Open Access

Astaxanthin inhibits integrin α5 expression by suppressing activation of JAK1/STAT3 in Helicobacter pylori‑stimulated gastric epithelial cells

  • Authors:
    • Jieun Woo
    • Joo Weon Lim
    • Hyeyoung Kim
  • View Affiliations

  • Published online on: May 16, 2023     https://doi.org/10.3892/mmr.2023.13014
  • Article Number: 127
  • Copyright: © Woo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Integrins act as cell‑matrix adhesion molecules involved in cell attachment to the extracellular matrix and generate signals that respond to cancer metastasis. Integrin α5β1 is a heterodimer with α5 and β1 subunits and mediates cell adhesion and migration of cancer cells. Integrins are transcriptionally regulated by the Janus kinase (JAK)/STAT signaling pathways. Our previous study revealed that Helicobacter pylori increased the levels of reactive oxygen species (ROS), which activate JAK1/STAT3 in gastric cancer AGS cells in vitro. Astaxanthin (ASX) has been reported to be an effective antioxidant and anticancer nutrient. The present study investigated whether ASX suppresses H. pylori‑induced integrin α5 expression, cell adhesion and migration and whether ASX reduces ROS levels and suppresses phosphorylation of JAK1/STAT3 in gastric cancer AGS cells stimulated with H. pylori. The effect of ASX was determined using a dichlorofluorescein fluorescence assay, western blot analysis, adhesion assay and wound‑healing assay in AGS cells stimulated with H. pylori. The results demonstrated that H. pylori increased the expression levels of integrin α5, without affecting integrin β1, and increased cell adhesion and migration of AGS cells. ASX reduced ROS levels and suppressed JAK1/STAT3 activation, integrin α5 expression, cell adhesion and migration of H. pylori‑stimulated AGS cells. In addition, both a JAK/STAT inhibitor, AG490, and an integrin α5β1 antagonist, K34C, suppressed cell adhesion and migration in H. pylori‑stimulated AGS cells. AG490 inhibited integrin α5 expression in AGS cells stimulated with H. pylori. In conclusion, ASX inhibited H. pylori‑induced integrin α5‑mediated cell adhesion and migration by decreasing the levels of ROS and suppressing JAK1/STAT3 activation in gastric epithelial cells.

Introduction

Helicobacter pylori is the major pathogen in atrophic gastritis, peptic ulcers and gastric cancer (1). The underlying mechanism of H. pylori-mediated gastric carcinogenesis is the production of reactive oxygen species (ROS) by neutrophil activation or by H. pylori itself in the infected gastric epithelial cells (2). Our previous studies indicated that H. pylori induced NADPH oxidase activation, resulting in increased production of ROS in human gastric epithelial cells (3,4).

ROS are composed of hydroxyl radicals, superoxide anion and hydrogen peroxide (5). The normal range of ROS levels can be strictly controlled by antioxidant defense systems such as superoxide dismutase (SOD), catalase and glutathione peroxidase in mammalian cells (6). However, excessive ROS generation potentially causes oxidative stress, which leads to tumor initiation, growth and progression, resulting in cancer development (68). ROS activate Janus kinase (JAK)/STAT signaling and phosphorylate STAT3 in stem cells and a human salivary gland cell line (9,10). Inflammatory stimuli such as cytokines potentially activate JAKs via phosphorylation; thereafter, activated JAKs recruit STATs (11). Activated/phosphorylated (p-)STATs form dimers that translocate from the cytoplasm to the nucleus, regulating the expression of pro-inflammatory genes such as interleukin-6 by binding to specific promoter sequences (12). Activated STAT3 mediates the expression of cancer-inducing genes related to tumor cell survival, migration and metastasis (13,14) such as integrin β6 (15).

Integrins are heterodimeric, transmembrane proteins that comprise α and β subunits (16). Mammals possess 18 α and eight β integrin subunits that combine to form 24 types of αβ integrin heterodimers (17). Different integrin heterodimers bind to different extracellular ligands and distinct integrin heterodimers are present in various tissues (18). Among these integrins, integrin α5β1 is a heterodimer with α5 and β1 subunits. Integrin α5β1 is related to tumor metastasis and carcinogenesis (17). Integrin-mediated cell adhesion serves an important role in cancer metastasis by inducing further cell migration and invasion (19,20). Integrin α5 promotes migration and invasion through STAT3 signaling in non-small cell lung cancer cells (21).

Our previous study showed that an H. pylori strain originally isolated from a Korean patient (HP99) increased integrin α5/β1 expression, which was inhibited by treatment with diphenyleneiodonium chloride, an inhibitor of NADPH oxidase in AGS cells (22). The results clearly demonstrated the involvement of NADPH oxidase in H. pylori-induced integrin expression. In addition, HP99 induces the expression of proteinase-activated receptor-2, which mediates α5/β1 expression and cell adhesion to fibronectin in gastric epithelial AGS cells (23). A study using a cDNA microarray of 352 cancer-related genes demonstrated that H. pylori (strain NCTC11637) increased integrin α5 expression in AGS cells (24). Since H. pylori increases ROS-mediated activation of the JAK1/STAT3 signaling pathway in AGS cells (25), JAK1/STAT3 may be involved in the upregulation of integrin expression in H. pylori-stimulated AGS cells.

Astaxanthin (ASX) is a red-orange and lipid-soluble oxycarotenoid pigment (26). It is found in seafood such as shrimp, lobster, trout, krill and salmon (26). ASX can scavenge ROS in both the inner and outer membrane of the cells (27). Therefore, ASX may inhibit development of oxidative stress-induced diseases, including inflammatory diseases (28). Furthermore, ASX has anticancer effects, inducing anti-proliferative activity and suppressing cell migration and invasion in colorectal cancer, breast cancer, melanoma, gastric cancer and oral cancer cells (2931). By inhibiting matrix metalloproteinase-1, −2 and −9 expression, ASX suppresses melanoma cancer cell migration and metastasis (30). ASX inhibits cancer development and progression by inhibiting STAT3 signaling in oral squamous carcinoma cells (31). Another study demonstrated that, in H. pylori-stimulated cells, ASX treatment activated peroxisome proliferator-activated receptor γ (PPAR-γ) to induce the expression of its downstream antioxidant gene catalase, and suppressed intracellular and mitochondrial ROS production (32). Furthermore, ASX prevents H. pylori-induced decreases in SOD2 levels and SOD activity, and reduces mitochondrial ROS in gastric epithelial cells (33). Therefore, ASX may suppress H. pylori-induced cell adhesion and migration by reducing ROS-mediated activation of STAT3 and expression of integrins in gastric epithelial cells.

