1. INTRODUCTION
Gastric cancer (GC), a common malignancy of the alimentary system, is the third leading cause of tumor-associated death worldwide [1]. It has the highest annual incidence in East Asian countries compared to other regions in Asia [2]. Approximately 679,000 patients are newly diagnosed with GC in China each year. Owing to its large population, China accounts for most patients with GC in East Asia [3]. Despite the substantial improvements made in surgical techniques and medications for GC over the past decade, the 5-year survival rate for patients with advanced GC is poor, at <15% [4]. Identifying new drugs remains the main strategy for exploring new treatment options for tumors, including GC.
Signal transducers and activators of transcription (STAT) is a family of nucleocytoplasmic shuttling transcription factors responsible for the signal transduction of extracellular cytokines and the activation of gene transcription [5]. This family includes STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b, and STAT6, which have relatively conserved homologous functional domains. All seven STAT proteins play central roles in cellular immune responses, cell survival, differentiation, angiogenesis, and motility [6]. STAT3 is the most widely studied member of the STAT family, and the most closely associated with tumorigenesis; it is a key regulatory molecule in various oncogenic signaling pathways [7]. Abnormally activated STAT3 is present in more than 70% of human cancers [8, 9], including breast, lung, liver, stomach, brain, colon, cervix, and prostate cancers, and acute myeloid leukemia and multiple myeloma [7]. The protein structure of STAT3 contains an N-terminal domain, a coiled-coil domain, a DNA-binding domain, a linker region, an SH2 domain, and a C-terminal transactivation domain, similarly to other STAT family members [10]. STAT3 exists as an inactive monomer in the cytoplasm. After the tyrosine residues of JAKs are phosphorylated by upstream growth factor receptors (e.g., gp130 and EGFR), the SH2 domain of STAT3 recognizes and binds these phosphorylated-tyrosine docking sites, thus placing STAT3 in proximity to activated JAKs; phosphorylation then occurs at Y705 in the C-terminal transactivation domain. Phosphorylation of Y705 promotes SH2 domain-mediated head-to-tail dimerization of STAT3 followed by translocation to the nucleus. After entry into the nucleus, STAT3 binds cognate response elements in target gene promoters, thereby regulating the expression of regulatory transcription factors promoting cell growth (such as cyclin D1), survival (such as survivin), and cell invasion and migration (such as MMP proteins and vimentin) [11]. In addition, studies increasingly indicate that constitutive activation of STAT3 promotes tumor-induced immune suppression at several levels [12]. Given the above characteristics, STAT3 has been widely studied in tumor malignant progression and as a tumor target.
The herbaceous perennial Macleaya cordata (Wild.) R. Br. belongs to the Papaveraceae family and is ubiquitously found in China, North America, and Europe [13, 14]. M. cordata is recorded in many ancient books as a traditional Chinese medicine. In recent years, studies have reported that it has a variety of pharmacological activities, such as antifungal, antimicrobial, anti-inflammatory, antioxidant, and antitumor activities [15–17]. Pharmacological studies have shown that the main biologically active ingredient of M. cordata is alkaloids. To date, 147 alkaloids have been identified and/or isolated in M. cordata. Most are isoquinoline alkaloids, including sanguinarine, chelidonine, allocryptine, dihydrochelerythrine, oxysanguinarine, and dihydrosanguinarine [18, 19]. The compound 6-ethoxydihydrosanguinarine (6-EDS, Figure 1a ) is a secondary transformation product formed after dihydrosanguinarine is extracted from M. cordata [20]. Because the effect and mechanism of 6-EDS in GC was unclear, herein, we investigated the effects of 6-EDS in GC.
2. MATERIALS AND METHODS
2.1 Reagents
The compound 6-EDS, with a purity of ≥ 98%, was purchased from BioBioPha Co., Ltd. (Kunming, China).
2.2 Cell culture
Human GC cell lines BGC823, MGC803, and AGS were purchased from the ATCC, and were maintained in Dulbecco’s modified Eagle’s medium (Gibco, USA) supplemented with 10% FBS and antibiotics, and incubated in a humidified atmosphere with 5% CO2 at 37°C.
