Open Access

Downregulation of miR‑7 and miR‑153 is involved in Helicobacter pylori CagA induced gastric carcinogenesis and progression

  • Authors:
    • Yu Song
    • Dong Guo
    • Jia-Fei Liu
    • Li-Na Ge
    • Peng Liu
    • Ye-Min Qu
    • Hai-Yan Cong
    • Tian Li
    • Xin Chang
    • Yi-Ran Wang
    • Li-Ying Shao
    • Zong-Jun Du
    • Ming-Yi Wang
  • View Affiliations

  • Published online on: May 24, 2023     https://doi.org/10.3892/ijo.2023.5527
  • Article Number: 79
  • Copyright: © Song 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

Helicobacter pylori (H. pylori) infection plays a pivotal role in the development of gastric cancer (GC). However, the association between aberrant microRNAs (miRNAs/miRs) expression and H. pylori‑induced GC remains poorly understood. The present study reported that repeated infection of H. pylori caused the oncogenicity of GES‑1 cells in BALB/c Nude mice. miRNA sequencing revealed that both miR‑7 and miR‑153 were significantly decreased in the cytotoxin‑associated gene A (CagA) positive GC tissues and this was further confirmed in a chronic infection model of GES‑1/HP cells. Further biological function experiments and in vivo experiments validated that miR‑7 and miR‑153 can promote apoptosis and autophagy, inhibit proliferation and inflammatory response in GES‑1/HP cells. All the associations between miR‑7/miR‑153 and their potential targets were revealed via bioinformatics prediction and dual‑luciferase reporter assay. Particularly, downregulation of both miR‑7 and miR‑153 obtained an improved sensitivity and specificity in diagnosing H. pylori (CagA+)‑induced GC. The present study identified that the combination of miR‑7 and miR‑153 may be regarded as novel therapeutic targets in H. pylori CagA (+)‑associated GC.

Introduction

Gastric carcinoma (GC) is the third commonest cause of cancer death globally (1). Multiple risk factors, including gene polymorphism, dietary habits and smoking, have been reported as key factors in GC. However, H. pylori infection is known as the main risk, accounting for 90% of new cases of non-cardia gastric cancer (2). Growing evidence shows that H. pylori virulence factors have important roles in the carcinogenesis pattern 'chronic gastritis-gastric mucosa atrophy-intestinal metaplasia-dysplasia-gastric cancer' (3-5). Particularly, CagA is demonstrated to be the critical risk factor for chronic infection (6,7). It can interact with a variety of intracellular proteins, perturb multiple intracellular signaling pathways and promote the malignant transformation of the host cells (6). However, the molecular mechanism of H. pylori (CagA+)-induced GC is still not fully understood.

microRNAs (miRNAs/miRs) serve crucial roles in a variety of biological processes, including tumorigenesis and progression (8). Aberrant regulation of miRNAs can function as oncogenes or anti-oncogenes by binding with its specific target mRNAs, resulting in mRNA degradation or/and translational repression (9). Studies have revealed the regulatory roles of miRNAs in H. pylori-induced inflammation and carcinogenesis (10-13).

As an ancient miRNA, miR-7 is involved in the fine-tuning of several signaling pathways, serving primarily as a tumor suppressor in various cancers (14). It can suppress tumor metastasis via neuro-oncological ventral antigen 2 (NOVA2) in non-small cell lung cancer (15), suppress the progression of pancreatic cancer via mitogen-activated protein kinase kinase 9 (MAP3K9) (16) and reduce breast cancer stem cell metastasis by suppressing RELA proto-oncogene, NF-κB subunit (RELA) and decreasing the expression of endothelial cell-selective adhesion molecule (ESAM) (17). miR-7 is downregulated in GC (18); it can inhibit proliferation and angiogenesis and promotes apoptosis of GC cells via Raf-1 (19), increase cisplatin sensitivity of GC cells through suppressing mTOR (20), inhibit the invasion and metastasis of GC cells through inhibitions of EGFR (21) and insulin-like growth factor-1 receptor (IGF1R) (22). Particularly, it can form a regulatory feedback circuit with NF-κB (23) and act as a potential therapeutic target for aberrant NF-κB-driven distant metastasis of GC (24).

miR-153 exerts opposite functions as a tumor promoter or suppressor in different types of cancer. It can improve sensitivity to gefitinib by regulating the expression of ABCE1 in lung cancer (25), inhibit triple-negative breast cancer by targeting zine finger E-box-binding homeobox2 (ZEB2)-induced epithelial-mesenchymal transition (EMT) (26), regulate the progression of ovarian carcinoma by targeting myeloid cell leukemia 1 (MCL1) (27) and decrease tryptophan catabolism and inhibit angiogenesis via the repression of indoleamine 2,3-dioxygenase 1 in bladder cancer (28). Meanwhile, high expression of miR-153 is relevant for a poor prognosis for prostate cancer patients (29). miR-153 also increases invasion and therapeutic resistance by pleiotropic effects in colorectal cancer progression (30). Nevertheless, relevant studies of miR-153 in GC are rare. miR-153 acts as a tumor suppressor by aiming at Krüppel-like factor 5 (KLF5) in GC (31).

To date, the synergistic roles of miRNAs have not been well elucidated in GC. In the present study, an integrated approach was adopted and identified that miR-7 and miR-153 were involved in H. pylori-induced gastric carcinogenesis and progression. The present study systematically validated that simultaneous overexpression of miR-7 and miR-153 significantly promoted apoptosis, inhibited proliferation and inflammatory response by multiple targets. In addition, it was discovered that a combination of miR-7 and miR-153 might serve as novel diagnostic and therapeutic targets in H. pylori CagA (+)-associated GC.

Materials and methods

Cell lines, bacterial strains and culture

Human normal gastric epithelial cell line GES-1(RRID:CVCL_EQ22) and human GC cell lines MKN-45 (RRID:CVCL_0434), MKN-28 (RRID:CVCL_1416), HGC-27 (RRID:CVCL_1279), SNU-1 (RRID:CVCL_0099) and AGS (RRID:CVCL_0139) were purchased from the BeNa Culture Collection. GC cell line NCL-87 was purchased from the National Collection of Authenticated Cell Cultures. Cell lines were cultured in RPMI-1640 medium (HyClone; Cytiva) and maintained in a humidified atmosphere containing 5% CO2 at 37°C. All cell cultures were supplemented with 10% fetal bovine serum (FBS; Biological Industries), 100 IU/ml penicillin and 100 µg/ml streptomycin (Gibco, Thermo Fisher Scientific, Inc.).

H. pylori clinical isolate SBK was isolated from a gastric ulcer patient and preserved in Central Lab of Weihai Municipal Hospital. CagA-knockout H. pylori SBK was established according to the previously described method (32). H. pylori strain ATCC 26695 and its corresponding CagA-knockout H. pylori (named 0547) were kindly provided by Professor Boqing Li of Binzhou Medical University (Yantai, China). All strains were maintained in Colombian agar plates supplemented with 8% sheep blood under a microaerophilic environment.

Cell chronic infection model

GES-1 cells were co-cultured with H. pylori strain ATCC 26695 or isolated SBK at a multiplicity of infection (MOI) of 100:1 to construct the H. pylori chronic infection model, as previously described (33). However, only the SBK-treated GES-1 survived and was termed GES-1/HP. GES-1cells without exposure to H. pylori underwent the same procedure and were designated as control cells.

