Open Access

Oxymatrine‑mediated maturation of dendritic cells leads to activation of FOXP3+/CD4+ Treg cells and reversal of cisplatin‑resistance in lung cancer cells

  • Authors:
    • Hui Liu
    • Manman Zou
    • Pei Li
    • Haifeng Wang
    • Xijun Lin
    • Jin Ye
  • View Affiliations

  • Published online on: March 20, 2019     https://doi.org/10.3892/mmr.2019.10064
  • Pages: 4081-4090
  • Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The dendritic cell (DC)‑regulatory T (Treg) system serves a leading role in the immunosuppression of the tumor microenvironment, which is not conducive to radiotherapy and chemotherapy treatment for lung cancer. The present study aimed to investigate the effect of oxymatrine (OMT) on the DC‑Treg system in the tumor microenvironment in vitro and to examine its mechanism. The expressions of CD83 antigen, T‑lymphocyte activation antigen CD86, CD11 antigen‑like family member C and major histocompatibility complex II in DCs were increased upon treatment with 1 mg/ml OMT, as detected by flow cytometry. Following pretreatment with OMT, the DCs mediated the forkhead box protein P3 overexpression in primitive cluster of differentiation 4+ T cells at the protein and mRNA expression levels. The expression levels of anti‑inflammatory factors, including interleukin (IL)‑10, tumor growth factor‑β, IL‑35, and pro‑inflammatory cytokines, including interferon‑γ, IL‑12 and IL‑2, in the co‑culture supernatant were increased as measured by ELISA. When DCs and DC‑Tregs were co‑cultured with cisplatin‑resistant A549 cells, the proportion of apoptosis in the co‑culture groups was increased under treatment with cisplatin, which was detected by Annexin V/propidium Iodide staining and western blotting. The present results suggested that OMT may promote the maturation of DCs, mediate the differentiation of T cells into Treg cells, and reverse the resistance of tumor cells to cisplatin in vitro. It was suggested that OMT is an important adjunct to chemotherapy through the regulation of antitumor responses.

Introduction

In China, herbs and their products are widely used in clinical treatment, with increasing popularity. Matrine (MT) and oxymatrine (OMT), two types of alkaloid components in the roots of Sophora species, have various pharmacological activities and anti-inflammatory, anti-allergic, anti-virus, anti-fibrotic and cardiovascular protective effects (1). At present, OMT has been widely used in the treatment of hepatitis B and liver fibrosis in China (2). Furthermore, OMT may exert its anticancer activities through various channels, primarily by inhibiting cancer cell proliferation (3), inducing cell cycle arrest (4) and differentiation (5), accelerating apoptosis (6), restraining agiogenesis (7), inhibiting metastasis and invasion (8), and preventing or reducing chemotherapy- and radiotherapy-induced toxicities (9). However, these previous studies are mostly limited to observations of superficial phenomenon and lack systematic investigation using modern molecular biology techniques. The precise mechanism underlying the anticancer activity of OMT remains largely unknown.

Dendritic cells (DCs) serve a critical role in antigen capturing, processing and presentation (10). In the event of infection or inflammation of the body, microbial infection and other factors may promote the maturation of DCs, and thus initiate a T cell-mediated immune response (11,12). There are a range of effector T cells, including immunogenic cluster of differentiation (CD)4+ T helper (Th) cells, cytotoxic CD8+ T cells and in particular, tolerogenic regulatory T cells (Tregs), termed the DC-Treg system. In principle, DCs are associated with the two principal types of immunity, innate and adaptive. Therefore, DCs may be an ideal target for the development of immunotherapies and an adjuvant to convert their function between tolerogenic and immunogenic may be desirable. It is important to identify and develop strategies that may improve the efficacy of DC-mediated antitumor immunotherapy.

The immune status of the systemic or local microenvironment in tumor hosts may determine the responsiveness to chemotherapy (13). The immunomodulatory activity of OMT has been demonstrated in rheumatoid arthritis (13), chronic hepatitis B (14) and colitis models by shifting the Th subsets (15). However, to the best of the authors' knowledge, the potential effect of OMT on the DC-mediated antitumor immune response has not yet been studied. In the present study, the effects of OMT on DC maturation, and the subsequent simulation of CD4+ T cell polarization and cytokine secretion in vitro were examined. Furthermore, whether the immunomodulatory ability of OMT may reverse drug-resistance in A549 lung cancer cells was investigated.

Materials and methods

Subjects

Male NSCLC patients and healthy controls between the age of 40–55 were enrolled in this study. The median age was 46.8 years (range: 40–54) in NSCLC patients (n=13), and 45.3 years (range: 41–52) in healthy controls (n=15). Inclusion criteria for the present study were patients with histological proven NSCLC staging II–IV, who were primarily diagnosed in The Third Affiliated Hospital of Sun Yat-sen University between January 2017 and December 2017. All enrolled patients had no previous treatment with molecular target therapy, chemotherapy or radiotherapy. Exclusion criteria were chronic systemic diseases (including hypertension, diabetes and coronary heart disease) or immune systemic diseases (including HIV, organ transplantation and tumors). All subjects joined voluntarily with informed consents. This present study received ethical approval from the Institutional Review Board of Sun Yat-sen University.