The present study aimed to determine whether ASX suppresses integrin α5/β1 expression, cell adhesion and migration by inhibiting the JAK1/STAT3 signaling pathway in H. pylori-stimulated gastric epithelial cells. In addition, integrin α5/β1 expression, cell adhesion and migration were determined after treatment with a JAK/STAT inhibitor, AG490, and an integrin α5β1 antagonist, K34C, in H. pylori-stimulated cells.

Materials and methods

Cell line and culture conditions

The AGS human gastric epithelial cell line (CRL 1739) was obtained from American Type Culture Collection. Cells were cultured in complete medium comprising RPMI 1640 medium (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 100 U/ml penicillin, 2 mM glutamine, 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) and 100 µg/ml streptomycin. Subsequently, the cells were cultured in a cell incubator at 37°C in a humidified atmosphere with 95% air and 5% CO2.

Bacterial strain

H. pylori, strain NCTC11637 [cytotoxin-associated gene (cag) A+, vacuolating cytotoxin gene (vac) A+ strain], was obtained from American Type Culture Collection. The bacterium was inoculated on chocolate agar plates (Becton, Dickinson and Company) at 37°C under microaerophilic conditions (low levels of dioxygen but CO2-enriched environment) using an anaerobic chamber (BBL Campy Pouch® System; Becton, Dickinson and Company).

Reagents

ASX (Sigma-Aldrich; Merck KGaA) was dissolved in DMSO at 4 mM and stored in nitrogen gas at −80°C. Before use for treatment, the ASX stock solution was thawed at 21–23°C and diluted in 100% FBS to the desired concentrations (final concentration of 1 and 5 µM). The JAK/STAT inhibitor AG490 (cat. no. T3434; R&D Systems, Inc.) and integrin α5β1 antagonist K34C (N-(2,6-Dimethylbenzoyl)-O-[3-(2-pyridinylamino)propyl]-L-tyrosine; cat. no. 5114; R&D Systems, Inc.) were dissolved in DMSO at 50 mM.

Culture of AGS cells with H. pylori

AGS cells (8×104/2 ml or 4×105/10 ml) were seeded and cultured overnight to achieve 80% confluency. Whole H. pylori was harvested from the chocolate agar plates, suspended in antibiotic-free RPMI 1640 medium supplemented with 10% FBS, and subsequently added to the cells. The cells were culture with H. pylori in a cell incubator at 37°C in a humidified atmosphere with 95% air and 5% CO2 at a ratio of 1:50 for 30 min (to determine the levels of intracellular ROS, p-JAK1, JAK1, p-STAT3 and STAT3), 6 h (to determine the expression levels of integrin α5 and β1), 20 h (for the migration assay) and 24 h (for the adhesion assay).

Experimental protocol

AGS cells [1.5×105/2 ml (for measurement of intracellular ROS levels) or 7.5×105/10 ml (for Western blot analysis)] were treated at 37°C with ASX (1 or 5 µM) for 3 h before stimulation with H. pylori. Thereafter, cells were stimulated with H. pylori at 37°C for 30 min (to determine the levels of intracellular ROS, p-JAK1, JAK1, p-STAT3 and STAT3), 6 h (to determine the expression levels of integrin α5 and β1), 20 h (for the migration assay) and 24 h (for the adhesion assay). To determine the involvement of JAK/STAT and integrin in cell adhesion and migration, the cells were pretreated with AG490 (40 µM) and K34C (20 µM) at 37°C for 2 h and stimulated with H. pylori at 37°C for 20 h (for the migration assay) and 24 h (for the adhesion assay). For each experiment, the untreated cells were cultured with H. pylori for 30 min (to determine the levels of intracellular ROS, p-JAK1, JAK1, p-STAT3 and STAT3), 6 h (to determine the expression levels of integrin α5 and β1), 20 h (for the migration assay) and 24 h (for the adhesion assay) with the vehicle DMSO (0.1%) alone instead of ASX, AG490 or K34C.

To determine whether AG490 and K34C have an effect on the inhibitory activity of ASX on integrin α5 and β1 expression in H. pylori-stimulated cells, cells were pretreated with ASX (5 µM), AG490 (40 µM) and K34C (20 µM) at 37°C, and then stimulated with H. pylori at 37°C for 6 h. The pretreatment period for ASX was 3 h, while that of AG490 or K34C was 2 h. Protein expression levels of integrin α5 and β1 were determined using western blot analysis as described below.

To determine the appropriate incubation time for ROS level, activation of JAK1/STAT3, integrin expression and adhesion assays, time-course experiments were performed after stimulation of AGS cells with H. pylori. AGS cells (1.5×105/2 ml/well in 6-well plates) were stimulated with H. pylori at 37°C for 20, 30 and 60 min (for intracellular ROS levels), 15, 30 and 60 min (for total and phospho-specific levels of JAK/STAT proteins), 2, 4, 6 and 8 h (for integrin expression), or 6, 12, 18 and 24 h (for the adhesion assay).

Measurement of intracellular ROS levels

The cells were treated with 10 µg/ml dichlorofluorescein diacetate (Sigma-Aldrich; Merck KGaA) at 37°C for 30 min. Dichlorofluorescein fluorescence (excitation, 495 nm; emission, 535 nm) was detected using a Victor3 Multi-label Counter (PerkinElmer, Inc.) [https://resources.perkinelmer.com/lab-solutions/resources/docs/bro_victor3.pdf]. Intracellular ROS levels were normalized to the cell numbers and expressed as the relative percentage of control cells. Normalization was achieved by dividing intracellular ROS levels by the cell numbers.

