1
|
Chuang YT, Yen CY, Tang JY, Wu KC, Chang FR, Tsai YH, Chien TM, Chang HW. Marine anticancer drugs in modulating miRNAs and antioxidant signaling. Chem Biol Interact 2024; 399:111142. [PMID: 39019423 DOI: 10.1016/j.cbi.2024.111142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
Several marine drugs exert anticancer effects by inducing oxidative stress, which becomes overloaded and kills cancer cells when redox homeostasis is imbalanced. The downregulation of antioxidant signaling induces oxidative stress, while its upregulation attenuates oxidative stress. Marine drugs have miRNA-modulating effects against cancer cells. However, the potential antioxidant targets of such drugs have been rarely explored. This review aims to categorize the marine-drug-modulated miRNAs that downregulate their antioxidant targets, causing oxidative stress in anticancer treatments. We also categorize the downregulation of oxidative-stress-inducing miRNAs in antioxidant protection among non-cancer cells. We summarize the putative antioxidant targets of miRNA-modulating marine drugs by introducing a bioinformatics tool (miRDB). Finally, the marine drugs affecting antioxidant targets are surveyed. In this way, the connections between marine drugs and their modulating miRNA and antioxidant targets are innovatively categorized to provide a precise network for exploring their potential anticancer functions and protective effects on non-cancer cells.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei, 11031, Taiwan; Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan, 71004, Taiwan.
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Kuo-Chuan Wu
- Department of Computer Science and Information Engineering, National Pingtung University, Pingtung, 900392, Taiwan.
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Yi-Hong Tsai
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung, 907101, Taiwan.
| | - Tsu-Ming Chien
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan; Department of Urology, Kaohsiung Gangshan Hospital, Kaohsiung Medical University, Kaohsiung 820111, Taiwan.
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
2
|
Liew YX, Karen-Ng LP, Vincent-Chong VK. A Comprehensive Review of Natural Products as Therapeutic or Chemopreventive Agents against Head and Neck Squamous Cell Carcinoma Cells Using Preclinical Models. Biomedicines 2023; 11:2359. [PMID: 37760799 PMCID: PMC10525836 DOI: 10.3390/biomedicines11092359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/09/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a type of cancer that arises from the epithelium lining of the oral cavity, hypopharynx, oropharynx, and larynx. Despite the advancement of current treatments, including surgery, chemotherapy, and radiotherapy, the overall survival rate of patients afflicted with HNSCC remains poor. The reasons for these poor outcomes are due to late diagnoses and patient-acquired resistance to treatment. Natural products have been extensively explored as a safer and more acceptable alternative therapy to the current treatments, with numerous studies displaying their potential against HNSCC. This review highlights preclinical studies in the past 5 years involving natural products against HNSCC and explores the signaling pathways altered by these products. This review also addresses challenges and future directions of natural products as chemotherapeutic and chemoprevention agents against HNSCC.
Collapse
Affiliation(s)
- Yoon Xuan Liew
- Oral Cancer Research & Coordinating Centre (OCRCC), Faculty of Dentistry, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Lee Peng Karen-Ng
- Oral Cancer Research & Coordinating Centre (OCRCC), Faculty of Dentistry, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Vui King Vincent-Chong
- Department of Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
3
|
Afrasiabi M, Tahmasebi G, Eslami E, Seydi E, Pourahmad J. Cold Atmospheric Plasma Versus Cisplatin Against Oral Squamous Cell Carcinoma: A Mitochondrial Targeting Study. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2022; 21:e124106. [PMID: 36942058 PMCID: PMC10024331 DOI: 10.5812/ijpr-124106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/08/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022]
Abstract
Plasma therapy and the study of the effects of cold atmospheric plasma (CAP) on tissues and living cells have been considered by scientific researchers in recent years. CAP is used in the treatment of cancer, but its anti-cancer mechanism has not been fully studied. Therefore, we studied the toxicity effect of CAP by using argon as feed gas and the synergistic effects of CAP with cisplatin on tumor cells and mitochondria isolated from tumor legions of the rat model of oral squamous cell carcinoma (OSCC). For this reason, we determined the possible toxic alterations of CAP on mitochondrial upstream events and activation of caspase-3 as the key major downstream event of apoptosis. Also, the effects of cisplatin (10 µM) as a positive control and its synergistic effects with CAP (IC50 concentration) were investigated. The results showed that CAP reduced mitochondrial dysfunction by reduction in succinate dehydrogenase (SDH) activity. Also, CAP in concentrations of 1200, 2400, and 4800 a.u. has been able to increase the level of reactive oxygen species (ROS), mitochondrial swelling, damage to the mitochondrial membrane, cytochrome c release, and activation of the final mediator of apoptosis (caspase-3) only in the OSCC group. CAP at 4800 a.u concentration had similar effects to cisplatin (10 µM). Synergistic effects between CAP (2400 a.u) and cisplatin (10 µM) have also been reported. Based on all results CAP showed positive and promising results on mitochondrial upstream parameters leading to activation of caspase-3, the final mediator of apoptosis only on OSCC cells and mitochondria without any significant effect on normal cells and mitochondria.