Construction and sequencing of miRNA libraries

High-throughput sequencing was performed to identify miRNAs as follows: steps. Total RNA isolated from H. pylori-infected GC tissues and paired adjacent normal tissues were fractionated with a 15% native polyacrylamide gel (Invitrogen; Thermo Fisher Scientific, Inc.) and small RNAs of 18-30 nt were purified. Small RNAs were reverse transcribed into cDNA and further amplified. The sequence of the amplified products was performed on the Illumina HiSeq 2500 platform (Guangzhou RiboBio Co., Ltd.).

miRNA/RNA extraction and reverse transcription-quantitative (RT-q) PCR

Total miRNAs or RNA were extracted from 1×107 cells with RNAiso for Small RNA or RNAiso Plus (Takara Bio, Inc.) according to the manufacturer's protocol, and the concentrations were measured with the NanoDrop ND-1000 spectrophotometer (Thermo Fisher Scientific, Inc.). Then, 1-3 µg miRNA or RNA was reverse transcribed. RT-qPCR was performed using TB Green Premix Ex Taq II (Takara Biotechnology Co., Ltd.) on the 7500 Real-Time PCR instrument (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. U6 small nuclear RNA and β-actin were used as an internal control for miRNA and mRNA assays, respectively. Specific primers used for SP1, Bcl-xL, Bcl2, β-actin, miR-7, miR-153 and U6 were as follows: 5′-CCT CAG TGC ATT GGG TAC TTC (forward) and 5′-ACC AAG CTG AGC TCC ATG AT (reverse) for SP1; 5′-GAG CTG GTG GTT GAC TTT CTC (forward) and 5′-TCC ATC TCC GAT TCA GTC CCT (reverse) for Bcl-xL; 5′-GGT GGG GTC ATG TGT GTG G (forward) and 5′-CGG TTC AGG TAC TCA GTC ATC C (reverse) for Bcl2; 5′-CAT GTA CGT TGC TAT CCA GGC (forward) and 5′CTC CTT AAT GTC ACG CAC GAT-(reverse) for β-actin; 5′-CCA CGT TGG AAG ACT AGT GAT TT (forward) and 5′TAT GGT TTT GAC GAC TGT GTG AT-(reverse) for miR-7; 5′-AAC GAA CTT GCA TAG TCA CAA AAG (forward) and 5′TAT GGT TTT GAC GAC TGT GTG AT-(reverse) for miR-153; 5′-CAG CAC ATA TAC TAA AAT TGG AAC G (forward) and 5′ACG AAT TTG CGT GTC ATC C-(reverse) for U6. Fold changes of target miRNAs or genes were calculated by the 2−ΔΔCq method from triplicate experiments (34).

Western blotting

Cells or tissue samples were collected and lysed with T-PER (Thermo Fisher, Inc.) containing protease inhibitor and phosphatase inhibitor cocktail (Biotool, LLC), then incubated 30 min on ice and centrifuged at 13,700 × g at 4°C for 15 min. The supernatants were collected carefully and determined by BCA protein concentration assay kit (Beyotime Institute of Biotechnology). Total proteins (30 µg) were separated by 10% SDS-PAGE. Gels were transferred onto PVDF membranes by wet transfer. Membranes were blocked in 5% BSA in TBST (20 mM Tris-HCl, 150 mM NaCl, 0.05% Tween-20, pH 8.0) at 37°C for 2 h, incubated with primary antibodies to Bax (1:1,000; cat. no. 5023), Bcl-xL (1:1,000; cat. no. 2764), Bcl-2 (1:1,000; cat. no. 15071), Cleaved Caspase-3 (1:1,000; cat. no. 9661), phosphorylated (p)-AKT (cat. no. 4060), IL-1β (1:1,000; cat. no. 12703S) and AKT (1:1,000; cat. no. 4685; Cell Signaling Technology, Inc.); P53 (1:1,000; cat. no. 60283-2-Ig), P27 (1:1,000; cat. no. 25614-1-AP), SP1 (1:1,000; cat. no. 21962-1-AP), Cyclin A2 (1:1,000; cat. no. 66391-1-Ig), Cyclin E2 (1:1,000; cat. no. 11935-1-AP), Toll-like receptor 4 (TLR4; 1:1,000; cat. no. 19811-1-AP), Krüppel-like factor (KLF)5 (1:1,000; cat. no. 21017-1-AP; Proteintech Group, Inc.) and Ki-67 (1:1,000; cat. no. AF1738) and NF-κB p65 (1:1,000; cat. no. AF0246; Beyotime Institute of Biotechnology) overnight at 4°C, and then incubated with appropriate HRP-conjugated secondary antibody (1:5,000; cat. nos. A0208 or A0216; Beyotime) at room temperature for 30 min. Finally, the specific bands were visualized using super sensitive ECL chemiluminescence detection reagent (Tiangen Biotech Co., Ltd.) and imaged with Chemidoc XRS+ (Bio-Rad Laboratories, Inc.).

Cell apoptosis assay

This was performed according to the manufacturer's protocol as described previously (33). Briefly, cells were harvested, resuspended and labelled using an Annexin V-FITC apoptosis detection kit (BD Biosciences). Then, cells were analyzed by flow cytometer BD FACSaria II (BD Biosciences), and the total percentage of early + late apoptotic cells were processed for the apoptotic rate using the CellQuest Pro software v8.0.1 (BD Biosciences).

EdU assay

This was performed in accordance with the manufacturer's protocol using the BeyoClick EdU Cell Proliferation kit with Alexa Fluor 488 (Beyotime Institute of Biotechnology). In brief, cells were incubated with 10 µM EdU reagent at 37°C for 2 h, fixed with 4% paraformaldehyde for 15 min and permeated with 0.3% Triton X-100 for another 15 min at room temperature. The cells were incubated with the Click Reaction Mixture at room temperature for 30 min and then incubated with Hoechst 33342 for 10 min in a dark place. The ratio of EdU-positive cells to the total number of Hoechst 33342-positive cells was calculated as the cell proliferation rate.

Cell transfection

GES-1/HP cells and HGC-27 cells under H. pylori infection (5×105 cells/well) were transfected with miR-7-5p mimics (50 nmol; cat. no. miR1181031074037-1-5), miR-153-3p mimics (50 nmol; cat. no. miR10000439-1-5), combination of miR-7-5p (25 nmol) and miR-153-3p mimics (25 nmol), mimics negative control (NC, 50 nmol; cat. no. miR1N0000001-1-5; Guangzhou RiboBio Co., Ltd.), SP1 short interfering (si)RNA (cat. no. A10001) and negative control siRNA (cat. no. A06001; Shanghai GenePharma Co., Ltd.) in 6-well plates using the Lipofectamine 2000® (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol at 37°C for 6 h. The sequences of validated siRNA for SP1 were: 5′-CCA GCA ACA UGG GAA UUA UTT (forward) and 5′-AUA AUU CCC AUG UUG CUG GTT (reverse) for si-1; 5′-GCC GUU GGC UAU AGC AAA UTT (forward) and 5′-AUU UGC UAU AGC CTT AAC GGC TT (reverse) for si-2; 5′-GCC CUU AUU ACC ACC AAU ATT (forward) and 5′-UAU UGGUGG UAA UAA GGG CTT (reverse) for si-3; the sequences of negative control were 5′-UUC UCC GAA CGU GUC ACG UTT (forward) and 5′-ACG UGA CAC GUU CGG AGA ATT (reverse). The time interval between transfection and subsequent experimentation was 48 h.

