You are on page 1of 15

RESEARCH ARTICLE

www.advancedscience.com

Bile Acid–Microbiome Interaction Promotes Gastric


Carcinogenesis
Shouli Wang, Junliang Kuang, Hongwei Zhang, Wenlian Chen, Xiaojiao Zheng,
Jieyi Wang, Fengjie Huang, Kun Ge, Mengci Li, Mingliang Zhao, Cynthia Rajani,
Jinshui Zhu, Aihua Zhao,* and Wei Jia*

1. Introduction
Bile reflux gastritis (BRG) is associated with the development of gastric cancer
(GC), but the specific mechanism remains elusive. Here, a comprehensive Gastric cancer (GC) is one of the most
common and lethal gastrointestinal ma-
study is conducted to explore the roles of refluxed bile acids (BAs) and
lignancies worldwide.[1] The development
microbiome in gastric carcinogenesis. The results show that conjugated BAs, of GC, especially the intestinal type, usu-
interleukin 6 (IL-6), lipopolysaccharide (LPS), and the relative abundance of ally occurs through chronic gastric inflam-
LPS-producing bacteria are increased significantly in the gastric juice of both mation, atrophic gastritis, and intestinal
BRG and GC patients. A secondary BA, taurodeoxycholic acid (TDCA), is metaplasia.[2] Bile acids (BAs) have been
shown to be associated with intestinal meta-
significantly and positively correlated with the LPS-producing bacteria in the
plasia in the cardia[3,4] and bile reflux gas-
gastric juice of these patients. TDCA promotes the proliferation of normal tritis (BRG) has been implicated in the
gastric epithelial cells (GES-1) through activation of the IL-6/JAK1/STAT3 genesis of gastritis and gastric cancer in
pathway. These results are further verified in two mouse models, one by humans and animals.[5–7] A recent clini-
gavage of TDCA, LPS, and LPS-producing bacteria (Prevotella cal cross-sectional study showed that bile
melaninogenica), respectively, and the other by bile reflux (BR) surgery, reflux (BR) is an independent risk factor
for the development of precancerous gas-
mimicking clinical bile refluxing. Moreover, the bile reflux induced gastric
tric lesions and GC.[8] In addition, in the
precancerous lesions observed in the post BR surgery mice can be prevented remnant stomach of rats after gastrectomy,
by treatment with cryptotanshinone, a plant-derived STAT3 inhibitor. These BAs, the main component of the duodenal
results reveal an important underlying mechanism by which bile reflux juice, have been implicated in gastric can-
promotes gastric carcinogenesis and provide an alternative strategy for the cer via duodenogastric reflux.[9] In humans,
duodenogastric reflux has also been impli-
prevention of GC associated with BRG.
cated in gastric stump carcinoma.[10–12] Re-
cently, it was reported that acidified BAs

S. Wang, J. Kuang, X. Zheng, J. Wang, F. Huang, K. Ge, M. Li, M. Zhao, W. Chen


A. Zhao, W. Jia Cancer Institute, Longhua Hospital
Center for Translational Medicine and Shanghai Key Laboratory of Shanghai University of Traditional Chinese Medicine
Diabetes Mellitus Shanghai 200233, China
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital C. Rajani, W. Jia
Shanghai 200233, China Cancer Biology Program
E-mail: zhah@sjtu.edu.cn; weijia1@hkbu.edu.hk University of Hawaii Cancer Center
H. Zhang Honolulu, HI 96813, USA
Department of Metabolic and Bariatric Surgery J. Zhu
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital Department of Gastroenterology
Shanghai 200233, China Shanghai Jiao Tong University Affiliated Sixth People’s Hospital
Shanghai 200233, China
W. Jia
School of Chinese Medicine
The ORCID identification number(s) for the author(s) of this article Hong Kong Baptist University
can be found under https://doi.org/10.1002/advs.202200263 Kowloon Tong, Hong Kong 999077, China
© 2022 The Authors. Advanced Science published by Wiley-VCH GmbH.
This is an open access article under the terms of the Creative Commons
Attribution License, which permits use, distribution and reproduction in
any medium, provided the original work is properly cited.
DOI: 10.1002/advs.202200263

Adv. Sci. 2022, 9, 2200263 2200263 (1 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

enhanced tumor progression and telomerase activity of gastric lower (FC = 1.46 for BRG/control, FC = 1.61 for GC/control) than
cancer in vivo[13] and that deoxycholic acid induced intestinal those of conjugated BAs (FC = 39.10 for BRG/control, FC = 6.62
metaplasia in gastric epithelial cells.[14,15] for GC/control).
Reflux of the BAs and intestinal bacteria into the stomach was The increased BAs led us to consider whether the inherent
suggested to induce changes in the microbial composition.[16,17] acid-base balance in the stomach was disturbed. We then mea-
According to the recent reports, eradication of Helicobacter py- sured the pH value of gastric juice and the results showed that
lori (H. pylori) cannot completely reverse intestinal metaplasia pH increased drastically in patients with BRG (3.46 ± 0.28) and
progression,[18,19] although H. pylori is considered an important GC (3.93 ± 0.33) compared with control (1.98 ± 0.09) (Figure 1E).
etiological factor in both the precursor event and subsequent GC Compared with the strong acid in the stomach, the bile acids
development.[20,21] Hence, there are predisposing factors other refluxing into the stomach from the duodenum are weak acids,
than H. pylori infection that may also play important roles in which can break the original acid–base balance and increase the
GC development and progression. Many studies have found pH value in the stomach. We further identified a strong and
that there is a close relationship between BAs and microbiota positive correlation between 7 out of 8 conjugated BAs and the
in gastrointestinal inflammation and carcinogenesis.[22,23] How- pH value of human gastric juice, in which TDCA showed the
ever, there are few studies focused on gastric microbiota induced strongest positive correlation with pH value (Figure 1F). Mean-
by bile refluxing and their influences on the gastric mucosa in- while, the levels of TDCA were significantly increased in the gas-
jury. tric juice of BRG and GC groups (Figure 1G).
In the present study, we found that elevated conjugated BAs Together, these results suggest that there were significantly el-
(the BAs are conjugated with glycine or taurine) were positively evated levels of conjugated BAs in the gastric juice of BRG and
associated with lipopolysaccharide (LPS)-producing bacteria in GC patients, leading to the increase of gastric pH value and prob-
patients with BRG and GC. Through in vitro and in vivo as- ably inducing precancerous lesions.
says, we further investigated the roles of taurodeoxycholic acid
(TDCA), LPS, and LPS-producing bacteria in promoting gastric
inflammation and precancerous lesions. Our findings suggest 2.2. Increased Conjugated BAs Were Associated with the
that bile reflux can promote gastric carcinogenesis through the Elevated Abundance of LPS-Producing Bacteria
activation of the IL-6/JAK1/STAT3 pro-inflammatory signaling
pathway and that STAT3 inhibition can alleviate this carcinogenic According to the reports, the BAs refluxing into the stomach
effect, thus providing an alternative strategy for the prevention of were closely related to chronic inflammation.[24,25] So, we mea-
gastric cancer associated with bile reflux. sured pro-inflammatory cytokines in gastric juice and the results
showed that compared with control group, the concentrations
2. Results of IL-6, interleukin-1𝛽 (IL-1𝛽), and tumor necrosis factor alpha
(TNF-𝛼) were significantly increased in the BRG and GC groups
2.1. The Levels of Conjugated BAs Were Highly Associated with (Figure 2A). We also found that the concentrations of LPS in gas-
Bile Reflux Gastritis tric juice were significantly increased in the BRG and GC groups
but no difference of LPS levels was detected between BRG and
To investigate the BA characteristic of bile reflux gastritis, we col- GC groups (Figure 2B).
lected gastric juice samples of control (n = 50), BRG (n = 50), To further determine whether the levels of LPS-producing bac-
and GC (n = 45). The esophagogastroduodenoscopy (EGD) exam- teria increased correspondingly, we assessed microbial diversity
ination revealed large amounts of yellow-green fluid that resided and richness of gastric juice samples via the analysis of full-
in the gastric cavity of patients with BRG, and the hematoxylin length 16S rRNA gene sequencing. By measuring 𝛼 diversity us-
and eosin (H&E) staining of the stomach showed gastric precan- ing the Shannon index, we found that there was no significant
cerous lesions (intestinal metaplasia) in the BRG group and gas- difference among the three groups. Next, we determined the sig-
tric cancer cell infiltration in GC group (Figure 1A). The BA pro- nificantly increased species in the BRG (Figure 2C) and GC (Fig-
files of the gastric juice samples among control, BRG, and GC ure 2D) groups by linear discriminant analysis effect size (LEfSe)
groups were measured using an ultra-performance liquid chro- differential analysis, compared to the control group. The relative
matography coupled with a triple quadrupole mass spectrome- abundance of Neisseria sicca (N. sicca), Prevotella melaninogenica
try (UPLC/TQMS) based targeted metabolomics approach. The (P. melaninogenica), P. jejuni, Veillonella parvula (V. parvula), V.
results revealed that the total BAs (the sum of all detected con- atypica, and P. pallens were significantly elevated in both BRG
jugated and unconjugated BAs (the BAs in the free form)), es- and GC groups with P. melaninogenica having the highest relative
pecially conjugated BAs, were significantly elevated in patients abundance in both BRG and GC groups, 5.15% and 5.88%, re-
with BRG and GC (Figure 1B–D), among which glycocholic acid spectively (Figure 2E). We next applied a metagenomic approach
(GCA), glycochenodeoxycholic acid (GCDCA), glycodeoxycholic and found that most of the species significantly increased in gas-
acid (GDCA), glycoursodeoxycholic acid (GUDCA), taurocholic tric juice of BRG (5 of 6, 83%) and GC (10 of 13, 77%) groups
acid (TCA), taurochenodeoxycholic acid (TCDCA), TDCA, and were LPS-producing bacteria (Figure 2C,D, marked with aster-
tauroursodeoxycholic acid (TUDCA) increased substantially (Fig- isks), consistent with the significant increase of LPS levels (Fig-
ure S1A, Supporting Information). Meanwhile, the unconjugated ure 2B). However, none of the five elevated species in the con-
BAs in the gastric juice of the BRG and GC groups also increased trol group was LPS-producing bacteria (Figure S1C, Supporting
significantly (Figure S1B, Supporting Information), but the abso- Information). In addition, the pathway analysis of metagenomic
lute concentrations and the fold change (FC) of them were much data showed that the activation of the LPS biosynthetic pathway