2.3 Cell cytotoxicity assays
Cell cytotoxicity was determined with MTT assays and xCELLigence real time cell analysis (RTCA) assays. For MTT assays, cells were seeded into 96-well plates, then treated with 6-EDS. The absorbance was determined at 490 nm with an automated microplate reader (BioTek, USA) [21]. For RTCA assays, the cells were seeded into 16-well E-Plates (ACEA Biosciences, USA), and an xCELLigence RTCA HT Instrument (ACEA Biosciences, USA) was used for label-free, real-time, automated monitoring. After 24 h, the cells were treated with 6-EDS. The cell index analysis (15 min interval detection) lasted for 60 h.
2.4 Soft-agar colony-formation assays
Soft-agar colony-formation analysis was performed as previously described [22].
2.5 High-content analysis
The Operetta CLS™ high-content analysis system (PerkinElmer, Germany) was used to monitor the movement potential of cells. Cell tracks were recorded in real time to dynamically determine the migration. Cells were seeded into 96-well plates, and cell movement was recorded overnight in real time. The paths of the cells were automatically analyzed in Harmony 4.1 software.
2.6 Annexin V/PI double staining and apoptosis detection
Cells were incubated with 6-EDS for 24 h. Cell apoptosis was detected through annexin V/PI detection with an annexin V/PI kit (Dojindo, Japan). Flow cytometry was performed with a flow cytometer (Beckman, USA) [23].
2.7 Mitochondrial membrane potential measurement
After incubation with 6-EDS for 24 h, cells were collected and stained with the JC-1 fluorescent probe. After incubation for 20 min, a flow cytometer (Beckman, USA) was used to detect fluorescence.
2.8 DAPI staining
For observation of nuclear morphology, cells were fixed and then stained with DAPI (Beyotime, China). The images of stained cells were obtained with a fluorescence microscope (Olympus, Japan).
2.9 Western blotting
Western blotting was conducted according to the standard method described previously [24]. The primary antibodies used were anti-GAPDH, anti-MMP9, anti-cyclin D1, anti-JAK2 (Santa Cruz Biotechnology, USA); anti-phospho-STAT3 (Y705), anti-phospho-STAT3 (S727), anti-STAT3, anti-phospho-JAK2 (Y1007/Y1008), anti-E-cadherin, anti-caspase-9, anti-PARP, anti-caspase-3, anti-PARP, anti-phospho-EGFR (T1068), anti-EGFR (Cell Signaling Technology, USA); and anti-c-Myc, anti-MMP2, anti-vimentin, anti-Lamin B1 (Proteintech Group, USA).
2.10 Wound-healing assays
Wound-healing assays were performed as previously described [25]. ImageJ software was used for measuring the wound gaps.
2.11 xCELLigence real-time cellular analysis assay for cell invasion
The upper chamber of a 16-well CIM-Plate (ACEA Biosciences, USA) was pre-coated with 1 mg/mL Matrigel (BD Biosciences, USA) for 4 h. Cells (8 × 103 cells per well) in serum-free medium were seeded in the upper chambers, and complete medium containing 20% FBS was added to the lower chambers. Subsequently, an RTCA DPlus Instrument (ACEA Biosciences, USA) was used to detect the cell index (15-minute interval detection) for 50 h.
2.13 Real-time quantitative PCR
Real-time quantitative PCR (qPCR) was performed on an ABI StepOnePlus™ Real-Time PCR System (ABI, USA) with SYBR® Green Real-time PCR Master Mix (Toyobo Co., Ltd., Japan). Primers are listed in Table 1 .
2.14 Isolation of nuclear and cytoplasmic fractions
Nuclear and cytosolic fractions were separated with a Nuclear and Cytoplasmic Protein Extraction Kit (Beyotime Biotechnology, China). Western blotting was used to detect the expression of STAT3, Lamin B1, and GAPDH.
2.15 Immunofluorescence assays
Cell or tissue sections were fixed in 4% formaldehyde, then blocked with 3% BSA/0.2% Triton X-100 in PBS. Subsequently, cells were incubated with antibody to STAT3, then with Alexa Fluor® 488-conjugated secondary antibody (Abbkine, China). Next, the slides were stained with DAPI (Beyotime Biotechnology, China). Fluorescence images were acquired on an Olympus laser scanning confocal microscope.
