1
|
Chuerduangphui J, Ekalaksananan T, Heawchaiyaphum C, Vatanasapt P, Teeramatwanich W, Phusingha P, Pientong C. Zinc-alpha-2-glycoprotein overexpression and maintaining anti-apoptotic function in oral squamous cell carcinoma. Arch Oral Biol 2025; 176:106298. [PMID: 40398100 DOI: 10.1016/j.archoralbio.2025.106298] [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: 01/26/2025] [Revised: 05/08/2025] [Accepted: 05/14/2025] [Indexed: 05/23/2025]
Abstract
OBJECTIVE Overexpression of zinc-alpha-2-glycoprotein (ZAG) can be induced by various factors and has potential to be a biomarker in certain malignancies. However, in oral squamous cell carcinoma (OSCC), the risks and effects associated with ZAG overexpression are still poorly known. Here, we investigated the effect of HPV16 oncogenes and arecoline on the expression levels of ZAG and the possible effects of ZAG in OSCC cell lines. DESIGN The level of ZAG expression was determined in protein extracted from exfoliated buccal cells from cancer-free control individuals and oral lesion cells from OSCC. Oral cell lines expressing HPV16E6/E7, and treated with arecoline were prepared to investigate ZAG expression. The effects of ZAG on cell biological activity and its targeting of UCP1 were determined in ZAG-overexpressing and ZAG-knockdown cells. RESULTS The expression of ZAG protein was significantly increased in oral lesion cells from OSCC relative to controls. Notably, the expression level of ZAG in OSCC positive for HPV, betel-quid chewing, and combination of both factors, was slightly higher than in cancer-free controls. ZAG expression was upregulated in oral cells treated with HPV16 oncoproteins E6 and/or E7, and treatment with arecoline (25 μg/ml). Interestingly, ZAG overexpression significantly increased UCP1 and decreased apoptosis, whereas decreased UCP1 and increased apoptosis were found in ZAG-knockdown cells. The mRNA expression levels of TP53, STAT3, BCL2, and NFKB1 corresponded to observed anti-apoptosis function. CONCLUSIONS HPV oncoproteins and high doses of arecoline are risk factors for an overexpressed ZAG protein that has an anti-apoptotic function in OSCC.
Collapse
Affiliation(s)
- Jureeporn Chuerduangphui
- Department of Microbiology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand; HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Tipaya Ekalaksananan
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chukkris Heawchaiyaphum
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Patravoot Vatanasapt
- Department of Otorhinolaryngology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Watchareporn Teeramatwanich
- Department of Otorhinolaryngology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Pensiri Phusingha
- National Institute of Health, Department of Medical Sciences, Tiwanond Road, Nonthaburi 11000, Thailand
| | - Chamsai Pientong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand.
| |
Collapse
|
2
|
Wu FC, Ting CC, Jeng JH, Chen H, Kim YD, Wu CC, Kao YH, Tseng CH, Chen YK, Ogasawara T, Hoshi K, Lo WL, Takahashi T, Yang YH, Ko EC. Hypoxia amplifies arecoline-induced invasion and metastasis in oral squamous cell carcinoma - Insights into TGF-β1 signaling and collagen production. J Dent Sci 2025; 20:1129-1138. [PMID: 40224083 PMCID: PMC11993037 DOI: 10.1016/j.jds.2024.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/25/2024] [Indexed: 04/15/2025] Open
Abstract
Background/Purpose Betel quid chewing is a major risk factor for oral squamous cell carcinoma (OSCC), largely due to arecoline, a key alkaloid. Hypoxia, common in the tumor microenvironment, also influences cancer progression. This study investigated the combined effects of arecoline and hypoxia on proliferation, migration, and protein expression in tongue squamous cell carcinoma (SCC-25) cells, focusing on the TGF-β1 signaling pathway and type I collagen production. Materials and methods SCC-25 cells were treated with arecoline and incubated for 24 h under normoxia or hypoxia. Cytotoxicity assays and Western blotting were performed to assess cell viability and protein expression. Results At 2.5 μg/mL, arecoline enhanced SCC-25 cell proliferation under normoxia, while hypoxia suppressed this effect. Arecoline significantly promoted cell migration that was further amplified by hypoxia. Western blotting revealed that arecoline upregulated TGF-β1, Smad2/3, phosphorylated Smad2/3, and type I collagen. Under hypoxia, HIF1-α expression increased along with TGF-β1 and type I collagen, indicating that hypoxia enhances arecoline-induced collagen production through TGF-β1 signaling. Conclusion Arecoline stimulates SCC-25 cell proliferation and migration, with hypoxia amplifying these effects by promoting TGF-β1 signaling and type I collagen production. These findings suggest that betel quid consumption, in combination with hypoxia, may exacerbate the invasion and metastasis of OSCC.
Collapse
Affiliation(s)
- Feng-Cheng Wu
- Liberty Lab of Tissue Engineering Takao, Kaohsiung, Taiwan
- Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Chan Ting
- Liberty Lab of Tissue Engineering Takao, Kaohsiung, Taiwan
- School of Dentistry, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jiiang-Huei Jeng
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hangshen Chen
- Liberty Lab of Tissue Engineering Takao, Kaohsiung, Taiwan
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yong-Deok Kim
- Department of Oral and Maxillofacial Surgery School of Dentistry and Dental Research Institute, and Institute of Translational Dental Sciences, Pusan National University, Yangsan, South Korea
| | - Chia-Chen Wu
- Liberty Lab of Tissue Engineering Takao, Kaohsiung, Taiwan
| | - Yu-Hsun Kao
- Liberty Lab of Tissue Engineering Takao, Kaohsiung, Taiwan
- Division of Oral and Maxillofacial Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chih-Huang Tseng
- Liberty Lab of Tissue Engineering Takao, Kaohsiung, Taiwan
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuk-Kwan Chen
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Toru Ogasawara
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuto Hoshi
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Wen-liang Lo
- College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tetsu Takahashi
- Oral and Maxillofacial Surgery, Southern Tohoku Fukushima Hospital, Koriyama, Fukushima, Japan
| | - Yi-Hsin Yang
- National Institute of Cancer Research, Tainan, Taiwan
| | - Edward Chengchuan Ko
- Liberty Lab of Tissue Engineering Takao, Kaohsiung, Taiwan
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Oral and Maxillofacial Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- Institute of Precise Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
3
|
Bai LY, Dokla EME, Chu PC, Feng CH, Hu JL, Wang LJ, Weng JR. A synthetic molecule targeting STAT3 against human oral squamous cell carcinoma cells. Int J Med Sci 2025; 22:1081-1091. [PMID: 40027184 PMCID: PMC11866527 DOI: 10.7150/ijms.105200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/17/2025] [Indexed: 03/05/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC), one of the most common cancers in Taiwan, needs new therapeutic agents and treatments. The aim of this study was to investigate the anti-proliferative activity of {N-[3-chloro-4-[5-[3-[[[4-[(cyclopropylcarbonyl)-amino]3-(trifluoromethyl)phenylamino]carbonyl]amino]phenyl]-1,2,4-oxadiazol-3-yl]phenyl]-3-pyridine-carboxamide} (COC), a synthetic molecule, in OSCC cells. COC exhibits potent tumor-suppressive efficacy with IC50 values of 195 nM and 204 nM toward SCC2095 and SCC4 OSCC cells, respectively. Our data revealed that COC caused caspase-dependent apoptosis and downregulated the MAPK signaling pathway. In addition, COC modulated the levels of E-cadherin and β-catenin and inhibited migration. COC also decreased p-STAT3 levels, and the overexpression of STAT3 partially attenuated COC-induced cytotoxicity. Therefore, our findings suggest the use of COC as a new approach to oral cancer treatment.
Collapse
Affiliation(s)
- Li-Yuan Bai
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
- College of Medicine, China Medical University, Taichung 404, Taiwan
| | - Eman M. E. Dokla
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo 115, Egypt
| | - Po-Chen Chu
- Department of Cosmeceutics and Graduate Institute of Cosmeceutics, China Medical University, Taichung 404, Taiwan
| | - Chia-Hsien Feng
- Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Jing-Lan Hu
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Liang-Jun Wang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Jing-Ru Weng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
4
|
Huang HH, Chang JT, You GR, Fu YF, Shen EYL, Huang YF, Shen CR, Cheng AJ. MiRNA Profiling of Areca Nut-Induced Carcinogenesis in Head and Neck Cancer. Cancers (Basel) 2024; 16:3710. [PMID: 39518147 PMCID: PMC11545612 DOI: 10.3390/cancers16213710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND While miRNAs are increasingly recognized for their role in tumorigenesis, their involvement in head and neck cancer (HNC) remains insufficiently explored. Additionally, the carcinogenic mechanisms of areca nut, a major habitual carcinogen in Southeast Asia, are not well understood. METHODS AND RESULTS This study adopts a systematic approach to identify miRNA profiles associated with areca nut-induced HNC. Using miRNA microarray analysis, we identified 292 miRNAs dysregulated in areca nut-treated HNC cells, with 136 upregulated and 156 downregulated. Bioinformatic analysis of the TCGA-HNSC dataset uncovered a set of 692 miRNAs relevant to HNC development, comprising 449 overexpressed and 243 underexpressed in tumor tissues. Integrating these datasets, we defined a signature of 84 miRNAs, including 39 oncogenic miRNAs (OncomiRs) and 45 tumor-suppressive miRNAs (TsmiRs), highlighting their pivotal role in areca nut-induced carcinogenesis. MultiMiR analysis identified 740 genes cross-regulated by eight hub TsmiRs, significantly impacting key cancer-related pathways (p53, PI3K-AKT, MAPK, and Ras) and critical oncogenic processes. Moreover, we validated miR-499a-5p as a vital regulator, demonstrating its ability to mitigate areca nut-induced cancer progression by reducing cell migration, invasion, and chemoresistance. CONCLUSIONS Thus, this miRNA signature addresses a crucial gap in understanding the molecular underpinnings of areca nut-induced carcinogenesis and offers a promising platform for clinical applications in risk assessment, diagnosis, and prognosis of areca nut-associated malignancies.