Dual-luciferase reporter assay

Bcl-xL-3′-UTR/wild-type (WT), Bcl-xL-3′-UTR/mutant (MUT), Bcl2-3′-UTR/WT and Bcl2-3′-UTR/Mut were cloned and embedded into the psiCHECK-2 plasmid. Cells were co-transfected with either WT or MUT luciferase reporter vector and either mimic miRNAs or negative control using Lipofectamine 2000® (Invitrogen; Thermo Fisher Scientific, Inc.), respectively. After 48 h transfection, cells were treated using Dual-Luciferase Reporter Gene Assay kit (Beyotime Institute of Biotechnology). Dual fluorescent signals were detected with a luminometer (GloMax Navigator; Promega Corporation) and each value for Renilla luciferase activity was normalized by firefly luciferase activity.

Cell cycle assay

The cell cycle assay was conducted using a cell cycle and apoptosis kit (Beyotime Institute of Biotechnology) according to the manufacturer's protocol. Cells transfected with miR-7, miR-153 or miR-7 and miR-153 together at 37°C for 48 h were digested and stained with propidium iodide at 37°C for 30 min in the dark. Then, the cell DNA content was determined by the flow cytometer BD FACSaria II (BD Biosciences), and the cell cycle was analyzed using the FlowJo software (v10.8.2; FlowJo LLC).

Fluorescence in situ hybridization (FISH) analysis

A FAM-labeled Locked Nucleic Acid (LNA) probe for detecting miR-7 and a Cy3-labeled LNA probe for detecting miR-153 were synthesized and performed on paraffin sections using a fluorescence in situ hybridization kit (Shanghai GenePharma Co., Ltd.). In brief, firstly, the dissected tissues were fixed with 10% formalin for 48 h at room temperature. After rinsing the tissues with running tap water and dehydrating the tissues in EtOH baths at 70, 95 and 100% ethanol, successively, the tissues were incubated in a 65°C paraffin bath twice, 30 min each. Then, the paraffin-embedded tissues were sectioned at 4 µm. Subsequently, the paraffin sections were dewaxed, rehydrated, digested with proteinase K at 37°C for 20 min, and dehydrated through a graded series of alcohol. Then, the sections were hybridized with the probes directed against miR-7 and miR-153 at 37°C for overnight in wet box, and further counterstained with 4,6-diamidino-2-phenylindole (DAPI) at room temperature for 10 min. Images were captured using fluorescence microscopy (Nikon Corporation).

Immunohistochemical (IHC) analysis

Paraffin sections were made from H. pylori-infected GC tissues, adjacent normal tissues and xenograft tumor samples according the following steps: First, the dissected tissues were fixed with 10% formalin for 48 h at room temperature. After rinsing the tissues with running tap water and dehydrating the tissues in EtOH baths in the 70, 95 and 100% ethanol, successively, the tissues were incubated in a 65°C paraffin bath twice, 30 min each. Then, the paraffin-embedded tissues were sectioned at 4 µm. The paraffin sections were dewaxed and underwent antigen retrieval with citrate buffer. The slides were blocked with 0.1% BSA and incubated overnight with the following antibodies: Bcl-xL (1:50; cat. no. 2764; Cell Signaling Technology, Inc.), Bcl-2 (1:50; cat. no. 15071; Cell Signaling Technology, Inc.), Ki-67 (1:100; cat. no. AF1738; Beyotime Institute of Biotechnology), SP1 (1:50; cat. no. 21962-1-AP; Proteintech Group, Inc.) or P53 (1:100; cat. no. 60283-2-Ig) at 4°C overnight, and then incubated with HRP-conjugated secondary antibodies (1:50; Beyotime Institute of Biotechnology) at 37°C for 1 h. Finally, the target proteins were visualized on-site with the 3,3′-diaminobenzidine substrate (Beyotime Institute of Biotechnology) and images captured using fluorescence microscopy (Nikon Corporation).

Chromatin immunoprecipitation (ChIP)

ChIP assay was conducted following the instructions of the ChIP Assay kit (Beyotime Institute of Biotechnology). Briefly, cultured cells (1×107 cells in 10 cm dish) were fixed with 1% formaldehyde at 37°C for 10 min, and then neutralized with 125 mM glycine solution (1.1 ml) at room temperature for 5 min. Chromatin solutions were sonicated and incubated with anti-SP1 antibody (1:100) or control IgG (1:100) overnight with rotation at 4°C. DNA-protein cross-links were disassembled and chromatin DNA was purified and subjected to PCR analysis with the primers: 5′-CCG CTC GAG CGG CCA GGT CGG CGA GAA TCC T-3′ and 5′-CCC AAG CTT GGG TTA GCG CCA GTC TTG AGC ACA T-3′ for SP1, 5′-TAC TAG CGG TTT TAC GGG CG-3′ and 5′-TCG AAC AGG AGG AGC AGA GAG CGA-3′ for GAPDH as a negative control. DNA isolated from total nuclear extract was used the PCR input control, and the PCR products were analyzed by gel electrophoresis using a 2% agarose gel. PrimeSTAR HS DNA Polymerase (Takara Bio Inc.) was used for the PCR process. Thermocycling conditions was initial denaturation: 98°C for 5 min; 30 cycles of denaturation (98°C for 10 sec), annealing (60°C for 15 sec) and elongation (72°C for 15 sec).

Nude mouse tumor xenograft model

A total of 20 Balb/c male nude mice, 4-6 weeks old, weighing 20-25 g, were obtained from Weihai Desheng Technology Testing Co., Ltd. and housed in standard pathogen-free conditions environment with a temperature of 22±1°C, relative humidity of 50±1% and a 12-h light/dark cycle. All animal studies (including the mouse euthanasia procedure) were approved by the Ethics Committee of Weihai Municipal Hospital (approval no. 20220103) and conducted in compliance with the regulations and guidelines of Weihai Municipal Hospital institutional animal care and conducted according to the AAALAC and the IACUC guidelines.

To establish a xenograft model, 1×107 HGC-27 cells were suspended in 100 µl PBS and inoculated subcutaneously into the right hind flank. After 2 weeks, the transplanted nude mice were randomly divided into three groups (n=5/group): i) MicrON hsa-miR-7 (cat. no. miR4180703093100-4-5; Guangzhou RiboBio Co., Ltd.), ii) micrON hsa-miR-153 (cat. no. miR40000439; Guangzhou RiboBio Co., Ltd.) and iii) MicrON hsa-miR-7 and micrON hsa-miR-153 together were injected into the unilateral implanted tumor at the dose of 5 nmol (in 20 µl PBS) per mouse under anesthesia with isoflurane inhalation (2-3% induction, 1-2% maintenance). The implanted tumor on the other side was injected with micrON agomir NC#22 (cat. no. miR4N0000001-2-5; Guangzhou RiboBio Co., Ltd.) as NC. Tumor volume were calculated as π/6 × width2 × length every 4 days for 5 times. After 6 weeks, mice were euthanized by cervical dislocation under anesthesia (1% sodium pentobarbital intraperitoneal injection, 30 mg/kg) and tumor tissues were excised, measured, weighed and used for IHC and FISH. The mice were euthanized when reaching the following humane endpoints: i) Tumor burden was >10% of body weight and the diameter of tumor was >20 mm; ii) tumor ulceration, infection or necrosis was observed; iii) tumor interferes with eating or walking; iv) rapid weight loss (loss of 15-20% of original weight). Notably, none of these endpoints were reached during the experiments. Animal health and behavior were monitored twice a day (at the beginning and end of the day) by an experienced individual. A comprehensive judgment on death was made by observing cessation of respiration, heartbeat and pupil and nerve reflexes.

IL-1β levels quantification (ELISA)

IL-1β levels in GES-1 cells treated with or without H. pylori infection and biopsy specimens obtained from the antrum of gastric patients with H. pylori infection and matched normal tissues were determined in duplicated samples by an ELISA (cat. no. PI305; Beyotime Institute of Biotechnology). Briefly, biopsy samples were immediately placed in 2 ml of PBS (pH 7.4), frozen on dry ice, homogenized and centrifuged (10,000 × g, 4°C for 10 min). Then, total protein in supernatants or cell culture supernatants was measured and IL-1β levels were analyzed according to the manufacturer's instructions.