2.16 Transfection of DNA
The pcDNA3.1-EGFP-STAT3-Myc expression plasmid was purchased from HeBio Co. Ltd. (Shanghai, China). Plasmids were transfected with Lipofectamine® 3000 transfection reagent (Invitrogen, USA).
2.17 Molecular docking
The structure of the phosphorylated STAT3 core protein bound to DNA was obtained from the RCSB PDB database (PDB ID: 6NJS). Wild-type STAT3 was obtained by back-mutation to unphosphorylated tyrosine with the mutagenesis model in PyMOL. The missing DNA strands were repaired by base-pairing and modeled with the 3DNA webserver [26]. Then the structure was prepared by removal of hydrogen assignments at pH 7.0 with academic Maestro [27]. Water molecules near small-molecule-binding pockets were retained. Ligand-protein docking experiments were conducted mainly with Autodock Vina. The search space at the binding pocket was a rectangle of 22.5 Å × 22.5 Å × 22.5 Å in the X, Y, and Z dimensions. The docked poses were clustered with a tolerance of 2 Å root-mean-square deviations and were evaluated for the final docking structure according to the docking score.
2.18 Molecular dynamic simulation
Molecular dynamics (MD) simulations were performed with the GROMACS 5.1.5 program [28]. We constructed four systems to estimate the effects of 6-EDS on STAT3-DNA binding: (i) wild-type STAT3 with DNA (wt-STAT3_DNA), (ii) 6-EDS compound with wild-type STAT3 and DNA (6-EDS_wt-STAT3_DNA), (iii) 6-EDS with phosphorylated STAT3 (6-EDS_Y2P-STAT3), and (iv) 6-EDS with phosphorylated STAT3 and DNA (6-EDS_Y2P-STAT3_DNA). Every simulation system was solved with TIP3P waters and neutralization by Na+ and Cl- ions. The periodic boundary conditions had a minimal distance of 1.0 Å between the molecules and the edge of the box. [29]. The ff99SB force field was used for STAT3 and DNA, and the GAFF force field was used for 6-EDS topology [30]. Simulation systems were subjected to constant number of particles, volume, and temperature (NVT) equilibration, and constant number of particles, pressure, and temperature (NPT) equilibration. Finally, MD was performed for a period of 50 ns with 2-fs steps. The trajectory information was collected every 2 ps [31].
2.19 Binding-free-energy calculation
The binding free energies (ΔGbind) were calculated with the MM/GBSA model [32]. The GROMACS output trajectory was correctly fitted, and PBC conditions were removed before the calculations were run with gmx_MMPBSA. ΔGbind was calculated as follows:
(1)
(2)
(3)
Here, ΔGsolv refers to solvation free energy; ΔGGAS refers to the gas-phase energy; TΔS refers to changes in the conformational entropy upon binding; and ΔEint refers to the bond, angle, and dihedral energies, which were usually taken as zero. ΔEele refers to electrostatic; ΔEvdw refers to van der Waals energies; and ΔGsolv refers to the sum of the no-electrostatic-solvation component (nonpolar contribution, e.g., ΔESURF) and the electrostatic solvation energy (polar contribution, e.g., ΔGPB). TΔS was also omitted in this method, owing to its intrinsically low prediction accuracy [33, 34].
2.20 Microscale thermophoresis assays
Purified human STAT3 protein was labeled with a Monolith™ NT.115 Protein Labeling Kit (NanoTemper Technologies GmbH, Germany). Cells transfected with EGFP-STAT3 expression plasmid were lysed in RIPA buffer to isolate total protein. Subsequently, 16 serial dilutions of 6-EDS were prepared, ranging from 1.25 mM to 0.038 μM. The labeled STAT3 or cell lysates containing EGFP-STAT3 protein were quantified, then incubated with 6-EDS for 10 min in the dark. Samples were loaded into standard capillaries in the NanoTemper Monolith™ NT (NanoTemper Technologies GmbH, Germany). The dissociation constant (K d) was then calculated.
