International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Epithelial-to-mesenchymal transition (EMT) is a critical pathological feature in fibrosis-related diseases, including intestinal fibrosis and pulmonary fibrosis (1), and represents a highly dynamic, multi-stage process regulated by multiple signaling pathways that promote acquisition of the mesenchymal phenotype. In detail, external stimulation of signaling pathways, including TGFβ/Wnt, triggers a transcriptional program during EMT that induces the expression of the E-cadherin transcriptional repressor SNAIL, which promotes cell migration, invasiveness and fibrosis. The core process of EMT is characterized by epithelial cells losing polarity and intercellular connections to acquire a mesenchymal phenotype with enhanced mesenchymal traits such as collagen, fibronectin (FN), vimentin and α-smooth muscle actin (α-SMA) expression, as well as reduced epithelial adhesion protein E-cadherin expression (2-4). Proton pump inhibitors (PPIs) have been shown to have anticancer activity against several types of cancer. For instance, omeprazole destroyed cyclic AMP response element-binding protein (CREB)-binding protein (CBP)/p300-mediated SNAIL protein acetylation to induce its degradation, leading to the inhibition of EMT in cancer cells (5), while pantoprazole was shown to inactivate the Wnt/β-catenin signaling pathway to block the EMT process (6-8). Moreover, rabeprazole was shown to reduce resistance to temozolomide in glioma via EMT inhibition (9). Despite the fact that previous studies showed that PPIs can inhibit EMT, to date, there is no direct evidence fully describing the role of rabeprazole in fibrosis.
It is well known that SMAD family member 3 (SMAD3) is an important mediator of TGFβ-induced fibrosis or EMT, which can crosstalk with other pathways to modulate pathological progression. For example, Y-box binding protein 1, a member of the DNA/RNA-binding protein family, was reported to upregulate SMAD7 transcription or interact with SMAD3 to overcome the effect of TGFβ stimulation (10,11). In addition, transcriptional intermediary factor 1γ (TIF1γ; also known as TRIM33), a direct target of CREB, interacted with SMAD3 to antagonize SMAD3-induced fibrosis (12), which was in line with studies reporting on the anti-EMT function of TIF1γ (13,14). Notably, the phosphorylation of SMAD3 linker (at the ser204, ser208, ser213 and Thr179 residues) was reported to accelerate nuclear shuttle of cytoplasmic SMAD3, which decreased the accessibility of non-phosphorylated SMAD3 to membrane-anchored TGFβ receptor type I (TGFβRI), leading to inhibition of SMAD3C phosphorylation (15,16). Moreover, TIF1γ has been shown to act as a SMAD4 ubiquitin ligase, either dissociating SMAD2/SMAD3-SMAD4 complexes or mediating polyubiquitylation and degradation of SMAD4, thereby promoting competitive binding of SMAD2/3 through the MH2 domain (17). In addition, TIF1γ was reported to occupy SMAD-binding elements to prevent SMAD2/3-mediated DNA binding (12). Based on these findings, it is hypothesized that this TIF1γ/SMAD3 complex may have an inhibitory effect on SMAD3 phosphorylation and nuclear translocation to ablate SMAD3-mediated gene transcription. However, the critical phosphorylation site of SMAD3 in response to the TIF1γ/SMAD3 complex remain unclear.
Rabeprazole, a classical PPI, was previously recognized to be involved in the modulation of inflammation (18,19), barrier function (20,21) and metabolism (22). The aim of the present study was to identify the novel biological function of rabepraozole in fibrosis and reveal the possible relationship between TIF1γ and SMAD3 through immunofluorescence (IF), immunoprecipitation (IP) and luciferase assays. providing significant implications for understanding the function of rabeprazole.
