1
|
Kudo Y, Nakamura K, Tsuzuki H, Hirota K, Kawai M, Takaya D, Fukuzawa K, Honma T, Yoshino Y, Nakamura M, Shiota M, Fujimoto N, Ikari A, Endo S. Docosahexaenoic acid enhances the treatment efficacy for castration-resistant prostate cancer by inhibiting autophagy through Atg4B inhibition. Arch Biochem Biophys 2024; 760:110135. [PMID: 39181384 DOI: 10.1016/j.abb.2024.110135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Autophagy induction in cancer is involved in cancer progression and the acquisition of resistance to anticancer agents. Therefore, autophagy is considered a potential therapeutic target in cancer therapy. In this study, we found that long-chain fatty acids (LCFAs) have inhibitory effects on Atg4B, which is essential for autophagosome formation, through screening based on the pharmacophore of 21f, a recently developed Atg4B inhibitor. Among these fatty acids, docosahexaenoic acid (DHA), a polyunsaturated fatty acid, exhibited the most potent Atg4B inhibitory activity. DHA inhibited autophagy induced by androgen receptor signaling inhibitors (ARSI) in LNCaP and 22Rv1 prostate cancer cells and significantly increased apoptotic cell death. Furthermore, we investigated the effect of DHA on resistance to ARSI by establishing darolutamide-resistant prostate cancer 22Rv1 (22Rv1/Dar) cells, which had developed resistance to darolutamide, a novel ARSI. At baseline, 22Rv1/Dar cells showed a higher autophagy level than parental 22Rv1 cells. DHA significantly suppressed Dar-induced autophagy and sensitized 22Rv1/Dar cells by inducing apoptotic cell death through mitochondrial dysfunction. These results suggest that DHA supplementation may improve prostate cancer therapy with ARSI.
Collapse
Affiliation(s)
- Yudai Kudo
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Kana Nakamura
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Honoka Tsuzuki
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Kotaro Hirota
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Mina Kawai
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Daisuke Takaya
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Kaori Fukuzawa
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Teruki Honma
- Center for Biosystems Dynamics Research, RIKEN, Kanagawa, 230-0045, Japan
| | - Yuta Yoshino
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Mitsuhiro Nakamura
- Laboratories of Drug Informatics, Department of Pharmacy Practice and Science, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Naohiro Fujimoto
- Department of Urology, University of Occupational and Environmental Health, Fukuoka, 807-8555, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Satoshi Endo
- The United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, 501-1194, Japan; Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu, 501-1193, Japan.
| |
Collapse
|
2
|
Senevirathna K, Pradeep R, Jayasinghe YA, Jayawickrama SM, Illeperuma R, Warnakulasuriya S, Jayasinghe RD. Carcinogenic Effects of Areca Nut and Its Metabolites: A Review of the Experimental Evidence. Clin Pract 2023; 13:326-346. [PMID: 36961055 PMCID: PMC10037666 DOI: 10.3390/clinpract13020030] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023] Open
Abstract
Oral cancers (OC) are among the most frequent malignancies encountered in Southeast Asia, primarily due to the prevalent habit of betel quid (BQ) and smokeless tobacco use in this region. Areca nut (AN), the primary ingredient in BQ, contains several alkaloids, including arecoline, arecaidine, guvacoline, and guvacine. These have been associated with both the AN abuse liability and carcinogenicity. Additionally, variations in AN alkaloid levels could lead to differences in the addictiveness and carcinogenic potential across various AN-containing products. Recent studies based on animal models and in vitro experiments show cellular and molecular effects induced by AN. These comprise promoting epithelial-mesenchymal transition, autophagy initiation, tissue hypoxia, genotoxicity, cytotoxicity, and cell death. Further, clinical research endorses these undesired harmful effects in humans. Oral submucosal fibrosis, a potentially malignant disease of the oral cavity, is predominantly reported from the geographical areas of the globe where AN is habitually chewed. OC in chronic AN users presents a more aggressive phenotype, such as resistance to anti-cancer drugs. The available evidence on the carcinogenicity of AN based on the findings reported in the recently published experimental studies is discussed in the present review.
