You are on page 1of 13

Life Sciences 248 (2020) 117456

Contents lists available at ScienceDirect

Life Sciences
journal homepage: www.elsevier.com/locate/lifescie

Mechanism of berberine in treating Helicobacter pylori induced chronic T


atrophic gastritis through IRF8-IFN-γ signaling axis suppressing
Tao Yanga, Ruilin Wangb, Jianzhong Zhangc, Chunmei Baod, Juling Zhangd, Ruisheng Lie,
Xing Chenf, Shihua Wuf, Jianxia Wenf, Shizhang Weig, Haotian Lig, Huadan Caig,
⁎ ⁎⁎
Xiangdong Yangh, , Yanling Zhaog,
a
College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, No. 37, 12 Bridge Road, Chengdu 610075, PR China
b
Integrative Medical Center, The Fifth Medical Center of PLA General Hospital, Beijing 100039, PR China
c
Center of Disease Control and Prevention, National Institute for Communicable Disease Control and Prevention, Beijing 100039, PR China
d
Division of Clinical Microbiology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, PR China
e
Research Center for Clinical and Translational Medicine, The Fifth Medical Center of PLA General Hospital, Beijing 100039, PR China
f
College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
g
Department of Pharmacy, The Fifth Medical Center of PLA General Hospital, Beijing 100039, PR China
h
Colorectal and Anal Surgery, Chengdu Anorectal Hospital, No 152 Daqiang East Street, Taisheng South Road, Chengdu 610075, PR China

A R T I C LE I N FO A B S T R A C T

Keywords: Aims: In this study, we will investigate the therapeutic effects of berberine (BBR) in Helicobacter pylori (H. pylori)
Chronic atrophic gastritis induced chronic atrophic gastritis (CAG). Furthermore, potential mechanisms of BBR in regulating IRF8-IFN-γ
Helicobacter pylori signaling axis will also be investigated.
Berberine Materials and methods: H. pylori were utilized to establish CAG model of rats. Therapeutic effects of BBR on
IRF8
serum supernatant indices, and histopathology of stomach were analyzed in vivo. Moreover, GES-1 cells were
IFN-γ
infected by H. pylori, and intervened with BBR in vitro. Cell viability, morphology, proliferation, and quanti-
tative analysis were detected by high-content screening (HCS) imaging assay. To further investigate the potential
mechanisms of BBR, relative mRNA, immunohistochemistry and protein expression in IRF8-IFN-γ signaling axis
were measured.
Key findings: Results showed serum supernatant indices including IL-17, CXCL1, and CXCL9 were downregulated
by BBR intervention, while, G-17 increased significantly. Histological injuries of gastric mucosa induced by H.
pylori also were alleviated. Moreover, cell viability and morphology changes of GES-1 cells were improved by
BBR intervention. In addition, proinflammatory genes and IRF8-IFN-γ signaling axis related genes, including
Ifit3, Upp1, USP18, Nlrc5, were suppressed by BBR administration in vitro and in vivo. The proteins expression
related to IRF8-IFN-γ signaling axis, including Ifit3, IRF1 and Ifit1 were downregulated by BBR intervention.

Significance system. Abdominal pain, bloating, abdominal discomfort, loss of ap-


petite and secondary anemia, weight loss are classical symptoms of
Berberine possesses therapeutic effects on CAG induced by H. pylori CAG. The pathological features of CAG are characterized by chronic
via chronic inflammation inhibition. The anti-inflammatory properties inflammatory processes of gastric mucosa, altered mucosal atrophy and
of BBR tightly related to suppression of IRF8-IFN-γ signaling axis, which intestinal metaplasia (especially incomplete colonization) [1]. CAG
is crucial in Th1 cell-dominant inflammatory response. have served as the main stages of precancerous lesion of gastric cancer
(PLGC) and may evolve into irreversible tumorigenesis [2]. Up to now,
1. Introduction the mechanisms of CAG have not been fully investigated. However,
there exist a lot of evidence declare Helicobacter pylori (H. pylori) is the
Chronic atrophic gastritis (CAG) is a prevailing disease in digestive leading risk factor of CAG [3–6]. H. pylori is a kind of Gram-negative


Corresponding author.
⁎⁎
Correspondence to: Y. Zhao, Department of Pharmacy, The Fifth Medical Center of PLA General Hospital, No 100 West Fourth Ring Middle Road, Beijing 100039,
PR China.
E-mail addresses: y-xd@vip.163.com (X. Yang), zhaoyl2855@126.com (Y. Zhao).

https://doi.org/10.1016/j.lfs.2020.117456
Received 11 January 2020; Received in revised form 15 February 2020; Accepted 19 February 2020
Available online 22 February 2020
0024-3205/ © 2020 Published by Elsevier Inc.
T. Yang, et al. Life Sciences 248 (2020) 117456

