Radian Sophorae flavescentis is a traditional Chinese medicine commonly used to treat cancer in China. However, its active components and underlying mechanism remain ambiguous. In this study, we have screened the pharmacokinetic parameters of the main chemical constituents of Radian Sophorae flavescentis by Traditional Chinese Medicine Systems Pharmacology (TCMSP) Database and Analysis Platform and have found that Sophoridine is one of the best antitumor active ingredients. We have found that MAPKAPK2 is a potential target for Sophoridine by the PharmMapper and KEGG databXase analysis. Moreover, we have found that Sophoridine selectively inactivates phospho-MAPKAPK2 (Thr222) and directly binds into the ATP site of MAPKAPK2 by molecular docking. Furthermore, we have found out a direct binding between MAPKAPK2 and Sophoridine by cellular thermal shift assay and drug affinity responsive targets stability assay. The inhibition effects are further confirmed by Western blot: Sophoridine significantly decreases phospho-MAPKAPK2 (Thr222) in a time-dependent manner, but there is no obvious change in its total expression in colorectal cancer cells. Clinical studies have shown that a higher level of MAPKAPK2 is associated with a poorer percent survival rate (prognosis). Furthermore, a higher level of MAPKAPK2 is positively associated with the enrichment of downregulation of apoptosis and autophagy by gene set enrichment analysis, as well as upregulation of proliferation and cell-cycle arrest. Taken together, our results suggest that the MAPKAPK2 plays a key role in Sophoridine-inhibited growth and invasion in colorectal cancers.

Implications:

These studies show that Sophoridine may be a promising therapeutic strategy that blocks tumorigenesis in colorectal cancers.

Colorectal cancer is one of the most frequent causes of cancer-related morbidity and mortality globally (1–3). Despite the benefits of high-quality early screening and detection, surgery, and new chemotherapeutic agents for improving the treatment of advanced and metastatic colorectal cancers, the 5-year survival rate for advanced colorectal cancers is less than 10% (4). There is still a lack of optimal treatment strategies for colorectal cancers. Currently, anti-EGFR agents (bevacizumab, ramucirumab, regorafenib, ziv-aflibercept) are used in combination with chemotherapy as a standard of care for the first-line therapy of metastatic colorectal cancer (5–7). Although these agents are designed to restrain tumor-selective proliferation, they can cause serious toxic effects, affect normal tissues, and sometimes severely interfere with therapeutic success and the quality of life of patients (8). Therefore, the development of novel, effective treatment approaches is urgently needed to improve clinical outcomes of colorectal cancer patients.

Traditional Chinese medicine (TCM) has a long history of application and a significant contribution to modern medicine (9, 10). As important sources of active natural products, TCM shows unique advantages (11–13). Radix Sophorae Flavescentis (the dried roots of Sophora Flavescens Ait) is widely used in China, Japan, and some European countries for its various physiologic functions (14, 15). It contains several major effective components such as Matrine, Sophoridine, Oxymatrine, etc. Sophoridine, an active quinolizidine alkaloid compound, displays various biological properties such as anticancer activity, antiviral activity, antifibrotic activity, antimicrobial activity, antiinflammatory activity, etc. (16–19). In particular, recent studies have revealed that it exhibits potent anticancer effects in different tumor cell lines and animal models (20); however, the exact underlying mechanism of the anticancer effect of Sophoridine is remaining unclear.

In this study, we screened the pharmacokinetic parameters of the main chemical constituents of Radian Sophorae flavescentis by Traditional Chinese Medicine Systems Pharmacology (TCMSP) Database and Analysis Platform database and found that Sophoridine is one of the best antitumor active ingredients. We have identified that Sophoridine is an effective inhibitor of MAPKAPK2 by directly interacting with the ATP site of MAPKAPK2, leading to the repression of multiple oncogenic processes in colorectal cancers. Clinical studies have shown that a higher level of MAPKAPK2 is associated with a poorer percent survival rate (prognosis). Furthermore, a higher level of MAPKAPK2 was positively associated with the enrichment of downregulation of apoptosis and autophagy by gene set enrichment analysis (GSEA), as well as upregulation of proliferation and cell-cycle arrest.

Taken all together, these results show that Sophoridine may be recognized as an attractive drug candidate by targeting MAPKAPK2 for colorectal cancers therapy.

Cell culture

Human colorectal cancer cell lines HCT116, RKO, and SW480 were obtained from Cell Resource Center of the Chinese Academy of Sciences in 2017 (CAMS, PUMC, Beijing, China). The cells being used were used within 1 month after resuscitation (passage number between 9 and 30). The cell lines were identified using a short tandem repeat analysis and tested for mycoplasma using MycoAlert (Lonza) in these cell lines. All cell lines were cultured in DMEM supplemented with 10% FBS and 1% penicillin/streptomycin in a humidified atmosphere under 5% CO2 at 37°C. Sophoridine (purity ≥98%) was purchased from Shanghai Yuanye Bio-Technology and dissolved in dimethyl sulfoxide (DMSO) to prepare a 10 mmol/L stock solution for storage at −20°C.

Cell viability assay

Cell viability was measured using CCK-8 assay (Dojindo). Human colorectal cancer cells were seeded into flat-bottom 96-well plates (5 × 103 cells/well) and treated with Sophoridine at indicated concentrations for another 48 hours after plating for 24 hours. Subsequently, medium was discarded and added solution of cell counting to each well, followed by 1 hour of incubation. The absorbance was detected at 450 nm using a microplate reader.

Colony formation assay

Human colorectal cancer cells (1 × 103) were seeded into 6-well plates. After 24 hours, cells were treated with Sophoridine at the indicated concentrations for 48 hours. Cells were then cultured in fresh medium for another week. Colonies were fixed with 4% paraformaldehyde and stained with 0.1% crystal violet. Photographs were acquired in indicated time periods, and the cell numbers were counted.

