1 Introduction
Gastric cancer (GC) is a prevalent malignancy globally, with around one million new cases diagnosed each year [
1,
2]. Despite advancements in treatment, the 5-year survival rate for GC remains below 40%, primarily due to late-stage diagnoses [
3,
4]. This underscores the urgent need for reliable early diagnostic and prognostic biomarkers, and a deeper understanding of the molecular mechanism for GC progression.
The transcription factor (TF) zinc finger and BTB domain-containing (ZBTB) 10 belongs to the ZBTB protein family, an emerging family of TFs with critical functions in development, differentiation, and oncogenesis [
5,
6]. ZBTB10 has been identified as a downstream target of microRNA-27a and acts as a potential suppressor of the transcription factor Sp1 in various cancers. For instance, Mertens-Talcott et al. found that miR-27a promoted breast cancer progression by regulating the expression of specificity protein TFs, ZBTB10 [
7]. Another study revealed that miR-27a regulated the endothelial differentiation of breast cancer stem-like cells through the ZBTB10/VEGF axis [
8]. However, recent studies have shown that ZBTB10 also exerted its functions through other mechanisms. Bluhm A et al. found that ZBTB10 could act as a novel variant repeat binding protein at the telomere region of alternative lengthening of telomere [
9]. Meanwhile, it has also been found that ZBTB10 transcription factor was crucial for murine cDC1 activation and cytokine secretion [
10]. More importantly, androgen-activated ZBTB10 has been confirmed to inhibit prostate cancer progression by negatively regulating PKLR expression [
11]. It follows from the above that the potential mechanisms by which ZBTB10 acts are varied. Although direct links between ZBTB10 and GC progression are not yet fully established, genetic variations in hsa-mir-27a that increase miR-27a expression levels and reduce ZBTB10 expression levels were associated with heightened GC risk and metastasis in the Chinese population [
12]. This suggests a potential role for ZBTB10 in GC progression and highlights the need for further investigation into its detailed roles and specific mechanisms.
Betulinic acid (BA), derived from the bark of white birch trees, is a pentacyclic triterpenoid with diverse biological activities, including anti-viral, anti-bacterial, anti-oxidant, anti-inflammatory, and anti-fibrotic effects [
13‐
15]. Liu et al. identified BA as a modulator of cannabinoid receptors CB1 and CB2 [
16], and it has demonstrated significant anti-tumor activity in cancers such as breast [
17], gastric [
18], and bladder [
19]. The anti-cancer effects of BA were thought to involve various pathways, including the Bmi-1/ROS/AMPK-mTOR-ULK1 pathway in bladder cancer [
20] and the NF-κB/VASP pathway in GC [
18]. However, there is still a lack of systematic research on the in vivo experiments and underlying mechanisms by which BA inhibits GC progression. Therefore, it is crucial to validate the effect of BA in vivo and explore its mechanisms more systematically.
In this study, we elucidated the inhibiting effects of ZBTB10 and BA in GC progression using both in vitro and in vivo models. We demonstrated that BA-induced upregulation of ZBTB10 transcriptionally activated ARRDC3, which in turn bound to ITGB4, leading to its ubiquitination and degradation. This process ultimately reduced the levels of PI3K and AKT phosphorylation. Our findings suggest that targeting the ZBTB10/ARRDC3/ITGB4/PI3K/AKT axis, alongside BA treatment, could offer promising strategies for GC therapy.
2 Materials and methods
2.1 Patients and tissue specimens
GC tissues and paired adjacent normal tissues were collected from patients diagnosed with GC who underwent surgical resection at the First Affiliated Hospital of Sun Yat-sen University. All tissue specimens were confirmed by pathologic examination and were separated and frozen at -80 °C or formalin-fixed. Written informed consents were obtained from all the patients, and the study was approved by the Ethics Committee of the First Affiliated Hospital of Sun Yat-sen University.