Preparation of cell extracts

The cells were harvested using trypsin-EDTA and pelleted by centrifugation at 1,000 × g at 4°C for 5 min. The cell pellets were resuspended with lysis buffer containing 10 mM Tris (pH 7.4), 15 mM NaCl, 1% NP-40 and commercial protease inhibitor complex (Complete; Roche Diagnostics GmbH), and lysed by drawing the cells through a 1-ml syringe with several rapid strokes. The mixture was then incubated at 0°C on ice for 30 min and centrifuged at 13,000 × g at 4°C for 15 min. The supernatant was collected and used as whole-cell extracts. The protein concentration was determined by using the Bradford assay (Bio-Rad Laboratories, Inc.).

Western blot analysis

Whole-cell extracts (6–40 µg protein/lane) were loaded per lane, separated using 8–10% SDS-PAGE under reductive conditions and transferred to nitrocellulose membranes (Amersham; Cytiva) by electroblotting. Protein transfer was verified using reversible Ponceau S staining. Membranes were blocked using 3% non-fat, dry milk in TBS with 0.2% Tween 20 (TBS-T) at 21–23°C for 1 h. The proteins were detected by incubation with antibodies against integrin α5 (1:500 dilution) (cat. no. 610633; BD Biosciences), integrin β1 (1:500 dilution) (cat. no. 610467; BD Biosciences), p-JAK1 (1:300 dilution) (cat. no. 3331S; Cell Signaling Technology, Inc.), JAK1 (1:500 dilution) (cat. no. 3332S; Cell Signaling Technology, Inc.), p-STAT3 (1:200 dilution) (cat. no. 9131S; Cell Signaling Technology, Inc.), STAT3 (1:500 dilution) (cat. no. 4904S; Cell Signaling Technology, Inc.) and β-actin (1:2,000 dilution) (cat. no. sc-47778; Santa Cruz Biotechnology, Inc.) in TBS-T containing 3% dry milk overnight at 4°C. After washing with TBS-T, the primary antibodies were detected using HRP-conjugated secondary antibodies [anti-mouse (1:2,000 dilution) (cat. no. sc-2005 Santa Cruz Biotechnology, Inc.) or anti-rabbit (1:2,000 dilution) (cat. no. sc-2357 Santa Cruz Biotechnology, Inc.)] and visualized using the enhanced chemiluminescence detection system (Santa Cruz Biotechnology, Inc.) based on exposure to BioMax MR film (Kodak). The enhanced chemiluminescence detection system is comprised of a two-component kit containing a bottle of luminol substrate and a bottle of peroxide solution. The solutions are equilibrated to 21–23°C and then mixed in equal volumes directly before use. The levels of integrins were compared with those of β-actin. The levels of p-JAK1 or p-STAT3 were compared with those of total JAK1 or total STAT3, respectively. Protein band density was semi-quantified using Image J software (version 1.46; National Institutes of Health). The data are presented as the mean ± standard error from three immunoblots and are shown as the relative density of the protein band normalized to the indicated protein (β-actin or total JAK1 or STAT3).

Adhesion assay

The adhesive potential of AGS cells stimulated with H. pylori was assessed using human fibronectin-coated 96-well strips (cat. no. ECM101; Sigma-Aldrich; Merck KGaA). Cells were pretreated with ASX at 37°C for 3 h and then stimulated with H. pylori at 37°C for 24 h. The cells were detached by incubation with 6.6 mM EDTA at 37°C for 5 min, washed with PBS and suspended in serum-free RPMI 1640 containing 0.02% BSA (Sigma-Aldrich; Merck KGaA) at a density of 1.5×105 cells/ml. The cell suspension (100 µl) was added to each well of the fibronectin-coated plates and cells were incubated at 37°C for 1 h. The medium was removed from the wells and the cells were gently washed twice with PBS containing 0.9 mM Ca2+ and 0.5 mM Mg2+. The adherent cells were stained with 0.2% crystal violet in 10% ethanol for 5 min at 21–23°C and subsequently washed three times with PBS. After washing, cells were solubilized using solubilization buffer (a 50/50 mixture of 0.1 M NaH2PO4, pH 4.5 and 50% ethanol) at 21–23°C for 10 min, and the absorbance at 570 nm was quantified using a microplate reader. The percentage of adherent cells in the 0 h or ‘None’ (uninfected cells without any treatment) groups was set as 100%.

Wound-healing assay

Cells were seeded in 12-well plates and allowed to grow in a complete medium until 90–100% confluency. Subsequently, individual wells were scratched using a 200-µl micropipette tip to create a denuded zone (the ‘wound space’ of an artificially ruptured monolayer of AGS cells) of constant width (1 mm). Complete media were removed from these cells and replaced with RPMI-1640 containing 4% FBS. The cells were pretreated with 5 or 10 µM ASX at 37°C for 3 h and then stimulated with H. pylori at 37°C for 20 h. To determine cell migration, cell images were captured at baseline (0 h) and after 20 h of H. pylori stimulation using phase-contrast light microscopy. The final gap width of the scratch was measured and compared with the initial gap. The wound area was quantified using ImageJ software (version 1.46; National Institutes of Health).

Statistical analysis

Statistical analysis was performed using SPSS software, version 14 (SPSS, Inc). All values are presented as the mean ± standard error of three independent experiments. For each experiment, four samples were used for each group (total number for each group was 12). One-way ANOVA, followed by Tukey's post-hoc test, was used for statistical analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

ASX inhibits integrin α5 expression in H. pylori-stimulated AGS cells

To determine whether H. pylori induces integrin α5 and β1 expression, AGS cells were stimulated with H. pylori for the indicated time periods. As shown in Fig. 1A, H. pylori increased the expression levels of integrin α5 at 2 h, which continued to 6 h. Therefore, to measure the effect of ASX on H. pylori-induced integrin α5 expression, a 6 h-culture period was used. However, integrin β1 expression was not altered following H. pylori stimulation. H. pylori-induced integrin α5 expression was suppressed by ASX treatment (final concentration, 1 and 5 µM) at 6 h (Fig. 1B).

