + MZY531
group. e body weights of the mice were measured twice every week. e CYP + LH group was given 40mg/
kg levamisole hydrochloride (LH), the CYP + MZY531 group was given B. coagulans MZY531, and the Control
and CYP groups were given the same dose of normal saline. All mice were given oral administration according
to the volume of 0.1mL/10g for 14days, once daily. e immunosuppression mouse model induced by CTX
was established according to the previous method. CYP (50mg/kg/days) was intraperitoneally injected into
the mice in CYP, CYP + LH, and CYP + MZY531 groups on days 15 and 16. Control group was administered
intraperitoneally with the same volume of physiological saline. Aer the last injection, the mice were starved
for 24h but given free access to water. e mice were sacriced by cervical dislocation, and the jejunum, ileum,
spleen, and feces were collected.
Determination of immune organ index. Before the mice were killed, the nal weight of the mice was
recorded. en, the thymus and spleen tissues were immediately dissected, washed in precooled normal saline
at 4°C, dried using lter paper, and weighed. e spleen and thymus index were calculated according to the fol-
lowing formula: spleen and thymus index = spleen and thymus weight (mg)/nal weight (g)15.
Determination of immune and inammatory factors in the ileum. e ileum of mice was quickly
collected and placed in an ice bath. An appropriate amount of ileum tissue was then selected and mixed with
normal saline according to the proportion of 1:9 to prepare 10% tissue homogenate. Next, the homogenate was
centrifuged (4000×g, 10min) at 4°C, and the supernatant was collected. e immunoglobulin (IgA, IgE, IgG,
and IgM) and inammatory factors (IL-2, IFN-γ, IL-4, and IL-10) concentrations were detected using ELISA
kits according to the instructions of Jiangsu Enzymatic Biology Co., Ltd. (Jiangsu, China). e optical density
(OD) value of the solution was measured at 450nm using an automatic microplate reader.
Pathological observation of jejunum. e fresh jejunum tissue was washed with normal saline, xed in
4% paraformaldehyde for 48h, embedded in paran, and prepared into 8-μm slices. en, samples were stained
with hematoxylin–eosin (H&E) for 5min, dehydrated, and sealed with neutral glue. e pathological changes in
the jejunum of each group were observed under light microscope (Nikon Corporation, Tokyo, Japan), and the
Content courtesy of Springer Nature, terms of use apply. Rights reserved
3
Vol.:(0123456789)
Scientic Reports | (2023) 13:11181 | https://doi.org/10.1038/s41598-023-38379-0
www.nature.com/scientificreports/
villus length (V) and crypt depth (C) were recorded16. Moreover, the images were acquired at a magnication
of × 25 and × 200 (Supplementary Information).
Western blotting. e ileal tissue was lysed by RIPA kit, and the supernatant was collected. e BCA
kit determined the total protein concentration in the supernatant. Samples were then mixed with the protein
sample buer at 1:1 and boiled in a water bath for 8min to collect the proteins, which were then separated by
sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to polyvinylidene uo-
ride (PVDF) membrane. e membrane was blocked for 60min in TBST solution containing 3% bovine serum
albumin (BSA) and then incubated with rabbit anti-ZO-1, Occludin, Claudin-1, TLR4, MyD88, NF-κB, IKBα,
and β-actin overnight at 4°C. Samples were then incubated with horseradish peroxidase (HRP) labeled second-
ary antibody for 60min at room temperature. Samples were then washed with TBST three times, and the protein
expression was detected by an enhanced chemiluminescence reagent. e gray values of the bands were detected
by Image Quant LAS 4000 (Shanghai, China) and standardized by β-actin.
Gut microbial analysis. Fresh fecal samples of the cecum were immediately frozen in liquid nitrogen and
stored at 80°C. QIAamp Fast DNA kit was used to extract total DNA from feces. According to the previous
study17, the V3 and V4 regions of 16S rDNA were amplied by universal primers using polymerase chain reac-
tion (PCR). en the amplied products were sequenced by Illumina MiSeq, and the sequences of high quality
with 97% similarity were incorporated into a taxon on QIIME soware, and the diversity of gut microbiota was
analyzed. e Chao1, Shannon, Simpson, and Pielou-e indices were used to investigate α diversity. e principal
coordinate method of weighted UniFrac phylogenetic distance matrix was used to analyze β diversity; the rela-
tive abundance at the gate level was used to indicate the dierence in bacterial colony structure among groups,
and the heat map analysis showed the dierence in dierent microorganisms at the genus level. In addition,
Spearman’s analysis revealed the correlation between gut microbiota and immune and inammatory levels in
mice. e original data and sequencing sample data obtained in this study can be obtained from the National
Center forBiotechnology Information (NCBI) database with the registration number: PRJNA884309.
Statistical analysis. All the experimental data were expressed as mean ± standard deviation (SD). SPSS20.0
and Origin8.0 were used for data processing and analysis. e overall signicant dierence was evaluated by
single factor analysis of variance (ANOVA) and Tukey multiple comparisons. A P value < 0.05 was considered to
be statistically signicant.
Institutional review board statement. e animal experiment procedures were approved by the Com-
mittee of Animal Experimental Ethical Inspection of Laboratory Animal Centre, Yanbian University (approved
number: SCXK-2020-0001).
ARRIVE guidelines. All the research methods contained in the manuscript are carried out in accordance
with the requirements of ARRIVE.
Figure1. A schedule of experimental procedures (A) and eects of B. coagulans MZY531 on spleen (B) and
thymus (C) index in immunosuppressed mice. All data were statistically analyzed using a one-way analysis of
variance and Tukey multiple comparison. #P < 0.05 and ##P < 0.01 vs. control group; *P < 0.05 and **P < 0.01 vs.
CYP group.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
4
Vol:.(1234567890)
Scientic Reports | (2023) 13:11181 | https://doi.org/10.1038/s41598-023-38379-0
www.nature.com/scientificreports/
Results
B. coagulans MZY531 increases the immune organ index of intestinal injury mice. As shown in
Fig.1, the intervention of CYP signicantly decreased the spleen (Fig.1B) and thymus (Fig.1C) index of mice
compared with the control group, while the treatment of LH and B. coagulans MZY531 signicantly increased
the spleen and thymus index (all P < 0.01). Aer B. coagulans MZY531 treatment, the spleen and thymus index
increased by 71.36% and 69.39%, respectively, compared with the CYP group (all P < 0.01). ese results indi-
cate that B. coagulans MZY531 could eectively alleviate immune organ atrophy induced by CYP. Additionally,
the spleen and thymus indexes of the B. coagulans MZY531 group were higher than in the CYP group, with the
indexes being close to those of the LH group. ese ndings suggest that B. coagulans MZY531 plays a crucial
role in preventing the atrophy of immune organs.
B. coagulans MZY531 increases the level of immune protein in the ileum of intestinal injury
mice. e results showed that (Fig.2A–D), the induction of CYP signicantly decreased the levels of IgG,
IgM, IgA, and IgE by 47.75%, 46.92%, 38.15%, and 39.50%, respectively, compared with the control group (all
P < 0.01). e levels of IgG, IgM, IgA, and IgE in the B. coagulans MZY531 treatment were signicantly higher
than those in the CYP group (P < 0.05), approaching the values of the control group. Aer the B. coagulans
MZY531 treatment, the levels of IgG and IgM were similar to those of the positive control LH group (P > 0.05).
ese results showed that the treatment of B. coagulans MZY531 could reverse the decrease of immune protein
level induced by CYP and improve the immunity of mice.
