Articles Service
Research article
Ganodermanontriol Suppresses the Progression of Lung Adenocarcinoma by Activating CES2 to Enhance the Metabolism of Mycophenolate Mofetil
1Respiratory Department, Longquan People’s Hospital, No. 699, Dongcha Road, Longquan City, Zhejiang Province, 323000, P.R. China
2Respiratory Department, The Sixth Affiliated Hospital of Wenzhou Medical University, No. 15 Dazhong Street, Liandu District, Lishui City, Zhejiang Province, 323000, P.R. China
3Longquan People’s Hospital, No. 699, Dongcha Road, Longquan City, Zhejiang Province, 323000, P.R. China
4Wenzhou Medical University, Wenzhou Chashan Higher Education Park, Wenzhou, Zhejiang Province, 325006, P.R. China
5School of Public Administration, Wenzhou Medical University, Wenzhou Chashan Higher Education Park, Wenzhou, Zhejiang Province, 325006, P.R. China
J. Microbiol. Biotechnol. 2024; 34(2): 249-261
Published February 28, 2024 https://doi.org/10.4014/jmb.2306.06020
Copyright © The Korean Society for Microbiology and Biotechnology.
Abstract
Keywords
Graphical Abstract
Introduction
Lung cancer is the second most common malignancy in the world and the leading cause of death in cancer patients [1]. Clinically, lung adenocarcinoma (LUAD), as the most frequent pathological type of non-small cell lung cancer (NSCLC), accounts for 85% of all lung cancer cases, with main characteristics of dense lymphocytic infiltration and metastasis in the early stage [2, 3]. Despite breakthroughs in early diagnosis and standard treatments for lung cancer over the decades, most patients are already at an advanced stage when they are diagnosed [4]. Additionally, the overall prognosis of LUAD still remains poor, with 5-year survival rates ranging from 68% in patients at TNM stage Ib to 0-10% in patients at stages IVa-IVb, due to occurrence of relapse, drug resistance, side effects of chemotherapy, etc. [5, 6]. Therefore, it is urgently needed to develop novel antineoplastic agents with minimal side effects.
Carboxylesterase (CES) belongs to the serine hydrolase superfamily, and can hydrolyze endogenous compounds and exogenous chemicals in human body [18]. CES2 is predominantly expressed in the small intestines, and is instrumental in the metabolism of pharmaceutical products containing ester and amide bonds [19]. Growing evidence has supported that the genetic variant of CES2 is strongly associated with drug resistance and recurrence of some tumors [20, 21]. Mycophenolate mofetil (MMF) is an immunosuppressant widely used in clinical practice to treat acute rejection of transplanted organs [22, 23]. Recently, mycophenolic acid (MPA), the active metabolite of MMF, has been reported to restrict the growth and metastatic development of LUAD [24], but its molecular mechanism in regulating lung cancer is still elusive. There is evidence that MPA is an immunosuppressive drug targeting inosine-5’-monophosphate dehydrogenases (IMPDHs) [25]. IMPDHs can be increased in tumors, and down-regulation of IMPDH can serve as an anticancer therapy [26]. Despite the confirmed effect of IMPDH on small lung cancer cells [26], its role in LUAD has not been explored.
In the present study, we investigated the effect of GDNT or MMF on cell apoptosis and cycle, as well as on tumor growth in LUAD in vitro and in vivo. In addition, we further probed into the mechanism of GDNT in influencing the anti-cancer effect of MMF. Based on our findings, GDNT can enhance the inhibitory effect of MMF on the progression of LUAD by activating CES2 to increase the production of MPA.
Methods
Reagent Preparation
GDNT (E2500) was purchased from Selleck Chemicals (USA). MMF (HY-B0199) was ordered from MedChemExpress (USA). Dimethyl sulfoxide (DMSO, GC203005, Servicebio, China) was utilized to dissolve GDNT or MMF to prepare stock solution and stored at -20°C.
Cell Culture and Treatment
Human lung cancer cell lines H1299 (CRL-5803) and A549 (CCL-185) were provided by the American Type Culture Collection (ATCC, USA). For cell culture, H1299 or A549 cells were grown in RPMI-1640 Medium (R8758, Sigma-Aldrich, USA) with supplement of 10% fetal bovine serum (10099141C, Thermo Fisher, USA) at 37°C in a humidified incubator (5% CO2, 95% air). For cell treatment, H1299 or A549 cells were incubated with GDNT at 0, 3.125, 6.25, 12.5, 25 or 50 μM, or with MMF at 0, 0.05, 0.5, 5, or 50 μg/ml in the culture medium for 24 h [15, 27].
Bioinformatics Analysis
UALCAN database (http://ualcan.path.uab.edu/) was applied to analyze differential expressions of IMPDH1 or IMPDH2 in LUAD based on 59 normal samples and 515 primary tumor samples [28]. Swisstargetprediction database (http://www.swisstargetprediction.ch/) was employed to analyze putative targets for GDNT [29].
Construction of Short Hairpin RNA (shRNA)
To knockdown CES2 in H1299 or A549 cells, shRNA targeting CES2 (shCES2; senses 5’-GGTCTCCAATTCTAGTTTA-3’, antisense: 5’-TAAACTAGAATTGGAGACC-3’) and its shRNA negative control (shNC) were synthesized by GenePharma (China). After culture, lung cancer cells were transferred into 6-well plates, and incubated overnight to reach 50-70% confluence. Then, cell transfection was performed with Escort IV Transfection Reagent (L3287, Sigma-Aldrich, USA) according to the manufacturer’s protocol. The cells transfected with shCES2 or shNC were harvested 48 h post transfection, and then subjected to examination of transfection efficiency using quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR).
RNA Isolation and qRT-PCR
Total RNA from lung cancer cells after cell transfection was isolated using RNeasy Mini Kit (74104, Qiagen, Germany). After quantification, complementary DNA (cDNA) was reversely transcribed from total RNA using RevertAid First Strand cDNA Synthesis Kit (K1622, Thermo Fisher, USA). The samples were mixed with Universal SYBR Green Supermix (1725120, Bio-Rad, USA), and then 7900HT Fast Real-Time PCR System (Applied Biosystems, USA) was used to conduct qRT-PCR. Primer sequences used in this reaction are as follows (5’-3’): CES2 (forward: CTAGGTCCGCTGCGATTTG, reverse: TGAGGTCCTGTAGACACATGG) and GAPDH (forward: GGAGCGAGATCCCTCCAAAAT, reverse: GGCTGTTGTCATACTTCTCATGG). The relative mRNA expression level of CES2 was calculated using 2-ΔΔCt method [30], with GAPDH serving as the internal control.
Methylthiazolyldiphenyl-Tetrazolium Bromide (MTT) Assay
Lung cancer cells with or without CES2 knockdown were seeded in 96-well plates (5 × 103 cells/well) and incubated overnight. Next, the cells were treated with GDNT or MMF at different concentrations, or co-treated with 6.25 μM GDNT and 5 μg/ml MMF at 37°C for 24 h. Thereafter, 20 μl MTT solution (G4101-1, Servicebio, China) was added into each well for further 4-h incubation. Following formazan dissolution by DMSO, MD SpectraMax M5 microplate reader (Molecular Devices, USA) was utilized to detect cell absorbance at 570 nm.
Assessment of Cell Apoptosis and Cycle
The determination of cell apoptosis was completed using Annexin V-FITC Apoptosis Detection Kit (CA1020, Solarbio, China). After cell transfection and/or cell treatment, lung cancer cells were suspended with 1×Binding Buffer at the concentration of 5 × 106 cells/ml and later incubated with 5 μl Annexin V-FITC solution at room temperature (RT) for 5 min in the dark. Subsequently, cells were cultured with 5 μl propidium iodide (PI) and 400 μl phosphate buffered saline (PBS; G4202, Servicebio, China), and finally CytoFLEX SRT flow cytometer (Beckman Coulter, USA) was employed to analyze the apoptotic cells.
PI staining was carried out to evaluate cell cycle. According the manual of Cell Cycle and Apoptosis Analysis Kit (C1052, Beyotime, China), 2 × 105 cells were suspended with pre-cooled PBS and fixed with 70% ethanol (G2350, Solarbio, China) at 4°C for 12 h. After 5 min of centrifugation (1,000 ×
Animals and Ethics Statement
Male immunodeficient nude mice (nu/nu, 6 weeks old) were reared in a controlled environment (26-28°C, 40-60% humidity, 12-h on/off light cycles) and allowed to access food and drinking water
Xenograft and Drug Administration
For performing in vivo xenograft, H1299 cells (5 × 106) were suspended with 0.2 ml RPMI-1640 Medium, and injected subcutaneously into the right flank of each mouse. After 7 days of injection, mice were randomly divided into four groups (
Immunohistochemistry (IHC)
Tumor samples were fixed with 10% formalin (E672001, Sangon Biotech, China) and embedded into paraffin according to standard procedures. Through microtomy, 5 μm sections were obtained, exposed to 3% hydrogen peroxide (H299581, Aladdin, China) for 20 min, washed with PBS and treated with 0.01 M citric acid (abs44109576, Absin, China). Subsequently, the sections were treated with 5% non-fat milk (abs9175, Absin, China) in 1×TBS with Tween-20 (TBST; G0004, Servicebio, China) at RT for 30 min. Then, the sections were incubated with CES2 antibody (PA5-102415, Thermo Fisher) at 4°C overnight, followed by hybridization with horseradish peroxidase (HRP)-conjugated mouse anti-rabbit IgG (D110065, Sangon Biotech, China) at RT for 1 h. After treatment with DAB (D12384, Sigma-Aldrich) and counterstaining with hematoxylin (G1004, Servicebio, China) in sequence, AXio Lab.A1 microscope (×100 magnification, Zeiss, ermany) was used to observe positive signals of CES2 in the sections.