2.21 Drug-affinity-responsive-target stability assays
Cells were harvested and lysed with lysis buffer (Sigma, USA). Lysates were treated with 6-EDS and then incubated with pronase (Sigma, USA) at 37°C for 30 min. The hydrolyzation of STAT3 was detected by western blotting [35].
2.22 Cellular thermal-shift assays
Cells were harvested and lysed with lysis buffer (Sigma, USA). Lysates were then heated at 35–55°C after incubation with 6-EDS for 30 min. The stabilizing effect of 6-EDS on STAT3 was detected by western blotting.
2.23 Xenograft experiments
Female nude mice 5–7 weeks old were purchased from Vital River Laboratory (Beijing, China) and monitored in an SPF environment. The mice were injected subcutaneously with 6 × 106 BGC823 cells. Before treatment, mice were randomly divided into two groups: a vehicle group (0.8% DMSO, 12% cremophor, and 8% ethanol in saline; n=7) and 6-EDS-treated group (intraperitoneal injection of 2.0 mg/kg 6-EDS; n=7). The mice were treated four times per week for a total of 4 weeks. Caliper measurements were performed twice per week with the following formula: 4π/3 × (width/2)2 × (length/2). Mice were sacrificed when their tumors reached 1.5 cm, or they appeared moribund. Subcutaneous tumors were excised for western blotting, or were fixed and then embedded in paraffin for H&E staining and immunohistochemistry.
2.24 Statistical analysis
All experiments were repeated at least three times, and the data are presented as mean ± SD. Differences between data groups were evaluated for significance with Student’s t-test for unpaired data or one-way analysis of variance and Bonferroni post-test. P < 0.05 indicates statistical significance.
3. RESULTS
3.1 6-EDS suppresses the growth of GC cells
We investigated the effects of 6-EDS on GC cell lines. The IC50 values of 6-EDS in AGS, BGC823, and MGC803 cells were 5.86 μM, 6.43 μM, and 5.48 μM, respectively, at 24 h ( Figure 1b ). We performed RTCA assays to monitor the proliferation ability of cells. The RTCA assays indicated that 6-EDS decreased the growth rates of AGS, BGC823, and MGC803 cells ( Figure 1c ). Next, colony-formation assays demonstrated that 6-EDS markedly decreased the clonogenic ability of AGS, BGC823, and MGC803 cells ( Figure 1d ). High-content imaging technology was used to track the morphology of cells. The number of tracked cells decreased after treatment with 6-EDS, and the mean roundness of cells treated with 6-EDS had increased at 24 h ( Figure 1h, 1i ).
3.2 Mitochondrially mediated apoptosis is induced by 6-EDS in GC cells
Apoptosis was further analyzed in AGS, BGC823, and MGC803 cells. Annexin V/PI staining and flow cytometry assays showed that 6-EDS induced apoptosis of AGS, BGC823, and MGC803 cells ( Figure 2a ). DAPI staining further confirmed that 6-EDS induced chromatin condensation and fragmentation in AGS, BGC823, and MGC803 cells ( Figure 2b ). Next, the mitochondrial membrane potential (MMP) was measured. The red JC-1 aggregates indicated intact MMP, whereas green JC-1 monomers indicated damaged MMP. Fluorescence detection showed that 6-EDS treatment decreased the number of red fluorescent cells and increased the number of green fluorescent cells. Therefore, the ratio of red/green JC-1 fluorescence decreased ( Figure 2c ), thus suggesting that 6-EDS induced MMP damage in GC cells. Furthermore, expression of caspase and apoptotic protein was detected by western blotting, thus indicating that 6-EDS induced a decrease in the precursor form of caspase-3 (pro-casp3) and caspase-9 (pro-casp9), and induced cleavage of PARP in AGS, BGC823, and MGC803 cells ( Figure 2d ). These data suggested that 6-EDS induced mitochondrially mediated intrinsic apoptosis in GC cells.