anaerobic bacterium and has been defined as I class of carcinogenic 2. Materials and methods
factors [7]. More than 50% of the world's population is infected with H.
pylori [8]. The urease apoenzyme encoded by ureA and ureB of H. pylori 2.1. Ethics statement
is capable of employing acid acclimation, combating gastric acidity,
allowing gastric colonization [9,10]. Colonization of H. pylori in hu- This study was conducted strictly in accordance with the Guidelines
mans stomach mucosa could induce various inflammatory processes, for the Care and Use of Laboratory Animals of the Ministry of Science
including innate and adaptive mucosal inflammation [11,12]. H. pylori and Technology of China. All breeding and experiments were under-
infection in stomach mucosa has been recognized as the primary cause taken with review and approval from the Animal Ethical and
of many stomach diseases, including chronic gastritis, peptic ulcers, Experimental Committee of the Fifth Medical Center of PLA General
lymphoma of mucosa-associated lymphoid tissue (MALT lymphoma) Hospital (Approval ID: IACUC-2018-010).
and gastric adenocarcinoma [13].
It has been reported that Interferon regulatory factor 8 (IRF8) in 2.2. Berberine preparation
stomach tissues when H. pylori infection became increased expression,
and Interferon-gamma (IFN-γ) acted as major regulator of IRF8 in H. Standards of BBR (purity ≥98%, Cat. No. CHB181028) was pur-
pylori induced inflammatory process [14]. IRF8 is a transcription factors chased from Chroma Biotechnology Co. Ltd. (Chengdu, China). BBR
that is induced by interferons (IFNs) in a wide range of cell types [15]. was dissolved in dimethyl sulfoxide (DMSO) and diluted to corre-
IRF8 plays essential roles in cellular differentiation and function sponding concentration when applied for human gastric epithelial cell
[16,17]. Developing lymphoid and myeloid cells express stable IRF8. (GES-1). Then, BBR (purity ≥95%, Cat. No. CHB180606) was dissolved
When binding to a partner molecule or specific DNA sequences, all in sodium chloride injection (NS 0.9%) and diluted to corresponding
activities of IRF8 are activated and asserts its transcriptional regulatory concentration when applied to SD rats.
activity strongly [18]. Previous studies have illustrated interferon (IFN)
is one of the strongest inducers of IRF8 transcription. IFN-c-activated 2.3. Rats and infection
factor (GAF), is the resultant dimer of IFN, could bind to the IFN-c
activated site (GAS) and then upregulate IRF8 [19,20]. IFN-γ is a kind A model using H. pylori-infected rats with chronic gastritis was
of T helper cell 1 (Th1) cytokines, may promote the release of proin- previously described [31]. Specific pathogen free (SPF) male Sprague-
flammatory cytokines, augmenting inflammation and apoptosis in- Dawley rats (180 ± 20 g) were purchased from Beijing Sibeifu Animal
duced by H. pylori [21]. IFN-γ plays double-dimension adjustment role Breeding Center (Permission No. SCXK-(jing) 2016–0002). All rats were
in the induction of suppressive Th1-like Treg cells [22]. During mi- negative for pathogenic bacteria including Helicobacter spp and para-
croorganism infection, IFN-γ promotes T-box transcription factor (T- sites. Rats were maintained under SPF conditions (temperature:
bet) expressing Treg cells, which restrain type I responses [23]. In 25 ± 0.5 °C, humidity: 55 ± 5%, 12 h:12 h light-dark cycle) in a
contrast, continuous exposure of IFN-γ can destabilize suppressive Th1- shelter sustained facility and provided with sterile food and water. In-
like Treg cells [24]. IRF8 is highly expressed by Treg cells and mainly fection of rats with the H. pylori strain (isolated from the stomach tissue
controls type I immune responses [25]. During this immune response of a male CAG patient (No. ZCDC111001), kindly provided by Prof.
process, IRF8 get increased in a way similar to T-bet and chemokine (C- Jianzhong Zhang, National Institute for Communicable Disease Control
X-C motif) receptor 3 (CXCR3) [25]. On the contrary, decreased IRF8 and Prevention, Beijing, China). H. pylori were grown in Campylobacter
level will cause aberrantly high expression of T helper cell 2 (Th2) [IL- agar base (Oxoid, UK) containing 1% H. pylori selective supplement
4, IL-5, IL-13, et al] and T helper cell 17 (Th17) [IL-21, et al] cytokines (Oxoid, UK) and 3% fetal bovine serum, cultured at 37 °C under mi-
in Treg cells, which leads to Treg cells losing suppressive function [26]. croaerophilic conditions (10% CO2, 5% O2, 85% N2). After culture for
Thus, there may exist feedback interaction between IRF8 and IFN-γ 4 days, live bacteria were collected and adjusted to 108 colony forming
when infection happens. Higher expression of IRF8 or IFN-γ may fa- units (CFU)/mL. Briefly, rats were inoculated with an oral dose of
cilitate chronic infection. 1.5 × 108 CFU/ml H. pylori at day 1, 3, 5 and 7. At 8 weeks following
H. pylori eradication could benefit millions of people all over the infection, stomach tissues were processed for rapid urease test, his-
world and inhibit the development of CAG and PLGC. However, due to tology and tissue culture for enumeration of H. pylori. Infection status
issues of soaring antibiotic resistance and changing epidemiology of H. and H. pylori-induced chronic atrophic gastritis in rats were confirmed.
pylori infection, treatment failure and high rates of recrudescence are After infection verified, rats were randomly assigned to three different
universal. New approaches and strategies are needed for effective groups, including H. pylori infection group, BBR with low-dose (14 mg/
management. Fortunately, Traditional Chinese medicine (TCM) may kg/d) and high-dose group (28 mg/kg/d), respectively.
contribute to H. pylori eradication and successful treatment of CAG.
Berberine (BBR), a natural isoquinoline alkaloid from various tradi- 2.4. Cells infection and cell viability evaluation
tional Chinese medicine herbs, such as Rhizoma coptidis, Cortex phello-
dendri, has been widely applied in clinical treatment for many cen- Human gastric epithelial cells (GES-1) line were purchased from
turies. Over the past decades, many studies have revealed BBR and its FuHeng Cell Center (Shanghai, China). The adherence of H. pylori
derivatives possess high activity against both inflammation and cancer (multiplicity of infection [MOI] of 10:1, 20:1, 50:1 and 100:1) to GES-1
in the digestive system [27,28]. Meanwhile, there are increasing reports for 24 h. The relative number of H. pylori was examined using a Synergy
elucidated BBR holds great potential to treat chronic gastritis induced H1 Hybrid Reader (Biotech, USA). The optical density (OD) at 600 nm
by H. pylori infection in stomach mucosa [29,30]. Nevertheless, po- was set to count colony forming units of H. pylori (1
tential mechanisms by which BBR exerts inhibitory effect on H. pylori OD600nm = 1.5 × 108 colony forming units/ml). The viability of GES-1
infection is not yet completely clarified. The potential mechanisms of after infection and processed by BBR, 10% (vol/vol) cell counting kit-8
BBR in IRF8-IFN-γ signaling axis was shown in Fig. 1. (CCK-8; Lot. PG658, Dojindo, Tokyo, Japan) was added into cells and
In this study, we will investigate the therapeutic effects of BBR in H. incubated for 15 min at 37 °C. Absorbance was measured at 450 nm.
pylori induced CAG. Furthermore, we also explore potential mechan- Cell viability was calculated as cell viability (%) = 100 × (OD treat-
isms of BBR in regulating IRF8-IFN-γ signaling axis, contributing to ment/OD control).
better understanding the protection roles of BBR mechanisms.
2.5. Morphological identification and quantitative statistics

Morphological identification and quantitative statistics of GES-1

2
T. Yang, et al. Life Sciences 248 (2020) 117456

Fig. 1. Potential mechanisms of BBR in


IRF8-IFN-γ signaling axis. means sti-
mulation, means inhibition.