Immunofluorescence assay

Cells were seeded in 24-well plates and treated with Sophoridine at indicated concentrations for 48 hours. The cells were washed in cold PBS and then fixed with 4% paraformaldehyde followed by 5 minutes of permeabilization with 0.1% Triton X-100. Then we blocked with 1% BSA containing 1% goat serum for 30 minutes. After incubation with anti-LC3 overnight at 4°C, cells were exposed to corresponding secondary antibodies for 1 hour at room temperature, and then stained with DAPI (4′,6-diamidino-2-phenylindole). Cells were observed by confocal laser scanning microscopy and quantified manually the acquired images with Image J software.

Western blot assays

Standard Western blot analysis was performed as previously described (21, 22). Antibodies against SQSTM1/p62 (D5E2), Bax (D2E11), Bcl-2, and Cyclin D1 were purchased from Cell Signaling Technology. Antibodies against p27, LC-3, and MAPKAPK2 were purchased from Proteintech. β-Actin and Phospho-MAPKAPK2 (Thr222) antibodies were purchased from ABClonal. Full scans of Western blot assays are shown in Supplementary Figs. S4 to S7.

Plasmids transfection

Expression vector of human MAPKAPK2 was designed and purchased from Genechem. We transfected with 0.8 mg of DNA construct using Lipofectamine 2000 (Invitrogen) according to the manufacturer's instructions.

Flow cytometry analysis

Colorectal cancer cells were treated with sop for 48 hours. The cells were collected with EDTA-free trypsin and washed with ice-cold PBS for 2 times. For cell apoptosis analysis, all cells were resuspended with binding buffer and incubated with Annexin V–FITC and propidium iodide (BD Biosciences) according to the manufacturer's instructions. In a limited time, the percentages of apoptotic cells were analyzed using flow cytometry (Attune NxT; Invitrogen). For cell-cycle analysis, harvested cells were fixed in 75% ethanol overnight at −20°C. On the following day, the cells were recovered by centrifugation and washed in cold PBS. Thereafter, they were incubated in 0.5 mL PI/RNase staining Solution (Invitrogen) in the dark at room temperature for 30 minutes. Cell cycle was determined and analyzed by flow cytometry.

Real-time PCR

Total RNA was extracted as previously described (23). RNA quantity and purity were determined by using a NanoDrop 2000 (Thermo Scientific). Total RNA was reverse transcribed with HiFiScript cDNA Synthesis Kit (Cowin Biotech). Then, real-time PCR was performed in triplicate with UltraSYBR mixture (Cowin Biotech) using 7500 RT-PCR System (Applied Biosystems, Life Technologies). The expression of genes was normalized to the Actin gene. The primers used are listed in the Supplementary Table S1.

Cellular thermal shift assays

Cellular thermal shift assays (CETSA) were performed to determine the direct binding between Sophoridine and MAPKAPK2 in cellular. Colorectal cancer cells were pretreated with DMSO or Sophoridine for 48 hours, chilled on ice, washed with PBS plus protease inhibitor cocktail, and then collected and heated for 3 minutes at appropriate temperature. Subsequently, cells were lysed and proteins were separated, and the corresponding index was determined by Western blot assays.

Drug affinity responsive targets stability assay

The drug affinity responsive targets stability (DARTS) assay was conducted as described above (24, 25). To prepare DARTS samples, 1 × 107 colorectal cancer cells were lysed in 2.4 mL M-PER buffer with protease inhibitors, centrifuged, collected proteins, and then added 10 × TNC buffer. Lysates were equally divided into two parts for 1 hour at room temperature with DMSO or Sophoridine, and incubated with 1 mg/mL pronase at room temperature for 5 minutes. The reaction was stopped by adding protease inhibitors, and samples were stored at −20°C standby.

Molecular docking

Docking simulations were operated using the DiscoveryStudio 2017 R2 molecular modeling software. The three-dimensional (3D) structures of the Sophoridine molecule were generated with ChemDraw and were energy minimized with CHARMm force field. The initial 3D geometric coordinates of MAPKAP kinase 2 (PDBcode: 2JBO) were obtained from the Protein Databank (PDB). Then, the protein structure was prepared by removing water molecules and adding hydrogen. CDOCKER protocols were employed as docking approaches and calculated the predicted binding energy (kcal mol−1). The complex structure with the most favorable binding-free energies was selected as the optimal docked conformation for later experimental verification.

Database of colorectal cancer patients

Clinical data can be obtained via the publically available The Cancer Genome Atlas and Gene Expression Omnibus (GEO) datasets. The expression level of MAPKAPK2 in colorectal cancer patients was analyzed by Kaplan–Meier estimate. GSEA was used to identify the association of MAPKAPK2 expression with biological processes of colorectal cancer cells by GSEA 3.0 software (http://www.broadinstitute.org/gsea/).

Statistical analysis

The data were represented as mean ± SD. Two-tailed unpaired Student t test was used for comparing two groups of data. One-ANOVA was used to compare multiple groups of data. Survival analysis was determined using the Kaplan–Meier estimates and the log-rank test. The variation (P < 0.05) was considered statistically significant.

Sophoridine suppresses growth and induces apoptosis in colorectal cancer cells

The photo of Radian Sophorae flavescentis is shown in Fig. 1A. The pharmacokinetic parameters of the main chemical constituents of Radian Sophorae flavescentis were screened by TCMSP database in Supplementary Table S2, and Sophoridine is one of the best antitumor active ingredients. As shown in Supplementary Fig. S1A and Supplementary Table S3, the pharmacokinetics properties were elucidated of Sophoridine in rat by high performance liquid chromatography. The result could provide meaningful reference for further clinical medication of Sophoridine. The chemical structure of Sophoridine is shown in Fig. 1B. As shown in Fig. 1C, clonogenicity of colorectal cancer cell lines (RKO, SW480, and HCT116) was dramatically reduced by Sophoridine for 48 hours. The CCK8 assay was used to detect the cytotoxic effects of Sophoridine against colorectal cancer cell lines and two nontumorigenic cell lines (HKC and LX-2). Consistently, as evidenced by decreased cell viability, Sophoridine strongly inhibited cell proliferation in colorectal cancer cells (Fig. 1D–F). However, Sophoridine did not affect the cell viability of HKC and LX-2 cells (Supplementary Fig. S1B and S1C). In vivo, the indexes had no statistical difference of routine blood test and serum biochemical measurements, compared with the control group, in Supplementary Tables S4 and S5, and no obvious abnormalities in histopathology after the Sophoridine administration in mice (Supplementary Fig. S1D). These results suggest that Sophoridine showed no obvious drug toxicity under conditions of potent antitumor efficacy. To investigate cell apoptosis regulation effect of Sophoridine on colorectal cancer cells, we performed Western blot assays. As shown in Fig. 1G, after exposure to Sophoridine, colorectal cancer cells showed a downregulation of Bcl-2 and an upregulation of Bax, which result in a dose-dependent increase of the ratio of Bax/Bcl-2. The apoptotic effects were further confirmed by employing Annexin V staining by treating colorectal cancer cells with Sophoridine (Fig. 1H). Taken together, these results suggest that Sophoridine suppresses growth and induces apoptosis of colorectal cancer cells.