Patient samples

All blood and paired samples of H. pylori-infected GC tissues and adjacent normal tissues were obtained from Weihai Municipal Hospital. All samples were clinically and pathologically confirmed.

Tissue microarray analysis

GC tissue microarrays (TMAs) consisting of 24 cases of gastric disease (including six cases of GC tissue with matched adjacent normal gastric tissue) were obtained from Shanghai Outdo Biotech Co., Ltd. FISH probes and specific antibodies or were applied to slides followed the protocol of IHC and FISH described previously. Slides were scanned with a confocal scanner and images captured under fluorescence microscopy (Nikon Corporation). The staining intensity of TMAs was evaluated and scored using Image-Pro Plus 6.0 (Media Cybernetics, Inc.).

Bioinformation analysis

TargetScan (https://www.targetscan.org/vert_80/), miRDB (https://mirdb.org/) and picTar (https://pictar.mdc-berlin.de/cgi-bin/PicTar_vertebrate.cgi) are widely applicable to predict biological targets of miRNAs. In this study, we used TargetScan, miRDB and picTar to synthetically analyzed the targets of miR-7 or miR-153. Pathway Commons (http://www.pathwaycommons.org/) is a web resource for biological pathway data. The present study used Pathway Commons to analyzed the interaction between Sp1 and p53.

Statistical analysis

For the majority of experiments, the two-tailed, unpaired, Student's test was applied to compare groups. For paired GC and adjacent non-tumor tissues, the two-tailed, paired, Student's test was used (GraphPad Prism V. 7.0 software; Dotmatics). Comparisons among multiple groups were analyzed using one-way ANOVA followed by Dunnett's post hoc test. Pearson correlation analysis was used to analyze correlation of two indexes. Data are presented as mean ± SD. P-values <0.05 was considered significant. receiver operating characteristic curve was introduced for assessing the diagnostic potential of miR-7, miR-153 and their combine in GC via Statistical Product and Service Solutions (SPSS) 22.0 (IBM, Armonk, NY, USA).

Results

miR-7 and miR-153 were downregulated in H. pylori-associated GC. Expression profile changes of miRNAs were analyzed between H. pylori-infected GC tissues and compared adjacent normal tissues using the Illumina HisSeq 2500 high-throughput sequencing (miRNA-seq) technique. A total of 331 miRNAs (159 upregulated and 172 downregulated, Table SI) were identified after fold change filtering and illustrated with hierarchical clustering analysis, scatter plots and volcano plots (Fig. 1A-C), respectively. Partially differently expressed miRNAs were further confirmed in our established chronic H. pylori-infected model GES-1/HP cells (Fig. 1D). Notably, both miR-153 and miR-7 were found to be significantly downregulated in H. pylori infected GC tissues and GES-1/HP cells, revealing that dysregulation of miR-7 and miR-153 is not only involved in H. pylori-induced gastric malt lymphoma (13).

CagA plays an important role on miRNA expression (35). To clarify the effect of CagA on the downregulation of miR-7 and miR-153, GES-1 cells were co-cultured with H. pylori strain 26695 and its corresponding CagA knockout strain 0547 with or without the addition of recombinant protein CagA, respectively. The results showed that infection of H. pylori strain 26695 significantly decreased the expression of miR-7 and miR-153 in GES-1 cells. Meanwhile, compared with infection of H. pylori strain 26695, a clear elevation was found in expression of miR-7 and miR-153 accompanied by knockout of CagA. The expression of miR-7 and miR-153 was further weakened with the addition of recombinant protein CagA, indicating that CagA might account for the downregulation of miR-7 and miR-153 (Fig. 1E). Subsequently, miR-7 and miR-153 were analyzed in GES-1 cells, GC cell lines and GES-1/HP cells and the results showed that miR-7 and miR-153 were both downregulated (Fig. 1F and G). The lower expression of miR-7 and miR-153 was confirmed by FISH analysis in H. pylori-infected GC tissues compared with adjacent normal tissues (Fig. 1H).

miR-7 and miR-153 combination significantly accelerates apoptosis and autophagy in H. pylori-associated GC cells. Numerous miRNAs regulate programmed cell death including apoptosis, autophagy and necroptosis. Particularly, some miRNAs serve important roles in crosstalk between apoptosis and autophagy. Our previous results demonstrated that apoptosis was inhibited in GES-1/HP cells. Some critical apoptosis-related proteins significantly changed, e.g., Bcl-xL, Bcl2 and P65 were significantly increased, while Bax and cleaved caspase-3 were decreased (33). Furthermore, as shown in Fig. 2A, an obvious increase in apoptosis was found when GES-1 cells were treated with SBK/ΔCagA. Western blotting showed that Bax and cleaved caspase-3 were enhanced in these cells (Fig. 2B).

To clarify the functional role of miR-7 and miR-153 on cell apoptosis, miR-7, miR-153, or their combination were overexpressed in GES-1/HP and H. pylori-infected HGC-27 cells (Fig. S1), respectively. As shown in Figs. 2C and D and S2A, miR-7 or miR-153 enhanced the apoptosis of the above cells. Western blotting showed that Bcl-xL and Bcl2 were more repressed, while Bax and cleaved caspase-3 were more upregulated in cells co-expressing both miR-7 and miR-153 compared to those overexpressing miR-7 or miR-153 alone (Figs. 2E and S2E). Moreover, it was found that autophagy marker Atg5 and LC3B increased with overexpression of miR-7 and miR-153 (Figs. 2E and S2E), consistent with previous reports (36,37). Collectively, these results demonstrated that miR-7 and miR-153 serve important roles in the crosstalk between apoptosis and autophagy.

Subsequently, bioinformatics algorithms (including TargetScan, miRDB and PicTar) indicated that there was a potential binding site of miR-7 in 3′UTR of Bcl-xL and a potential binding site of miR-153 in 3′UTR of Bcl2 (Fig. 2F and G, left). Dual-luciferase reporter assay showed that overexpression of miR-7 or miR-153 inhibited luciferase activity in Bcl-xL-w3′UTR reporter gene constructs or BCL-2-w3′UTR reporter gene constructs, respectively, but did not affect mutation reporter gene constructs (right of Fig. 2F and G, right). These results proved that Bcl-xL was a direct target of miR-7 and BCL-2 was a direct target of miR-153 in H. pylori-associated GC cells.

Combination of miR-7 and miR-153 significantly represses the proliferation of H. pylori-associated GC cells. Furthermore, the effects of miR-7 and miR-153 on cell proliferation were determined in GES-1/HP and H. pylori-infected HGC-27 cells, respectively. The results of EdU assay (Figs. 3A and S2B) and clone formation assay (Figs. 3B and S2C) confirmed that cell growth and proliferation ability were decreased with overexpression of miR-7, miR-153, or their combination. Western blotting results showed that the expression of p-AKT was significantly downregulated. Meanwhile, cell-cycle associated protein P53 and P27 were significantly upregulated when GES-1/HP and H. pylori-infected HGC-27 cells were overexpressed with miR-7, miR-153, or their combination (Figs. 3C and S2E). Cell cycle detection also showed that overexpression of miR-7, miR-153, or their combination could induce G1 phase arrest (Figs. 3D and S2D).