Collapse
Affiliation(s)
- Mona Afrasiabi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Tahmasebi
- Department of Atomic/Molecular Physics, Faculty of Physics, Iran University of Science and Technology, Tehran, Iran
| | - Esmaeil Eslami
- Department of Atomic/Molecular Physics, Faculty of Physics, Iran University of Science and Technology, Tehran, Iran
| | - Enayatollah Seydi
- Department of Occupational Health and Safety Engineering, School of Health, Alborz University of Medical Sciences, Karaj, Iran
- Research Center for Health, Safety and Environment, Alborz University of Medical Sciences, Karaj, Iran
- Corresponding Author: Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Jalal Pourahmad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Corresponding Author: Department of Occupational Health and Safety Engineering, School of Health, Alborz University of Medical Sciences, Karaj, Iran; Research Center for Health, Safety and Environment, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
4
|
Naeem A, Hu P, Yang M, Zhang J, Liu Y, Zhu W, Zheng Q. Natural Products as Anticancer Agents: Current Status and Future Perspectives. Molecules 2022; 27:molecules27238367. [PMID: 36500466 PMCID: PMC9737905 DOI: 10.3390/molecules27238367] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Natural products have been an invaluable and useful source of anticancer agents over the years. Several compounds have been synthesized from natural products by modifying their structures or by using naturally occurring compounds as building blocks in the synthesis of these compounds for various purposes in different fields, such as biology, medicine, and engineering. Multiple modern and costly treatments have been applied to combat cancer and limit its lethality, but the results are not significantly refreshing. Natural products, which are a significant source of new therapeutic drugs, are currently being investigated as potential cytotoxic agents and have shown a positive trend in preclinical research and have prompted numerous innovative strategies in order to combat cancer and expedite the clinical research. Natural products are becoming increasingly important for drug discovery due to their high molecular diversity and novel biofunctionality. Furthermore, natural products can provide superior efficacy and safety due to their unique molecular properties. The objective of the current review is to provide an overview of the emergence of natural products for the treatment and prevention of cancer, such as chemosensitizers, immunotherapeutics, combinatorial therapies with other anticancer drugs, novel formulations of natural products, and the molecular mechanisms underlying their anticancer properties.
Collapse
Affiliation(s)
- Abid Naeem
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Pengyi Hu
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Ming Yang
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jing Zhang
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yali Liu
- Key Laboratory of Pharmacodynamics and Safety Evaluation, Health Commission of Jiangxi Province, Nanchang Medical College, Nanchang 330006, China
- Key Laboratory of Pharmacodynamics and Quality Evaluation on Anti-Inflammatory Chinese Herbs, Jiangxi Administration of Traditional Chinese Medicine, Nanchang Medical College, Nanchang 330006, China
| | - Weifeng Zhu
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Qin Zheng
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
- Correspondence:
| |
Collapse
|
5
|
Aengenvoort J, Sekeres M, Proksch P, Fritz G. Targeting Mechanisms of the DNA Damage Response (DDR) and DNA Repair by Natural Compounds to Improve cAT-Triggered Tumor Cell Death. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113567. [PMID: 35684504 PMCID: PMC9182506 DOI: 10.3390/molecules27113567] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/11/2022] [Accepted: 05/26/2022] [Indexed: 12/20/2022]
Abstract
Recently, we identified secalonic acid F (SA), 5-epi-nakijiquinone Q (NQ) and 5-epi-ilimaquinone (IQ) as natural compounds (NC) affecting mechanisms of the DNA damage response (DDR). Here, we further characterized their effects on DDR, DNA repair and cytotoxicity if used in mono- and co-treatment with conventional anticancer therapeutics (cAT) (cisplatin (Cis), doxorubicin (Doxo)) in vitro. All three NC influence the phosphorylation level of selected DDR-related factors (i.e., pCHK1, pKAP1, pP53, pRPA32) in mono- and/or co-treatment. Both SA and NQ attenuate the Cis- and Doxo-induced G2/M-phase arrest and effectively stimulate caspase-mediated apoptosis. Notably, SA impacts DNA repair as reflected by enhanced steady-state levels of Cis-(1,2-GpG)-DNA adducts and Doxo-induced DNA double-strand breaks (DSB). Moreover, SA decreased the mRNA and protein expression of the homologous recombination (HR)-related DSB repair factors RAD51 and BRCA1. Both SA and NQ promote Cis- and Doxo-induced cytotoxicity in an additive to synergistic manner (CI ≤ 1.0). Summarizing, we conclude that SA promotes cAT-driven caspase-dependent cell death by interfering with DSB repair and DDR-related checkpoint control mechanisms. Hence, SA is considered as the most promising lead compound to evaluate its therapeutic window in forthcoming pre-clinical in vivo studies.