Gibco™ BASIC DMEM (cat. no. C11995500BT) and fetal bovine serum (FBS) (Premium Plus; cat. no. A5669701) were purchased from Thermo Fisher Scientific, Inc. Rabeprazole (cat. no. HY-B0656) was purchased from MedChemExpress. Lipo8000™ transfection reagent (cat. no. C0533), nuclear and cytoplasmic protein extraction kit (cat. no. P0028) and cell lysis buffer for western blotting and immunoprecipitation (IP; cat. no. P0013) were purchased from Beyotime Institute of Biotechnology. shRNA-TIF1γ plasmids and TIF1γ promoter plasmid, were constructed and obtained from Youbio Biotech Co., Ltd. Protein A/G magnetic beads (cat. no. B23202) were purchased from Selleck Chemicals. EZ-press RNA Purification Kit (cat. no. B0004D), 4X EZscript Reverse Transcription Mix II (with gDNA Remover; cat. no. EZB-RT2G) and 2X Color SYBR Green qPCR Master Mix (cat. no. A0012) were purchased from EZBioscience. A dual-luciferase reporter assay system (cat. no. E1910) was obtained from Promega Corporation. Antibodies including α-SMA specific monoclonal antibody (mAb) (cat. no. 67735-1-Ig), FN mAb (cat. no. 66042-1-Ig), vimentin polyclonal antibody (pAb) (cat. no. 10366-1-AP), collagen type I (Col1a1) mAb (cat. no. 67288-1-Ig), SMAD3 mAb (cat. no. 66516-1-Ig), lamin A/C pAb (cat. no. 10298-1-AP) and α-tubulin mAb (cat. no. 66031-1-Ig) were purchased from Proteintech Group, Inc. TIF1γ mouse mAb (cat. no. YM1108), SMAD3 (phospho Ser204) rabbit pAb (cat. no. YP0363), SMAD3 (phospho Ser213) rabbit pAb (cat. no. YP0364), SMAD3 (phospho Thr179) rabbit pAb (cat. no. YP0745) and SMAD3 (phospho Ser208) rabbit pAb (cat. no. YP0746) were purchased from Immunoway Biotechnology Co., Ltd.; peroxidase affiniPure™ goat anti-rabbit IgG (H+L) (cat. no. 111-035-003) and peroxidase-conjugated affiniPure goat anti-mouse IgG (H+L) (cat. no. 115-035-003) were obtained from Jackson ImmunoResearch Laboratories, Inc.
Gastric epithelial cells, including AGS (cat. no. JNO-H0238) and GES-1 (cat. no. JNO-H0240), were purchased from Guangzhou Jennio Biotech Co. Ltd. and maintained in DMEM medium containing 10% FBS, 100 mg/ml streptomycin and 100 U/ml penicillin. Rabeprazole was dissolved in DMSO and stored at -80˚C. The concentration of rabeprazole used was 100 µM. For cell transfection of each well in a 6-well plate, when cells reached 50% confluence, 2 µg plasmids were gently mixed with 4 µl Lipo8000 transfection reagent in 125 µl Opti-MEM® medium at room temperature according to the manufacturer's instructions. Subseqently, the mix was added into each well for 48 h. pLVX-shRNA-PURO plasmids (cat. no. L28550-L28552) targeting TIF1γ (also known as TRIM33) were constructed and purchased from Youbio Biotech Co., Ltd.
Following treatment, total RNA was extracted from 106 cells per group with the EZ-press RNA purification kit according to the manufacturer's instructions. cDNA was synthesized using 4X EZscript reverse transcription mix II (with gDNA Remover) to analyze the indicated gene expression using the 2X color SYBR Green qPCR Master Mix. The following primer pairs were used for qPCR: FN forward, 5'-TCAGCTTCCTGGCACTTCTG-3' and reverse, 5'-TCTTGTCCTACATTCGGCGG-3'; vimentin forward, 5'-GGACCAGCTAACCAACGACA-3' and reverse, 5'-AAGGTCAAGACGTGCCAGAG-3'; COl1a1 forward, 5'-TCGGAGGAGAGTCAGGAAGG-3' and reverse, 5'-CCCGGTGACACATCAAGACA-3'; α-SMA forward, 5'-CTATGAGGGCTATGCCTTGCC-3' and reverse, 5'-GCTCAGCAGTAGTAACGAAGGA-3'; TIF1γ forward, 5'-AGCACTACTATACAGCAAGCGA-3' and reverse, 5'-CAGAAGGTGGGATCACAATGG-3'; β-actin forward, 5'-CTTCGCGGGCGACGAT-3' and reverse, 5'-CCACATAGGAATCCTTCTGACC-3'.