Collapse
Affiliation(s)
- Hung-Han Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (H.-H.H.); (C.-R.S.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (G.-R.Y.); (Y.-F.F.)
| | - Joseph T. Chang
- Department of Radiation Oncology and Proton Therapy Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan; (J.T.C.); (E.Y.-L.S.)
- School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Guo-Rung You
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (G.-R.Y.); (Y.-F.F.)
| | - Yu-Fang Fu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (G.-R.Y.); (Y.-F.F.)
| | - Eric Yi-Liang Shen
- Department of Radiation Oncology and Proton Therapy Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan; (J.T.C.); (E.Y.-L.S.)
| | - Yi-Fang Huang
- Department of General Dentistry, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan;
- Graduate Institute of Dental and Craniofacial Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chia-Rui Shen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (H.-H.H.); (C.-R.S.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (G.-R.Y.); (Y.-F.F.)
| | - Ann-Joy Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (H.-H.H.); (C.-R.S.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (G.-R.Y.); (Y.-F.F.)
- Department of Radiation Oncology and Proton Therapy Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan; (J.T.C.); (E.Y.-L.S.)
| |
Collapse
|
5
|
Kizhakkoottu S, Ramani P, Tilakaratne WM. Role of Stem Cells in the Pathogenesis and Malignant Transformation of Oral Submucous Fibrosis. Stem Cell Rev Rep 2024; 20:1512-1520. [PMID: 38837114 DOI: 10.1007/s12015-024-10744-0] [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] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Pathogenesis and malignant potential of Oral submucous fibrosis(OSMF) have always been a topic of interest among the researchers. Despite OSMF being a collagen metabolic disorder, the alterations occurring in the connective tissue stroma affects the atrophic surface epithelium in later stages and progresses to malignant phenotypes. The present review aims to summarize the role of stem cells in the pathogenesis and malignant transformation of oral submucous fibrosis. MATERIALS AND METHODS A literature search was carried out using data banks like Medline and Embase, google scholar and manual method with no time frame, pertinent to the role of mucosal stem cells in OSMF and its malignisation. The relevant literature was reviewed, critically appraised by all the authors and compiled in this narrative review. RESULTS Critical appraisal and evaluation of the data extracted from the selected articles were compiled in this review. The collated results highlighted the upregulation and downregulation of various stem cell markers during the progression and malignisation of OSMF were depicted in a descriptive and detail manner in the present review. CONCLUSION We highlight the potential of mucosal stem cells in the regulation and malignisation of OSMF. However, future large-scale clinical studies will be needed to support whether manipulation of this stem cells at molecular level will be sufficient for the treatment and preventing the malignant transformation of OSMF.
Collapse
Affiliation(s)
- Suvarna Kizhakkoottu
- Department of Oral Pathology and Microbiology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai, India
| | - Pratibha Ramani
- Department of Oral Pathology and Microbiology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai, India.
| | | |
Collapse
|
6
|
Lu MY, Hsieh PL, Chao SC, Fang CY, Ohiro Y, Liao YW, Yu CC, Chang MT. Targeting MetaLnc9/miR-143/FSCN1 axis inhibits oxidative stress and myofibroblast transdifferentiation in oral submucous fibrosis. J Dent Sci 2024; 19:1416-1425. [PMID: 39035266 PMCID: PMC11259661 DOI: 10.1016/j.jds.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/10/2024] [Indexed: 07/23/2024] Open
Abstract
Background/purpose Persistent activation of myofibroblasts is attributed to various dysregulated biological events conferring multiple types of fibrosis diseases, including oral submucous fibrosis (OSF). Although the significance of non-coding RNAs (ncRNAs) in the occurrence of fibrosis has been appreciated, the detailed mechanisms still have not been fully elucidated. The aim of this study was to identify key dysregulated ncRNAs and elucidate their pro-fibrotic mechanisms in promoting myofibroblast activation and the pathological development of OSF. Materials and methods Expression of non-coding RNAs and mRNAs in OSF cohort was determined using RNA sequencing and qRT-PCR. The molecular axis of pro-fibrotic ncRNAs were exploited via luciferase reporter activity assay and RNA expression rescue experiments. Functional assays, including collagen gel contraction, wound healing ability, cell migration, and reactive oxygen species (ROS) production, were conducted to assess the changes in the myofibroblastic phenotypes of primary human buccal mucosal fibroblasts. Results Herein, we found that long non-coding RNA MetaLnc9 was upregulated in OSF specimens and positively associated with several fibrosis markers. Silencing of MetaLnc9 diminished the features of activated myofibroblasts and the production of ROS. We not only showed that MetaLnc9 functioned as a competitive endogenous RNA of microRNA (miR)-143, but also demonstrated that the pro-fibrosis effect of MetaLnc9 on myofibroblast activities was mediated by suppression of miR-143. Moreover, our data showed that fascin actin-bundling protein 1 (FSCN1) was a direct target of miR-143 and positively related to MetaLnc9. Conclusion Upregulation of MetaLnc9 may enhance the activation of myofibroblasts by sponging miR-143 and titrating its inhibitory property on FSCN1.
Collapse
Affiliation(s)
- Ming-Yi Lu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Pei-Ling Hsieh
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Shih-Chi Chao
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chih-Yuan Fang
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Wan Fang Hospital, Taipei, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yoichi Ohiro
- Oral and Maxillofacial Surgery, Division of Oral Pathobiological Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Yi-Wen Liao
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Cheng-Chia Yu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Min-Te Chang
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan, Taiwan
| |
Collapse
|
7
|
Hao P, Zhang P, Liu Y, Cao Y, Du L, Gao L, Dong Q. Network pharmacology and experiment validation investigate the potential mechanism of triptolide in oral squamous cell carcinoma. Front Pharmacol 2024; 14:1302059. [PMID: 38259290 PMCID: PMC10800448 DOI: 10.3389/fphar.2023.1302059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Objective: This study aimed to investigate the molecular mechanism of triptolide in the treatment of oral squamous cell carcinoma (OSCC) via network pharmacology and experimental validation. Methods: The network pharmacological method was used to predict the key targets, detect the signal pathways for the treatment of OSCC, and screen the critical components and targets for molecular docking. Predicted targets were validated in cellular and xenograft mouse model. Results: In this study, we predicted action on 17 relevant targets of OSCC by network pharmacology. PPI network demonstrated that Jun, MAPK8, TP53, STAT3, VEGFA, IL2, CXCR4, PTGS2, IL4 might be the critical targets of triptolide in the treatment of OSCC. These potential targets are mainly closely related to JAK-STAT and MAPK signaling pathways. The analysis of molecular docking showed that triptolide has high affinity with Jun, MAPK8 and TP53. Triptolide can suppress the growth of OSCC cells and xenograft mice tumor, and downregulate the expression of Jun, MAPK8, TP53, STAT3, VEGFA, IL2, CXCR4, PTGS2 to achieve the therapeutic effect of OSCC. Conclusion: Through network pharmacological methods and experimental studies, we predicted and validated the potential targets and related pathways of triptolide for OSCC treatment. The results suggest that triptolide can inhibit the growth of OSCC via several key targets.
Collapse
Affiliation(s)
- Puyu Hao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
- Environmental and Operational Medicine Research Department, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Pengcheng Zhang
- Environmental and Operational Medicine Research Department, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Ying Liu
- Environmental and Operational Medicine Research Department, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Yang Cao
- Environmental and Operational Medicine Research Department, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Lianqun Du
- Environmental and Operational Medicine Research Department, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Li Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
| | - Qingyang Dong
- Environmental and Operational Medicine Research Department, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| |
Collapse
|
8
|
Su YF, Lin CS, Shen PC, Chuang SE, Dai YH, Huang TW, Lin CY, Hung YJ, Shieh YS. MiR-34a functions as a tumor suppressor in oral cancer through the inhibition of the Axl/Akt/GSK-3β pathway. J Dent Sci 2024; 19:428-437. [PMID: 38303867 PMCID: PMC10829669 DOI: 10.1016/j.jds.2023.08.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/14/2023] [Indexed: 02/03/2024] Open
Abstract
Background/purpose Oral cancer is a prevalent malignancy affecting men globally. This study aimed to investigate the regulatory role of miR-34a in oral cancer cells through the Axl/Akt/glycogen synthase kinase-3β (GSK-3β) pathway and its impact on cellular malignancy. Materials and methods We examined the effects of miR-34a overexpression on the malignancy of oral cancer cells. Multiple oral cancer cell lines were assessed to determine the correlation between endogenous miR-34a and Axl levels. Transfection experiments with miR-34a were conducted to analyze its influence on Axl mRNA and protein expression. Luciferase reporter assays were performed to investigate miR-34a's modulation of Axl gene transcription. Manipulation of miR-34a expression was utilized to demonstrate its regulatory effects on oral cancer cells through the Axl/Akt/GSK-3β pathway. Results Overexpression of miR-34a significantly suppressed the malignancy of oral cancer cells. We observed an inverse correlation between endogenous miR-34a and Axl levels across multiple oral cancer cell lines. Transfection of miR-34a resulted in decreased Axl mRNA and protein expression, and luciferase reporter assays confirmed miR-34a-mediated modulation of Axl gene transcription. The study revealed regulatory effects of miR-34a on oral cancer cells through the Axl/Akt/GSK-3β pathway, leading to alterations in downstream target genes involved in cellular proliferation and tumorigenesis. Conclusion Our findings highlight the significance of the miR-34a/Axl/Akt/GSK-3β signaling axis in modulating the malignancy of oral cancer cells. Targeting miR-34a may hold therapeutic potential in oral cancer treatment, as manipulating its expression can attenuate the aggressive behavior of oral cancer cells via the Axl/Akt/GSK-3β pathway.