In the xenograft model, miR-7 or miR-153 significantly reduced tumor volumes and weights and the miR-7 and miR-153 combination group had a much greater inhibitory effect than miR-7 or miR-153 alone (Fig. 3E and F). IHC analysis revealed that miR-7, or miR-153, or miR-7 and miR-153 treatment reduced proliferation via P53 and P27 (Fig. 3G).

miR-7 induces cell cycle arrest in H. pylori-associated GC cells via the Sp1/p53 axis. Bioinformatics algorithms and Pathway Commons (http://www.pathwaycommons.org/) implied that miR-7 might induce cell cycle arrest via the Sp1/p53 axis. Bioinformatics algorithms (including TargetScan, miRDB and PicTar) indicated a potential binding site in 3′UTR of Sp1 (Fig. 4A). Dual-luciferase reporter assay was performed in GES-1/HP cells and the result showed that miR-7 overexpression inhibited luciferase activity in Sp1-wild-type (w)3′UTR reporter gene constructs, but did not affect Sp1-mutant (m)3′UTR reporter gene constructs (Fig. 4B). These results proved that sp1 was a direct target of miR-7 in H. pylori-associated GC cells.

Considering the puzzling interplay between p53 and Sp1, Sp1 was overexpressed in GES-1 cell and Sp1-siRNA was adopted in GES-1/HP cells. Cell cycle analysis showed that SP1 is mainly involved in the G1 phase of the cell cycle (Fig. 4C and D). Western blotting showed that overexpression of Sp1 downregulated the expression of P53 and P27. Nevertheless, P53 and P27 were increased with Sp1 knockdown (Fig. 4D-F). Further promoter analysis showed three putative binding sites of SP1 in the approximate transcription start site of P53 (Fig. 4H). Dual luciferase reporter gene assay showed that SP1 overexpression significantly reduced the relative luciferase activity, while the relative luciferase activity was enhanced with SP1 knockdown (Fig. 4G). In addition, the ChIP assay showed that SP1 could directly bind to the P53 promoter region. All the findings suggested that cell cycle arrest was enhanced via the Sp1/p53 axis when miR-7 was overexpressed in H. pylori-associated GC cells.

miR-7 and miR-153 regulates NF-kB-induced inflammation via TLR4 and KLF5/IL-1β. Some evidence shows an association between IL-1β gene polymorphisms and H. pylori infection (38). Meanwhile, the results of the present study also showed that expression of miR-7 and miR-153 were decreased with H. pylori infection. To further uncover whether miR-7 and miR-153 control the activity of IL-1β-mediated NF-kB signaling pathway, the present study assessed the IL-1β levels in biopsy specimens obtained from the antrum of gastric patients with H. pylori infection and matched normal tissues. ELISA results showed that IL-1β was significantly enhanced in patients with H. pylori infection (Fig. 5A). Same samples were used to analyze the expression of miR-7 and miR-153. The RT-qPCR results showed that miR-7 and miR-153 were weakened with H. pylori infection (Fig. 5B and C). Furthermore, Pearson correlation analysis showed that the expression of miR-7 or miR-153 was negatively associated with the expression of IL-1β in H. pylori-infected GC specimens (Fig. 5D and E). Upregulation of IL-1β level was also detected when GES-1 cells were treated with H. pylori infection (Fig. 5F). Moreover, IL-1β was significantly reduced at both mRNA and protein levels when miR-7 and miR-153 were overexpressed in GES-1/HP cells (Fig. 5E and F). Meanwhile, TLR4 and KLF5 were decreased (Fig. 5H), which is consistent with previous reports, implying that miR-7 and miR-153 regulate NF-kB-induced inflammation via TLR4 and KLF5.

Potentially diagnostic value of miR-7 and miR-153 in H. pylori-associated gastric GC. To investigate the correlation between the expression of miR-7 and miR-153 levels and clinicopathological features of H. pylori-associated GC patients, 60 tissue specimens were adopted to analyze the clinical correlation. It was found that low expression of miR-7 and miR-153 was significantly associated with the TNM stage and lymph node metastasis (Table SII). Moreover, the expression changes of miR-7, miR-153, Bcl-xL, SP1 and Bcl2 were analyzed in 20 cases of GC tissues and paired adjacent normal tissues. Downregulation of miR-7 and miR-153 and upregulation of Bcl-xL, Sp1 and Bcl2 were found in GC tissues (Fig. 6A and B). Associations between miR-7, miR-153 and their target Bcl-xL, SP1 and Bcl2 were further confirmed by FISH and ISH analysis on tissue microarray (Fig. 6C).

As shown in Fig. 6D, the area under the curve (AUC) value for combined detection of miR-7 and miR-153 (AUC=0.9675, 95% Confidence Interval (CI): 0.9192-1.106) indicated a higher sensitivity and specificity than miR-7 (AUC=0.9675, 95% CI: 0.8019-0.9981) or miR-153 (AUC=0.9325, 95% CI: 0.8598-1.005) alone in GC tissues. Furthermore, based on the critical role of H. pylori in the carcinogenesis pattern 'chronic gastritis-gastric mucosa atrophy-intestinal metaplasia-dysplasia-gastric cancer', GC tissues from different phases of patients and paired adjacent normal tissues were obtained by endoscopic biopsy or surgical samples and the expression of miR-7 and miR-153 was detected. Results showed that miR-7 and miR-153 were gradually decreased in the process of carcinogenesis (Fig. 6E).

Finally, both miR-7 and miR-153 were detected in serum between healthy individuals and GC patients with H. pylori infection. The results showed that miR-7 and miR-153 were significantly downregulated (Fig. 6F and G) and the AUC value for combined detection of miR-7 and miR-153 (AUC=0.9500, 95% CI: 0.8788-1.021) also indicated a higher sensitivity and specificity than miR-7 (AUC=0.8750, 95% CI: 0.7493-1.001) or miR-153 (AUC=0.7800, 95% CI: 0.5823-0.9777) alone in GC serum (Fig. 6H). Collectively, the results showed the potential diagnostic value of miR-7 and miR-153 in H. pylori-associated gastric GC.

Discussion

H. pylori is considered a class I carcinogen and an inducer of the long-term evolution of gastric tissue from malignant precancerous lesions to GC (39). Growing evidence has shown the interaction between H. pylori infection and dysregulation of miRNA expression (40-42). In the present study, a total of 331 miRNAs were identified as abnormal expression between H. pylori-infected GC tissues and compared adjacent normal tissues. Among them, a number of miRNAs have been identified as possessing an association with H. pylori-induced gastric carcinogenesis. For example, miR-375 is downregulated in H. pylori-induced GC (43), it can inhibit H. pylori-induced gastric carcinogenesis by blocking JAK2-STAT3 signaling (44) and reduce the stemness of GC cells through triggering ferroptosis (45); miR-196a-5p is increased in H. pylori-positive GC tissue (13,46). In the present study, miR-153 and miR-7 were both downregulated with infection of H. pylori and demonstrated to be associated with the virulence factor CagA. Further analysis showed that downregulation of miR-7 and miR-153 can be detected in both serums and clinical tissues of H. pylori-associated patients with GC and has been associated with the TNM stage and lymph node metastasis; therefore, conjoint analysis of miR-7 and miR-153 might serve as potential candidate marker in early diagnosis of H. pylori-associated GC.

Bcl-2 families have essential roles in apoptotic regulation and have garnered interest as therapeutic targets. The present study identified that Bcl-xL is the target gene of miR-7 and Bcl2 is the target gene of miR-153. Particularly, the combined application of miR-7 and miR-153 can significantly enhance the apoptosis of GES-1/HP cells. Growing evidence has shown that Bcl2 is target of miR-7 in different cancer cells (47,48). However, there was no obvious change on the expression of Bcl2 when miR-7 was overexpressed in GES-1/HP cells, consistent with the previous result of genome-wide screenings (23). It was hypothesized that there is cell specificity. In addition, Bcl2 has been reported to be the target of miR-153 in human glioma (49), colorectal cancer (50) and chronic myeloid leukemia cells (51). The results of the present study confirmed that Bcl2 also can be negative-regulated by miR-153 in GES-1/HP cells.