Figure 1.

Sophoridine suppresses growth and induces apoptosis in CRC cells. A, The photo of Radian Sophorae flavescentis. B, The Chemical structure of Sophoridine. C, The clonogenicity of RKO, SW480, and HCT116 cells was determined after treatment with Sophoridine at indicated concentrations for 14 days. D–H, CRC cells were treated with indicated concentrations of Sophoridine for 48 hours. D–F, Cell viability was determined using CCK8 assay. G, The protein levels of Bcl-2 and Bax were detected by Western blot assays. H, The percentage of apoptotic cells was determined by flow cytometer. For C–G, data are shown as mean ± SD (n = 3); *, P < 0.05; **, P < 0.01; and ***, P < 0.001 compared with control (Student t test). All the Western data shown are representative of at least three independent experiments.

Figure 1.

Sophoridine suppresses growth and induces apoptosis in CRC cells. A, The photo of Radian Sophorae flavescentis. B, The Chemical structure of Sophoridine. C, The clonogenicity of RKO, SW480, and HCT116 cells was determined after treatment with Sophoridine at indicated concentrations for 14 days. D–H, CRC cells were treated with indicated concentrations of Sophoridine for 48 hours. D–F, Cell viability was determined using CCK8 assay. G, The protein levels of Bcl-2 and Bax were detected by Western blot assays. H, The percentage of apoptotic cells was determined by flow cytometer. For C–G, data are shown as mean ± SD (n = 3); *, P < 0.05; **, P < 0.01; and ***, P < 0.001 compared with control (Student t test). All the Western data shown are representative of at least three independent experiments.

Close modal

Sophoridine induces cell-cycle arrest and promotes autophagy in colorectal cancer cells

To investigate cell-cycle arrest promotion effect of Sophoridine on colorectal cancer cells, we performed Western blot assays. As indicated in Fig. 2A and B, Sophoridine significantly reduced the expression of Cyclin D1, while markedly increased the expression of p27 in a dose-dependent manner. The cell-cycle arrest effects were further confirmed by employing flow cytometry detection. As shown in Fig. 2C, Sophoridine significantly raised cell number at G0–G1 phase after 48-hour exposure, accompanied by decreased cell number at G2–M phase. Moreover, we also explored the protein levels of classical autophagy markers by Western blot assays and immunofluorescence staining. As shown in Fig. 2D–F, Sophoridine markedly reduced the expression of p62, whereas dramatically increased the expression of LC-3B puncta dose-dependently. Collectively, these results indicate that Sophoridine induces cell-cycle arrest and promotes autophagy of colorectal cancer cells.

Figure 2.

Sophoridine induces cell-cycle arrest and promotes autophagy in CRC cells. A and B, Western blotting analysis shows that the protein expression of Cyclin D1 and p27 was determined in SW480 and RKO cells treated with indicated doses of Sophoridine for 48 hours. C, Cells were treated with indicated concentrations of Sophoridine for 48 hours. Cell-cycle distribution was detected by flow cytometer. D and E, The protein levels of p62 and LC-3 were determined by Western blot assays. F, Cells were treated with DMSO or Sophoridine. The expression of LC-3 was determined by immunofluorescence staining. Right, relative level of LC-3.

Figure 2.

Sophoridine induces cell-cycle arrest and promotes autophagy in CRC cells. A and B, Western blotting analysis shows that the protein expression of Cyclin D1 and p27 was determined in SW480 and RKO cells treated with indicated doses of Sophoridine for 48 hours. C, Cells were treated with indicated concentrations of Sophoridine for 48 hours. Cell-cycle distribution was detected by flow cytometer. D and E, The protein levels of p62 and LC-3 were determined by Western blot assays. F, Cells were treated with DMSO or Sophoridine. The expression of LC-3 was determined by immunofluorescence staining. Right, relative level of LC-3.

Close modal

Potential target prediction and screening by network pharmacology

To investigate potential targets of Sophoridine in colorectal cancer cells, 116 pharmacophore candidates were predicted via pharmMapper (http://www.lilab-ecust.cn/pharmmapper/). The ranked list of hit target pharmacophore models is sorted by normalized fit score in descending order (Supplementary Table S6), and the top ten were displayed in Table 1. To improve the specificity, 3,298 colorectal cancer–associated genes were retrieved from the disGeNET (http://www.disgenet.org) database (Supplementary Table S7). A total of 67 potential targets of Sophoridine identified in colorectal cancer–associated genes were selected for constructing the drug-target (D-T) network (Fig. 3A). Thus, we examined changes in the transcriptional levels of these genes in colon cancer cells after treatment with Sophoridine. As shown in Fig. 3B, Sophoridine can significantly inhibit the expression of MAPK14, BRAF, FGFR1, and other genes. To make a deep exploring of the action mechanism of Sophoridine in colorectal cancer cells, we used String (https://string-db.org/) to obtain protein interactions and then constructed protein–protein interaction (PPI) network of genes associated with drug mediated by Cytoscape 3.2.1 (Fig. 3C), and the key topological parameter degree was analyzed. The biological functions of these potential targets were performed by Cytoscape plugin, ClueGO. These results suggest that these genes are involved in the development of various cancers and are closely related to MAPK VENTS, signaling through FGFR, VEGF pathways, etc. (Fig. 3D). Based on the above analysis, we predict that Sophoridine can inhibit the development of colorectal cancer cells by targeting MAPKAPK2.