cells were examined by a High-Content System array scan (Thermo anti-IRF8 Ab (Cat No.: 18977-1-AP, Proteintech, 1:100) and anti-IFN-γ
Scientific, Massachusetts, USA), which was utilized by Wen, et al. [32] Ab (Cat No.: NBP2-66900, Novusbio, 1:100) were utilized. Light mi-
before. Fluorescent dyes, including Hoechst 33342 (H3570, Invitrogen), croscopy (Olympus, Japan) at 200×, 400× magnification was applied
aiming to mark nuclear area, Calcein AM (C3099, Invitrogen) to spot to photograph images.
cytoplasm, and ethidium homodimer-1 (EthD-1) (L3224, Invitrogen) to
detect nucleic acids, were employed to identify morphology of GES-1 2.9. Real-time quantitative PCR for mRNA expressions in stomach tissues
cells. The cell health profiling assay module was selected in the HCS
system, the parameters and forma setting were reported previously by Microarray analysis was done using RNA extracts from frozen sto-
O'Brien, et al. [33] and wave length in different channels were set to mach tissues of infected and control rats or cells. TRIzol reagent (Nordic
collect fluorescent images. Configure acquisition parameters, including Bioscience, Beijing, China) were applied and converted into cDNA by
objective (10×), camera configuration (×1) and acquisition camera reverse transcription kit (Promega, Madison, USA) according to man-
mode (1104 × 1104, 2 × 2 binning), were employed. Ultimately, the ufacturer's protocol. The primers sequences of IRF8, IFN-γ, Ifit3, Upp-1,
Array Scan XTI system was utilized to quantify the mean fluorescence USP-18, Nlrc5, NF-κB and TNF-α were listed in Table 1. Quantitative
intensity of GES-1 cells by software algorithm. Real-time PCR for these mRNA was performed and analyzed using
cDNA and SYBR Green PCR Master Mix (Nordic Bioscience, Beijing,
2.6. Histopathological examination China). RT-PCR was performed on a 7500 fast real-time PCR system
(Applied Biosystems, Foster City, CA, USA). 7500 software (Applied
Stomach tissue samples were fixed and preserved in 10% neutral Biosystems for 7500 and 7500 Fast Real-Time PCR Products, version
buffered formalin solution, cleared in xylene, embedded in paraffin, 2.0.5) was used to present results and export data. The relative amounts
and cut into 5 μm thick slices by using a microtome. Stomach tissue of mRNA were determined based on 2−ΔΔCt calculations with β-actin as
sections were stained with hematoxylin-eosin (HE). Histopathological a controlling reference.
determination of the stomach mucosa injury was conducted by photo-
graphy with light microscopy and 200×, 400× magnification was used 2.10. Western blotting analysis for protein expression
in the microscopy analysis.
Stomach tissues were homogenized with ice-cold RIPA lysis buffer
2.7. Measurement of specific serum biomarkers (tissues to buffer ratio, 1: 10, w/v) supplemented with a protease in-
hibitor cocktail. The homogenates were centrifuged at 4 °C for 10 min
The Synergy H1 Hybrid Reader (Biotech, USA) was employed to at 10,000g, and the supernatants were stored at −20 °C. Bicinchoninic
measure serum biochemical indices, including gastrin-17 (G-17), in- acid (BCA) protein assay kit (Solarbio, Beijing, China) was used to
terleukin-17 (IL-17), chemokines 1 (CXCL1), chemokines 9 (CXCL9). measure the protein concentration. Proteins (50 mg) were boiled with
The enzyme-linked immunosorbent assay (ELISA) kits of G-17 and IL-17 SDS sample buffer, separated by SDS-PAGE (25 min, 80 V; 50 min,
were purchased from MLBIO biotechnology Co., Ltd. (Shanghai, China). 120 V), and then electrophoretic ally transferred onto PVDF membranes
CXCL1 and CXCL9 kits were purchased from JYMBIO biotechnology (Millipore, MA, USA; 200 mA, 2 h). Tris-buffered saline (TBS) with 5%
Co., Ltd. (Wuhan, China). All assays were performed rigorously ac- non-fat dry milk (NFDM) was applied to seal PVDF membranes. Next,
cording to the manufacturer's instructions. these membranes were incubated overnight at 4 °C with rabbit anti-
IRF8 Ab (Proteintech, Cat No.: 18977-1-AP, dilution: 1:1000), rabbit
2.8. Immunohistochemistry (IHC) anti-IFN gamma Ab (Novusbio, Cat No.: NBP2-66900, dilution: 1:500),
rabbit anti-Ifit3 Ab (BIOSS, Cat No.: bs-15515R, dilution: 1:500), rabbit
Paraffin sections of stomach tissues were stained with a polyclonal anti-Ifit1 Ab (Proteintech, Cat No.: 23247-1-AP, dilution: 1:500), rabbit

3
T. Yang, et al. Life Sciences 248 (2020) 117456

Table 1
Primers sequences of real-time PCR analyses for mRNA expression.
Genes Forward Reverse

SD rats
IRF8 AGAGCCGCCTGCCTGATAGC TGCCCGCTTCCTCCACCATC
IFN-γ ACAACCCACAGATCCAGCACAAAG GCTTCCTTAGGCTAGATTCTGGTGAC
Ifit3 ACAGCTTGGTCATGTGCCGTTAC TCTTCTCTCACTCTTGTCAGCCTCTC
Upp1 ATCATCCGCATTGGCACTTCAGG GTTTCGAGTCACCCGCTTTCCC
USP18 TGCTGCTCATCTCTTCCTTACAATCTG TCCTGCTGCTCTCTGCTCCAC
Nlrc5 CACCTTGCCACCAACCTCACG GCCAACTCAGCAGCCATCTCATC
β-Actin CACTATCGGCAATGAGCGGTTCC ACTGTGTTGGCATAGAGGTCTTTACG

GES-1
IRF8 CAGCCACTTGGAAGACGAGGTTAC ACTTCATTCACGCAGCCAGCAG
IFN-γ AGTGATGGCTGAACTGTCGC ACTGGGATGCTCTTCGACCT
USP18 CCACCTCATGCGATTCTCCATCAG GCTCCTCAGCATCACAAGACTCTC
Upp1 CGGTGCTGTCTGTCAGTCATGG ACCAGAAGTGCCAATGCGGATG
NF-κB GACATGGTGGTCGGCTTCGC CGCCTCTGTCATTCGTGCTTCC
TNF-α CCAATGGCGTGGAGCTGAGAG TCTGGTAGGAGACGGCGATGC
β-Actin AGGAAGGACCTGTATGCCAACA GCGCGGTGATCTCTTTCTG

Fig. 2. Disease performance of chronic atrophic gastritis in rats. (A) Weight of rats (n = 6), data were shown as mean ± SD, ##P < .01 versus control group; (B)
Morphology of rats stomach; (C) Rapid urease test of stomach tissues. Ctrl, control group.

Fig. 3. Effect of BBR on cell viability of


GES-1 cells. (A) Cell viability of GES-1 cells
in different concentrations of BBR (10
μΜ–100 μΜ); (B) Cell viability of GES-1
cells in different MOI of H. pylori
(10:1–100:1); (C) Cell viability of GES-1
cells in the intervention of BBR. ##P < .01
versus control group, ⁎⁎P < .01 versus H.
pylori group. Results were expressed as
percentages of the control group. Data were
shown as mean ± SD. BBR, berberine; Ctrl,
control group; MOI, multiplicity of infec-
tion.

4
T. Yang, et al. Life Sciences 248 (2020) 117456

(caption on next page)

5
T. Yang, et al. Life Sciences 248 (2020) 117456

Fig. 4. Morphological identification and quantitative analysis of HCS imaging assay for GES-1 cells. Intensity of fluorescence staining reflected the survival cells
(green fluorescence) and dead cells (red fluorescence), Scale bar = 50 μm. (A) Valid cell counts of HCS analysis for GES-1 cells (% of control). (B) Survival cell counts
of GES-1 cells (MEAN_TargetAvgIntenCh2). (C) Dead cell counts of GES-1 cells (MEAN_TargetAvgIntenCh3). N·S, none-significant, ##P < .01 versus control group.

P < .05 versus H. pylori infection group. ⁎⁎P < .01 versus H. pylori infection group. The results are expressed as percentages of control group. Data were shown as
mean ± SD. Ctrl, control group; BBR, berberine. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this
article.)