2.2 Cell lines, cell culture, and reagents
Human GC cell lines (AGS, MKN1, MKN28, HGC-27 and MGC803) and normal human gastric epithelial cells-1 (GES-1) were purchased from the Chinese Academy of Sciences, Shanghai Branch Cell Bank. All human cell lines have been authenticated using STR profiling within the last three years, and all experiments were performed with mycoplasma-free cells. AGS cells were cultured in DMEM/F12 (Gibco, Waltham, MA, USA), and MKN1, MKN28, HGC-27, MGC803, and GES-1 cells were cultured in RPMI-1640 (Gibco).
BA (B8936, Sigma Aldrich, St. Louis, MO, USA) was dissolved in DMSO (Sigma Aldrich) as a 20 mM stock solution and stored at -20 °C. LY294002 (HY-10108, MCE, Shanghai, China) was dissolved in DMSO as a 5 mM stock solution and stored at -80 °C. MG-132 (HY-13259, MCE) was dissolved in DMSO as a 10 mM stock solution and stored at -80 °C.
2.3 Construction of stable cell lines and transfection
For the construction of stable overexpression and knockdown GC cell lines, we purchased lentivirus from iGene Biotechnology (Guangzhou, Guangdong, China) and infected GC cells with it. Subsequently, complete media containing 3 µg/mL puromycin (Sigma-Aldrich) were used to screen stably overexpressing and knocked down GC cell lines. Overexpression plasmids were constructed using pEZ-Lv201 (iGene Biotechnology), and knockdown plasmids were constructed using LVRU6GP (iGene Biotechnology). All plasmids and siRNAs were constructed from iGene Biotechnology (Supplementary Table
2). Plasmids and siRNAs transfection were performed using Lipofectamine 3000 (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions.
2.4 Cell viability assay
For the proliferation assays, GC cells (1 × 103/well) were seeded in 96-well plates with six replicates. Proliferation rates were determined using the Cell Counting Kit-8 (CCK-8, Boster Biological Technology, Wuhan, Hubei, China), and measure was performed on a microplate reader (Bio-TEK, Winooski, VT, USA). For the cell viability assay, 5 × 103 cells per well were inoculated into 96-well plates. After attaching for about 24 h, the culture medium was replaced with complete media containing different concentrations of BA and LY294002 for 24 h. Finally, the absorbance was measured at 450 nm.
2.5 5-Ethynyl-2’-deoxyuridine (EdU) incorporation assay
5 × 103 cells per well were inoculated into 96-well plates. The EdU incorporation assay was conducted using an EdU Staining Kit (RiboBio, Guangzhou, Guangdong, China) according to the manufacturer’s instructions. Finally, the cell nucleus was visualized using DAPI, and fluorescence signals were acquired using a fluorescence microscope (Leica, Wetzlar, Germany).
Cells were seeded separately in 6-well plates at a density of 500/well. The medium was changed twice per week. After 14 days, cells were fixed in 4% paraformaldehyde for 30 min, stained with 0.5% crystal violet for 15 min, rinsed three times with PBS to remove excess dye, photographed, and counted.
2.7 Transwell cell migration and invasion assay
5 × 104 cells resuspended in 400 µL serum-free medium were plated in the upper chamber, while the lower chamber was filled with the complete culture medium. Finally, the cells were fixed with 4% paraformaldehyde and stained with 0.5% crystal violet. Images were acquired using a microscope (Olympus, Tokyo, Japan).
2.8 Cell apoptosis analysis
Cell apoptosis was evaluated by flow cytometric analysis using annexin V-FITC/PI staining (BD Biosciences, San Jose, CA, USA). Apoptotic cells were analyzed using FACSCalibur flow cytometry (BD Biosciences), and FlowJo software (Tree Star Corp, Ashland, WI, USA) was used to analyze the results.
2.9 RNA isolation and quantitative real-time PCR (qRT-PCR)
Total RNA was extracted from tissues and cells using TRIzol reagent (Takara, Beijing, China) according to the manufacturer’s protocol. cDNA was generated using the Master Mix cDNA Synthesis Kit (Accurate Biotechnology, Changsha, Hunan, China). qPCR was performed using SYBR Green I (Accurate Biotechnology). The expression levels of the target genes were measured using the 2–ΔΔCt method and normalized to GAPDH as a reference. Each experiment was performed in triplicate and qPCR primers are listed in Supplementary Table 3.