Collapse
Affiliation(s)
- Jana Aengenvoort
- Institute of Toxicology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany; (J.A.); (M.S.)
| | - Marlena Sekeres
- Institute of Toxicology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany; (J.A.); (M.S.)
| | - Peter Proksch
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich-Heine University Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany;
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany; (J.A.); (M.S.)
- Correspondence: ; Tel.: +49-211-8113022
| |
Collapse
|
6
|
Synthetic Tryptanthrin Derivatives Induce Cell Cycle Arrest and Apoptosis via Akt and MAPKs in Human Hepatocellular Carcinoma Cells. Biomedicines 2021; 9:biomedicines9111527. [PMID: 34829756 PMCID: PMC8615277 DOI: 10.3390/biomedicines9111527] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/12/2021] [Accepted: 10/20/2021] [Indexed: 12/20/2022] Open
Abstract
Trytanthrin, found in Ban-Lan-Gen, is a natural product containing an indoloquinazoline moiety and has been shown to possess anti-inflammatory and anti-viral activities. Chronic inflammation and hepatitis B are known to be associated with the progression of hepatocellular carcinoma (HCC). In this study, a series of tryptanthrin derivatives were synthesized to generate potent anti-tumor agents against HCC. This effort yielded two compounds, A1 and A6, that exhibited multi-fold higher cytotoxicity in HCC cells than the parent compound. Flow cytometric analysis demonstrated that A1 and A6 caused S-phase arrest and downregulated the expression of cyclin A1, B1, CDK2, and p-CDC2. In addition to inducing caspase-dependent apoptosis, A1 and A6 exhibited similar regulation of the phosphorylation or expression of multiple signaling targets, including Akt, NF-κB, and mitogen-activated protein kinases. The anti-tumor activities of A1 and A6 were also attributable to the generation of reactive oxygen species, accompanied by an increase in p-p53 levels. Therefore, A1 and A6 have potential clinical applications since they target diverse aspects of cancer cell growth in HCC.