Western Blot
Cells or fresh tumor samples were homogenized in RIPA Buffer (R0010, China) as per the manufacturer’s specification to extract total protein which then was quantified by BCA Protein Assay Kit (PC0020, China). Equal amounts of protein samples (15 μg/lane) were separated using 10% SDS-PAGE and loaded onto polyvinylidene difluoride membranes (0.45 μm, 88585, Thermo Fisher), followed by blocking with TBST-diluted 5% non-fat milk (GC310001, Servicebio, China). Primary antibodies against CES2 (ab184957, 62 kDa), IMPDH1 (ab33039, 55 kDa), IMPDH2 (ab129165, 56 kDa) and GAPDH (ab181603, 36 kDa) were incubated with the membranes at 4°C overnight. Next, the membranes were hybridized with HRP-conjugated secondary antibody (ab6721) at RT for 1 h. All antibodies used in Western blot were provided by Abcam (UK). After that, the stripped membranes were treated with ECL Western Blotting Detection Kit (SW2040, Solarbio, China), and immunoblots were detected by ChemiDoc XRS+ imaging system (Bio-Rad). Bandscan software (Bio-Rad) was exploited to analyze relative protein expression (represented as the ratio of the gray-scale value of the target band to that of the GAPDH band).
Statistical Analysis
Data from all experiments performed thrice were expressed as mean ± standard deviation. Comparisons between two groups or among multiple groups were carried out using independent-samples
-
Fig. 3. The expression of IMPDH1/2 in LUAD and the target prediction of GDNT.
(A, B) Expressions of IMPDH1 and IMPDH2 in LUAD based on 59 normal samples and 515 primary tumor samples were analyzed using UALCAN database (http://ualcan.path.uab.edu/). (C) Swisstargetprediction database (http://www.swisstargetprediction.ch/) was employed to analyze putative targets for GDNT.
Results
GDNT Enhanced the Effects of MMF on Suppressing Viability, Promoting Apoptosis and Inducing Cell Cycle Arrest in Lung Cancer Cells
Previous evidence has shown that GDNT (Fig. 1A), a lanostanoid triterpene extracted from
-
Fig. 1. The effects of GDNT and/or MMF on viability and apoptosis of lung cancer cells.
(A) The chemical structure of GDNT. (B-E) H1299 or A549 cells were treated with GDNT (0, 3.125, 6.25, 12.5, 25, 50 μM) or MMF (0, 0.05, 0.5, 5, 50 μg/ml) for 24 h, and MTT assay was performed to examine cell viability. (F, G) H1299 or A549 cells were treated with 6.25 μM GDNT and/or 5 μg/ml MMF for 24 h, and MTT assay was conducted to test cell viability. (H-J) After treatment with 6.25 μM GDNT and/or 5 μg/ml MMF for 24 h, flow cytometry was used to analyze cell apoptosis. The horizontal axis in the H graph represented the number of FITC stained cells, while the vertical axis represented the number of PI stained cells. Data from all experiments performed thrice were expressed as mean ± standard deviation. *
p < 0.05, **p < 0.01, ***p < 0.001, vs. 0; ^p < 0.05, ^^p < 0.01, ^^^p < 0.001, vs. Control; θθθp < 0.001, vs. MMF; ###p < 0.001, vs. GDNT. MMF, mycophenolate mofetil; GDNT, ganodermanontriol; MTT, methylthiazolyldiphenyl-tetrazolium bromide.
-
Fig. 2. The effect of GDNT and/or MMF on lung cancer cell cycle.
(A) H1299 or A549 cells were treated with 6.25 μM GDNT and/or 5 μg/ml MMF for 24 h, and flow cytometry was applied to analyze cell cycle. (B, C) Bar chart of cell cycle proportion in the Control, GDNT, MMF and MMF+GDNT groups. Data from all experiments performed thrice were expressed as mean ± standard deviation. ^^^
p < 0.001, vs. Control; θθθp < 0.001, vs. MMF; ###p < 0.001, vs. GDNT.
IMPDH1 and IMPDH2 Were Highly Expressed in LUAD Samples and CES2 Was a Potential Target for GDNT
Based on TCGA, UALCAN database predicted that IMPDH1 expression was higher in primary tumors from LUAD samples than in normal samples (Fig. 3A,
GDNT Up-Regulated CES2 Expression and Enhanced the Effect of MMF on Down-Regulating IMPDH1 and IMPDH2 Expressions in Lung Cancer Cells
CES2 has been indicated to influence MMF metabolism, given the genetic polymorphism of this metabolic enzyme [22]. In H1299 or A549 cells, it was determined that the protein level of CES2 was up-regulated by GDNT treatment (Fig. 4A-4C,
-
Fig. 4. The effect of GDNT and/or MMF on CES2, IMPDH1 and IMPDH2 expressions in lung cancer cells.
(A-C) H1299 or A549 cells were treated with 6.25 μM GDNT and/or 5 μg/ml MMF for 24 h, and Western blot was performed to detect protein level of CES2 in the Control, GDNT, MMF and MMF+GDNT groups. (D-F) After the cell treatment for 24 h, the protein expression levels of IMPDH1 and IMPDH2 in H1299 cells were measured by Western blot. (G-I) Following the cell treatment for 24 h, IMPDH1 and IMPDH2 protein expressions in A549 cells were determined by Western blot. Relative expression levels were normalized with GAPDH. Data from all experiments performed thrice were expressed as mean ± standard deviation. ^^
p < 0.01, ^^^p < 0.001, vs. Control; θp < 0.05, θθp < 0.01, θθθp < 0.001, vs. MMF; ###p < 0.001, vs. GDNT. CES2, carboxylesterase 2; IMPDH, inosine-5’-monophosphate dehydrogenase.
CES2 Knockdown Reversed the Synergistic Effect of GDNT and MMF against Lung Cancer In Vitro
Next, we explored the effect of CES2 on the development of lung cancer cells in the presence of MMF and GDNT. After transfection with shCES2, the mRNA and protein expressions of CES2 in either H1299 or A549 cells were strikingly decreased (Fig. 5A-5E,
-
Fig. 5. CES2 knockdown reversed the synergistic effect of GDNT and MMF on viability and apoptosis of lung cancer cells.
(A, B) H1299 or A549 cells were transfected with shCES2 or shNC for 48 h, and qRT-PCR was performed to examine the mRNA expression of CES2, with GAPDH used as the internal control. (C-E) The protein expression of CES2 in the Control, shNC and shCES2 groups was determined by Western blot, with GAPDH used as the internal control. (F, G) After cell transfection and co-treatment with 6.25 μM GDNT and/or 5 μg/ml MMF, the viability of H1299 and A549 cells was evaluated by MTT assay. (H-J) The apoptosis of the indicated cells was tested by flow cytometry. Data from all experiments performed thrice were expressed as mean ± standard deviation. +++
p < 0.001, vs. shNC; θθθp < 0.001, vs. MMF; ΔΔΔp < 0.001, vs. MMF + GDNT + shNC. shCES2, short hairpin RNA (shRNA) targeting CES2; shNC, shRNA negative control.
-
Fig. 6. CES2 knockdown offset the synergistic effect of GDNT and MMF on cell cycle and IMPDH1/2 expression in lung cancer cells.
(A-C) After cell transfection with shCES2 and co-treatment with 6.25 μM GDNT and/or 5 μg/ml MMF, the cell cycle of H1299 and A549 cells was analyzed by flow cytometry. (D-I) Protein levels of IMPDH1 and IMPDH2 in the indicated cells were measured by Western blot. Relative expression levels were normalized with GAPDH. Data from all experiments performed thrice were expressed as mean ± standard deviation. θθ
p < 0.01, θθθp < 0.001, vs. MMF; ΔΔp < 0.01, ΔΔΔp < 0.001, vs. MMF + GDNT + shNC. shCES2, short hairpin RNA (shRNA) targeting CES2; shNC, shRNA negative control.