Isolation and culture of DCs

Blood samples (50 ml) were obtained from each subjects. Peripheral blood mononuclear cells (PBMCs) were isolated by Ficoll (GE Healthcare Life Sciences, Little Chalfont, UK) density gradient centrifugation at 1,200 × g and 4°C for 20 min. PBMCs were cultured for 2 h and non-adherent cells were removed. The DC cell culture medium was prepared by adding recombinant human (rh) granulocyte-macrophage colony-stimulating factor (GM-CSF; 100 ng/ml) and rh interleukin (IL)-4 (100 ng/ml) to RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 10% fetal bovine serum (FBS; Hyclone; GE Healthcare Life Sciences, Logan, UT, USA). PBMCs were incubated for 8 days at 37°C and prepared for subsequent experiments.

Detection of DC surface markers

Cells were treated with lipopolysaccharide (LPS; 100 ng/ml), OMT (1 mg/ml) and OMT (1 mg/ml) + LPS (100 ng/ml) for 48 h. According to preliminary experiments in the present study, oxymatrine inhibited the proliferation of dendritic cells, and the effect of inhibiting proliferation was observed at OMT concentration of 0.8 mg/ml. However, the inhibitory effect was not significantly increased with the increase of concentration, so the concentration of OMT in this experiment was chosen to be 1.0 mg/ml (data not shown). The cultured DCs were collected and centrifuged at 1,000 × g for 5 min at 4°C. The supernatant was discarded and the cells were washed three times with PBS. In total, 5 µl CD83 antigen (CD83)-phycoerythrin (PE; 556855; BD Biosciences, San Jose, CA, USA), T-lymphocyte activation antigen CD86 (CD86)-PE (560957; BD Biosciences), CD11 antigen-like family member C (CD11c)-PE (555392; BD Biosciences) and major histocompatibility complex II (MHC II)-PE(555812, BD Biosciences) were added to each tube. Subsequent to gentle mixing, the cells were incubated at room temperature in the dark for 30 min. The cells were washed twice with PBS and resuspended in 100 µl PBS, and the DC surface markers were detected using a flow cytometer (BD Accuri C6; BD Biosciences).

Isolation and culture of CD4+ T cells from patients with non-small cell lung cancer (NSCLC)

A total of 10 patients with newly diagnosed NSCLC were enrolled to collect 20 ml blood prior to any treatment. Subsequently, PBMCs were isolated using the Ficoll density gradient method at 1,200 × g, 4°C for 20 min. In total, 1×106 cells were used to isolate CD4+ T cells using a magnetic cell separation system. The selected cells were cultured in RPMI-1640 medium containing 10% FBS for subsequent experiments.

Co-culture of DCs and T cells

The DCs were treated with control, LPS (100 ng/ml), OMT (1 mg/ml) and OMT (1 mg/ml)+LPS (100 ng/ml) for 48 h. Cells were subsequently centrifuged at 1,000 × g for 5 min at 4°C and the supernatant was discarded. Subsequent to washing three times with PBS, the DCs and peripheral blood-derived CD4+ T cells from patients with NSCLC were implanted into Matrigel-coated Transwell chambers; DCs were seeded into the lower chambers and CD4+ T cells into the upper chambers at 1:5, 1:10 and 1:20 ratios. Cells were cultured in RPMI-1640 medium containing 10% FBS for 48 h; a non-contact cell co-culture system was established.

qPCR for forkhead box protein P3

DC cells and T cells were co-cultured at a ratio of 1:5, 1:10 and 1:20, respectively, as described above. CD4+ T cells were collected and total RNA was extracted using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocols. cDNA were synthesized using PrimeScript™ RT reagent kit with gDNA Eraser (Takara Bio, Inc., Otsu, Japan), strictly following the manufacturer's protocols. The target genes and controls were analyzed by RT-qPCR using SYBR-Green PCR Master Mix (Toyobo Life Science, Osaka, Japan) and the reactions were performed on ABI 7500 system (Applied Biosystems; Thermo Fisher Scientific, Inc.). Primers used in the study were as follows, U6 (housekeeping) primers: 5′-CGCTTCACGAATTTGCGTGTCAT-3′ (forward) and 5′-GCTTCGGCAGCACATATACTAAAAT-3′ (reverse); FOXP3 primers: 5′-CTGACCAAGGCTTCATCTGTG-3′ (forward) and 5′-ACTCTGGGAATGTGCTGTTTC−3′ (reverse). Reagents in the reaction included 0.5 µl of cDNA (1:20), 0.5 µl of forward primer and 0.5 µl of reverse primer, 10 µl of PCR Master Mix and 4.0 µl dH2O. Thermocycling conditions for PCR were 95°C for 5 min, and 40 cycles of 95°C for 15 sec, 65°C for 15 sec and 72°C for 32 sec. The average value was calculated by the relative quantitative method, and the gene expressions in the treatment group relative to the control group were calculated by the formula 2−ΔΔCq following Livak's method (16).