3.3 The migratory and invasive behavior of GC cells is suppressed by 6-EDS
High-content imaging technology was used to track the movement of AGS, BGC823, and MGC803 cells. The cell migration and displacement area decreased significantly with increasing 6-EDS concentration ( Figure 3a ). Wound-healing and Transwell assays were also performed to measure the migratory ability. The 6-EDS treatment significantly inhibited the migration of AGS, BGC823, and MGC803 cells ( Figure 3b, 3c ). Next, we used RTCA assays to monitor real-time changes in cell invasion by examining the cell index. Under increasing 6-EDS concentrations, the cell-invasion curve decreased significantly ( Figure 3d ). In Transwell invasion assays, 6-EDS treatment significantly decreased the number of invasive cells ( Figure 3e ). Therefore, 6-EDS significantly decreased the metastatic ability of GC cells.
3.4 GC progression is suppressed by 6-EDS through inhibition of STAT3 activity
The mechanism of 6-EDS in inhibiting the malignant progression of GC was further investigated. Western blotting analysis indicated that 6-EDS down-regulated the expression of several proteins associated with cell malignant proliferation, migration and invasion, and anti-apoptosis, including cyclin D1, c-Myc, MMP2, MMP9, and survivin ( Figure 4a ). Moreover, 6-EDS reversed the EMT of GC cells, as manifested primarily in the down-regulation of vimentin and the up-regulation of E-cadherin expression ( Figure 4a ). Furthermore, qPCR revealed that 6-EDS inhibited the mRNA levels of MMP2, vimentin, c-Myc, and cyclin D1 ( Figure 4b ). These four genes share a common upstream transcription factor, STAT3 [36]. Therefore, we investigated the expression and activation of STAT3. The results suggested that 6-EDS significantly decreased the phosphorylation of Y705 of STAT3, but did not alter the phosphorylation of S727 or the total protein expression level of STAT3 ( Figure 4c ). Moreover, the expression and phosphorylation of JAK2, a direct regulatory molecule upstream of STAT3, was not affected by 6-EDS ( Figure 4c ), thus suggesting that 6-EDS may directly suppress the activation of STAT3. The transcriptional activity of STAT3 was further detected by nucleocytoplasmic separation, and 6-EDS was found to inhibit the levels of phosphorylated STAT3 in the nucleus and cytoplasm ( Figure 4d ). Cytokines and growth factors (e.g., IL-6 and EGF) promote the phosphorylation and activation of STAT3 through their receptors (gp130 and EGFR), thereby promoting the malignant progression of tumors [6]. Further experiments indicated that exogenous IL-6 stimulation promoted STAT3 activation in GC cells, whereas pretreatment with 6-EDS overcame the effects of IL-6 ( Figure 4e, 4f ). Moreover, 6-EDS overcame the promoting effect of IL-6 on the expression of STAT3 target genes (cyclin D1, c-Myc, and vimentin) ( Figure 4e ). Although 6-EDS had little effect on EGF-stimulated phosphorylation of EGFR ( Figure 4g ), this treatment overcame the activation effect of EGF on STAT3, and further decreased the EGF-mediated promotion of the expression of cyclin D1, c-Myc, and vimentin ( Figure 4g ). These data demonstrated that 6-EDS inhibited the expression of downstream target genes associated with tumor malignant progression by inhibiting STAT3 activity. Next, we determined whether the sensitivity of cells to 6-EDS cytotoxicity was associated with the level of STAT3 expression. GC cells were transfected with the EGFP-STAT3-Myc expression plasmid ( Figure 4h ). Overexpression of STAT3 significantly antagonized the inhibitory effects of 6-EDS on GC cell proliferation, migration, and invasion ( Figure 4i, 4j ). Together, the above results suggested that 6-EDS inhibited the malignant progression of GC by directly inhibiting the activity of STAT3.