Collapse
Affiliation(s)
- Yu-Fu Su
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Shu Lin
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Po-Chien Shen
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shuang-En Chuang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yang-Hong Dai
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tsai-Wang Huang
- Division of Thoracic Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Che-Yi Lin
- Department of Oral and Maxillofacial Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Yi-Jen Hung
- Division of Endocrinology and Metabolism, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Shing Shieh
- Department of Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department and Graduate Institute of Biochemistry, National Defense Medical Center, Taipei, Taiwan
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
9
|
Mishra SR, Behera BP, Singh VK, Mahapatra KK, Mundkinajeddu D, Bhat D, Minz AM, Sethi G, Efferth T, Das S, Bhutia SK. Anticancer activity of Bacopa monnieri through apoptosis induction and mitophagy-dependent NLRP3 inflammasome inhibition in oral squamous cell carcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155157. [PMID: 37951147 DOI: 10.1016/j.phymed.2023.155157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 11/13/2023]
Abstract
BACKGROUND Bacopa monnieri (BM) is traditionally used in human diseases for its antioxidant, anti-inflammatory and neuroprotective effects. However, its anticancer potential has been poorly understood. AIM The aim of this study was to explore the detailed anticancer mechanism of BM against oral cancer and to identify the bioactive BM fraction for possible cancer therapeutics. RESULTS We performed bioactivity-guided fractionation and identified that the aqueous fraction of the ethanolic extract of BM (BM-AF) had a potent anticancer potential in both in vitro and in vivo oral cancer models. BM-AF inhibited cell viability, colony formation, cell migration and induced apoptotic cell death in Cal33 and FaDu cells. BM-AF at low doses promoted mitophagy and BM-AF mediated mitophagy was PARKIN dependent. In addition, BM-AF inhibited arecoline induced reactive oxygen species production in Cal33 cells. Moreover, BM-AF supressed arecoline-induced NLR family pyrin domain containing 3 (NLRP3) inflammasome activation through mitophagy in Cal33 cells. The in vivo antitumor effect of BM-AF was further validated in C57BL/6J mice through a 4-nitroquinolin-1-oxide and arecoline-induced oral cancer model. The tumor incidence was significantly reduced in the BM-AF treated group. Further, data obtained from western blot and immunohistochemistry analysis showed increased expression of apoptotic markers and decreased expression of inflammasome markers in the tongue tissue obtained from BM-AF treated mice in comparison with the non-treated tumor bearing mice. CONCLUSION In conclusion, BM-AF exhibited potent anticancer activity through apoptosis induction and mitophagy-dependent inhibition of NLRP3 inflammasome activation in both in vitro and in vivo oral cancer models. Moreover, we have investigated apoptosis and mitophagy-inducing compounds from this plant extract having anticancer activity against oral cancer cells.
Collapse
Affiliation(s)
- Soumya Ranjan Mishra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Bishnu Prasad Behera
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | | | - Kewal Kumar Mahapatra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India; Current affiliation: Department of Agriculture and Allied Sciences (Zoology), C. V Raman Global University, Bhubaneswar, 752054, Odisha, India
| | | | - Deeksha Bhat
- Research and Development Department, Natural Remedies Pvt. Ltd, India
| | - Aruna Mukti Minz
- Department of Pathology, Ispat General Hospital, Rourkela, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Surajit Das
- Laboratory of Environmental Microbiology and Ecology, Department of Life Science, National Institute of Technology Rourkela, 769008, Odisha, India
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| |
Collapse
|
10
|
Gil-Martín E, Ramos E, López-Muñoz F, Egea J, Romero A. Potential of melatonin to reverse epigenetic aberrations in oral cancer: new findings. EXCLI JOURNAL 2023; 22:1280-1310. [PMID: 38234969 PMCID: PMC10792176 DOI: 10.17179/excli2023-6624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/22/2023] [Indexed: 01/19/2024]
Abstract
It is now an accepted principle that epigenetic alterations cause cellular dyshomeostasis and functional changes, both of which are essential for the initiation and completion of the tumor cycle. Oral carcinogenesis is no exception in this regard, as most of the tumors in the different subsites of the oral cavity arise from the cross-reaction between (epi)genetic inheritance and the huge challenge of environmental stressors. Currently, the biochemical machinery is put at the service of the tumor program, halting the cell cycle, triggering uncontrolled proliferation, driving angiogenesis and resistance to apoptosis, until the archetypes of the tumor phenotype are reached. Melatonin has the ability to dynamically affect the epigenetic code. It has become accepted that melatonin can reverse (epi)genetic aberrations present in oral and other cancers, suggesting the possibility of enhancing the oncostatic capacity of standard multimodal treatments by incorporating this indolamine as an adjuvant. First steps in this direction confirm the potential of melatonin as a countermeasure to mitigate the detrimental side effects of conventional first-line radiochemotherapy. This single effect could produce synergies of extraordinary clinical importance, allowing doses to be increased and treatments not to be interrupted, ultimately improving patients' quality of life and prognosis. Motivated by the urgency of improving the medical management of oral cancer, many authors advocate moving from in vitro and preclinical research, where the bulk of melatonin cancer research is concentrated, to systematic randomized clinical trials on large cohorts. Recognizing the challenge to improve the clinical management of cancer, our motivation is to encourage comprehensive and robust research to reveal the clinical potential of melatonin in oral cancer control. To improve the outcome and quality of life of patients with oral cancer, here we provide the latest evidence of the oncolytic activity that melatonin can achieve by manipulating epigenetic patterns in oronasopharyngeal tissue.
Collapse
Affiliation(s)
- Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, University of Vigo, 36310 Vigo, Spain
| | - Eva Ramos
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Francisco López-Muñoz
- Faculty of Health, Camilo José Cela University of Madrid (UCJC), 28692 Madrid, Spain
- Neuropsychopharmacology Unit, Hospital 12 de Octubre Research Institute, 28041 Madrid, Spain
| | - Javier Egea
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006 Madrid, Spain
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
11
|
Gocol H, Zeng JH, Chang S, Koh BY, Nguyen H, Cirillo N. A Critical Interpretive Synthesis of the Role of Arecoline in Oral Carcinogenesis: Is the Local Cholinergic Axis a Missing Link in Disease Pathophysiology? Pharmaceuticals (Basel) 2023; 16:1684. [PMID: 38139811 PMCID: PMC10748297 DOI: 10.3390/ph16121684] [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: 10/10/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Arecoline is the primary active carcinogen found in areca nut and has been implicated in the pathogenesis of oral squamous cell carcinoma (OSCC) and oral submucous fibrosis (OSF). For this study, we conducted a stepwise review process by combining iterative scoping reviews with a post hoc search, with the aim of identifying the specific mechanisms by which arecoline initiates and promotes oral carcinogenesis. Our initial search allowed us to define the current trends and patterns in the pathophysiology of arecoline-induced OSF and OSCC, which include the induction of cell proliferation, facilitation of invasion, adhesion, and migration, increased collagen deposition and fibrosis, imbalance in immune and inflammatory mechanisms, and genotoxicity. Key molecular pathways comprise the activation of NOTCH1, MYC, PRDX2, WNT, CYR61, EGFR/Pl3K, DDR1 signaling, and cytokine upregulation. Despite providing a comprehensive overview of potential pathogenic mechanisms of OSF, the involvement of molecules functioning as areca alkaloid receptors, namely, the muscarinic and nicotinic acetylcholine receptors (AChRs), was not elucidated with this approach. Accordingly, our search strategy was refined to reflect these evidence gaps. The results of the second round of reviews with the post hoc search highlighted that arecoline binds preferentially to muscarinic AChRs, which have been implicated in cancer. Consistently, AChRs activate the signaling pathways that partially overlap with those described in the context of arecoline-induced carcinogenesis. In summary, we used a theory-driven interpretive review methodology to inform, extend, and supplement the conventional systematic literature assessment workflow. On the one hand, the results of this critical interpretive synthesis highlighted the prevailing trends and enabled the consolidation of data pertaining to the molecular mechanisms involved in arecoline-induced carcinogenesis, and, on the other, brought up knowledge gaps related to the role of the local cholinergic axis in oral carcinogenesis, thus suggesting areas for further investigation.