Detection of forkhead box protein P3 (FOXP3)+/CD4+ Tregs

The co-cultured CD4+ T cells were collected and centrifuged at 1,000 × g for 5 min at 4°C; the supernatant was discarded. Anti-Human FoxP3 Staining kit with CD4-Fluorescein Isothiocyanate and FOXP3-PE (560132; BD Biosciences) was used in the following experiments. The cells were washed three times with PBS, and CD4-Fluorescein Isothiocyanate (1:20) was added to each tube and incubated for 30 min at room temperature in the dark. For antibodies to enter the intracellular structure, cells were resuspended in fixation/permeabilization buffer (00–5223/00-5123; eBioscience; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocols. Following two washes with PBS, the cells were incubated for 30 min at room temperature in the dark. FOXP3-PE (1:20) was added and incubated for another 30 min in the same condition. Cells were subsequently washed twice and resuspended with the fixation/permeabilization buffer. The ratio of FOXP3+/CD4+ cells was detected using a flow cytometer (BD Accuri C6; BD Biosciences).

Detection of cytokines in supernatant of the co-culture system

The DCs were treated with control, LPS (100 ng/ml), OMT (1 mg/ml) and OMT (1 mg/ml) + LPS (100 ng/ml), and mixed with peripheral blood-derived CD4+ T cells from patients with NSCLC at ratios of 1:5, 1:10 and 1:20. After 48 h, the supernatant of the co-culture system was collected. The concentrations of IL-10, transforming growth factor (TGF)-β, IL-35, interferon (IFN)-γ, IL-2 and IL-12 in the supernatant were detected using ELISA kits: IL-10 (E-EL-H0103c), TGF-β (E-EL-0110c), IL-35 (E-EL-H2443c), IFN-γ (E-EL-0108c), IL-2 (E-EL-H0099c) and IL-12 (E-EL-H0150c; all from R&D Systems, Inc., Minneapolis, MN, USA).

Detection of A549 apoptosis following co-culture

The induction of A549 resistant cells was performed by gradually increasing the concentration of cisplatin from a starting concentration of 1.0 µM, and after 4 weeks, it was increased to 2.0 µM, and serially subcultured for ~100 passages. Then, 6 months later, the A549/DDP resistant cell line was established. The human lung adenocarcinoma cells A549 were purchased from Procell Life Science & Technology Co., Ltd. (Wuhan, China; cat. no. CL-0016). The cells were maintained in RPMI-1640 (Gibco; Thermo Fisher Scientific, Inc.) medium with 10% fetal bovine serum (FBS; Hyclone; GE Healthcare Life Sciences) and 1% penicillin-streptomycin solution (Beyotime Institute of Biotechnology, Haimen, China), and cells were cultured in a humidified atmosphere at 37°C with 5% CO2. Co-culture systems of A549/DDP plus DC or DC-T were established. OMT (1 mg/ml)-treated DCs and the FOXP3+/CD4+ T cells induced by DC-T co-culture (cell ratio, 1:5) were incubated with A549/DDP cells. The cells were seeded for 24 h in the upper and lower chambers of Transwell plates at ratios of 1:1, 1:5, 1:10 and 1:20. At the same time, 22 µM DDP was added to the A549/DDP medium for 24 h. The collected cells were stained using an Annexin V/Propidium Iodide kit (BD Biosciences) according to the manufacturer's protocol, and the apoptotic ratio was analyzed using a flow cytometer (BD Accuri C6; BD Biosciences).

Western blotting

A549/DDP cells were lysed using radioimmunoprecipitation assay buffer (50 mM Tris, 150 mM NaCl, 1% NPNP-40, 1% sodium deoxycholate and 0.05% SDS; pH 7.4) following co-culture with OMT-treated DCs/DC-T and treatment with DDP. Total protein was extracted and subsequently quantified using a bicinchoninic acid kit (Bio-Rad Laboratories, Inc., Hercules, CA, USA). For western blotting, protein loading was set at 40 µg and an 12% SDS-PAGE gel was used for protein separation. The proteins were transferred to a nitrocellulose membrane and incubated with primary antibodies against FOXP3 (1:3,000; cat. no. ab191416), apoptosis regulator BAX, (Bax; 1:1,000; cat. no. ab32503), B cell lymphoma-2 (Bcl-2; 1:1,000; cat. no. ab32124) and β-actin (1:1,000; cat. no. ab8224) overnight at 4°C, and subsequently incubated with Goat Anti-Rabbit (1:1,000; cat. no. ab6702) or Anti-Mouse (1:1,000; cat. no. ab6708) (all from Abcam, Cambridge, UK) secondary antibodies for 1 h at room temperature. Chemiluminescence was used to develop the color of the membrane using an ECL kit according to the manufacturer's instructions (cat. no. FD8030; Guangzhou Fude Biological Technology Co., Ltd., Guangzhou, China). ImageJ 8.0 software (National Institutes of Health, Bethesda, MD, USA) was used to perform densitometric analysis.

Statistical analysis

Each experiment was repeated at least 3 times. Data were presented as the mean ± standard deviation. Statistical significance was determined by one-way analysis of variance with Bonferroni Correction using SPSS software version 19.0 (IBM Corp., Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

OMT promotes DC maturation

The monocytes were prepared from the peripheral blood of healthy adult volunteers. To investigate the effect of OMT, the immature DCs were cultured for 8 days containing 1 mg/ml OMT in medium supplemented with rhGM-CSF (100 ng/ml) and rhIL-4 (100 ng/ml). Bacterial LPS is the most common ‘maturation’ signal for DCs (17). With the LPS-treated (100 ng/ml) group (LPS-DCs) as a positive control and mononuclear cells as a negative control, the expression of DC surface markers was detected by flow cytometry analysis, as presented in Fig. 1. Compared with the negative control group, treatment with OMT (1 mg/ml) significantly increased the expression of CD83, CD86, CD11c and MHC II (P<0.001), and the expression level of these markers was higher when LPS and OMT were combined. These results demonstrated that OMT may promote DC maturation.