GDNT Potentiated the Tumor-Suppressive Effect of MMF on Lung Cancer In Vivo
Considering the prominent role of GDNT in suppressing the development of lung cancer cells by increasing apoptosis, the regulatory effect of GDNT cooperated with MMF on tumorigenesis in lung cancer should be explored. Thus, we performed xenograft experiment through subcutaneously injecting H1299 cells into nude mice, followed by administration with GDNT and/or MMF. 45 days later, it was observed that either GDNT or MMF markedly reduced the volume and weight of tumors excised from the mice (Fig. 7A-7C,
-
Fig. 7. GDNT enhanced the effect of MMF on suppressing tumor growth of lung cancer in vivo.
(A) Xenograft experiment was performed by subcutaneously injecting H1299 cells into mice, followed by treatment of MMF (20 mg/kg/2d) and/or GDNT (3 mg/kg/1d). After 45 days, all tumor-bearing mice were sacrificed, and the tumors were excised and photographed. (B) The volume of tumors was calculated. (C) The weight of tumors was measured. Data from all experiments performed thrice were expressed as mean ± standard deviation. ^^
p < 0.01, ^^^p < 0.001, vs. Control; θθθp < 0.001, vs. MMF; ###p < 0.001, vs. GDNT.
-
Fig. 8. GDNT strengthened the effect of MMF on down-regulating CES2 and IMPDH1/2 expressions in lung cancer in vivo.
(A, B) After xenograft experiment, immunohistochemistry was carried out to detect the expression of CES2 in the excised tumors. (C-E) Protein levels of IMPDH1 and IMPDH2 in the excised tumors were determined by Western blot. Relative expression levels were normalized with GAPDH. Data from all experiments performed thrice were expressed as mean ± standard deviation. ^^^
p < 0.001, vs. Control; θp < 0.05, θθp < 0.01, vs. MMF; ###p < 0.001, vs. GDNT.
Discussion
Currently, radiotherapy and first-line antineoplastic agents remain the basic treatments for most patients with advanced NSCLC, but the overall survival rate is affected by factors including side effects, non-targeted cytotoxicity and drug resistance [33]. With the rise of integrative therapies for malignant tumors,
Ample evidence at home and abroad has supported that
As an immunosuppressant, MMF, which can be hydrolyzed into MPA in the body, is widely used in the clinical treatment after solid organ transplantation to prevent rejection reaction through inhibiting IMPDH [41], but has been recently evidenced to repress the tumor progression via targeting tumor-associated fibroblast [31]. Although the anti-proliferative role of MMF has been confirmed in lung cancer, its underlying mechanism in LUAD is not fully expounded. Of note, this study unveiled that MMF treatment alone boosted apoptosis of LUAD cells and blocked intercellular DNA replication by augmenting the percentage of S phase, consistent with the findings of Ling
Based on the above-mentioned findings, we subsequently investigated the possible molecular mechanism of GDNT enhancing the anti-cancer effect of MMF. Through bioinformatics analysis, we found that CES2 was predicted as a potential target for GDNT. Reportedly, CES2 impacts the clinical results of cancer patients by affecting chemosensitivity [43]. In the study of graft rejection, CES2 has been suggested to function as a mediator in the metabolism of MMF to MPA [22]. In lung cancer cells, the expression of CES2 was increased by GDNT with/without MMF but was not significantly affected in the presence of MMF alone. MPA can target IMPDH which has been confirmed to be highly expressed in many tumors [44, 45]. In this study, the results of Western blot revealed that both IMPDH1 and IMPDH2 expressions were down-regulated by MMF in the cells, which was further strengthened by GDNT. IMPDH1 and IMPDH2 are two isoforms of IMPDH that is implicated in cell metabolism and proliferation by modulating
Conclusion
Despite the importance of IMPDH in anti-cancer treatment, the development and application of IMPDH inhibitors are limited by their side effects at high doses [25]. The present study unravels the anti-cancer mechanism of GDNT in LUAD, and further provides a new direction for developing adjuvant agents to enhance the efficacy of IMPDH inhibitors in treating lung cancer.
Supplemental Materials
Acknowledgments
This work was supported by the Zhejiang Provincial Medical and Health Project [2023587787]; the Zhejiang Provincial Science and Technology Department Project [LGF22H010012]; the Longquan Science and Technology Bureau Project [2021KJ-003]; the Zhejiang Provincial Medical and Health Research Project [2021KY1235]; the Lishui Science and Technology Bureau Project [2020077571].
Author Contributions
Substantial contributions to conception and design: Qingfeng Xie and Zhuo Cao
Data acquisition, data analysis and interpretation: Weiling You, Xiaoping Cai, Mei Shen, Zhangyong Yin, Yiwei Jiang, Xin Wang, Siyu Ye
Drafting the article or critically revising it for important intellectual content: Qingfeng Xie, Zhuo Cao, Weiling You and Xiaoping Cai, Mei Shen, Zhangyong Yin, Yiwei Jiang, Xin Wang, Siyu Ye
Final approval of the version to be published: All authors
Agreement to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of the work are appropriately investigated and resolved: Qingfeng Xie, Zhuo cao, Weiling You, Xiaoping Cai, Mei Shen, Zhangyong Yin, Yiwei Jiang, Xin Wang, Siyu Ye
Conflict of Interest
The authors have no financial conflicts of interest to declare.
References
- Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A,
et al . 2021. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries.CA: Cancer J. Clin. 71 : 209-249. - Herbst RS, Morgensztern D, Boshoff C. 2018. The biology and management of non-small cell lung cancer.
Nature 553 : 446-454. - Luo C, Lei M, Zhang Y, Zhang Q, Li L, Lian J,
et al . 2020. Systematic construction and validation of an immune prognostic model for lung adenocarcinoma.J. Cell. Mol. Med. 24 : 1233-1244. - Nooreldeen R, Bach H. 2021. Current and future development in lung cancer diagnosis.
Int. J. Mol. Sci. 22 : 8661. - Kadara H, Scheet P, Wistuba II, Spira AE. 2016. Early events in the molecular pathogenesis of lung cancer.
Cancer Prev. Res. 9 : 518-527. - Duma N, Santana-Davila R, Molina JR. 2019. Non-small cell lung cancer: epidemiology, screening, diagnosis, and treatment.
Mayo Clin. Proc. 94 : 1623-1640. - Soccol CR, Bissoqui LY, Rodrigues C, Rubel R, Sella SR, Leifa F,
et al . 2016. Pharmacological properties of biocompounds from spores of the lingzhi or reishi medicinal mushroomGanoderma lucidum (Agaricomycetes ): a review.Int. J. Med. Mushrooms 18 : 757-767. - Chan SW, Tomlinson B, Chan P, Lam CWK. 2021. The beneficial effects of
Ganoderma lucidum on cardiovascular and metabolic disease risk.Pharm. Biol. 59 : 1161-1171. - Sohretoglu D, Huang S. 2018.
Ganoderma lucidum polysaccharides as an anti-cancer agent.Anticancer Agents Med. Chem. 18 : 667-674. - Shao Y, Qiao L, Wu L, Sun X, Zhu D, Yang G,
et al . 2016. Structure identification and anti-cancer pharmacological prediction of triterpenes fromGanoderma lucidum .Molecules 21 : 678. - Barbieri A, Quagliariello V, Del Vecchio V, Falco M, Luciano A, Amruthraj NJ,
et al . 2017. Anticancer and anti-inflammatory properties ofGanoderma lucidum extract effects on melanoma and triple-negative breast cancer treatment.Nutrients 9 : 210. - Liu W, Yuan R, Hou A, Tan S, Liu X, Tan P,
et al . 2020.Ganoderma triterpenoids attenuate tumour angiogenesis in lung cancer tumour-bearing nude mice.Pharm. Biol. 58 : 1061-1068. - Zhu J, Xu J, Jiang LL, Huang JQ, Yan JY, Chen YW,
et al . 2019. Improved antitumor activity of cisplatin combined withGanoderma lucidum polysaccharides in U14 cervical carcinoma-bearing mice.Kaohsiung J. Med. Sci. 35 : 222-229. - Li X, Xie Y, Yang BB. 2018. Characterizing novel anti-oncogenic triterpenoids from ganoderma.
Cell Cycle 17 : 527-528. - Jiang J, Jedinak A, Sliva D. 2011. Ganodermanontriol (GDNT) exerts its effect on growth and invasiveness of breast cancer cells through the down-regulation of CDC20 and uPA.
Biochem. Biophys. Res. Commun. 415 : 325-329. - Jedinak A, Thyagarajan-Sahu A, Jiang J, Sliva D. 2011. Ganodermanontriol, a lanostanoid triterpene from
Ganoderma lucidum , suppresses growth of colon cancer cells through ß-catenin signaling.Int. J. Oncol. 38 : 761-767. - Xu C, Guo H, Kong D, Pang D, Ding Y. 2018. Ganodermanontriol inhibits expression of special AT rich sequence binding protein 1 gene in human hepatocellular carcinoma.
J. Cancer Res. Ther. 14 : S964-s968. - Zou LW, Jin Q, Wang DD, Qian QK, Hao DC, Ge GB,
et al . 2018. Carboxylesterase inhibitors: an update.Curr. Med. Chem. 25 : 1627-1649. - Chen F, Zhang B, Parker RB, Laizure SC. 2018. Clinical implications of genetic variation in carboxylesterase drug metabolism.