As described in Niu et al (23), after treatment, cells were harvested to extract the total protein using cell lysis buffer for western blotting and IP. Briefly, 15 µg total protein was separated by 10% SDS-PAGE and transferred into polyvinylidene fluoride (PVDF) membranes. The protein bands were blocked with PBST (0.5% Tween-20) (cat. no. BL345A) containing 2% skim milk (cat. no. BS102) plus 3% BSA (cat. no. BS114; all from Biosharp) for 1 h at room temperature. After washing with PBST (0.5% Tween-20), the membranes were incubated with the primary antibodies with a dilution of 1:2,000 overnight at 4˚C. Following incubation with primary antibodies, the membranes were incubated with the secondary antibodies at 1:2,000 for 1 h at room temperature. Finally, after washing with PBST, the bands were visualized and captured using Chemiluminescence imaging instrument (MiniChemi 610, Sinsage) after incubation with an ECL chemiluminescent substrate (cat. no. NEL105001EA; Revvity, Inc.). the density was quantified by Image J software (version: 1.8.0_351; National Institutes of Health) and normalized to internal control.
According to the manufacturer's instructions, after serum starvation and when the cells reached 80% confluence, cells were treated with or without rabeprazole for 1 h at 37˚C and the cytosolic and nuclear fractions were extracted using the nuclear and cytoplasmic protein extraction kit aforementioned. Western blotting was performed to analyze the protein expression.
As described in Li et al (24), when cells reached 80% confluence in a 6-cm dish, the medium was replaced with serum-free DMEM for 18 h. The cells were incubated with or without rabeprazole for 1 h, after which the medium was removed and the cells were washed with ice-cold PBS. The cells were then incubated with cell lysis buffer for western blotting and immunoprecipitation for 10 min on ice, scraped, and transferred into 1.5-ml tubes for an additional 30 min of lysis on a rotator at 4˚C. The lysates were centrifuged at 12,000 x g for 2 min at 4˚C to collect the supernatant. A total of 10 µl of anti-SMAD3 antibody was added to the supernatant and incubated overnight at 4˚C. Subsequently, 20 µl of protein A/G magnetic beads (cat. no. B23202; Selleck Chemicals) were added and incubated for 1 h at 4˚C to pull down the immune complexes. After three washes with cell lysis buffer for western blotting and IP (cat. no. P0013), the complexes were eluted with SDS loading buffer and boiled at 100˚C for 10 min for immunoblotting analysis.
Cells were digested and reseeded into an 8-well slide overnight and incubated at 37˚C with or without rabeprazole for 1 h. After fixation with 3% PFA (cat. no. BL3786A) for 10 min at room temperature and permeabilization with 0.3% Triton X-100 (cat. no. BL935B; both from Biosharp; Beijing Labgic Technology Co., Ltd.) for 5 min at 4˚C, cells were blocked with 3% milk at room temperature for 1 h. The incubation with anti-SMAD3 at a dilution of 1:400 was conducted overnight at 4˚C. Subsequently, further incubation was performed for another 1 h at room temperature with goat anti-mouse IgG (H+L) (AbFluor 594) (cat. no. RS3608; Immunoway Biotechnology Co., Ltd.) at a dilution of 1:300. After washing and staining the nuclei with DAPI buffer (cat. no. C1006; Beyotime Insitute of Biotechnology) for 5 min at room temperature, the coverslips were covered with glass slides. Stained cells were visualized using a laser scanning fluorescent microscope (Leica Microsystems GmbH).
Briefly, the reporter plasmid containing 0.5 µg TIF1γ promoter in PGL3-Basic (cat. no. 56089) and 10 ng pRL-TK Renilla luciferase plasmid (cat. no. V1079; both from Youbio Biotech Co., Ltd.) at a ratio of 50:1 were co-transfected into cells in a 24-well plate at room temperature. Cells were maintained for 24 h with complete medium in a cell incubator using Lipo8000 transfection reagent (C0533; Beyotime Institute of Biotechnology), followed by incubation with 100 µM rabeprazole for another 24 h in serum-free medium at 37˚C. An equal precentage of DMSO was used as the vehicle control. Each group consisted of three replicates. The cells were lysed with 100 µl Passive Lysis Buffer, and samples were collected and measured using a plate-reading luminometer to calculate relative light units determined by the ratio of firely/Renilla to reflect TIF1γ transactivation, using the dual-luciferase reporter assay system (cat. no. E1910; Promega Corporation) according to the manufacturer's instructions.