3.5 The compound 6-EDS directly binds and inhibits STAT3
We further investigated the above in vitro results in silico. The protein structure of STAT3 is shown in Figure 5a . Molecular docking suggested that 6-EDS binds the SH2 domain of the STAT3-DNA complex ( Figure 5b, 5c ), and several water molecules mediate the binding of 6-EDS to STAT3. Two water molecules form water bridges between the side chain of K591 and the oxethyl group of 6-EDS. One water mediates the binding of S611 and S613 with the 1,3-benzodioxole of 6-EDS via a water bridge. Another 1,3-benzodioxole group forms a hydrogen bond with the side chain of R609. Hydrophobic interactions occur between the nitrogen methyl group of 6-EDS and the side chains of residues V637 and P639 ( Figure 5d ). The binding energy of 6-EDS to STAT3 is −6.8 kcal/mol. Next, we performed MD simulations on the 6-EDS–STAT3 complex. The root-mean-square deviation was used to explore the effects of 6-EDS on the binding stability of STAT3 protein. No significant structural differences were observed in short-term simulations. The four systems showed similar trends in RMSD of the protein skeleton, and their average values of RMSD maintained approximately 0.45 nm in the second half of the simulation ( Figure 5e ). Although previous studies have shown that STAT3 binds DNA only after phosphorylation, recent studies have reported that unphosphorylated STAT3 can also bind DNA [37]. Next, the structural alignment showed that phosphorylated STAT3 and unphosphorylated STAT3 are structurally very similar, except for Y705 ( Figure 5f ). We additionally estimated the effects of 6-EDS on the DNA binding of STAT3. As shown in Table 2 , ΔGbind indicated that unphosphorylated STAT3 (wt-STAT3_DNA) and phosphorylated STAT3 (Y2P-STAT3_DNA) bind DNA with similar energies. Interestingly, after 6-EDS binding, the binding energy of DNA with both systems decreased. The binding energy of phosphorylated STAT3 with DNA decreased from –225.66 to –114.57 kcal/mol. The binding energy of unphosphorylated STAT3 with DNA decreased from –217.85 to –105.36 kcal/mol ( Figure 5g ). These results indicated that the binding of 6-EDS indeed affects the binding ability of phosphorylated or unphosphorylated STAT3 and DNA. From the viewpoint of energy decomposition, the binding of 6-EDS mainly affects the van der Waals interactions and electrostatic interactions between STAT3 and 6-EDS. Collectively, in silico validation through dynamics simulations of the docked drug-target complex confirmed the in vitro results of the effective inhibition of STAT3 by 6-EDS.
Energy component | Systems | |||||
---|---|---|---|---|---|---|
wt-STAT3_DNA | Y2P-STAT3_DNA | 6-EDS_wt-STAT3 | 6-EDS_Y2P-STAT3 | 6-EDS_wt-STAT3_DNA | 6-EDS_Y2P-STAT3_DNA | |
ΔEvdw | −63.86 ± 4.44 | −66.19 ± 14.26 | −40.10 ± 3.30 | −29.81 ± 3.60 | −46.14 ± 4.81 | −35.55 ± 7.94 |
ΔEele | −1035.82 ± 96.01 | −1270.16 ± 111.34 | −18.06 ± 7.89 | −15.96 ± 3.77 | −1270.52 ± 76.17 | −890.26 ± 87.68 |
ΔEGB | 890.92 ± 94.95 | 1119.66 ± 106.10 | 19.23 ± 5.46 | 8.77 ± 3.93 | 1219.46 ± 71.51 | 816.27 ± 87.97 |
ΔESURF | −9.08 ± 0.47 | −8.99 ± 1.92 | −5.04 ± 0.30 | −3.48 ± 0.45 | −7.16 ± 0.46 | −5.06 ± 1.23 |
ΔGGAS | −1099.69 ± 96.42 | −1336.34 ± 103.66 | −59.17 ± 8.84 | −45.77 ± 5.51 | −916.66 ± 76.46 | −925.78 ± 92.47 |
ΔGsolv | 881.83 ± 94.97 | 1110.68 ± 107.01 | 14.19 ± 5.32 | 5.28 ± 3.79 | 811.31 ± 71.31 | 811.21 ± 81.27 |
ΔGbind | −217.85 ± 5.81 | −225.66 ± 9.00 | −44.98 ± 4.63 | −40.49 ± 3.00 | −105.36 ± 8.19 | −114.57 ± 10.55 |