Table 1.

Top ten pharmacophore candidates identified by PharmMapper

Pharma modelNorm fitSampleNameUniplot
2p3g_v 0.9707 MAPKAPK2 MAP kinase-activated protein kinase 2 P49137 
3gam_v 0.8849 NQO2 Ribosyldihydronicotinamide dehydrogenase (quinone) P16083 
1shj_v 0.8818 CASP7 Caspase-7 CASP7_HUMAN 
1e7a_v 0.8008 ALB Serum albumin ALBU_HUMAN 
2zas_v 0.7294 ESRRG Estrogen-related receptor gamma P62508 
2o65_v 0.702 PIM1 Proto-oncogene serine/threonine-protein kinase Pim-1 PIM1_HUMAN 
2ipw_v 0.6914 AKR1B1 Aldose reductase ALDR_HUMAN 
3fzk_v 0.6896 HSPA8 Heat shock cognate 71 kDa protein P11142 
1fdu_v 0.6888 HSD17B1 Estradiol 17-beta-dehydrogenase 1 P14061 
Pharma modelNorm fitSampleNameUniplot
2p3g_v 0.9707 MAPKAPK2 MAP kinase-activated protein kinase 2 P49137 
3gam_v 0.8849 NQO2 Ribosyldihydronicotinamide dehydrogenase (quinone) P16083 
1shj_v 0.8818 CASP7 Caspase-7 CASP7_HUMAN 
1e7a_v 0.8008 ALB Serum albumin ALBU_HUMAN 
2zas_v 0.7294 ESRRG Estrogen-related receptor gamma P62508 
2o65_v 0.702 PIM1 Proto-oncogene serine/threonine-protein kinase Pim-1 PIM1_HUMAN 
2ipw_v 0.6914 AKR1B1 Aldose reductase ALDR_HUMAN 
3fzk_v 0.6896 HSPA8 Heat shock cognate 71 kDa protein P11142 
1fdu_v 0.6888 HSD17B1 Estradiol 17-beta-dehydrogenase 1 P14061 
Figure 3.

Network pharmacology analysis of the candidate targets. A, D-T network is the intersection of drug targets and CRC-associated genes. B, The mRNA levels of the candidate targets were detected by real-time PCR. C, PPI network of potential target proteins (color distribution according to topological parameter degree). D, Relative drug-mediated genes was analyzed by ClueGO.

Figure 3.

Network pharmacology analysis of the candidate targets. A, D-T network is the intersection of drug targets and CRC-associated genes. B, The mRNA levels of the candidate targets were detected by real-time PCR. C, PPI network of potential target proteins (color distribution according to topological parameter degree). D, Relative drug-mediated genes was analyzed by ClueGO.

Close modal

Sophoridine promotes the apoptotic and autophagic capacities and induces cell-cycle arrest via MAPKAPK2 inactivation

To detect whether MAPKAPK2 is a direct target of Sophoridine, we employed the CETSAs. As shown in Fig. 4A–C, Sophoridine treatment significantly shifted the MAPKAPK2 melting curve compared with control. Moreover, our DARTS data suggested that Sophoridine binds to MAPKAPK2, protecting it from proteolytic cleavage (Fig. 4D). To search the promising target for binding mode of Sophoridine in MAPKAPK2, molecular docking simulation experiments were performed between Sophoridine and MAPKAPK2 by employing Discovery Studio 2017 R2 software. The CDOCKER docking result revealed that Sophoridine can bound into the ATP site of MAPKAP kinase 2 (PDBcode: 2JBO), and extend into the sub pockets for the adenine moiety and the α-phosphate, surrounded by key residues (LEU193, LEU70, ALA91, VAL78, LYS93), thus blocking the ATP-binding site fully (Fig. 4E–G). Furthermore, to further evaluate the MAPKAPK2 inhibitory effect, we detected the constitutive activation of MAPKAPK2 in colorectal cancer cells by the specific antibodies against phospho-MAPKAPK2 Thr222. As shown in Fig. 4H and Supplementary Fig. S2A, Sophoridine significantly reduced the phosphorylation level of MAPKAPK2 (Thr222) and MAPKAPK2 activity, but there was no big difference in its total expression in colorectal cancer cells. Interestingly, Sophoridine did not affect the p38 (upstream activators of MAPKAPK2) activity and significantly reduced the docking interaction of p38-MAPKAPK2 in Supplementary Fig. S2B and S2C.

Figure 4.

Sophoridine targeting inhibits phosphorylation of MAPKAPK2. A–C, Cells were treated with or without 160 μmol/L of Sophoridine for 48 hours and subsequently heated at different temperature for 3 minutes. After freeze-thaw cycles for cell lysis, the soluble MAPKAPK2 protein levels bound to a drug were visualized by Western blot assays. Right, relative band intensity of MAPKAPK2. D, Cells were incubated with 160 μmol/L of Sophoridine for or PBS for 1 hour at room temperature and digested with Pronase for 5 minutes at room temperature. MAPKAPK2 protein levels were tested by western blot assays. E–G, Docking model of Sophoridine with MAPKAPK2. E, The interaction pattern of Sophoridine with the residues. F, 2D diagram between the receptor and ligand. G, Sophoridine binding with the pocket is composed of hydrogen bonds. H, SW480 and RKO cells were treated with indicated concentration of Sophoridine for 48 hours. The protein levels of p-MAPKAPK2 were detected by Western blot assays. Total MAPKAPK2 expressions were detected as the internal control.

Figure 4.