Collapse
Affiliation(s)
| | | | | | | | | | - Nicola Cirillo
- Melbourne Dental School, The University of Melbourne, Carlton, VIC 3053, Australia (B.Y.K.)
| |
Collapse
|
12
|
Wolf CL, Pruett C, Lighter D, Jorcyk CL. The clinical relevance of OSM in inflammatory diseases: a comprehensive review. Front Immunol 2023; 14:1239732. [PMID: 37841259 PMCID: PMC10570509 DOI: 10.3389/fimmu.2023.1239732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/30/2023] [Indexed: 10/17/2023] Open
Abstract
Oncostatin M (OSM) is a pleiotropic cytokine involved in a variety of inflammatory responses such as wound healing, liver regeneration, and bone remodeling. As a member of the interleukin-6 (IL-6) family of cytokines, OSM binds the shared receptor gp130, recruits either OSMRβ or LIFRβ, and activates a variety of signaling pathways including the JAK/STAT, MAPK, JNK, and PI3K/AKT pathways. Since its discovery in 1986, OSM has been identified as a significant contributor to a multitude of inflammatory diseases, including arthritis, inflammatory bowel disease, lung and skin disease, cardiovascular disease, and most recently, COVID-19. Additionally, OSM has also been extensively studied in the context of several cancer types including breast, cervical, ovarian, testicular, colon and gastrointestinal, brain,lung, skin, as well as other cancers. While OSM has been recognized as a significant contributor for each of these diseases, and studies have shown OSM inhibition is effective at treating or reducing symptoms, very few therapeutics have succeeded into clinical trials, and none have yet been approved by the FDA for treatment. In this review, we outline the role OSM plays in a variety of inflammatory diseases, including cancer, and outline the previous and current strategies for developing an inhibitor for OSM signaling.
Collapse
Affiliation(s)
- Cody L. Wolf
- Department of Biomolecular Sciences, Boise State University, Boise, ID, United States
| | - Clyde Pruett
- Department of Biological Sciences, Boise State University, Boise, ID, United States
| | - Darren Lighter
- Department of Biological Sciences, Boise State University, Boise, ID, United States
| | - Cheryl L. Jorcyk
- Department of Biomolecular Sciences, Boise State University, Boise, ID, United States
- Department of Biological Sciences, Boise State University, Boise, ID, United States
| |
Collapse
|
13
|
Ko AMS, Tu HP, Ko YC. Systematic Review of Roles of Arecoline and Arecoline N-Oxide in Oral Cancer and Strategies to Block Carcinogenesis. Cells 2023; 12:1208. [PMID: 37190117 PMCID: PMC10137008 DOI: 10.3390/cells12081208] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/11/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Betel quid and areca nut are complex mixture carcinogens, but little is known about whether their derived single-agent arecoline or arecoline N-oxide (ANO) is carcinogenic, and the underlying mechanisms remain unclear. In this systematic review, we analyzed recent studies on the roles of arecoline and ANO in cancer and strategies to block carcinogenesis. In the oral cavity, flavin-containing monooxygenase 3 oxidizes arecoline to ANO, and both alkaloids conjugate with N-acetylcysteine to form mercapturic acid compounds, which are excreted in urine, reducing arecoline and ANO toxicity. However, detoxification may not be complete. Arecoline and ANO upregulated protein expression in oral cancer tissue from areca nut users compared to expression levels in adjacent normal tissue, suggesting a causal relationship between these compounds and oral cancer. Sublingual fibrosis, hyperplasia, and oral leukoplakia were diagnosed in mice subjected to oral mucosal smearing of ANO. ANO is more cytotoxic and genotoxic than arecoline. During carcinogenesis and metastasis, these compounds increase the expression of epithelial-mesenchymal transition (EMT) inducers such as reactive oxygen species, transforming growth factor-β1, Notch receptor-1, and inflammatory cytokines, and they activate EMT-related proteins. Arecoline-induced epigenetic markers such as sirtuin-1 hypermethylation, low protein expression of miR-22, and miR-886-3-p accelerate oral cancer progression. Antioxidants and targeted inhibitors of the EMT inducers used reduce the risk of oral cancer development and progression. Our review findings substantiate the association of arecoline and ANO with oral cancer. Both of these single compounds are likely carcinogenic to humans, and their mechanisms and pathways of carcinogenesis are useful indicators for cancer therapy and prognosis.
Collapse
Affiliation(s)
- Albert Min-Shan Ko
- Department of Biomedical Sciences, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
- Cardiovascular Department, Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan
- Health Aging Research Center, Chang Gung University, Taoyuan 33302, Taiwan
| | - Hung-Pin Tu
- Department of Public Health and Environmental Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Ying-Chin Ko
- Department of Medical Research, China Medical University Hospital, China Medical University, No. 2 Yu-Der Road, Taichung 40447, Taiwan
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 106216, Taiwan
| |
Collapse
|
14
|
RNA N6-Methyladenosine (m6A) Methyltransferase-like 3 Facilitates Tumorigenesis and Cisplatin Resistance of Arecoline-Exposed Oral Carcinoma. Cells 2022; 11:cells11223605. [PMID: 36429032 PMCID: PMC9688745 DOI: 10.3390/cells11223605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/08/2022] [Accepted: 11/12/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Arecoline is known as the main active carcinogen found in areca nut extract that drives the pathological progression of oral squamous cell carcinoma (OSCC). Studies have revealed that dysregulation of RNA N6-methyladenosine (m6A) methyltransferase components is intimately linked to cancer initiation and progression, including oral cancer. METHODS The arecoline-induced dysregulated methyltransferase-like 3 (METTL3) gene was identified using RNA-seq transcriptome assay. Using in vitro and in vivo models, the biological roles of METTL3 in arecoline-transformed oral cancer were examined. RESULTS We found that METTL3 was markedly elevated in arecoline-exposed OSCC cell lines and OSCC tissues of areca nut chewers. We identified that hypoxia-inducible factor 1-alpha (HIF-1α) stimulated METTL3 expression at the transcriptional level and further proved that METTL3-MYC-HIF-1α formed a positive autoregulation loop in arecoline-transformed OSCC cells. Subsequently, we manifested that METTL3 depletion profoundly reduced cell proliferation, cell migration, oncogenicity, and cisplatin resistance of arecoline-exposed OSCC cells. CONCLUSIONS Developing novel strategies to target METTL3 may be a potential way to treat OSCC patients, particularly those with areca nut chewing history and receiving cisplatin treatment.
Collapse
|
15
|
The World of Oral Cancer and Its Risk Factors Viewed from the Aspect of MicroRNA Expression Patterns. Genes (Basel) 2022; 13:genes13040594. [PMID: 35456400 PMCID: PMC9027895 DOI: 10.3390/genes13040594] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/17/2022] [Accepted: 03/23/2022] [Indexed: 01/27/2023] Open
Abstract
Oral cancer is one of the leading causes of death worldwide, with a reported 5-year survival rate of around 50% after treatment. Epigenetic modifications are considered to have a key role in oral carcinogenesis due to histone modifications, aberrant DNA methylation, and altered expression of miRNAs. MicroRNAs (miRNAs) are small non-coding RNAs that have a key role in cancer development by regulating signaling pathways involved in carcinogenesis. MiRNA deregulation identified in oral cancer has led to the idea of using them as potential biomarkers for early diagnosis, prognosis, and the development of novel therapeutic strategies. In recent years, a key role has been observed for risk factors in preventing and treating this malignancy. The purpose of this review is to summarize the recent knowledge about the altered mechanisms of oral cancer due to risk factors and the role of miRNAs in these mechanisms.
Collapse
|
16
|
Li X, Chen W, Gao Y, Song J, Gu Y, Zhang J, Cheng X, Ai Y. FTO Regulates Arecoline-exposed Oral Cancer Immune Response through PD-L1. Cancer Sci 2022; 113:2962-2973. [PMID: 35289035 PMCID: PMC9459271 DOI: 10.1111/cas.15332] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/28/2022] [Accepted: 03/09/2022] [Indexed: 12/01/2022] Open
Abstract
The high prevalence of oral squamous cell carcinoma (OSCC) in South Asia is associated with habitual areca nut chewing. Arecoline, a primary active carcinogen within areca nut extract, is known to promote OSCC pathological development. Dysregulation of N6‐methyladenosine (m6A) modification has begun to emerge as a significant contributor to cancer development and progression. However, the biological effects and molecular mechanisms of m6A modification in arecoline‐promoted OSCC malignance remain elusive. We reveal that chronic arecoline exposure substantially induces upregulation of fat mass and obesity‐associated protein (FTO), MYC, and programmed cell death‐ligand 1 (PD‐L1) in OSCC cells. Moreover, upregulation of PD‐L1 is observed in OSCC cell lines and tissues and is associated with areca nut chewing in OSCC patients. We also demonstrate that arecoline‐induced FTO promotes the stability and expression levels of PD‐L1 transcripts through mediating m6A modification and MYC activity, respectively. PD‐L1 upregulation confers superior cell proliferation, migration, and resistance to T‐cell killing to OSCC cells. Blockage of PD‐L1 by administration of anti‐PD‐L1 antibody shrinks tumor size and improves mouse survival by elevating T‐cell‐mediated tumor cell killing. Therefore, targeting PD‐L1 might be a potential therapeutic strategy for treating PD‐L1‐positive OSCC patients, especially those with habitual areca nut chewing.