B. coagulans MZY531 increases the level of anti-inammatory cytokines in the ileum of intes-
tinal injury mice. As shown in Fig.3A–D, the levels of IFN-γ, IL-2, IL-4, and IL-10 in the CYP group were
signicantly lower than those in the control group (P < 0.05), indicating that CYP could signicantly inhibit
the production of anti-inammatory cytokines. Compared with the CYP group, the treatment of B. coagu-
lans MZY531 signicantly increased the levels of IFN-γ, IL-2, IL-4, and IL-10 by 18.86%, 89.03%, 29.81%, and
51.14%, respectively (P < 0.05). e above results also showed that B. coagulans MZY531 could improve the
anti-inammatory ability of CYP-induced intestinal injury model mice. Furthermore, B. coagulans MZY531,
in particular, signicantly improved the secretion of IL-10. ese results indicated that B. coagulans MZY531
could improve inammatory responses by increasing the secretion of anti-inammatory cytokines in ileum of
CYP-induced immunosuppressed mice.
Figure2. Eects of B. coagulans MZY531 on the levels of immune proteins level in ileum of mice with
intestinal injury induced by CYP. IgG (A); IgM (B); IgA (C) and IgE (D). All data were statistically analyzed
using a one-way analysis of variance and Tukey multiple comparison. #P < 0.05 and ##P < 0.01 vs. control group;
*P < 0.05 and **P < 0.01 vs. CYP group.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
5
Vol.:(0123456789)
Scientic Reports | (2023) 13:11181 | https://doi.org/10.1038/s41598-023-38379-0
www.nature.com/scientificreports/
B. coagulans MZY531 improves the histomorphological changes of jejunum in intestinal injury
mice. e results of HE staining of the jejunum of mice are shown in Fig.4A, while no obvious pathologi-
cal changes in jejunum were found in the blank group. In the CYP group, the jejunal villi were shortened and
exfoliated (black arrow), the intestinal epithelium of the local mucous layer was missing, the lamina propria was
exposed (yellow arrow), slight edema could be seen locally, and the gap between the intestinal epithelium and
lamina propria was seen (red arrow). However, the villi length increased, and the intestinal epithelial structure
was signicantly recovered in LH and B. coagulans MZY531 groups. In addition, the treatment of MZY531
signicantly increased villus length (Fig.4B), crypt depth (Fig.4C), and the V/C ratio (Fig.4D), which were
153.85%, 26.44%, and 101.37%, respectively, higher than those of the CYP group (all P < 0.01). ese results
also suggested that B. coagulans MZY531 could improve the pathological intestinal damage in CYP-induced
intestinal injury model mice.
B. coagulans MZY531 improves intestinal barrier function in intestinal injury mice. e eect
of B. coagulans MZY531 on proteins (ZO-1, occludin, and claudin-1) of the intestinal barrier pathway in the
ileum of mice was evaluated by Western blotting. As shown in Fig.5, compared with the blank group, the induc-
tion of CYP signicantly decreased the protein levels of ZO-1, occludin, and claudin-1 (all P < 0.05). Compared
with CYP group, Intervention with B. coagulans MZY531 signicantly increased the protein expression of ZO-1,
occluding, and claudin-1 by 61.87% (P < 0.01), 47.80% (P < 0.05), and 17.32% (P < 0.05), respectively. Based on
this result, B. coagulans MZY531 could repair the intestinal barrier damage induced by CYP in mice.
B. coagulans MZY531 inhibits the level of inammation in intestinal injury mice. e results
presented in Fig.6 shows that the expression of TLR4 inammatory pathway protein in mouse jejunum. e
induction of CYP signicantly upregulated the levels of TLR4, MyD88, NF-κB, and IKBα. On the contrary,
the treatment of LH and B. coagulans MZ531 signicantly inhibited the expression of TLR4, MyD88, NF-κB
and IKBα, and the level of MZY531 group was down-regulated by 34.55% (P < 0.01), 29.95% (P < 0.05), 20.71%
(P < 0.01), and 52.36% (P < 0.01) compared with CYP group, respectively. us, these results revealed that the
treatment of B. coagulans MZY531 markedly resisted the expression of intestinal inammation induced by CYP
through downregulation of TLR4/MyD88/NF-κB inammatory signaling pathways.
B. coagulans MZY531 remodels the intestinal microora of intestinal injury mice. We evalu-
ated the eect of B. coagulans MZY531 on gut microbiota by 16SrRNA high-throughput sequencing. e results
of α diversity showed that (Table1), the indexes of Chao1, Shannon, Simpson, and Pielou-e in the B. coagulans
Figure3. Eects of B. coagulans MZY531 on the levels of anti-inammatory factors in ileum of
immunosuppressed mice. IFN-γ (A); IL-2 (B); IL-4 (C) and IL-10 (D). All data were statistically analyzed
using a one-way analysis of variance and Tukey multiple comparison. #P < 0.05 and ##P < 0.01 vs. control group;
*P < 0.05 and **P < 0.01 vs. CYP group.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
6
Vol:.(1234567890)
Scientic Reports | (2023) 13:11181 | https://doi.org/10.1038/s41598-023-38379-0
www.nature.com/scientificreports/
MZY531 group were signicantly higher than those in the CYP group (all P < 0.01), which suggested that the
intervention of B. coagulans MZY531 increased the richness and diversity of gut microbiota. Venn diagram
(Fig.7A) further showed that the four groups shared 273 OTU, while the number of unique OTU in the blank
group, CYP group, LH group, and MZY531 group was 451, 549, 783, and 666, respectively, indicating that the
treatment of B. coagulans MZY531 increases the number of OTU induced by CYP. In addition, in PCoA analysis
(Fig.7B), the CYP group was far away from the blank group, LH group, and MZY531 group, while the treatment
of LH and B. coagulans MZY531 made the diversity of gut microbiota of mice more inclined to the blank group.
In order to further evaluate the specic changes in gut microbiota, we investigated the relative abundance of gut
microbiota (Fig.7C) at the gate level. We found that B. coagulans MZY531 treatment increased Firmicutes but
decreased Bacteroidetes abundance. In genus-level thermographic analysis (Fig.7D), the intervention of B. coag-
ulans MZY531 increased the abundance of probiotic, including Lactobacillus, Prevotella and Bidobacterium,
and decreased the level of harmful bacteria Odoribacter and Shigella. ese results showed that the intervention
of B. coagulans MZY531 could reshape the structure of gut microbiota, increase the abundance of probiotics, and
reduce the level of pathogenic bacteria.
Next, Spearman analysis was used to analyze the correlation between single strains of gut microbiota and
immune and anti-inammatory proteins in mice (Fig.7E), revealing that Prevotella and Bidobacterium were
positively correlated with immune proteins, including IgG, IgM, IgA, and IgE, and anti-inammatory factors,
including IFN-γ, IL-2, IL-4, and IL-10, while Bacteroidetes and Odoribacter was negatively correlated with
immune proteins and anti-inammatory factors. ese results further reveal that the intervention of B. coagu-
lans MZY531 could increase the immunity and anti-inammatory ability of mice by increasing the abundance
of probiotics.
Discussion
CYP is a widely used chemotherapeutic drug for cancer treatment. Yet, CYP can seriously damage the body’s
immunity and induce the disorder of intestinal microora, thus increasing the risk of immune deciency and
intestinal injury diseases18. It has been found that probiotics can improve the function of intestinal microora
by regulating the value of specic microora in the intestinal tract, thus having a benecial eect on the body19.