Adv. Sci. 2022, 9, 2200263 2200263 (2 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

Figure 1. Conjugated BAs were highly associated with bile reflux gastritis. Human gastric sections and juice are analyzed in control (n = 50), BRG
(n = 50), and GC (n = 45) groups. A) Representative images of EGD and H&E staining, bars, 50 μm. B) Heatmaps of normalized concentrations of
the conjugated BAs in human gastric juice. The color of each cell in the heatmap corresponds to the normalized and log-transformed raw abundance
of the BAs in each sample. C) Concentrations of total BAs in human gastric juice. D) Concentrations of conjugated BAs in human gastric juice. E)
pH values of human gastric juice. F) Heatmaps of Spearman correlation coefficients of 8 elevated conjugated BAs and pH value in human gastric
juice. The color of the cells indicates the Spearman correlation coefficients (r). G) Concentrations of TDCA in human gastric juice. Data are shown
as mean with SEM. Differences between groups were assessed using the Kruskal-Wallis test, #p < 0.005, *p < 0.05. BRG: bile reflux gastritis; GC:
gastric cancer; EGD: esophagogastroduodenoscopy; H&E: hematoxylin and eosin; GCA: glycocholic acid; GCDCA: glycochenodeoxycholic acid; GDCA:
glycodeoxycholic acid; GUDCA: glycoursodeoxycholic acid; TCA: taurocholic acid; TCDCA: taurochenodeoxycholic acid; TDCA: taurodeoxycholic acid;
TUDCA: tauroursodeoxycholic acid; BAs: bile acids.

Adv. Sci. 2022, 9, 2200263 2200263 (3 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

Figure 2. Increased conjugated BAs were associated with the elevated abundance of LPS-producing bacteria. A) Concentrations of inflammatory cy-
tokines in human gastric juice from control, BRG, and GC groups. B) Concentrations of LPS in human gastric juice from control, BRG, and GC groups.
C) Elevated species in gastric juice of BRG group based on the LEfSe analysis between BRG and control groups. The species marked with asterisks are
those with LPS biosynthesis function in the metagenomic analysis. D) Elevated species in gastric juice of GC group based on the LEfSe analysis between
GC and control groups. The species marked with asterisks are those with LPS biosynthesis function in the metagenomic analysis. E) Relative abundance
of the 5 LPS-producing species elevated in both BRG and GC groups. F) Relative abundance of the LPS biosynthesis pathway in human gastric micro-
biota based on metagenomic analysis. G) Heatmaps of Spearman correlation coefficients of 8 elevated conjugated BAs and Prevotella melaninogenica in
human gastric juice. The color of the cells indicates the Spearman correlation coefficients (r). Data are shown as mean with SEM. Differences between
groups were assessed using the Kruskal-Wallis test, #p < 0.005, *p < 0.05. BRG: bile reflux gastritis; GC: gastric cancer; LEfSe: linear discriminant analysis
effect size; LPS: lipopolysaccharide; TDCA: taurodeoxycholic acid; GDCA: glycodeoxycholic acid; GCA: glycocholic acid; GCDCA: glycochenodeoxycholic
acid; TCA: taurocholic acid; GUDCA: glycoursodeoxycholic acid; TUDCA: tauroursodeoxycholic acid; TCDCA: taurochenodeoxycholic acid.

Adv. Sci. 2022, 9, 2200263 2200263 (4 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

was significantly enriched in both BRG and GC groups compared 43 weeks of intervention, there were only slight differences in
with control group (Figure 2F). In order to explore the relation- the mice body weights (Figure S3A, Supporting Information) and
ship between the refluxed BAs and LPS-producing bacteria, we food intake (Figure S3B, Supporting Information). Obvious in-
performed Spearman correlation analysis and found that except flammatory cell infiltration in gastric tissues was found in mice
N. sicca, the other four LPS-producing bacteria had a significant treated with 43 weeks of TDCA, LPS, and TDCA with LPS (Fig-
positive correlation with the elevated conjugated BAs (Figure 2G; ure 4A), suggesting that long-term intervention of TDCA and
Figure S1D, Supporting Information), in which TDCA had the LPS promoted gastric inflammation in mice. Moreover, levels of
strongest positive correlation with P. melaninogenica (Figure 2G). LPS and LPS-producing bacteria (P. melaninogenica) were signif-
Collectively, our results demonstrated that bile reflux may have icantly increased in gastric contents of TDCA, LPS, and TDCA
induced significant changes in the composition of gastric micro- with LPS groups (Figure 4B,C), which was consistent with our
biota, mainly manifested as an increase in the relative abundance clinical findings. The concentrations of TDCA were significantly
of LPS-producing bacteria, a result consistent with the observed increased in the contents and tissues of the TDCA and TDCA
increased amounts of LPS in gastric juice. with LPS groups (Figure S3C–J, Supporting Information). These
results provided supporting evidence that TDCA and LPS were
capable of promoting gastric inflammation in mice.
2.3. TDCA and LPS Promoted Growth of Gastric Epithelial Cells To assess whether TDCA and LPS induced the increase of
IL-6 level, we performed enzyme-linked immunosorbent assay
To verify whether the conjugated BAs and LPS influence the (ELISA) and quantitative real-time polymerase chain reaction
growth of gastric epithelial cells in vitro, we performed cell pro- (RT-qPCR) and the results showed that both IL-6 concentrations
liferation assays to determine the effects of the conjugated BAs and gene expression were significantly increased in TDCA, LPS,
(0, 50, 100 μm final concentrations) and LPS (0, 50, 100 ng mL−1 and TDCA with LPS treated groups, with the highest level in
final concentrations) on the growth of the normal gastric epithe- TDCA with LPS group (Figure 4D,E). Activation of glycoprotein
lial cell line (GES-1) (Figure 3A) and the gastric cancer cell line 130 (GP130), also known as IL-6 signal transducer, promotes
(AGS) (Figure 3B). The results showed that TDCA had the great- the phosphorylation of Janus kinase 1 (JAK1), which in turn
est effect on promoting cell growth of both GES-1 and AGS in phosphorylates signal transducer and activator of transcription
a dose-dependent manner. We then used TDCA (100 μm), LPS 3 (STAT3) on tyrosine-705 (p-STAT3 (Y705)). Constitutive activa-
(100 ng mL−1 ), and TDCA (100 μm) with LPS (100 ng mL−1 ) tion of STAT3 protein is a common pro-inflammatory oncogenic
to treat both GES-1 and AGS cells. The results showed that the feature identified in numerous solid tumors including gastric
TDCA with LPS treated group exhibited a higher proliferation cancer.[26,27] To confirm if the increase of IL-6 levels induced by
rate (Figure 3C,D). Consistent with the results of proliferation TDCA and LPS can activate the STAT3 pathway, we examined
assays, the number of clones of GES-1 (Figure 3E) and AGS (Fig- the levels of STAT3 phosphorylation using Western blot anal-
ure 3F) increased significantly after TDCA (100 μm), LPS (100 ng ysis and immunohistochemistry staining. The results showed
mL−1 ), and TDCA (100 μm) with LPS (100 ng mL−1 ) respective that the levels of STAT3 phosphorylation were significantly in-
interventions. These results revealed that TDCA with LPS pro- creased in TDCA, LPS, and TDCA with LPS groups (Figure 4F;
moted the most cell growth of GES-1 and AGS cells. Figure S4, Supporting Information). Furthermore, the mRNA of
To further investigate how TDCA and LPS directly promote the the STAT3 target genes (c-Myc, Vegf, Bcl2 and Cxcl10) were upreg-
cell proliferation, we performed the Western blot analysis of the ulated correspondingly (Figure 4G). These results suggested that
proliferation markers, Ki-67 and proliferating cell nuclear anti- TDCA and LPS promoted gastric inflammation in mice, probably
gen (PCNA) in the GES-1 cells with TDCA (100 μm), LPS (100 ng through the activation of STAT3 signaling pathway. In addition,
mL−1 ), and TDCA (100 μm) with LPS (100 ng mL−1 ) interven- the immunohistochemistry results showed that the protein lev-
tion for 24 h. The results showed that the protein levels of Ki- els of Ki-67 were significantly increased in the TDCA, LPS, and
67 were significantly increased after treatment with TDCA, LPS, TDCA with LPS groups (Figure S5, Supporting Information).
and TDCA with LPS, while no significant changes were found
in PCNA (Figure S2, Supporting Information). Furthermore, we
found the protein levels of Cyclin D1, a regulator of cell cycle 2.5. P. melaninogenica Induced Gastric Inflammation in Mice
progression, were significantly upregulated by TDCA, LPS, and
TDCA with LPS (Figure S2, Supporting Information), which sug- To verify whether LPS-producing bacteria can promote gastric in-
gested that TDCA and LPS may promote cell proliferation by ac- flammation, we used P. melaninogenica (PM) (1 × 109 cfu/day)
celerating cell cycle. and PM (1 × 109 cfu/day) with TDCA (120 mg/kg/day) to treat
C57BL/6J mice for 43 weeks, and saline for the control group.
Body weights (Figure S6A, Supporting Information) and food in-
2.4. TDCA and LPS Promoted Gastric Inflammation in Mice takes (Figure S6B, Supporting Information) had slight changes
between control, PM, and PM with TDCA groups. Compared
Our clinical results showed that bile reflux can significantly in- with the control group, there was obvious inflammatory cell in-
crease the levels of inflammatory factors. To further investigate filtration in gastric tissues of PM and PM with TDCA groups
whether TDCA and LPS can promote gastric inflammation in (Figure 5A). Accordingly, the levels of LPS and LPS-producing
vivo, we used saline (control vehicle), TDCA (120 mg/kg/day), bacteria, P. melaninogenica in gastric contents were substantially
LPS (0.05 mg/kg/day), and TDCA (120 mg/kg/day) with LPS elevated in PM and PM with TDCA groups, in which PM with
(0.05 mg/kg/day) to treat C57BL/6J mice for 43 weeks. During TDCA group had a higher level (Figure 5B,C). The concentra-