Sophoridine targeting inhibits phosphorylation of MAPKAPK2. A–C, Cells were treated with or without 160 μmol/L of Sophoridine for 48 hours and subsequently heated at different temperature for 3 minutes. After freeze-thaw cycles for cell lysis, the soluble MAPKAPK2 protein levels bound to a drug were visualized by Western blot assays. Right, relative band intensity of MAPKAPK2. D, Cells were incubated with 160 μmol/L of Sophoridine for or PBS for 1 hour at room temperature and digested with Pronase for 5 minutes at room temperature. MAPKAPK2 protein levels were tested by western blot assays. E–G, Docking model of Sophoridine with MAPKAPK2. E, The interaction pattern of Sophoridine with the residues. F, 2D diagram between the receptor and ligand. G, Sophoridine binding with the pocket is composed of hydrogen bonds. H, SW480 and RKO cells were treated with indicated concentration of Sophoridine for 48 hours. The protein levels of p-MAPKAPK2 were detected by Western blot assays. Total MAPKAPK2 expressions were detected as the internal control.

Close modal

Next, we found that MAPKAPK2 overexpression in colorectal cancer cells strongly attenuated the inhibitory effect of Sophoridine on colony formation and cell viability (Fig. 5A–C). In addition, ectopic MAPKAPK2 expression dramatically recovered MAPKAPK2-regulated cell-cycle arrest and MAPKAPK2-induced apoptosis and autophagy (Fig. 5D–I). Moreover, we found that knockdown of endogenous MAPKAPK2 by siRNA has a similar effect with Sophoridine-regulated cell-cycle arrest, apoptosis, and autophagy. Furthermore, knockdown of endogenous MAPKAPK2 further enhanced the anticolorectal cancer effect of Sophoridine (Supplementary Fig. S3A–S3D). Taken together, these results indicate that Sophoridine promotes apoptosis and autophagy and induces cell-cycle arrest through targeting MAPKAPK2.

Figure 5.

Sophoridine promotes the apoptotic and autophagic capacities and induces cell-cycle arrest via MAPKAPK2 inactivation. A–I, Cells transfected with MAPKAPK2 (MAPKAPK2 Vec) or empty vector (Control Vec) followed by Sophoridine treatment. A, D, F, and H, The protein levels of MAPKAPK2, Bcl-2, Bax, Cyclin D1, p27, and LC-3 were detected by Western blot assays. B, The cell viability was measured by CCK8 assay. C, The colony formation capability was detected by clonogenic assay. E, The percentage of apoptotic cells was measured by flow cytometer. G, Cell-cycle distribution was detected by flow cytometer. I, The LC-3 was detected by IF analysis. Right, relative level of LC-3. For B, C, G, and I, data are shown as mean ± SD (n = 3); *, P < 0.01; **, P < 0.005; and ***, P < 0.001 compared with Vector control; #, P < 0.01 and ###, P < 0.001 compared with Vector control–transfected cells treated with Sophoridine (Student t test). All the Western blot data shown are representative of at least three independent experiments.

Figure 5.

Sophoridine promotes the apoptotic and autophagic capacities and induces cell-cycle arrest via MAPKAPK2 inactivation. A–I, Cells transfected with MAPKAPK2 (MAPKAPK2 Vec) or empty vector (Control Vec) followed by Sophoridine treatment. A, D, F, and H, The protein levels of MAPKAPK2, Bcl-2, Bax, Cyclin D1, p27, and LC-3 were detected by Western blot assays. B, The cell viability was measured by CCK8 assay. C, The colony formation capability was detected by clonogenic assay. E, The percentage of apoptotic cells was measured by flow cytometer. G, Cell-cycle distribution was detected by flow cytometer. I, The LC-3 was detected by IF analysis. Right, relative level of LC-3. For B, C, G, and I, data are shown as mean ± SD (n = 3); *, P < 0.01; **, P < 0.005; and ***, P < 0.001 compared with Vector control; #, P < 0.01 and ###, P < 0.001 compared with Vector control–transfected cells treated with Sophoridine (Student t test). All the Western blot data shown are representative of at least three independent experiments.

Close modal

Clinical significance of the MAPKAPK2 in colorectal cancer

To further investigate the clinical outcome of MAPKAPK2 in colorectal cancer patient, we subjected them to Kaplan–Meier analysis in GEO data set. Data revealed that higher MAPKAPK2 expression was associated with poorer percent disease-specific survival (DSS) (GSE17536, P = 0.020, Fig. 6A), disease-free survival (DFS) (GSE17536, P = 0.039, Fig. 6B), and overall survival (OS) (GSE17536, P = 0.025, Fig. 6C). Moreover, GEO database revealed that MAPKAPK2 level is high in colorectal cancer tissue compared with that in normal colon tissue (GSE110225, P = 0.043, Fig. 6D). Furthermore, according to the level of MAPKAPK2 from GSE17536, we estimated that higher level of MAPKAPK2 was positively correlated with enrichment of downregulation of apoptosis and autophagy by GSEA, as well as upregulation of proliferation and cell-cycle arrest (Fig. 6E–H). Taken together, these results indicate that MAPKAPK2 may be a prognosis marker in colorectal cancer patients.

Figure 6.

Clinical significance of the MAPKAPK2 in CRC. A–C, Kaplan–Meier plots of the PFS, DFS, and OS of CRC patients, stratified by expression of MAPKAPK2. Data obtained from the dataset of GEO (GSE17536). D, MAPKAPK2 expression level in CRC tissue compared with in normal colon tissue (GSE110225). E–H, Identification of gene sets enriched in phenotypes associated with MAPKAPK2 by GSEA using GSE17536 data.

Figure 6.

Clinical significance of the MAPKAPK2 in CRC. A–C, Kaplan–Meier plots of the PFS, DFS, and OS of CRC patients, stratified by expression of MAPKAPK2. Data obtained from the dataset of GEO (GSE17536). D, MAPKAPK2 expression level in CRC tissue compared with in normal colon tissue (GSE110225). E–H, Identification of gene sets enriched in phenotypes associated with MAPKAPK2 by GSEA using GSE17536 data.

Close modal

Collectively, our results show that the MAPKAPK2 plays an important role in Sophoridine-regulated apoptosis, autophagy, and cell-cycle arrest in colorectal cancer.