Collapse
Affiliation(s)
- Kalpani Senevirathna
- Centre for Research in Oral Cancer (CROC), Faculty of Dental Sciences, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Roshan Pradeep
- Centre for Research in Oral Cancer (CROC), Faculty of Dental Sciences, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Yovanthi Anurangi Jayasinghe
- Centre for Research in Oral Cancer (CROC), Faculty of Dental Sciences, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Shalindu Malshan Jayawickrama
- Centre for Research in Oral Cancer (CROC), Faculty of Dental Sciences, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Rasika Illeperuma
- Centre for Research in Oral Cancer (CROC), Faculty of Dental Sciences, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Saman Warnakulasuriya
- Faculty of Dentistry, Oral and Craniofacial Sciences, King's College, London SE1 9RA, UK
| | - Ruwan Duminda Jayasinghe
- Centre for Research in Oral Cancer (CROC), Faculty of Dental Sciences, University of Peradeniya, Peradeniya 20400, Sri Lanka
- Department of Oral Medicine and Periodontology, Faculty of Dental Sciences, University of Peradeniya, Peradeniya 20400, Sri Lanka
| |
Collapse
|
3
|
Chen L, Yuan F, Chen S, Li X, Kong L, Zhang W. Potential Role of Host Microbiome in Areca Nut-Associated Carcinogenesis and Addiction. Molecules 2022; 27:8171. [PMID: 36500264 PMCID: PMC9739811 DOI: 10.3390/molecules27238171] [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: 06/28/2022] [Revised: 08/21/2022] [Accepted: 08/30/2022] [Indexed: 11/25/2022] Open
Abstract
Areca nut (AN) is widely consumed all over the world, bringing great harm to human health and economy. Individuals with AN chewing are at high risk of cardiovascular disease and impaired immune system and metabolic system. Despite a growing number of studies having reported on the adverse effects brought by AN chewing, the exact mechanism of it is limited and the need for additional exploration remains. In recent years, the interaction between microorganisms, especially intestinal microorganism and host, has been extensively studied. AN chewing might disrupt the oral and intestinal microbiota communities through direct connect with the microbes it contains, altering PH, oxygen of oral and intestinal microenvironment, and disturbing the immune homeostasis. These mechanisms provide insights into the interplay between areca nut and host microbiota. Emerging studies have proposed that bidirectional interaction between polyphenols and intestinal microbes might play a potential role in the divergence of polyphenol, extracted from AN, among individuals with or without AN-induced cancer development and progression. Although some AN chewers have been aware of the harmful effects brought by AN, they cannot abolish this habit because of the addiction of AN. Increasing studies have tried to revealed that gut microbiota might influence the onset/development of addictive behaviors. Altogether, this review summarizes the possible reasons for the disturbance of host microbiota caused by areca nut chewing and clarifies the complex interaction between human microbiome and major constituents and the addiction and carcinogenicity of AN, tempting to provide novel insights into the development and utilization of it, and to control the adverse consequences caused by AN chewing.
Collapse
Affiliation(s)
- Lihui Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410078, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, China
| | - Fulai Yuan
- Health Management Center, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Sifang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Xiong Li
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510060, China
| | - Lingyu Kong
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410078, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, China
| |
Collapse
|
4
|
Kudo Y, Endo S, Fujita M, Ota A, Kamatari YO, Tanaka Y, Ishikawa T, Ikeda H, Okada T, Toyooka N, Fujimoto N, Matsunaga T, Ikari A. Discovery and Structure-Based Optimization of Novel Atg4B Inhibitors for the Treatment of Castration-Resistant Prostate Cancer. J Med Chem 2022; 65:4878-4892. [PMID: 35244402 DOI: 10.1021/acs.jmedchem.1c02113] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Autophagy inhibition is an attractive target for cancer therapy. In this study, we discovered inhibitors of Atg4B essential for autophagosome formation and evaluated their potential as therapeutics for prostate cancer. Seventeen compounds were identified as candidates after in silico screening and a thermal shift assay. Among them, compound 17 showed the most potent Atg4B inhibitory activity, inhibited autophagy induced by anti-castration-resistant prostate cancer (CRPC) drugs, and significantly enhanced apoptosis. Although 17 has been known as a phospholipase A2 (PLA2) inhibitor, other PLA2 inhibitors had no effect on Atg4B and autophagy. We then performed structural optimization based on molecular modeling and succeeded in developing 21f (by shortening the alkyl chain of 17), which was a potent competitive inhibitor for Atg4B (Ki = 3.1 μM) with declining PLA2 inhibitory potency. Compound 21f enhanced the anticancer activity of anti-CRPC drugs via autophagy inhibition. These findings suggest that 21f can be used as an adjuvant drug for therapy with anti-CRPC drugs.