Collapse
Affiliation(s)
- Xia Li
- Department of Oral Medicine, Foshan Stomatological Hospital, Medical College of Foshan University, Foshan, Guangdong, 528000, China
| | - Wuya Chen
- Department of Oral Medicine, Foshan Stomatological Hospital, Medical College of Foshan University, Foshan, Guangdong, 528000, China
| | - Yijun Gao
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Jing Song
- Department of Oral Maxillofacial Surgery, Foshan Stomatological Hospital, Medical College of Foshan University, Foshan, Guangdong, 528000, China
| | - Yangcong Gu
- Department of Oral Maxillofacial Surgery, Foshan Stomatological Hospital, Medical College of Foshan University, Foshan, Guangdong, 528000, China
| | - Jianming Zhang
- Department of Preventive Dentistry, Foshan Stomatological Hospital, Medical College of Foshan University, Foshan, Guangdong, 528000, China
| | - Xiufeng Cheng
- Department of Oral Medicine, Foshan Stomatological Hospital, Medical College of Foshan University, Foshan, Guangdong, 528000, China
| | - Yilong Ai
- Department of Oral Medicine, Foshan Stomatological Hospital, Medical College of Foshan University, Foshan, Guangdong, 528000, China
| |
Collapse
|
17
|
Overexpression of PTPRZ1 Regulates p120/ -Catenin Phosphorylation to Promote Carcinogenesis of Oral Submucous Fibrosis. JOURNAL OF ONCOLOGY 2022; 2022:2352360. [PMID: 35251170 PMCID: PMC8890887 DOI: 10.1155/2022/2352360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/22/2021] [Accepted: 01/03/2022] [Indexed: 11/17/2022]
Abstract
Background Oral submucous fibrosis (OSF) is a potentially malignant disease of the oral cavity. New molecular predictors are needed to identify the high risk of malignant transformation in potentially malignant oral lesions. Our purpose is to explore PTPRZ1 and p120/β-catenin pathogenesis in the carcinogenesis of OSF to identify novel drug targets. Methods The expression of PTPRZ1, p120, and β-catenin in clinical tissues was detected. Then, PTPRZ1, p120, β-catenin, RhoA, Rac1, CDC42, cyclin D1, and c-myc expressions were detected by qRT-PCR and western blot. CCK-8 was applied to measure hOMF cells viability. Wound healing and transwell assay were applied to measure cell migration and invasion. Western blot and IF detected the distribution of p-p120 and p-β-catenin. Tumor formation experiment explored PTPRZ1 effects on OSF. Results PTPRZ1, p120, and β-catenin were abnormally expressed in cancer tissues. PTPRZ1 regulated the phosphorylation of p120/β-catenin. Western blot and IF showed that in the oe-NC group, p-p120 and p-β-catenin were expressed in the cell membrane. p-p120 and p-β-catenin were expressed in the cytoplasm and nucleus of the oe-PTPRZ1 group. In vitro experimental results revealed overexpression of PTPRZ1 and β-catenin, and silencing of p120 promoted cell proliferation, migration, and invasion. The tumor volume and weight in the sh-PTPRZ1 group were significantly reduced. IHC revealed the positive rate of PTPRZ1 was also low. Conclusions Overexpression of PTPRZ1 regulated the phosphorylation of p120/β-catenin to promote OSF malignancy.
Collapse
|
18
|
Li M, Jin S, Zhang Z, Ma H, Yang X. Interleukin-6 facilitates tumor progression by inducing ferroptosis resistance in head and neck squamous cell carcinoma. Cancer Lett 2021; 527:28-40. [PMID: 34902522 DOI: 10.1016/j.canlet.2021.12.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/16/2021] [Accepted: 12/08/2021] [Indexed: 12/18/2022]
Abstract
Ferroptosis resistance is an important mechanism of tumor progression. Interleukin-6 (IL-6) is a representative inflammatory cytokine during chronic inflammation; however, our current understanding of its regulatory role of ferroptosis during carcinogenesis of head and neck squamous cell carcinoma is limited. Chromatin immunoprecipitation and functional observations were performed to investigate xCT-regulatory function of IL-6. We observed a gradual increase in lipid peroxide 4-hydroxynonenal and IL-6 levels during progression from normal oral mucosa to leukoplakia and HNSCC. Meanwhile, the expression of xCT, a key amino acid antiporter assisting ferroptosis resistance, was correlated with IL-6 levels. The upregulated expression of xCT in HNSCC is associated with poor prognosis. Silencing of xCT inhibited HNSCC cell proliferation in vitro and tumor growth in vivo, inducing ferroptosis. Mechanistically, IL-6 transcriptionally activates xCT expression through the JAK2/STAT3 pathway. Furthermore, IL-6 reversed ferroptosis and growth suppression that was induced by xCT knockdown or ferroptosis inducer erastin. Our results demonstrate the critical role of IL-6-induced ferroptosis resistance during HNSCC carcinogenesis. The IL-6/STAT3/xCT axis acts as a novel mechanism driving tumor progression and thus may potentially be utilized as a target for tumor prevention and therapy.
Collapse
Affiliation(s)
- Mingyu Li
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, No. 639, Zhizaoju Rd, Shanghai, 200011, China; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| | - Shufang Jin
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, No. 639, Zhizaoju Rd, Shanghai, 200011, China; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| | - Zhiyuan Zhang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, No. 639, Zhizaoju Rd, Shanghai, 200011, China; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| | - Hailong Ma
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, No. 639, Zhizaoju Rd, Shanghai, 200011, China; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| | - Xi Yang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, No. 639, Zhizaoju Rd, Shanghai, 200011, China; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| |
Collapse
|
19
|
Porcheri C, Mitsiadis TA. New Scenarios in Pharmacological Treatments of Head and Neck Squamous Cell Carcinomas. Cancers (Basel) 2021; 13:cancers13215515. [PMID: 34771677 PMCID: PMC8583200 DOI: 10.3390/cancers13215515] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/31/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most frequent types of cancer with a lethal outcome in half of the diagnosed cases. Mostly, HNSCC develops in the oral cavity, and its development is associated with tobacco and areca nut/betel quid usage, alcohol consumption, and HPV infection. Oral squamous cell carcinoma, as other head and neck cancers, presents a high degree of intratumor heterogeneity, which makes their treatment difficult, and directly correlates with drug resistance. Since the classical treatments for HNSCC oftentimes do not resolve the clinical picture, there is great need for novel therapeutic approaches, models for drug testing, and new drug delivery systems.
Collapse
|
20
|
Li X, Xie X, Gu Y, Zhang J, Song J, Cheng X, Gao Y, Ai Y. Fat mass and obesity-associated protein regulates tumorigenesis of arecoline-promoted human oral carcinoma. Cancer Med 2021; 10:6402-6415. [PMID: 34378866 PMCID: PMC8446412 DOI: 10.1002/cam4.4188] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 11/06/2022] Open
Abstract
Arecoline, a major alkaloid within areca nut extract, is recognized as the primary active carcinogen promoting oral squamous cell carcinoma (OSCC) pathological development. Dysregulation of N6-methyladenosine (m6A) methyltransferase components (e.g., Fat mass and obesity-associated protein [FTO] and methyltransferase-like 3 [METTL3]) are closely associated with multiple cancer progression, including oral cancer. However, the biological function role of FTO in arecoline-induced oral cancer is largely unknown. We identified that FTO was significantly upregulated in OSCC tissues from patients with areca nut chewing habits and chronic arecoline-treated OSCC cell lines. Depletion of FTO attenuated the arecoline-promoted stemness, chemoresistance, and oncogenicity of OSCC cells. Finally, we revealed that FTO was negatively regulated by a transcription factor forkhead box protein A2 (FOXA2) in OSCC cells. This study, for the first time, demonstrated that FTO plays an oncogenic role in arecoline-induced OSCC progression. Thus, developing new therapeutic agents targeting FTO may serve as a promising method to treatment OSCC patients, especially those with areca nut chewing habits.