Collapse
|
7
|
Natural Merosesquiterpenes Activate the DNA Damage Response via DNA Strand Break Formation and Trigger Apoptotic Cell Death in p53-Wild-type and Mutant Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13133282. [PMID: 34209047 PMCID: PMC8268692 DOI: 10.3390/cancers13133282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/19/2021] [Accepted: 06/27/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Bowel cancer is a serious disease, which affects many people worldwide. Unfortunately, the disease is often diagnosed in an advanced stage, which impairs the chance of survival. Furthermore, resistance to therapy occurs frequently. Thus, novel therapeutic approaches are required to improve cancer therapy. Here, we studied whether merosesquiterpenes might be useful for cancer treatment. These compounds occur in marine sponges and were isolated by our group. We were able to identify three compounds with potent cytotoxic activity in different cell lines established from human large bowel cancer. Our experiments provided evidence that the compounds cause DNA damage and trigger cell death, so-called mitochondrial apoptosis, which was attested in cancer cells with expression of wild-type and mutated p53 tumor suppressor. Finally, we show that merosesquiterpenes also kill intestinal tumor organoids, an ex vivo model of large bowel cancer. Abstract Colorectal cancer (CRC) is a frequently occurring malignant disease with still low survival rates, highlighting the need for novel therapeutics. Merosesquiterpenes are secondary metabolites from marine sponges, which might be useful as antitumor agents. To address this issue, we made use of a compound library comprising 11 isolated merosesquiterpenes. The most cytotoxic compounds were smenospongine > ilimaquinone ≈ dactylospontriol, as shown in different human CRC cell lines. Alkaline Comet assays and γH2AX immunofluorescence microscopy demonstrated DNA strand break formation in CRC cells. Western blot analysis revealed an activation of the DNA damage response with CHK1 phosphorylation, stabilization of p53 and p21, which occurred both in CRC cells with p53 knockout and in p53-mutated CRC cells. This resulted in cell cycle arrest followed by a strong increase in the subG1 population, indicative of apoptosis, and typical morphological alterations. In consistency, cell death measurements showed apoptosis following exposure to merosesquiterpenes. Gene expression studies and analysis of caspase cleavage revealed mitochondrial apoptosis via BAX, BIM, and caspase-9 as the main cell death pathway. Interestingly, the compounds were equally effective in p53-wild-type and p53-mutant CRC cells. Finally, the cytotoxic activity of the merosesquiterpenes was corroborated in intestinal tumor organoids, emphasizing their potential for CRC chemotherapy.
Collapse
|
8
|
Abstract
Possessing the quinone moiety, ilimaquinone (1), a sponge–derived sesquiterpene quinone, has been hypothesised to express its cytotoxicity through a redox cycling process, yielding active product(s) that can cause DNA damage. To determine the DNA damaging effects of 1 and examine whether a redox transformation may participate in its functions, the DNA damaging properties of 1, the corresponding hydroquinone (2) and hydroquinone triacetates (3) and their 5-epimeric counterparts (4–6) were tested and compared. When incubated directly with plasmid DNA, the hydroquinones were the only active species capable of cleaving the DNA. In cell-based assays, however, the quinones and hydroquinone triacetates were active in the same range as that of the corresponding hydroquinones, and all damaged the cellular DNA in a similar manner. The in situ reduction of 1 and 4 were supported by the decreases in the cytotoxicity when cells were pre-exposed to dicoumarol, an NAD(P)H:quinone oxidoreductase 1 (NQO1) inhibitor. The results confirmed the DNA damaging activities of the ilimaquinones 1 and 4, and indicated the necessity to undergo an in-situ transformation into the active hydroquinones, thereby exerting the DNA damaging properties as parts of the cytotoxic mechanisms.
Collapse
|
9
|
Antitumor Effects of a Sesquiterpene Derivative from Marine Sponge in Human Breast Cancer Cells. Mar Drugs 2021; 19:md19050244. [PMID: 33925873 PMCID: PMC8144972 DOI: 10.3390/md19050244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/27/2022] Open
Abstract
In this study, the anti-proliferative effect of ilimaquinone, a sesquiterpene derivative from the marine sponge, in breast cancer cells was investigated. Ilimaquinone inhibited the proliferation of MCF-7 and MDA-MB-231 breast cancer cells with IC50 values of 10.6 μM and 13.5 μM, respectively. Non-tumorigenic human breast epithelial cells were less sensitive to ilimaquinone than breast cancer cells. Flow cytometric and Western blot analysis showed that ilimaquinone induced S-phase arrest by modulating the expression of p-CDC-2 and p21. Ilimaquinone induces apoptosis, which is accompanied by multiple biological biomarkers, including the downregulation of Akt, ERK, and Bax, upregulation of p38, loss of mitochondrial membrane potential, increased reactive oxygen species generation, and induced autophagy. Collectively, these findings suggest that ilimaquinone causes cell cycle arrest as well as induces apoptosis and autophagy in breast cancer cells.