ASX inhibits H. pylori-induced cell adhesion and migration

To determine whether H. pylori increases the number of adherent cells, the cells were stimulated with H. pylori up to 24 h. H. pylori stimulation significantly increased the number of adherent cells at 12 h, which continued to 24 h (Fig. 2A). At 24 h of culture, the number of adherent cells in the H. pylori-stimulated group was two times higher compared with that of the unstimulated group (‘H. pylori -, ASX -’) (Fig. 2B). The H. pylori-stimulated increase in adherent cells was reduced by ASX treatment (both 1 and 5 µM) at 24 h (Fig. 2B).

Since the percentage of wound closure reflects the migration rate, images were captured at 0 and 20 h after wounding. As shown in Fig. 2C, H. pylori increased the rate of wound closure, which was suppressed by ASX at 20 h.

ASX reduces ROS levels and suppresses activation of JAK1/STAT3 in H. pylori-stimulated cells

As shown in Fig. 3A, H. pylori increased intracellular ROS levels at 30 min in AGS cells. Therefore, a 30-min culture period was used to examine the effect of ASX on ROS levels in H. pylori-stimulated cells (Fig. 3B). Both 1 and 5 µM ASX reduced the ROS levels, which were increased by H. pylori stimulation at 30 min.

Stimulation with H. pylori increased the p-JAK1/total-JAK1 ratio at 30 min, and this decreased at 60 min (Fig. 3C). The p-STAT3/total-STAT3 ratio was significantly increased by H. pylori stimulation up to 60 min. However, the total levels of JAK1 and STAT3 were not changed by H. pylori. ASX markedly reduced the increased ratio of p-JAK1/total-JAK1 and p-STAT3/total-STAT3 in H. pylori-stimulated cells (Fig. 3D).

AG490 or K34C suppress H. pylori-induced integrin α5 expression, cell adhesion and migration

To determine the effect of JAK/STAT on integrin α5 expression in H. pylori-stimulated AGS cells, a JAK/STAT inhibitor, AG490, was used to treat the H. pylori-stimulated cells. AG490 reduced the H. pylori-induced increase in integrin α5 levels (Fig. 4A). To assess whether H. pylori-induced cell adhesion and migration are mediated by JAK/STAT activation and integrin α5β1, AG490 and K34C, an integrin α5β1 antagonist, were used. AG490 and K34C reduced the H. pylori-induced increase in adherent cells and wound closure levels (Fig. 4B and C). Therefore, H. pylori may increase cell adhesion and migration via JAK1/STAT3 activation and integrin α5 expression in AGS cells.

Figure 4.

AG490 and K34C suppress Helicobacter pylori-induced integrin α5 expression, cell adhesion and migration. (A) Cells were pretreated with 40 µM AG490 for 2 h, and subsequently stimulated with H. pylori for 6 h. Protein levels of integrin α5 and β1 were determined using western blot analysis, with β-actin as the loading control. (B) Cells were pretreated with 40 µM AG490 or 20 µM K34C for 2 h, and subsequently stimulated with H. pylori for 24 h. Adherent cells were stained and absorbance was detected at 570 nm. Results are presented as the mean ± standard error (the total number for each group was 12). The percentage of adherent cells in the ‘None’ group was set as 100%. (C) Cells were pretreated with 40 µM AG490 or 20 µM K34C for 2 h, and subsequently stimulated with H. pylori for 20 h. The wound healing assay was performed to detect cell migration. Representative images of wounds in AGS cells were captured at baseline (0 h) and at 20 h (magnification, ×100). Wound closure was evaluated by measuring the remaining cell-free area and expressed as a percentage of the initial cell-free area. The bar graph indicates wound closure rate (%). Results are presented as the mean ± SE (the total number for each group was 12). (D) Cells were pretreated with ASX (5 µM), AG490 (40 µM) and K34C (20 µM), and then stimulated with H. pylori for 6 h. The pretreatment period for ASX was 3 h, while that of AG490 or K34C was 2 h. Protein levels of integrin α5 and β1 were determined using western blot analysis, with β-actin as the loading control. The densitometry data are presented as the mean ± standard error from three immunoblots, and are shown as the relative density of protein bands normalized to β-actin. *P<0.05 vs. ‘None’ (unstimulated cells without treatment of AG490, K34C or ASX). #P<0.05 vs. ‘Control’ (H. pylori-stimulated cells without treatment of AG490, K34C or ASX). ASX, astaxanthin.

To determine whether AG490 and K34C have an effect on the inhibitory activity of ASX on the expression of integrin α5 in H. pylori-stimulated cells, the cells were pretreated with ASX, AG490 and K34C, followed by H. pylori stimulation for 6 h. The inhibitory activity of ASX on expression of integrin α5 was not changed by addition of AG490 in H. pylori-stimulated cells (Fig. 4D). As shown in Figs. 1B and 3D, the levels of integrin α5 expression, p-JAK1/total-JAK1 ratio, and p-STAT3/total-STAT3 ratio in H. pylori-stimulated cells treated with ASX (5 µM) were lower than those of unstimulated cells (H. pylori -, ASX -). As shown in Fig. 4A, the level of integrin α5 expression in H. pylori-stimulated cells treated with a JAK/STAT inhibitor AG490 was similar to unstimulated cells. The results demonstrate that ASX act as an inhibitor of JAK1/STAT3 like AG490, but the inhibitory effect of ASX (5 µM) for integrin α5 expression was higher than the effect of AG490 (40 µM). Thus, the addition of AG490 to ASX did not have synergistic effect on inhibition by ASX on integrin α5 expression in H. pylori-stimulated cells. K34C is a selective ligand for integrin α5β1 (34), which inhibits functions of integrin α5β1 including migration in glioma cells (35). As shown in Fig. 4B and C, cell adhesion and migration in H. pylori-stimulated cells treated with of K34C (20 µM) were similar to the levels in unstimulated cells. Similarly, ASX treatment reduced H. pylori-induced increases in adherent cells and migration (Fig. 1B and C). These results indicate that ASX may inhibit functions of integrin α5β1 since ASX reduced the expression of integrin α5 in H. pylori-stimulated cells. Even though K34C is a selective functional inhibitor of integrin α5β1, further study is necessary to determine whether K34C reduces expression of integrin α5 in H. pylori-stimulated cells.