Data analysis was performed with GraphPad Prism 10.0 (Dotmatics). Statistical comparisons between two groups were performed using the unpaired Student's t-test. One-sample t-test was conducted to analyze the RT-q PCR results. Statistical comparisons among three groups were performed using one-way ANOVA followed by the Dunnett's post hoc test. P<0.05 was considered to indicate a statistically significant difference.
AGS and GES-1 gastric epithelial cells were employed as in vitro models to assess the impact of rabeprazole on fibrotic processes. The expression levels of genes involved in fibrosis were analyzed in gastric epithelial cells after incubation for 24 h with rabeprazole at 10 and 100 µM. Compared with the control group (Fig. 1A and B), a significant fibrosis inhibition was observed in GES-1 and AGS cells in response to 100 µM rabeprazole stimulation as indicated by the largely decreased FN and Col1a1 mRNA levels, whereas no significant change was observed in vimentin mRNA expression regardless of treatment with 10 and 100 µM. Moreover, western blotting was performed to detect the indicated protein expression levels, and quantification of the indicated proteins revealed that rabeprazole treatment at 100 µM induced a downregulation of FN and Col1a1 expression in GES-1 and AGS cells, respectively, while no influence on vimentin expression was observed with treatment at 10 µM. In addition, the expression of α-SMA was downregulated in GES-1 cells (Fig. 1C and D). Taken together, these findings indicated that rabeprazole has a potent suppressive effect in inhibiting fibrosis.
Since TIF1γ was reported to suppress extracellular matrix (ECM) production by inhibiting TGF-β1 transcriptional response (25-27), the present study focused on investigating the possible effect of rabeprazole on TIF1γ expression in gastric epithelial cells. Based on the previous results, treatment with 100 µM rabeprazole was selected for further experiments. Compared with the control group (Fig. 2A and B), the mRNA levels of TIF1γ were significantly increased in AGS and GES-1 cells in response to rabeprazole treatment. In addition, the protein expression level of TIF1γ was significantly enhanced by rabeprazole treatment (Fig. 2C). Moreover, dual luciferase reporter experiments further revealed that TIF1γ transactivation was significantly increased in response to rabeprazole treatment (Fig. 2D). Most importantly, depletion of TIF1γ expression by shRNA plasmid-targeted TIF1γ could rescue the effect of rabeprazole on FN and Col1a1 protein expression (Fig. 2E). These results indicated that rabeprazole inhibited the ECM through upregulation of TIF1γ expression.
It is well known that SMAD3 phosphorylation, whether in the linker domain or at the C-terminus, promotes SMAD3 nuclear translocation to initiate gene transcription (16). Notably, a reciprocal inhibition existed between phosphorylation of the SMAD3 linker domain and C-terminus, which was attributed to the fact that SMAD3 phosphorylation at the linker domain could induce SMAD3 nuclear shuttling, thereby inhibiting the accessibility of non-phosphorylated SMAD3 to membrane-anchored TGFβRI (28).
The present results demonstrated that rabeprazole attenuated fibrosis, prompting an exploration of the influence of rabeprazole on SMAD3 activation. Through immunoblotting quantification, it was found that, in gastric epithelial cells, rabeprazole treatment led to a downregulation of SMAD3 phosphorylation at Ser204, Ser208 and Ser213, while no significant changes were observed at Thr179 of the SMAD3 linker domain (Fig. 3A-E). Moreover, subcellular fraction combined with immunofluorescence analysis further showed that the nuclear SMAD3 level was reduced in response to rabeprazole (Fig. 3F). Overall, rabeprazole inhibited SMAD3 phosphorylation and nuclear translocation.
The present study aimed to elucidate the mechanism by which rabeprazole influences fibrotic processes. SMAD3 was identified as a key transcriptional regulator of Col1a1, contributing to fibrosis via TGFβ-dependent signaling pathways (29). Notably, TIF1γ was shown to mitigate ECM accumulation by interacting with SMAD3, thereby attenuating fibrotic responses (12). These findings suggest that SMAD3 may play a pivotal role in the antifibrotic effects mediated by rabeprazole-induced TIF1γ activity. On this basis, the present study aimed to explore the possible relationship between SMAD3 and TIF1γ. As shown in Fig. 4A, endogenous SMAD3 interacted with TIF1γ, and this interaction was enhanced in response to rabeprazole. In line with this, immunoprecipitation assays further revealed that rabeprazole treatment led to a significantly enhanced SMAD3/TIF1γ complex, which in turn further inhibited SMAD3 linker phosphorylation, including Ser204, Ser208 and Ser213 (Fig. 4A-E), consistent with our other research work showing that overexpression of TIF1γ led to inhibition of SMAD3 phosphorylation (Li et al unpublished data). These results indicated that rabeprazole treatment led to an antifibrotic TIF1γ/SMAD3 complex.