Fig. 5. Expression of proinflammatory genes in GES-1 with H. pylori infection. All data are presented as mean ± SD. ∗∗P < .01 versus control group; Ctrl, control
group; IRF8, interferon regulatory factor 8; IFN-γ, interferon-gamma; NF-κB, nuclear factor-κ-gene binding; TNF-α, tumor necrosis factor-α.

anti-IRF1 Ab (BIOSS, Cat No.: bsm-52114R, dilution: 1:1000) and anti- 3. Results
β-actin monoclonal antibody (BIOSS, Cat No.: bs-0061R, dilution:
1:3000), respectively. Then, membranes were washed five times for 3.1. Disease performance of chronic atrophic gastritis in H. pylori-induced
5 min each with TBS-0.1% Tween 20 (TBST) and incubated with HRP- rats
conjugated secondary antibody (goat anti-mouse IgG (H + L)) (ZSGB-
BIO, Cat No.: ZB-2301, dilution, 1:25000) for 1 h at room temperature Eight weeks after H. pylori infection, the rats infected with H. pylori
on a shaker. Western blotting detection system (Quantity One, Bio-Rad exhibited CAG symptoms, such as loss of appetite, diarrhea and weight
Laboratories, USA) was applied to obtain images. Quantification of loss (Fig. 2A). The surface of the gastric mucosa was rough and pale, the
bands was performed by densitometric analysis through Bio-Rad stomach wrinkles are disordered microvascular exposure under the
Quantity One. The β-actin was set as a loading control. mucosa, many bleeding points were seen directly by visual inspection
(Fig. 2B). The rapid urease test of stomach tissues (from body of sto-
mach and pyloric antrum) was positive (Fig. 2C). Those changes are
2.11. Statistics analysis similar to those populations who have CAG caused by H. pylori in clinic.

All data were presented as mean ± standard deviation (SD) and


analyzed with the SPSS software program (version 17.0; SPSS Inc., 3.2. Cell viability
Chicago, IL, USA). Data were presented using one-way analysis of
variance (ANOVA) followed by Bonferroni method. P < .05 was CCK-8 kit was utilized to determine cell viability of GES-1. The
considered statistically significant and P < .01 was highly significant. optimum concentrations of BBR for protecting GES-1 cells against H.
GraphPad Prism software for Windows (version 6.02; Inc., San Diego, pylori-induced injury were explored firstly. The concentrations of BBR
USA) were utilized for visible presentation of all results. were set as 0, 10, 20, 30, 40, 50 and 100 μM. The results showed that
BBR with the concentrations of 100 μM could significantly inhibit the
cell viability (52.31 ± 5.74) compared with the control groups
(100 ± 17.93) (P < .01). Meanwhile, cell viability of GES-1 with
50 μM concentrations of BBR was lower than 90% (66.33 ± 4.35).
When given at 40 μM concentrations of BBR, cell viability of GES-1 was
higher than 90% (94.91 ± 7.92). Accordingly, 40 μM of BBR showed a

6
T. Yang, et al. Life Sciences 248 (2020) 117456

Fig. 6. Effects of BBR on serum supernatant indices of CAG rats (n = 6). Data were shown as mean ± SD. ##P < .01 versus control group; ∗∗P < .01 versus H.
pylori group; Ctrl, control group; L, low-dose group; H, high-dose group. G-17, gastrin-17; IL-17, interleukin-17; CXCL1, chemokines 1; CXCL9, chemokines 9.

Fig. 7. Effects of BBR on stomach histological changes at low dose and high dose. Representative photomicrographs and summary data for HE staining of stomach
histological changes were represented. Red arrows indicated neutrophil infiltration. Ctrl, control group; L, low-dose group; H, high-dose group. (For interpretation of
the references to colour in this figure legend, the reader is referred to the web version of this article.)

7
T. Yang, et al. Life Sciences 248 (2020) 117456

Fig. 8. Effects of BBR on the IRF8-IFN-γ signaling axis mRNA expressions in vitro with H. pylori infection. All data are presented as mean ± SD. ##P < .01 versus
control group; ∗P < .05 versus H. pylori infection group; ∗∗P < .01 versus H. pylori infection group. Ctrl, control group; Combination, AX-024 + BBR 40 μM; IRF8,
interferon regulatory factor 8; IFN-γ, interferon-gamma; USP18, ubiquitin specific peptidase 18; Upp1, uridine phosphorylase 1.

relatively protective effect and was used to investigate the protective nuclear area and cytoplasm. The intensity of green fluorescence (sur-
effect of BBR on GES-1 injury induced by H. pylori (Fig. 3A). Then, the vival cell staining) was significantly increased and the red fluorescence
adherence of H. pylori (multiplicity of infection [MOI] of 10:1, 20:1, (dead cell staining) was significantly decreased. In terms of quantitative
50:1 and 100:1) to GES-1 co-cultivated for 24 h. There were lot of dead statistics, GES-1 cell counts were significantly decreased after infected
GES-1 cells through observation of naked eyes directly when infected with H. pylori for 24 h. Compared with H. pylori infection group, the
with MOI 100:1, and the cell viability (39.81 ± 8.06) was decreased reduction rate of cell counts in BBR group declined remarkably. Results
significantly compared with the control groups (100 ± 12.91). Cell were shown in Fig. 4 (A–C). All data suggested that BBR may sig-
viability of GES-1 with MOI 50:1 was about (50.03 ± 7.95). Conse- nificantly keep GES-1 from H. pylori infection and reduce injury.
quently, MOI 50:1 was chosen in this study for subsequent experiment
(Fig. 3B). Next, BBR with the concentrations of 40 μM and 20 μM with 3.4. Expression of proinflammatory genes in GES-1 with H. pylori infection
MOI 50:1 were cultivated together for 24 h. Results demonstrated cell
viability of GES-1 was increased in 40 μM (83.44 ± 7.46) and 20 μM An ocean of studies reported expression of proinflammatory genes
BBR group (75.47 ± 9.31) compared with the H. pylori infection group increased when H. pylori infection. In this paper, expression of proin-
(Fig. 3C). Overall, all results revealed BBR was capable of protecting flammatory genes were probed in GES-1 when H. pylori infection. The
GES-1 from H. pylori infection. expression of proinflammatory genes encoding IRF-8, IFN-γ, NF-κB and
TNF-α were tested with MOI of 10:1, 20:1, 50:1 and 100:1. As described
3.3. Morphological identification and quantitative statistics of GES-1 cells previously, there was multitude of floating GES-1 cells when infected
with MOI 100:1 for 24 h. As a result, proinflammatory genes expression
In order to identify the effects of BBR on cell morphological influ- dramatically upregulated with MOI of 10:1, 20:1, 50:1 and 100:1 for
ence, high-content survival cell imaging assays were utilized to quali- 24 h compared with H. pylori uninfected group. Obviously, the highest-
tatively and quantitatively assay cell counts, morphology and cell via- level expression of proinflammatory genes (almost higher than 2-fold
bility. As displayed in Fig. 4, GES-1 cells in the control group presented increasing) when infected with MOI 50:1 in GES-1 cells. Accordingly,
Hoechst and Calcein, AM fluorescence in the nuclear area and cyto- MOI 50:1 was selected in this study for following exploration. Results
plasm homogeneously. However, morphological changes of GES-1 cells were shown in Fig. 5.
in H. pylori infection group were remarkable, the nucleus tinted by
Hoechst 33342 were featured with malformations, deeper staining and 3.5. Detection of biomarkers in serum
coarse-grained. The cytoplasm stained by Calcein, AM was character-
ized with bright spots (green fluorescence) and fractured cytoskeleton. To clarify the activities of several specific markers of H. pylori-in-
Dead cells stained by EthD-1 with red fluorescence increased phe- duced CAG rats, the serum supernatant levels of gastrin-17 (G-17), in-
nomenally. On the contrary, intervened by BBR with 20 μM and 40 μM, terleukin-17 (IL-17), CXCL1, and CXCL9 were estimated. As showed in
the morphological changes of GES-1 cells were slight, no matter in Fig. 6, IL-17, CXCL1, and CXCL9 increased markedly in H. pylori-