Differentiation of FOXP3+/CD4+ Tregs induced by OMT-DCs

To examine the response of allogeneic primary CD4+ T cells to DC stimulation pretreated with OMT (1 mg/ml; OMT-DC), the following experiments were performed. CD4+ T cells were isolated from the peripheral blood of patients with NSCLC. DCs were co-cultured with the primary CD4+ T cells for 48 h in Transwell plates at ratios of 1:5, 1:10 and 1:20. The expression levels of FOXP3 in CD4+ T cells were analyzed by flow cytometry, western blotting and qPCR.

As presented in Fig. 2, OMT-DCs stimulated the primary CD4+ T cells to express FOXP3, and the mRNA expression level of FOXP3 decreased as the DCs were more diluted. The FOXP3 protein assay showed that the total amount of FOXP3 was increased when LPS and OMT were used separately or in combination (especially when used in combination). However, in the case of the same treatment, the dilution of DC cells had no apparent effect on FOXP3 protein amount when groups of different DC cell dilution ratios were compared among each other. Whether there are translation level and turnover level factors regulating the expression of FOXP3 protein in this case deserves further investigation. Altogether, these results demonstrated that OMT-pretreated DCs may enhance FOXP3+/CD4+ Treg differentiation.

Cytokine secretion stimulated by OMT-pretreated DCs

Inflammatory factors serve an important role in the immune response. ELISA was used to quantitatively analyze inflammatory factors in the co-culture supernatants. When OMT-DCs were co-cultured with T cells, a large number of anti-inflammatory factors, including IL-10, TGF-β and IL-35, and pro-inflammatory cytokines, including IFN-γ, IL-12 and IL-2, were secreted. Furthermore, the expression levels of those inflammatory factors decreased as the DCs were more diluted with T cells (Fig. 3). When LPS-DCs were co-cultured with T cells, it additionally promoted the secretion of inflammatory factors, in a similar manner to OMT-DCs. When LPS and OMT were applied simultaneously, the secretion of inflammatory factors reached the highest concentration. These results suggested that OMT-pretreated DCs may promote the primary T cells to polarize into FOXP3+/CD4+ Tregs, and enhance the anti-inflammatory and pro-inflammatory cytokines secretion. Based on these results, the co-culture ratio of DCs to CD4+ T cells was set at 1:5 for subsequent experiments.

Reversal of DDP-resistance in A549/DDP by OMT-DC/OMT-DC-T

A DDP-resistant cell line, A549/DDP, was established. As presented in Fig. 4A, A549/DDP has a half maximal inhibitory concentration of 22 µM for DDP. In order to investigate the effect of OMT-pretreated DCs and OMT-DCs induced CD4+ T cells on the sensitivity of A549/DDP to antitumor drugs, OMT-stimulated DCs (OMT-DC) and OMT-DC-stimulated primary CD4+ T cells (OMT-DCs-T) were co-cultured with the A549/DDP cells at ratios of 1:1, 1:5, 1:10 or 1:20 for 24 h. At the same time, the A549/DDP cells were treated with 22 µM DDP for 24 h. Annexin V/PI staining was used to analyze the apoptosis of the A549/DDP cells.

As presented in Fig. 4B, OMT-DC and OMT-DC-T may increase the percentage of apoptotic cells in A549/DDP, and the apoptotic rate decreased accompanied by the gradual dilution of the OMT-DCs/OMT-DCs-T with A549/DDP.

The western blotting results (Fig. 5) demonstrated that the expression of apoptosis-associated protein Bcl-2 increased as the A549/DDP cell apoptosis decreased; however, the protein expression level of Bax decreased. These results suggested that OMT-pretreated DCs and DC-induced T cells may reverse DDP-resistance in a non-contactable co-culture cell model.

Discussion

Carcinogenesis is a multistep process; agents that are able to target one or more of these processes maybe ideal cancer chemopreventive agents. According to previous studies (18,19), OMT may exert anticancer activities through various channels. Notably, it was observed that all of these functions were a result of direct contact of OMT with tumor cells. The present study is, to the best of the authors' knowledge, the first study to demonstrate that the therapeutic efficacy of OMT may be indirectly achieved by activating the DC-mediated antitumor immune response in circulation and/or in the tumor microenvironment.

Monocyte-derived mature DCs serve a key role in the immune response. Activated DCs may release various cytokines that mediate T cell activation and efficiently present a subset of antigens to T cells (20). At present, the potential impact of OMT on the maturation and function of DCs has not yet been fully investigated. In the present study, it was identified that OMT may enhance DC maturation in the process of differentiation of mononuclear cells derived from human peripheral blood. The positive effect of OMT on proliferation was significant at 1 mg/ml, and the surface markers of DCs, including CD83, CD86, CD11c and MHC II, were increased, suggesting that OMT may promote DC maturation. Immature DCs facilitate tolerance toward cancer cells, whereas, fully mature DCs may strongly promote anticancer immunity (21). The immune inhibitory microenvironment of the tumor inhibits DC differentiation and maturation (22,23). Efficiency and the success of such an interaction is dependent on the maturation status of the DCs (24). These data provide insight to support the use of OMT-mediated anticancer immunotherapy in human cancer.