To validate the interaction between 6-EDS and STAT3, and to quantify the binding affinity, we performed microscale thermophoresis (MST) assays. The MST quantification analysis revealed the binding ability of 6-EDS and STAT3 with a K d of 76.42 μM and a signal-to-noise ratio of 11.4 ( Figure 5h ). The binding of small molecules promotes the stability of the target protein. Drug-affinity-responsive-target stability assays were conducted to detect the stability of STAT3. Incubation with 6-EDS led to a concentration-dependent decrease in STAT3 proteolysis ( Figure 5i ). Cellular thermal-shift assays (CETSAs), performed to further confirm the effect of 6-EDS on STAT3 stability, indicated that 6-EDS facilitated STAT3 thermotolerance in a temperature gradient (35–55°C) ( Figure 5j ). Collectively, these data demonstrated that 6-EDS directly bound STAT3. To further confirm the critical amino acid residues involved in STAT3 binding to 6-EDS, we performed site-directed mutagenesis affecting the K591, R609, S611, and S613 residues in the EGFP-STAT3 plasmid, according to the molecular docking prediction. The EGFP, EGFP-STAT3, EGFP-STAT3K591A, EGFP-STAT3R609A, EGFP-STAT3S611A, and EGFP-STAT3S613A plasmids were transfected into HEK293T cells, and the total protein was extracted and incubated with 6-EDS. MST assays indicated that EGFP-STAT3 bound 6-EDS, whereas EGFP did not ( Figure 5k ). In addition, the K591A, R609A, S613A mutations had no significant effects on the binding of STAT3 and 6-EDS. K591A and R609A of STAT3 still bound 6-EDS, but the K d of binding increased significantly, thus indicating a weakened binding effect. In contrast, the S611A mutation inhibited the binding of STAT3 to 6-EDS, thereby suggesting that S611 plays a key role in the binding of STAT3 to 6-EDS. Conclusively, the above results indicated that 6-EDS suppresses the activity of STAT3 and its binding DNA by directly binding the SH2 domain.
3.6 The compound 6-EDS hinders tumor growth in vivo
We next detected the inhibitory effect of 6-EDS on tumor growth in vivo. After generation of BGC823 murine xenograft models, 6-EDS (2 mg/kg, i.p.) was administered for 4 weeks. Subsequently, the mice were sacrificed ( Figure 6a ). In agreement with the in vitro proliferation data, 6-EDS significantly inhibited the of tumor growth in vivo ( Figure 6b–e ). As shown in Figure 6f , treatment with 6-EDS did not cause clear changes in the weights of mice. Diminished Ki67 intensity was observed in the 6-EDS-treated xenograft tumors, as demonstrated by immunohistochemical staining ( Figure 6g ). In addition, fluorescence immunohistochemistry and western blotting suggested that 6-EDS inhibited the phosphorylation of STAT3 ( Figure 6h, 6i ). Furthermore, the expression of target genes downstream of STAT3 associated with proliferation, invasion, and migration in tumor tissue was detected by qPCR, thus indicating that 6-EDS significantly decreased the mRNA expression of MMP2, vimentin, cyclin D1, and c-Myc in tumor tissue ( Figure 6j ). The above results suggested that 6-EDS inhibited tumor progression in vivo through STAT3.
Overall, we concluded that 6-EDS inhibits the malignant progression of GC mainly by directly binding the SH2 domain of STAT3, thus inhibiting the phosphorylation and transcriptional activity of STAT3.
4. DISCUSSION
The critical role of STAT3 in malignant tumorigenesis and progression has sparked research in targeted drug discovery to screen small molecules that perturb STAT3 activity. Here, we examined a novel small-molecule STAT3 inhibitor, 6-EDS, which has shown great promise in pre-clinical evaluations of GC treatment. We observed clear toxicity effects of 6-EDS on GC cell survival, including promoting cell apoptosis, and decreasing invasion and migration. Moreover, 6-EDS decreased tumor burden in GC xenografts in vivo. These anti-tumor effects were exerted by 6-EDS binding the SH2 domain, and inhibiting the binding of phosphorylated and non-phosphorylated STAT3 to DNA.