Discussion

In the present study, ASX inhibited H. pylori-induced integrin α5 expression, cell adhesion and migration by suppressing ROS-mediated activation of JAK1/STAT3 in gastric epithelial AGS cells. In addition, both a JAK/STAT inhibitor (AG490) and an integrin α5β1 antagonist (K34C) suppressed cell adhesion and migration in H. pylori-stimulated AGS cells.

For the virulence factor of H. pylori, cag A is a unique genomic fragment containing ~30 genes and is required for the secretion of the cag A protein (36). Vac A induces cytoplasmic vacuolation in gastric epithelial cells (37) and is responsible for apoptosis in gastric epithelial cells (38). There are different allelic variants in the vac A sequence; three different families of vacA signal sequences (s1a, s1b and s2) and two different families of middle-region alleles (m1 and m2) (39). These variants are related to the differences in cytotoxin production and clinical outcomes of the H. pylori infection (4042).

Regarding integrin expression in H. pylori-stimulated gastric epithelial cells, H. pylori in a Korean isolate (HP99) stimulated integrin α5 and β1 expression in gastric epithelial cells (22,23). However, H. pylori (NCTC11637) upregulated integrin α5, but not integrin β1, in gastric epithelial cells (24), which supports the present findings using NCTC11637. In a previous study by our group, HP99 was identified as H. pylori cag A+, vac A+ (s1, m2) strain (43), while NCTC11637 is cag A+, vac A+ (s1, m1) strain (44). These different allelic variants in the vac A sequence may affect the expression of integrin α5 and β1 expression in gastric epithelial cells.

In regard to integrin expression, cell adhesion and the virulence factor of H. pylori, Su et al (45) revealed that antibodies against α5- and β1-integrin decreased adherence in AGS cells. Miyata et al (46) also demonstrated the relation of integrin α5β1 expression and adhesion to fibronectin in gastric cancer cells. In H. pylori-stimulated cells, the virulence factor cag A mediates integrin α5β1 expression in infected gastric epithelial cells (47). These studies showed the close relation of cag A, integrin α5 expression and cell adhesion in gastric epithelial cells.

To investigate the levels of ROS and JAK/STAT3, a 30 min-culture period was used, since increased ROS levels and activation of JAK1/STAT3 were observed after 30 min of stimulation with H. pylori. As aforementioned, cag A phosphorylation is important for integrin-mediated cell adhesion in gastric epithelial cells (46). Backert et al (48) found that tyrosine phosphorylation of the cag A protein occurred at 15 min and increased until 180 min in H. pylori-stimulated gastric epithelial cells. Tsugawa et al (49) demonstrated that phosphorylated cag A was dephosphorylated by the H. pylori vac A, which exerted its effect by reducing intracellular glutathione and increasing ROS in gastric epithelial cells. Further studies should be performed to determine whether H. pylori cag A is translocated into AGS cells after 30 min of stimulation with H. pylori.

ROS contribute to the development of gastric cancer and ROS levels are elevated in patients with gastric cancer (50,51). H. pylori stimulation increases ROS levels in gastric epithelial cells (5254), which activates the JAK1/STAT3 signaling pathway to induce cytokine IL-8 expression in gastric epithelial cells (55).

JAK/STAT signaling serves a ciritcal role in the pathogenesis of solid tumour development (11). Furthermore, constitutively activated STAT3 increases integrin β6 expression, which mediates tumorigenesis and enhances cell motility of prostate epithelial cells (15). JAK2-STAT5 signaling mediates the integrin-dependent migration of mesenchymal stem cells in ischemic cerebral lesions in vivo (56). These findings suggest a potential association among JAK/STAT, integrins and adhesion/migration in gastric cancer development (11,15,56). In the present study, integrin α5 expression was decreased by treatment with AG490, a JAK/STAT inhibitor. These results suggest that the JAK1/STAT3 signaling pathway may regulate integrin α5 expression. AG490 also inhibited integrin-mediated cell adhesion and migration of H. pylori-stimulated AGS cells. The present findings demonstrated the activation of JAK1/STAT3, as examined by detecting phosphorylation of JAK1/STAT3, in H. pylori-stimulated cells. Further studies are necessary to determine the translocation of STAT3 to the nucleus to induce integrin α5 expression in H. pylori-stimulated AGS cells.

ASX suppressed JAK1/STAT-3 activation to inhibit invasion and angiogenesis in an oral cancer model (57). Another study reported the protective effect of ASX against breast cancer cells in vitro through the suppression of proliferation and migration (58). To the best of our knowledge, the novel finding of the present study is that the antioxidant effect of ASX may attenuate JAK1/STAT3 activation, integrin α5 protein expression, cell adhesion and migration in H. pylori-stimulated gastric epithelial cells.

Regarding the dosage of ASX, 5 µM ASX inhibits oxidative stress-induced cell death by reducing ROS levels and inhibiting the degradation of DNA repair protein Ku in gastric epithelial cells (59). Furthermore, 5 µM ASX inhibits H. pylori-induced IL-8 expression by activating PPAR-γ and its target gene catalase in AGS cells (32). Our previous study demonstrated that 5 µM ASX did not induce cell death, while 10 and 20 µM ASX decreased the viable cell numbers in gastric epithelial AGS cells (60). Therefore, 1 and 5 µM ASX were used to determine the effect of ASX on integrin expression in H. pylori-stimulated gastric epithelial cells in the present study.

In the present study, AGS cells were cultured with H. pylori. JAK1/STAT3 signaling was activated in H. pylori-stimulated cells. Backert et al (48) and Tsugawa et al (49) determined the presence of cag A or vac A in gastric epithelial cells after stimulation with H. pylori. Therefore, further studies should be performed to determine the presence of H. pylori virulence factors in H. pylori-stimulated AGS cells to confirm the infection of H. pylori in AGS cells.