8
T. Yang, et al. Life Sciences 248 (2020) 117456

Fig. 9. Effects of BBR on the IRF8-IFN-γ signaling axis mRNA expressions in vivo with H. pylori infection (n = 6). All data are presented as mean ± SD. ##P < .01
versus control group; ∗P < .05 versus H. pylori infection group; ∗∗P < .01 versus H. pylori infection group. Ctrl, control group; IRF8, interferon regulatory factor 8;
IFN-γ, interferon-gamma; Ifit3, interferon induced protein with tetratricopeptide repeats 3; Upp1, uridine phosphorylase 1; USP18, ubiquitin specific peptidase 18;
Nlrc5, NLR family CARD domain containing 5.

induced CAG group compared to control group. On the contrary, G-17 3.7. Berberine suppressed IRF8-IFN-γ signaling axis when H. pylori
decreased distinctly in H. pylori infection group compared with non- infection in vitro
infection group. In the administration of BBR group (14 mg/kg and
28 mg/kg), IL-17, CXCL1, and CXCL9 significantly reduced, and the Previous genome-wide analysis of IRF8 target genes by employing
high dosage of compatibility group (28 mg/kg) had lower level of IL-17, ChIP-seq and RNA-seq demonstrated USP18 and Upp1 were relevant
CXCL1, and CXCL9 compared with group with low dosage and H. pylori downstream genes [14]. The role of BBR in IRF8-IFN-γ signaling axis in
infection group. Conversely, the high dosage of compatibility group GES-1 infected by H. pylori (MOI 50:1) was further investigated in this
maintained a higher level of G-17 compared with group with low do- examine. Selective inhibitor of IFN-γ (AX-024, MedChem Express,
sage and H. pylori infection group. Altogether, these results in vivo Shanghai, China, 10 ng/ml) was applied to black the expression of IFN-
imply that the activities of several specific markers, including IL-17, γ. As seen in Fig. 8, the expression of IFN-γ was significantly suppressed
CXCL1, and CXCL9, might get suppressed by BBR intervention. by exposure to AX-024 in the presence of H. pylori. Additionally, we
Nevertheless, G-17 raised significantly following administration of BBR observed that proinflammatory genes including NF-κB and TNF-α were
when H. pylori infection. decreased significantly with AX-024 blocking. Furthermore, IRF8 and
its downstream genes (USP18, Upp1) also were suppressed significantly
by AX-024 inhabitation. Accordingly, these findings suggested that IFN-
3.6. Histological examination of gastric mucosa γ could induce IRF8 expression in GES-1 cells. Intervened by 40 μM BBR
subsequently, the expression of NF-κB, TNF-α, IFN-γ, IRF8, USP18, and
Histological features of gastric mucosa were the direct evidence for Upp1 also decreased significantly compared with H. pylori infected
the therapeutic effect of BBR against CAG infected by H. pylori. In the group. Next, AX-024 plus BBR (combination group) was utilized to treat
current study, hematoxylin-eosin (HE) staining was performed to GES-1 cells with H. pylori infection, data showed all proinflammatory
evaluate morphological features, which were induced by inflammation genes mentioned above were suppressed obviously. Together, these
accumulation in stomach tissues. Rats in the control group showed findings suggested that BBR could suppress IRF8-IFN-γ signaling axis
normal morphology, signed with tightly and orderly gastric glandular and expression of proinflammatory genes, protect GES-1 cells from in-
and parietal cells. In the H. pylori infected group, pathologic changes of fection induced by H. pylori.
rats are characteristic with atrophy and irregular arrangement of gastric
glandular, infiltrated with large amounts of lymphocytes and plasma 3.8. Berberine attenuated IRF8-IFN-γ signaling axis when H. pylori
cells, and loss of parietal cells. Conversely, those changes in BBR groups infection in vivo
were more slightly than in H. pylori infected group in terms of the
gastric glandular and membranes of parietal cells. Administration of We then evaluated the effects of BBR on expression of IRF8 and its
BBR at high dose (28 mg/kg) exhibited lower infiltration of in- downstream genes in the gastric mucosa of H. pylori-infected rats by
flammatory cell and other histological injuries noticeably. Results were qRT-PCR firstly. Results were presented in Fig. 9. Evidently, the ex-
presented in Fig. 7. pression of IFN-γ, IRF8 and its related downstream genes including

9
T. Yang, et al. Life Sciences 248 (2020) 117456

Fig. 10. Effects of BBR on immunohistochemical analysis with low dose and high dose. Representative photomicrographs and summary data for IHC staining of
stomach histological changes were represented. Red arrows indicated positive expression. IHC, immunohistochemistry; Ctrl, control group; L, low-dose group; H,
high-dose group. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

Ifit3, Upp1, USP18, Nlrc5 were increased significantly in H. pylori-in- Subsequently, the protein expressions of IFN-γ, IRF8, Ifit3, IRF1 and
fected rats compared with the normal rats. Administration of BBR at Ifit1 in stomach tissues were estimated by western blotting analysis. As
high dose (28 mg/kg) expressed lower level of IFN-γ, IRF8 and its shown in Fig. 11, IFN-γ, IRF8, Ifit3, IRF1 and Ifit1 expressed at a re-
downstream genes noticeably compared with the H. pylori-infected latively high level in H. pylori-infected group compared with control
group and low dose (14 mg/kg) group. Consequently, those results group. However, BBR given at a high dosage could significantly inhibit
mentioned above revealed that BBR could down-regulate genes ex- the expression of IFN-γ, IRF8 and its associated downstream proteins
pression bound to IRF8-IFN-γ signaling axis and hold anti-inflammatory including Ifit3, IRF1 and Ifit1. Remarkably, the expressions of these
properties in H. pylori-infected gastric tissues. proteins were exceedingly significant differences in low-dose group and
high-dose group. Findings from western blotting analysis provided
3.9. Berberine inhibited protein expression in IRF8-IFN-γ signaling axis stronger convincement that BBR is able to treat H. pylori induced CAG
when H. pylori infection through restraining IRF8-IFN-γ signaling axis.