Exp. Opin. Drug Metab. Txicol. 14 : 131-142. - Lam SW, van der Noort V, van der Straaten T, Honkoop AH, Peters GJ, Guchelaar HJ,
et al . 2018. Single-nucleotide polymorphisms in the genes of CES2, CDA and enzymatic activity of CDA for prediction of the efficacy of capecitabine-containing chemotherapy in patients with metastatic breast cancer.Pharm. Res. 128 : 122-129. - Zhang Y, Sun L, Sun Y, Chen Y, Wang X, Xu M,
et al . 2020. Overexpressed CES2 has prognostic value in CRC and knockdown CES2 reverses L-OHP-resistance in CRC cells by inhibition of the PI3K signaling pathway.Exp. Cell Res. 389 : 111856. - Cilião HL, Camargo-Godoy RBO, Souza MF, Zanuto A, Delfino VDA, Cólus IMS. 2018. Polymorphisms in IMPDH2, UGT2B7, and CES2 genes influence the risk of graft rejection in kidney transplant recipients taking mycophenolate mofetil.
Mut. Res. Genet. Toxicol. Environ. Mutagen. 836 : 97-102. - Parlakpinar H, Gunata M. 2021. Transplantation and immunosuppression: a review of novel transplant-related immunosuppressant drugs.
Immunopharmacol. Immunotoxicol. 43 : 651-665. - Chen X, Zhao Y, Wang D, Lin Y, Hou J, Xu X,
et al . 2021. The HNF4α-BC200-FMR1-positive feedback loop promotes growth and metastasis in invasive mucinous lung adenocarcinoma.Cancer Res. 81 : 5904-5918. - Naffouje R, Grover P, Yu H, Sendilnathan A, Wolfe K, Majd N,
et al . 2019. Anti-tumor potential of IMP dehydrogenase inhibitors: a century-long story.Cancers 11 : 1346. - Keppeke GD, Andrade LEC, Barcelos D, Fernandes M, Landman G. 2020. IMPDH-based cytoophidium structures as potential theranostics in cancer.
Mol. Ther. 28 : 1557-1558. - Stracke S, Ramudo L, Keller F, Henne-Bruns D, Mayer JM. 2006. Antiproliferative and overadditive effects of everolimus and mycophenolate mofetil in pancreas and lung cancer cells in vitro.
Transplant. Proc. 38 : 766-770. - Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi B,
et al . 2017. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses.Neoplasia 19 : 649-658. - Daina A, Michielin O, Zoete V. 2019. SwissTargetPrediction: updated data and new features for efficient prediction of protein targets of small molecules.
Nucleic Acids Res. 47 : W357-w364. - Livak KJ, Schmittgen TD. 2001. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method.
Methods 25 : 402-408. - Yang Z, Zhang L, Zhu H, Zhou K, Wang H, Wang Y,
et al . 2021. Nanoparticle formulation of mycophenolate mofetil achieves enhanced efficacy against hepatocellular carcinoma by targeting tumour-associated fibroblast.J. Cell. Mol. Med. 25 : 3511-3523. - Kennedy EM, P'Pool SJ, Jiang J, Sliva D, Minto RE. 2011. Semisynthesis and biological evaluation of ganodermanontriol and its stereoisomeric triols.
J. Nat. Prod. 74 : 2332-2337. - Wang M, Herbst RS, Boshoff C. 2021. Toward personalized treatment approaches for non-small-cell lung cancer.
Nat. Med. 27 : 1345-1356. - Guo J, Yuan C, Huang M, Liu Y, Chen Y, Liu C,
et al . 2018.Ganoderma lucidum -derived polysaccharide enhances coix oil-based microemulsion on stability and lung cancer-targeted therapy.Drug Deliv. 25 : 1802-1810. - Feng L, Yuan L, Du M, Chen Y, Zhang MH, Gu JF,
et al . 2013. Anti-lung cancer activity through enhancement of immunomodulation and induction of cell apoptosis of total triterpenes extracted fromGanoderma luncidum (Leyss. ex Fr.) Karst.Molecules (Basel, Switzerland) 18 : 9966-9981. - Zolj S, Smith MP, Goines JC, Ali TS, Huff MO, Robinson DL,
et al . 2018. Antiproliferative effects of a rriterpene-enriched extract from lingzhi or reishi medicinal mushroom,Ganoderma lucidum (Agaricomycetes), on human lung cancer cells.Int. J. Med. Mushrooms 20 : 1173-1183. - Wang T, Xie ZP, Huang ZS, Li H, Wei AY, Di JM,
et al . 2015. Total triterpenoids fromGanoderma Lucidum suppresses prostate cancer cell growth by inducing growth arrest and apoptosis.J. Huazhong Univ. Sci. Technol. Med. Sci. 35 : 736-741. - Wang H, Liu Z, Wu P, Wang H, Ren W. 2021. NUSAP1 accelerates osteosarcoma cell proliferation and cell cycle progression via upregulating CDC20 and cyclin A2.
OncoTargets Ther. 14 : 3443-3454. - Chu Z, Zhang X, Li Q, Hu G, Lian CG, Geng S. 2019. CDC20 contributes to the development of human cutaneous squamous cell carcinoma through the Wnt/β-catenin signaling pathway.
Int. J. Oncol. 54 : 1534-1544. - Hu H, Tou FF, Mao WM, Xu YL, Jin H, Kuang YK,
et al . 2022. microRNA-1321 and microRNA-7515 contribute to the progression of non-small cell lung cancer by targeting CDC20.Kaohsiung J. Med. Sci. 38 : 425-436. - Plunk MA, Quintana JM, Darden CM, Lawrence MC, Naziruddin B, Kane RR. 2021. Design and catalyzed activation of mycophenolic acid prodrugs.
ACS Med. Chem. Lett. 12 : 812-816. - Ling G, Lamprecht S, Shubinsky G, Osyntsov L, Yerushalmi B, Pinsk I,
et al . 2018. Mycophenolate mofetil alone and in combination with tacrolimus inhibits the proliferation of HT-29 human colonic adenocarcinoma cell line and might interfere with colonic tumorigenesis.Anticancer Res. 38 : 3333-3339. - Silvestris N, Simone G, Partipilo G, Scarpi E, Lorusso V, Brunetti AE,
et al . 2014. CES2, ABCG2, TS and Topo-I primary and synchronous metastasis expression and clinical outcome in metastatic colorectal cancer patients treated with first-line FOLFIRI regimen.Int. J. Mol. Sci. 15 : 15767-15777. - Winnicki W, Fichtenbaum A, Mitulovič G, Herkner H, Regele F, Baier M,
et al . 2022. Individualization of mycophenolic acid therapy through pharmacogenetic, pharmacokinetic and pharmacodynamic testing.Biomedicines 10 : 2882. - Siebert A, Deptuła M, Cichorek M, Ronowska A, Cholewiński G, Rachon J. 2021. Anticancer properties of amino acid and peptide derivatives of mycophenolic acid.
Antcancer Agents Med. Chem. 21 : 462-467. - Keppeke GD, Chang CC, Peng M, Chen LY, Lin WC, Pai LM,
et al . 2018. IMP/GTP balance modulates cytoophidium assembly and IMPDH activity.Cell Div. 13 : 5. - Valvezan AJ, McNamara MC, Miller SK, Torrence ME, Asara JM, Henske EP,
et al . 2020. IMPDH inhibitors for antitumor therapy in tuberous sclerosis complex.JCI Insight 5 : e135071. - Huang F, Ni M, Chalishazar MD, Huffman KE, Kim J, Cai L,
et al . 2018. Inosine monophosphate dehydrogenase dependence in a subset of small cell lung cancers.Cell Metab. 28 : 369-382.e365. - Ruan H, Song Z, Cao Q, Ni D, Xu T, Wang K,
et al . 2020. IMPDH1/YB-1 positive feedback loop assembles cytoophidia and represents a therapeutic target in metastatic tumors.Mole. Ther. 28 : 1299-1313.
Related articles in JMB
Article
Research article
J. Microbiol. Biotechnol. 2024; 34(2): 249-261
Published online February 28, 2024 https://doi.org/10.4014/jmb.2306.06020
Copyright © The Korean Society for Microbiology and Biotechnology.
Ganodermanontriol Suppresses the Progression of Lung Adenocarcinoma by Activating CES2 to Enhance the Metabolism of Mycophenolate Mofetil
Qingfeng Xie1†, Zhuo Cao2†, Weiling You1, Xiaoping Cai2, Mei Shen3, Zhangyong Yin2, Yiwei Jiang4, Xin Wang4, and Siyu Ye5*
1Respiratory Department, Longquan People’s Hospital, No. 699, Dongcha Road, Longquan City, Zhejiang Province, 323000, P.R. China
2Respiratory Department, The Sixth Affiliated Hospital of Wenzhou Medical University, No. 15 Dazhong Street, Liandu District, Lishui City, Zhejiang Province, 323000, P.R. China
3Longquan People’s Hospital, No. 699, Dongcha Road, Longquan City, Zhejiang Province, 323000, P.R. China
4Wenzhou Medical University, Wenzhou Chashan Higher Education Park, Wenzhou, Zhejiang Province, 325006, P.R. China
5School of Public Administration, Wenzhou Medical University, Wenzhou Chashan Higher Education Park, Wenzhou, Zhejiang Province, 325006, P.R. China
Correspondence to:Siyu Ye, yesiyu_ysy1@163.com
†These authors contributed equally to this work.