Figure4. Eects of B. coagulans MZY531 on the histomorphological changes of jejunum in immunosuppressed
mice. e pathological changes of jejunum (magnication × 25 and × 200) (A). Black arrows indicated the
shorterning of intestinal villi, yellow arrows indicated exposed lamina propria, and black arrows indicated mild
edema and enlarged spaces. Villus height (B), Crypt depth (C). e ratio of villus height to crypt depth (D). All
data were statistically analyzed using a one-way analysis of variance and Tukey multiple comparison. #P < 0.05
and ##P < 0.01 vs. control group; *P < 0.05 and **P < 0.01 vs. CYP group.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
7
Vol.:(0123456789)
Scientic Reports | (2023) 13:11181 | https://doi.org/10.1038/s41598-023-38379-0
www.nature.com/scientificreports/
erefore, we speculate that probiotics may be a new therapeutic way to alleviate CYP-induced intestinal injury.
is study focused on the protective eect of B. coagulans MZY531 on intestinal injury induced by CYP.
As important immune organs, spleen and thymus have an important role in regulating systemic immune
function20. Our study showed that the intervention of B. coagulans MZY531 signicantly increases the spleen and
thymus index of mice induced by CYP. Coincidentally, Awad etal. reported that the use of probiotics increases the
spleen and thymus index of broilers21; similar results were obtained by Kabir etal.22. In addition, the occurrence
of immune dysfunction seems to be regulated by IFN-γ and IL-4 levels, and the decrease of their levels may lead
to impaired immune function23. Studies have shown that CYP, as an eective immunosuppressant, can induce the
decrease of IFN-γ and IL-4 levels, thus destroying immune homeostasis and leading to immunosuppression24.
It is worth noting that B. coagulans MZY531 treatment signicantly increases the levels of IFN-γ and IL-4 and
increases the expression of immune-related cytokines, including IgG, IgM, IgA, and IgE. Furthermore, Bomko
etal. found that B. coagulans had normalised both the quantitative parameters of the immune system and the
cells’ functional activity by decreasing the level of immune-related cytokines25. is study further suggests that
treating B. coagulans MZY531 can resist the immune function damage induced by CYP by improving the func-
tion of immune organs and the level of immune protein.
As the main food digestive organ most easily aected by foreign antigens or microorganisms, the intesti-
nal tract is the rst line of defense against pathogenic microorganisms26. A harmful environment may induce
oxidation and inammation in the intestinal tract, damaging intestinal mucosal. Studies have shown that CYP
can induce the shortening and shedding of intestinal villi and, in turn, lead to intestinal mucosal damage27. In
this study, the pathological results showed that the treatment of B. coagulans MZY531 signicantly reduces the
shedding of intestinal villi, increases the crypt depth, and reduces the edema of intestinal endothelial cells, which
indicates that the intervention of B. coagulans MZY531 could alleviate the intestinal mucosal injury induced by
CYP. It is worth noting that the impairment of intestinal mucosal barrier function seems to be the main inducing
factor of intestinal mucosal injury28. erefore, increasing intestinal barrier function seems to be a good means
to prevent or treat intestinal injury. As intestinal tight junction proteins, ZO-1, occludin, and claudin-1 have
an important role in maintaining intestinal barrier permeability and constitute intestinal mucosal barrier with
intestinal epithelial cells29,30. According to previous studies, B. coagulans SCC-19 improves the intestinal barrier
Figure5. Eects of B. coagulans MZY531 on ZO-1 intestinal barrier pathway in ileum of immunosuppressed
mice. e protein expression of Claudin-1, Occludin and ZO-1 in the ileal were detected by western blot (A).
e ratios of ZO-1/β-actin (B), Occludin/β-actin (C) and Claudin-1/β-actin (D) protein bands for each region
were quantied using densitometry and presented in the graph. All data were statistically analyzed using a one-
way analysis of variance and Tukey multiple comparison. #P < 0.05 and ##P < 0.01 vs. control group; *P < 0.05 and
**P < 0.01 vs. CYP group.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
8
Vol:.(1234567890)
Scientic Reports | (2023) 13:11181 | https://doi.org/10.1038/s41598-023-38379-0
www.nature.com/scientificreports/
of carp induced by heavy metal cadmium (Cd) by up-regulating the mRNA expression of ZO-1, occluding,
and claudin-131. In addition, Zhou etal. conrmed that NCU116 extracellular polysaccharides of Lactobacillus
plantarum could increase the expression of the ZO-1 tight junction protein pathway by promoting the binding
of STAT3 to occludin and ZO-1 promoters, thus repairing the intestinal mechanical barrier function induced by
dextransodiumsulfate (DSS)32. Importantly, our study also found that MZY531 activates the expression of ZO-1,
occluding, and claudin-1 proteins, thus restoring intestinal mucosal barrier function. erefore, we speculate that
MZY531 may improve intestinal permeability by activating the expression of intestinal tight junction protein,
thus resisting intestinal mucosal injury induced by CYP.
In recent years, increasing evidence has shown that intestinal injury can also induce intestinal leakage (leaky
gut), causing bacteria and their metabolites to translocate to the blood, releasing inammatory factors, includ-
ing LPS and TNF-α33. In addition, the accumulation of pro-inammatory mediators can break the balance of
anti-inammatory and pro-inammatory factors and further aggravate the inammatory cascade and intestinal
injury34. As a specic anti-inammatory factor, IL-10 has an important role in enhancing the anti-inammatory
ability of the body35. It has been reported that probiotics can reduce advocated inammatory expression by acti-
vating IL-10-mediated immune pathways36. Interestingly, similar results were obtained in this study. erefore,
enhancing the expression of the anti-inammatory factor IL-10 may be one of the keys to controlling intestinal
Figure6. Eects of B. coagulans MZY531 on TLR4/MyD88 inammatory pathway in ileum of
immunosuppressed mice. e protein expression of IKBα, NF-κB, MyD88 and TLR4 in the ileal were detected
by western blot (A). e ratios of TLR4/β-actin (B), MyD88/β-actin (C), NF-κB/β-actin (D) and IKBα/β-actin
(E) protein bands for each region were quantied using densitometry and presented in the graph. All data
were statistically analyzed using a one-way analysis of variance and Tukey multiple comparison. #P < 0.05 and
##P < 0.01 vs. control group; *P < 0.05 and **P < 0.01 vs. CYP group.
Table 1. Eects of MZY531 on α-diversity of gut microbiota in mice with intestinal injury induced by CYP.
All data were statistically analyzed using a one-way analysis of variance and Tukey multiple comparison.