Collapse
Affiliation(s)
- Xia Li
- Department of Oral MedicineFoshan Stomatological HospitalMedical College of Foshan UniversityFoshanChina
| | - Xiaoli Xie
- Department of EndodonticsHunan Xiangya Stomatological HospitalCentral South UniversityChangshaChina
| | - Yangcong Gu
- Department of Oral Maxillofacial SurgeryFoshan Stomatological HospitalMedical College of Foshan UniversityFoshanChina
| | - Jianming Zhang
- Department of Preventive DentistryFoshan Stomatological HospitalMedical College of Foshan UniversityFoshanChina
| | - Jiang Song
- Department of Oral Maxillofacial SurgeryFoshan Stomatological HospitalMedical College of Foshan UniversityFoshanChina
| | - Xiufeng Cheng
- Department of Oral MedicineFoshan Stomatological HospitalMedical College of Foshan UniversityFoshanChina
| | - Yijun Gao
- Department of StomatologyThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Yilong Ai
- Department of Oral MedicineFoshan Stomatological HospitalMedical College of Foshan UniversityFoshanChina
| |
Collapse
|
21
|
Ghosh S, Talukdar PD, Bhattacharjee A, Giri S, Bhattacharyya NP, Chatterji U. JunD accentuates arecoline-induced disruption of tight junctions and promotes epithelial-to-mesenchymal transition by association with NEAT1 lncRNA. Oncotarget 2021; 12:1520-1539. [PMID: 34316331 PMCID: PMC8310672 DOI: 10.18632/oncotarget.28026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/11/2021] [Indexed: 12/02/2022] Open
Abstract
Head and neck cancers are highly prevalent in south-east Asia, primarily due to betel nut chewing. Arecoline, the primary alkaloid is highly carcinogenic; however its role in promoting tumorigenesis by disrupting junctional complexes and increasing risk of metastasis is not well delineated. Subsequently, the effects of low and high concentrations of arecoline on the stability of tight junctions and EMT induction were studied. A microarray analysis confirmed involvement of a MAPK component, JunD, in regulating tight junction-associated genes, specifically ZO-1. Results established that although arecoline-induced phosphorylation of JunD downregulated expression of ZO-1, JunD itself was modulated by the lncRNA-NEAT1 in presence of arecoline. Increased NEAT1 in tissues of HNSCC patients significantly correlated with poor disease prognosis. Here we show that NEAT1-JunD complex interacted with ZO-1 promoter in the nuclear compartment, downregulated expression of ZO-1 and destabilized tight junction assembly. Consequently, silencing NEAT1 in arecoline-exposed cells not only downregulated the expression of JunD and stabilized expression of ZO-1, but also reduced expression of the EMT markers, Slug and Snail, indicating its direct regulatory role in arecoline-mediated TJ disruption and disease progression.
Collapse
Affiliation(s)
- Subarna Ghosh
- Cancer Research Laboratory, Department of Zoology, University of Calcutta, Kolkata 700019, West Bengal, India
| | - Priyanka Dey Talukdar
- Cancer Research Laboratory, Department of Zoology, University of Calcutta, Kolkata 700019, West Bengal, India
| | | | - Sarbani Giri
- Department of Life Sciences, Assam University, Silchar 788011, Assam, India
| | | | - Urmi Chatterji
- Cancer Research Laboratory, Department of Zoology, University of Calcutta, Kolkata 700019, West Bengal, India.,Centre for Research in Nanoscience and Nanotechnology, University of Calcutta, Kolkata 700098, West Bengal, India
| |
Collapse
|
22
|
Zhang Y, Wang X, Han S, Wang Y, Liu R, Meng F, Su Z, Huo F. Suppression of miR-886-3p mediated by arecoline (ARE) contributes to the progression of oral squamous cell carcinoma. J Investig Med 2020; 69:377-381. [PMID: 33310761 DOI: 10.1136/jim-2020-001405] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2020] [Indexed: 02/04/2023]
Abstract
Previous studies have reported that as the main extract of areca nut, arecoline (ARE) causes DNA damage and in turn contributes to the carcinogenesis of oral epithelial cells. It has been reported that ARE can inhibit the expression of miR-886-3p. In the current study, we aimed to explore the expression and biological functions of miR-886-3p in oral squamous cell carcinoma (OSCC). Herein, we demonstrated that in OSCC cells treated with ARE, the expression level of miR-886-3p was negatively correlated with the concentration of ARE. Compared with adjacent tissue, the expression level of miR-886-3p in OSCC tissue was remarkably downregulated. Transfection of miR-886-3p mimics markedly decreased viability, migration and invasion of OSCC cells. These experimental data implied that miR-886-3p suppression mediated by ARE took part in the proliferation and metastasis of OSCC. This study can help elucidate the mechanism by which areca nut chewing contributes to the malignant transformation of oral epithelial cells.
Collapse
Affiliation(s)
- Yanbo Zhang
- Department of Stomatology, Chengde Medical University Affiliated Hospital, Chengde, Hebei, China
| | - Xuefeng Wang
- Department of Stomatology, Chengde Medical University Affiliated Hospital, Chengde, Hebei, China
| | - Shangzhi Han
- Department of Stomatology, Chengde Medical University Affiliated Hospital, Chengde, Hebei, China
| | - Yangyang Wang
- Department of Stomatology, Chengde Medical University Affiliated Hospital, Chengde, Hebei, China
| | - Rui Liu
- Department of Stomatology, Chengde Medical University Affiliated Hospital, Chengde, Hebei, China
| | - Fanli Meng
- Department of Stomatology, Chengde Medical University Affiliated Hospital, Chengde, Hebei, China
| | - Zhejun Su
- Department of Stomatology, Chengde Medical University Affiliated Hospital, Chengde, Hebei, China
| | - Feng Huo
- Department of Stomatology, Chengde Medical University Affiliated Hospital, Chengde, Hebei, China
| |
Collapse
|
23
|
Oncostatin M: A mysterious cytokine in cancers. Int Immunopharmacol 2020; 90:107158. [PMID: 33187910 DOI: 10.1016/j.intimp.2020.107158] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/04/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
Oncostatin M (OSM), as a member of the Interleukin-6 family cytokines, plays a significant role in inflammation, autoimmunity, and cancers. It is mainly secreted by T lymphocytes, neutrophils, and macrophages and was initially introduced as anti-cancer agent. However, in some cases, it promotes cancer progression. Overexpression of OSM and OSM receptor has been detected in various cancers including colon cancer, breast cancer, pancreatic cancer, myeloma, brain tumors, chronic lymphocytic leukemia, and hepatoblastoma. STAT3 is the main downstream signaling molecule of OSM, which operates the leading role in modifications of cancer cells and enhancing cell growth, invasion, survival, and all other hallmarks of cancer cells. However, due to the presence of multiple signaling pathways, it can act contradictory in some cancers. In this review, we will discuss the emerging roles of OSM in cancer and elucidate its function in tumor control or progression and finally discuss therapeutic approaches designed to manipulate this cytokine in cancer.
Collapse
|
24
|
Genetic toxicology and toxicokinetics of arecoline and related areca nut compounds: an updated review. Arch Toxicol 2020; 95:375-393. [PMID: 33097969 DOI: 10.1007/s00204-020-02926-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/05/2020] [Indexed: 01/13/2023]
Abstract
Areca nut (AN) is consumed by more than 600 million of individuals, particularly in some regions of South Asia, East Africa, and tropical Pacific, being classified as carcinogenic to humans. The most popular way of exposure consists of chewing a mixture of AN with betel leaf, slaked lime, and other ingredients that may also contain tobacco named betel quid (BQ). Arecoline is the principal active compound of AN, and, therefore, has been systematically studied over the years in several in vitro and in vivo genotoxicity endpoints. However, much of this information is dispersed, justifying the interest of an updated and comprehensive review article on this topic. In this sense, it is thus pertinent to describe and integrate the genetic toxicology data available as well as to address key toxicokinetics aspects of arecoline. This review also provides information on the effects induced by arecoline metabolites and related compounds, including other major AN alkaloids and nitrosation derivatives. The complexity of the chemicals involved renders this issue a challenge in genetic toxicology. Overall, positive results in several endpoints have been reported, some of them suggesting a key role for arecoline metabolites. Nevertheless, some negative genotoxicity findings for this alkaloid in short-term assays have also been reported in the literature. Finally, this article also collates information on the potential mechanisms of arecoline-induced genotoxicity, and suggests further approaches to tackle this important toxicological issue.
Collapse
|
25
|
Li P, Zhang S, Mo Y, Zhang L, Wang Y, Xiong F, Zhang S, Liu J, Xu Y, Zeng Z, Xiong W, Li Y, Gong Z. Long non-coding RNA expression profiles and related regulatory networks in areca nut chewing-induced tongue squamous cell carcinoma. Oncol Lett 2020; 20:302. [PMID: 33093911 DOI: 10.3892/ol.2020.12165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
Areca nut chewing is an important risk factor for developing tongue squamous cell carcinoma (TSCC), although the underlying molecular mechanism is unknown. To determine the potential molecular mechanisms of areca nut chewing-induced TSCC, the present study performed whole-genome detection with five pairs of TSCC and adjacent normal tissues, via mRNA- and long non-coding (lnc)RNA-gene chip analysis. A total of 3,860 differentially expressed genes were identified, including 2,193 lncRNAs and 1,667 mRNAs. Gene set-enrichment analysis revealed that the differentially expressed mRNAs were enriched in chromosome 22q13, 8p21 and 3p21 regions, and were regulated by nuclear factor kappa B (NF-κB) and interferon regulatory factors (IRFs). The results of ingenuity pathway analysis revealed that these mRNAs were significantly enriched for inflammatory immune-related signaling pathways. A co-expression network of mRNAs and lncRNAs was constructed by performing weighted gene co-expression network analysis. The present study focused on NF-κB-, IRF- and Th cell-signaling pathway-related lncRNAs and the corresponding mRNA-lncRNA regulatory networks. To the best of our knowledge, the present study was the first to investigate differential mRNA- and lncRNA-expression profiles in TSCCs induced by areca nut chewing. Inflammation-related mRNA-lncRNA regulatory networks driven by IRFs and NF-κB were identified, as well as the Th cell-related signaling pathways that play important carcinogenic roles in areca nut chewing-induced TSCC. These differentially expressed mRNAs and lncRNAs, and their regulatory networks provide insight for further analysis on the molecular mechanism of areca nut chewing-induced TSCC, candidate molecular markers and targets for further clinical intervention.