Adv. Sci. 2022, 9, 2200263 2200263 (5 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

Figure 3. TDCA and LPS promoted the growth of gastric epithelial cells. A) The effects of 8 conjugated BAs on regulating cell proliferation of normal
gastric epithelial cells (GES-1) were evaluated by a Cell Counting Kit-8 (CCK-8) assay. B) The effects of 8 conjugated BAs on regulating cell proliferation
of gastric cancer cell line (AGS) were evaluated by a CCK-8 assay. C) TDCA, LPS, and TDCA+LPS significantly increased cell proliferation of GES-1. D)
TDCA, LPS, and TDCA+LPS significantly increased cell proliferation of AGS. E) TDCA, LPS, and TDCA+LPS significantly increased colony formation of
GES-1. F) TDCA, LPS, and TDCA+LPS significantly increased colony formation of AGS. Data are shown as mean with SEM. Differences between groups
(all groups compared to the control group) were assessed using the one-way ANOVA test, #p < 0.005, *p < 0.05. TDCA: taurodeoxycholic acid; GDCA:
glycodeoxycholic acid; TUDCA: tauroursodeoxycholic acid; GUDCA: glycoursodeoxycholic acid; TCA: taurocholic acid; GCA: glycocholic acid; TCDCA:
taurochenodeoxycholic acid; GCDCA: glycochenodeoxycholic acid; LPS: lipopolysaccharide.

tions of TDCA in gastric contents and tissues were significantly mation in mice and activation of the STAT3 signaling pathway.
increased in PM with TDCA group (Figure S6C–J, Supporting In- The immunohistochemistry results showed that the protein lev-
formation). Meanwhile, IL-6 concentration and gene expression els of Ki-67 were significantly increased in the PM and PM with
showed similar group differences (Figure 5D,E). The results of TDCA groups (Figure S8, Supporting Information).
both Western blot analysis and immunohistochemistry staining
showed that the levels of STAT3 phosphorylation were signifi-
cantly increased in PM and PM with TDCA groups (Figure 5F; 2.6. TDCA and LPS Promoted Gastric Carcinogenesis by
Figure S7, Supporting Information). Additionally, the mRNA lev- Activating IL-6/JAK1/STAT3 Pathway in Gastric Epithelial Cells
els of STAT3 target genes were also significantly upregulated in
the two interventions groups (Figure 5G). Taken together, LPS- To determine whether TDCA and LPS can also promote the ac-
producing bacteria, P. melaninogenica, promoted gastric inflam- tivation of IL-6 and STAT3 pathways in vitro, we used TDCA

Adv. Sci. 2022, 9, 2200263 2200263 (6 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

Figure 4. TDCA and LPS promoted gastric inflammation in mice. A) Representative images of H&E staining of gastric tissues from control (n = 6),
TDCA (n = 8), LPS (n = 8), and TDCA+LPS (n = 8) groups, bars, 50 μm. B) Concentrations of LPS in gastric contents from control, TDCA, LPS, and
TDCA+LPS groups. C) Relative gene expression of P. melaninogenica in gastric contents from control, TDCA, LPS, and TDCA+LPS groups by RT-qPCR.
D) Concentrations of IL-6 in gastric tissues from control, TDCA, LPS, and TDCA+LPS groups. E) Relative mRNA expression of Il6 in gastric tissues from
control, TDCA, LPS, and TDCA+LPS groups. F) The gastric protein expression of STAT3 in the mice from control, TDCA, LPS, and TDCA+LPS groups.
G) Relative mRNA expression of STAT3 target genes in gastric tissues from control, TDCA, LPS, and TDCA+LPS groups. Data are shown as mean with
SEM. Differences between groups were assessed using the one-way ANOVA test or Kruskal-Wallis test, #p < 0.005, *p < 0.05. H&E: hematoxylin and
eosin; TDCA: taurodeoxycholic acid; LPS: lipopolysaccharide.

(100 μm), LPS (100 ng mL−1 ), and TDCA (100 μm) with LPS (100 μm) with LPS (100 ng mL−1 ) for 24 h. The results indi-
(100 ng mL−1 ) to intervene GES-1 cells for 4 h. We found that both cated that both TDCA and LPS activated the phosphorylation of
the concentrations of IL-6 in the supernatant and the mRNA ex- JAK1(Y1022), STAT3 (Y705), and the mRNA expression levels of
pression levels were significantly increased after treatment with STAT3 target genes (VEGF, C-MYC, BCL-2, and CXCL10) (Fig-
TDCA, LPS, and TDCA with LPS (Figure 6A,B), which were con- ure 6C,D). These results suggested that TDCA and LPS promoted
sistent with the results of our clinical and in vivo experiments the growth of gastric epithelial cells possibly through the activa-
(Figures 2A, 4D, and 5D). These results suggested that TDCA tion of the IL-6/JAK1/STAT3 pathway.
and LPS promote the secretion of IL-6 in GES-1 cells. To further verify the effects of TDCA and LPS on STAT3 ac-
To examine whether the increase in IL-6 levels induced by tivation, GES-1 cells were treated with an inhibitor of STAT3,
TDCA and LPS can activate the STAT3 pathway, GES-1 cells were cryptotanshinone (15 μm) for 30 min before the 24-h inter-
treated with TDCA (100 μm), LPS (100 ng mL−1 ), and TDCA vention with TDCA (100 μm), LPS (100 ng mL−1 ), and TDCA

Adv. Sci. 2022, 9, 2200263 2200263 (7 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

Figure 5. P. melaninogenica induced gastric inflammation in mice. A) Representative images of H&E staining of gastric tissues from control (n = 6),
PM (n = 8), and PM+TDCA (n = 7) groups, bars, 50 μm. B) Concentrations of LPS in gastric contents from control, PM, and PM+TDCA groups. C)
Relative gene expression of P. melaninogenica in gastric contents from control, PM, and PM+TDCA groups by RT-qPCR. D) Concentrations of IL-6 in
gastric tissues from control, PM, and PM+TDCA groups. E) Relative mRNA expression of Il6 in gastric tissues from control, PM, and PM+TDCA groups.
F) The gastric protein expression of STAT3 in the mice from control, PM, and PM+TDCA groups. G) Relative mRNA expression of STAT3 target genes
in gastric tissues from control, PM, and PM+TDCA groups. Data are shown as mean with SEM. Differences between groups were assessed using the
one-way ANOVA test or Kruskal-Wallis test, #p < 0.005, *p < 0.05. H&E: hematoxylin and eosin; PM: P. melaninogenica; TDCA: taurodeoxycholic acid;
LPS: lipopolysaccharide.