We demonstrated previously that the mRNA levels of IFN-γ, IRF8 4. Discussion


and its controlled downstream genes including Ifit3, Upp1, USP18,
Nlrc5 were significantly decreased following intervention of BBR. In the current research, findings revealed that berberine could
Immediately after, the protein expression related to IRF8-IFN-γ sig- suppress the IRF8-IFN-γ signaling axis to exhibit anti-inflammatory
naling axis were measured. As reported by Yan M, et al. previously properties in H. pylori-induced chronic atrophic gastritis (CAG). It could
[14], genes increased most in H. pylori-infected gastric tissues was Ifit3 attenuate the inflammatory response by suppressing the production of
(4.7 fold change), followed by IRF1 (2.1 fold change) and Ifit1 (2.2 fold pro-Th1 cytokines via inhibiting IFN-γ expression, which is involved in
change). All those genes were found to be direct targets of IRF8 and the the anti-inflammatory role of BBR in CAG with H. pylori infection. Our
expressions were IRF8-dependent fashion. Consistent with the findings works pave a theoretical cornerstone for exploring the pharmacological
of mRNA, immunohistochemical analyses revealed that non-infected effects of berberine in the treatment of CAG induced by H. pylori.
stomach tissues expressed low levels of IRF8 as evidenced by barely > 50% of the world's population is infected with H. pylori [8]. H.
positive staining of IRF8 in nucleated glandular cells. However, H. py- pylori can colony in the gastric mucosa for decades and trigger vigorous
lori infected tissues embraced a robust increase of IRF8. Correspond- host innate and adaptive immune responses [34]. Chronic inflammation
ingly, H. pylori infected tissues hold higher level of IFN-γ compared process in stomach mucosa is the principally pathological changes of
with non-infected tissues. Interfered by BBR, the expression of IRF8 and CAG with H. pylori infection [35]. When inflammations or injury come,
IFN-γ decreased significantly. Administration of BBR at high dosage immune response of T-cell and B-cell was activated in order to clear
(28 mg/kg) exhibited slight infiltration of IRF8 and IFN-γ observably bacteria, including H. pylori, either contributing to mucosa damage in
compared with the H. pylori-infected tissues and low dosage tissues. inflammation process. Furthermore, neutrophil and macrophages re-
Results were presented in Fig. 10. cruitment also got triggered in early stage in controlling tissue

10
T. Yang, et al. Life Sciences 248 (2020) 117456

Fig. 11. Proteins expression in IRF8-IFN-γ signaling axis estimated by western blotting analysis. (A) Western blotting images of IFN-γ, IRF8, Ifit3, IRF1 and Ifit1. (B)
Relative IFN-γ protein level in stomach. (C) Relative IRF8 protein level in stomach. (D) Relative Ifit3 protein level in stomach. (E) Relative IRF1 protein level in
stomach. (F) Relative Ifit1 protein level in stomach. All data are presented as the mean ± SD. Results were from three independent experiments. #P < .05 versus
control group; ##P < .01 versus control group; ⁎P < .05 versus H. pylori infection group; ⁎⁎P < .01 versus H. pylori infection group. Ctrl, control group; L, low-dose
group; H, high-dose group. IRF8, interferon regulatory factor 8; IFN-γ, interferon-gamma; Ifit3, interferon induced protein with tetratricopeptide repeats 3; IRF1,
interferon regulatory factor 1; Ifit1, interferon induced protein with tetratricopeptide repeats 1.

infections or injury [36]. Previous study has demonstrated that H. pylori promotes the differentiation of monocytes into granulocytes [38]. IRF8
infection mainly induced Th1 cell-dominant immune response in gastric also acts as a key controller of differentiation and function in dendritic
inflammation [37]. Interferon regulatory factor 8 (IRF8), a transcrip- cell, establishing host defense and innate immunity [39]. It is uni-
tion factors, plays a vital role in the differentiation of myeloid cells and versally known that interferon (IFN) is one of the strongest inducers of