Collapse
|
10
|
Zhang X, Cheng J, Liu S, Li R. Down-regulating circular RNA_0004674 delays the progression of oral squamous cell carcinoma through microRNA-377-3p/THBS1 axis. Life Sci 2021:119236. [PMID: 33621591 DOI: 10.1016/j.lfs.2021.119236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 02/05/2021] [Accepted: 02/12/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Circular RNAs (CircRNAs) are of great significance in oral squamous cell carcinoma (OSCC) cell progression. Insufficiently, the performance of Circ_0004674 has not been specified in the disease, which alighted our desire to unmask its actions in OSCC cell progression with microRNA (miR)-377-3p and thrombospondin-1 (THBS1). METHODS OSCC expression chip were collected through GEO database and analyzed. The upstream mechanism of THBS1 was predicted through databases. OSCC cancer tissues and normal tissues were resected, in which Circ_0004674, miR-377-3p and THBS1 expression were examined. The relationship of Circ_0004674, miR-377-3p and THBS1 was identified. Circ_0004674- and/or miR-377-3p-related oligonucleotides were transfected into CAL27 cells for detecting cell biological behaviors. Tumors in mice were implanted to monitor the tumor-forming ability of cells. RESULTS THBS1 showed high expression in the three OSCC chips, and it was enriched in PI3K-AKT signaling pathway. The upstream mechanism of THBS1 predicted that Circ_0004674 regulated THBS1 through miR-377-3p. Circ_0004674 and THBS1 levels were enhanced while miR-377-3p level was reduced in OSCC. Down-regulating Circ_0004674 restricted the growth of CAL27 cells in vivo and in vitro. Restoring miR-377-3p, the target gene of Circ_0004674, destroyed CAL27 cell progression and tumor growth. miR-377-3p suppression rescued the effects of down-regulated Circ_0004674 on OSCC. THBS1 was negatively mediated by miR-377-3p. CONCLUSION It is clarified that depleting Circ_0004674 mediates miR-377-3p to restrain THBS1, after which OSCC cell progression can be suppressed. It widens the way to control OSCC from a novel perspective.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Junying Cheng
- Department of Magnetic resonance, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Sirui Liu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Rui Li
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
11
|
Jhou AJ, Chang HC, Hung CC, Lin HC, Lee YC, Liu WT, Han KF, Lai YW, Lin MY, Lee CH. Chlorpromazine, an antipsychotic agent, induces G2/M phase arrest and apoptosis via regulation of the PI3K/AKT/mTOR-mediated autophagy pathways in human oral cancer. Biochem Pharmacol 2021; 184:114403. [PMID: 33388284 DOI: 10.1016/j.bcp.2020.114403] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/26/2020] [Accepted: 12/29/2020] [Indexed: 02/08/2023]
Abstract
Chlorpromazine (CPZ), an FDA-approved phenothiazine derivative used to treat schizophrenia and other psychiatric disorders, has been demonstrated to have potential anti-tumor effects. However, the potential effects of CPZ on human oral cancer cells and the underlying molecular mechanisms remain unknown. In this study, treatment of human oral cancer cells with CPZ inhibited their proliferation and induced G2/M phase arrest. Treatment with CPZ induced apoptosis through the extrinsic death receptor and the intrinsic mitochondrial pathways. In addition, the induction of autophagy was observed by the formation of autophagosomes, the expression of autophagy-related proteins and activation of the PI3K/Akt/mTOR/p70S6K pathway. The CPZ-induced cell death was reversed by the pan-caspase inhibitor Z-VAD-FMK, by the autophagy inhibitor 3-MA and by the knockdown of LC3B using a shRNA (shLC3B), suggesting that autophagy promoted CPZ-induced apoptosis. Finally, CPZ significantly suppressed tumor growth in both a zebrafish oral cancer xenotransplantation model and in a murine model of 4-nitroquinoline-1-oxide (4NQO)-induced oral cancer. Overall, this evidence demonstrated that CPZ is a novel promising strategy for the treatment of oral cancer.
Collapse
Affiliation(s)
- An-Jie Jhou
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hao-Chiun Chang
- Department of Orthopaedics, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Chih-Chang Hung
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Han-Chen Lin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Department of Medical Research, Kaohsiung Medical University Hospital, Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yi-Chen Lee
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Department of Medical Research, Kaohsiung Medical University Hospital, Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wang-Ta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80708 Taiwan
| | - Kuang-Fen Han
- Department of Nursing, Min-Hwei Junior College of Health Care Management, Tainan 73658, Taiwan
| | - Yu-Wei Lai
- Division of Urology, Taipei City Hospital Renai Branch, Department of Urology, National Yang-Ming University School of Medicine, Taipei 11221, Taiwan
| | - Mei-Ying Lin
- Community Health Promotion Center, Kaohsiung Municipal Ci-Jin Hospital, Kaohsiung 80708, Taiwan
| | - Chien-Hsing Lee
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Pharmacology, School of Medicine; School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
| |
Collapse
|