Collapse
Affiliation(s)
- Panchun Li
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Shanshan Zhang
- Department of Stomatology, The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yongzhen Mo
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, Hunan 410078, P.R. China
| | - Lishen Zhang
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, Hunan 410078, P.R. China
| | - Yumin Wang
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, Hunan 410078, P.R. China
| | - Fang Xiong
- Department of Stomatology, The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Shuai Zhang
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jiang Liu
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Yuming Xu
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, Hunan 410078, P.R. China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, Hunan 410078, P.R. China
| | - Yong Li
- Department of Medicine, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Zhaojian Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
26
|
Wang F, Ke Y, Yang L, Wang FJ. Quercetin protects human oral keratinocytes from lipopolysaccharide-induced injury by downregulating microRNA-22. Hum Exp Toxicol 2020; 39:1310-1317. [PMID: 32329368 DOI: 10.1177/0960327120918291] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Quercetin exerts anti-inflammatory effects, but whether it can benefit patients with the chronic inflammatory disease of oral lichen planus (OLP), which is a common chronic mucocutaneous disorder with an immune-mediated pathogenesis, is unclear. The present research examined the impacts of quercetin in a cell-based OLP model in which human oral keratinocytes (HOKs) were treated with lipopolysaccharide (LPS). METHODS Effects of quercetin on viability, proliferation, and apoptosis of HOKs were assessed using the Cell Counting Kit-8 assay, Western blotting, and flow cytometry, respectively. Effects of treatment on levels of microRNA-22 (miR-22) were measured using stem-loop reverse transcription polymerase chain reaction, while levels of proteins and phosphorylation in the PI3K/AKT and JAK1/STAT3 cascades were analyzed by Western blot. RESULTS Quercetin mitigated LPS-induced reduction in HOK viability and elevation of apoptosis. It also weakened LPS-induced upregulation of miR-22. Quercetin treatment led to significantly higher levels of p-PI3K, p-AKT, p-JAK1, and p-STAT3. These effects of quercetin were enhanced when miR-22 was knocked down and partly reversed when miR-22 was overexpressed. CONCLUSION Quercetin can mitigate LPS-induced injury in HOKs by downregulating miR-22, thereby activating PI3K/AKT and JAK1/STAT3 cascades.
Collapse
Affiliation(s)
- F Wang
- Department of Stomatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Y Ke
- Department of Stomatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - L Yang
- Department of Stomatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - F J Wang
- Department of Stomatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| |
Collapse
|
27
|
Islam S, Uehara O, Matsuoka H, Kuramitsu Y, Adhikari BR, Hiraki D, Toraya S, Jayawardena A, Saito I, Muthumala M, Nagayasu H, Abiko Y, Chiba I. DNA hypermethylation of sirtuin 1 (SIRT1) caused by betel quid chewing-a possible predictive biomarker for malignant transformation. Clin Epigenetics 2020; 12:12. [PMID: 31931863 PMCID: PMC6958620 DOI: 10.1186/s13148-019-0806-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 12/30/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND DNA hypermethylation of tumor suppressor genes is observed in precancerous lesions and oral cancer of individuals with the habits of betel quid (BQ) chewing. SIRT1 has been identified as playing a role in the maintenance of epithelial integrity, and its alteration is often related to carcinogenesis. However, the methylation and transcription status of SIRT1 in patients with BQ chewing-related oral cancer has not been investigated. We examined the methylation status of SIRT1 in paraffin-embedded tissue samples of oral squamous cell carcinoma (OSCC) obtained from BQ chewing and non-chewing patients and in tissue samples from healthy control subjects. In addition, we examined whether the hypermethylation of SIRT1 followed by its transcriptional downregulation in the human gingival epithelial cells could be caused by arecoline, a major component of BQ. Furthermore, we investigated the methylation status of SIRT1 in smear samples of macroscopically healthy buccal mucosa from subjects with a habit of BQ chewing. RESULTS SIRT1 was significantly hypermethylated in tissue samples of OSCC from BQ chewers and non-chewers than in oral mucosa from healthy control subjects. Results also showed that the hypermethylation level of SIRT1 was significantly higher in OSCC of patients with BQ chewing habits than in those of non-chewing habits (p < 0.05). Our in vitro model showed that hypermethylation is followed by downregulation of the transcriptional level of SIRT1 (p < 0.05). The methylation levels of SIRT1 in the smear samples obtained from BQ chewing individuals were significantly higher than those in the samples obtained from individuals that did not chew BQ. The duration of BQ chewing habits was correlated positively to the frequency of SIRT1 hypermethylation (p < 0.05). CONCLUSIONS Our results suggest that DNA hypermethylation of SIRT1 is involved in the occurrence of oral cancer in BQ chewing patients and that hypermethylation in the oral mucosa of BQ chewers could be a predictive marker for the occurrence of malignant transformation. This is the first report that showed DNA hypermethylation in clinically healthy oral epithelium of BQ chewers. Our study shows evidence that DNA hypermethylation may be an early event of oral carcinogenesis prior to observable clinical changes.
Collapse
Affiliation(s)
- Shajedul Islam
- Division of Disease Control and Molecular Epidemiology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan.,Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan
| | - Osamu Uehara
- Division of Disease Control and Molecular Epidemiology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan.,Research Institute of Cancer Prevention, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan
| | - Hirofumi Matsuoka
- Division of Disease Control and Molecular Epidemiology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan
| | - Yasuhiro Kuramitsu
- Research Institute of Cancer Prevention, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan
| | - Bhoj Raj Adhikari
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan
| | - Daichi Hiraki
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan
| | - Seiko Toraya
- Division of Disease Control and Molecular Epidemiology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan
| | - Asiri Jayawardena
- Department of General Education, School of Dental Medicine, Tsurumi University, Kanagawa, 230-8501, Japan
| | - Ichiro Saito
- Department of Pathology, School of Dental Medicine, Tsurumi University, Kanagawa, 230-8501, Japan
| | - Malsantha Muthumala
- Department of Oral and Maxillofacial Surgery, Army Hospital, Colombo, Sri Lanka
| | - Hiroki Nagayasu
- Division of Oral and Maxillofacial Surgery, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan
| | - Yoshihiro Abiko
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan.
| | - Itsuo Chiba
- Division of Disease Control and Molecular Epidemiology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan
| |
Collapse
|
28
|
Chuerduangphui J, Ekalaksananan T, Heawchaiyaphum C, Vatanasapt P, Pientong C. Peroxiredoxin 2 is highly expressed in human oral squamous cell carcinoma cells and is upregulated by human papillomavirus oncoproteins and arecoline, promoting proliferation. PLoS One 2020; 15:e0242465. [PMID: 33332365 PMCID: PMC7746188 DOI: 10.1371/journal.pone.0242465] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 11/02/2020] [Indexed: 01/19/2023] Open
Abstract
Peroxiredoxin 2 (PRDX2) is upregulated in various cancers including oral squamous cell carcinoma (OSCC). It is a known tumor promoter in some cancers, but its role in OSCC is unclear. This study aimed to investigate the effect of arecoline, an alkaloid of the betel nut, and human papillomavirus type 16 (HPV16) E6/E7 oncoproteins on induction of PRDX2 expression, and also the effects of PRDX2 overexpression in oral cell lines. Levels of PRDX2 protein were determined using western blot analysis of samples of exfoliated normal oral cells (n = 75) and oral lesion cells from OSCC cases (n = 75). Some OSCC cases were positive for HPV infection and some patients had a history of betel quid chewing. To explore the level of PRDX2 by western blot, the proteins were extracted from oral cell lines that were treated with arecoline or retroviruses containing HPV16 E6 gene and HPV16 E6/E7 expressing vector. For analysis of PRDX2 functions, cell proliferation, cell-cycle progression, apoptosis and migration was compared between oral cells overexpressing PRDX2 and cells with PRDX2-knockdown. PRDX2 expression levels tended to be higher in OSCC samples that were positive for HPV infection and had history of betel quid chewing. Arecoline treatment in vitro at low concentrations and overexpression of HPV16 E6 or E6/E7 in oral cells induced PRDX2 overexpression. Interestingly, in oral cells, PRDX2 promoted cell proliferation, cell-cycle progression (G2/M phase), cell migration and inhibited apoptosis. Upregulation of PRDX2 in oral cells was induced by arecoline and HPV16 oncoproteins and promoted growth of OSCC cells.