Previous studies have shown that there is negative association between miR-7 and SP1 and that the miR-7/SP1 axis exerts pivotal roles via different downstream pathways. The miR-7/SP1/TP53BP1 axis may serve a key role in the radiosensitivity of non-small cell lung cancer (52) and miR-7/SP1 axis acts as an effector of MTA2 to affect KLK10 levels and mobility of cervical cancer cells (53). The present study showed that SP1 also can be repressed in miR-7-overexpressed GES-1/HP cells and cell proliferation was significantly attenuated via P53-mediated cell cycle arrest. Previous studies show that there is a puzzling interplay between p53 and Sp1. As transcription factors, both p53 and Sp1 regulate critical cellular life and death decisions. Intriguingly, p53 and Sp1 have similar consensus sequences on GC-boxes and may compete in binding to specific promoters and act in opposite directions (54,55). Li et al (56) report that Sp1 is a crucial factor for p53-mediated apoptosis but not cell cycle arrest, which conflicts with the presumed function of Sp1 as an oncogene. The present study showed that SP1 can repress the expression of P53 by directly binding to the promoter of p53.

Studies have shown the connection between inflammation and cancer (57,58). IL-1β, one of the inflammatory factors, is a potent pro-inflammatory cytokine and has been associated with an increased risk of gastritis and non-cardia GC (59). The present study confirmed its obvious elevation in GC with H. pylori infection (Fig. 5A). TLR4 has reported to possess a role in the host response to H. pylori infection (60,61), KLF5 expression is also significantly upregulated following co-culture of H. pylori (62). TLR4 and KLF5 are both involved in IL-1β activated NF-κB cascade (63,64). In the present study, overexpression of miR-7 and miR-153 inhibited the expression of NF-κB, miR-7 downregulated TLR4 and miR-153 decreased the expression of KLF5 in accordance with previous studies (65,66).

In conclusion, the present study uncovered a more comprehensive understanding of miR-7 and miR-153 in apoptosis, proliferation and inflammation of H. pylori-associated GC cells and provided insights into miR-7 and miR-153 as potential diagnostic marker of GC. Furthermore, its findings suggested that miR-7 and miR-153-targeted therapeutics may represent a potential strategy for H. pylori CagA-associated gastric cancer.

Supplementary Data

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request. The raw data sequencing data from this study have been deposited in the National Genomics Data Center, China National Center for Bioinformation/Beijing Institute of Genomics Chinese Academy of Sciences repository (https://ngdc.cncb.ac.cn/gas-human/; accession no. HRA003675).

Authors' contributions

YS, DG, JFL, LNG, PL, YMQ, HYC, TL, XC, LYS and YRW designed and performed experiments, analyzed data, interpreted results and prepared the manuscript. ZJD helped with designing experiments and evaluating the manuscript. MYW designed experiments, analyzed data and wrote the manuscript. DG, LYS and XC revised the manuscript. MYW and PL confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The studies involving human participants were reviewed and approved by the Ethics Committee of Weihai Municipal Hospital (approval no. 2021021). Informed consent to be included in the study was obtained from all patients.

Patient consent of publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors are grateful to Professor Boqing Li of Binzhou Medical University (Yantai, China) for H. pylori strain ATCC 26695 and its corresponding CagA-knockout strain.

Funding

The present study was funded by the National Natural Science Foundation of China (grant no. 82102398), Shandong Provincial Natural Science Foundation (grant nos. ZR2020QC073, ZR2021MH409 and ZR2020MH296), Shandong Province Medical and Health Science and Technology Development Plan (grant nos. 2018WS106 and 202211000314) and the Incubation Project of Weihai Municipal Hospital (grant nos. FH-2021-XZ02 and FH-2021-JY16).

References

1 

Smyth EC, Nilsson M, Grabsch HI, van Grieken NC and Lordick F: Gastric cancer. Lancet. 396:635–648. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Plummer M, Franceschi S, Vignat J, Forman D and de Martel C: Global burden of gastric cancer attributable to Helicobacter pylori. Int J Cancer. 136:487–490. 2015. View Article : Google Scholar

3 

Wen S and Moss SF: Helicobacter pylori virulence factors in gastric carcinogenesis. Cancer Lett. 282:1–8. 2009. View Article : Google Scholar :

4 

Nejati S, Karkhah A, Darvish H, Validi M, Ebrahimpour S and Nouri HR: Influence of Helicobacter pylori virulence factors CagA and VacA on pathogenesis of gastrointestinal disorders. Microb Pathog. 117:43–48. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Javed S, Skoog EC and Solnick JV: Impact of Helicobacter pylori virulence factors on the host immune response and gastric pathology. Curr Top Microbiol Immunol. 421:21–52. 2019.PubMed/NCBI

6 

Takahashi-Kanemitsu A, Knight CT and Hatakeyama M: Molecular anatomy and pathogenic actions of Helicobacter pylori CagA that underpin gastric carcinogenesis. Cell Mol Immunol. 17:50–63. 2020. View Article : Google Scholar :

7 

Gao S, Song D, Liu Y, Yan H and Chen X: Helicobacter pylori CagA PROTEIN ATTENUates 5-Fu sensitivity of gastric cancer cells through upregulating cellular glucose metabolism. Onco Targets Ther. 13:6339–6349. 2020. View Article : Google Scholar : PubMed/NCBI

8 

Lujambio A and Lowe SW: The microcosmos of cancer. Nature. 482:347–355. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Dastmalchi N, Safaralizadeh R and Banan Khojasteh SM: The correlation between microRNAs and Helicobacter pylori in gastric cancer. Pathog Dis. 77:ftz0392019. View Article : Google Scholar : PubMed/NCBI

11 

Parizadeh SM, Jafarzadeh-Esfehani R, Avan A, Ghandehari M, Goldani F and Parizadeh SM: The prognostic and predictive value of microRNAs in patients with H. pylori-positive gastric cancer. Curr Pharm Des. 24:4639–4645. 2018. View Article : Google Scholar

12 

Zou D, Xu L, Li H, Ma Y, Gong Y, Guo T, Jing Z, Xu X and Zhang Y: Role of abnormal microRNA expression in Helicobacter pylori associated gastric cancer. Crit Rev Microbiol. 45:239–251. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Blosse A, Levy M, Robe C, Staedel C, Copie-Bergman C and Lehours P: Deregulation of miRNA in Helicobacter pylori-induced gastric MALT lymphoma: From mice to human. J Clin Med. 8:8452019. View Article : Google Scholar : PubMed/NCBI

14 

Korać P, Antica M and Matulić M: MiR-7 in cancer development. Biomedicines. 9:3252021. View Article : Google Scholar

15 

Xiao H: MiR-7-5p suppresses tumor metastasis of non-small cell lung cancer by targeting NOVA2. Cell Mol Biol Lett. 24:602019. View Article : Google Scholar : PubMed/NCBI

16 

Xia J, Cao T, Ma C, Shi Y, Sun Y, Wang ZP and Ma J: miR-7 suppresses tumor progression by directly targeting MAP3K9 in pancreatic cancer. Mol Ther Nucleic Acids. 13:121–132. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Li M, Pan M, Wang J, You C, Zhao F, Zheng D, Guo M, Xu H, Wu D, Wang L and Dou J: miR-7 reduces breast cancer stem cell metastasis via inhibiting RELA to decrease ESAM expression. Mol Ther Oncolytics. 18:70–82. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Kong D, Piao YS, Yamashita S, Oshima H, Oguma K, Fushida S, Fujimura T, Minamoto T, Seno H, Yamada Y, et al: Inflammation-induced repression of tumor suppressor miR-7 in gastric tumor cells. Oncogene. 31:3949–3960. 2012. View Article : Google Scholar