DCs are key regulators of adaptive immunity with the potential to induce T cell activation/immunity or T cell suppression/tolerance. DCs are capable of differentiating naïve T cells into a range of effector cells, including immunogenic CD4+ Th cells, cytotoxic CD8+ T cells and tolerogenic Tregs (25). In the present study, similar to LPS, OMT-activated DCs were able to subsequently promote the differentiation of T cells into FOXP3+/CD4+ Treg cells in vitro. In agreement with the present results, Ma et al (26) demonstrated that OMT significantly upregulated FOXP3 and downregulated nuclear receptor ROR-γ expression, thereby inducing Treg/Th17 imbalance and inhibiting inflammation of rheumatoid arthritis. In contrast, it was identified that treatment with OMT triggers an increase in Th1 cytokines and a decrease in Th2 cytokines, which enhances the immune response and improves inhibitory activities to hepatitis B virus (14). Accordingly, OMT may promote the immune response to remove viruses and relieve hyperinflammation to protect organ function in autoimmune disorders (26). Therefore, it was hypothesized that OMT may have bilateral modulation on DC-T interaction, which may be switched mutually, according to the specific pathophysiological setting.

It is generally accepted that FOXP3+/CD4+ Treg infiltration in tumor tissue serves a role in inhibiting antitumor immunity and contributing to immune tolerance (2729), thus promoting the occurrence and progression of tumors. However, a number of previous studies demonstrated that FOXP3+/CD4+ Treg infiltration was positively associated with good prognosis in colorectal, head and neck cancer, NSCLC and breast cancer (3033). Immunosuppression or immune escape has been implicated as a primary mechanism in the initial establishment of tumors. Along with the secondary infection and cytotoxic drug usage in advanced stages, the resultant chronic systemic inflammation may alter the antigenic landscape of tumor cells, rewire oncogenic signaling networks, protect against cell death and reprogramme immune cell functions (34). It represents a shared resistance mechanism to different therapy (35,36). At this stage, a higher proportion of FOXP3+/CD4+ Tregs may relieve hyperinflammation and therapy-induced injury, thereby contributing to improve the overall prognosis of patients. Therefore, the DC-T response to treatment with OMT may vary at each stage of tumor progression.

The supernatant of the DC-T co-culture medium contained anti-inflammatory factors (including IL-10, TGF-β and IL-35) and pro-inflammatory cytokines (IFN-γ, IL-12 and IL-2). Furthermore, pro-inflammatory and anti-inflammatory factor secretion decreased when the co-cultured DCs were more diluted with the primitive T cells. This suggested that the DC-T co-culture system may affect the final anti-/pro-inflammatory concentration in the tumor microenvironment. The therapeutic effect of OMT is partly achieved by immune modulation. The protection of OMT is associated with inhibiting pro-inflammatory cytokines in acute lung injury (37), pulmonary fibrosis (38), rheumatoid arthritis (13) and organic ischemia/reperfusion injury (39,40). However, Ning et al (41) demonstrated that OMT was able to directly induce antiviral cytokine secretions in the peripheral lymphocytes isolated from patients with chronic hepatitis B. Therefore, further study is required to clarify the specific pattern of OMT regulating DC-T interaction, and determining the ultimate immune balance seems to be particularly critical.

In the past 5 years, MT and OMT have been extensively studied for their cancer chemopreventive potential against various cancer types. However, only a number of previous studies have investigated the underlying mechanisms of drug-resistance reversion. It was demonstrated that OMT and its derivatives may reverse DDP resistance by inhibiting multidrug resistance-associated protein 1 expression and the downregulation of Bax/Bcl-2 in the human nasopharyngeal carcinoma cell line HONE1 (42). Wang et al (43) observed that MT induced mitochondrial apoptosis in DDP-resistant NSCLC cells via suppression of β-catenin/survivin signaling (43). In addition, OMT may additionally reverse taxol resistance in NCI-H520/TAX25 human lung cancer cells (44), whereas, MT was able to induce apoptotic effects in doxorubicin resistant K562 cells (45). Therefore, OMT and its derivatives maybe used as ideal adjuncts for cancer chemotherapy.

In contrast to a previous study (46), it was demonstrated that OMT-pretreated DCs and DCs-T coculture may enhance apoptosis of DDP/A549 cells in vitro, without direct contact of OMT with tumor cells. Therefore, it was hypothesized that OMT-induced DC maturation and activation may be key in promoting DDP/A549 apoptosis. At present, it is well documented that mature DCs are able to induce strong antitumor responses mediated by effective candidates, including cytotoxic CD8+ T cells and large numbers of cytokines, to enhance sensitivity of tumor cells to various chemotherapeutic drugs (47). Immunogenic cell death of tumor cells starts from the interaction between dendritic cells and cancer cells. Cytokines released during apoptosis of tumor cells promote the maturation of dendritic cells. Mature dendritic cells activate tumor-specific cytotoxic T cells, which shows the antitumor effect (48). Further investigations on the molecular mechanisms of DC-mediated antitumor capacity following OMT administration are required.