Collapse
Affiliation(s)
- Yudai Kudo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Mei Fujita
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Atsumi Ota
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Yuji O Kamatari
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan.,United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Takeshi Ishikawa
- Graduate School of Science and Engineering, Kagoshima University, 1-21-40 Korimoto, Kagoshima 890-0065, Japan
| | - Hayato Ikeda
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-8555, Japan
| | - Takuya Okada
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-8555, Japan
| | - Naoki Toyooka
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-8555, Japan
| | - Naohiro Fujimoto
- Department of Urology, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Toshiyuki Matsunaga
- Education Center of Green Pharmaceutical Sciences, Gifu Pharmaceutical University, Gifu 502-8585, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| |
Collapse
|
5
|
Autophagy-A Hidden but Important Actor on Oral Cancer Scene. Int J Mol Sci 2020; 21:ijms21239325. [PMID: 33297472 PMCID: PMC7729760 DOI: 10.3390/ijms21239325] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/26/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022] Open
Abstract
The duration of denture use, oral hygiene, smoking and male sex were identified as risk factors for oral mucosal lesions. As it is well known, all the oral mucosal lesions associated with risk factors have an important degree of malignity. Chronic mechanical irritation can be another cause of oral cancer and it is produced by the constant action of a deleterious agent from the oral cavity. Autophagy represents a complex evolutionary conserved catabolic process in which cells self-digest intracellular organelles in order to regulate their normal turnover and remove the damaged ones with compromised function to further maintain homeostasis. Autophagy is modulated by mTOR kinase and indirectly by PI3K/AKT survival pathway. Due to its dual capacity to either induce cell death or promote cell survival, important evidence pointed that autophagy has a two-faced role in response to chemotherapy in cancer. In conclusion, understanding how to overcome cytoprotective autophagy and how to take advantage of autophagic cell death is critical in order to enhance the cancer cells sensitivity to particular therapeutic agents.
Collapse
|
6
|
Yang WF, Qin N, Song X, Jiang C, Li T, Ji P, Li Y, Ding D, Wang C, Dai J, Jin G, Chen TW, Chang YS, Ouyang DQ, Liao GQ, Hu Z, Chang KP, Su YX, Ma H. Genomic Signature of Mismatch Repair Deficiency in Areca Nut-Related Oral Cancer. J Dent Res 2020; 99:1252-1261. [PMID: 32527169 DOI: 10.1177/0022034520930641] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Areca nut (AN) chewing contributes to an increase of oral squamous cell carcinoma (OSCC) cases in South and Southeast Asia; however, genomic events underlying the carcinogenesis process of AN-related OSCC remain unclear. Here, we comprehensively describe the genomic and transcriptome alterations of 113 Chinese OSCC patients (89 AN related and 24 AN negative) by whole-exome sequencing and RNA sequencing, and we compared the genomic differences between AN-related and AN-negative samples by integrating sequencing data of 325 OSCC patients from The Cancer Genome Atlas database and 50 from a published Taiwanese study. We identified 11 significantly mutated genes for OSCC, including 4 novel ones (ATG2A, WEE1, DST, and TSC2), of which WEE1 and ATG2A mutated with significantly higher rates in AN-related samples (P = 0.04 and P = 0.003, respectively). Mutational signature analysis revealed that AN-related OSCCs were specially characterized by the genomic signature of mismatch repair deficiency (dMMR), which could also predict the prognosis status of AN-related OSCC. In addition, an elevated PD-L1 expression was also observed in both AN-related patients (P = 3.71 × 10-11) and those with a high dMMR level (P = 1.99 × 10-4). Further differential expression analysis and in vitro experiments confirmed the role of dMMR in the development of OSCC induced by AN exposure. Taken together, this study first revealed the molecular profiles and highlighted the role of dMMR in AN-related OSCC among the Chinese population and identified that AN-related OSCC may represent a potential cohort for effective anti-PD-1/L1 immunotherapy.