Abstract
New anti-lung cancer therapies are urgently required to improve clinical outcomes. Since ganodermanontriol (GDNT) has been identified as a potential antineoplastic agent, its role in lung adenocarcinoma (LUAD) is investigated in this study. Concretely, lung cancer cells were treated with GDNT and/or mycophenolate mofetil (MMF), after which MTT assay, flow cytometry and Western blot were conducted. Following bioinformatics analysis, carboxylesterase 2 (CES2) was knocked down and rescue assays were carried out in vitro. Xenograft experiment was performed on mice, followed by drug administration, measurement of tumor growth and determination of CES2, IMPDH1 and IMPDH2 expressions. As a result, the viability of lung cancer cells was reduced by GDNT or MMF. GDNT enhanced the effects of MMF on suppressing viability, promoting apoptosis and inducing cell cycle arrest in lung cancer cells. GDNT up-regulated CES2 level, and strengthened the effects of MMF on down-regulating IMPDH1 and IMPDH2 levels in the cells. IMPDH1 and IMPDH2 were highly expressed in LUAD samples. CES2 was a potential target for GDNT. CES2 knockdown reversed the synergistic effect of GDNT and MMF against lung cancer in vitro. GDNT potentiated the role of MMF in inhibiting tumor growth and expressions of CES2 and IMPDH1/2 in lung cancer in vivo. Collectively, GDNT suppresses the progression of LUAD by activating CES2 to enhance the metabolism of MMF.
Keywords: Lung adenocarcinoma, ganodermanontriol, mycophenolate mofetil, carboxylesterase 2
Introduction
Lung cancer is the second most common malignancy in the world and the leading cause of death in cancer patients [1]. Clinically, lung adenocarcinoma (LUAD), as the most frequent pathological type of non-small cell lung cancer (NSCLC), accounts for 85% of all lung cancer cases, with main characteristics of dense lymphocytic infiltration and metastasis in the early stage [2, 3]. Despite breakthroughs in early diagnosis and standard treatments for lung cancer over the decades, most patients are already at an advanced stage when they are diagnosed [4]. Additionally, the overall prognosis of LUAD still remains poor, with 5-year survival rates ranging from 68% in patients at TNM stage Ib to 0-10% in patients at stages IVa-IVb, due to occurrence of relapse, drug resistance, side effects of chemotherapy, etc. [5, 6]. Therefore, it is urgently needed to develop novel antineoplastic agents with minimal side effects.
Carboxylesterase (CES) belongs to the serine hydrolase superfamily, and can hydrolyze endogenous compounds and exogenous chemicals in human body [18]. CES2 is predominantly expressed in the small intestines, and is instrumental in the metabolism of pharmaceutical products containing ester and amide bonds [19]. Growing evidence has supported that the genetic variant of CES2 is strongly associated with drug resistance and recurrence of some tumors [20, 21]. Mycophenolate mofetil (MMF) is an immunosuppressant widely used in clinical practice to treat acute rejection of transplanted organs [22, 23]. Recently, mycophenolic acid (MPA), the active metabolite of MMF, has been reported to restrict the growth and metastatic development of LUAD [24], but its molecular mechanism in regulating lung cancer is still elusive. There is evidence that MPA is an immunosuppressive drug targeting inosine-5’-monophosphate dehydrogenases (IMPDHs) [25]. IMPDHs can be increased in tumors, and down-regulation of IMPDH can serve as an anticancer therapy [26]. Despite the confirmed effect of IMPDH on small lung cancer cells [26], its role in LUAD has not been explored.
In the present study, we investigated the effect of GDNT or MMF on cell apoptosis and cycle, as well as on tumor growth in LUAD in vitro and in vivo. In addition, we further probed into the mechanism of GDNT in influencing the anti-cancer effect of MMF. Based on our findings, GDNT can enhance the inhibitory effect of MMF on the progression of LUAD by activating CES2 to increase the production of MPA.
Methods
Reagent Preparation
GDNT (E2500) was purchased from Selleck Chemicals (USA). MMF (HY-B0199) was ordered from MedChemExpress (USA). Dimethyl sulfoxide (DMSO, GC203005, Servicebio, China) was utilized to dissolve GDNT or MMF to prepare stock solution and stored at -20°C.
Cell Culture and Treatment
Human lung cancer cell lines H1299 (CRL-5803) and A549 (CCL-185) were provided by the American Type Culture Collection (ATCC, USA). For cell culture, H1299 or A549 cells were grown in RPMI-1640 Medium (R8758, Sigma-Aldrich, USA) with supplement of 10% fetal bovine serum (10099141C, Thermo Fisher, USA) at 37°C in a humidified incubator (5% CO2, 95% air). For cell treatment, H1299 or A549 cells were incubated with GDNT at 0, 3.125, 6.25, 12.5, 25 or 50 μM, or with MMF at 0, 0.05, 0.5, 5, or 50 μg/ml in the culture medium for 24 h [15, 27].
Bioinformatics Analysis
UALCAN database (http://ualcan.path.uab.edu/) was applied to analyze differential expressions of IMPDH1 or IMPDH2 in LUAD based on 59 normal samples and 515 primary tumor samples [28]. Swisstargetprediction database (http://www.swisstargetprediction.ch/) was employed to analyze putative targets for GDNT [29].
Construction of Short Hairpin RNA (shRNA)
To knockdown CES2 in H1299 or A549 cells, shRNA targeting CES2 (shCES2; senses 5’-GGTCTCCAATTCTAGTTTA-3’, antisense: 5’-TAAACTAGAATTGGAGACC-3’) and its shRNA negative control (shNC) were synthesized by GenePharma (China). After culture, lung cancer cells were transferred into 6-well plates, and incubated overnight to reach 50-70% confluence. Then, cell transfection was performed with Escort IV Transfection Reagent (L3287, Sigma-Aldrich, USA) according to the manufacturer’s protocol. The cells transfected with shCES2 or shNC were harvested 48 h post transfection, and then subjected to examination of transfection efficiency using quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR).
RNA Isolation and qRT-PCR
Total RNA from lung cancer cells after cell transfection was isolated using RNeasy Mini Kit (74104, Qiagen, Germany). After quantification, complementary DNA (cDNA) was reversely transcribed from total RNA using RevertAid First Strand cDNA Synthesis Kit (K1622, Thermo Fisher, USA). The samples were mixed with Universal SYBR Green Supermix (1725120, Bio-Rad, USA), and then 7900HT Fast Real-Time PCR System (Applied Biosystems, USA) was used to conduct qRT-PCR. Primer sequences used in this reaction are as follows (5’-3’): CES2 (forward: CTAGGTCCGCTGCGATTTG, reverse: TGAGGTCCTGTAGACACATGG) and GAPDH (forward: GGAGCGAGATCCCTCCAAAAT, reverse: GGCTGTTGTCATACTTCTCATGG). The relative mRNA expression level of CES2 was calculated using 2-ΔΔCt method [30], with GAPDH serving as the internal control.
Methylthiazolyldiphenyl-Tetrazolium Bromide (MTT) Assay
Lung cancer cells with or without CES2 knockdown were seeded in 96-well plates (5 × 103 cells/well) and incubated overnight. Next, the cells were treated with GDNT or MMF at different concentrations, or co-treated with 6.25 μM GDNT and 5 μg/ml MMF at 37°C for 24 h. Thereafter, 20 μl MTT solution (G4101-1, Servicebio, China) was added into each well for further 4-h incubation. Following formazan dissolution by DMSO, MD SpectraMax M5 microplate reader (Molecular Devices, USA) was utilized to detect cell absorbance at 570 nm.
Assessment of Cell Apoptosis and Cycle
The determination of cell apoptosis was completed using Annexin V-FITC Apoptosis Detection Kit (CA1020, Solarbio, China). After cell transfection and/or cell treatment, lung cancer cells were suspended with 1×Binding Buffer at the concentration of 5 × 106 cells/ml and later incubated with 5 μl Annexin V-FITC solution at room temperature (RT) for 5 min in the dark. Subsequently, cells were cultured with 5 μl propidium iodide (PI) and 400 μl phosphate buffered saline (PBS; G4202, Servicebio, China), and finally CytoFLEX SRT flow cytometer (Beckman Coulter, USA) was employed to analyze the apoptotic cells.