As DCs are largely responsible for the ability of the immune response to recognize and eliminate cancer cells, DCs have attracted interest for antitumor immunotherapy in recent decades (49). In order for DC-based immunotherapy to elicit potent antitumor immune responses, an immune stimulatory response, as opposed to a tolerogenic immune response, is required following the administration of DCs. At present, research has focused on identifying the key factors responsible for therapeutic success/failure to determine the full clinical potential use of DCs in cancer immunotherapy. Due to the low toxicity and wide range of biological activities, OMT and specific natural products, which have therapeutic selectivity or may preferentially kill cancer cells without significant toxicity to normal cells, are being considered for future cancer therapy.

In conclusion, the multi-targeted chemopreventive efficacy of OMT was demonstrated in the present study, focusing on its immunoregulatory capacity. OMT may increase the apoptosis of drug-resistant tumor cells by activating DC differentiation and function, thereby enhancing the antitumor immune response. The present data suggested that OMT is a promising reagent for the development of more effective DC-based immunotherapy or cancer vaccines. The results of the present study are not only applicable to patients with tumors; however, additionally have reference value for infectious and immune diseases, and provide a theoretical basis for the use of OMT in the clinical setting.

Acknowledgements

Not applicable.

Funding

The present study was supported by funds from the China National Natural Science Funds (grant no. 81472760) and the Science and Technology Planning Project of Guangdong Province, China (grant no. 2014A020212078).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

HL designed all the experiments, applied for project funding and wrote the final version of the paper. MZ performed the in vitro cell culture, including isolation and culture of DCs and CD4+ T cells, as well as co-culture of DCs and T cells. PL performed flow cytometeric detection, including detection of surface markers of DCs and the ratio of FOXP3+ Tregs/CD4+. HW enrolled and collected the baseline data for the NSCLC patients and healthy controls. HW completed the detection of cytokines in supernatants using ELISA kits. XL completed the qPCR and western blotting in overall experiments, and completed the detection of A549 apoptosis. JY performed the statistical analysis and wrote the paper in cooperation with HL. JY and HL dealt with editorial queries and the overall submission. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All participants provided written consent, and the study was approved by the Institutional Review Board of Sun Yat-sen University.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Liu Y, Xu Y, Ji W, Li X, Sun B, Gao Q and Su C: Antitumor activities of matrine and oxymatrine: Literature review. Tumour Biol. 35:5111–5119. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Parvez MK, Arbab AH, Al-Dosari MS and Al-Rehaily AJ: Antiviral natural products against chronic hepatitis B: Recent developments. Curr Pharm Des. 22:286–293. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Liang CZ, Zhang JK, Shi Z, Liu B, Shen CQ and Tao HM: Matrine induces caspase-dependent apoptosis in human osteosarcoma cells in vitro and in vivo through the upregulation of Bax and Fas/FasL and downregulation of Bcl-2. Cancer Chemother Pharmacol. 69:317–331. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Zhao B, Li B, Bai S, Shen L, Ren R, Jonas JB, Xu X, Lu Q and Liu Q: Effects of matrine on proliferation and apoptosis of cultured retinoblastoma cells. Graefes Arch Clin Exp Ophthalmol. 250:897–905. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Xing Y, Yan F, Liu Y, Liu Y and Zhao Y: Matrine inhibits 3T3-L1 preadipocyte differentiation associated with suppression of ERK1/2 phosphorylation. Biochem Biophys Res Commun. 396:691–695. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Zhang S, Zhang Y, Zhuang Y, Wang J, Ye J, Zhang S, Wu J, Yu K and Han Y: Matrine induces apoptosis in human acute myeloid leukemia cells via the mitochondrial pathway and Akt inactivation. PLoS One. 7:e468532012. View Article : Google Scholar : PubMed/NCBI

7 

Chen H, Zhang J, Luo J, Lai F, Wang Z, Tong H, Lu D, Bu H, Zhang R and Lin S: Antiangiogenic effects of oxymatrine on pancreatic cancer by inhibition of the NF-κB-mediated VEGF signaling pathway. Oncol Rep. 30:589–595. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Yu H, Zhang H, Li D, Zhang X, Xue H and Zhao S: Matrine inhibits matrix metalloproteinase-9 expression and invasion of human hepatocellular carcinoma cells. J Asian Nat Prod Res. 13:242–250. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Sun Q, Ma W, Gao Y, Zheng W, Zhang B and Peng Y: Meta-analysis: Therapeutic effect of transcatheter arterial chemoembolization combined with compound kushen injection in hepatocellular carcinoma. Afr J Tradit Complement Altern Med. 9:178–188. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Villadangos JA, Schnorrer P and Wilson NS: Control of MHC class II antigen presentation in dendritic cells: A balance between creative and destructive forces. Immunol Rev. 207:191–205. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Merad M, Sathe P, Helft J, Miller J and Mortha A: The dendritic cell lineage: Ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol. 31:563–604. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Sallusto F and Lanzavecchia A: Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med. 179:1109–1118. 1994. View Article : Google Scholar : PubMed/NCBI