11
T. Yang, et al. Life Sciences 248 (2020) 117456

IRF8 transcription. IFN-γ is a kind of Th1 cytokines and produced by results suggested that BBR which could block the course of IRF8-IFN-γ
activated T lymphocytes (T cells) and natural killer (NK) cells. It may signaling axis to H. pylori-infected procession in vivo and in vitro. The
promote the release of proinflammatory cytokines, augmenting in- therapeutic effects of BBR associated intimately to Th1 cell-dominant
flammation and apoptosis induced by H. pylori [21]. During micro- immune responses in gastric inflammation, and changed the adaptive
organism infection, IFN-γ promotes T-bet expressing Treg cells, which immunity responses to H. pylori by converting CD4+ T cells into Th1
restrain type I responses [23]. At the same time, IRF8 is highly ex- differentiation.
pressed by Treg cells and mainly controls type I immune responses
[25]. Earlier study has illustrated that expression of IFN-γ was IRF8- 5. Conclusions
dependent in H. pylori-infected GECs cells, meanwhile, expression of
IRF8 was induced by IFN-γ in GSM06 cells via a time-dependent All in all, conclusions from this study suggest that berberine pos-
manner. Briefly, IFN-γ upregulates IRF8 expression, which, in turn, sesses therapeutic effects on H. pylori-induced chronic atrophic gastritis,
promotes IFN-γ production [14]. IFN-γ-intermediated immunity in the supporting the role of BBR as an anti-inflammatory agent in CAG in-
infection of early phase is expected to be exaggerated through this duced by H. pylori. The anti-inflammatory property of BBR tightly re-
positive feedback loop between IRF8 and IFN-γ [40]. lated to suppression of IRF8-IFN-γ signaling axis, which is crucial in
In this current examination, the feedback interaction between IRF8 Th1 cell-dominant inflammatory response. The findings from this re-
and IFN-γ is verified one more time in vitro and in vivo. This regulatory search pave a theoretical cornerstone for investigating the pharmaco-
circuit may boost inflammatory responses in gastric mucosa to promote logical effects of berberine in human CAG induced by H. pylori.
bacterial clearance. Consequently, it is potential that stimulation of
IRF8 and IFN-γ may facilitate the process of chronic infection and in- Author contributions
crease the likelihood of malignant transformation induced by H. pylori.
In this study, the expression of proinflammatory genes, including NF- Tao Yang, Xiangdong Yang and Yanling Zhao conceived the project
κB, TNF-α, IFN-γ, IRF8, USP18, and Upp1 were probed in GES-1 cells and wrote the manuscript. Tao Yang performed main part of the ex-
when H. pylori infection. We found the expression of all proin- periments, with contributions from Ruilin Wang, Chunmei Bao, Juling
flammatory genes, such as NF-κB, TNF-α, was activated in H. pylori Zhang and Ruisheng Li. Jianzhong Zhang provided us with Helicobacter
infected GES-1 cells. Furthermore, genes in IRF8-IFN-γ signaling axis pylori. Xing Chen, Shihua Wu, Jianxia Wen performed and analyzed the
also got increased when H. pylori infection. Next, the genes related to high content analysis and real-time quantitative PCR experiments data.
IRF8-IFN-γ regulatory circuit were quested in the gastric mucosa of H. Haotian Li, Shizhang Wei and Huadan Cai contributed to the data
pylori-infected rats. Accordingly, the expression of IFN-γ, IRF8 and collection and analysis. Xiangdong Yang and Yanling Zhao participated
IRF8-bound genes including Ifit3, Upp1, USP18, Nlrc5 were increased in the project design as well as manuscript draft preparation and revi-
significantly in H. pylori-infected gastric mucosa. We also revealed the sion. All authors read and approved the final manuscript.
protein expression in IRF8-IFN-γ signaling axis got escalated in H. py-
lori-infected gastric mucosa. There is no denying that IRF8-IFN-γ sig- Declaration of competing interest
naling axis exerts pivotal effects in chronic inflammation process in-
duced by H. pylori. All authors of this manuscript state that they do not have any
Berberine (BBR) is a natural isoquinoline alkaloid from various conflict of interests, and there is nothing to disclose.
traditional Chinese medicine herbs, such as Rhizoma coptidis, Cortex
phellodendri, has been widely applied in clinical treatment for many Acknowledgments
centuries. Over the past decades, many studies have revealed BBR and
its derivatives possess high activity against both inflammations and This research was financially supported by the National Key
cancers in the digestive system, and with utterly slight side effects and Research and Development Program (No.2018YFC1704500).
resistance in clinical application [27,30]. In this work, we explored the
pharmacological effects of BBR in the treatment of H. pylori-induced References
CAG. The mechanism of BBR in treating H. pylori-infected CAG were
also explored by analyzing the interaction between IRF8 and IFN-γ [1] D. Campana, D. Ravizza, P. Ferolla, et al., Risk factors of type 1 gastric neu-
response. In our findings, we observed that BBR was capable of pro- roendocrine neoplasia in patients with chronic atrophic gastritis. A retrospective,
multicentre study, Endocrine. 56 (3) (2017) 633–638, https://doi.org/10.1007/
tecting GES-1 from H. pylori infection. Cell viability, morphological s12020-016-1099-y.
identification of GES-1 cells was ameliorated directly following BBR [2] H. Song, I.G. Ekheden, Z. Zheng, et al., Incidence of gastric cancer among patients
administration. The pathologic changes of gastric mucosa, including with gastric precancerous lesions: observational cohort study in a low risk Western
population, BMJ. 351 (2015) h3867, https://doi.org/10.1136/bmj.h3867.
arrangement of gastric glandular, infiltration of lymphocytes and [3] B.N. Parsons, U.Z. Ijaz, R. D’Amore, et al., Comparison of the human gastric mi-
plasma cells and loss of parietal cells, also were better than in H. pylori crobiota in hypochlorhydric states arising as a result of Helicobacter pylori-induced
infected tissues. Several specific markers in serum, such as IL-17, atrophic gastritis, autoimmune atrophic gastritis and proton pump inhibitor use,
PLoS Pathog. 13 (11) (2017) e1006653, , https://doi.org/10.1371/journal.ppat.
CXCL1, and CXCL9, significantly reduced after intervened by BBR. [4] K. Fagan-Garcia, J. Geary, H.J. Chang, et al., Burden of disease from Helicobacter
Conversely, G-17 raised significantly following administration of BBR pylori infection in western Canadian Arctic communities, BMC Public Health 19 (1)
when H. pylori infection. We speculated that with H. pylori clearance (2019) 730, https://doi.org/10.1186/s12889-019-7065-x.
[5] L. Gao, M.N. Weck, A. Michel, et al., Association between chronic atrophic gastritis
intervened by BBR, the secretory function of gland cells (G cells) re-
and serum antibodies to 15 Helicobacter pylori proteins measured by multiplex
turned to normal action and levels. In terms of mechanism of BBR in serology, Cancer Res. 69 (7) (2009) 2973–2980, https://doi.org/10.1158/0008-
treating H. pylori-infected CAG, the proinflammatory genes, encoding 5472.
NF-κB, TNF-α, IFN-γ, IRF8 and its downstream genes were either de- [6] X. Gao, Y. Zhang, H. Brenner, Associations of Helicobacter pylori infection and
chronic atrophic gastritis with accelerated epigenetic ageing in older adults, Br. J.
creased significantly followed by AX-024, a selective inhibitor of IFN-γ, Cancer 117 (8) (2017) 1211–1214, https://doi.org/10.1038/bjc.2017.314.
or downregulated by BBR intervention in GES-1 cells. These data sup- [7] M. Hatakeyama, Helicobacter pylori CagA and gastric cancer: a paradigm for hit-
port the role of BBR as an anti-inflammatory agent in GES-1 cells injury and-run carcinogenesis, Cell Host Microbe 15 (3) (2014) 306–316, https://doi.org/
10.1016/j.chom.2014.02.008.
induced by H. pylori. We also demonstrated the expression of IFN-γ, [8] J.K.Y. Hooi, W.Y. Lai, W.K. Ng, et al., Global prevalence of helicobacter pylori in-
IRF8 and its related downstream genes including Ifit3, Upp1, USP18, fection: systematic review and meta-analysis, Gastroenterology. 153 (2) (2017)
Nlrc5 were suppressed significantly in H. pylori-infected gastric mucosa. 420–429, https://doi.org/10.1053/j.gastro.2017.04.022.
[9] D.R. Scott, E.A. Marcus, D.L. Weeks, et al., Mechanisms of acid resistance due to the
Meanwhile, we revealed the protein expression in IRF8-IFN-γ signaling urease system of Helicobacter pylori, Gastroenterology. 123 (1) (2002) 187–195,
axis were downregulated after exposure to BBR. Taken together, these