PI staining was carried out to evaluate cell cycle. According the manual of Cell Cycle and Apoptosis Analysis Kit (C1052, Beyotime, China), 2 × 105 cells were suspended with pre-cooled PBS and fixed with 70% ethanol (G2350, Solarbio, China) at 4°C for 12 h. After 5 min of centrifugation (1,000 ×
Animals and Ethics Statement
Male immunodeficient nude mice (nu/nu, 6 weeks old) were reared in a controlled environment (26-28°C, 40-60% humidity, 12-h on/off light cycles) and allowed to access food and drinking water
Xenograft and Drug Administration
For performing in vivo xenograft, H1299 cells (5 × 106) were suspended with 0.2 ml RPMI-1640 Medium, and injected subcutaneously into the right flank of each mouse. After 7 days of injection, mice were randomly divided into four groups (
Immunohistochemistry (IHC)
Tumor samples were fixed with 10% formalin (E672001, Sangon Biotech, China) and embedded into paraffin according to standard procedures. Through microtomy, 5 μm sections were obtained, exposed to 3% hydrogen peroxide (H299581, Aladdin, China) for 20 min, washed with PBS and treated with 0.01 M citric acid (abs44109576, Absin, China). Subsequently, the sections were treated with 5% non-fat milk (abs9175, Absin, China) in 1×TBS with Tween-20 (TBST; G0004, Servicebio, China) at RT for 30 min. Then, the sections were incubated with CES2 antibody (PA5-102415, Thermo Fisher) at 4°C overnight, followed by hybridization with horseradish peroxidase (HRP)-conjugated mouse anti-rabbit IgG (D110065, Sangon Biotech, China) at RT for 1 h. After treatment with DAB (D12384, Sigma-Aldrich) and counterstaining with hematoxylin (G1004, Servicebio, China) in sequence, AXio Lab.A1 microscope (×100 magnification, Zeiss, ermany) was used to observe positive signals of CES2 in the sections.
Western Blot
Cells or fresh tumor samples were homogenized in RIPA Buffer (R0010, China) as per the manufacturer’s specification to extract total protein which then was quantified by BCA Protein Assay Kit (PC0020, China). Equal amounts of protein samples (15 μg/lane) were separated using 10% SDS-PAGE and loaded onto polyvinylidene difluoride membranes (0.45 μm, 88585, Thermo Fisher), followed by blocking with TBST-diluted 5% non-fat milk (GC310001, Servicebio, China). Primary antibodies against CES2 (ab184957, 62 kDa), IMPDH1 (ab33039, 55 kDa), IMPDH2 (ab129165, 56 kDa) and GAPDH (ab181603, 36 kDa) were incubated with the membranes at 4°C overnight. Next, the membranes were hybridized with HRP-conjugated secondary antibody (ab6721) at RT for 1 h. All antibodies used in Western blot were provided by Abcam (UK). After that, the stripped membranes were treated with ECL Western Blotting Detection Kit (SW2040, Solarbio, China), and immunoblots were detected by ChemiDoc XRS+ imaging system (Bio-Rad). Bandscan software (Bio-Rad) was exploited to analyze relative protein expression (represented as the ratio of the gray-scale value of the target band to that of the GAPDH band).
Statistical Analysis
Data from all experiments performed thrice were expressed as mean ± standard deviation. Comparisons between two groups or among multiple groups were carried out using independent-samples
-
Figure 3. The expression of IMPDH1/2 in LUAD and the target prediction of GDNT.
(A, B) Expressions of IMPDH1 and IMPDH2 in LUAD based on 59 normal samples and 515 primary tumor samples were analyzed using UALCAN database (http://ualcan.path.uab.edu/). (C) Swisstargetprediction database (http://www.swisstargetprediction.ch/) was employed to analyze putative targets for GDNT.
Results
GDNT Enhanced the Effects of MMF on Suppressing Viability, Promoting Apoptosis and Inducing Cell Cycle Arrest in Lung Cancer Cells
Previous evidence has shown that GDNT (Fig. 1A), a lanostanoid triterpene extracted from
-
Figure 1. The effects of GDNT and/or MMF on viability and apoptosis of lung cancer cells.
(A) The chemical structure of GDNT. (B-E) H1299 or A549 cells were treated with GDNT (0, 3.125, 6.25, 12.5, 25, 50 μM) or MMF (0, 0.05, 0.5, 5, 50 μg/ml) for 24 h, and MTT assay was performed to examine cell viability. (F, G) H1299 or A549 cells were treated with 6.25 μM GDNT and/or 5 μg/ml MMF for 24 h, and MTT assay was conducted to test cell viability. (H-J) After treatment with 6.25 μM GDNT and/or 5 μg/ml MMF for 24 h, flow cytometry was used to analyze cell apoptosis. The horizontal axis in the H graph represented the number of FITC stained cells, while the vertical axis represented the number of PI stained cells. Data from all experiments performed thrice were expressed as mean ± standard deviation. *
p < 0.05, **p < 0.01, ***p < 0.001, vs. 0; ^p < 0.05, ^^p < 0.01, ^^^p < 0.001, vs. Control; θθθp < 0.001, vs. MMF; ###p < 0.001, vs. GDNT. MMF, mycophenolate mofetil; GDNT, ganodermanontriol; MTT, methylthiazolyldiphenyl-tetrazolium bromide.
-
Figure 2. The effect of GDNT and/or MMF on lung cancer cell cycle.
(A) H1299 or A549 cells were treated with 6.25 μM GDNT and/or 5 μg/ml MMF for 24 h, and flow cytometry was applied to analyze cell cycle. (B, C) Bar chart of cell cycle proportion in the Control, GDNT, MMF and MMF+GDNT groups. Data from all experiments performed thrice were expressed as mean ± standard deviation. ^^^
p < 0.001, vs. Control; θθθp < 0.001, vs. MMF; ###p < 0.001, vs. GDNT.
IMPDH1 and IMPDH2 Were Highly Expressed in LUAD Samples and CES2 Was a Potential Target for GDNT
Based on TCGA, UALCAN database predicted that IMPDH1 expression was higher in primary tumors from LUAD samples than in normal samples (Fig. 3A,
GDNT Up-Regulated CES2 Expression and Enhanced the Effect of MMF on Down-Regulating IMPDH1 and IMPDH2 Expressions in Lung Cancer Cells
CES2 has been indicated to influence MMF metabolism, given the genetic polymorphism of this metabolic enzyme [22]. In H1299 or A549 cells, it was determined that the protein level of CES2 was up-regulated by GDNT treatment (Fig. 4A-4C,
-
Figure 4. The effect of GDNT and/or MMF on CES2, IMPDH1 and IMPDH2 expressions in lung cancer cells.
(A-C) H1299 or A549 cells were treated with 6.25 μM GDNT and/or 5 μg/ml MMF for 24 h, and Western blot was performed to detect protein level of CES2 in the Control, GDNT, MMF and MMF+GDNT groups. (D-F) After the cell treatment for 24 h, the protein expression levels of IMPDH1 and IMPDH2 in H1299 cells were measured by Western blot. (G-I) Following the cell treatment for 24 h, IMPDH1 and IMPDH2 protein expressions in A549 cells were determined by Western blot. Relative expression levels were normalized with GAPDH. Data from all experiments performed thrice were expressed as mean ± standard deviation. ^^
p < 0.01, ^^^p < 0.001, vs. Control; θp < 0.05, θθp < 0.01, θθθp < 0.001, vs. MMF; ###p < 0.001, vs. GDNT. CES2, carboxylesterase 2; IMPDH, inosine-5’-monophosphate dehydrogenase.
CES2 Knockdown Reversed the Synergistic Effect of GDNT and MMF against Lung Cancer In Vitro
Next, we explored the effect of CES2 on the development of lung cancer cells in the presence of MMF and GDNT. After transfection with shCES2, the mRNA and protein expressions of CES2 in either H1299 or A549 cells were strikingly decreased (Fig. 5A-5E,
-
Figure 5. CES2 knockdown reversed the synergistic effect of GDNT and MMF on viability and apoptosis of lung cancer cells.
(A, B) H1299 or A549 cells were transfected with shCES2 or shNC for 48 h, and qRT-PCR was performed to examine the mRNA expression of CES2, with GAPDH used as the internal control. (C-E) The protein expression of CES2 in the Control, shNC and shCES2 groups was determined by Western blot, with GAPDH used as the internal control. (F, G) After cell transfection and co-treatment with 6.25 μM GDNT and/or 5 μg/ml MMF, the viability of H1299 and A549 cells was evaluated by MTT assay. (H-J) The apoptosis of the indicated cells was tested by flow cytometry. Data from all experiments performed thrice were expressed as mean ± standard deviation. +++
p < 0.001, vs. shNC; θθθp < 0.001, vs. MMF; ΔΔΔp < 0.001, vs. MMF + GDNT + shNC. shCES2, short hairpin RNA (shRNA) targeting CES2; shNC, shRNA negative control.
-
Figure 6. CES2 knockdown offset the synergistic effect of GDNT and MMF on cell cycle and IMPDH1/2 expression in lung cancer cells.