# P < 0.05 and ##P < 0.01 vs. control group; *P < 0.05 and **P < 0.01 vs. CYP group.
Groups Chao1 Shannon Simpson Pielou_e
Control 516.75 ± 112.82** 5.57 ± 0.66** 0.9326 ± 0.027** 0.6286 ± 0.05*
CYP 341.79 ± 82.56## 4.33 ± 0.52## 0.8397 ± 0.04## 0.5254 ± 0.05#
CYP + LH 548.66 ± 39.71** 5.83 ± 0.49** 0.9488 ± 0.024** 0.651 ± 0.05**
CYP + MZY531 615.62 ± 72.18** 6.00 ± 0.41** 0.9516 ± 0.02** 0.658 ± 0.04**
Content courtesy of Springer Nature, terms of use apply. Rights reserved
9
Vol.:(0123456789)
Scientic Reports | (2023) 13:11181 | https://doi.org/10.1038/s41598-023-38379-0
www.nature.com/scientificreports/
inammation. Nevertheless, some studies have reported that the expression of IL-10 is aected by the TLR4
pathway. As a type I transmembrane protein expressed on the cell membrane, TLR4 has an important role in
regulating the balance of inammation37. e NF-κB pathway is a downstream signal transduction pathway
dependent on TLR4/MyD88 pathway. When the body is in normal homeostasis, NF-κB will bind to I-κB and
remain static38. However, external stimulation can activate the expression of the TLR4/MyD88 pathway and
further induce the activation of the I-κB complex, thus regulating the expression of target genes, including TNF-
α, IL-1β, IL-6, and IL-1039. It is reported that B. coagulans TL3 inhibits intestinal inammation induced by LPS
through TLR4/MyD88, which suggests that the TLR4/MyD88 signal pathway may be the signal transduction
mechanism of B. coagulans inhibiting intestinal inammation expression13. In addition, some scholars have
reported that the combined treatment of several probiotics, including B. coagulans, suppresses DSS-induced
colitis by up-regulating the level of IL-1040. Interestingly, it has been reported that E5564, an antagonist of TLR4,
can competitively bind to TLR4-MD2, further inhibit the activation of downstream NF-κB and promote the
release of anti-inammatory factor IL-1041. In this study, we found that MZY531 down-regulates the expression
of the TLR4/MyD88 pathway and increases the level of IL-10. is further suggests that B. coagulans MZY531
may be an inhibitor of TLR4, i.e., it increases the level of IL-10 in the intestine of mice by inhibiting the TLR4/
MyD88 pathway, thus improving the anti-inammatory ability of mice and nally improving the intestinal
inammatory injury induced by CYP.
B. coagulans can regulate the disorder of intestinal microora, which has a benecial eect on the host42. Stud-
ies have shown increased abundance and diversity of probiotics in feces collected from elderly taking B. coagulans
GBI-30 and 6086 for 28 days43. Our study also obtained consistent results; we found that the intervention of B.
coagulans MZY531 increases the total number of bacteria in the feces of mice with intestinal injury induced by
CYP, which also indicates that B. coagulans MZY531 restores the abundance of intestinal ora. In addition, we
also found that B. coagulans MZY531 can increase the abundance of probiotics (Bidobacterium, Prevotella and
Firmicutes) and reduce the number of bacteria causing inammation (Bacteroides and Shigella). Of note, it has
also been reported that taking B. coagulans 13002 increases intestinal damage induced by cyclophosphamide by
increasing the abundance of probiotics12. In addition, Xie etal. reported that taking L. plantarum NCU116 may
increase the number of Bidobacteria in the feces of mice, and further reduce the disorder of gut microbiota,
thus improving the intestinal mucosal damage induced by CYP8. is indirectly conrms that B. coagulans
MZY531 has an important role in improving the intestinal injury induced by CYP. Interestingly, we examined
the correlation between intestinal ora and intestinal immune function and anti-inammatory ability by Spear-
man analysis, nding that Bidobacterium and Prevotella were positively correlated with immune proteins and
anti-inammatory factors in the intestinal tract; similar conclusions were reached by Li etal.44. is further
Figure7. Eects of B. coagulans MZY531 on the changes of gut microbiota in immunosuppressed mice.
Venn diagram (A); PCoA analysis (B). e species compositions analysis at phylum level (C). e heat map at
genus level (D). e correlation analysis between gut microbiota and intestinal immune proteins and anti-
inammatory factors at genus level by Spearman analysis (E). #P < 0.05 and ##P < 0.01 vs. control group; *P < 0.05
and **P < 0.01 vs. CYP group.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
10
Vol:.(1234567890)
Scientic Reports | (2023) 13:11181 | https://doi.org/10.1038/s41598-023-38379-0
www.nature.com/scientificreports/
suggests that the intervention of B. coagulans MZY531 can improve the immune and anti-inammatory ability
of the intestine by increasing the abundance of probiotics in the intestine (Fig.8), thus reducing the intestinal
injury caused by intestinal inammation and immunosuppression caused by CYP.
Conclusion
To sum up, B. coagulans MZY531 treatment improves intestinal barrier function and inammatory expression
in CYP-induced immunosuppressive mice, and its possible mechanism is related to the ZO-1 intestinal barrier
pathway and TLR4/MyD88 inammatory pathway. In addition, B. coagulans MZY531 also improves the disorder
of intestinal microora by increasing the abundance of probiotics in the intestine and further improving the
immune function and anti-inammatory ability of mice. erefore, this study provides a new research idea for
treating intestinal injury in CYP-induced immunosuppressive mice and a solid theoretical basis for the develop-
ment and utilization of B. coagulans.
Data availability
e data presented in this study are available on request from the corresponding author.
Received: 10 March 2023; Accepted: 7 July 2023
References
1. McGuckin, M. A., Eri, R., Simms, L. A., Florin, T. H. & Radford-Smith, G. Intestinal barrier dysfunction in inammatory bowel
diseases. Inamm. Bowel Dis. 15, 100–113. https:// doi. org/ 10. 1002/ ibd. 20539 (2009).
2. Cardoso-Silva, D. et al. Intestinal barrier function in gluten-related disorders. Nutrients 11, 2325. https:// doi. org/ 10. 3390/ nu111
02325 (2019).
3. Odenwald, M. A. & Turner, J. R. e intestinal epithelial barrier: A therapeutic target? Nat. Rev. Gastroenterol. Hepatol. 14, 9–21.
https:// doi. org/ 10. 1038/ nrgas tro. 2016. 169 (2017).
4. Teshima, C. W., Dieleman, L. A. & Meddings, J. B. Abnormal intestinal permeability in Crohn’s disease pathogenesis. Ann. N. Y.
Acad. Sci. 1258, 159–165. https:// doi. org/ 10. 1111/j. 1749- 6632. 2012. 06612.x (2012).
5. Tilg, H., Zmora, N., Adolph, T. E. & Elinav, E. e intestinal microbiota fuelling metabolic inammation. Nat. Rev. Immunol. 20,
40–54. https:// doi. org/ 10. 1038/ s41577- 019- 0198-4 (2020).
Figure8. Schematic illustration showing the mechanisms of B. coagulans MZY531 alleviates intestinal mucosal
injury in immunosuppressive mice.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
11
Vol.:(0123456789)
Scientic Reports | (2023) 13:11181 | https://doi.org/10.1038/s41598-023-38379-0
www.nature.com/scientificreports/
6. Allam-Ndoul, B., Castonguay-Paradis, S. & Veilleux, A. Gut microbiota and intestinal trans-epithelial permeability. Int. J. Mol. Sci.
21, 6402. https:// doi. org/ 10. 3390/ ijms2 11764 02 (2020).
7. Viaud, S. et al. e intestinal microbiota modulates the anticancer immune eects of cyclophosphamide. Science 342, 971–976.
https:// doi. org/ 10. 1126/ scien ce. 12405 37 (2013).
8. Xie, J. H. et al. Lactobacillus plantarum NCU116 attenuates cyclophosphamide-induced intestinal mucosal injury, metabolism and
intestinal microbiota disorders in mice. Food Funct. 7, 1584–1592. https:// doi. org/ 10. 1039/ c5fo0 1516b (2016).
9. Ohland, C. L. & Macnaughton, W. K. Probiotic bacteria and intestinal epithelial barrier function. Am. J. Physiol. Gastrointest. Liver
Physiol. 298, G807–G819. https:// doi. org/ 10. 1152/ ajpgi. 00243. 2009 (2010).
10. Shinde, T. et al. Probiotic Bacillus coagulans MTCC 5856 spores exhibit excellent in-vitro functional ecacy in simulated gastric
survival, mucosal adhesion and immunomodulation. J. Funct. Foods 52, 100–108. https:// doi. org/ 10. 1016/j. j. 2018. 10. 031 (2019).