Fibrosis is a common pathological change in various diseases, including chronic hepatitis B-related liver diseases (30) and gastrointestinal-related diseases (31). Previous studies demonstrated that rabeprazole has various biological functions (9,29,32,33), however, the present study further revealed a novel mechanism of rabeprazole in fibrosis. The present study found that rabeprazole exerted an inhibitory effect on fibrosis, which was attributed to enhanced TIF1γ expression, while enhanced TIF1γ expression could inhibit SMAD3 linker phosphorylation. Further analysis showed that TIF1γ interacted with SMAD3, leading to suppression of SMAD3 signaling characterized by decreased phosphorylation of SMAD3 linker at Ser213, Ser204 and Ser208, and rabeprazole treatment enhanced this interaction between TIF1γ and SMAD3 to aggravate SMAD3 linker phosphorylation inhibition to alleviate fibrosis. This finding not only enriched the knowledge on the biological function of rabeprazole, but also the provided a possible therapeutic strategy for fibrosis.
Rabeprazole, a well-known PPI, was reported to induce M2-type adipose tissue macrophages, alleviating chronic inflammation (18), and to improve the survival rate and ameliorate pathological damage in Clostridium perfringens or perfringolysin O (PFO)-treated Galleria mellonella (34). Moreover, rabeprazole was shown to have anti-Trypanosoma cruzi activity by targeting cellular triosephosphate isomerase (35). Rabeprazole has been demonstrated to destroy gastric epithelial barrier function in vivo and in vitro (20,36), while exhibiting nephroprotective effects through inhibition of organic cation transporter 2(37) as well as promoting vascular impairment through hypoxia-inducible factor 1-α (38). The present study further investigated the antifibrotic function of rabeprazole. In previous studies, rabeprazole was demonstrated to induce enhanced TIF1γ expression, leading to suppression of TGFβ signaling. Notably, phosphorylation of TIF1γ at Tyr-524, Tyr-610, and Tyr-1048 was found to destroy the complex between TIF1γ and SMAD3, which could enhance TGFβ signaling (17,39). Therefore, further investigation was required to reveal the mechanism through which rabeprazole regulated TIF1γ expression. However, the influence of rabeprazole on TIF1γ phosphorylation remains unclear and will require further investigations. In addition, the present study lacked in vivo experiments confirming the possible role of rabeprazole in fibrosis. Furthermore, phosphorylation of SMAD3, at both the linker domain and C-terminus, is a critical event for the initiation of fibrosis or EMT (40). Phosphorylation of SMAD3 at Ser204, Ser208, and Ser213 was observed to be significantly reduced in response to rabeprazole stimulation, and this phenomenon was attributed to the formation of an enhanced complex between TIF1γ and SMAD3 caused by rabeprazole. However, the specific domain and phosphorylation status of TIF1γ that facilitate its interaction with SMAD3, particularly under rabeprazole treatment, remain unclear. In addition, considering that the disease and tumor microenvironment are enriched with diverse inflammatory and immune cells, including macrophages and fibroblasts, our future work will employ progressively complex in vitro systems, ranging from spheroid and organoid-based co-cultures to air-liquid interface organoids and microfluidic organoid-on-a-chip platforms. These models will be used to investigate whether rabeprazole plays a role in remodeling the cellular microenvironment (41,42).
In summary, the present study revealed a novel antifibrotic role of rabeprazole in vitro, mediated through the formation of a TIF1γ-SMAD3 complex. These findings offer novel insights into the biological functions of rabeprazole and suggest its potential as an alternative therapeutic strategy for fibrosis-related diseases.
Not applicable.
Funding: The present study was supported by the Science and Technology Project in the Field of Social Development of Zhuhai (grant. no. 2220004000296).
The data generated in the present study are included in the figures and/or tables of this article.