19 

Lin J, Liu Z, Liao S, Li E, Wu X and Zeng W: Elevated microRNA-7 inhibits proliferation and tumor angiogenesis and promotes apoptosis of gastric cancer cells via repression of Raf-1. Cell Cycle. 19:2496–2508. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Xu N, Lian YJ, Dai X and Wang YJ: miR-7 increases cisplatin sensitivity of gastric cancer cells through suppressing mTOR. Technol Cancer Res Treat. 16:1022–1030. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Xie J, Chen M, Zhou J, Mo MS, Zhu LH, Liu YP, Gui QJ, Zhang L and Li GQ: miR-7 inhibits the invasion and metastasis of gastric cancer cells by suppressing epidermal growth factor receptor expression. Oncol Rep. 31:1715–1722. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Zhao X, Dou W, He L, Liang S, Tie J, Liu C, Li T, Lu Y, Mo P, Shi Y, et al: MicroRNA-7 functions as an anti-metastatic microRNA in gastric cancer by targeting insulin-like growth factor-1 receptor. Oncogene. 32:1363–1372. 2013. View Article : Google Scholar

23 

Zhao XD, Lu YY, Guo H, Xie HH, He LJ, Shen GF, Zhou JF, Li T, Hu SJ, Zhou L, et al: MicroRNA-7/NF-κB signaling regulatory feedback circuit regulates gastric carcinogenesis. J Cell Biol. 210:613–627. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Ye T, Yang M, Huang D, Wang X, Xue B, Tian N, Xu X, Bao L, Hu H, Lv T and Huang Y: MicroRNA-7 as a potential therapeutic target for aberrant NF-κB-driven distant metastasis of gastric cancer. J Exp Clin Cancer Res. 38:552019. View Article : Google Scholar

25 

Wang L, Lv X, Fu X, Su L, Yang T and Xu P: MiR-153 inhibits the resistance of lung cancer to gefitinib via modulating expression of ABCE1. Cancer Biomark. 25:361–369. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Shi D, Li Y, Fan L, Zhao Q, Tan B and Cui G: Upregulation of miR-153 inhibits triple-negative breast cancer progression by targeting ZEB2-mediated EMT and contributes to better prognosis. Onco Targets Ther. 12:9611–9625. 2019. View Article : Google Scholar

27 

Li C, Zhang Y, Zhao W, Cui S and Song Y: miR-153-3p regulates progression of ovarian carcinoma in vitro and in vivo by targeting MCL1 gene. J Cell Biochem. 120:19147–19158. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Zhang W, Mao S, Shi D, Zhang J, Zhang Z, Guo Y, Wu Y, Wang R, Wang L, Huang Y and Yao X: MicroRNA-153 decreases tryptophan catabolism and inhibits angiogenesis in bladder cancer by targeting indoleamine 2,3-dioxygenase 1. Front Oncol. 9:6192019. View Article : Google Scholar : PubMed/NCBI

29 

Bi CW, Zhang GY, Bai Y, Zhao B and Yang H: Increased expression of miR-153 predicts poor prognosis for patients with prostate cancer. Medicine (Baltimore). 98:e167052019. View Article : Google Scholar : PubMed/NCBI

30 

Zhang L, Pickard K, Jenei V, Bullock MD, Bruce A, Mitter R, Kelly G, Paraskeva C, Strefford J, Primrose J, et al: miR-153 supports colorectal cancer progression via pleiotropic effects that enhance invasion and chemotherapeutic resistance. Cancer Res. 73:6435–6447. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Ouyang Y, Yuan W and Qiu S: MicroRNA-153 functions as a tumor suppressor in gastric cancer via targeting Kruppel-like factor 5. Exp Ther Med. 16:473–482. 2018.PubMed/NCBI

32 

Guo Y, Zhang T, Shi Y, Zhang J, Li M, Lu F, Zhang J, Chen X and Ding S: Helicobacter pylori inhibits GKN1 expression via the CagA/p-ERK/AUF1 pathway. Helicobacter. 25:e126652020. View Article : Google Scholar

33 

Liu JF, Guo D, Kang EM, Wang YS, Gao XZ, Cong HY, Liu P, Zhang NQ and Wang MY: Acute and chronic infection of H. pylori caused the difference in apoptosis of gastric epithelial cells. Microb Pathog. 150:1047172021. View Article : Google Scholar : PubMed/NCBI

34 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

35 

Hayashi Y, Tsujii M, Wang J, Kondo J, Akasaka T, Jin Y, Li W, Nakamura T, Nishida T, Iijima H, et al: CagA mediates epigenetic regulation to attenuate let-7 expression in Helicobacter pylori-related carcinogenesis. Gut. 62:1536–1546. 2013. View Article : Google Scholar

36 

Yuan J, Li Y, Liao J, Liu M, Zhu L and Liao K: MicroRNA-7 inhibits hepatocellular carcinoma cell invasion and metastasis by regulating Atg5-mediated autophagy. Transl Cancer Res. 9:3965–3972. 2020. View Article : Google Scholar : PubMed/NCBI

37 

Hou S, Guo M, Xi H, Zhang L, Zhao A, Hou H and Fang W: MicroRNA-153-3p sensitizes melanoma cells to dacarbazine by suppressing ATG5-mediated autophagy and apoptosis. Transl Cancer Res. 9:5626–5636. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Ren HY, Wen LS, Geng YH, Huang JB, Liu JF, Shen DY and Meng JR: Association between IL-1B gene polymorphisms and Helicobacter pylori infection: A meta-analysis. Microb Pathog. 137:1037692019. View Article : Google Scholar : PubMed/NCBI

39 

Wang F, Meng W, Wang B and Qiao L: Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Lett. 345:196–202. 2014. View Article : Google Scholar

40 

Han T, Jing X, Bao J, Zhao L, Zhang A, Miao R, Guo H, Zhou B, Zhang S, Sun J and Shi J: H. pylori infection alters repair of DNA double-strand breaks via SNHG17. J Clin Invest. 130:3901–3918. 2020. View Article : Google Scholar : PubMed/NCBI

41 

So JBY, Kapoor R, Zhu F, Koh C, Zhou L, Zou R, Tang YC, Goo PCK, Rha SY, Chung HC, et al: Development and validation of a serum microRNA biomarker panel for detecting gastric cancer in a high-risk population. Gut. 70:829–837. 2021. View Article : Google Scholar

42 

Liu Y, Zhu J, Ma X, Han S, Xiao D, Jia Y and Wang Y: ceRNA network construction and comparison of gastric cancer with or without Helicobacter pylori infection. J Cell Physiol. 234:7128–7140. 2019. View Article : Google Scholar

43 

Zhang Z, Chen S, Fan M, Ruan G, Xi T, Zheng L, Guo L, Ye F and Xing Y: Helicobacter pylori induces gastric cancer via down-regulating miR-375 to inhibit dendritic cell maturation. Helicobacter. 26:e128132021. View Article : Google Scholar : PubMed/NCBI

44 

Miao L, Liu K, Xie M, Xing Y and Xi T: miR-375 inhibits Helicobacter pylori-induced gastric carcinogenesis by blocking JAK2-STAT3 signaling. Cancer Immunol Immunother. 63:699–711. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Ni H, Qin H, Sun C, Liu Y, Ruan G, Guo Q, Xi T, Xing Y and Zheng L: MiR-375 reduces the stemness of gastric cancer cells through triggering ferroptosis. Stem Cell Res Ther. 12:3252021. View Article : Google Scholar : PubMed/NCBI