11. Liu, J. J., Reid, G., Jiang, Y., Turner, M. S. & Tsai, C. C. Activity of HIV entry and fusion inhibitors expressed by the human vaginal
colonizing probiotic Lactobacillus reuteri RC-14. Cell. Microbiol. 9, 120–130. https:// doi. org/ 10. 1111/j. 1462- 5822. 2006. 00772.x
(2007).
12. Zhao, S. et al. Bacillus coagulans 13002 and fructo-oligosaccharides improve the immunity of mice with immunosuppression
induced by cyclophosphamide through modulating intestinal-derived and fecal microbiota. Food Res. Int. 140, 109793. https://
doi. org/ 10. 1016/j. foodr es. 2020. 109793 (2021).
13. Wang, Y. et al. Bacillus coagulans TL3 inhibits LPS-induced caecum damage in rat by regulating the TLR4/MyD88/NF-κB and
Nrf2 signal pathways and modulating intestinal microora. Oxid. Med. Cell. Longev. 2022, 5463290. https:// doi. org/ 10. 1155/ 2022/
54632 90 (2022).
14. Ou, Z. et al. Betulinic acid attenuates cyclophosphamide-induced intestinal mucosa injury by inhibiting the NF-κB/MAPK signal-
ling pathways and activating the Nrf2 signalling pathway. Ecotoxicol. Environ. Saf. 225, 112746. https:// doi. org/ 10. 1016/j. ecoenv.
2021. 112746 (2021).
15. Guo, M. Z., Meng, M., Feng, C. C., Wang, X. & Wang, C. L. A novel polysaccharide obtained from Craterellus cornucopioides
enhances immunomodulatory activity in immunosuppressive mice models via regulation of the TLR4-NF-κB pathway. Food
Funct. 10, 4792–4801. https:// doi. org/ 10. 1039/ c9fo0 0201d (2019).
16. Salva, S., Marranzino, G., Villena, J., Agüero, G. & Alvarez, S. Probiotic Lactobacillus strains protect against myelosuppression and
immunosuppression in cyclophosphamide-treated mice. Int. Immunopharmacol. 22, 209–221. https:// doi. org/ 10. 1016/j. intimp.
2014. 06. 017 (2014).
17. Wang, L. et al. Lactobacillus plantarum DP189 reduces α-SYN aggravation in MPTP-induced Parkinson’s disease mice via regulat-
ing oxidative damage, inammation, and gut microbiota disorder. J. Agric. Food Chem. 70, 1163–1173. https:// doi. org/ 10. 1021/
acs. jafc. 1c077 11 (2022).
18. Yu, Q. et al. Molecular mechanism underlying chemoprotective eects of Ganoderma atrum polysaccharide in cyclophosphamide-
induced immunosuppressed mice. J. Funct. Foods 15, 52–60. https:// doi. org/ 10. 1016/j. j. 2015. 03. 015 (2015).
19. Lahtinen, S. J. et al. Probiotics modulate the Bidobacterium microbiota of elderly nursing home residents. Age 31, 59–66. https://
doi. org/ 10. 1007/ s11357- 008- 9081-0 (2009).
20. Wu, Y., Zhu, C. P., Zhang, Y., Li, Y. & Sun, J. R. Immunomodulatory and antioxidant eects of pomegranate peel polysaccharides
on immunosuppressed mice. Int. J. Biol. Macromol. 137, 504–511. https:// doi. org/ 10. 1016/j. ijbio mac. 2019. 06. 139 (2019).
21. Awad, W. A., Ghareeb, K., Abdel-Raheem, S. & Böhm, J. Eects of dietary inclusion of probiotic and synbiotic on growth per-
formance, organ weights, and intestinal histomorphology of broiler chickens. Poult. Sci. 88, 49–56. https:// doi. org/ 10. 3382/ ps.
2008- 00244 (2009).
22. Emami, N. K. & Dalloul, R. A. Centennial review: Recent developments in host-pathogen interactions during necrotic enteritis
in poultry. Poult. Sci. 100, 101330. https:// doi. org/ 10. 1016/j. psj. 2021. 101330 (2021).
23. Liu, Y. et al. Ameliorative eects of Antrodia cinnamomea polysaccharides against cyclophosphamide-induced immunosuppression
related to Nrf2/HO-1 signaling in BALB/c mice. Int. J. Biol. Macromol. 116, 8–15. https:// doi. org/ 10. 1016/j. ijbio mac. 2018. 04. 178
(2018).
24. Lis, M. & Obmińska-Mrukowicz, B. Modulatory eects of bestatin on T and B lymphocyte subsets and the concentration of
cytokines released by 1/2 lymphocytes in cyclophosphamide-treated mice. Central Eur. J. Immunol. 38, 42–53. https:// doi.
org/ 10. 5114/ ceji. 2013. 34357 (2013).
25. Bomko, T. V., Nosalskaya, T. N., Kabluchko, T. V., Lisnyak, Y. V. & Martynov, A. V. Immunotropic aspect of the Bacillus coagulans
probiotic action. J. Pharm. Pharmacol. 69, 1033–1040. https:// doi. org/ 10. 1111/ jphp. 12726 (2017).
26. Wu, Y. et al. e role of autophagy in maintaining intestinal mucosal barrier. J. Cell. Physiol. 234, 19406–19419. https:// doi. org/
10. 1002/ jcp. 28722 (2019).
27. Ying, M. et al. Cultured Cordyceps sinensis polysaccharides attenuate cyclophosphamide-induced intestinal barrier injur y in mice.
J. Funct. Foods 62, 103523. https:// doi. org/ 10. 1016/j. j. 2019. 103523 (2019).
28. Park, C. M. & Song, Y. S. Luteolin and luteolin-7-O-glucoside inhibit lipopolysaccharide-induced inammatory responses through
modulation of NF-κB/AP-1/PI3K-Akt signaling cascades in RAW 2647 cells. Nutr. Res. Pract. 7, 423–429. https:// doi. org/ 10. 4162/
nrp. 2013.7. 6. 423 (2013).
29. Krug, S. M., Schulzke, J. D. & Fromm, M. Tight junction, selective permeability, and related diseases. Semin. Cell Dev. Biol. 36,
166–176. https:// doi. org/ 10. 1016/j. semcdb. 2014. 09. 002 (2014).
30. Zhuang, Y. et al. Resveratrol attenuates oxidative stress-induced intestinal barrier injury through PI3K/Akt-mediated Nrf2 signal-
ing pathway. Oxid. Med. Cell. Longev. 2019, 7591840. https:// doi. org/ 10. 1155/ 2019/ 75918 40 (2019).
31. Chang, X. et al. Bacillus coagulans SCC-19 maintains intestinal health in cadmium-exposed common carp (Cyprinus carpio L.)
by strengthening the gut barriers, relieving oxidative stress and modulating the intestinal microora. Ecotoxicol. Environ. Saf. 228,
112977. https:// doi. org/ 10. 1016/j. ecoenv. 2021. 112977 (2021).
32. Zhou, X. et al. Exopolysaccharides from Lactobacillus plantarum NCU116 regulate intestinal barrier function via STAT3 signaling
pathway. J. Agric. Food Chem. 66, 9719–9727. https:// doi. org/ 10. 1021/ acs. jafc. 8b033 40 (2018).
33. Liu, B. et al. Raw bowl tea (Tuocha) polyphenol prevention of nonalcoholic fatty liver disease by regulating intestinal function in
mice. Biomolecules 9, 435. https:// doi. org/ 10. 3390/ biom9 090435 (2019).