(100 μm) with LPS (100 ng mL−1 ). Cryptotanshinone is a bioac- 2.7. Bile Reflux Caused Gastric Carcinogenesis and
tive compound isolated from the roots of Salvia miltiorrhiza that Cryptotanshinone Achieved Targeted Therapeutic Effects in Mice
specifically blocks STAT3 phosphorylation at Tyr705.[28] The re-
sults showed that the effects of TDCA and LPS on activating To further approach the effects of clinical BRG on gastric car-
STAT3 were abolished in the presence of cryptotanshinone (Fig- cinogenesis, we conducted a BR surgery model of C57BL/6J
ure 6E), supporting our hypothesis that TDCA and LPS promote mice. Briefly, side-to-side anastomosis was performed between
the proliferation of gastric epithelial cells by IL-6/JAK1/STAT3 the greater curvature of the stomach and the upper jejunum
signaling. located approximately 1.5 cm distally from the pylorus ring

Adv. Sci. 2022, 9, 2200263 2200263 (8 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

Figure 6. TDCA and LPS promoted gastric carcinogenesis by activating IL-6/JAK1/STAT3 pathway in gastric epithelial cells. A) Concentrations of IL-6
in the supernatant of GES-1 cells increased significantly in TDCA, LPS, and TDCA+LPS groups. B) Relative mRNA expression of IL6 in GES-1 cells
increased significantly in TDCA, LPS, and TDCA+LPS groups. C) Western blot analysis showing the activation of IL-6/JAK1/STAT3 pathway in TDCA,
LPS, and TDCA+LPS groups. D) Relative mRNA expression of STAT3 target genes in GES-1 cells increased significantly in TDCA, LPS, and TDCA+LPS
groups. E) Western Blot results showed STAT3 inhibitor (cryptotanshinone) markedly attenuated the activation effect of IL-6/JAK1/STAT3 pathway in
GES-1 cells. Data are shown as mean with SEM. Differences between groups were assessed using the one-way ANOVA test, #p < 0.005, *p < 0.05.
TDCA: taurodeoxycholic acid; LPS: lipopolysaccharide.

(Figure 7A). Mice in the BR surgery with cryptotanshinone group the sham group (Figure 7B–D). These results indicated that we
received intraperitoneal injections of cryptotanshinone. For 50 successfully established a bile reflux mouse model through the
weeks after surgery, all mice were fasted overnight before be- BR surgery. In addition, total BAs, conjugated BAs, and TDCA
ing euthanized. Overall, the body weights and food intakes of the concentrations had a similar group difference in gastric tissues
sham group had slight differences relative to the BR surgery and (Figure S9C–H, Supporting Information).
BR surgery with cryptotanshinone groups, which indicated BR The results of H&E showed that 50 weeks after BR surgical
surgery had no significant effects on the body weights (Figure intervention, 3 out of 8 mice in the BR surgery group developed
S9A, Supporting Information) and slight changes on food intakes gastric lesions, of which 1 was a precancerous lesion (dysplasia),
of mice (Figure S9B, Supporting Information). As expected, the and the remaining 2 were atypical hyperplasia (Figure 7E). How-
total BAs, conjugated BAs, and TDCA in gastric contents were ever, the 6 mice in the BR surgery with cryptotanshinone group
greatly increased in the BR surgery and BR surgery with cryp- showed no obvious gastric lesions, only inflammatory cell infil-
totanshinone groups, which were more than ten times of that in tration, which suggested that cryptotanshinone has a potential

Adv. Sci. 2022, 9, 2200263 2200263 (9 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

Figure 7. Bile reflux caused gastric carcinogenesis and cryptotanshinone achieved targeted therapeutic effects in mice. A) Schematic diagram of the BR
surgery in C57BL/6J mice. B) Concentrations of total BAs in gastric contents from sham (n = 8), BR surgery (n = 8), and BR surgery+cryptotanshinone
(n = 6) groups. C) Concentrations of conjugated BAs in gastric contents from sham, BR surgery, and BR surgery+cryptotanshinone groups. D) Con-

Adv. Sci. 2022, 9, 2200263 2200263 (10 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

therapeutic effect on gastric injury caused by bile reflux in mice. while, the levels of LPS and IL-6 in gastric juice also increased sig-
These results demonstrated that bile reflux promoted gastric car- nificantly in BRG and GC groups. Further, we provided multiple
cinogenesis in mice which could be markedly attenuated by the lines of evidence, using in vitro and in vivo models that supported
STAT3 inhibitor, cryptotanshinone. a role for TDCA and LPS in the upregulation of IL-6/JAK1/STAT3
Consistent with our clinical study, the level of P. melanino- pathway activity. Finally, we established a BR model in mice by
genica was greatly increased in the gastric contents of the BR gastrojejunostomy, and verified the results found in our clinical
surgery and BR surgery with cryptotanshinone groups (Fig- study.
ure 7F), which implied that BR surgery promoted the growth of In previous studies, deoxycholic acid or chenodeoxycholic acid
P. melaninogenica. Accordingly, the LPS level in gastric contents was commonly selected directly for animal or cell experiments
had similar group differences (Figure 7G). Meanwhile, both the to explore the effect of bile reflux on gastric cancer,[14,37,38] how-
IL-6 concentrations and mRNA expression were significantly in- ever, the rationality for BAs selection was not discussed. Our
creased in the BR surgery and BR surgery with cryptotanshinone study identified specifically that conjugated BAs were increased
groups (Figure 7H,I). Both the results of Western blot analysis in the BRG and GC groups with only minimal changes in uncon-
and immunohistochemistry staining showed that the phospho- jugated BAs. In addition, the concentrations of conjugated BAs
rylation levels of STAT3 significantly increased in the BR surgery were much higher than that of unconjugated BAs. Therefore, we
group compared to the sham group (Figure 7J; Figure S10, Sup- used the significantly elevated TDCA in the BRG group as an in-
porting Information). However, when the BR surgery with cryp- tervention in our cell and animal experiments to be consistent
totanshinone group was compared with the BR surgery group, with the clinical findings.
the level of STAT3 phosphorylation was significantly decreased, Many existing studies have focused on the effect of BAs on the
indicating that cryptotanshinone had an offsetting effect on the gastric mucosa after BRG,[13,39,40] but less attention has been paid
activation of STAT3 induced by bile reflux. The RT-qPCR results to the effect of significantly changed intragastric bacteria, which
revealed similar group differences for the mRNA expression of may play an important role in the development of gastric mu-
STAT3 target genes (Figure 7K). Consistent with the previous cell cosa lesions. It was generally perceived that microbial coloniza-
and animal results, BR surgery induced an increase in the protein tion of the stomach was unlikely since gastric conditions are in-
levels of Ki-67 compared with the sham group (Figure S11, Sup- hospitable to microorganisms.[41] In the present study, increased
porting Information). However, when the BR surgery with cryp- conjugated BAs led to a decreased gastric acidity and thus, we
totanshinone group was compared with the BR surgery group, hypothesized that the microbiota composition had significantly
the levels of Ki-67 were obviously decreased (Figure S11, Sup- changed in the stomach of patients with BRG and GC. Using
porting Information). full-length 16S rRNA gene sequences analysis, we found that
the bacterial community structure in the BRG and GC groups
changed significantly. The most significant change was an in-
3. Discussion
crease in the relative abundance of LPS-producing bacteria, such
Bile reflux was found to be closely associated with the develop- as P. melaninogenica and P. jejuni. Furthermore, we applied a
ment of gastric precancerous lesions and gastric cancer,[29–31] but metagenomics approach that revealed the enrichment of the LPS
the reason for this connection remains unclear. The length of biosynthesis pathway in the BRG and GC groups. In addition, the
time for BAs exposure correlated with the severity of patholog- concentrations of LPS increased significantly in the gastric juice
ical changes in the gastric mucosa.[24] In addition, BAs directly of the BRG and GC groups, which were consistent with the re-
induced intestinal metaplasia and progression to neoplasia of sults from the 16S rRNA gene sequencing and metagenome anal-
the stomach.[29,32,33] Although BAs are thought to be critical in ysis. Additionally, there was no significant difference in Chao1
the pathogenesis of gastric mucosal diseases, the mechanisms or Shannon index among the control, BRG, and GC groups. The
by which BAs induce transformation in the stomach are not yet changes of microbial 𝛼 diversity in gastric cancer are not consis-
clear.[34] Previously, some researchers used deoxycholic acid to es- tent in previous studies,[42–45] which might be due to the antibi-
tablish chronic gastritis animal models.[35] Other previous stud- otic treatment or dietary habits of the patients, or the limitation
ies proposed that bile reflux functioned as an initiator of gastric of sample size in different studies.
carcinogenesis.[4,36] According to previous reports, researchers have established a
In this study, we have shown that BAs especially conjugated model of BR using Wistar or Fischer rats through surgery and
BAs were significantly increased in gastric juice of patients with used this model to further study the relationship between BRG
BRG and GC. The increased BAs were found to be associated and gastric cancer,[5,30] but few studies have reported establish-
with the elevated abundance of LPS-producing bacteria. Mean- ing a BR model in mice. Rats do not possess a gallbladder and