2.10 Protein extracting and western blot (WB)
Western blot was performed as described by Liu [
21]. Proteins were extracted from tissues and cells using RIPA lysis buffer (Beyotime, Guangzhou, Guangdong, China) following the manufacturer’s protocol and quantified with the bicinchoninic acid protein assay kit (Beyotime). GAPDH was used as a loading control. The antibodies are listed in Supplementary Table 4.
2.11 Phospho-proteomic profiling
ZBTB10-overexpression AGS cells and control cells were washed thrice with PBS and solubilized in lysis buffer. The lysates were resuspended at 4 °C for 30 min by gently pipetting and rocking. Then, the lysates were centrifuged at 14,000 ×g for 10 min and transfer the supernatant into a clean test tube. After quantifying the protein concentration with the BCA assay kit (Beyotime), cell lysates containing 400 µg of total protein were firstly diluted and then detected in Proteome Profiler Human Phospho-Kinase Array (R&D Systems, Minneapolis, MN, USA) according to the manufacturer’s instructions.
2.12 Enzyme-linked immunosorbent assay (ELISA)
Concentrations of p-PI3K in AGS and MKN1 cell lysates with ZBTB10 overexpression and knockdown were measured by the ELISA Kits (ml060625, Mlbio, Shanghai, China) according to the manufacturer’s instructions.
2.13 Chromatin immunoprecipitation (ChIP)
The ChIP assay was performed using a ChIP assay kit (Cell Signaling Technology, Boston, MA, USA) according to the manufacturer’s instructions. Briefly, GC cells were crosslinked with 1% formaldehyde and quenched with glycine (Solarbio, Beijing, China) at room temperature, after which they were collected, washed and resuspended in lysis buffer (Cell Signaling Technology). The digested chromatin was incubated with anti-IgG (DIA-AN, Wuhan, Hubei, China) and anti-ZBTB10 (Abcam) for immunoprecipitation. DNA was purified and analyzed by qRT-PCR.
2.14 Luciferase reporter assay
Plasmid preparation was completed by Genechem (Shanghai, China). AGS and MKN1 cells were transfected with corresponding plasmids using Lipofectamine 3000 (Invitrogen). After 48 h, 20 µM BA was added to the medium and treated for 24 h. Luciferase activity was detected by the Multifunctional enzyme marker (Thermo).
2.15 Co-immunoprecipitation (Co-IP)
The cells were collected and were lysed on ice with IP lysate. Lysates were centrifuged for 10 min (12,000 ×g, 4 °C), and the supernatant was collected. Subsequently, the supernatant was incubated with corresponding primary antibodies overnight at 4 °C. The next day, protein A/G Magnetic Beads were added to the samples and incubated for 4 h at 4 °C. The beads were washed five times and then denatured at 97 °C for 7 min prior to WB analysis.
2.16 MG-132 treatment
For the ubiquitination assay, the GC cells were pretreated with the indicated plasmids for 48 h. After treated with 20 µM MG-132 for 6 h, the cells were lysed by IP lysis buffer. Next, immunoprecipitation against the corresponding protein was performed. Finally, the immunoprecipitants were subjected to WB.
2.17 Cellular immunofluorescence (IF)
AGS cells with stable ARRDC3 overexpression and negative control were seeded onto the chamber slides. After attaching for about 24 h, the cells on chamber slides were washed with PBS and fixed in 4% paraformaldehyde for 15 min at room temperature, followed by exposure to 0.3% Triton X-100 for 20 min. Subsequently, the cells were blocked in 2% goat serum for 1 h and incubated with specific primary antibodies overnight at 4 °C. The next day, cells were incubated fluorescent secondary antibodies corresponding to the species in dark for 2 h and DAPI was used for nuclear staining. Finally, images were acquired using a confocal microscope (FV3000, Olympus).