Collapse
Affiliation(s)
- Jureeporn Chuerduangphui
- Department of Microbiology, Faculty of Science, Kasetsart University, Bangkok, Thailand
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
| | - Tipaya Ekalaksananan
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Chukkris Heawchaiyaphum
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Patravoot Vatanasapt
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
- Department of Otorhinolaryngology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Chamsai Pientong
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- * E-mail:
| |
Collapse
|
29
|
Han Z, Zhan R, Chen S, Deng J, Shi J, Wang W. miR-181b/Oncostatin m axis inhibits prostate cancer bone metastasis via modulating osteoclast differentiation. J Cell Biochem 2019; 121:1664-1674. [PMID: 31680294 DOI: 10.1002/jcb.29401] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022]
Abstract
The activation of osteoblasts is significantly correlated to prostate tumor bone metastasis and bone loss. Oncostatin M (OSM) could promote breast cancer metastasis to bone. However, its role and mechanism in prostate cancer bone metastasis remain unclear. MicroRNAs (miRNAs) could play important roles in cancers via post-transcriptionally regulating target genes via binding to specific sequences in the 3' UTR of downstream target genes. In the present study, we performed microarray profiling analyses to identify differentially-expressed miRNAs in preosteoclast before and after osteoclast differentiation that could target OSM. miR-181b-5p was downregulated during Raw264.7 cells differentiation into osteoclast. By direct targeting OSM 3' UTR, miR-181b-5p inhibited OSM messenger RNA expression and protein levels, subsequently decreasing IL-6 and AREG and increasing OPG, while OSM overexpression exerted an opposing effect. More importantly, co-culture with miR-181b-5p-overexpressing differentiated Raw264.7 cells suppressed proliferation, migration, and invasion of mouse prostate cancer RM-1 cells, while co-culture with OSM-overexpressing Raw264.7 cells led to opposing cellular effects. More importantly, the effects of miR-181b-5p on osteoclastogenic factors and RM-1 cells could be significantly reversed by OSM overexpression. In summary, miR-181b-5p/OSM axis could be a viable therapeutic target for patients with surgically removed primary tumors to reduce bone metastasis and prevent bone loss.
Collapse
Affiliation(s)
- Ziwei Han
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ruisen Zhan
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shijie Chen
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia Deng
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Shi
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weiguo Wang
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
30
|
Zhang C, Hao Y, Sun Y, Liu P. Quercetin suppresses the tumorigenesis of oral squamous cell carcinoma by regulating microRNA-22/WNT1/β-catenin axis. J Pharmacol Sci 2019; 140:128-136. [DOI: 10.1016/j.jphs.2019.03.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/19/2019] [Accepted: 03/22/2019] [Indexed: 12/11/2022] Open
|
31
|
How Each Component of Betel Quid Is Involved in Oral Carcinogenesis: Mutual Interactions and Synergistic Effects with Other Carcinogens—a Review Article. Curr Oncol Rep 2019; 21:53. [DOI: 10.1007/s11912-019-0800-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
32
|
Gupta A, Ajith A, Singh S, Panday RK, Samaiya A, Shukla S. PAK2-c-Myc-PKM2 axis plays an essential role in head and neck oncogenesis via regulating Warburg effect. Cell Death Dis 2018; 9:825. [PMID: 30068946 PMCID: PMC6070504 DOI: 10.1038/s41419-018-0887-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/07/2018] [Accepted: 07/18/2018] [Indexed: 12/21/2022]
Abstract
The histone modifiers (HMs) are crucial for chromatin dynamics and gene expression; however, their dysregulated expression has been observed in various abnormalities including cancer. In this study, we have analyzed the expression of HMs in microarray profiles of head and neck cancer (HNC), wherein a highly significant overexpression of p21-activated kinase 2 (PAK2) was identified which was further validated in HNC patients. The elevated expression of PAK2 positively correlated with enhanced cell proliferation, aerobic glycolysis and chemoresistance and was associated with the poor clinical outcome of HNC patients. Further, dissection of molecular mechanism revealed an association of PAK2 with c-Myc and c-Myc-dependent PKM2 overexpression, wherein we showed that PAK2 upregulates c-Myc expression and c-Myc thereby binds to PKM promoter and induces PKM2 expression. We observed that PAK2-c-Myc-PKM2 axis is critical for oncogenic cellular proliferation. Depletion of PAK2 disturbs the axis and leads to downregulation of c-Myc and thereby PKM2 expression, which resulted in reduced aerobic glycolysis, proliferation and chemotherapeutic resistance of HNC cells. Moreover, the c-Myc complementation rescued PAK2 depletion effects and restored aerobic glycolysis, proliferation, migration and invasion in PAK2-depleted cells. The global transcriptome analysis of PAK2-depleted HNC cells revealed the downregulation of various genes involved in active cell proliferation, which indicates that PAK2 overexpression is critical for HNC progression. Together, these results suggest that the axis of PAK2-c-Myc-PKM2 is critical for HNC progression and could be a therapeutic target to reduce the cell proliferation and acquired chemoresistance and might enhance the efficacy of standard chemotherapy which will help in better management of HNC patients.
Collapse
Affiliation(s)
- Amit Gupta
- Epigenetics and RNA Processing Lab, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
| | - Athira Ajith
- Epigenetics and RNA Processing Lab, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
- Lab No. 315, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology, Madras, Tamil Nadu, 600036, India
| | - Smriti Singh
- Epigenetics and RNA Processing Lab, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
| | | | - Atul Samaiya
- Department of Surgical Oncology, Bansal Hospital, Bhopal, Madhya Pradesh, 462016, India
| | - Sanjeev Shukla
- Epigenetics and RNA Processing Lab, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India.
| |
Collapse
|
33
|
Sari LM, Subita GP, Auerkari EI. Areca nut extract demonstrated apoptosis-inducing mechanism by increased caspase-3 activities on oral squamous cell carcinoma. F1000Res 2018; 7:723. [PMID: 31448077 PMCID: PMC6668048 DOI: 10.12688/f1000research.14856.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/04/2018] [Indexed: 12/11/2022] Open
Abstract
Background: Oral squamous cell carcinoma is a neoplasm of keratinocyte cells of oral mucosa epithelium that can potentially spread through lymphatic tissue or blood vessel. Although areca nut is one of the plants with a risk of inducing that cancer, areca nut is believed to have high antioxidant properties. Due to the current interest in the apoptosis effects from areca nut for oral cancer treatment, we investigated its ability to induce apoptosis and caspase-3 activity in oral cancer cell lines: HSC-2 and HSC-3. Methods: We examined the effect of areca nut on apoptosis and caspase-3 activity in HSC-2 and HSC-3 cells. Flow cytometry was conducted for the quantification of the cells that were apoptotic and expressing the caspase-3 enzyme for 24 and 48 hours. Results: Areca nut induced a significant increase (p<0.01) in late apoptosis of HSC-2 cells and mostly occurred over 48 hours. The study also found that in HSC-3, there were significant increases (p<0.01) the percentage of cells in early apoptosis after 24 hours and late apoptosis at 48 hours. Caspase-3 activity increased after 24 and 48 hours of areca nut exposure in both cells. Conclusions: The study showed that areca nut could be considered as a potential anticancer agent through its capability in inducing a caspase-dependent apoptosis.
Collapse
Affiliation(s)
- Liza Meutia Sari
- Oral Medicine Department, Faculty of Dentistry, University of Syiah Kuala, Banda Aceh, 23111, Indonesia
| | - Gus Permana Subita
- Oral Medicine Department, Faculty of Dentistry, University of Indonesia, Jakarta, 10430, Indonesia
| | - Elza Ibrahim Auerkari
- Oral Biology Department, Faculty of Dentistry, University of Indonesia, Jakarta, 10430, Indonesia
| |
Collapse
|
34
|
Sari LM, Subita GP, Auerkari EI. Areca nut extract demonstrated apoptosis-inducing mechanism by increased caspase-3 activities on oral squamous cell carcinoma. F1000Res 2018; 7:723. [PMID: 31448077 PMCID: PMC6668048 DOI: 10.12688/f1000research.14856.5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2019] [Indexed: 01/03/2023] Open
Abstract
Background: Oral squamous cell carcinoma is a neoplasm of keratinocyte cells of oral mucosa epithelium that can potentially spread through lymphatic tissue or blood vessel. Although areca nut is one of the plants with a risk of inducing that cancer, areca nut is believed to have high antioxidant properties. Due to the current interest in the apoptosis effects from areca nut for oral cancer treatment, we investigated its ability to induce apoptosis and caspase-3 activity in oral cancer cell lines: HSC-2 and HSC-3. Methods: We examined the effect of areca nut on apoptosis and caspase-3 activity in HSC-2 and HSC-3 cells. Flow cytometry was conducted for the quantification of the cells that were apoptotic and expressing the caspase-3 enzyme for 24 and 48 hours. Results: Areca nut induced a significant increase (p<0.01) in late apoptosis of HSC-2 cells and mostly occurred over 48 hours. The study also found that in HSC-3, there were significant increases (p<0.01) the percentage of cells in early apoptosis after 24 hours and late apoptosis at 48 hours. Caspase-3 activity increased after 24 and 48 hours of areca nut exposure in both cells. Conclusions: The study showed that areca nut could be considered as a potential anticancer agent through its capability in inducing a caspase-dependent apoptosis.
Collapse
Affiliation(s)
- Liza Meutia Sari
- Oral Medicine Department, Faculty of Dentistry, University of Syiah Kuala, Banda Aceh, 23111, Indonesia
| | - Gus Permana Subita
- Oral Medicine Department, Faculty of Dentistry, University of Indonesia, Jakarta, 10430, Indonesia
| | - Elza Ibrahim Auerkari
- Oral Biology Department, Faculty of Dentistry, University of Indonesia, Jakarta, 10430, Indonesia
| |
Collapse
|