centrations of TDCA in gastric contents from sham, BR surgery, and BR surgery+cryptotanshinone groups. E) Representative images of H&E staining
of gastric tissues from sham, BR surgery, and BR surgery+cryptotanshinone groups, bars, 50 μm. F) Relative gene expression of P. melaninogenica in
gastric contents from sham, BR surgery, and BR surgery+cryptotanshinone groups by RT-qPCR. G) Concentrations of LPS in gastric contents from
sham, BR surgery, and BR surgery+cryptotanshinone groups. H) Relative mRNA expression of Il6 in gastric tissues from sham, BR surgery, and BR
surgery+cryptotanshinone groups. I) Concentrations of IL-6 in gastric tissues from sham, BR surgery, and BR surgery+cryptotanshinone groups. J) The
gastric protein expression of STAT3 in the mice from sham, BR surgery, and BR surgery+cryptotanshinone groups. K) Relative mRNA expression of
STAT3 target genes in gastric tissues from sham, BR surgery, and BR surgery+cryptotanshinone groups. Data are shown as mean with SEM. Differences
between groups were assessed using the one-way ANOVA test or Kruskal-Wallis test, #p < 0.005, *p < 0.05. BR: bile reflux; LPS: lipopolysaccharide;
TDCA: taurodeoxycholic acid; H&E: hematoxylin and eosin.

Adv. Sci. 2022, 9, 2200263 2200263 (11 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

therefore the use of BR mice which do have one was thought systems based on primary human gastric cells may serve as more
to be a closer pre-clinical model. In this study, we successfully realistic model systems for future studies.
constructed a surgical model of BR in C57BL/6 J mice and used
this model to further confirm the results of our clinical and cel-
lular studies. Only 37.5% of mice in the BR surgery group de-
veloped gastric lesions after 50 weeks post-surgery, which may 4. Experimental Section
be due to the long time required for bile reflux to induce gastric Experimental Design: The main objective of this study was to investi-
disease. The mice developing lesions did respond to the STAT3 gate the changes in the BA profiles of patients with BRG and to explore
inhibitor, which confirmed the results of our cell studies regard- the effects of refluxed BAs and microbiome on gastric carcinogenesis. Pa-
ing IL-6/JAK1/STAT3 involvement in GC development. tients’ sample collection and use were approved by the ethical committee
Bile acids could induce proliferation of rat hepatic stellate of Shanghai Jiao Tong University Affiliated Sixth People’s Hospital (ethics
approval number: No. 2020–097). All subjects were provided with a full ex-
cells[46] or human colon cancer cell line[47] via activation of the planation regarding the nature of the study, and written informed consent
epidermal growth factor receptor. Several studies have reported was obtained from each of the patients prior to starting the endoscopy
that BAs regulate the proliferation or intestinal metaplasia of gas- (Chinese Clinical Trial Register (ChiCTR) number: ChiCTR2000035004).
tric epithelial cells by directly interacting with their receptors, The subjects in the control group were those without bile reflux gastri-
such as the Farnesoid X receptor (FXR)[14] and G protein-coupled tis, gastric cancer, or the following conditions: other cancers, acute and
bile acid receptor 1 (TGR5).[36] In our study, we found that TDCA chronic gastrointestinal diseases, acute infectious disease, receipt of any
antibiotic treatment within 3 months before sample collection, regularly
and LPS could promote the proliferation of gastric cells. Further-
taking prescription Chinese or Western medicines, pregnancy, H. pylori
more, the results of Western Blot showed that the protein levels treatment, history of surgery, history of taking proton pump inhibitors or
of Ki67 (a proliferation marker) and Cyclin D1 (a regulator of cell histamine-2 receptor blockers.
cycle progression) were significantly upregulated by TDCA, LPS, All animal procedures and testing were approved by the national legisla-
and TDCA with LPS, suggesting that TDCA and LPS may pro- tion and local guidelines of the laboratory animals center at Shanghai Jiao
mote cell proliferation by accelerating cell cycle. In the future, Tong University Affiliated Sixth People’s Hospital, Shanghai, China (ethics
approval number: No. 2017-0038). Sample sizes for animal studies were
more mechanistic experiments should be conducted to further
determined by statistical analysis of variance and on the basis of our ex-
verify whether TDCA and LPS promote cell proliferation by ac- perience with similar studies. The sample size (n) for each experimental
celerating the cell cycle and ultimately promote gastric carcino- group is indicated in the corresponding figure legends and was between
genesis. six and eight mice per group. The number of replicates for each experiment
Consistently with previous studies that there was a signifi- is described in the relevant figure legends.
cant increase in the potentially pathogenic bacteria in gastric Human Gastric Juices: A total of 145 patient cases who underwent an
cancer,[44,45,48] such as Prevotella and Veillonella. In our study, P. EGD examination were enrolled in the present study. Patients with a his-
tory of gastric surgery, a history of H. pylori eradication, pregnant patients,
melaninogenica, an LPS-producing bacteria, was observed to be and patients who were judged by the attending physicians to be unsuitable
significantly increased in the gastric juice of BRG and GC groups. for endoscopy were excluded from the study. Endoscopy was performed
In a cohort of 276 patients with gastric cancer,[45] the researchers in the early morning in participants who had not taken any food, water,
have found that P. melaninogenica was significantly increased in or drugs since the previous night. Gastric juice in the gastric mucous lake
the tumoral tissues compared to peritumoral tissues. As an oral was aspirated as much as possible immediately before the endoscopic ob-
and respiratory pathogen, P. melaninogenica can colonize and servation. Gastric juice was aspirated through the forceps channel of the
endoscope into a recovery vessel. The clinical characteristics of the 145
overgrow in the stomach of the BRG group, which may be due
subjects are listed in online Table S1, Supporting Information, and the de-
to a large amount of bile acids reflux into the stomach, changing tailed procedures of gastric juice collection is in Supporting Information.
the intragastric environment, such as the increase of pH value. Animal Experiment 1: Four-week-old C57BL/6J male mice were pur-
Recently published study on Gut, 2021[49] showed that gastric mi- chased from Shanghai Sippr-BK Laboratory Animal Co. Ltd. (Shanghai,
crobiota from patients with intestinal metaplasia and gastric can- China). All the mice were maintained in a specific-pathogen-free (SPF)
cer induced premalignant lesions in the gastric mucosa of recipi- environment with controlled conditions, a 12 h light/dark cycle at 20—
22 °C and 45 ± 5% humidity. The mice were acclimated with a normal
ent germ-free mice after one-year post inoculation, providing the
diet for 1 week and subsequently divided into 4 groups: Mice were orally
causality of associations of human gastric microbiome in the on- gavaged with saline solution (control, n = 6), TDCA (120 mg/kg/day, n =
set of gastric cancer. The germ-free mice model will be applied 8), and LPS (0.05 mg/kg/day, n = 8) or TDCA (120 mg/kg/day) plus LPS
in future studies to investigate the role of the microbiota in the (0.05 mg/kg/day) (n = 8) for 43 weeks.
onset of gastric cancer. Animal Experiment 2: Four-week-old C57BL/6J male mice were pur-
In our study, conjugated BAs and LPS increased significantly chased from Shanghai Sippr-BK Laboratory Animal Co. Ltd. (Shanghai,
in the stomach of the BRG and GC groups, which, in turn, China). All the mice were maintained in an SPF environment with con-
trolled conditions, a 12 h light/dark cycle at 20—22 °C and 45 ± 5% hu-
promoted gastric lesions by upregulating the IL-6/JAK1/STAT3 midity. The mice were acclimated with a normal diet for 1 week and sub-
pathway. In the future, the number of clinical and animal sam- sequently divided into 3 groups: Mice were orally gavaged saline solution
ples should be expanded to verify the current findings. In addi- (control, n = 6), PM (1 × 109 cfu/day, n = 8), and TDCA (120 mg/kg/day)
tion, the correlation of conjugated BAs and gastric microbiota plus PM (1 × 109 cfu/day) (n = 7) respectively, for 43 weeks.
should also be investigated, such as the mechanism through Animal Experiment 3: Six-week-old C57BL/6J male mice were pur-
which BAs increase the abundance of LPS-producing bacteria. chased from Shanghai Sippr-BK Laboratory Animal Co. Ltd. (Shanghai,
China) and used for constructing a BR model. All the mice were main-
Meanwhile, GES-1, the SV40 transformed human fetal GES, has
tained in a SPF environment with controlled conditions, a 12 h light/dark
been shown to be non-tumorigenic in nude mice and is consid- cycle at 20–22 °C and 45 ± 5% humidity. The mice were acclimated with a
ered a non-malignant cell line that can be used to mimic human normal diet for 1 week and subsequently divided into 3 groups: sham (n
gastric epithelial cells suffering from BAs reflux. Other model = 8), BR surgery (n = 8), and BR surgery with cryptotanshinone (n = 6)