34. Xavier, R. J. & Podolsky, D. K. Unravelling the pathogenesis of inammatory bowel disease. Nature 448, 427–434. https:// doi. org/
10. 1038/ natur e06005 (2007).
35. Neurath, M. F. Cytokines in inammatory bowel disease. Nat. Rev. Immunol. 14, 329–342. https:// doi. org/ 10. 1038/ nri36 61 (2014).
36. Sun, M. et al. Microbiota-derived short-chain fatty acids promote 1 cell IL-10 production to maintain intestinal homeostasis.
Nat. Commun. 9, 3555. https:// doi. org/ 10. 1038/ s41467- 018- 05901-2 (2018).
37. Xu, H. et al. Invitro immune toxicity of ochratoxin A in porcine alveolar macrophages: A role for the ROS-relative TLR4/MyD88
signaling pathway. Chem. Biol. Interact. 272, 107–116. https:// doi. org/ 10. 1016/j. cbi. 2017. 05. 016 (2017).
38. Parlato, M. & Yeretssian, G. NOD-like receptors in intestinal homeostasis and epithelial tissue repair. Int. J. Mol. Sci. 15, 9594–9627.
https:// doi. org/ 10. 3390/ ijms1 50695 94 (2014).
39. Sun, L. et al. A novel role of OS-9 in the maintenance of intestinal barrier function from hypoxia-induced injury via p38-dependent
pathway. Int. J. Biol. Sci. 11, 664–671. https:// doi. org/ 10. 7150/ ijbs. 10783 (2015).
40. Wang, Y. et al. Combination of probiotics with dierent functions alleviate DSS-induced colitis by regulating intestinal microbiota,
IL-10, and barrier function. Appl. Microbiol. Biotechnol. 104, 335–349. https:// doi. org/ 10. 1007/ s00253- 019- 10259-6 (2020).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
12
Vol:.(1234567890)
Scientic Reports | (2023) 13:11181 | https://doi.org/10.1038/s41598-023-38379-0
www.nature.com/scientificreports/
41. Mullarkey, M. et al. Inhibition of endotoxin response by e5564, a novel Toll-like receptor 4-directed endotoxin antagonist. J.
Pharmacol. Exp. er. 304, 1093–1102. https:// doi. org/ 10. 1124/ jpet. 102. 044487 (2003).
42. Bindels, L. B., Delzenne, N. M., Cani, P. D. & Walter, J. Towards a more comprehensive concept for prebiotics. Nat. Rev. Gastroen-
terol. Hepatol. 12, 303–310. https:// doi. org/ 10. 1038/ nrgas tro. 2015. 47 (2015).
43. Nyangale, E. P., Farmer, S., Keller, D., Cherno, D. & Gibson, G. R. Eect of prebiotics on the fecal microbiota of elderly volunteers
aer dietary supplementation of Bacillus coagulans GBI-30, 6086. Anaerobe 30, 75–81. https:// doi. org/ 10. 1016/j. anaer obe. 2014.
09. 002 (2014).
44. Li, S., Wang, L., Liu, B. & He, N. Unsaturated alginate oligosaccharides attenuated obesity-related metabolic abnormalities by
modulating gut microbiota in high-fat-diet mice. Food Funct. 11, 4773–4784. https:// doi. org/ 10. 1039/ c9fo0 2857a (2020).
Acknowledgements
is work was sponsored by 2022 Jilin Province Science and Technology Development Plan, Natural Science
Foundation of Jilin Province (20220101310JC).
Author contributions
Methodology, Soware, Formal analysis, Investigation, Writing—original dra, Writing—review & editing,
Visualization, Z.Z.; Methodology, Supervision, Writing—review & editing, M.S.; Project administration, Data
curation, Writing-review & editing, X.C.; Writing—review & editing, J.C.; Conceptualization, Methodology,
C.L.; Project administration, Resources, Formal analysis, Investigation, Writing—original dra, Writing—review
& editing, Funding acquisition, Supervision, X.Z. All authors have read and agreed to the published version of
the manuscript.
Competing interests
e authors declare no competing interests.
Additional information
Supplementary Information e online version contains supplementary material available at https:// doi. org/
10. 1038/ s41598- 023- 38379-0.
Correspondence and requests for materials should be addressed to X.Z.
Reprints and permissions information is available at www.nature.com/reprints.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access is article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons licence, and indicate if changes were made. e images or other third party material in this
article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the
material. If material is not included in the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/.
© e Author(s) 2023
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Tight junctions are vital components of the intestinal mucosal barrier against harmful pathogens, predominantly made up of membrane protein complexes, including zonula occludens proteins (ZOs), occludin, and claudins (109). Alterations in the structure and function of tight junctions may directly impair the integrity of the intestinal mucosal barrier, leading to the infiltration of pathogens and other undesirable substances through the epithelial layer (110). Previous studies have confirmed the regulatory role of Bacillus spp. on intestinal tight junction proteins in piglets (35,75,105,(111)(112)(113). ...
Article
Full-text available
The application of Bacillus spp. as probiotics in the swine industry, particularly for piglet production, has garnered significant attention in recent years. This review aimed to summarized the role and mechanisms of Bacillus spp. in promoting growth and maintaining gut health in piglets. Bacillus spp. can enhance intestinal barrier function by promoting the proliferation and repair of intestinal epithelial cells and increasing mucosal barrier integrity, thereby reducing the risk of pathogenic microbial invasion. Additionally, Bacillus spp. can activate the intestinal immune system of piglets, thereby enhancing the body’s resistance to diseases. Moreover, Bacillus spp. can optimize the gut microbial community structure, enhance the activity of beneficial bacteria such as Lactobacillus, and inhibit the growth of harmful bacteria such as Escherichia coli, ultimately promoting piglet growth performance and improving feed efficiency. Bacillus spp. has advantages as well as challenges as an animal probiotic, and safety evaluation should be conducted when using the newly isolated Bacillus spp. This review provides a scientific basis for the application of Bacillus spp. in modern piglet production, highlighting their potential in improving the efficiency of livestock production and animal welfare.
... These findings align with previous reports of W. coagulans strains colonizing the intestinal mucosa, whereby they enhance mucosal barrier function, modulate the mucosal immune system, reduce inflammatory factor production, alleviate intestinal inflammation and inflammation-related diseases, promote secretory IgA production, and facilitate antigen-specific immune responses. 41 There were four limitations to this study. Firstly, only one gender of mice was used in the infectious model of H. pylori. ...
Article
Full-text available
The widespread prevalence of Helicobacter pylori infection, particularly in China, contributes to the development of gastrointestinal diseases. Antibiotics have limitations, including adverse reactions and increased antibiotic resistance. Therefore, identification of novel gastrogenic probiotics capable of surviving the acidic gastric environment and effectively combating H. pylori infection has potential in restoring gastric microbiota homeostasis. Five novel strains of human gastrogenic Weizmannia coagulans (BCF-01–05) were isolated from healthy gastric mucosa and characterized using 16S rDNA identification. Acid resistance, H. pylori inhibition, and adherence to gastric epithelial cells were evaluated in in-vitro experiments and the molecular mechanism explored in in-vivo experiments. Among the gastric-derived W. coagulans strains, BCF-01 exhibited the strongest adhesion and H. pylori inhibition, warranting further in-vivo safety evaluation. Through 16S rRNA sequencing of a mouse model, BCF-01 was determined to significantly restore H. pylori-associated gastric dysbiosis and increase the abundance of potential probiotic bacteria. Furthermore, BCF-01 enhanced mucosal tight junction protein expression and inhibited the TLR4-NFκB-pyroptosis signaling pathway in macrophages, as demonstrated by qRT-PCR and western blotting. These findings highlight the potential of BCF-01 in the prevention and control of H. pylori infection. Specifically, treatment with BCF-01 effectively restored gastric microecology and improved H. pylori-mediated mucosal barrier destruction while reducing inflammation through inhibition of the TLR4-NFκB-pyroptosis signaling pathway in macrophages. BCF-01 is a promising alternative to traditional triple therapy for H. pylori infections, offering minimal side effects with high suitability for high-risk individuals.