Collapse
Affiliation(s)
- W F Yang
- Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - N Qin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Mainland China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Mainland China
| | - X Song
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Mainland China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Mainland China
| | - C Jiang
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, Mainland China
| | - T Li
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, Mainland China
| | - P Ji
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Mainland China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Mainland China
| | - Y Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Mainland China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Mainland China
| | - D Ding
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Mainland China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Mainland China
| | - C Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Mainland China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Mainland China.,Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Mainland China
| | - J Dai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Mainland China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Mainland China
| | - G Jin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Mainland China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Mainland China
| | - T W Chen
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Y S Chang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.,Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - D Q Ouyang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Mainland China
| | - G Q Liao
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Mainland China
| | - Z Hu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Mainland China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Mainland China
| | - K P Chang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.,Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Y X Su
- Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - H Ma
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Mainland China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Mainland China
| |
Collapse
|
7
|
Xu Z, Lü FY, Jiang EH, Zhao XP, Shang ZJ. [Relationship among areca nut, intracellular reactive oxygen species, and autophagy]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2020; 38:80-85. [PMID: 32037771 PMCID: PMC7184295 DOI: 10.7518/hxkq.2020.01.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 09/27/2019] [Indexed: 12/31/2022]
Abstract
The relationship between areca nut as a primary carcinogen and oral cancer has been widely concerned. Areca can change the levels of reactive oxygen species (ROS) and autophagy in cells, and the levels of ROS and autophagy are closely related to the occurrence and development of tumors. This paper reviewed the relationships among areca nut, intracellular ROS, and autophagy.
Collapse
Affiliation(s)
- Zhi Xu
- Dept. of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feng-Yuan Lü
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Er-Hui Jiang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology Hubei-MOST & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Xiao-Ping Zhao
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zheng-Jun Shang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology Hubei-MOST & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
8
|
Chemistry, metabolism and pharmacology of carcinogenic alkaloids present in areca nut and factors affecting their concentration. Regul Toxicol Pharmacol 2019; 110:104548. [PMID: 31805361 DOI: 10.1016/j.yrtph.2019.104548] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/27/2019] [Accepted: 12/01/2019] [Indexed: 12/28/2022]
Abstract
Areca Nut (AN), the seed of tropical palm tree Areca catechu, is a widely chewed natural product with estimated 600 million users across the world. Various AN products, thriving in the market, portray 'Areca nut' or 'Supari' as mouth freshener and safe alternative to smokeless tobacco. Unfortunately, AN is identified as a Group 1 human carcinogen by International Agency for Research on Cancer (IARC). Wide variation in the level of alkaloids, broadly ranging from 2 to 10 mg/gm dry weight, is observed in diverse variety of AN sold worldwide. For the first time, various factors influencing the formation of carcinogenic alkaloids in AN at various stages, including during the growth, processing, and storage of the nut, are discussed. Current review illustrates the mechanism of cancer induction by areca alkaloids in humans and also compiles dose-dependent pharmacology and toxicology data of arecoline, the most potent carcinogenic alkaloid in AN. Careful monitoring of the arecoline content in AN can potentially be used as a tool in product surveillance studies to identify the variations in characteristics of various AN sample sold worldwide. The article will help to generate public awareness and sensitize the government bodies to initiate campaigns against AN use and addiction.