We further demonstrated that 6-EDS inhibited GC cell viability and induced GC cell apoptosis by affecting the mitochondrial apoptosis pathway mediated by caspase-9. The down-regulation of the pro-survival molecule survivin downstream of STAT3 by 6-EDS may be involved in the activation of the above apoptotic signals [38]. We used RTCA and a high-content imaging system to demonstrate that 6-EDS significantly inhibited the viability, area, and motility of GC cells. Increased cell roundness indicates decreased cellular activity and migratory ability [39]. Various cytokines, growth factors, and intracellular signaling pathways promote tumor cell survival and malignant progression by activating STAT3 signaling [10]. We detected IL-6 and EGF signaling upstream of STAT3 and the key regulatory molecule JAK2, thus demonstrating that 6-EDS down-regulated the expression of downstream target genes associated with invasion, migration, and EMT by directly inhibiting STAT3 activity. Knockdown of STAT3 has been widely reported to significantly inhibit GC cell growth, EMT, and consequent tumor invasion and migration [40, 41]. Our investigation further demonstrated that overexpression of STAT3 significantly antagonized the inhibitory effect of 6-EDS on GC cell malignant progression, thus suggesting that 6-EDS inhibits GC malignant progression at least partly by inhibiting STAT3.
Currently, the core strategies for targeting STAT3 include suppressing STAT3 phosphorylation, binding the SH2 domain, disrupting STAT3-DNA binding, or suppressing STAT3 transcriptional activity [42]. Targeting the SH2 domain is the main method to screen for STAT3 inhibitors, but targeting the SH2 domain to inhibit STAT3 phosphorylation and dimerization may not completely inhibit abnormal STAT3 signal transduction. Recently, studies have reported that STAT3 participates in transcriptional regulation without tyrosine phosphorylation; that is, non-phosphorylated STAT3 dimers bind DNA and promote transcription [43, 44]. Inhibition of phosphorylated and non-phosphorylated STAT3 binding DNA may represent another effective method to eliminate STAT3 signaling. Molecular docking analysis showed that the best docking orientation for 6-EDS binding STAT3 was the SH2 domain. The 6-EDS bound the SH2 domain and inhibited the phosphorylation of Y705, thereby repressing STAT3 dimerization. The dimerization region of STAT3 covers the SH2 domain and the 688–722 aa region of the transactivation domain, where Y705 resides. The phosphorylation of Y705 is necessary for dimerization [42]. Binding of 6-EDS decreased the dimerization and nuclear entry of STAT3, and may also be detrimental to the phosphorylation of STAT3 through structural changes. The energy dissipation-cum-signaling mechanism suggests that local structural changes or binding ligands are thermodynamically coupled to the distal activation site [45]. The conformational changes caused by mutation or ligand binding can be 20 Å or more, thus resulting in identifiable functional consequences on kinase activity [46]. The binding of 6-EDS in the SH2 domain may be thermodynamically coupled to the Y705 site through distal residues, thereby affecting the degree of phosphorylation of Y705. Moreover, MD simulations indicated that, although 6-EDS did not bind the DNA binding domain, it decreased the binding free energy of phosphorylated STAT3 and non-phosphorylated STAT3 to target gene DNA. Binding free energy is dominated by electrostatic and hydrophobic interactions. Moreover, 6-EDS has strong electronegativity and thus binds basic amino acids, such as K591 and R609. With the participation of water molecules, the side chain of S611 formed a hydrogen bond with 6-EDS. As a polar hydrophilic amino acid, S611 has no other interactions, an aspect potentially important in recognizing intermolecular binding. Furthermore, we demonstrated the direct binding effect of 6-EDS and STAT3 and key amino acid sites through MST, CETSA, and drug-affinity-responsive-target stability assays. Although site-directed mutagenesis indicated that the S611 site of STAT3 played a critical role in binding 6-EDS, the affinity of 6-EDS toward the STAT3 protein must be further improved.
Conclusively, our data reveal that 6-EDS is a novel STAT3 inhibitor, and demonstrate that 6-EDS has a significant inhibitory effect on the survival, invasion, and migration of GC cells, and activates mitochondrial apoptosis. These effects are associated with direct inhibition of STAT3 by 6-EDS. This study may provide an experimental foundation for antitumor targeted therapy based on 6-EDS.