Article
Full-text available
While there is evidence supporting the immune-stimulating properties of Weizmannia coagulans, the specific mechanism by which its supernatant may enhance immunity and its relationship with the gut microbiota remains unclear....
Article
Full-text available
Probiotics are defined as living or dead bacteria and their byproducts that maintain the balance of the intestinal microbiome. They are non-toxic, non-pathogenic, and do not release any toxins either within or outside the body. Adequate consumption of probiotics can enhance metabolite production, increase immunity, maintain a balanced intestinal flora, and stimulate growth. Probiotics do not have negative antibiotic effects and help maintain the natural flora in animals in a balanced state or prevent dysbacteriosis. Heyndrickxia coagulans (H. coagulans) is a novel probiotic species that is gradually being used for the improvement of human health. Compared to commonly used probiotic lactic acid bacteria, H. coagulans can produce spores, which provide the species with high resistance to adverse conditions. Even though they are transient residents of the gut, beneficial bacteria can have a significant impact on the microbiota because they can outnumber harmful germs, and vice versa. This article discusses the probiotic mechanisms of H. coagulans and outlines the requirements for a substance to be classified as a probiotic. It also addresses how to assess strains that have recently been discovered to possess probiotic properties.
Article
Purpose of review Gastrointestinal mucositis (GIM) is a significant complication of cancer therapy. Whilst inflammation is a central feature of GIM, studies attempting to mitigate mucosal damage via this mechanism are scarce. This review describes the relation between GIM, local and systemic inflammation, and the microbiome and its metabolites, and explores recent research on therapeutics that target this relationship. Recent findings Recent literature underscores the pivotal role of inflammation in GIM, elucidating its bidirectional relation with disturbance of the gut microbiota composition and intestinal permeability. These events cause a heightened risk of bloodstream infections and lead to systemic inflammation. While studies investigating risk prediction models or therapeutics targeting GIM-related inflammation remain scarce, results have shown promise in finding biomarkers and alleviating GIM and its accompanying clinical symptoms. Summary The findings underscore the important role of inflammation and the microbiome in GIM. Understanding the inflammatory pathways driving GIM is crucial for developing effective treatments. Further research is needed using genomics, epigenomics, and microbiomics to explore better risk prediction models or therapeutic strategies aimed at mitigating GIM-related inflammation.
Article
Bovine colostrum encapsulated by liposomes improves the bioavailability of IgG and enhances the immune regulatory function in immunosuppressed mice.
Article
Full-text available
Background: Bacillus coagulans has been widely used in food and feed additives, which can effectively inhibit the growth of harmful bacteria, improve intestinal microecological environment, promote intestinal development, and enhance intestinal function, but its probiotic mechanism is not completely clear. Aim: The aim of this study is to discuss the effect and mechanism of Bacillus coagulans TL3 on oxidative stress and inflammatory injury of cecum induced by LPS. Method: The Wistar rats were randomly divided into four groups, each containing 7 animals. Two groups were fed with basic diet (the LPS and control, or CON, groups). The remaining groups were fed with basic diet and either a intragastric administration high or low dose of B. coagulans, forming the HBC and LBC groups, respectively. The rats were fed normally for two weeks. On the 15th day, those in the LPS, HBC, and LBC groups were injected intraperitoneally with LPS-the rats in the CON group were injected intraperitoneally with physiological saline. After 4 hours, all the rats were anesthetized and sacrificed by cervical dislocation, allowing samples to be collected and labeled. The inflammatory and antioxidant cytokine changes of the cecum were measured, and the pathological changes of the cecum were observed, determining the cecal antioxidant, inflammation, and changes in tight junction proteins and analysis of intestinal flora. Result: The results show that LPS induces oxidative damage in the cecal tissues of rats and the occurrence of inflammation could also be detected in the serum. The Western blot results detected changes in the NF-κB- and Nrf2-related signaling pathways and TJ-related protein levels. Compared with the LPS group, the HBC group showed significantly downregulated levels of expression of Nrf2, NQO1, HO-1, GPX, and GCLC. The expression of TLR4, MYD88, NF-κB, IL-6, TNFα, and IL-1β was also significantly downregulated, while the expression of other proteins (ZO-1, occludin, and claudin-1) increased significantly. Bacillus coagulans TL3 was also found to increase the relative abundance of the beneficial bacterium Akkermansia muciniphila in the intestines. There is also a significant reduction in the number of harmful bacteria Escherichia coli and Shigella (Enterobacteriaceae). Conclusion: Bacillus coagulans TL3 regulates the TLR4/MyD88/NF-κB and Nrf2 signaling pathways in the cecal tissue of rats, protects the intestine from inflammation and oxidative damage caused by LPS, and inhibits the reproduction of harmful bacteria and promotes beneficial effects by regulating the intestinal flora bacteria grow, thereby enhancing intestinal immunity.
Article
Full-text available
Necrotic enteritis (NE) is a significant enteric disease in commercial poultry with considerable economic effect on profitability manifested by an estimated $6 billion in annual losses to the global industry. NE presents a unique challenge, being a complex enteric disease that often leads to either clinical (acute) or subclinical (chronic) form. The latter typically results in poor performance (reduced feed intake, weight gain and eventually higher feed conversion ratio (FCR) with low mortality rates, and represents the greatest economic impact on poultry production. The use of antibiotic growth promoters (AGPs) has been an effective tool in protecting birds from enteric diseases by maintaining gut health and modifying gut microbiota, thus improving broilers’ production efficiency and improving enteric health. The removal of AGPs presented the poultry industry with several challenges, including reduced bird health and immunity as well as questioning the safety of poultry products. Consequently, research on antibiotic alternatives that can support gut health was intensified. Probiotics, prebiotics, essential oils and organic acids were among various additives that have been tested for their efficacy against NE with some being effective but not to the level of AGPs. The focus of this review is on the relationship between NE pathogenesis, microbiome, and host immune responses, along with references to recent reviews addressing production aspects of NE. With a comprehensive understanding of these dynamic changes, new and programmed strategies could be developed to make use of the current products more effectively or build a stepping stone towards the development of a new generation of supplements.
Article
Full-text available
Constant remodeling of tight junctions to regulate trans-epithelial permeability is essential in maintaining intestinal barrier functions and thus preventing diffusion of small molecules and bacteria to host systemic circulation. Gut microbiota dysbiosis and dysfunctional gut barrier have been correlated to a large number of diseases such as obesity, type 2 diabetes and inflammatory bowel disease. This led to the hypothesis that gut bacteria-epithelial cell interactions are key regulators of epithelial permeability through the modulation of tight junctions. Nevertheless, the molecular basis of host-pathogen interactions remains unclear mostly due to the inability of most in vitro models to recreate the differentiated tissue structure and components observed in the normal intestinal epithelium. Recent advances have led to the development of a novel cellular model derived from intestinal epithelial stem cells, the so-called organoids, encompassing all epithelial cell types and reproducing physiological properties of the intestinal tissue. We summarize herein knowledge on molecular aspects of intestinal barrier functions and the involvement of gut bacteria-epithelial cell interactions. This review also focuses on epithelial organoids as a promising model for epithelial barrier functions to study molecular aspects of gut microbiota-host interaction.