46 

Lee JW, Kim N, Park JH, Kim HJ, Chang H, Kim JM, Kim JW and Lee DH: Differential MicroRNA expression between gastric cancer tissue and non-cancerous gastric mucosa according to Helicobacter pylori status. J Cancer Prev. 22:33–39. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Hong T, Ding J and Li W: miR-7 reverses breast cancer resistance to chemotherapy by targeting MRP1 And BCL2. Onco Targets Ther. 12:11097–11105. 2019. View Article : Google Scholar

48 

Xiong S, Zheng Y, Jiang P, Liu R, Liu X and Chu Y: MicroRNA-7 inhibits the growth of human non-small cell lung cancer A549 cells through targeting BCL-2. Int J Biol Sci. 7:805–814. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Sun D, Mu Y and Piao H: MicroRNA-153-3p enhances cell radiosensitivity by targeting BCL2 in human glioma. Biol Res. 51:562018. View Article : Google Scholar : PubMed/NCBI

50 

He Y, Zhang L, Tan F, Wang LF, Liu DH, Wang RJ and Yin XZ: MiR-153-5p promotes sensibility of colorectal cancer cells to oxaliplatin via targeting Bcl-2-mediated autophagy pathway. Biosci Biotechnol Biochem. 84:1645–1651. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Li YL, Tang JM, Chen XY, Luo B, Liang GH, Qu Q and Lu ZY: MicroRNA-153-3p enhances the sensitivity of chronic myeloid leukemia cells to imatinib by inhibiting B-cell lymphoma-2-mediated autophagy. Hum Cell. 33:610–618. 2020. View Article : Google Scholar : PubMed/NCBI

52 

Guo G, Li L, Song G, Wang J, Yan Y and Zhao Y: miR-7/SP1/TP53BP1 axis may play a pivotal role in NSCLC radiosensitivity. Oncol Rep. 44:2678–2690. 2020. View Article : Google Scholar : PubMed/NCBI

53 

Lin CL, Ying TH, Yang SF, Wang SW, Cheng SP, Lee JJ and Hsieh YH: Transcriptional suppression of miR-7 by MTA2 induces Sp1-mediated KLK10 expression and metastasis of cervical cancer. Mol Ther Nucleic Acids. 20:699–710. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Wang MJ, Pei DS, Qian GW, Yin XX, Cheng Q, Li LT, Li HZ and Zheng JN: p53 regulates Ki-67 promoter activity through p53- and Sp1-dependent manner in HeLa cells. Tumour Biol. 32:905–912. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Drayman N, Ben-Nun-Shaul O, Butin-Israeli V, Srivastava R, Rubinstein AM, Mock CS, Elyada E, Ben-Neriah Y, Lahav G and Oppenheim A: p53 elevation in human cells halt SV40 infection by inhibiting T-ag expression. Oncotarget. 7:52643–52660. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Li H, Zhang Y, Ströse A, Tedesco D, Gurova K and Selivanova G: Integrated high-throughput analysis identifies Sp1 as a crucial determinant of p53-mediated apoptosis. Cell Death Differ. 21:1493–1502. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Balkwill F and Mantovani A: Inflammation and cancer: Back to Virchow? Lancet. 357:539–545. 2001. View Article : Google Scholar : PubMed/NCBI

58 

Coussens LM and Werb Z: Inflammation and cancer. Nature. 420:860–867. 2002. View Article : Google Scholar : PubMed/NCBI

59 

El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, Young HA, Herrera J, Lissowska J, Yuan CC, Rothman N, et al: Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature. 404:398–402. 2000. View Article : Google Scholar : PubMed/NCBI

60 

Kawahara T, Kuwano Y, Teshima-Kondo S, Kawai T, Nikawa T, Kishi K and Rokutan K: Toll-like receptor 4 regulates gastric pit cell responses to Helicobacter pylori infection. J Med Invest. 48:190–197. 2001.PubMed/NCBI

61 

Kawahara T, Teshima S, Oka A, Sugiyama T, Kishi K and Rokutan K: Type I Helicobacter pylori lipopolysaccharide stimulates toll-like receptor 4 and activates mitogen oxidase 1 in gastric pit cells. Infect Immun. 69:4382–4389. 2001. View Article : Google Scholar : PubMed/NCBI

62 

Noto JM, Khizanishvili T, Chaturvedi R, Piazuelo MB, Romero-Gallo J, Delgado AG, Khurana SS, Sierra JC, Krishna US, Suarez G, et al: Helicobacter pylori promotes the expression of Krüppel-like factor 5, a mediator of carcinogenesis, in vitro and in vivo. PLoS One. 8:e543442013. View Article : Google Scholar

63 

Xie Z, Jie Z, Wang G, Sun X, Tang P, Chen S, Qin A, Wang J and Fan S: TGF-β synergizes with ML264 to block IL-1β-induced matrix degradation mediated by Krüppel-like factor 5 in the nucleus pulposus. Biochim Biophys Acta Mol Basis Dis. 1864:579–589. 2018. View Article : Google Scholar

64 

Tang J, Xu L, Zeng Y and Gong F: Effect of gut microbiota on LPS-induced acute lung injury by regulating the TLR4/NF-kB signaling pathway. Int Immunopharmacol. 91:1072722021. View Article : Google Scholar

65 

Zhang XD, Fan QY, Qiu Z and Chen S: MiR-7 alleviates secondary inflammatory response of microglia caused by cerebral hemorrhage through inhibiting TLR4 expression. Eur Rev Med Pharmacol Sci. 22:5597–5604. 2018.PubMed/NCBI

66 

Yu L, Xu Q, Yu W, Duan J and Dai G: LncRNA cancer susceptibility candidate 15 accelerates the breast cancer cells progression via miR-153-3p/KLF5 positive feedback loop. Biochem Biophys Res Commun. 506:819–825. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2023
Volume 63 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Song Y, Guo D, Liu J, Ge L, Liu P, Qu Y, Cong H, Li T, Chang X, Wang Y, Wang Y, et al: Downregulation of miR‑7 and miR‑153 is involved in <em>Helicobacter pylori</em> CagA induced gastric carcinogenesis and progression. Int J Oncol 63: 79, 2023
APA
Song, Y., Guo, D., Liu, J., Ge, L., Liu, P., Qu, Y. ... Wang, M. (2023). Downregulation of miR‑7 and miR‑153 is involved in <em>Helicobacter pylori</em> CagA induced gastric carcinogenesis and progression. International Journal of Oncology, 63, 79. https://doi.org/10.3892/ijo.2023.5527
MLA
Song, Y., Guo, D., Liu, J., Ge, L., Liu, P., Qu, Y., Cong, H., Li, T., Chang, X., Wang, Y., Shao, L., Du, Z., Wang, M."Downregulation of miR‑7 and miR‑153 is involved in <em>Helicobacter pylori</em> CagA induced gastric carcinogenesis and progression". International Journal of Oncology 63.1 (2023): 79.
Chicago
Song, Y., Guo, D., Liu, J., Ge, L., Liu, P., Qu, Y., Cong, H., Li, T., Chang, X., Wang, Y., Shao, L., Du, Z., Wang, M."Downregulation of miR‑7 and miR‑153 is involved in <em>Helicobacter pylori</em> CagA induced gastric carcinogenesis and progression". International Journal of Oncology 63, no. 1 (2023): 79. https://doi.org/10.3892/ijo.2023.5527