13 

Ghiringhelli F and Apetoh L: The interplay between the immune system and chemotherapy: emerging methods for optimizing therapy. Expert Rev Clin Immunol. 10:19–30. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Dong Y, Xi H, Yu Y, Wang Q, Jiang K and Li L: Effects of oxymatrine on the serum levels of T helper cell 1 and 2 cytokines and the expression of the S gene in hepatitis B virus S gene transgenic mice: A study on the anti-hepatitis B virus mechanism of oxymatrine. J Gastroenterol Hepatol. 17:1299–1306. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Zhou Y, Wang HL, Zhao WC, Chen Y and Deng HZ: Total alkaloids of Sophora alopecuroides increases the expression of CD4+ CD25+ Tregs and IL-10 in rats with experimental colitis. Am J Chin Med. 38:265–277. 2010. View Article : Google Scholar : PubMed/NCBI

16 

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

17 

Stein J, Steven S, Bros M, Sudowe S, Hausding M, Oelze M, Münzel T, Grabbe S, Reske-Kunz A and Daiber A: Role of protein kinase C and Nox2-derived reactive oxygen species formation in the activation and maturation of dendritic cells by phorbol ester and lipopolysaccharide. Oxid Med Cell Longev. 2017:41572132017. View Article : Google Scholar : PubMed/NCBI

18 

Li S, Zhang Y, Liu Q, Zhao Q, Xu L, Huang S, Huang S and Wei X: Oxymatrine inhibits proliferation of human bladder cancer T24 cells by inducing apoptosis and cell cycle arrest. Oncol Lett. 13:4453–4458. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Lin B, Li D and Zhang L: Oxymatrine mediates Bax and Bcl-2 expression in human breast cancer MCF-7 cells. Pharmazie. 71:154–157. 2016.PubMed/NCBI

20 

Sarhan D, Palma M, Mao Y, Adamson L, Kiessling R, Mellstedt H, Österborg A and Lundqvist A: Dendritic cell regulation of NK-cell responses involves lymphotoxin-α, IL-12, and TGF-β. Eur J Immunol. 45:1783–1793. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Dudek AM, Martin S, Garg AD and Agostinis P: Immature, semi-mature, and fully mature dendritic cells: Toward a DC-cancer cells interface that augments anticancer immunity. Front Immunol. 4:4382013. View Article : Google Scholar : PubMed/NCBI

22 

Menetrier-Caux C, Montmain G, Dieu MC, Bain C, Favrot MC, Caux C and Blay JY: Inhibition of the differentiation of dendritic cells from CD34(+) progenitors by tumor cells: Rrole of interleukin-6 and macrophage colony-stimulating factor. Blood. 92:4778–4791. 1998.PubMed/NCBI

23 

Bell D, Chomarat P, Broyles D, Netto G, Harb GM, Lebecque S, Valladeau J, Davoust J, Palucka KA and Banchereau J: In breast carcinoma tissue, immature dendritic cells reside within the tumor, whereas mature dendritic cells are located in peritumoral areas. J Exp Med. 190:1417–1426. 1999. View Article : Google Scholar : PubMed/NCBI

24 

Sittig S, de Vries I and Schreibelt G: Primary human blood dendritic cells for cancer immunotherapy-tailoring the immune response by dendritic cell maturation. Biomedicines. 3:282–303. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Joffre O, Nolte MA, Spörri R and Reis e Sousa C: Inflammatory signals in dendritic cell activation and the induction of adaptive immunity. Immunol Rev. 227:234–247. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Ma A, Yang Y, Wang Q, Yin W, Jing W and Zhang Y: Anti-inflammatory effects of oxymatrine on rheumatoid arthritis in rats via regulating the imbalance between Treg and Th17 cells. Mol Med Rep. 15:3615–3622. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Mao Y, Poschke I and Kiessling R: Tumour-induced immune suppression: Role of inflammatory mediators released by myelomonocytic cells. J Intern Med. 276:154–170. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Noy R and Pollard JW: Tumor-associated macrophages: From mechanisms to therapy. Immunity. 41:492014. View Article : Google Scholar : PubMed/NCBI

29 

Jandus C, Bioley G, Speiser D and Romero P: Selective accumulation of differentiated FOXP3(+) CD4 (+) T cells in metastatic tumor lesions from melanoma patients compared to peripheral blood. Cancer Immunol Immunother. 57:1795–1805. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Yeong J, Thike AA, Lim JC, Lee B, Li H, Wong SC, Hue SS, Tan PH and Iqbal J: Higher densities of Foxp3+ regulatory T cells are associated with better prognosis in triple-negative breast cancer. Breast Cancer Res Treat. 163:21–35. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Jackute J, Zemaitis M, Pranys D, Sitkauskiene B, Miliauskas S, Bajoriunas V, Lavinskiene S and Sakalauskas R: The prognostic influence of tumor infiltrating Foxp3(+)CD4(+), CD4(+) and CD8(+) T cells in resected non-small cell lung cancer. J Inflamm (Lond). 12:632015. View Article : Google Scholar : PubMed/NCBI

32 

West NR, Kost SE, Martin SD, Milne K, Deleeuw RJ, Nelson BH and Watson PH: Tumour-infiltrating FOXP3(+) lymphocytes are associated with cytotoxic immune responses and good clinical outcome in oestrogen receptor-negative breast cancer. Br J Cancer. 108:155–162. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Badoual C, Sandoval F, Pere H, Hans S, Gey A, Merillon N, Van Ryswick C, Quintin-Colonna F, Bruneval P, Brasnu D, et al: Better understanding tumor-host interaction in head and neck cancer to improve the design and development of immunotherapeutic strategies. Head Neck. 32:946–958. 2010.PubMed/NCBI