Article
Full-text available
Oxidative stress is implicated in a wide range of intestinal disorders and closely associated with their pathological processes. Resveratrol (RSV), a plant extract, plays a vital role in protecting various organs in vitro and in vivo . However, the benefits of RSV are controversial, and underlying mechanisms for its antioxidant effects on intestinal epithelial cells remain unclear. In this study, we evaluated the effects of RSV on oxidative stress induced by H 2 O 2 in IPEC-J2 cells. We found that pretreatment with RSV significantly increased cell viability; increased expression levels of tight junction (TJ) proteins (claudin-1, occludin, and ZO-1); improved activities of superoxide dismutase-1 (SOD-1), catalase (CAT), and glutathione peroxidase (GSH-Px); and decreased intracellular reactive oxygen species (ROS) levels and apoptosis induced by H 2 O 2 ( P<0.05 ). In addition, RSV upregulated Akt phosphorylation, Nrf2 phosphorylation, and expression levels of antioxidant genes HO-1, SOD-1, and CAT in a dose-dependent manner ( P<0.05 ) under oxidative stress. Knockdown of Nrf2 by short-hairpin RNA (shRNA) abrogated RSV-mediated protection against H 2 O 2 -induced apoptosis, RSV-induced increase of TJ protein levels, and antioxidant gene expression (SOD-1, CAT, and GSH-Px) ( P<0.05 ). Consistent with Nrf2 knockdown, the PI3K/Akt inhibitor LY294002 significantly suppressed RSV-induced Nrf2 phosphorylation and RSV-induced increase of TJ protein levels and antioxidant gene expression under H 2 O 2 treatment ( P<0.05 ). Collectively, these results demonstrate that RSV can directly protect IPEC-J2 cells against oxidative stress through the PI3K/Akt-mediated Nrf2 signaling pathway, suggesting that RSV may be an effective feed additive against intestinal damage in livestock production.
Article
Heavy metal cadmium (Cd) pollution is a serious problem affecting the sustainable development of aquaculture and the safety of aquatic foods. Research about the use of probiotics to attenuate toxic damage caused by Cd²⁺ in aquatic animals has received widespread attention. Bacillus coagulans (B. coagulans), a kind of probiotics commonly used in aquaculture, has been shown to adsorb Cd²⁺ both in vivo and vitro. Here, we aimed to determine the effects of B. coagulans on Cd²⁺ bioaccumulation, gut barrier function, oxidative stress and gut microbiota in common carp following Cd²⁺ exposure. The fish were exposure to Cd²⁺ at 0 and 0.5 mg/L and/or fed a B. coagulans-containing diet at 10⁷, 10⁸ and 10⁹ CFU/g for 8 weeks. The results indicated that B. coagulans can maintain gut barrier function in Cd²⁺-exposed fish by reducing Cd²⁺ bioaccumulation, increasing the mRNA levels of tight junction protein genes (occludin, claudin-2 and zonula occludens-1), and decreasing the levels of diamine oxidase and D-lactic acid. In addition, B. coagulans could relieve oxidative stress in Cd²⁺-exposed fish by restoring the activities of glutathione peroxidase, catalase and superoxide dismutase. Moreover, Cd²⁺ exposure decreased the intestinal microbiota diversity and changed the intestinal microbiota compositions in common carp. However, supplementation with B. coagulans could reverse the altered intestinal microbiota diversity and composition after Cd²⁺ exposure, decrease the abundance of some pathogens (Shewanella and Vibrio), and increase the abundance of probiotics (Bacillus and Lactobacillus). These results indicate that B. coagulans may serve as a potential antidote for alleviating Cd²⁺ toxicity.
Article
Betulinic acid (BA), a pentacyclic triterpenoid, has been associated with several biological effects, such as antioxidant, anti-inflammatory and antiviral activities. Previous studies have demonstrated that BA has the ability to alleviate intestinal mucosal damage, however, the potential mechanism associated with the effect has not been reported. This study aimed to investigate the possible protective mechanism of BA against cyclophosphamide (CYP)-induced intestinal mucosal damage. Here, we found that BA pretreatment prevented intestinal mucosal barrier dysfuction from CYP-challenged mice by repairing the intestinal physical, chemical, and immune barriers. Moreover, BA treatment suppressed the CYP-induced oxidative stress by activating the nuclear factor erythroid 2 [NF-E2]-related factor (Nrf2) pathway blocked reactive oxygen species (ROS) accumulation. In addition, BA inhibited CYP-triggered intestinal inflammation through down-regulating the nuclear transcription factor kappa B (NF-κB)/mitogen-activating protein kinase (MAPK) pathways. Furthermore, BA pretreatment reduced intestinal apoptosis by blocking ROS-activated mitochondrial apoptotic pathway. Overall, the current study demonstrated the protective effect of BA against CYP-caused intestinal mucosal damage by regulating the Nrf2 and NF-κB/MAPK signalling pathways, which may provide new therapeutic targets to attenuate intestinal impairment and maintain intestinal health.
Article
This study aims to evaluate the effects of probiotic Bacillus coagulans 13,002 (BCS) and prebiotic fructo-oligosaccharides (FOS) on mice treated with the alkylating agent cyclophosphamide (CTX). We found that both BCS and FOS, especially BCS, significantly alleviated CTX-induced injury by modulating intestinal-derived and fecal microbiota. BCS and BCS + FOS increased serum immunoglobulin levels, which were reduced by CTX. In addition, BCS and BCS + FOS upregulated IFN-γ and IL-4, which protect mucosal barriers and the balance of Th1/Th2. BCS promoted the growth of some beneficial bacteria, such as Bacteroides, Coprococcus, Enterococcus, Oscillospira, and Ruminococcus in mouse gut. In addition, BCS + FOS inhibited the growth of several harmful bacteria, including Acinetobacter, Arthrobacter, Brachybacterium, Corynebacterium, Jeotgalicoccus, Sporosarcina, and Staphylococcus. Furthermore, BCS potentially improved the growth of Anaerotruncus bacteria, which can promote the production of butyrate acids. In summary, according our results suggest that BCS and FOS improved the immunity of mice with immunosuppression induced by CTX through modulating intestinal-derived and fecal microbiota.
Article
Gut microbiota plays an important role in the high-fat diet (HFD)-induced obesity and related metabolic syndrome (MetS). Our previous study has demonstrated that unsaturated alginate oligosaccharides (UAOS) degraded by alginate lyase possess significant anti-obesity effects in HFD-fed mice. Herein, we further established that UAOS could significantly ameliorate obesity-related metabolic abnormalities, including hyperlipidemia, insulin resistance and low-grade inflammation. Particularly, the beneficial effect of UAOS on these metabolic abnormalities could be significantly reversed by antibiotic supplementation. Subsequently, the microbiological analysis has revealed that UAOS treatment can modulate the overall composition of the gut microbiota, which is highly associated with metabolic parameters. UAOS supplementation can partially reverse the gut dysbiosis induced by HFD-diet or antibiotics. Specifically, UAOS treatment selectively increased the relative abundance of beneficial intestinal bacteria (e.g. Lactobacillus and Akkermansia genus) and decreased the abundance of inflammogenic bacteria (e.g. Bacteroides and Parabacteroides). These results suggest that UAOS can attenuate the HFD-induced obesity and related abnormalities through modulating gut microbiota, indicating that UAOS can act as potent prebiotic agents in treating obesity and related metabolic diseases.