Adv. Sci. 2022, 9, 2200263 2200263 (12 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

groups. After 24 h of fasting, a midline laparotomy incision was performed platform (Illumina, USA) with PE150 strategy at Personal Biotechnology
under anesthesia with 60 mg kg−1 sodium pentobarbital. The following Co., Ltd. (Shanghai, China). The sequencing data of the metagenomic ap-
procedure was performed according to previously reported methods used proach are listed in online Table S3, Supporting Information. The detailed
to construct rat reflux models.[50,51] Briefly, side-to-side anastomosis was description of the pipeline of sequencing data analysis is in Supporting
performed between the greater curvature of the stomach and the upper Information.
jejunum located approximately 1.5 cm distally from the pylorus ring. The P. melaninogenica Abundance Analysis: The microbial genomic DNA
saline solution was injected into the peritoneal cavity to prevent adhesion of gastric contents in mice was extracted using the DNeasyPowerSoil Kit
and as postoperative fluid replenishment, and the wound was closed. Mice (QIAGEN, Inc., Netherlands), following the manufacturer’s instructions.
in the BR surgery with cryptotanshinone group received an intraperitoneal The quantity and quality of extracted DNAs were measured using a Nan-
injection of cryptotanshinone (20 mg/kg/day). For the construction of the oDrop ND-1000 spectrophotometer (Thermo Fisher Scientific, Waltham,
sham surgery group, the same pre- and postoperative preparations were MA, USA) and agarose gel electrophoresis, respectively. The extracted mi-
implemented as for the reflux model. Experimental mice were allowed ac- crobial DNA of all samples was first diluted to the same concentration
cess to water 24 h, and to food 36 h, postoperatively, and not treated with and then to construct SYBR Green-based quantitative real-time PCR. The
any known carcinogens. Sequential morphological changes of the stom- extracted microbial DNA was processed to construct SYBR Green-based
ach of experimental mice were studied. quantitative real-time PCR. The P. melaninogenica fraction as part of the
All the mice in the three experiments were raised with free access to nor- whole bacterial population was calculated by dividing the gene copy num-
mal diet and water, and their body weights and food intakes were recorded ber of P. melaninogenica primers[52] by the total gene copy number using
once a week. All mice were fasted overnight before being euthanized. Sam- 16S rRNA primers. The primers used in this study are listed in online Table
ples of gastric tissues and gastric contents were carefully collected and S4, Supporting Information.
kept in liquid nitrogen and then stored at −80 °C until analysis. Luminex Multiplex Assay: The collected gastric juice of human and
Cell Culture and Reagents: GES-1 cells, a normal gastric epithelial cell the gastric tissues of mice were immediately cryopreserved (−80 °C),
line, purchased from Fuheng Biotechnology Co., Ltd., Shanghai, were cul- and the concentrations of pro-inflammatory cytokines were quantified us-
tured in Dulbecco’s modified Eagle’s medium (DMEM, Gibco). AGS, the ing Luminex multiplex assay (R&D systems, LXSAHM-06 for human and
gastric cancer cell line purchased from American Tissue Culture Collec- LXSAMSM-05 for mice) according to the manufacturer’s instructions.
tion (ATCC), was cultured in DMEM/F-12. 10% v/v fetal bovine serum Bacterial Strain and Culture Condition: P. melaninogenica was pur-
(Gibco, qualified, Australia origin) supplemented with a 1% v/v Peni- chased from the ATCC (ATCC, 25845) and was cultured in the ATCC
cillin/Streptomycin mix. All cells were incubated at 37 °C in an atmosphere Medium 2863 at 37 °C under anaerobic conditions.
containing 5% CO2 . The reagents used in the cell experiments included BAs Analysis: The collected gastric juices were immediately cryopre-
TCA (J&K Scientific, 909688), TCDCA (Matrix Scientific, 100646), TDCA served (−80 °C), and the BA concentrations were quantitatively deter-
(J&K Scientific, 423806), TUDCA (J&K Scientific, 496672), GCA (Aladdin, mined using UPLC/TQMS (Waters, Milford, MA) according to a protocol
G131002), GCDCA (J&K Scientific, 107563), GDCA (Aladdin, S102123), we previously established.[6,53–55]
GUDCA (J&K Scientific, G0459), LPS (Sigma, L6529) and cryptotanshi- LPS Analysis: The collected human gastric juice, and mouse gastric
none (Selleck, S2285). The ELISA kit used in the cell experiments was ac- tissues and gastric contents were immediately cryopreserved (−80 °C),
quired from LEGEND MAX Human IL-6 ELISA Kit (BioLegend, 430507). and the LPS concentrations were quantitatively measured using ELISA kits
Cell Viability Assay: Cell viability was determined by the Cell Counting (Mlbio, ml9022423 for human, ml037221-C for mouse) according to the
Kit-8 (CCK-8, Dojindo, Japan) assay. For each well in a 96-well plate, 1500 manufacturer’s instructions.
cells were seeded with 100 μL of culture medium and treated with the con- RT-qPCR: Total RNA of cell samples and gastric tissues homogenized
jugated BAs or LPS for 72 h. Then 10 μL of CCK-8 solution was added to the with TissueLyzer (QIAGEN) was isolated using TRIzol Reagent (Invitro-
cells and cells were incubated for 3 h at 37 °C. The reaction product was gen, Life Technology, USA). The total RNA concentration was measured
quantitatively measured according to the manufacturer’s instructions. using a NanoDrop 2000C spectrophotometer (Thermo Fisher Scientific,
Cell Colony Formation Assay: For each well in a 6-well plate, 250 cells Waltham, MA, USA). A purified, 500 ng sample of total RNA from each
were seeded with 3 mL of culture medium and treated with the conjugated stomach sample was reverse transcribed using random hexamer primers
BAs or LPS for 12 days. The number of cell colonies was counted after to form the cDNA templates by employing a Prime Script RT Reagent Kit
crystal violet staining. (TAKARA, Kusatsu, Japan). The qPCR primers were designed and synthe-
Full-Length 16S rRNA Gene Sequencing: Total microbial genomic DNA sized (Sangon Biotech, Shanghai, China). The qPCR reaction mixture was
samples were extracted using the OMEGA DNA isolation kit (Omega, set up using Power Up SYBR Green PCR Master Mix (Applied Biosystems,
D5625-01, USA), following the manufacturer’s instructions. The quantity Thermo Fisher Scientific, USA) and the reaction was run in an ABI 7900HT
and quality of extracted DNA were measured using a NanoDrop NC-1000 Real-Time PCR System (Applied Biosystems Instruments, Thermo Fisher
spectrophotometer (Thermo Fisher Scientific, Waltham, MA, USA) and Scientific, USA). All the procedures were handled following the manufac-
agarose gel electrophoresis, respectively. Polymerase chain reaction (PCR) turer’s instructions. The values of the target genes were normalized to
amplification of the full-length bacterial 16S rRNA gene was performed glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and the relative ex-
using the forward primer 27F (5″-AGAGTTTGATCMTGGCTCAG-3″) and pression level was shown as fold changes relative to the average value in
the reverse primer 1492R (5″-ACCTTGTTACGACTT-3″). The Quantitative the control group. The primers used in this study are listed in online Table
Insights Into Microbial Ecology (QIIME2) pipeline was employed to pro- S4, Supporting Information.
cess the sequencing data at Shanghai Personal Biotechnology Co., Ltd. Western Blot Analysis: Cell samples and gastric tissues were lysed
(Shanghai, China). The sequencing data of the full-length 16S rRNA gene with RIPA buffer (Beyotime Technology, Shanghai, China) containing
sequences are listed in online Table S2, Supporting Information. The de- 1mM PMSF (Beyotime Technology, Shanghai, China), Protease Inhibitor
tailed description of the pipeline of sequencing data analysis is in Support- Cocktail Set III (Merck Millipore), and Phosphatase Inhibitor Cocktail
ing Information. (Sigma) in an ice bath for 30 min followed by centrifugation at 14000 g for
Metagenomic Analysis: Total microbial genomic DNA samples were 10 min. The supernatants were collected, and protein concentrations were
extracted using the OMEGA DNA isolation kit (Omega, D5625-01, USA), measured using a BCA Protein Assay Kit (Pierce, Rockford, IL, USA). A 5 μg
following the manufacturer’s instructions. The quantity and quality of ex- μL−1 of protein extract was combined with loading buffer (Beyotime Tech-
tracted DNAs were measured using a NanoDrop ND-1000 spectropho- nology, Shanghai, China) and denatured by boiling at 100 °C for 10 min.
tometer (Thermo Fisher Scientific, Waltham, MA, USA) and agarose gel The denatured proteins were resolved by 12% SDS-PAGE and transferred
electrophoresis, respectively. The extracted microbial DNA samples were to Immobilon-P Transfer Membranes (Millipore Corporation, Tullagreen,
processed to construct metagenome shotgun sequencing libraries with IRL). The membranes were blocked with 5% BSA (Beyotime Technology,
insert sizes of 400 bp by using the Illumina TruSeq Nano DNA LT Library Shanghai, China) at room temperature for 1 h, incubated with primary
Preparation Kit. Each library was sequenced by the Illumina HiSeq X-ten antibodies overnight at 4 °C, and then incubated with horseradish perox-