LL, XT and JC designed the experiments. LL, ZL and LF performed the experiments and data collection. LL, YC and XT analyzed the data and generated the figures. LL, XT and JC drafted the manuscript and revised the manuscript. LL and XT confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Luo L, Zhang W, You S, Cui X, Tu H, Yi Q, Wu J and Liu O: The role of epithelial cells in fibrosis: Mechanisms and treatment. Pharmacol Res. 202(107144)2024.PubMed/NCBI View Article : Google Scholar | |
|
Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Thiery JP, Acloque H, Huang RY and Nieto MA: Epithelial-mesenchymal transitions in development and disease. Cell. 139:871–890. 2009.PubMed/NCBI View Article : Google Scholar | |
|
Kalluri R and Weinberg RA: The basics of epithelial-mesenchymal transition. J Clin Invest. 119:1420–1428. 2009.PubMed/NCBI View Article : Google Scholar | |
|
Li Y, Ren BX, Li HM, Lu T, Fu R and Wu ZQ: Omeprazole suppresses aggressive cancer growth and metastasis in mice through promoting Snail degradation. Acta Pharmacol Sin. 43:1816–1828. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Zhang B, Ling T, Zhaxi P, Cao Y, Qian L, Zhao D, Kang W, Zhang W, Wang L, Xu G and Zou X: Proton pump inhibitor pantoprazole inhibits gastric cancer metastasis via suppression of telomerase reverse transcriptase gene expression. Cancer Lett. 452:23–30. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Feng S, Zheng Z, Feng L, Yang L, Chen Z, Lin Y, Gao Y and Chen Y: Proton pump inhibitor pantoprazole inhibits the proliferation, self-renewal and chemoresistance of gastric cancer stem cells via the EMT/β-catenin pathways. Oncol Rep. 36:3207–3214. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Zhang B, Yang Y, Shi X, Liao W, Chen M, Cheng AS, Yan H, Fang C, Zhang S, Xu G, et al: Proton pump inhibitor pantoprazole abrogates adriamycin-resistant gastric cancer cell invasiveness via suppression of Akt/GSK-β/β-catenin signaling and Epithelial-mesenchymal transition. Cancer Lett. 356:704–712. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Babu D, Mudiraj A, Yadav N, Y B V K C, Panigrahi M and Prakash Babu P: Rabeprazole has efficacy per se and reduces resistance to temozolomide in glioma via EMT inhibition. Cell Oncol (Dordr). 44:889–905. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Dooley S, Said HM, Gressner AM, Floege J, En-Nia A and Mertens PR: Y-box protein-1 is the crucial mediator of antifibrotic interferon-gamma effects. J Biol Chem. 281:1784–1795. 2006.PubMed/NCBI View Article : Google Scholar | |
|
Higashi K, Inagaki Y, Fujimori K, Nakao A, Kaneko H and Nakatsuka I: Interferon-gamma interferes with transforming growth factor-beta signaling through direct interaction of YB-1 with Smad3. J Biol Chem. 278:43470–43479. 2003.PubMed/NCBI View Article : Google Scholar | |
|
Lee EJ, Hwang I, Lee JY, Park JN, Kim KC, Kim I, Moon D, Park H, Lee SY, Kim HS, et al: Hepatic stellate cell-specific knockout of transcriptional intermediary factor 1γ aggravates liver fibrosis. J Exp Med. 217(e20190402)2020.PubMed/NCBI View Article : Google Scholar | |
|
Ikeuchi Y, Dadakhujaev S, Chandhoke AS, Huynh MA, Oldenborg A, Ikeuchi M, Deng L, Bennett EJ, Harper JW, Bonni A and Bonni S: TIF1γ protein regulates epithelial-mesenchymal transition by operating as a small ubiquitin-like modifier (SUMO) E3 ligase for the transcriptional regulator SnoN1. J Biol Chem. 289:25067–25078. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Su Z, Sun Z, Wang Z, Wang S, Wang Y, Jin E, Li C, Zhao J, Liu Z, Zhou Z, et al: TIF1γ inhibits lung adenocarcinoma EMT and metastasis by interacting with the TAF15/TBP complex. Cell Rep. 41(111513)2022.PubMed/NCBI View Article : Google Scholar | |