2.18 Immunohistochemistry (IHC)
The tissues were fixed with 10% paraformaldehyde and embedded in paraffin. After deparaffinization, rehydration, and antigen retrieval, the samples were incubated with primary antibodies using the corresponding antibodies. Images were taken with an Axio Scope.A1 398 vertical microscope (Leica). The final immunoreactive score (IRS) was obtained by multiplication of the intensity score and the quantity score. Briefly, two investigators independently scored the percentage of positive-stained cells (%PC) from 0 to 4 (0: no positive cells; 1: <10%; 2: 10–50%; 3: 51–80%; 4: >80%) and the staining intensity from 0 to 3 (0: no staining; 1: weak staining; 2: moderate staining; 3: strong staining). The IRS was then calculated as the product of these two scores (range, 0–12). We defined an IRS score of less than 6 as low expression and the rest as high expression.
2.19 Animal studies
Female BALB/c nude mice (5 weeks old) were obtained from Specific Pathogen-Free Biotechnology Co., Ltd. (Beijing, China) and housed under specific pathogen-free conditions. All animal experiments were performed with the approval of the Animal Care and Use Committee of the Sun Yat-sen University.
For the xenograft model, MGC803 cells (5 × 10
6 in 100 µL PBS) with ZBTB10 overexpression and negative control were subcutaneously injected into each flank of nude mice (six mice/group). Tumor size was measured every 3 days (volume = length × width
2 × 1/2). The mice were sacrificed 21 days after tumor cell implantation. The tumors were weighed, imaged, fixed in 4% paraformaldehyde. To investigate the function of BA, xenograft models were established using MGC803 cells with knockdown ZBTB10 and negative control according to the methods in previous studies [
17]. Seven days later, the tumors grew to approximately 100 mm
3, and tumor-bearing mice were randomly assigned to four groups.
For the lung metastasis model, MGC803 cells (1 × 10
6 in 100 µL PBS) overexpressing ZBTB10 and the negative control were injected into the tail vein of nude mice. The mice were sacrificed 8 weeks after GC cell injection and pulmonary metastatic nodules were counted. Finally, the lungs were resected, photographed, and fixed in 4% paraformaldehyde for further analyses. Similarly, we explored the effect of BA on lung metastasis according to a previous study [
22].
For the popliteal lymph node metastasis model, MGC803 cells (1 × 106 in 20 µL PBS) with ZBTB10 overexpression and negative control were injected into the left footpad of mice (twice in 3 days). The mice were sacrificed after 8 weeks, and the lymph node transfer ratio was determined. The footpad and popliteal lymph nodes of the left leg were resected, photographed, and fixed in 4% paraformaldehyde for further analyses.
2.20 Statistical analysis
All statistical analyses were performed using SPSS 22.0 (IBM, Chicago, IL, USA) and GraphPad Prism 8.0 (GraphPad, La Jolla, CA, USA). All results were presented as the mean ± standard deviation (SD) of data from independent bio-triplications. Data analysis in each group was performed using Student’s t-test, and one-way or two-way analysis of variance. Kaplan-Meier analysis was used to determine the survival differences. P < 0.05 was considered to indicate a statistically significant difference.
4 Discussion
Most patients with GC are diagnosed at advanced stages, which significantly complicates treatment options [
32]. Despite numerous efforts to explore for the early diagnostic methods, reliable molecular biomarkers for GC remain largely unidentified. The ZBTB protein family, characterized by their zinc finger domains, encompasses a group of TFs involved in various biological processes, including gene regulation, DNA recognition, RNA packaging, apoptosis regulation, and protein folding and assembly [
33,
34]. Moreover, these proteins can act as either tumor promoters or suppressors depending on the context [
35]. In our study, RNA-sequencing identified ZBTB10 as one of the most downregulated genes in GC. ZBTB10 is a novel member of the ZBTB transcription factors, and its biological role has not been fully elucidated. Previous studies have reported that ZBTB10 functions as a transcriptional inhibitor of SP1 and plays a role in inhibiting tumor activities, suggesting that ZBTB10 should be considered a tumor suppressor [
36]. Notably, in a pivotal investigation examining the nexus between mutations in miRNA and susceptibility of GC, reduced ZBTB10 mRNA level was found to be associated with the increased risk and metastasis of GC [
12], suggesting that ZBTB10 may inhibit GC progression. Our findings confirmed that ZBTB10 was significantly downregulated in GC tissues compared to normal tissues. Further analysis revealed that down-regulated ZBTB10 expression in patients with GC was an independent risk factor and closely correlated with lower survival rate. Similarly, we also draw the same conclusion in cell function and animal experiments. To the best of our knowledge, this is the first study to determine the role of ZBTB10 in GC progression.