Collapse
|
9
|
Li YC, Cheng AJ, Lee LY, Huang YC, Chang JTC. Multifaceted Mechanisms of Areca Nuts in Oral Carcinogenesis: the Molecular Pathology from Precancerous Condition to Malignant Transformation. J Cancer 2019; 10:4054-4062. [PMID: 31417650 PMCID: PMC6692602 DOI: 10.7150/jca.29765] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 03/29/2019] [Indexed: 02/06/2023] Open
Abstract
Oral cancer is one of the most frequent malignant diseases worldwide, and areca nut is a primary carcinogen causing this cancer in Southeast Asia. It has been widely reported that areca nut induced several cytotoxic effects in oral cells, including ROS generation, inflammation, tissue hypoxia, DNA damage, and cell invasion. Recently, through chronic exposure model, more extensive pathological effects due to areca nut have been found. These include the induction of autophagy, promotion of epithelial- mesenchymal transition, and facilitation of cancer stemness conversion. Clinical findings support these adverse effects. Oral submucosal fibrosis, a premalignant condition, is prevalent in the area with habitual chewing of areca nuts. Consistently, oral cancer patients with habitual chewing areca nut exhibit more aggressive phenotypes, including resistance to chemo-radiotherapy. In this review, we comprehensively discuss and concisely summarize the up-to-date molecular and cellular mechanisms by which areca nuts contribute to malignant transformation. This review may provide critical information regarding clinical applications in risk assessment, disease prevention, diagnosis, and personalized therapeutics for areca nut-induced oral malignancy.
Collapse
Affiliation(s)
- Yi-Chen Li
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Ann-Joy Cheng
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan.,Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan 333, Taiwan
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan 333, Taiwan
| | - Yu-Chen Huang
- Department of Oral Maxillofacial Surgery, Chang Gung Memorial Hospital-Linkou, Taoyuan 333, Taiwan
| | - Joseph Tung-Chieh Chang
- Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan 333, Taiwan.,Department of Radiation Oncology, Xiamen Chang Gung Memorial Hospital, Xiamen, Fujian, China
| |
Collapse
|
10
|
Lin MH, Liu YC, Liu SY, Chen FC, Yang PJ, Li GH, Liu PY, Yen CY. Clathrin-mediated endocytosis is required for ANE 30-100K-induced autophagy. J Oral Pathol Med 2017; 47:25-31. [PMID: 28520088 DOI: 10.1111/jop.12593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2017] [Indexed: 01/30/2023]
Abstract
BACKGROUND We identified an autophagy-inducing areca nut (AN) ingredient (AIAI) in the 30-100 kDa fraction of AN extract (ANE 30-100K). This study was to analyze the role of endocytosis in ANE 30-100K-induced autophagy. METHODS We used benzyl alcohol, dynasore, and shRNA of clathrin and dynamin to assess whether ANE 30-100K-induced cytotoxicity and accumulation of microtubule-associated protein 1 light chain 3 (LC3)-II were affected in oral (OECM-1) and esophageal (CE81T/VGH) carcinoma cells. RESULTS Both benzyl alcohol and dynasore effectively reduced ANE 30-100K-induced cytotoxicity and LC3-II accumulation in OECM-1 and CE81T/VGH cells. Downregulated protein expression of both clathrin and dynamin by their shRNA also significantly attenuated ANE 30-100K-induced elevation of LC3-II levels in CE81T/VGH cells. CONCLUSIONS These results indicate that AIAI may be engulfed by cells through clathrin-mediated endocytosis, which promotes the execution of the following autophagy program.