In conclusion, ASX suppressed H. pylori-induced cell adhesion and migration by suppressing integrin α5 expression via the ROS-mediated JAK1/STAT3 signaling pathway in human gastric epithelial cells.

Acknowledgements

Not applicable.

Funding

The present study was supported by the BK21 FOUR project, Yonsei University, Republic of Korea.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

HK conceived and designed the experiments. JWL assisted in the experimental design. JW performed the experiments. JW and JWL analyzed the data. JW and JWL confirmed the authenticity of all raw data. JW wrote the manuscript. JWL and HK reviewed and edited the manuscript. JWL designed the additional experiments for the revised manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Kusters JG, van Vliet AH and Kuipers EJ: Pathogenesis of Helicobacter pylori infection. Clin Microbiol Rev. 19:449–490. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Handa O, Naito Y and Yoshikawa T: Helicobacter pylori: A ROS-inducing bacterial species in the stomach. Inflamm Res. 59:997–1003. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Cha B, Lim JW, Kim KH and Kim H: 15-deoxy-D12,14-prostaglandin J2 suppresses RANTES expression by inhibiting NADPH oxidase activation in Helicobacter pylori-infected gastric epithelial cells. J Physiol Pharmacol. 62:167–174. 2011.PubMed/NCBI

4 

Cha B, Lim JW, Kim KH and Kim H: HSP90beta interacts with Rac1 to activate NADPH oxidase in Helicobacter pylori-infected gastric epithelial cells. Int J Biochem Cell Biol. 42:1455–1461. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Halliwell B: Reactive oxygen species in living systems: Source, biochemistry, and role in human disease. Am J Med. 91:14S–22S. 1991. View Article : Google Scholar : PubMed/NCBI

6 

Snezhkina AV, Kudryavtseva AV, Kardymon OL, Savvateeva MV, Melnikova NV, Krasnov GS and Dmitriev AA: ROS generation and antioxidant defense systems in normal and malignant cells. Oxid Med Cell Longev. 2019:61758042019. View Article : Google Scholar : PubMed/NCBI

7 

Fiedor J and Burda K: Potential role of carotenoids as antioxidants in human health and disease. Nutrients. 6:466–488. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Sies H and Jones DP: Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol. 21:363–383. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Venkatabalasubramanian S: The complex interplay between JAK-STAT pathway and ROS in regulating stem cells during inflammation and cancer. Chakraborti S: Handbook of Oxidative Stress in Cancer: Therapeutic Aspects. Springer; Singapore: pp. 1–12. 2022

10 

Charras A, Arvaniti P, Le Dantec C, Dalekos GN, Zachou K, Bordron A and Renaudineau Y: JAK inhibitors and oxidative stress control. Front Immunol. 10:28142019. View Article : Google Scholar : PubMed/NCBI

11 

Thomas SJ, Snowden JA, Zeidler MP and Danson SJ: The role of JAK/STAT signalling in the pathogenesis, prognosis and treatment of solid tumours. Br J Cancer. 113:365–371. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Pfitzner E, Kliem S, Baus D and Litterst CM: The role of STATs in inflammation and inflammatory diseases. Curr Pharm Des. 10:2839–2850. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Yu H, Kortylewski M and Pardoll D: Crosstalk between cancer and immune cells: Role of STAT3 in the tumour microenvironment. Nat Rev Immunol. 7:41–51. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Kamran MZ, Patil P and Gude RP: Role of STAT3 in cancer metastasis and translational advances. Biomed Res Int. 2013:4218212013. View Article : Google Scholar : PubMed/NCBI

15 

Azare J, Leslie K, Al-Ahmadie H, Gerald W, Weinreb PH, Violette SM and Bromberg J: Constitutively activated Stat3 induces tumorigenesis and enhances cell motility of prostate epithelial cells through integrin beta 6. Mol Cell Biol. 27:4444–4453. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Park EJ, Myint PK, Ito A, Appiah MG, Darkwah S, Kawamoto E and Shimaoka M: Integrin-ligand interactions in inflammation, cancer, and metabolic disease: Insights into the multifaceted roles of an emerging ligand irisin. Front Cell Dev Biol. 8:5880662020. View Article : Google Scholar : PubMed/NCBI

17 

Hou J, Yan D, Liu Y, Huang P and Cui H: The roles of integrin α5β1 in human cancer. Onco Targets Ther. 13:13329–13344. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Hynes RO: Integrins: Bidirectional, allosteric signaling machines. Cell. 110:673–687. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Gallant ND, Michael KE and García AJ: Cell adhesion strengthening: Contributions of adhesive area, integrin binding, and focal adhesion assembly. Mol Biol Cell. 16:4329–4340. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Bendas G and Borsig L: Cancer cell adhesion and metastasis: Selectins, integrins, and the inhibitory potential of heparins. Int J Cell Biol. 2012:6767312012. View Article : Google Scholar : PubMed/NCBI

21 

Yang Y, Wang Y, Che X, Hou K, Wu J, Zheng C, Cheng Y, Liu Y, Hu X and Zhang J: Integrin α5 promotes migration and invasion through the FAK/STAT3/AKT signaling pathway in icotinib-resistant non-small cell lung cancer cells. Oncol Lett. 22:5562021. View Article : Google Scholar : PubMed/NCBI