(A-C) After cell transfection with shCES2 and co-treatment with 6.25 μM GDNT and/or 5 μg/ml MMF, the cell cycle of H1299 and A549 cells was analyzed by flow cytometry. (D-I) Protein levels of IMPDH1 and IMPDH2 in the indicated cells were measured by Western blot. Relative expression levels were normalized with GAPDH. Data from all experiments performed thrice were expressed as mean ± standard deviation. θθ
p < 0.01, θθθp < 0.001, vs. MMF; ΔΔp < 0.01, ΔΔΔp < 0.001, vs. MMF + GDNT + shNC. shCES2, short hairpin RNA (shRNA) targeting CES2; shNC, shRNA negative control.
GDNT Potentiated the Tumor-Suppressive Effect of MMF on Lung Cancer In Vivo
Considering the prominent role of GDNT in suppressing the development of lung cancer cells by increasing apoptosis, the regulatory effect of GDNT cooperated with MMF on tumorigenesis in lung cancer should be explored. Thus, we performed xenograft experiment through subcutaneously injecting H1299 cells into nude mice, followed by administration with GDNT and/or MMF. 45 days later, it was observed that either GDNT or MMF markedly reduced the volume and weight of tumors excised from the mice (Fig. 7A-7C,
-
Figure 7. GDNT enhanced the effect of MMF on suppressing tumor growth of lung cancer in vivo.
(A) Xenograft experiment was performed by subcutaneously injecting H1299 cells into mice, followed by treatment of MMF (20 mg/kg/2d) and/or GDNT (3 mg/kg/1d). After 45 days, all tumor-bearing mice were sacrificed, and the tumors were excised and photographed. (B) The volume of tumors was calculated. (C) The weight of tumors was measured. Data from all experiments performed thrice were expressed as mean ± standard deviation. ^^
p < 0.01, ^^^p < 0.001, vs. Control; θθθp < 0.001, vs. MMF; ###p < 0.001, vs. GDNT.
-
Figure 8. GDNT strengthened the effect of MMF on down-regulating CES2 and IMPDH1/2 expressions in lung cancer in vivo.
(A, B) After xenograft experiment, immunohistochemistry was carried out to detect the expression of CES2 in the excised tumors. (C-E) Protein levels of IMPDH1 and IMPDH2 in the excised tumors were determined by Western blot. Relative expression levels were normalized with GAPDH. Data from all experiments performed thrice were expressed as mean ± standard deviation. ^^^
p < 0.001, vs. Control; θp < 0.05, θθp < 0.01, vs. MMF; ###p < 0.001, vs. GDNT.
Discussion
Currently, radiotherapy and first-line antineoplastic agents remain the basic treatments for most patients with advanced NSCLC, but the overall survival rate is affected by factors including side effects, non-targeted cytotoxicity and drug resistance [33]. With the rise of integrative therapies for malignant tumors,
Ample evidence at home and abroad has supported that
As an immunosuppressant, MMF, which can be hydrolyzed into MPA in the body, is widely used in the clinical treatment after solid organ transplantation to prevent rejection reaction through inhibiting IMPDH [41], but has been recently evidenced to repress the tumor progression via targeting tumor-associated fibroblast [31]. Although the anti-proliferative role of MMF has been confirmed in lung cancer, its underlying mechanism in LUAD is not fully expounded. Of note, this study unveiled that MMF treatment alone boosted apoptosis of LUAD cells and blocked intercellular DNA replication by augmenting the percentage of S phase, consistent with the findings of Ling
Based on the above-mentioned findings, we subsequently investigated the possible molecular mechanism of GDNT enhancing the anti-cancer effect of MMF. Through bioinformatics analysis, we found that CES2 was predicted as a potential target for GDNT. Reportedly, CES2 impacts the clinical results of cancer patients by affecting chemosensitivity [43]. In the study of graft rejection, CES2 has been suggested to function as a mediator in the metabolism of MMF to MPA [22]. In lung cancer cells, the expression of CES2 was increased by GDNT with/without MMF but was not significantly affected in the presence of MMF alone. MPA can target IMPDH which has been confirmed to be highly expressed in many tumors [44, 45]. In this study, the results of Western blot revealed that both IMPDH1 and IMPDH2 expressions were down-regulated by MMF in the cells, which was further strengthened by GDNT. IMPDH1 and IMPDH2 are two isoforms of IMPDH that is implicated in cell metabolism and proliferation by modulating
Conclusion
Despite the importance of IMPDH in anti-cancer treatment, the development and application of IMPDH inhibitors are limited by their side effects at high doses [25]. The present study unravels the anti-cancer mechanism of GDNT in LUAD, and further provides a new direction for developing adjuvant agents to enhance the efficacy of IMPDH inhibitors in treating lung cancer.
Supplemental Materials
Acknowledgments
This work was supported by the Zhejiang Provincial Medical and Health Project [2023587787]; the Zhejiang Provincial Science and Technology Department Project [LGF22H010012]; the Longquan Science and Technology Bureau Project [2021KJ-003]; the Zhejiang Provincial Medical and Health Research Project [2021KY1235]; the Lishui Science and Technology Bureau Project [2020077571].
Author Contributions
Substantial contributions to conception and design: Qingfeng Xie and Zhuo Cao
Data acquisition, data analysis and interpretation: Weiling You, Xiaoping Cai, Mei Shen, Zhangyong Yin, Yiwei Jiang, Xin Wang, Siyu Ye
Drafting the article or critically revising it for important intellectual content: Qingfeng Xie, Zhuo Cao, Weiling You and Xiaoping Cai, Mei Shen, Zhangyong Yin, Yiwei Jiang, Xin Wang, Siyu Ye
Final approval of the version to be published: All authors
Agreement to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of the work are appropriately investigated and resolved: Qingfeng Xie, Zhuo cao, Weiling You, Xiaoping Cai, Mei Shen, Zhangyong Yin, Yiwei Jiang, Xin Wang, Siyu Ye
Conflict of Interest
The authors have no financial conflicts of interest to declare.
Fig 1.
Fig 2.
Fig 3.
Fig 4.
Fig 5.
Fig 6.
Fig 7.
Fig 8.
References
- Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A,
et al . 2021. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries.CA: Cancer J. Clin. 71 : 209-249. - Herbst RS, Morgensztern D, Boshoff C. 2018. The biology and management of non-small cell lung cancer.
Nature 553 : 446-454. - Luo C, Lei M, Zhang Y, Zhang Q, Li L, Lian J,
et al . 2020. Systematic construction and validation of an immune prognostic model for lung adenocarcinoma.J. Cell. Mol. Med. 24 : 1233-1244. - Nooreldeen R, Bach H. 2021. Current and future development in lung cancer diagnosis.
Int. J. Mol. Sci. 22 : 8661. - Kadara H, Scheet P, Wistuba II, Spira AE. 2016. Early events in the molecular pathogenesis of lung cancer.
Cancer Prev. Res. 9 : 518-527. - Duma N, Santana-Davila R, Molina JR. 2019. Non-small cell lung cancer: epidemiology, screening, diagnosis, and treatment.
Mayo Clin. Proc. 94 : 1623-1640. - Soccol CR, Bissoqui LY, Rodrigues C, Rubel R, Sella SR, Leifa F,
et al . 2016. Pharmacological properties of biocompounds from spores of the lingzhi or reishi medicinal mushroomGanoderma lucidum (Agaricomycetes ): a review.Int. J. Med. Mushrooms 18 : 757-767. - Chan SW, Tomlinson B, Chan P, Lam CWK. 2021. The beneficial effects of
Ganoderma lucidum on cardiovascular and metabolic disease risk.Pharm. Biol. 59 : 1161-1171. - Sohretoglu D, Huang S. 2018.
Ganoderma lucidum polysaccharides as an anti-cancer agent.Anticancer Agents Med. Chem. 18 : 667-674. - Shao Y, Qiao L, Wu L, Sun X, Zhu D, Yang G,
et al . 2016. Structure identification and anti-cancer pharmacological prediction of triterpenes fromGanoderma lucidum .Molecules 21 : 678. - Barbieri A, Quagliariello V, Del Vecchio V, Falco M, Luciano A, Amruthraj NJ,
et al . 2017. Anticancer and anti-inflammatory properties ofGanoderma lucidum extract effects on melanoma and triple-negative breast cancer treatment.Nutrients 9 : 210. - Liu W, Yuan R, Hou A, Tan S, Liu X, Tan P,
et al . 2020.Ganoderma triterpenoids attenuate tumour angiogenesis in lung cancer tumour-bearing nude mice.Pharm. Biol. 58 : 1061-1068. - Zhu J, Xu J, Jiang LL, Huang JQ, Yan JY, Chen YW,
et al . 2019. Improved antitumor activity of cisplatin combined withGanoderma lucidum polysaccharides in U14 cervical carcinoma-bearing mice.Kaohsiung J. Med. Sci. 35 : 222-229. - Li X, Xie Y, Yang BB. 2018. Characterizing novel anti-oncogenic triterpenoids from ganoderma.
Cell Cycle 17 : 527-528. - Jiang J, Jedinak A, Sliva D. 2011. Ganodermanontriol (GDNT) exerts its effect on growth and invasiveness of breast cancer cells through the down-regulation of CDC20 and uPA.