Sophoridine, an active quinolizidine alkaloid compound, has been proven to possess extensive physiologic activities (16, 17). Previous studies have revealed that Sophoridine displays prominent anticancer biological effects (20). However, the underlying molecular mechanisms are still elucidated. In this study, we have illustrated that Sophoridine promotes apoptosis and autophagy and induces cell-cycle arrest via targeting MAPKAPK2, leading to the blocking of the growth and development of colorectal cancers.

Network pharmacology is one of the strategies for discovering new drugs (26–28). In recent years, it has played an increasingly important role in the research and development of new drugs, such as target identification, mechanism of action, discovery and optimization of drug lead, and preclinical efficacy and safety evaluation (29–31). Network pharmacology is often studied by integrating multidisciplinary molecular networks, such as chemical informatics, bioinformatics, and systems biology (32, 33). Radix Sophorae Flavescentis is very commonly used in China, Japan, and some European countries for its various physiologic functions (17). Sophoridine is one of major bioactive components from Radix Sophorae Flavescentis. Previous studies have identified Sophoridine contains several biological properties (16, 20). In this study, we predicted 116 pharmacophore candidates via PharmMapper (http://lilab.ecust.edu.cn/pharmmapper) and found 67 potentially targets of Sophoridine from 3,298 colorectal cancer–associated genes, and then we constructed the D-T network through these targets. Moreover, we have found that Sophoridine can significantly inhibit the expression of MAPK14, BRAF, FGFR1, and other genes by real-time PCR. Furthermore, we used String (https://string-db.org/) to obtain protein interactions and then constructed PPI network of genes associated with drug mediated by Cytoscape 3.2.1. And we analyzed the key topological parameter degree and performed the biological functions of these potential targets by Cytoscape plugin, ClueGO. Taken together, the results suggest that these genes are involved in the development of various cancers and are closely related to MAPK EVENTS, signaling through FGFR, VEGF pathways, etc., and we predicted that MAPKAPK2 may target colorectal cancer cells involved in the treatment with Sophoridine.

MAPKAPK2, also called MK2, is known to be acted as a downstream signaling protein of p38MAPK and regulates a cascade of critical biological processes including inflammatory responses, nuclear export, stress, and DNA damage (34). Depending on these processes, MAPKAPK2 regulates transcript stability, expression of diverse proteins, and the phosphorylation involved in numerous important cellular phenomenon, such as cell cycle, senescence, cell migration, cell proliferation, and apoptosis (35–37). Systemic side effects are a major obstacle to the conversion of developed p38MAPK inhibitors into successful new drugs. This is the foremost cause of failure in the clinical trials. To solve this problem and effectively inhibit p38MAPK, researchers turned their focus to many of its downstream targets, such as MAPKAPK2. Previous studies have shown that targeting MAPKAPK2 to interdict its downstream events is as good as direct upstream inhibition of the p38MAPK pathway without obvious side effects of p38MAPK inhibitors (38–40). In the present study, we have demonstrated a direct binding between MAPKAPK2 and Sophoridine by DARTS assay and CETSA. Furthermore, we have found that Sophoridine selectively inactivates phospho-MAPKAPK2 (Thr222) and directly binds into the ATP site of MAPKAPK2 by molecular docking. The blockage effects are further determined by Western blot: Sophoridine markedly reduces phospho-MAPKAPK2 (Thr222) in a dose-dependent manner, but there is no obvious deference in its total expression in colorectal cancer cells.

Past reports have demonstrated the expression of MAPKAPK2 in a multitude of cell types like cancers, smooth muscle cells, and endothelial cells (41–44). A recent study has elucidated that MAPKAPK2 plays a key role in colon cancer processes via axis inhibition of Hsp27, which finally results in promoting cell angiogenesis, migration, survival, and proliferation (35). Literature reports have shown that deletion of MAPKAPK2 conduces to DNA damage and apoptosis through impaired phosphorylation of MDM2 and subsequently enhances the primary regulator of p53 stability in skin cancer (45). Past studies have reported that MAPKAPK2 promotes the invasion and metastasis via regulation of MMP-2/9 mRNA half-life in bladder cancer (46). In the present study, we have found that ectopic MAPKAPK2 expression significantly blocks Sophoridine-regulated apoptosis, autophagy, and cell-cycle arrest of colorectal cancer. Clinical studies have shown that MAPKAPK2 expression is associated with poor prognosis. Furthermore, we estimated that a higher level of MAPKAPK2 was positively associated with the enrichment of downregulation of apoptosis and autophagy by GSEA, as well as upregulation of proliferation and cell-cycle arrest. Collectively, our results demonstrate that the MAPKAPK2 plays a major role in Sophoridine-regulated tumorigenesis in colorectal cancers.

In summary, we have screened the pharmacokinetic parameters of the main chemical constituents of Radian Sophorae flavescentis by TCMSP database and have found that Sophoridine is one of the best antitumor active ingredients. Our study elucidates that Sophoridine induces apoptosis, autophagy, and cell-cycle arrest through targeting MAPKAPK2, which leads to inhibiting the tumor development and progression of colorectal cancers. These studies show that Sophoridine may be a promising therapeutic strategy that blocks tumorigenesis in colorectal cancers.

No potential conflicts of interest were disclosed.