22 

Cho SO, Kim KH, Yoon JH and Kim H: Signaling for integrin alpha5/beta1 expression in Helicobacter pylori-infected gastric epithelial AGS cells. Ann N Y Acad Sci. 1090:298–304. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Seo JH, Lim JW, Yoon JH and Kim H: Proteinase-activated receptor-2 mediates the expression of integrin alpha5 and beta1 in Helicobacter pylori-infected gastric epithelial AGS cells. Digestion. 80:40–49. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Lim JW, Kim H and Kim KH: Cell adhesion-related gene expression by Helicobacter pylori in gastric epithelial AGS cells. Int J Biochem Cell Biol. 35:1284–1296. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Cha B, Lim JW and Kim H: Jak1/Stat3 is an upstream signaling of NF-κB activation in Helicobacter pylori-induced IL-8 production in gastric epithelial AGS cells. Yonsei Med J. 56:862–866. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Higuera-Ciapara I, Félix-Valenzuela L and Goycoolea FM: Astaxanthin: A review of its chemistry and applications. Crit Rev Food Sci Nutr. 46:185–196. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Hussein G, Sankawa U, Goto H, Matsumoto K and Watanabe H: Astaxanthin, a carotenoid with potential in human health and nutrition. J Nat Prod. 69:443–449. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Sztretye M, Dienes B, Gönczi M, Czirják T, Csernoch L, Dux L, Szentesi P and Keller-Pintér A: Astaxanthin: A potential mitochondrial-targeted antioxidant treatment in diseases and with aging. Oxid Med Cell Longev. 2019:38496922019. View Article : Google Scholar : PubMed/NCBI

29 

Faraone I, Sinisgalli C, Ostuni A, Armentano MF, Carmosino M, Milella L, Russo D, Labanca F and Khan H: Astaxanthin anticancer effects are mediated through multiple molecular mechanisms: A systematic review. Pharmacol Res. 155:1046892020. View Article : Google Scholar : PubMed/NCBI

30 

Chen YT, Kao CJ, Huang HY, Huang SY, Chen CY, Lin YS, Wen ZH and Wang HMD: Astaxanthin reduces MMP expressions, suppresses cancer cell migrations, and triggers apoptotic caspases of in vitro and in vivo models in melanoma. J Func Foods. 31:20–31. 2017. View Article : Google Scholar

31 

Kowshik J, Nivetha R, Ranjani S, Venkatesan P, Selvamuthukumar S, Veeravarmal V and Nagini S: Astaxanthin inhibits hallmarks of cancer by targeting the PI3K/NF-κΒ/STAT3 signalling axis in oral squamous cell carcinoma models. IUBMB Life. 71:1595–1610. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Kim SH, Lim JW and Kim H: Astaxanthin inhibits mitochondrial dysfunction and interleukin-8 expression in Helicobacter pylori-infected gastric epithelial cells. Nutrients. 10:13202018. View Article : Google Scholar : PubMed/NCBI

33 

Kim SH, Lim JW and Kim H: Astaxanthin prevents decreases in superoxide dismutase 2 level and superoxide dismutase activity in Helicobacter pylori-infected gastric epithelial cells. J Cancer Preven. 24:54–58. 2019. View Article : Google Scholar

34 

Martinkova E, Maglott A, Leger DA, Bonnet D, Stiborova M, Takeda K, Martin S and Dontenwill M: alpha5beta1 integrin antagonists reduce chemotherapy-induced premature senescence and facilitate apoptosis in human glioblastoma cells. Int J Cancer. 127:1240–1248. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Renner G, Noulet F, Mercier MC, Choulier L, Etienne-Selloum N, Gies JP, Lehmann M, Lelong-Rebel I, Martin S and Dontenwill M: Expression/activation of α5β1 integrin is linked to the β-catenin signaling pathway to drive migration in glioma cells. Oncotarget. 7:62194–62207. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Diaz MI, Valdivia A, Martinez P, Palacios JL, Harris P, Novales J, Garrido E, Valderrama D, Shilling C, Kirberg A, et al: Helicobacter pylori vacA s1a and s1b alleles from clinical isolates from different regions of Chile show a distinct geographic distribution. World J Gastroenterol. 11:6366–6372. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Leunk RD, Johnson PT, David BC, Kraft WG and Morgan DR: Cytotoxic activity in broth-culture filtrates of Campylobacter pylori. J Med Microbiol. 26:93–99. 1988. View Article : Google Scholar : PubMed/NCBI

38 

Chiozzi V, Mazzini G, Oldani A, Sciullo A, Ventura U, Romano M, Boquet P and Ricci V: Relationship between Vac A toxin and ammonia in Helicobacter pylori-induced apoptosis in human gastric epithelial cells. J Physiol Pharmacol. 60:23–30. 2009.PubMed/NCBI

39 

Atherton JC, Cao P, Peek RM Jr, Tummuru MK, Blaser MJ and Cover TL: Mosaicism in vacuolating cytotoxin alleles of Helicobacter pylori: Association of specific vacA types with cytotoxin production and peptic ulceration. J Biol Chem. 270:17771–17777. 1995. View Article : Google Scholar : PubMed/NCBI

40 

Atherton JC, Peek RM Jr, Tham KT, Cover TL and Blaser MJ: Clinical and pathological importance of heterogeneity in vacA, the vacuolating cytotoxin gene of Helicobacter pylori. Gastroenterology. 112:92–99. 1997. View Article : Google Scholar : PubMed/NCBI

41 

McClain MS, Cao P, Iwamoto H, Vinion-Dubiel AD, Szabo G, Shao Z and Cover TL: A 12-amino-acid segment, present in type s2 but not type s1 Helicobacter pylori VacA proteins, abolishes cytotoxin activity and alters membrane channel formation. J Bacteriol. 183:6499–6508. 2001. View Article : Google Scholar : PubMed/NCBI

42 

Telford JL, Ghiara P, Dell'Orco M, Comanducci M, Burroni D, Bugnoli M, Tecce MF, Censini S, Covacci A, Xiang Z, et al: Gene structure of the Helicobacter pylori cytotoxin and evidence of its key role in gastric disease. J Exp Med. 179:1653–1658. 1994. View Article : Google Scholar : PubMed/NCBI

43 

Seo JH, Lim JW, Kim H and Kim KH: Helicobacter pylori in a Korean isolate activates mitogen-activated protein kinases, AP-1, and NF-kappaB and induces chemokine expression in gastric epithelial AGS cells. Lab Invest. 84:49–62. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Xerry J and Owen RJ: Conservation and microdiversity of the phospholipase A (pldA) gene of Helicobacter pylori infecting dyspeptics from different countries. FEMS Immunol Med Microbiol. 32:17–25. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Su B, Johansson S, Fällman M, Patarroyo M, Granström M and Normark S: Signal transduction-mediated adherence and entry of Helicobacter pylori into cultured cells. Gastroenterology. 117:595–604. 1999. View Article : Google Scholar : PubMed/NCBI