|
Matsuzaki K: Smad3 phosphoisoform-mediated signaling during sporadic human colorectal carcinogenesis. Histol Histopathol. 21:645–662. 2006.PubMed/NCBI View Article : Google Scholar | |
|
Ooshima A, Park J and Kim SJ: Phosphorylation status at Smad3 linker region modulates transforming growth factor-β-induced Epithelial-mesenchymal transition and cancer progression. Cancer Sci. 110:481–488. 2019.PubMed/NCBI View Article : Google Scholar | |
|
He W, Dorn DC, Erdjument-Bromage H, Tempst P, Moore MA and Massague J: Hematopoiesis controlled by distinct TIF1gamma and Smad4 branches of the TGFbeta pathway. Cell. 125:929–941. 2006.PubMed/NCBI View Article : Google Scholar | |
|
Li Y, Hao J, Kong X, Yuan W, Shen Y, Hui Z and Lu X: Rabeprazole mitigates obesity-induced chronic inflammation and insulin resistance associated with increased M2-type macrophage polarization. Biochim Biophys Acta Mol Basis Dis. 1870(167142)2024.PubMed/NCBI View Article : Google Scholar | |
|
Chen SQ, Hu BF, Yang YR, He Y, Yue L, Guo D, Wu TN, Feng XW, Li Q, Zhang W and Wen JG: The protective effect of rabeprazole on cisplatin-induced apoptosis and necroptosis of renal proximal tubular cells. Biochem Biophys Res Commun. 612:91–98. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Yang F, Li L, Zhou Y, Pan W, Liang X, Huang L, Huang J, Cheng Y, Geng L, Xu W and Gong S: Rabeprazole destroyed gastric epithelial barrier function through FOXF1/STAT3-mediated ZO-1 expression. Clin Exp Pharmacol Physiol. 50:516–526. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Son M, Park IS, Kim S, Ma HW, Kim JH, Kim TI, Kim WH, Han J, Kim SW and Cheon JH: Novel Potassium-competitive acid blocker, tegoprazan, protects against colitis by improving gut barrier function. Front Immunol. 13(870817)2022.PubMed/NCBI View Article : Google Scholar | |
|
Zhou Y, Chen S, Yang F, Zhang Y, Xiong L, Zhao J, Huang L, Chen P, Ren L, Li H, et al: Rabeprazole suppresses cell proliferation in gastric epithelial cells by targeting STAT3-mediated glycolysis. Biochem Pharmacol. 188(114525)2021.PubMed/NCBI View Article : Google Scholar | |
|
Niu R, Lan J, Liang D, Xiang L, Wu J, Zhang X, Li Z, Chen H, Geng L, Xu W, et al: GZMA suppressed GPX4-mediated ferroptosis to improve intestinal mucosal barrier function in inflammatory bowel disease. Cell Commun Signal. 22(474)2024.PubMed/NCBI View Article : Google Scholar | |
|
Li P, Wu Y, Deng Z, Samad A, Xi Y, Song J, Zhang Y, Li J, Zhou YA, Xiong Q and Wu C: Two novel SH3TC2 mutations predispose to Charcot-Marie-Tooth disease type 4C by mistargeting away from TFRC. Cell Signal. 130(111669)2025.PubMed/NCBI View Article : Google Scholar | |
|
Hesling C, Fattet L, Teyre G, Jury D, Gonzalo P, Lopez J, Vanbelle C, Morel AP, Gillet G, Mikaelian I and Rimokh R: Antagonistic regulation of EMT by TIF1γ and Smad4 in mammary epithelial cells. EMBO Rep. 12:665–672. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Qi G, Lu G, Yu J, Zhao Y, Wang C, Zhang H and Xia Q: Up-regulation of TIF1γ by valproic acid inhibits the epithelial mesenchymal transition in prostate carcinoma through TGF-β/Smad signaling pathway. Eur J Pharmacol. 860(172551)2019.PubMed/NCBI View Article : Google Scholar | |
|
Yin X, Xu C, Zheng X, Yuan H, Liu M, Qiu Y and Chen J: SnoN suppresses TGF-β-induced epithelial-mesenchymal transition and invasion of bladder cancer in a TIF1γ-dependent manner. Oncol Rep. 36:1535–1541. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Sun YM, Wu Y, Li GX, Liang HF, Yong TY, Li Z, Zhang B, Chen XP, Jin GN and Ding ZY: TGF-β downstream of Smad3 and MAPK signaling antagonistically regulate the viability and partial epithelial-mesenchymal transition of liver progenitor cells. Aging (Albany NY). 16:6588–6612. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Zheng M, Li H, Sun L, Cui S, Zhang W, Gao Y and Gao R: Calcipotriol abrogates TGF-β1/pSmad3-mediated collagen 1 synthesis in pancreatic stellate cells by downregulating RUNX1. Toxicol Appl Pharmacol. 491(117078)2024.PubMed/NCBI View Article : Google Scholar | |