Conception and design: H. Yu

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): R. Wang, H. Liu, Y. Shao, K. Wang

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Yin, Y. Qiu, H. Wu, E. Liu, T. Wang, X. Gao, H. Yu

Writing, review, and/or revision of the manuscript: H. Yu

Study supervision: H. Yu

This work was supported by grants from National Natural Science Foundation of China (81603253, 21711540293, and 81873089 to H. Yu, and 81602614 and 81973570 to Y. Qiu), Important Drug Development Fund, Ministry of Science and Technology of China (2018ZX09735-002 to T. Wang), and Natural Science Foundation of Tianjin City (No. 15PTCYSY00030 to Z. Li).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Bray
F
,
Ferlay
J
,
Soerjomataram
I
,
Siegel
RL
,
Torre
LA
,
Jemal
A
. 
Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries
.
CA Cancer J Clin
2018
;
68
:
394
424
.
2.
Ferlay
J
,
Colombet
M
,
Soerjomataram
I
,
Mathers
C
,
Parkin
DM
,
Piñeros
M
, et al
Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods
.
Int J Cancer
2019
;
144
:
1941
53
.
3.
Arnold
M
,
Sierra
MS
,
Laversanne
M
,
Soerjomataram
I
,
Jemal
A
,
Bray
F
. 
Global patterns and trends in colorectal cancer incidence and mortality
.
Gut
2017
;
66
:
683
91
.
4.
Van Cutsem
E
,
Cervantes
A
,
Adam
R
,
Sobrero
A
,
Van Krieken
JH
,
Aderka
D
, et al
ESMO consensus guidelines for the management of patients with metastatic colorectal cancer
.
Ann Oncol
2016
;
27
:
1386
422
.
5.
Goldstein
DA
,
Ahmad
BB
,
Chen
Q
,
Ayer
T
,
Howard
DH
,
Lipscomb
J
, et al
Cost-effectiveness analysis of regorafenib for metastatic colorectal cancer
.
J Clin Oncol
2015
;
33
:
3727
32
.
6.
Cassidy
S
,
Syed
BA
. 
Colorectal cancer drugs market
.
Nat Rev Drug Discov
2015
;
16
:
525
6
.
7.
Clarke
JM
,
Hurwitz
HI
,
Rangwala
F
. 
Understanding the mechanisms of action of antiangiogenic agents in metastatic colorectal cancer: a clinician's perspective
.
Cancer Treat Rev
2014
;
40
:
1065
72
.
8.
Gharwan
H
,
Groninger
H
. 
Kinase inhibitors and monoclonal antibodies in oncology: clinical implications
.
Nat Rev Clin Oncol
2016
;
13
:
209
27
.
9.
Rodrigues
T
,
Reker
D
,
Schneider
P
,
Schneider
G
. 
Counting on natural products for drug design
.
Nat Chem
2016
;
8
:
531
41
.
10.
David
B
,
Wolfender
JL
,
Dias
DA
. 
The pharmaceutical industry and natural products: historical status and new trends
.
Phytochem Rev
2015
;
14
:
299
315
.
11.
Harvey
AL
,
Edrada-Ebel
RA
,
Quinn
RJ
. 
The re-emergence of natural products for drug discovery in the genomics era
.
Nat Rev Drug Discov
2015
;
14
:
111
29
.
12.
Wright
GD
. 
Opportunities for natural products in 21st century antibiotic discovery
.
Nat Prod Rep
2017
;
34
:
694
701
.
13.
Atanasov
AG
,
Waltenberger
B
,
Pferschy-Wenzig
EM
,
Linder
T
,
Wawrosch
C
,
Uhrin
P
, et al
Discovery and resupply of pharmacologically active plant-derived natural products: a review
.
Biotechnol Adv
2015
;
33
:
1582
614
.
14.
Wang
W
,
You
RL
,
Qin
WJ
,
Hai
LN
,
Fang
MJ
,
Huang
GH
, et al
Anti-tumor activities of active ingredients in compound Kushen injection
.
Acta Pharmacol Sin
2015
;
36
:
676
9
.
15.
He
X
,
Fang
J
,
Huang
L
,
Wang
J
,
Huang
X
. 
Sophora flavescens Ait.: traditional usage, phytochemistry and pharmacology of an important traditional Chinese medicine
.
J Ethnopharmacol
2015
;
172
:
10
29
.
16.
Cai
XH
,
Guo
H
,
Xie
B
. 
Structural modifications of matrine-type alkaloids
.
Mini Rev Med Chem
2018
;
18
:
730
44
.
17.
Sun
M
,
Cao
H
,
Sun
L
,
Dong
S
,
Bian
Y
,
Han
J
, et al
Antitumor activities of kushen: literature review
.
Evid Based Complement Alternat Med
2012
;
2012
:
373219
.
18.
Huang
J
,
Matrine
Xu H
. 
Bioactivities and structural modifications
.
Curr Top Med Chem
2016
;
16
:
3365
78
.
19.
Ni
W
,
Li
C
,
Liu
Y
,
Song
H
,
Wang
L
,
Song
H
, et al
Various bioactivity and relationship of structure-activity of matrine analogues
.
J Agric Food Chem
2017
;
65
:
2039
47
.
20.
Rashid
HU
,
Xu
Y
,
Muhammad
Y
,
Wang
L
,
Jiang
J
. 
Research advances on anticancer activities of matrine and its derivatives: an updated overview
.
Eur J Med Chem
2019
;
161
:
205
38
.
21.
Yu
H
,
Yin
S
,
Zhou
S
,
Shao
Y
,
Sun
J
,
Pang
X
, et al
Magnolin promotes autophagy and cell cycle arrest via blocking LIF/Stat3/Mcl-1 axis in human colorectal cancers
.
Cell Death Dis
2018
;
9
:
702
.
22.
Yu
H
,
Qiu
Y
,
Pang
X
,
Li
J
,
Wu
S
,
Yin
S
, et al
Lycorine promotes autophagy and apoptosis via TCRP1/Akt/mTOR axis inactivation in human hepatocellular carcinoma
.
Mol Cancer Ther
2017
;
16
:
2711
23
.
23.
Yu
H
,
Yue
X
,
Zhao
Y
,
Li
X
,
Wu
L
,
Zhang
C
, et al