46 

Miyata S, Koshikawa N, Yasumitsu H and Miyazaki K: Trypsin stimulates integrin alpha(5)beta(1)-dependent adhesion to fibronectin and proliferation of human gastric carcinoma cells through activation of proteinase-activated receptor-2. J Biol Chem. 275:4592–4598. 2000. View Article : Google Scholar : PubMed/NCBI

47 

Yeh YC, Cheng HC, Yang HB, Chang WL and Sheu BS: H. pylori CagL-Y58/E59 Prime Higher Integrin α5β1 in Adverse pH Condition to Enhance Hypochlorhydria Vicious Cycle for Gastric Carcinogenesis. PLoS One. 8:e727352013. View Article : Google Scholar : PubMed/NCBI

48 

Backert S, Ziska E, Brinkmann V, Zimny-Arndt U, Fauconnier A, Jungblut PR, Naumann M and Meyer TF: Translocation of the Helicobacter pylori CagA protein in gastric epithelial cells by a type IV secretion apparatus. Cell Microbiol. 2:155–164. 2000. View Article : Google Scholar : PubMed/NCBI

49 

Tsugawa H, Suzuki H, Saya H, Hatakeyama M, Hirayama T, Hirata K, Nagano O, Matsuzaki J and Hibi T: Reactive oxygen species-induced autophagic degradation of Helicobacter pylori CagA is specifically suppressed in cancer stem-like cells. Cell Host Microbe. 12:764–777. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Gu H, Huang T, Shen Y, Liu Y, Zhou F, Jin Y, Sattar H and Wei Y: Reactive oxygen species-mediated tumor microenvironment transformation: The mechanism of radioresistant gastric cancer. Oxid Med Cell Longev. 2018:58012092018. View Article : Google Scholar : PubMed/NCBI

51 

Liou GY and Storz P: Reactive oxygen species in cancer. Free Radic Res. 44:479–496. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Ding SZ, Minohara Y, Fan XJ, Wang J, Reyes VE, Patel J, Dirden-Kramer B, Boldogh I, Ernst PB and Crowe SE: Helicobacter pylori infection induces oxidative stress and programmed cell death in human gastric epithelial cells. Infect Immun. 75:4030–4039. 2007. View Article : Google Scholar : PubMed/NCBI

53 

Smoot DT, Elliott TB, Verspaget HW, Jones D, Allen CR, Vernon KG, Bremner T, Kidd LCR, Kim KS, Groupman JD and Ashktorab H: Influence of Helicobacter pylori on reactive oxygen-induced gastric epithelial cell injury. Carcinogenesis. 21:2091–2095. 2000. View Article : Google Scholar : PubMed/NCBI

54 

Shimoyama T, Fukuda S, Liu Q, Nakaji S, Fukuda Y and Sugawara K: Production of chemokines and reactive oxygen species by human neutrophils stimulated by Helicobacter pylori. Helicobacter. 7:170–174. 2002. View Article : Google Scholar : PubMed/NCBI

55 

Choi JH, Cho SO and Kim H: α-Lipoic acid inhibits expression of IL-8 by suppressing activation of MAPK, Jak/Stat, and NF-κB in H. pylori-infected gastric epithelial AGS cells. Yonsei Med J. 57:260–264. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Zhang Y, Zheng J, Zhou Z, Zhou H, Wang Y, Gong Z and Zhu J: Fractalkine promotes chemotaxis of bone marrow-derived mesenchymal stem cells towards ischemic brain lesions through Jak2 signaling and cytoskeletal reorganization. FEBS J. 282:891–903. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Kowshik J, Baba AB, Giri H, Deepak Reddy G, Dixit M and Nagini S: Astaxanthin inhibits JAK/STAT-3 signaling to abrogate cell proliferation, invasion and angiogenesis in a hamster model of oral cancer. PLoS One. 9:e1091142014. View Article : Google Scholar : PubMed/NCBI

58 

McCall B, McPartland CK, Moore R, Kamenetskii AF and Booth BW: Effects of astaxanthin on the proliferation and migration of breast cancer cells in vitro. Antioxidants (Basel). 7:1352018. View Article : Google Scholar : PubMed/NCBI

59 

Lee J, Lim JW and Kim H: Astaxanthin inhibits oxidative stress-induced Ku protein degradation and apoptosis in gastric epithelial cells. Nutrients. 14:39392022. View Article : Google Scholar : PubMed/NCBI

60 

Kim S, Lee H, Lim JW and Kim H: Astaxanthin induces NADPH oxidase activation and receptor-interacting protein kinase 1-mediated necroptosis in gastric cancer AGS cells. Mol Med Rep. 24:8372021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2023
Volume 27 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Woo J, Lim JW and Kim H: Astaxanthin inhibits integrin α5 expression by suppressing activation of JAK1/STAT3 in Helicobacter pylori‑stimulated gastric epithelial cells. Mol Med Rep 27: 127, 2023
APA
Woo, J., Lim, J.W., & Kim, H. (2023). Astaxanthin inhibits integrin α5 expression by suppressing activation of JAK1/STAT3 in Helicobacter pylori‑stimulated gastric epithelial cells. Molecular Medicine Reports, 27, 127. https://doi.org/10.3892/mmr.2023.13014
MLA
Woo, J., Lim, J. W., Kim, H."Astaxanthin inhibits integrin α5 expression by suppressing activation of JAK1/STAT3 in Helicobacter pylori‑stimulated gastric epithelial cells". Molecular Medicine Reports 27.6 (2023): 127.
Chicago
Woo, J., Lim, J. W., Kim, H."Astaxanthin inhibits integrin α5 expression by suppressing activation of JAK1/STAT3 in Helicobacter pylori‑stimulated gastric epithelial cells". Molecular Medicine Reports 27, no. 6 (2023): 127. https://doi.org/10.3892/mmr.2023.13014