Collapse
Affiliation(s)
- Mei-Huei Lin
- Department of Biotechnology, Chia Nan University of Pharmacy, Tainan, Taiwan.,Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Young-Chau Liu
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan.,Division of Natural Science, College of Liberal Education, Shu-Te University, Kaohsiung City, Taiwan
| | - Shyun-Yeu Liu
- Department of Dentistry, Taipei Medical University, Taipei, Taiwan.,Department of Dentistry, National Defense Medical Center, Taipei, Taiwan.,Oral and Maxillofacial Surgery Section, Chi Mei Medical Center, Tainan, Taiwan
| | - Fang-Chi Chen
- Department of Biotechnology, Chia Nan University of Pharmacy, Tainan, Taiwan
| | - Pei-Jung Yang
- Department of Biotechnology, Chia Nan University of Pharmacy, Tainan, Taiwan
| | - Guan-Hua Li
- Department of Biotechnology, Chia Nan University of Pharmacy, Tainan, Taiwan
| | - Pang-Yen Liu
- Department of Biotechnology, Chia Nan University of Pharmacy, Tainan, Taiwan
| | - Ching-Yu Yen
- Department of Dentistry, Taipei Medical University, Taipei, Taiwan.,Department of Dentistry, National Defense Medical Center, Taipei, Taiwan.,Oral and Maxillofacial Surgery Section, Chi Mei Medical Center, Tainan, Taiwan
| |
Collapse
|
11
|
Tang JC, Feng YL, Liang X, Cai XJ. Autophagy in 5-Fluorouracil Therapy in Gastrointestinal Cancer: Trends and Challenges. Chin Med J (Engl) 2017; 129:456-63. [PMID: 26879020 PMCID: PMC4800847 DOI: 10.4103/0366-6999.176069] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective: 5-Fluorouracil (5-FU)-based combination therapies are standard treatments for gastrointestinal cancer, where the modulation of autophagy is becoming increasingly important in offering effective treatment for patients in clinical practice. This review focuses on the role of autophagy in 5-FU-induced tumor suppression and cancer therapy in the digestive system. Data Sources: All articles published in English from 1996 to date those assess the synergistic effect of autophagy and 5-FU in gastrointestinal cancer therapy were identified through a systematic online search by use of PubMed. The search terms were “autophagy” and “5-FU” and (“colorectal cancer” or “hepatocellular carcinoma” or “pancreatic adenocarcinoma” or “esophageal cancer” or “gallbladder carcinoma” or “gastric cancer”). Study Selection: Critical reviews on relevant aspects and original articles reporting in vitro and/or in vivo results regarding the efficiency of autophagy and 5-FU in gastrointestinal cancer therapy were reviewed, analyzed, and summarized. The exclusion criteria for the articles were as follows: (1) new materials (e.g., nanomaterial)-induced autophagy; (2) clinical and experimental studies on diagnostic and/or prognostic biomarkers in digestive system cancers; and (3) immunogenic cell death for anticancer chemotherapy. Results: Most cell and animal experiments showed inhibition of autophagy by either pharmacological approaches or via genetic silencing of autophagy regulatory gene, resulting in a promotion of 5-FU-induced cancer cells death. Meanwhile, autophagy also plays a pro-death role and may mediate cell death in certain cancer cells where apoptosis is defective or difficult to induce. The dual role of autophagy complicates the use of autophagy inhibitor or inducer in cancer chemotherapy and generates inconsistency to an extent in clinic trials. Conclusion: Autophagy might be a therapeutic target that sensitizes the 5-FU treatment in gastrointestinal cancer.
Collapse
Affiliation(s)
| | | | | | - Xiu-Jun Cai
- Department of General Surgery, Zhejiang Province Key Laboratory of Laparosopic Technology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang 310016, China
| |
Collapse
|
12
|
Abstract
Macroautophagy/autophagy is a conserved lysosomal degradation process essential for cell physiology and human health. By regulating apoptosis, inflammation, pathogen clearance, immune response and other cellular processes, autophagy acts as a modulator of pathogenesis and is a potential therapeutic target in diverse diseases. With regard to oral disease, autophagy can be problematic either when it is activated or impaired, because this process is involved in diverse functions, depending on the specific disease and its level of progression. In particular, activated autophagy functions as a cytoprotective mechanism under environmental stress conditions, which regulates tumor growth and mediates resistance to anticancer treatment in established tumors. During infections and inflammation, activated autophagy selectively delivers microbial antigens to the immune systems, and is therefore connected to the elimination of intracellular pathogens. Impaired autophagy contributes to oxidative stress, genomic instability, chronic tissue damage, inflammation and tumorigenesis, and is involved in aberrant bacterial clearance and immune priming. Hence, substantial progress in the study of autophagy provides new insights into the pathogenesis of oral diseases. This review outlines the mechanisms of autophagy, and highlights the emerging roles of this process in oral cancer, periapical lesions, periodontal diseases, and oral candidiasis.