LIF negatively regulates tumour-suppressor p53 through Stat3/ID1/MDM2 in colorectal cancers
.
Nat Commun
2014
;
5
:
5218
.
24.
Wu
S
,
Qiu
Y
,
Shao
Y
,
Yin
S
,
Wang
R
,
Pang
X
, et al
Lycorine displays potent antitumor efficacy in colon carcinoma by targeting STAT3
.
Front Pharmacol
2018
;
9
:
881
.
25.
Yin
S
,
Qiu
Y
,
Jin
C
,
Wang
R
,
Wu
S
,
Liu
H
, et al
7-deoxynarciclasine shows promising antitumor efficacy by targeting Akt against hepatocellular carcinoma
.
Int J Cancer
2019
;
145
:
3334
46
.
26.
Kibble
M
,
Saarinen
N
,
Tang
J
,
Wennerberg
K
,
Mäkelä
S
,
Aittokallio
T
. 
Network pharmacology applications to map the unexplored target space and therapeutic potential of natural products
.
Nat Prod Rep
2015
;
32
:
1249
66
.
27.
Hopkins
AL
. 
Network pharmacology: the next paradigm in drug discovery
.
Nat Chem Biol
2008
;
4
:
682
90
.
28.
Fotis
C
,
Antoranz
A
,
Hatziavramidis
D
,
Sakellaropoulos
T
,
Alexopoulos
LG
. 
Network-based technologies for early drug discovery
.
Drug Discov Today
2018
;
23
:
626
35
.
29.
Zhao
S
,
Iyengar
R
. 
Systems pharmacology: network analysis to identify multiscale mechanisms of drug action
.
Annu Rev Pharmacol Toxicol
2012
;
52
:
505
21
.
30.
Hsin
KY
,
Matsuoka
Y
,
Asai
Y
,
Kamiyoshi
K
,
Watanabe
T
,
Kawaoka
Y
, et al
SystemsDock: a web server for network pharmacology-based prediction and analysis
.
Nucleic Acids Res
2016
;
44
:
W507
13
.
31.
Moffat
JG
,
Vincent
F
,
Lee
JA
,
Eder
J
,
Prunotto
M
. 
Opportunities and challenges in phenotypic drug discovery: an industry perspective
.
Nat Rev Drug Discov
2017
;
16
:
531
43
.
32.
Gartner
ZJ
,
Prescher
JA
,
Lavis
LD
. 
Unraveling cell-to-cell signaling networks with chemical biology
.
Nat Chem Biol
2017
;
13
:
564
8
.
33.
Korcsmaros
T
,
Schneider
MV
,
Superti-Furga
G
. 
Next generation of network medicine: interdisciplinary signaling approaches
.
Integr Biol (Camb)
2017
;
9
:
97
108
.
34.
Soni
S
,
Anand
P
,
Padwad
YS
. 
MAPKAPK2: the master regulator of RNA-binding proteins modulates transcript stability and tumor progression
.
J Exp Clin Cancer Res
2019
;
38
:
121
.
35.
Henriques
A
,
Koliaraki
V
,
Kollias
G
. 
Mesenchymal MAPKAPK2/HSP27 drives intestinal carcinogenesis
.
Proc Natl Acad Sci U S A
2018
;
115
:
E5546
55
.
36.
Kumar
B
,
Koul
S
,
Petersen
J
,
Khandrika
L
,
Hwa
JS
,
Meacham
RB
, et al
p38 mitogen-activated protein kinase-driven MAPKAPK2 regulates invasion of bladder cancer by modulation of MMP-2 and MMP-9 activity
.
Cancer Res
2010
;
70
:
832
41
.
37.
Herranz
N
,
Gallage
S
,
Mellone
M
,
Wuestefeld
T
,
Klotz
S
,
Hanley
CJ
, et al
mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype
.
Nat Cell Biol
2015
;
17
:
1205
17
.
38.
Murali
B
,
Ren
Q
,
Luo
X
,
Faget
DV
,
Wang
C
,
Johnson
RM
, et al
Inhibition of the stromal p38MAPK/MK2 pathway limits breast cancer metastases and chemotherapy-induced bone loss
.
Cancer Res
2018
;
78
:
5618
30
.
39.
Soni
S
,
Saroch
MK
,
Chander
B
,
Tirpude
NV
,
Padwad
YS
. 
MAPKAPK2 plays a crucial role in the progression of head and neck squamous cell carcinoma by regulating transcript stability
.
J Exp Clin Cancer Res
2019
;
38
:
175
.
40.
Li
Y
,
Köpper
F
,
Dobbelstein
M
. 
Inhibition of MAPKAPK2/MK2 facilitates DNA replication upon cancer cell treatment with gemcitabine but not cisplatin
.
Cancer Lett
2018
;
428
:
45
54
.
41.
Taniyama
Y
,
Ushio-Fukai
M
,
Hitomi
H
,
Rocic
P
,
Kingsley
MJ
,
Pfahnl
C
, et al
Role of p38 MAPK and MAPKAPK-2 in angiotensin II-induced Akt activation in vascular smooth muscle cells
.
Am J Physiol Cell Physiol
2004
;
287
:
C494
9
.
42.
Brophy
CM
,
Woodrum
D
,
Dickinson
M
,
Beall
A
. 
Thrombin activates MAPKAP2 kinase in vascular smooth muscle
.
J Vasc Surg
1998
;
27
:
963
9
.
43.
Chang
E
,
Heo
KS
,
Woo
CH
,
Lee
H
,
Le
NT
,
Thomas
TN
, et al
MK2 SUMOylation regulates actin filament remodeling and subsequent migration in endothelial cells by inhibiting MK2 kinase and HSP27 phosphorylation
.
Blood
2011
;
117
:
2527
37
.
44.
Kayyali
US
,
Pennella
CM
,
Trujillo
C
,
Villa
O
,
Gaestel
M
,
Hassoun
PM
. 
Cytoskeletal changes in hypoxic pulmonary endothelial cells are dependent on MAPK-activated protein kinase MK2
.
J Biol Chem
2002
;
277
:
42596
602
.
45.
Johansen
C
,
Vestergaard
C
,
Kragballe
K
,
Kollias
G
,
Gaestel
M
,
Iversen
L
. 
MK2 regulates the early stages of skin tumor promotion
.
Carcinogenesis
2009
;
30
:
2100
8
.
46.
Kumar
B
,
Koul
S
,
Petersen
J
,
Khandrika
L
,
Hwa
JS
,
Meacham
RB
, et al
p38 mitogen-activated protein kinase-driven MAPKAPK2 regulates invasion of bladder cancer by modulation of MMP-2 and MMP-9 activity
.
Cancer Res
2010
;
70
:
832
41
.