12
T. Yang, et al. Life Sciences 248 (2020) 117456

https://doi.org/10.1053/gast.2002.34218. via K48-linked deubiquitination in regulatory T cells, FEBS Lett. 591 (12) (2017)
[10] E.A. Marcus, G. Sachs, D.R. Scott, The role of ExbD in periplasmic pH homeostasis 1677–1686, https://doi.org/10.1002/1873-3468.12668.
in Helicobacter pylori, Helicobacter. 18 (5) (2013) 363–372, https://doi.org/10. [27] K. Zou, Z. Li, Y. Zhang, et al., Advances in the study of berberine and its derivatives:
1111/hel.12055. a focus on anti-inflammatory and anti-tumor effects in the digestive system, Acta
[11] Y. Zhuang, P. Cheng, X.F. Liu, et al., A pro-inflammatory role for Th22 cells in Pharmacol. Sin. 38 (2) (2017) 157–167, https://doi.org/10.1038/aps.2016.125.
Helicobacter pylori-associated gastritis, Gut. 64 (9) (2015) 1368–1378, https://doi. [28] Q. Hu, L. Li, X. Zou, et al., Berberine attenuated proliferation, invasion and mi-
org/10.1136/gutjnl-2014-307020. gration by targeting the AMPK/HNF4alpha/WNT5A pathway in gastric carcinoma,
[12] M. Howlett, H.V. Chalinor, J.N. Buzzelli, et al., IL-11 is a parietal cell cytokine that Front. Pharmacol. 9 (2018) 1150, https://doi.org/10.3389/fphar.2018.01150.
induces atrophic gastritis, Gut. 61 (10) (2012) 1398–1409, https://doi.org/10. [29] Y.H. Lin, J.H. Lin, S.C. Chou, et al., Berberine-loaded targeted nanoparticles as
1136/gutjnl-2011-300539. specific Helicobacter pylori eradication therapy: in vitro and in vivo study,
[13] D.B. Polk, R.M. Peek Jr., Helicobacter pylori: gastric cancer and beyond, Nat. Rev. Nanomedicine. 10 (1) (2015) 57–71, https://doi.org/10.2217/nnm.14.76.
Cancer 10 (6) (2010) 403–414, https://doi.org/10.1038/nrc2857. [30] X. Wu, X. Li, Z. Dang, et al., Berberine demonstrates anti-inflammatory properties in
[14] M. Yan, H. Wang, J. Sun, et al., Cutting edge: expression of IRF8 in gastric epithelial Helicobacter pylori-infected mice with chronic gastritis by attenuating the Th17
cells confers protective innate immunity against Helicobacter pylori infection, J. response triggered by the B cell-activating factor, J. Cell. Biochem. 119 (7) (2018)
Immunol. 196 (5) (2016) 1999–2003, https://doi.org/10.4049/jimmunol.1500766. 5373–5381, https://doi.org/10.1002/jcb.26681.
[15] T. Taniguchi, K. Ogasawara, A. Takaoka, et al., IRF family of transcription factors as [31] D. Werawatganon, Simple animal model of Helicobacter pylori infection, World J.
regulators of host defense, Annu. Rev. Immunol. 19 (2001) 623–655, https://doi. Gastroenterol. 20 (21) (2014) 6420–6424, https://doi.org/10.3748/wjg.v20.i21.
org/10.1146/annurev.immunol.19.1.623. 6420.
[16] H. Wang, H.C. Morse 3rd., IRF8 regulates myeloid and B lymphoid lineage di- [32] J. Wen, J. Wang, P. Li, et al., Protective effects of higenamine combined with [6]-
versification, Immunol. Res. 43 (1–3) (2009) 109–117, https://doi.org/10.1007/ gingerol against doxorubicin-induced mitochondrial dysfunction and toxicity in
s12026-008-8055-8. H9c2 cells and potential mechanisms, Biomed. Pharmacother. 115 (2019) 108936,
[17] R. Lu, Interferon regulatory factor 4 and 8 in B-cell development, Trends Immunol. https://doi.org/10.1016/j.biopha.2019.108881.
29 (10) (2008) 487–492, https://doi.org/10.1016/j.it.2008.07.006. [33] P.J. O’Brien, W. Irwin, D. Diaz, et al., High concordance of drug-induced human
[18] P. Tailor, T. Tamura, H.C. Morse 3rdet al., The BXH2 mutation in IRF8 differentially hepatotoxicity with in vitro cytotoxicity measured in a novel cell-based model using
impairs dendritic cell subset development in the mouse, Blood. 111 (4) (2008) high content screening, Arch. Toxicol. 80 (9) (2006) 580–604, https://doi.org/10.
1942–1945, https://doi.org/10.1182/blood-2007-07-100750. 1007/s00204-006-0091-3.
[19] N. Nelson, Y. Kanno, C. Hong, et al., Expression of IFN regulatory factor family [34] A. Razavi, N. Bagheri, F. Azadegan-Dehkordi, M. Shirzad, G. Rahimian, Rafieian-
proteins in lymphocytes. Induction of Stat-1 and IFN consensus sequence binding Kopaei M, et al, Comparative Immune Response in Children and Adults with H.
protein expression by T cell activation, J. Immunol. 156 (10) (1996) 3711–3720. Pylori Infection. J Immunol Res. 2015 (2015) 315957, https://doi.org/10.1155/
[20] K. Takahashi, T. Sugiyama, S. Tokoro, et al., Inhibition of interferon-gamma-in- 2015/315957.
duced nitric oxide production by 10-hydroxy-trans-2-decenoic acid through in- [35] J. Zhang, H. Wang, Morroniside protects against chronic atrophic gastritis in rat via
hibition of interferon regulatory factor-8 induction, Cell. Immunol. 273 (1) (2012) inhibiting inflammation and apoptosis, Am. J. Transl. Res. 11 (9) (2019)
73–78, https://doi.org/10.1016/j.cellimm.2011.11.004. 6016–6023.
[21] C.A. Figueiredo, C.R. Marques, S. Costa Rdos, et al., Cytokines, cytokine gene [36] K. De Filippo, A. Dudeck, M. Hasenberg, et al., Mast cell and macrophage chemo-
polymorphisms and helicobacter pylori infection: friend or foe? World J. kines CXCL1/CXCL2 control the early stage of neutrophil recruitment during tissue
Gastroenterol. 20 (18) (2014) 5235–5243, https://doi.org/10.3748/wjg.v20.i18. inflammation, Blood. 121 (24) (2013) 4930–4937, https://doi.org/10.1182/blood-
5235. 2013-02-486217.
[22] A.O.H. Hall, D.P. Beiting, C. Tato, et al., The cytokines interleukin 27 and inter- [37] K.A. Eaton, M. Mefford, T. Thevenot, The role of T cell subsets and cytokines in the
feron-γ promote distinct Treg cell populations required to limit infection-induced pathogenesis of Helicobacter pylori gastritis in mice, J. Immunol. 166 (12) (2001)
pathology, Immunity. 37 (3) (2012) 511–523, https://doi.org/10.1016/j.immuni. 7456–7461, https://doi.org/10.4049/jimmunol.166.12.7456.
2012.06.014. [38] T. Tamura, T. Nagamura-Inoue, Z. Shmeltzer, et al., ICSBP directs bipotential
[23] M.A. Koch, G. Tucker-Heard, N.R. Perdue, et al., The transcription factor T-bet myeloid progenitor cells to differentiate into mature macrophages, Immunity. 13
controls regulatory T cell homeostasis and function during type 1 inflammation, (2) (2000) 155–165, https://doi.org/10.1016/s1074-7613(00)00016-9.
Nat. Immunol. 10 (6) (2009) 595–602, https://doi.org/10.1038/ni.1731. [39] D.-M. Shin, C.-H. Lee, H.C. Morse 3rd., IRF8 governs expression of genes involved in
[24] L.F. Lu, M.P. Boldin, A. Chaudhry, et al., Function of miR-146a in controlling Treg innate and adaptive immunity in human and mouse germinal center B cells, PLoS
cell-mediated regulation of Th1 responses, Cell 142 (6) (2010) 914–929, https:// One 6 (11) (2011) e27384, , https://doi.org/10.1371/journal.pone.0027384.
doi.org/10.1016/j.cell.2010.08.012. [40] J.F. Marquis, O. Kapoustina, D. Langlais, et al., Interferon regulatory factor 8 reg-
[25] W. Lee, H.S. Kim, S.Y. Baek, et al., Transcription factor IRF8 controls Th1-like ulates pathways for antigen presentation in myeloid cells and during tuberculosis,
regulatory T-cell function, Cell Mol Immunol. 13 (6) (2016) 785–794, https://doi. PLoS Genet. 7 (6) (2011) e1002097, , https://doi.org/10.1371/journal.pgen.
org/10.1038/cmi.2015.72. 1002097.
[26] R. Lin, J. Nie, J. Ren, et al., USP4 interacts and positively regulates IRF8 function

13

You might also like