Adv. Sci. 2022, 9, 2200263 2200263 (13 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

idase conjugated secondary antibodies. The bands were visualized using Data Availability Statement
a SuperSignal West Pico Chemiluminescent Substrate (Thermo Scientific,
Rockford, IL, USA) with a Tanon 5500 Chemiluminescent Imaging System The data that support the findings of this study are available from the cor-
(Tanon Science & Technology Co., Shanghai, China). The gray values of responding author upon reasonable request.
the bands were calculated using ImageJ software and were normalized to
𝛽-Actin. The antibodies used for the present study and the dilutions ratios
were as follows: 1:1000 for rabbit anti-STAT3 (Cell Signaling Technology,
MA, 12640S), 1:2000 for rabbit anti-p-STAT3 (T705) (Cell Signaling Tech- Keywords
nology, MA, 9145S), 1:1000 for rabbit anti-JAK1 (Cell Signaling Technology,
bile acid, bile reflux, gastric carcinogenesis, microbiome
MA, 3344S), 1:1000 for rabbit anti-p-JAK1 (T1022) (Cell Signaling Technol-
ogy, MA, 3331S), 1:1000 for rabbit anti-Cyclin D1 (Cell Signaling Technol-
ogy, MA, 2978S), 1:1000 for rabbit anti-PCNA (Cell Signaling Technology, Received: January 14, 2022
MA, 13110S), 1:100 for rabbit anti-Ki67 (Abcam, UK, ab16667), and 1:1000 Revised: February 21, 2022
for rabbit anti-𝛽-Actin (Cell Signaling Technology, MA, 4970S). Published online: March 14, 2022
Immunohistochemistry Analysis: For the immunohistochemistry stain-
ing of p-STAT3 and Ki67, the gastric tissues of the C57BL/6J mice were
fixed in 4% paraformaldehyde and embedded in paraffin according to the
manufacturer’s instructions. Then slides were deparaffinized, rehydrated,
and stained with rabbit anti-p-STAT3 (T705) (Cell Signaling Technology, [1] W. Chen, R. Zheng, P. D. Baade, S. Zhang, H. Zeng, F. Bray, A. Jemal,
MA, 9145S, 1:200), and rabbit anti-Ki67 (Abcam, UK, ab16667, 1:200). X. Q. Yu, J. He, Ca-Cancer J. Clin. 2016, 66, 115.
Statistical Analysis: Raw data of BAs quantification was obtained by [2] S. Sue, W. Shibata, S. Maeda, Biomed Res. Int. 2015, 2015, 737621.
MassLynx v4.1 and analyzed by TargetLynx v4.1 (Waters, Milford, MA). [3] M. F. Dixon, N. P. Mapstone, P. M. Neville, P. Moayyedi, A. T. Axon,
Data are expressed as mean with SEM. SPSS 26.0 (IBM SPSS, USA) and Gut 2002, 51, 351.
GraphPad Prism 8.0 (GraphPad Software, San Diego, USA) were used for [4] M. Tatsugami, M. Ito, S. Tanaka, M. Yoshihara, H. Matsui, K. Haruma,
statistical analyses and graphic generation. The sample distribution was K. Chayama, Cancer Epidemiol., Biomarkers Prev. 2012, 21, 2101.
determined using the Kolmogorov–Smirnov normality test. For statistical [5] M. Oba, K. Miwa, T. Fujimura, S. Harada, S. Sasaki, T. Hattori, Int. J.
comparisons, one-way analysis of variance (ANOVA) test or Kruskal-Wallis Cancer 2008, 123, 1491.
test followed by the post-hoc tests (Dunnett’s test) for the normal or non- [6] A. Zhao, S. Wang, W. Chen, X. Zheng, F. Huang, X. Han, K. Ge, C.
normal distributed data, respectively. Spearman’s rank correlation coeffi- Rajani, Y. Huang, H. Yu, J. Zhu, W. Jia, Sci. Rep. 2020, 10, 11601.
cients were calculated to examine the association of BAs and the pH val- [7] W. Jia, G. Xie, W. Jia, Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 111.
ues or the LPS-producing bacteria. All the p-values were adjusted by the [8] L. Y. Zhang, J. Zhang, D. Li, Y. Liu, D. L. Zhang, C. F. Liu, N. Wang, S.
false discovery rate (FDR) using the Benjamini−Hochberg method. The R. Wu, W. Q. Lu, J. Z. Guo, Y. Q. Shi, J. Dig. Dis. 2021, 22, 282.
corrected p-value of 0.05 was taken as a significance level. The 16S rRNA
[9] A. Kuwahara, T. Saito, M. Kobayashi, J. Cancer Res. Clin. Oncol. 1989,
gene sequencing analysis was performed using QIIME2[56] and R pack-
115, 423.
ages (v3.2.0). LEfSe was performed to detect differentially abundant taxa
[10] Y. Sugiyama, H. Sohma, M. Ozawa, R. Hada, Y. Mikami, M. Konn, K.
across groups.
Ono, Am. J. Surg. 1987, 153, 399.
[11] P. Bechi, A. Amorosi, R. Mazzanti, P. Romagnoli, L. Tonelli, Gastroen-
Supporting Information terology 1987, 93, 335.
[12] K. Kondo, Gastric Cancer 2002, 5, 16.
Supporting Information is available from the Wiley Online Library or from [13] X. Wang, L. Sun, X. Wang, H. Kang, X. Ma, M. Wang, S. Lin, M. Liu,
the author. C. Dai, Z. Dai, Cancer Med. 2017, 6, 788.
[14] T. Li, H. Guo, H. Li, Y. Jiang, K. Zhuang, C. Lei, J. Wu, H. Zhou, R. Zhu,
X. Zhao, Y. Lu, C. Shi, Y. Nie, K. Wu, Z. Yuan, D.-M. Fan, Y. Shi, Gut
Acknowledgements 2019, 68, 1751.
[15] N. Wang, S. Wu, J. Zhao, M. Chen, J. Zeng, G. Lu, J. Wang, J. Zhang,
This work was funded by the National Natural Science Foundation of China
J. Liu, Y. Shi, Cell Oncol. 2021, 44, 1119.
(81772530, 81974073, 82170833, 82122012) and Natural Science Founda-
tion of Shanghai (21ZR1448800). The authors also thank Wenming Zhang [16] M. Igarashi, H. Nakae, T. Matsuoka, S. Takahashi, T. Hisada, J.
for helping to collect the gastric juice samples. The authors also thank di- Tomita, Y. Koga, BMJ Open Gastroenterol. 2017, 4, e000144.
rector of Wenqi Tao (Shanghai Jiao Tong University Affiliated Sixth People’s [17] V. A. Poxon, D. L. Morris, D. J. Youngs, E. C. Albutt, M. R. Keighley,
Hospital) for helping pathological section analysis. World J. Surg. 1986, 10, 981.
[18] J. J. Y. Sung, O. O. Coker, E. Chu, C. H. Szeto, S. T. Y. Luk, H. C. H.
Lau, J. Yu, Gut 2020, 69, 1572.
Conflict of Interest [19] P. Correa, E. T. Fontham, J. C. Bravo, L. E. Bravo, B. Ruiz, G. Zarama, J.
L. Realpe, G. T. Malcom, D. Li, W. D. Johnson, R. Mera, J. Natl. Cancer
The authors declare no conflict of interest. Inst. 2000, 92, 1881.
[20] S. Asonuma, A. Imatani, N. Asano, T. Oikawa, H. Konishi, K. Iijima,
T. Koike, S. Ohara, T. Shimosegawa, Am. J. Physiol. Gastrointest. Liver
Author Contributions Physiol. 2009, 297, G312.
W.J. conceptualized the study and designed the research. W.J. and A.Z. [21] J. Houghton, T. C. Wang, Gastroenterology 2005, 128, 1567.
organized all the in vivo and in vitro studies and critical discussions of [22] S. R. Sinha, Y. Haileselassie, L. P. Nguyen, C. Tropini, M. Wang, L.
the results. S.W. performed the experiments and the overall analysis. S.W. S. Becker, D. Sim, K. Jarr, E. T. Spear, G. Singh, H. Namkoong, K.
and W.C. performed the cell experiments. A.Z., S.W., J.K., K.G., and J.Z. Bittinger, M. A. Fischbach, J. L. Sonnenburg, A. Habtezion, Cell Host
measured the BAs of all human samples. S.W., J.K., H.Z., X.Z., J.W., F.H., Microbe 2020, 27, 659.
K.G., M.L., and M.Z. contributed to the animal experiments. S.W. and A.Z. [23] H. Cao, M. Xu, W. Dong, B. Deng, S. Wang, Y. Zhang, S. Wang, S. Luo,
drafted the manuscript and produced the figures. W.J., A.Z., C.R., J.K., X.Z., W. Wang, Y. Qi, J. Gao, X. Cao, F. Yan, B. Wang, Int. J. Cancer 2017, 140,
J.W., F.H., M.L., and M.Z. critically revised the manuscript. 2545.