The PI3K/AKT pathway is one of the major cellular signaling pathways that plays an important role in basic intracellular functions [
37]. It is crucial for regulating cell growth, survival, and metabolism, and its dysregulation is implicated in numerous human cancers [
38]. Furthermore, researches have also confirmed the involvement of PI3K/AKT pathway in GC progression and the association with poor outcomes [
39,
40]. Our study was the first to demonstrate that ZBTB10 inhibited the PI3K/AKT pathway activation. More importantly, inactivating the PI3K/AKT pathway via LY294002 and siRNAs targeting AKT was proved to effectively counteract the malignant behaviors regulated by ZBTB10, which validated ZBTB10-hindering GC progression through the PI3K/AKT pathway inactivation.
However, the detailed mechanism of ZBTB10-mediated the PI3K/AKT pathway inactivation in GC remains unclear. Through comprehensive analysis of sequencing data, we identified ARRDC3 as one of the pivotal targets of ZBTB10. ARRDC3, a member of the arrestin superfamily, is known to inhibit tumor progression by negatively regulating ITGB4, a protein linked to aggressive tumor behavior [
25,
41]. For instance, it has been reported that ARRDC3 inhibited breast cancer progression by negatively regulating ITGB4 [
25]. Another study showed that ARRDC3 inhibited the progression of prostate cancer through ARRDC3/ITGB4 pathway [
26]. Given that integrins, including ITGB4, are known regulators of the PI3K/AKT pathway [
27], and that ARRDC3 has been shown to inhibit liver fibrosis and epithelial-to-mesenchymal transition via the ITGB4/PI3K/AKT pathway [
24], we hypothesized that ZBTB10 inactivates the PI3K/AKT pathway through the ARRDC3/ITGB4 axis. Further investigation revealed that ARRDC3 downregulation in GC tissues was correlated with poor prognosis and that ARRDC3 inhibited GC cell proliferation and metastasis
in vitro. Moreover, we demonstrated that ZBTB10 transcriptionally activated ARRDC3, which then directly bound to ITGB4, leading to its ubiquitination and degradation. This process ultimately reduced the phosphorylation levels of PI3K and AKT. In all, ARRDC3 is indispensable for the inhibition of PI3K/AKT pathway and GC progression regulated by ZBTB10, elucidating the role of the ZBTB10/ARRDC3/ITGB4 axis in this context.
Currently, the prognosis for the patients with advanced GC remains poor due to high recurrence rates, metastasis, and the lack of effective treatments [
42]. The search for novel, safe, and effective therapeutic agents is ongoing. Recent studies have highlighted the potential of small natural compounds as effective anti-tumor agents [
43]. ZBTB10 has also been identified as a target for several anti-cancer drugs [
29,
30], prompting us to investigate its potential as a target for anti-GC drugs using the CMap database. The results suggested that ZBTB10 might be a key target for BA in inhibiting GC progression. BA, a natural pentacyclic terpene, has demonstrated anti-cancer activity against various malignancies, including GC [
18‐
20]. Moreover, derivatives of BA have been used for the clinical treatment of numerous cancers [
44,
45]. Notably, BA has also been reported to play a role by promoting ZBTB10 expression in other cancers [
16,
46]. However, this has not been reported in GC. Furthermore, the effect of BA on GC and its biosafety in vivo have not been reported. Our study revealed, for the first time, that BA inhibited GC tumorigenicity and metastasis in vivo and was safe in mice, providing strong evidence for its potential clinical use. In addition, we also demonstrated that ZBTB10 was one of the targets for BA in inhibiting GC progression. Combined with all of the above data, we believe that ZBTB10 is a novel therapeutic target and BA might act as feasible therapeutic strategy for GC.
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.