34 

Musolino C, Allegra A, Pioggia G and Gangemi S: Immature myeloid-derived suppressor cells: A bridge between inflammation and cancer. Oncol Rep. 37:671–683. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Gao F, Liang B, Reddy S, Farias-Eisner R and Su X: Role of inflammation-associated microenvironment in tumorigenesis and metastasis. Curr Cancer Drug Targets. 14:30–45. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Beatty GL: Overcoming therapeutic resistance by targeting cancer inflammation. Am Soc Clin Oncol Educ Book. 35:e168–e173. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Xu G, Yao L, Rao S, Gong Z, Zhang S and Yu S: Attenuation of acute lung injury in mice by oxymatrine is associated with inhibition of phosphorylated p38 mitogen-activated protein kinase. J Ethnopharmacol. 98:177–183. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Chen X, Sun R, Hu J, Mo Z, Yang Z, Liao D and Zhong N: Attenuation of bleomycin-induced lung fibrosis by oxymatrine is associated with regulation of fibroblast proliferation and collagen production in primary culture. Basic Clin Pharmacol Toxicol. 103:278–286. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Hu ST, Tang Y, Shen YF, Ao HH, Bai J, Wang YL and Yang YJ: Protective effect of oxymatrine on chronic rat heart failure. J Physiol Sci. 61:363–372. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Jiang G, Liu X, Wang M, Chen H, Chen Z and Qiu T: Oxymatrine ameliorates renal ischemia-reperfusion injury from oxidative stress through Nrf2/HO-1 pathway. Acta Cir Bras. 30:422–429. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Ning Y and Xin W: Effect on Toll-like receptor 7 signaling pathway in PBMCs of chronic hepatitis B patients by oxymatrine in vitro. Chin Arch Trad Chin Med. 36:1196–1199. 2018.

42 

Zhang J and Tang A: Reversal effect of matrine on drug resistance of human nasopharyngeal carcinoma cell line. Zhong Yao Cai. 35:1989–1994. 2012.(In Chinese). PubMed/NCBI

43 

Wang HQ, Jin JJ and Wang J: Matrine induces mitochondrial apoptosis in cisplatin-resistant non-small cell lung cancer cells via suppression of β-catenin/survivin signaling. Oncol Rep. 33:2561–2566. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Luo SX, Deng WY, Wang XF, Lü HF, Han LL, Chen BB, Chen XB and Li N: Molecular mechanism of indirubin-3′-monoxime and Matrine in the reversal of paclitaxel resistance in NCI-H520/TAX25 cell line. Chin Med J (Eng). 126:925–929. 2013.

45 

Chai S, To KK and Lin G: Circumvention of multi-drug resistance of cancer cells by Chinese herbal medicines. Chin Med. 5:262010. View Article : Google Scholar : PubMed/NCBI

46 

Wang B, Han Q and Zhu Y: Oxymatrine inhibited cell proliferation by inducing apoptosis in human lung cancer A549 cells. Biomed Mater Eng. 26 (Suppl 1):S165–S172. 2015.PubMed/NCBI

47 

Truxova I, Hensler M, Skapa P, Halaska MJ, Laco J, Ryska A, Spisek R and Fucikova J: Rationale for the combination of dendritic cell-based vaccination approaches with chemotherapy agents. Int Rev Cell Mol Biol. 330:115–156. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Di Blasio S, Wortel IM, van Bladel DA, de Vries LE, Duiveman-de Boer T, Worah K, de Haas N, Buschow SI, de Vries IJ, Figdor CG and Hato SV: Human CD1c(+) DCs are critical cellular mediators of immune responses induced by immunogenic cell death. Oncoimmunology. 5:e11927392016. View Article : Google Scholar : PubMed/NCBI

49 

Palucka K and Banchereau J: Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 12:265–277. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2019
Volume 19 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu H, Zou M, Li P, Wang H, Lin X and Ye J: Oxymatrine‑mediated maturation of dendritic cells leads to activation of FOXP3+/CD4+ Treg cells and reversal of cisplatin‑resistance in lung cancer cells. Mol Med Rep 19: 4081-4090, 2019
APA
Liu, H., Zou, M., Li, P., Wang, H., Lin, X., & Ye, J. (2019). Oxymatrine‑mediated maturation of dendritic cells leads to activation of FOXP3+/CD4+ Treg cells and reversal of cisplatin‑resistance in lung cancer cells. Molecular Medicine Reports, 19, 4081-4090. https://doi.org/10.3892/mmr.2019.10064
MLA
Liu, H., Zou, M., Li, P., Wang, H., Lin, X., Ye, J."Oxymatrine‑mediated maturation of dendritic cells leads to activation of FOXP3+/CD4+ Treg cells and reversal of cisplatin‑resistance in lung cancer cells". Molecular Medicine Reports 19.5 (2019): 4081-4090.
Chicago
Liu, H., Zou, M., Li, P., Wang, H., Lin, X., Ye, J."Oxymatrine‑mediated maturation of dendritic cells leads to activation of FOXP3+/CD4+ Treg cells and reversal of cisplatin‑resistance in lung cancer cells". Molecular Medicine Reports 19, no. 5 (2019): 4081-4090. https://doi.org/10.3892/mmr.2019.10064