|
Xiao J, Liu H, Yao J, Yang S, Shen F, Bu K, Wang Z, Liu F, Xia N and Yuan Q: The characterization of serum proteomics and metabolomics across the cancer trajectory in chronic hepatitis B-related liver diseases. View: 5, 2024. | |
|
Ito T and Kayama H: Roles of fibroblasts in the pathogenesis of inflammatory bowel diseases and IBD-associated fibrosis. Int Immunol. 37:377–392. 2025.PubMed/NCBI View Article : Google Scholar | |
|
Xie J, Liang X, Xie F, Huang C, Lin Z, Xie S, Yang F, Zheng F, Geng L, Xu W, et al: Rabeprazole suppressed gastric intestinal metaplasia through activation of GPX4-mediated ferroptosis. Front Pharmacol. 15(1409001)2024.PubMed/NCBI View Article : Google Scholar | |
|
Gu M, Zhang Y, Zhou X, Ma H, Yao H and Ji F: Rabeprazole exhibits antiproliferative effects on human gastric cancer cell lines. Oncol Lett. 8:1739–1744. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Wang G, Liu Y, Deng L, Liu H, Deng X, Li Q, Feng H, Guo Z and Qiu J: Repurposing rabeprazole sodium as an anti-Clostridium perfringens drug by inhibiting perfringolysin O. J Appl Microbiol. 134:2023.PubMed/NCBI View Article : Google Scholar | |
|
Garcia-Torres I, De la Mora-De la Mora I, Lopez-Velazquez G, Cabrera N, Flores-López LA, Becker I, Herrera-López J, Hernández R, Pérez-Montfort R and Enríquez-Flores S: Repurposing of rabeprazole as an anti-Trypanosoma cruzi drug that targets cellular triosephosphate isomerase. J Enzyme Inhib Med Chem. 38(2231169)2023.PubMed/NCBI View Article : Google Scholar | |
|
Takashima S, Tanaka F, Kawaguchi Y, Usui Y, Fujimoto K, Nadatani Y, Otani K, Hosomi S, Nagami Y, Kamata N, et al: Proton pump inhibitors enhance intestinal permeability via dysbiosis of gut microbiota under stressed conditions in mice. Neurogastroenterol Motil. 32(e13841)2020.PubMed/NCBI View Article : Google Scholar | |
|
Sharaf G, E ME and El-Sayed EK: Augmented nephroprotective effect of liraglutide and rabeprazole via inhibition of OCT2 transporter in cisplatin-induced nephrotoxicity in rats. Life Sci. 321(121609)2023.PubMed/NCBI View Article : Google Scholar | |
|
Evans CE, Peng Y, Zhu MM, Dai Z, Zhang X and Zhao YY: Rabeprazole promotes vascular repair and resolution of Sepsis-induced inflammatory lung injury through HIF-1α. Cells. 11(1425)2022.PubMed/NCBI View Article : Google Scholar | |
|
Yuki R, Tatewaki T and Yamaguchi N, Aoyama K, Honda T, Kubota S, Morii M, Manabe I, Kuga T, Tomonaga T and Yamaguchi N: Desuppression of TGF-β signaling via nuclear c-Abl-mediated phosphorylation of TIF1γ/TRIM33 at Tyr-524, -610, and -1048. Oncogene. 38:637–655. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Matsuzaki K: Smad phospho-isoforms direct context-dependent TGF-β signaling. Cytokine Growth Factor Rev. 24:385–399. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Zhou PZ, Gao L, Wang LW, Zhang YF, Song WL and Hao YX: Clinical observation of magnesium aluminum carbonate combined with rabeprazole-based triple therapy in the treatment of helicobacter pylori-positive gastric ulcer associated with hemorrhage. Pak J Med Sci. 38:1271–1277. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Yuan K, Du X, Dong L, Pan J and Xue W: Modelling the tumor microenvironment in vitro in prostate cancer: Current and future perspectives. VIEW:. 5(20240074)2024. |