Adv. Sci. 2022, 9, 2200263 2200263 (14 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

[24] S. L. Chen, J. Z. Mo, Z. J. Cao, X. Y. Chen, S. D. Xiao, World J. Gas- [43] C. S. Eun, B. K. Kim, D. S. Han, S. Y. Kim, K. M. Kim, B. Y. Choi, K. S.
troenterol. 2005, 11, 2834. Song, Y. S. Kim, J. F. Kim, Helicobacter 2014, 19, 407.
[25] P. Dewar, M. F. Dixon, D. Johnston, J. Surg. Res. 1984, 37, 277. [44] R. M. Ferreira, J. Pereira-Marques, I. Pinto-Ribeiro, J. L. Costa, F.
[26] J. Bromberg, J. Clin. Invest. 2002, 109, 1139. Carneiro, J. C. Machado, C. Figueiredo, Gut 2018, 67, 226.
[27] M. Ernst, M. Najdovska, D. Grail, T. Lundgren-May, M. Buchert, H. [45] X. Liu, L. Shao, X. Liu, F. Ji, Y. Mei, Y. Cheng, F. Liu, C. Yan, L. Li, Z.
Tye, V. B. Matthews, J. Armes, P. S. Bhathal, N. R. Hughes, E. G. Mar- Ling, EBioMedicine 2019, 40, 336.
cusson, J. G. Karras, S. Na, J. D. Sedgwick, P. J. Hertzog, B. J. Jenkins, [46] G. Svegliati-Baroni, F. Ridolfi, R. Hannivoort, S. Saccomanno, M.
J. Clin. Invest. 2008, 118, 1727. Homan, S. De Minicis, P. L. Jansen, C. Candelaresi, A. Benedetti, H.
[28] D. S. Shin, H. N. Kim, K. D. Shin, Y. J. Yoon, S. J. Kim, D. C. Han, B. Moshage, Gastroenterology 2005, 128, 1042.
M. Kwon, Cancer Res. 2009, 69, 193. [47] K. Cheng, J. P. Raufman, Biochem. Pharmacol. 2005, 70, 1035.
[29] T. Matsuhisa, T. Arakawa, T. Watanabe, T. Tokutomi, K. Sakurai, S. [48] Z. Wang, X. Gao, R. Zeng, Q. Wu, H. Sun, W. Wu, X. Zhang, G. Sun,
Okamura, S. Chono, T. Kamada, A. Sugiyama, Y. Fujimura, K. Mat- B. Yan, L. Wu, R. Ren, M. Guo, L. Peng, Y. Yang, Front. Microbiol. 2020,
suzawa, M. Ito, M. Yasuda, H. Ota, K. Haruma, Dig. Endosc. 2013, 11, 997.
25, 519. [49] S. K. Kwon, J. C. Park, K. H. Kim, J. Yoon, Y. Cho, B. Lee, J. J. Lee, H.
[30] K. Miwa, H. Hasegawa, T. Fujimura, H. Matsumoto, R. Miyata, T. Jeong, Y. Oh, S. H. Kim, S. D. Lee, B. R. Hwang, Y. Chung, J. F. Kim,
Kosaka, I. Miyazaki, T. Hattori, Carcinogenesis 1992, 13, 2313. K. T. Nam, Y. C. Lee, Gut. 2021, https://doi.org/10.1136/gutjnl-2021-
[31] D. Li, J. Zhang, W. Z. Yao, D. L. Zhang, C. C. Feng, Q. He, H. H. Lv, 324489.
Y. P. Cao, J. Wang, Y. Qi, S. R. Wu, N. Wang, J. Zhao, Y. Q. Shi, J. Dig. [50] K. H. Chen, K. Mukaisho, H. Sugihara, Y. Araki, G. Yamamoto, T. Hat-
Dis. 2020, 21, 222. tori, Cancer Sci. 2007, 98, 1683.
[32] T. Matsuhisa, T. Tsukui, J. Clin. Biochem. Nutr. 2012, 50, 217. [51] T. Tatsuta, K. Mukaisho, H. Sugihara, K. Miwa, T. Tani, T. Hattori, Dig.
[33] P. M. Newberne, G. Charnley, K. Adams, M. Cantor, D. Roth, V. Dis. Sci. 2005, 50, 425.
Supharkarn, L. Fong, Food Chem. Toxicol. 1986, 24, 1111. [52] M. A. Nadkarni, G. V. Browne, K. L. Chhour, R. Byun, K. A. Nguyen,
[34] Y. Yuasa, Nat. Rev. Cancer 2003, 3, 592. C. C. Chapple, N. A. Jacques, N. Hunter, Eur. J. Clin. Microbiol. Infect.
[35] Z. Xiang, J. M. Si, H. D. Huang, World J. Gastroenterol. 2004, 10, 3212. Dis. 2012, 31, 2989.
[36] W. Cao, W. Tian, J. Hong, D. Li, R. Tavares, L. Noble, S. F. Moss, M. [53] F. Huang, X. Zheng, X. Ma, R. Jiang, W. Zhou, S. Zhou, Y. Zhang,
B. Resnick, Am. J. Physiol. Gastrointest. Liver Physiol. 2013, 304, G322. S. Lei, S. Wang, J. Kuang, X. Han, M. Wei, Y. You, M. Li, Y. Li,
[37] Y. Shi, Y. Wei, T. Zhang, J. Zhang, Y. Wang, S. Ding, Gastroenterol. Res. D. Liang, J. Liu, T. Chen, C. Yan, R. Wei, C. Rajani, C. Shen, G.
Pract. 2016, 2016, 9638963. Xie, Z. Bian, H. Li, A. Zhao, W. Jia, Nat. Commun. 2019, 10,
[38] Y. C. Wu, C. F. Chiu, C. T. Hsueh, C. T. Hsueh, Cancer Cell Int. 2018, 4971.
18, 75. [54] G. Xie, R. Jiang, X. Wang, P. Liu, A. Zhao, Y. Wu, F. Huang, Z. Liu, C.
[39] J. H. Yu, J. B. Zheng, J. Qi, K. Yang, Y. H. Wu, K. Wang, C. B. Wang, X. Rajani, X. Zheng, J. Qiu, X. Zhang, S. Zhao, H. Bian, X. Gao, B. Sun,
J. Sun, Int. J. Oncol. 2019, 54, 879. W. Jia, EBioMedicine 2021, 66, 103290.
[40] X. Wang, P. Zhou, X. Sun, J. Zheng, G. Wei, L. Zhang, H. Wang, J. Yao, [55] W. Jia, M. Wei, C. Rajani, X. Zheng, Protein Cell 2021, 12, 411.
S. Lu, P. Jia, Oncol. Rep. 2015, 33, 3038. [56] E. Bolyen, J. R. Rideout, M. R. Dillon, N. A. Bokulich, C. C. Abnet, G.
[41] M. Rajilic-Stojanovic, C. Figueiredo, A. Smet, R. Hansen, J. Kupcin- A. Al-Ghalith, H. Alexander, E. J. Alm, M. Arumugam, F. Asnicar, Y.
skas, T. Rokkas, L. Andersen, J. C. Machado, G. Ianiro, A. Gasbarrini, Bai, J. E. Bisanz, K. Bittinger, A. Brejnrod, C. J. Brislawn, C. T. Brown,
M. Leja, J. P. Gisbert, G. L. Hold, Aliment. Pharmacol. Ther. 2020, 51, B. J. Callahan, A. M. Caraballo-Rodríguez, J. Chase, E. K. Cope, R. Da
582. Silva, C. Diener, P. C. Dorrestein, G. M. Douglas, D. M. Durall, C.
[42] X. H. Chen, A. Wang, A. N. Chu, Y. H. Gong, Y. Yuan, Front. Microbiol. Duvallet, C. F. Edwardson, M. Ernst, M. Estaki, J. Fouquier, et al., Nat.
2019, 10, 1261. Biotechnol. 2019, 37, 852.

Adv. Sci. 2022, 9, 2200263 2200263 (15 of 15) © 2022 The Authors. Advanced Science published by Wiley-VCH GmbH

You might also like