Collapse
Affiliation(s)
- Ya-Qin Tan
- a The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education , School and Hospital of Stomatology, Wuhan University , Wuhan , Hubei , China
| | - Jing Zhang
- a The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education , School and Hospital of Stomatology, Wuhan University , Wuhan , Hubei , China.,b Department of Oral Medicine , School and Hospital of Stomatology, Wuhan University , Wuhan , Hubei , China
| | - Gang Zhou
- a The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education , School and Hospital of Stomatology, Wuhan University , Wuhan , Hubei , China.,b Department of Oral Medicine , School and Hospital of Stomatology, Wuhan University , Wuhan , Hubei , China
| |
Collapse
|
13
|
Cosway B, Lovat P. The role of autophagy in squamous cell carcinoma of the head and neck. Oral Oncol 2016; 54:1-6. [PMID: 26774913 DOI: 10.1016/j.oraloncology.2015.12.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 12/01/2015] [Accepted: 12/19/2015] [Indexed: 01/07/2023]
Abstract
Half a million new head and neck cancers are diagnosed each year worldwide. Although traditionally thought to be triggered by alcohol and smoking abuse, there is a growing subset of oropharyngeal cancers driven by the oncogenic human papilloma virus (HPV). Despite advances in both surgical and non-surgical treatment strategies, survival rates have remained relatively static emphasising the need for novel therapeutic approaches. Autophagy, the principal catabolic process for the lysosomal--mediated breakdown of cellular products is a hot topic in cancer medicine. Increasing evidence points towards the prognostic significance of autophagy biomarkers in solid tumours as well as strategies through which to harness autophagy modulation to promote tumour cell death. However, the role of autophagy in head and neck cancers is less well defined. In the present review, we summarise the current understanding of autophagy in head and neck cancers, revealing key areas for future translational research.
Collapse
Affiliation(s)
- Benjamin Cosway
- Institute for Cellular Medicine, Newcastle University, United Kingdom.
| | - Penny Lovat
- Institute for Cellular Medicine, Newcastle University, United Kingdom
| |
Collapse
|
14
|
Li YC, Chang JT, Chiu C, Lu YC, Li YL, Chiang CH, You GR, Lee LY, Cheng AJ. Areca nut contributes to oral malignancy through facilitating the conversion of cancer stem cells. Mol Carcinog 2015; 55:1012-23. [PMID: 26087469 DOI: 10.1002/mc.22344] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/08/2015] [Accepted: 05/13/2015] [Indexed: 01/06/2023]
Abstract
Oral cancer is one of the most frequent malignant diseases worldwide, and areca nut is a primary carcinogen causing this cancer in Southeast Asia. Previous studies to examine the effects of this carcinogen often used short-term and high-dose treatment of area nut extract as a research model, which do not recapitulate the conditions of patients with long-term and habitual use of this substance. To approach authentic mechanism of areca nut-induced oral carcinogenesis that occurs in human, we established four isogenic sublines of oral cells which were chronic exposed to areca nut extract. Without eliciting cytotoxicity or senescence, these four sublines cells exhibited significant increase in invasive ability, along with epithelial-mesenchymal transition. These cells also showed resistance to chemotherapeutic drug and irradiation, accompanying with the augmentation of ABCG2 protein efflux and increased ROS clearance. Moreover, these sublines possessed the characteristics of cancer stemness, as demonstrated by enriched CD24-/CD44+ and CD133+ sub-populations, enhanced spheroid cell formation, and induced expressions of pluripotent stemness regulators, including Gp96, Grp78, Slug, Sox9, Snail, and Foxc2. These stemness regulators were further shown up-regulations in oral cancer patients with areca nut-chewing habit, and were statistically correlated with CD44 expression, a stemness marker. In conclusion, our findings suggested that areca nut contributes to oral malignancy through facilitating the conversion of cancer stem cells. This study may further contribute to clinical applications in disease prevention, risk assessment or molecular therapeutics on areca nut- associated diseases.