Biochem. Biophys. Res. Commun. 415 : 325-329. - Jedinak A, Thyagarajan-Sahu A, Jiang J, Sliva D. 2011. Ganodermanontriol, a lanostanoid triterpene from
Ganoderma lucidum , suppresses growth of colon cancer cells through ß-catenin signaling.Int. J. Oncol. 38 : 761-767. - Xu C, Guo H, Kong D, Pang D, Ding Y. 2018. Ganodermanontriol inhibits expression of special AT rich sequence binding protein 1 gene in human hepatocellular carcinoma.
J. Cancer Res. Ther. 14 : S964-s968. - Zou LW, Jin Q, Wang DD, Qian QK, Hao DC, Ge GB,
et al . 2018. Carboxylesterase inhibitors: an update.Curr. Med. Chem. 25 : 1627-1649. - Chen F, Zhang B, Parker RB, Laizure SC. 2018. Clinical implications of genetic variation in carboxylesterase drug metabolism.
Exp. Opin. Drug Metab. Txicol. 14 : 131-142. - Lam SW, van der Noort V, van der Straaten T, Honkoop AH, Peters GJ, Guchelaar HJ,
et al . 2018. Single-nucleotide polymorphisms in the genes of CES2, CDA and enzymatic activity of CDA for prediction of the efficacy of capecitabine-containing chemotherapy in patients with metastatic breast cancer.Pharm. Res. 128 : 122-129. - Zhang Y, Sun L, Sun Y, Chen Y, Wang X, Xu M,
et al . 2020. Overexpressed CES2 has prognostic value in CRC and knockdown CES2 reverses L-OHP-resistance in CRC cells by inhibition of the PI3K signaling pathway.Exp. Cell Res. 389 : 111856. - Cilião HL, Camargo-Godoy RBO, Souza MF, Zanuto A, Delfino VDA, Cólus IMS. 2018. Polymorphisms in IMPDH2, UGT2B7, and CES2 genes influence the risk of graft rejection in kidney transplant recipients taking mycophenolate mofetil.
Mut. Res. Genet. Toxicol. Environ. Mutagen. 836 : 97-102. - Parlakpinar H, Gunata M. 2021. Transplantation and immunosuppression: a review of novel transplant-related immunosuppressant drugs.
Immunopharmacol. Immunotoxicol. 43 : 651-665. - Chen X, Zhao Y, Wang D, Lin Y, Hou J, Xu X,
et al . 2021. The HNF4α-BC200-FMR1-positive feedback loop promotes growth and metastasis in invasive mucinous lung adenocarcinoma.Cancer Res. 81 : 5904-5918. - Naffouje R, Grover P, Yu H, Sendilnathan A, Wolfe K, Majd N,
et al . 2019. Anti-tumor potential of IMP dehydrogenase inhibitors: a century-long story.Cancers 11 : 1346. - Keppeke GD, Andrade LEC, Barcelos D, Fernandes M, Landman G. 2020. IMPDH-based cytoophidium structures as potential theranostics in cancer.
Mol. Ther. 28 : 1557-1558. - Stracke S, Ramudo L, Keller F, Henne-Bruns D, Mayer JM. 2006. Antiproliferative and overadditive effects of everolimus and mycophenolate mofetil in pancreas and lung cancer cells in vitro.
Transplant. Proc. 38 : 766-770. - Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi B,
et al . 2017. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses.Neoplasia 19 : 649-658. - Daina A, Michielin O, Zoete V. 2019. SwissTargetPrediction: updated data and new features for efficient prediction of protein targets of small molecules.
Nucleic Acids Res. 47 : W357-w364. - Livak KJ, Schmittgen TD. 2001. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method.
Methods 25 : 402-408. - Yang Z, Zhang L, Zhu H, Zhou K, Wang H, Wang Y,
et al . 2021. Nanoparticle formulation of mycophenolate mofetil achieves enhanced efficacy against hepatocellular carcinoma by targeting tumour-associated fibroblast.J. Cell. Mol. Med. 25 : 3511-3523. - Kennedy EM, P'Pool SJ, Jiang J, Sliva D, Minto RE. 2011. Semisynthesis and biological evaluation of ganodermanontriol and its stereoisomeric triols.
J. Nat. Prod. 74 : 2332-2337. - Wang M, Herbst RS, Boshoff C. 2021. Toward personalized treatment approaches for non-small-cell lung cancer.
Nat. Med. 27 : 1345-1356. - Guo J, Yuan C, Huang M, Liu Y, Chen Y, Liu C,
et al . 2018.Ganoderma lucidum -derived polysaccharide enhances coix oil-based microemulsion on stability and lung cancer-targeted therapy.Drug Deliv. 25 : 1802-1810. - Feng L, Yuan L, Du M, Chen Y, Zhang MH, Gu JF,
et al . 2013. Anti-lung cancer activity through enhancement of immunomodulation and induction of cell apoptosis of total triterpenes extracted fromGanoderma luncidum (Leyss. ex Fr.) Karst.Molecules (Basel, Switzerland) 18 : 9966-9981. - Zolj S, Smith MP, Goines JC, Ali TS, Huff MO, Robinson DL,
et al . 2018. Antiproliferative effects of a rriterpene-enriched extract from lingzhi or reishi medicinal mushroom,Ganoderma lucidum (Agaricomycetes), on human lung cancer cells.Int. J. Med. Mushrooms 20 : 1173-1183. - Wang T, Xie ZP, Huang ZS, Li H, Wei AY, Di JM,
et al . 2015. Total triterpenoids fromGanoderma Lucidum suppresses prostate cancer cell growth by inducing growth arrest and apoptosis.J. Huazhong Univ. Sci. Technol. Med. Sci. 35 : 736-741. - Wang H, Liu Z, Wu P, Wang H, Ren W. 2021. NUSAP1 accelerates osteosarcoma cell proliferation and cell cycle progression via upregulating CDC20 and cyclin A2.
OncoTargets Ther. 14 : 3443-3454. - Chu Z, Zhang X, Li Q, Hu G, Lian CG, Geng S. 2019. CDC20 contributes to the development of human cutaneous squamous cell carcinoma through the Wnt/β-catenin signaling pathway.
Int. J. Oncol. 54 : 1534-1544. - Hu H, Tou FF, Mao WM, Xu YL, Jin H, Kuang YK,
et al . 2022. microRNA-1321 and microRNA-7515 contribute to the progression of non-small cell lung cancer by targeting CDC20.Kaohsiung J. Med. Sci. 38 : 425-436. - Plunk MA, Quintana JM, Darden CM, Lawrence MC, Naziruddin B, Kane RR. 2021. Design and catalyzed activation of mycophenolic acid prodrugs.
ACS Med. Chem. Lett. 12 : 812-816. - Ling G, Lamprecht S, Shubinsky G, Osyntsov L, Yerushalmi B, Pinsk I,
et al . 2018. Mycophenolate mofetil alone and in combination with tacrolimus inhibits the proliferation of HT-29 human colonic adenocarcinoma cell line and might interfere with colonic tumorigenesis.Anticancer Res. 38 : 3333-3339. - Silvestris N, Simone G, Partipilo G, Scarpi E, Lorusso V, Brunetti AE,
et al . 2014. CES2, ABCG2, TS and Topo-I primary and synchronous metastasis expression and clinical outcome in metastatic colorectal cancer patients treated with first-line FOLFIRI regimen.Int. J. Mol. Sci. 15 : 15767-15777. - Winnicki W, Fichtenbaum A, Mitulovič G, Herkner H, Regele F, Baier M,
et al . 2022. Individualization of mycophenolic acid therapy through pharmacogenetic, pharmacokinetic and pharmacodynamic testing.Biomedicines 10 : 2882. - Siebert A, Deptuła M, Cichorek M, Ronowska A, Cholewiński G, Rachon J. 2021. Anticancer properties of amino acid and peptide derivatives of mycophenolic acid.
Antcancer Agents Med. Chem. 21 : 462-467. - Keppeke GD, Chang CC, Peng M, Chen LY, Lin WC, Pai LM,
et al . 2018. IMP/GTP balance modulates cytoophidium assembly and IMPDH activity.Cell Div. 13 : 5. - Valvezan AJ, McNamara MC, Miller SK, Torrence ME, Asara JM, Henske EP,
et al . 2020. IMPDH inhibitors for antitumor therapy in tuberous sclerosis complex.JCI Insight 5 : e135071. - Huang F, Ni M, Chalishazar MD, Huffman KE, Kim J, Cai L,
et al . 2018. Inosine monophosphate dehydrogenase dependence in a subset of small cell lung cancers.Cell Metab. 28 : 369-382.e365. - Ruan H, Song Z, Cao Q, Ni D, Xu T, Wang K,
et al . 2020. IMPDH1/YB-1 positive feedback loop assembles cytoophidia and represents a therapeutic target in metastatic tumors.Mole. Ther. 28 : 1299-1313.