Collapse
Affiliation(s)
- Yi-Chen Li
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Joseph T Chang
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Crystal Chiu
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ya-Ching Lu
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yan-Liang Li
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chang-Hsu Chiang
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Guo-Rung You
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ann-Joy Cheng
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
15
|
Yen CY, Chiang WF, Liu SY, Lin CC, Liao KA, Lin CY, Hsieh WF, Cheng YC, Hsu KC, Lin PY, Chen TC, Lee IL, Lin MH, Liu YC. Impacts of autophagy-inducing ingredient of areca nut on tumor cells. PLoS One 2015; 10:e0128011. [PMID: 26017803 PMCID: PMC4445909 DOI: 10.1371/journal.pone.0128011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/21/2015] [Indexed: 12/17/2022] Open
Abstract
Areca nut (AN) is a popular carcinogen used by about 0.6-1.2 billion people worldwide. Although AN contains apoptosis-inducing ingredients, we previously demonstrated that both AN extract (ANE) and its 30-100 kDa fraction (ANE 30-100K) predominantly induce autophagic cell death in both normal and malignant cells. In this study, we further explored the action mechanism of ANE 30-100K-induced autophagy (AIA) in Jurkat T lymphocytes and carcinoma cell lines including OECM-1 (mouth), CE81T/VGH (esophagus), SCC25 (tongue), and SCC-15 (tongue). The results showed that chemical- and small hairpin RNA (shRNA)-mediated inhibition of AMP-activated protein kinase (AMPK) resulted in the attenuation of AIA in Jurkat T but not in OECM-1 cells. Knockdown of Atg5 and Beclin 1 expressions ameliorated AIA in OECM-1/CE81T/VGH/Jurkat T and OECM-1/SCC25/SCC-15, respectively. Furthermore, ANE 30-100K could activate caspase-3 after inhibition of Beclin 1 expression in OECM-1/SCC25/SCC15 cells. Meanwhile, AMPK was demonstrated to be the upstream activator of the extracellular-regulated kinase (ERK) in Jurkat T cells, and inhibition of MEK attenuated AIA in Jurkat T/OECM-1/CE81T/VGH cells. Finally, we also found that multiple myeloma RPMI8226, lymphoma U937, and SCC15 cells survived from long-term non-cytotoxic ANE 30-100K treatment exhibited stronger resistance against serum deprivation through upregulated autophagy. Collectively, our studies indicate that Beclin-1 and Atg5 but not AMPK are commonly required for AIA, and MEK/ERK pathway is involved in AIA. Meanwhile, it is also suggested that long-term AN usage might increase the resistance of survived tumor cells against serum-limited conditions.
Collapse
Affiliation(s)
- Ching-Yu Yen
- Department of Dentistry, Taipei Medical University, Taipei, Taiwan
- Department of Dentistry, National Defense Medical Center, Taipei, Taiwan
- Oral and Maxillofacial Surgery Section, Chi Mei Medical Center, Tainan, Taiwan
| | - Wei-Fan Chiang
- Oral and Maxillofacial Surgery Section, Chi Mei Hospital, Liouying, Taiwan
- School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Shyun-Yeu Liu
- Department of Dentistry, Taipei Medical University, Taipei, Taiwan
- Department of Dentistry, National Defense Medical Center, Taipei, Taiwan
- Oral and Maxillofacial Surgery Section, Chi Mei Medical Center, Tainan, Taiwan
| | - Chung-Chih Lin
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Kuo-An Liao
- Oral and Maxillofacial Surgery Section, Chi Mei Hospital, Liouying, Taiwan
| | - Che-Yi Lin
- Oral and Maxillofacial Surgery Section, Chi Mei Hospital, Liouying, Taiwan
| | - Wan-Fang Hsieh
- Department of Biotechnology, Chia Nan University of Pharmacy, Tainan, Taiwan
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Yon-Chi Cheng
- Department of Biotechnology, Chia Nan University of Pharmacy, Tainan, Taiwan
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Kai-Cheng Hsu
- Department of Biotechnology, Chia Nan University of Pharmacy, Tainan, Taiwan
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Pin-Yen Lin
- Department of Biotechnology, Chia Nan University of Pharmacy, Tainan, Taiwan
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Tai-Chi Chen
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - I-Ling Lee
- Oral and Maxillofacial Surgery Section, Chi Mei Hospital, Liouying, Taiwan
| | - Mei-Huei Lin
- Department of Biotechnology, Chia Nan University of Pharmacy, Tainan, Taiwan
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
- * E-mail: (MHL); (YCL)
| | - Young-Chau Liu
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
- Division of Natural Science, College of Liberal Education, Shu-Te University, Kaohsiung, Taiwan
- * E-mail: (MHL); (YCL)
| |
Collapse
|